1
|
Klatt D, Sereni L, Liu B, Genovese P, Schambach A, Verhoeyen E, Williams DA, Brendel C. Engineered packaging cell line for the enhanced production of baboon-enveloped retroviral vectors. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102389. [PMID: 39679008 PMCID: PMC11638596 DOI: 10.1016/j.omtn.2024.102389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024]
Abstract
The baboon endogenous retrovirus (BaEV) glycoprotein is superior to the commonly used vesicular stomatitis virus glycoprotein (VSVg) for retroviral gene transfer into resting hematopoietic stem cells and lymphocyte populations. The derivative BaEVRLess (lacking the R domain) produces higher viral titers compared with wild-type BaEV, but vector production is impaired by syncytia formation and cell death of the HEK293T cells due to the high fusogenic activity of the glycoprotein. This lowers viral titers, leads to increased batch-to-batch variability, and impedes the establishment of stable packaging cell lines essential for the economical production of viral supernatants. Here, we show that knockout of the entry receptor ASCT2 in HEK293T producer cells eliminates syncytia formation, resulting in a 2-fold increase in viral titers, reduced toxicity of viral supernatants, and enables the generation of stable packaging cell lines. In successive steps, we stably integrated BaEVRLess and α-retroviral a.Gag/Pol expression cassettes and isolated clones supporting titers up to 108 to 109 infectious particles/mL, a 10-fold increase in concentrated viral titers. The additional overexpression of CD47 and knockout of β2-microglobulin in the packaging cell line are tailored for future use in in vivo gene therapy applications by reducing non-specific uptake by macrophages and the immunogenicity of viral particles.
Collapse
Affiliation(s)
- Denise Klatt
- Gene Therapy Program, Dana Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lucia Sereni
- Gene Therapy Program, Dana Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Boya Liu
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pietro Genovese
- Gene Therapy Program, Dana Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Axel Schambach
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Els Verhoeyen
- Centre International de Recherche en Infectiologie (CIRI), Université Lyon, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR 5308, Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Université Côte d'Azur, INSERM U1065, Centre Méditerranéen de Médecine Moléculaire, 06200 Nice, France
| | - David A. Williams
- Gene Therapy Program, Dana Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christian Brendel
- Gene Therapy Program, Dana Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
2
|
Kitawi R, Ledger S, Kelleher AD, Ahlenstiel CL. Advances in HIV Gene Therapy. Int J Mol Sci 2024; 25:2771. [PMID: 38474018 DOI: 10.3390/ijms25052771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Early gene therapy studies held great promise for the cure of heritable diseases, but the occurrence of various genotoxic events led to a pause in clinical trials and a more guarded approach to progress. Recent advances in genetic engineering technologies have reignited interest, leading to the approval of the first gene therapy product targeting genetic mutations in 2017. Gene therapy (GT) can be delivered either in vivo or ex vivo. An ex vivo approach to gene therapy is advantageous, as it allows for the characterization of the gene-modified cells and the selection of desired properties before patient administration. Autologous cells can also be used during this process which eliminates the possibility of immune rejection. This review highlights the various stages of ex vivo gene therapy, current research developments that have increased the efficiency and safety of this process, and a comprehensive summary of Human Immunodeficiency Virus (HIV) gene therapy studies, the majority of which have employed the ex vivo approach.
Collapse
Affiliation(s)
- Rose Kitawi
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Scott Ledger
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Anthony D Kelleher
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
- St. Vincent's Hospital, Darlinghurst, NSW 2010, Australia
- UNSW RNA Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Chantelle L Ahlenstiel
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
- UNSW RNA Institute, University of New South Wales, Kensington, NSW 2052, Australia
| |
Collapse
|
3
|
Valeri E, Unali G, Piras F, Abou-Alezz M, Pais G, Benedicenti F, Lidonnici MR, Cuccovillo I, Castiglioni I, Arévalo S, Spinozzi G, Merelli I, Behrendt R, Oo A, Kim B, Landau NR, Ferrari G, Montini E, Kajaste-Rudnitski A. Removal of innate immune barriers allows efficient transduction of quiescent human hematopoietic stem cells. Mol Ther 2024; 32:124-139. [PMID: 37990494 PMCID: PMC10787167 DOI: 10.1016/j.ymthe.2023.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/29/2023] [Accepted: 11/17/2023] [Indexed: 11/23/2023] Open
Abstract
Quiescent human hematopoietic stem cells (HSC) are ideal targets for gene therapy applications due to their preserved stemness and repopulation capacities; however, they have not been exploited extensively because of their resistance to genetic manipulation. We report here the development of a lentiviral transduction protocol that overcomes this resistance in long-term repopulating quiescent HSC, allowing their efficient genetic manipulation. Mechanistically, lentiviral vector transduction of quiescent HSC was found to be restricted at the level of vector entry and by limited pyrimidine pools. These restrictions were overcome by the combined addition of cyclosporin H (CsH) and deoxynucleosides (dNs) during lentiviral vector transduction. Clinically relevant transduction levels were paired with higher polyclonal engraftment of long-term repopulating HSC as compared with standard ex vivo cultured controls. These findings identify the cell-intrinsic barriers that restrict the transduction of quiescent HSC and provide a means to overcome them, paving the way for the genetic engineering of unstimulated HSC.
Collapse
Affiliation(s)
- Erika Valeri
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Giulia Unali
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Francesco Piras
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Monah Abou-Alezz
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulia Pais
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Maria Rosa Lidonnici
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ivan Cuccovillo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ilaria Castiglioni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sergio Arévalo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulio Spinozzi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Rayk Behrendt
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Adrian Oo
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Baek Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nathaniel R Landau
- Department of Microbiology, NYU School of Medicine, New York, NY 10016, USA
| | - Giuliana Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9A, 27100 Pavia, Italy.
| |
Collapse
|
4
|
Manesia JK, Maganti HB, Almoflehi S, Jahan S, Hasan T, Pasha R, McGregor C, Dumont N, Laganière J, Audet J, Pineault N. AA2P-mediated DNA demethylation synergizes with stem cell agonists to promote expansion of hematopoietic stem cells. CELL REPORTS METHODS 2023; 3:100663. [PMID: 38070507 PMCID: PMC10783628 DOI: 10.1016/j.crmeth.2023.100663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 06/28/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023]
Abstract
Small molecules have enabled expansion of hematopoietic stem and progenitor cells (HSPCs), but limited knowledge is available on whether these agonists can act synergistically. In this work, we identify a stem cell agonist in AA2P and optimize a series of stem cell agonist cocktails (SCACs) to help promote robust expansion of human HSPCs. We find that SCACs provide strong growth-promoting activities while promoting retention and function of immature HSPC. We show that AA2P-mediated HSPC expansion is driven through DNA demethylation leading to enhanced expression of AXL and GAS6. Further, we demonstrate that GAS6 enhances the serial engraftment activity of HSPCs and show that the GAS6/AXL pathway is critical for robust HSPC expansion.
