1
|
Mudde ACA, Kuo CY, Kohn DB, Booth C. What a Clinician Needs to Know About Genome Editing: Status and Opportunities for Inborn Errors of Immunity. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:1139-1149. [PMID: 38246560 DOI: 10.1016/