Collapse
Affiliation(s)
- Javed K Manesia
- Canadian Blood Services, Centre for Innovation, Ottawa, ON, Canada
| | - Harinad B Maganti
- Canadian Blood Services, Centre for Innovation, Ottawa, ON, Canada; Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, Canada
| | - Sakhar Almoflehi
- Canadian Blood Services, Centre for Innovation, Ottawa, ON, Canada; Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, Canada
| | - Suria Jahan
- Canadian Blood Services, Centre for Innovation, Ottawa, ON, Canada; Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, Canada
| | - Tanvir Hasan
- Canadian Blood Services, Centre for Innovation, Ottawa, ON, Canada; Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, Canada
| | - Roya Pasha
- Canadian Blood Services, Centre for Innovation, Ottawa, ON, Canada
| | - Chelsea McGregor
- Canadian Blood Services, Centre for Innovation, Ottawa, ON, Canada
| | | | | | - Julie Audet
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Nicolas Pineault
- Canadian Blood Services, Centre for Innovation, Ottawa, ON, Canada; Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
5
|
Rai R, Naseem A, Vetharoy W, Steinberg Z, Thrasher AJ, Santilli G, Cavazza A. An improved medium formulation for efficient ex vivo gene editing, expansion and engraftment of hematopoietic stem and progenitor cells. Mol Ther Methods Clin Dev 2023; 29:58-69. [PMID: 36950452 PMCID: PMC10025975 DOI: 10.1016/j.omtm.2023.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/24/2023] [Indexed: 03/04/2023]
Abstract
Gene editing has emerged as a powerful tool for the therapeutic correction of monogenic diseases. CRISPR-Cas9 applied to hematopoietic stem and progenitor cells (HSPCs) has shown great promise in proof-of-principle preclinical studies to treat hematological disorders, and clinical trials using these tools are now under way. Nonetheless, there remain important challenges that need to be addressed, such as the efficiency of targeting primitive, long-term repopulating HSPCs and their in vitro expansion for clinical application. In this study, we assessed the effect of different culture medium compositions on the ability of HSPCs to proliferate and undergo homology-directed repair-mediated knock-in of a reporter gene, while preserving their stemness features during ex vivo culture. We demonstrated that by supplementing the culture medium with stem cell agonists and by fine-tuning its cytokine composition it is possible to achieve high levels of gene targeting in long-term repopulating HSPCs both in vitro and in vivo, with a beneficial balance between preservation of stemness and cell expansion. Overall, the implementation of this optimized ex vivo HSPC culture protocol can improve the efficacy, feasibility, and applicability of gene editing as a key step to unlocking the full therapeutic potential of this powerful technology.
Collapse
Affiliation(s)
- Rajeev Rai
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Asma Naseem
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Winston Vetharoy
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Zohar Steinberg
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Adrian J. Thrasher
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Giorgia Santilli
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Alessia Cavazza
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
6
|
Allen D, Kalter N, Rosenberg M, Hendel A. Homology-Directed-Repair-Based Genome Editing in HSPCs for the Treatment of Inborn Errors of Immunity and Blood Disorders. Pharmaceutics 2023; 15:1329. [PMID: 37242571 PMCID: PMC10220672 DOI: 10.3390/pharmaceutics15051329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Genome engineering via targeted nucleases, specifically CRISPR-Cas9, has revolutionized the field of gene therapy research, providing a potential treatment for diseases of the blood and immune system. While numerous genome editing techniques have been used, CRISPR-Cas9 homology-directed repair (HDR)-mediated editing represents a promising method for the site-specific insertion of large transgenes for gene knock-in or gene correction. Alternative methods, such as lentiviral/gammaretroviral gene addition, gene knock-out via non-homologous end joining (NHEJ)-mediated editing, and base or prime editing, have shown great promise for clinical applications, yet all possess significant drawbacks when applied in the treatment of patients suffering from inborn errors of immunity or blood system disorders. This review aims to highlight the transformational benefits of HDR-mediated gene therapy and possible solutions for the existing problems holding the methodology back. Together, we aim to help bring HDR-based gene therapy in CD34+ hematopoietic stem progenitor cells (HSPCs) from the lab bench to the bedside.
Collapse
Affiliation(s)
| | | | | | - Ayal Hendel
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (D.A.); (N.K.); (M.R.)
| |
Collapse
|
7
|
Hematopoietic Stem Cell Mobilization: Current Collection Approaches, Stem Cell Heterogeneity, and a Proposed New Method for Stem Cell Transplant Conditioning. Stem Cell Rev Rep 2021; 17:1939-1953. [PMID: 34661830 DOI: 10.1007/s12015-021-10272-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 10/20/2022]
Abstract
Hematopoietic stem cells naturally traffic out of their bone marrow niches into the peripheral blood. This natural trafficking process can be enhanced with numerous pharmacologic agents - a process termed "mobilization" - and the mobilized stem cells can be collected for transplantation. We review the current state of mobilization with an update on recent clinical trials and new biologic mechanisms regulating stem cell trafficking. We propose that hematopoietic mobilization can be used to answer questions regarding hematopoietic stem cell heterogeneity, can be used for non-toxic conditioning of patients receiving stem cell transplants, and can enhance gene editing and gene therapy strategies to cure genetic diseases.
Collapse
|
8
|
Christopher AC, Venkatesan V, Karuppusamy KV, Srinivasan S, Babu P, Azhagiri MKK, C K, Bagchi A, Rajendiran V, Ravi NS, Kumar S, Marepally SK, Mohankumar KM, Srivastava A, Velayudhan SR, Thangavel S. Preferential expansion of human CD34+CD133+CD90+ hematopoietic stem cells enhances gene-modified cell frequency for gene therapy. Hum Gene Ther 2021; 33:188-201. [PMID: 34486377 DOI: 10.1089/hum.2021.089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
CD34+CD133+CD90+ hematopoietic stem cells (HSCs) are responsible for long-term multi-lineage hematopoiesis and the high frequency of gene-modified HSCs is crucial for the success of hematopoietic stem and progenitor cell (HSPC) gene therapy. However, the ex vivo culture and gene manipulation steps of HSPC graft preparation significantly reduce the frequency of HSCs, thus necessitating large doses of HSPCs and reagents for the manipulation. Here, we identified a combination of small molecules, Resveratrol, UM729, and SR1 that preferentially expands CD34+CD133+CD90+ HSCs over other subpopulations of adult HSPCs in ex vivo culture. The preferential expansion enriches the HSCs in ex vivo culture, enhances the adhesion and results in a 6-fold increase in the long-term engraftment in NSG mice. Further, the culture enriched HSCs are more responsive to gene modification by lentiviral transduction and gene editing, increasing the frequency of gene-modified HSCs up to 10-fold in vivo. The yield of gene-modified HSCs obtained by the culture enrichment is similar to the sort-purification of HSCs and superior to Cyclosporin-H treatment. Our study addresses a critical challenge of low frequency of gene-modified HSCs in HSPC graft by developing and demonstrating a facile HSPC culture condition that increases the frequency of gene-modified cells in vivo. This strategy will improve the outcome of HSPC gene therapy and also simplify the gene manipulation process.
Collapse
Affiliation(s)
| | - Vigneshwaran Venkatesan
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India.,Manipal Academy of Higher Education, 76793, Manipal, Karnataka, India;
| | - Karthik V Karuppusamy
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India.,Manipal Academy of Higher Education, 76793, Manipal, Karnataka, India;
| | | | - Prathibha Babu
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India.,Manipal Academy of Higher Education, 76793, Manipal, Karnataka, India;
| | - Manoj Kumar K Azhagiri
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India.,Manipal Academy of Higher Education, 76793, Manipal, Karnataka, India;
| | - Karthik C
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India;
| | - Abhirup Bagchi
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India;
| | | | - Nithin Sam Ravi
- Center for Stem Cell Research, 302927, Vellore, Tamil Nadu, India;
| | - Sanjay Kumar
- Christian Medical College and Hospital Vellore, 30025, Center for Stem Cell Research, Vellore, Tamil Nadu, India;
| | | | | | - Alok Srivastava
- Christian Medical College, Centre for Stem Cell Research, CMC Campus, Bagayam, Vellore, Tamilnadu, India, 632002.,Christian Medical College, Haematology, Ida Scudder Road, Vellore, Tamil Nadu, India, 632004;
| | | | - Saravanabhavan Thangavel
- Center for Stem Cell Research, 302927, Christian Medical College Campus Bagayam,, Vellore, Tamil nadu, India, 632002;
| |
Collapse
|
9
|
Piras F, Kajaste-Rudnitski A. Antiviral immunity and nucleic acid sensing in haematopoietic stem cell gene engineering. Gene Ther 2021; 28:16-28. [PMID: 32661282 PMCID: PMC7357672 DOI: 10.1038/s41434-020-0175-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
Abstract
The low gene manipulation efficiency of human hematopoietic stem and progenitor cells (HSPC) remains a major hurdle for sustainable and broad clinical application of innovative therapies for a wide range of disorders. Given that all current and emerging gene transfer and editing technologies are bound to expose HSPC to exogenous nucleic acids and most often also to viral vectors, we reason that host antiviral factors and nucleic acid sensors play a pivotal role in the efficacy of HSPC genetic manipulation. Here, we review recent progress in our understanding of vector-host interactions and innate immunity in HSPC upon gene engineering and discuss how dissecting this crosstalk can guide the development of more stealth and efficient gene therapy approaches in the future.
Collapse
Affiliation(s)
- Francesco Piras
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
10
|
Johnson C, Belluschi S, Laurenti E. Beyond “to divide or not to divide”: Kinetics matters in hematopoietic stem cells. Exp Hematol 2020; 92:1-10.e2. [DOI: 10.1016/j.exphem.2020.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/06/2020] [Accepted: 11/07/2020] [Indexed: 01/03/2023]
|
11
|
K562 cell-derived exosomes suppress the adhesive function of bone marrow mesenchymal stem cells via delivery of miR-711. Biochem Biophys Res Commun 2020; 521:584-589. [DOI: 10.1016/j.bbrc.2019.10.096] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 10/11/2019] [Indexed: 01/20/2023]
|
12
|
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
|
13
|
Petrillo C, Calabria A, Piras F, Capotondo A, Spinozzi G, Cuccovillo I, Benedicenti F, Naldini L, Montini E, Biffi A, Gentner B, Kajaste-Rudnitski A. Assessing the Impact of Cyclosporin A on Lentiviral Transduction and Preservation of Human Hematopoietic Stem Cells in Clinically Relevant Ex Vivo Gene Therapy Settings. Hum Gene Ther 2019; 30:1133-1146. [PMID: 31037976 PMCID: PMC6761585 DOI: 10.1089/hum.2019.016] [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] [Indexed: 02/06/2023] Open
Abstract
Improving hematopoietic stem and progenitor cell (HSPC) permissiveness to lentiviral vector (LV) transduction without compromising their biological properties remains critical for broad-range implementation of gene therapy as a treatment option for several inherited diseases. This study demonstrates that the use of one-hit ex vivo LV transduction protocols based on either cyclosporin A (CsA) or rapamycin enable as efficient gene transfer as the current two-hit clinical standard into bone marrow-derived CD34+ cells while better preserving their engraftment capacity in vivo. CsA was additive with another enhancer of transduction, prostaglandin E2, suggesting that tailored enhancer combinations may be applied to overcome multiple blocks to transduction simultaneously in HSPC. Interestingly, besides enhancing LV transduction, CsA also significantly reduced HSPC proliferation, preserving the quiescent G0 fraction and the more primitive multipotent progenitors, thereby yielding the highest engraftment levels in vivo. Importantly, no alterations in the vector integration profiles could be detected between CsA and control transduced HSPC. Overall, the present findings contribute to the development of more efficient and sustainable LV gene therapy protocols, underscoring the benefits of scaling down required vector doses, as well as shortening the HSPC ex vivo culture time.
Collapse
Affiliation(s)
- Carolina Petrillo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Piras
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Alessia Capotondo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giulio Spinozzi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ivan Cuccovillo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandra Biffi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy.,Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts.,Program for Gene Therapy in Rare Diseases, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
14
|
Gene therapy targeting haematopoietic stem cells for inherited diseases: progress and challenges. Nat Rev Drug Discov 2019; 18:447-462. [DOI: 10.1038/s41573-019-0020-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
Li C, Psatha N, Gil S, Wang H, Papayannopoulou T, Lieber A. HDAd5/35 ++ Adenovirus Vector Expressing Anti-CRISPR Peptides Decreases CRISPR/Cas9 Toxicity in Human Hematopoietic Stem Cells. Mol Ther Methods Clin Dev 2018; 9:390-401. [PMID: 30038942 PMCID: PMC6054697 DOI: 10.1016/j.omtm.2018.04.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/26/2018] [Indexed: 01/12/2023]
Abstract
We generated helper-dependent HDAd5/35++ adenovirus vectors expressing CRISPR/Cas9 for potential hematopoietic stem cells (HSCs) gene therapy of β-thalassemia and sickle cell disease through re-activation of fetal γ-globin expression (HDAd-globin-CRISPR). The process of CRISPR/Cas9 gene transfer using these vectors was not associated with death of human CD34+ cells and did not affect their in vitro expansion and erythroid differentiation. However, functional assays for primitive HSCs, e.g., multi-lineage progenitor colony formation and engraftment in irradiated NOD/Shi-scid/interleukin-2 receptor γ (IL-2Rγ) null (NSG) mice, revealed toxicity of HDAd-globin-CRISPR vectors related to the prolonged expression and activity of CRISPR/Cas9. To control the duration of CRISPR/Cas9 activity, we generated an HDAd5/35++ vector that expressed two anti-CRISPR (Acr) peptides (AcrII4 and AcrII2) capable of binding to the CRISPR/Cas9 complex (HDAd-Acr). CD34+ cells that were sequentially infected with HDAd-CRISPR and HDAd-Acr engrafted at a significantly higher rate. Target site disruption frequencies in engrafted human cells were similar to those in pre-transplantation CD34+ cells, indicating that genome-edited primitive HSCs survived. In vitro differentiated HSCs isolated from transplanted mice demonstrated increased γ-globin expression as a result of genome editing. Our data indicate that the HDAd-Acr vector can be used as a tool to reduce HSC cytotoxicity of the CRISPR/Cas9 complex.
Collapse
Affiliation(s)
- Chang Li
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
| | - Nikoletta Psatha
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Sucheol Gil
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
| | - Hongjie Wang
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
| | - Thalia Papayannopoulou
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
16
|
Li C, Psatha N, Wang H, Singh M, Samal HB, Zhang W, Ehrhardt A, Izsvák Z, Papayannopoulou T, Lieber A. Integrating HDAd5/35++ Vectors as a New Platform for HSC Gene Therapy of Hemoglobinopathies. Mol Ther Methods Clin Dev 2018; 9:142-152. [PMID: 29766024 PMCID: PMC5948227 DOI: 10.1016/j.omtm.2018.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/08/2018] [Indexed: 12/22/2022]
Abstract
We generated an integrating, CD46-targeted, helper-dependent adenovirus HDAd5/35++ vector system for hematopoietic stem cell (HSC) gene therapy. The ∼12-kb transgene cassette included a β-globin locus control region (LCR)/promoter driven human γ-globin gene and an elongation factor alpha-1 (EF1α)-mgmtP140K expression cassette, which allows for drug-controlled increase of γ-globin-expressing erythrocytes. We transduced bone marrow lineage-depleted cells from human CD46-transgenic mice and transplanted them into lethally irradiated recipients. The percentage of γ-globin-positive cells in peripheral blood erythrocytes in primary and secondary transplant recipients was stable and greater than 90%. The γ-globin level was 10%-20% of adult mouse globin. Transgene integration, mediated by a hyperactive Sleeping Beauty SB100x transposase, was random, without a preference for genes. A second set of studies was performed with peripheral blood CD34+ cells from mobilized donors. 10 weeks after transplantation of transduced cells, human cells were harvested from the bone marrow and differentiated ex vivo into erythroid cells. Erythroid cells expressed γ-globin at a level of 20% of adult α-globin. Our studies suggest that HDAd35++ vectors allow for efficient transduction of long-term repopulating HSCs and high-level, almost pancellular γ-globin expression in erythrocytes. Furthermore, our HDAd5/35++ vectors have a larger insert capacity and a safer integration pattern than currently used lentivirus vectors.
Collapse
Affiliation(s)
- Chang Li
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
| | - Nikoletta Psatha
- Division of Hematology Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Hongjie Wang
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
| | - Manvendra Singh
- Max-Delbrück-Center for Molecular Medicine, Berlin, 13092 Germany
| | | | - Wenli Zhang
- Witten/Herdecke University, Witten, 58448, Germany
| | | | - Zsuzsanna Izsvák
- Max-Delbrück-Center for Molecular Medicine, Berlin, 13092 Germany
| | - Thalia Papayannopoulou
- Division of Hematology Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
17
|
Sii-Felice K, Giorgi M, Leboulch P, Payen E. Hemoglobin disorders: lentiviral gene therapy in the starting blocks to enter clinical practice. Exp Hematol 2018; 64:12-32. [PMID: 29807062 DOI: 10.1016/j.exphem.2018.05.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/18/2018] [Accepted: 05/19/2018] [Indexed: 01/19/2023]
Abstract
The β-hemoglobinopathies, transfusion-dependent β-thalassemia and sickle cell disease, are the most prevalent inherited disorders worldwide and affect millions of people. Many of these patients have a shortened life expectancy and suffer from severe morbidity despite supportive therapies, which impose an enormous financial burden to societies. The only available curative therapy is allogeneic hematopoietic stem cell transplantation, although most patients do not have an HLA-matched sibling donor, and those who do still risk life-threatening complications. Therefore, gene therapy by one-time ex vivo modification of hematopoietic stem cells followed by autologous engraftment is an attractive new therapeutic modality. The first proof-of-principle of conversion to transfusion independence by means of a lentiviral vector expressing a marked and anti-sickling βT87Q-globin gene variant was reported a decade ago in a patient with transfusion-dependent β-thalassemia. In follow-up multicenter Phase II trials with an essentially identical vector (termed LentiGlobin BB305) and protocol, 12 of the 13 patients with a non-β0/β0 genotype, representing more than half of all transfusion-dependent β-thalassemia cases worldwide, stopped red blood cell transfusions with total hemoglobin levels in blood approaching normal values. Correction of biological markers of dyserythropoiesis was achieved in evaluated patients. In nine patients with β0/β0 transfusion-dependent β-thalassemia or equivalent severity (βIVS1-110), median annualized transfusion volume decreased by 73% and red blood cell transfusions were stopped in three patients. Proof-of-principle of therapeutic efficacy in the first patient with sickle cell disease was also reported with LentiGlobin BB305. Encouraging results were presented in children with transfusion-dependent β-thalassemia in another trial with the GLOBE lentiviral vector and several other gene therapy trials are currently open for both transfusion-dependent β-thalassemia and sickle cell disease. Phase III trials are now under way and should help to determine benefit/risk/cost ratios to move gene therapy toward clinical practice.
Collapse
Affiliation(s)
- Karine Sii-Felice
- UMR E007, Service of Innovative Therapies, Institute of Biology François Jacob and University Paris Saclay, CEA Paris Saclay, Fontenay-aux-Roses, France
| | - Marie Giorgi
- UMR E007, Service of Innovative Therapies, Institute of Biology François Jacob and University Paris Saclay, CEA Paris Saclay, Fontenay-aux-Roses, France
| | - Philippe Leboulch
- UMR E007, Service of Innovative Therapies, Institute of Biology François Jacob and University Paris Saclay, CEA Paris Saclay, Fontenay-aux-Roses, France; Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Emmanuel Payen
- UMR E007, Service of Innovative Therapies, Institute of Biology François Jacob and University Paris Saclay, CEA Paris Saclay, Fontenay-aux-Roses, France; INSERM, Paris, France.
| |
Collapse
|
18
|
Reactivation of γ-globin in adult β-YAC mice after ex vivo and in vivo hematopoietic stem cell genome editing. Blood 2018; 131:2915-2928. [PMID: 29789357 DOI: 10.1182/blood-2018-03-838540] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022] Open
Abstract
Disorders involving β-globin gene mutations, primarily β-thalassemia and sickle cell disease, represent a major target for hematopoietic stem/progenitor cell (HSPC) gene therapy. This includes CRISPR/Cas9-mediated genome editing approaches in adult CD34+ cells aimed toward the reactivation of fetal γ-globin expression in red blood cells. Because models involving erythroid differentiation of CD34+ cells have limitations in assessing γ-globin reactivation, we focused on human β-globin locus-transgenic (β-YAC) mice. We used a helper-dependent human CD46-targeting adenovirus vector expressing CRISPR/Cas9 (HDAd-HBG-CRISPR) to disrupt a repressor binding region within the γ-globin promoter. We transduced HSPCs from β-YAC/human CD46-transgenic mice ex vivo and subsequently transplanted them into irradiated recipients. Furthermore, we used an in vivo HSPC transduction approach that involves HSPC mobilization and the intravenous injection of HDAd-HBG-CRISPR into β-YAC/CD46-transgenic mice. In both models, we demonstrated efficient target site disruption, resulting in a pronounced switch from human β- to γ-globin expression in red blood cells of adult mice that was maintained after secondary transplantation of HSPCs. In long-term follow-up studies, we did not detect hematological abnormalities, indicating that HBG promoter editing does not negatively affect hematopoiesis. This is the first study that shows successful in vivo HSPC genome editing by CRISPR/Cas9.
Collapse
|
19
|
Wang H, Richter M, Psatha N, Li C, Kim J, Liu J, Ehrhardt A, Nilsson SK, Cao B, Palmer D, Ng P, Izsvák Z, Haworth KG, Kiem HP, Papayannopoulou T, Lieber A. A Combined In Vivo HSC Transduction/Selection Approach Results in Efficient and Stable Gene Expression in Peripheral Blood Cells in Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 8:52-64. [PMID: 29255741 PMCID: PMC5722719 DOI: 10.1016/j.omtm.2017.11.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/21/2022]
Abstract
We recently reported on an in vivo hematopoietic stem cell (HSC) gene therapy approach. It involves the subcutaneous injections of G-CSF/AMD3100 to mobilize HSCs from the bone marrow into the peripheral blood stream and the intravenous injection of an integrating helper-dependent adenovirus vector system. HSCs transduced in the periphery homed back to the bone marrow, where they persisted long-term. However, high transgene marking rates found in primitive bone marrow HSCs were not reflected in peripheral blood cells. Here, we tested small-molecule drugs to achieve selective mobilization and transduction of HSCs. We found more efficient GFP marking in bone marrow HSCs but no increased marking in the peripheral blood cells. We then used an in vivo HSC chemo-selection based on a mutant of the O6-methylguanine-DNA methyltransferase (mgmtP140K) gene that confers resistance to O6-BG/BCNU and should give stably transduced HSCs a proliferation stimulus and allow for the selective survival and expansion of progeny cells. Short-term exposure of G-CSF/AMD3100-mobilized, in vivo-transduced mice to relatively low selection drug doses resulted in stable GFP expression in up to 80% of peripheral blood cells. Overall, the further improvement of our in vivo HSC transduction approach creates the basis for a simpler HSC gene therapy.
Collapse
Affiliation(s)
- Hongjie Wang
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA 98195, USA
| | - Maximilian Richter
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA 98195, USA
| | - Nikoletta Psatha
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| | - Chang Li
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA 98195, USA
| | - Jiho Kim
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA 98195, USA
| | - Jing Liu
- Witten/Herdecke University, Witten, 58448, Germany
| | | | - Susan K Nilsson
- Biomedical Manufacturing, CSIRO, Clayton, VIC 3800, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Benjamin Cao
- Biomedical Manufacturing, CSIRO, Clayton, VIC 3800, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Donna Palmer
- Baylor College of Medicine, Houston, TX 77046, USA
| | - Philip Ng
- Baylor College of Medicine, Houston, TX 77046, USA
| | - Zsuzsanna Izsvák
- Max-Delbrück-Center for Molecular Medicine, Berlin 13092, Germany
| | - Kevin G Haworth
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Hans-Peter Kiem
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Thalia Papayannopoulou
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| | - André Lieber
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA 98195, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| |
Collapse
|
20
|
Bone Marrow Homing and Engraftment Defects of Human Hematopoietic Stem and Progenitor Cells. Mediterr J Hematol Infect Dis 2017; 9:e2017032. [PMID: 28512561 PMCID: PMC5419183 DOI: 10.4084/mjhid.2017.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 03/18/2017] [Indexed: 12/31/2022] Open
Abstract
Homing of hematopoietic stem cells (HSC) to their microenvironment niches in the bone marrow is a complex process with a critical role in repopulation of the bone marrow after transplantation. This active process allows for migration of HSC from peripheral blood and their successful anchoring in bone marrow before proliferation. The process of engraftment starts with the onset of proliferation and must, therefore, be functionally dissociated from the former process. In this overview, we analyze the characteristics of stem cells (SCs) with particular emphasis on their plasticity and ability to find their way home to the bone marrow. We also address the problem of graft failure which remains a significant contributor to morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). Within this context, we discuss non-malignant and malignant hematological disorders treated with reduced-intensity conditioning regimens or grafts from human leukocyte antigen (HLA)-mismatched donors.
Collapse
|
21
|
Zonari E, Desantis G, Petrillo C, Boccalatte FE, Lidonnici MR, Kajaste-Rudnitski A, Aiuti A, Ferrari G, Naldini L, Gentner B. Efficient Ex Vivo Engineering and Expansion of Highly Purified Human Hematopoietic Stem and Progenitor Cell Populations for Gene Therapy. Stem Cell Reports 2017; 8:977-990. [PMID: 28330619 PMCID: PMC5390102 DOI: 10.1016/j.stemcr.2017.02.010] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/13/2017] [Accepted: 02/13/2017] [Indexed: 12/22/2022] Open
Abstract
Ex vivo gene therapy based on CD34+ hematopoietic stem cells (HSCs) has shown promising results in clinical trials, but genetic engineering to high levels and in large scale remains challenging. We devised a sorting strategy that captures more than 90% of HSC activity in less than 10% of mobilized peripheral blood (mPB) CD34+ cells, and modeled a transplantation protocol based on highly purified, genetically engineered HSCs co-infused with uncultured progenitor cells. Prostaglandin E2 stimulation allowed near-complete transduction of HSCs with lentiviral vectors during a culture time of less than 38 hr, mitigating the negative impact of standard culture on progenitor cell function. Exploiting the pyrimidoindole derivative UM171, we show that transduced mPB CD34+CD38- cells with repopulating potential could be expanded ex vivo. Implementing these findings in clinical gene therapy protocols will improve the efficacy, safety, and sustainability of gene therapy and generate new opportunities in the field of gene editing.
Collapse
Affiliation(s)
- Erika Zonari
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan 20132, Italy
| | - Giacomo Desantis
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan 20132, Italy
| | - Carolina Petrillo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | | | | | | | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy; Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCSS Ospedale San Raffaele, Milan 20132, Italy
| | - Giuliana Ferrari
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan 20132, Italy; Hematology and Bone Marrow Transplantation Unit, IRCSS Ospedale San Raffaele, Milan 20132, Italy.
| |
Collapse
|
22
|
In vivo transduction of primitive mobilized hematopoietic stem cells after intravenous injection of integrating adenovirus vectors. Blood 2016; 128:2206-2217. [PMID: 27554082 DOI: 10.1182/blood-2016-04-711580] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/10/2016] [Indexed: 12/31/2022] Open
Abstract
Current protocols for hematopoietic stem/progenitor cell (HSPC) gene therapy, involving the transplantation of ex vivo genetically modified HSPCs are complex and not without risk for the patient. We developed a new approach for in vivo HSPC transduction that does not require myeloablation and transplantation. It involves subcutaneous injections of granulocyte-colony-stimulating factor/AMD3100 to mobilize HSPCs from the bone marrow (BM) into the peripheral blood stream and the IV injection of an integrating, helper-dependent adenovirus (HD-Ad5/35++) vector system. These vectors target CD46, a receptor that is uniformly expressed on HSPCs. We demonstrated in human CD46 transgenic mice and immunodeficient mice with engrafted human CD34+ cells that HSPCs transduced in the periphery home back to the BM where they stably express the transgene. In hCD46 transgenic mice, we showed that our in vivo HSPC transduction approach allows for the stable transduction of primitive HSPCs. Twenty weeks after in vivo transduction, green fluorescent protein (GFP) marking in BM HSPCs (Lin-Sca1+Kit- cells) in most of the mice was in the range of 5% to 10%. The percentage of GFP-expressing primitive HSPCs capable of forming multilineage progenitor colonies (colony-forming units [CFUs]) increased from 4% of all CFUs at week 4 to 16% at week 12, indicating transduction and expansion of long-term surviving HSPCs. Our approach was well tolerated, did not result in significant transduction of nonhematopoietic tissues, and was not associated with genotoxicty. The ability to stably genetically modify HSPCs without the need of myeloablative conditioning is relevant for a broader clinical application of gene therapy.
Collapse
|
23
|
Early production of human neutrophils and platelets posttransplant is severely compromised by growth factor exposure. Exp Hematol 2016; 44:635-40. [PMID: 27090409 DOI: 10.1016/j.exphem.2016.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 01/24/2023]
Abstract
The critical human cells that produce neutrophils and platelets within 3 weeks in recipients of hematopoietic transplants are thought to produce these mature blood cells with the same kinetics in sublethally irradiated immunodeficient mice. Quantification of their numbers indicates their relative underrepresentation in cord blood (CB), likely explaining the clinical inadequacy of single CB units in rescuing hematopoiesis in myelosuppressed adult patients. We here describe that exposure of CD34(+) CB cells ex vivo to growth factors that markedly expand their numbers and colony-forming cell content also rapidly (within 24 hours) produce a significant and sustained net loss of their original short-term repopulating activity. This loss of short-term in vivo repopulating activity affects early platelet production faster than early neutrophil output, consistent with their origin from distinct input populations. Moreover, this growth factor-mediated loss is not abrogated by published strategies to increase progenitor homing despite evidence that the effect on rapid neutrophil production is paralleled in time and amount by a loss of the homing of their committed clonogenic precursors to the bone marrow. These results highlight the inability of in vitro or phenotype assessments to reliably predict clinical engraftment kinetics of cultured CB cells.
Collapse
|
24
|
Yang Y, Wang S, Miao Z, Ma W, Zhang Y, Su L, Hu M, Zou J, Yin Y, Luo J. miR-17 promotes expansion and adhesion of human cord blood CD34(+) cells in vitro. Stem Cell Res Ther 2015; 6:168. [PMID: 26345634 PMCID: PMC4562375 DOI: 10.1186/s13287-015-0159-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 07/14/2015] [Accepted: 08/17/2015] [Indexed: 01/11/2023] Open
Abstract
Introduction We have recently found that miR-17 is necessary in the cell-extrinsic control of cord blood (CB) CD34+ cell function. Here, we demonstrated that the proper level of miR-17 is also necessary in the cell-intrinsic control of the hematopoietic properties of CB CD34+ cells. Methods The miR-17 overexpression and knockdown models were created using primary CB CD34+ cells transfected by the indicated vectors. Long-term culture, colony forming, adhesion and trans-well migration assays were carried out to investigate the function of miR-17 on CB CD34+ cells in vitro. NOD prkdcscid Il2rgnull mice were used in a SCID repopulating cell assay to investigate the function of miR-17 on CB CD34+ cells in vivo. A two-tailed Student’s t-test was used for statistical comparisons. Results In vitro assays revealed that ectopic expression of miR-17 promoted long-term expansion, especially in the colony-forming of CB CD34+ cells and CD34+CD38− cells. Conversely, downregulation of miR-17 inhibited the expansion of CB CD34+ cells. However, the overexpression of miR-17 in vivo reduced the hematopoietic reconstitution potential of CB CD34+ cells compared to that of control cells. The increased expression of major adhesion molecules in miR-17 overexpressed CB CD34+ cells suggests that the adhesion between miR-17 overexpressed CB CD34+ cells and their niche in vivo is regulated abnormally, which may further lead to the reduced hematopoietic reconstitution capability of 17/OE cells in engrafted mice. Conclusion We conclude that the proper expression of miR-17 is required, at least partly, for normal hematopoietic stem cell–niche interaction and for the regulation of adult hematopoiesis. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0159-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuxia Yang
- Department of Medical & Research Technology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
| | - Saifeng Wang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Zhenchuan Miao
- Beijing Vitalstar Biotechnology Co., Ltd., Beijing, China.
| | - Wei Ma
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Yanju Zhang
- Tianjin Central Hospital for Obstetrics and Gynecology, Tianjin, China.
| | - Li Su
- Center of Medical and Health Analysis, Peking University, Beijing, China.
| | - Mengyu Hu
- Department of Medical & Research Technology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
| | - Junhua Zou
- Department of Medical & Research Technology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Jianyuan Luo
- Department of Medical & Research Technology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA. .,Department of Medical & Research Technology, Department of Pathology, School of Medicine, University of Maryland, College Park, USA.
| |
Collapse
|
25
|
Heterogeneity in hematopoietic stem cell populations: implications for transplantation. Curr Opin Hematol 2013; 20:257-64. [PMID: 23615054 DOI: 10.1097/moh.0b013e328360aaf6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Transplantation of hematopoietic cells is now a well established clinical procedure, although optimal outcomes are not always obtained. This reflects insufficient knowledge of the different subsets of primitive cells required to achieve a rapid and permanent recovery of mature blood cell production. Here we review recent findings that extend our understanding of these cells and their regulation, and implications for the ex-vivo expansion of these cells. RECENT FINDINGS Separate subsets of platelet and neutrophil lineage-restricted human hematopoietic cells with rapid but transient repopulating activities have been identified, thus adding to previous evidence of short-term repopulating cells that generate both of these lineages. New studies also suggest intrinsically determined heterogeneity in differentiation potentialities that are sustained at the stem cell level, and have revealed new ways their self-renewal can be influenced. SUMMARY Hematopoietic repopulation posttransplant is highly complex both in terms of the differing numbers and types of cells required for optimal hematopoietic recoveries and the factors that will determine the composition and behavior of a given inoculum. Successful ex-vivo expansion protocols will, thus, need to incorporate conditions that will produce adequate numbers of all cell types required with retention of their full functionality.
Collapse
|
26
|
Chiu YG, Ritchlin CT. Characterization of DC-STAMP+ Cells in Human Bone Marrow. JOURNAL OF BONE MARROW RESEARCH 2013; 1:1000127. [PMID: 25419541 PMCID: PMC4238037 DOI: 10.4172/2329-8820.1000127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Osteoclasts (OC), specialized cells derived from monocytes, maintain skeletal homeostasis under normal conditions but degrade bone in patients with rheumatoid (RA) and psoriatic arthritis (PsA). Monocytes initially develop in the bone marrow (BM), circulate in peripheral blood, and differentiate into distinct cell types with diverse functions. Imaging studies in (RA) patients and murine arthritis models demonstrate that bone marrow edema detected on MRI is the result of enhanced myelopoiesis which precedes the development of bone erosions detected on plain radiographs several years later. A major knowledge gap, however, is whether OC develop in the BM and circulate to the joint and if the differentiation to OC takes place in the joint space in response to differentiation signals such as RANKL and TNF. We have previously demonstrated that osteoclast precursors (OCP) are increased in the circulaton of patients with RA and PsA. We showed that DC-STAMP (Dendritic Cell-Specific Transmembrane protein), a 7-pass transmembrane protein expressed on the surface of monocytes, is essential for cell-to-cell fusion during OC differentiation and is a valid biomarker of OCP. Herein, we examined OCP in human bone marrow and identified one novel subset of DC-STAMP+CD45intermediate monocytes which was absent in the blood. We also found that OCPs reside in human BM with a higher frequency than in the peripheral blood. These findings support the notion that the BM is a major reservoir of circulating OCPs. In addition, we demonstrated that a higher frequency of DC-STAMP+ cells in the BM have detectable intracellular IFN-γ, IL-4 and IL-17A than DC-STAMP+ cells circulating in the peripheral blood. Finally, the frequency of DC-STAMP+ monocytes and T cells is signficantly higher in PsA BM compared to healthy controls, suggesting an enhanced myelopoiesis is a central event in inflammatory arthritis.
Collapse
Affiliation(s)
- Yahui Grace Chiu
- Allergy/Immunology and Rheumatology Division, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Christopher T Ritchlin
- Allergy/Immunology and Rheumatology Division, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| |
Collapse
|
27
|
Enhanced normal short-term human myelopoiesis in mice engineered to express human-specific myeloid growth factors. Blood 2012; 121:e1-4. [PMID: 23233660 DOI: 10.1182/blood-2012-09-456566] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Better methods to characterize normal human hematopoietic cells with short-term repopulating activity cells (STRCs) are needed to facilitate improving recovery rates in transplanted patients.We now show that 5-fold more human myeloid cells are produced in sublethally irradiated NOD/SCID-IL-2Receptor-γchain-null (NSG) mice engineered to constitutively produce human interleukin-3, granulocyte-macrophage colony-stimulating factor and Steel factor (NSG-3GS mice) than in regular NSG mice 3 weeks after an intravenous injection of CD34 human cord blood cells. Importantly, the NSG-3GS mice also show a concomitant and matched increase in circulating mature human neutrophils. Imaging NSG-3GS recipients of lenti-luciferase-transduced cells showed that human cells being produced 3 weeks posttransplant were heterogeneously distributed, validating the blood as a more representative measure of transplanted STRC activity. Limiting dilution transplants further demonstrated that the early increase in human granulopoiesis in NSG-3GS mice reflects an expanded output of differentiated cells per STRC rather than an increase in STRC detection. KEY POINTS NSG-3GS mice support enhanced clonal outputs from human short-term repopulating cells (STRCs) without affecting their engrafting efficiency. Increased human STRC clone sizes enable their more precise and efficient measurement by peripheral blood monitoring.
Collapse
|
28
|
Park GB, Kim MJ, Vasileva EA, Mishchenko NP, Fedoreyev SA, Stonik VA, Han J, Lee HS, Kim D, Jeong JY. Comparison of two-stage epidermal carcinogenesis initiated by 7,12-dimethylbenz(a)anthracene or N-methyl-N'-nitro-N-nitrosoguanidine in newborn and adult SENCAR and BALB/c mice. Cancer Res 1981; 17:md17090526. [PMID: 31505769 PMCID: PMC6780187 DOI: 10.3390/md17090526] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 12/19/2022]
Abstract
In order to define factors which determine susceptibility to chemical carcinogenesis, mice sensitive (SENCAR) and resistant (BALB/c) to epidermal carcinogenesis were studied under several treatment conditions for sensitivity to initiation by 7,12-dimethylbenz(a)anthracene or N-methyl-N'-nitro-N-nitrosoguanidine and promotion by 12-O-tetradecanoylphorbol-13-acetate. In newborns of both strains, topical application of initiator was much less effective than in adults. However, initiation by i.p. injection of 7,12-dimethylbenz(a)anthracene is at least as effective in newborns as in adults, which may indicate that topically applied carcinogen is not delivered effectively to target cells in newborns. Thus, newborn epidermis can respond to 7,12-dimethylbenz(a)anthracene as well as adult epidermis when the initiator is appropriately administered. SENCAR mice are much more sensitive than are BALB/c mice to both initiators, which suggests that enhanced metabolic activation of hydrocarbon carcinogens by SENCAR mice is unlikely to account for their sensitivity. Newborn male SENCAR's developed approximately 50% more papillomas than did females in all groups. BALB/c newborn mice developed so few tumors that a meaningful comparison of sensitivity of males and females could not be made. Thus, the increased sensitivity of SENCAR's was apparent regardless of route of administration of initiator or the age or sex of the mice. SENCAR mice also developed a significant number of papillomas and squamous cell carcinomas with 12-O-tetradecanoylphorbol-13-acetate promotion in the absence of an exogenous initiator. Therefore, the skin of SENCAR mice may contain an initiated population of cells capable of responding to tumor promoters.
Collapse
Affiliation(s)
- Ga-Bin Park
- Department of Biochemistry, Cancer Research Institute, Kosin University College of Medicine, Busan 49267, Korea
| | - Min-Jung Kim
- Department of Biochemistry, Cancer Research Institute, Kosin University College of Medicine, Busan 49267, Korea
| | - Elena A Vasileva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Natalia P Mishchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Sergey A Fedoreyev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Valentin A Stonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Cardiovascular and Metabolic Disease Center, Inje University College of Medicine, Busan 47392, Korea.
| | - Ho Sup Lee
- Department of Internal Medicine, Kosin University College of Medicine, Busan 49267, Korea.
| | - Daejin Kim
- Department of Anatomy, Inje University College of Medicine, Busan 47392, Korea.
| | - Jee-Yeong Jeong
- Department of Biochemistry, Cancer Research Institute, Kosin University College of Medicine, Busan 49267, Korea.
| |
Collapse
|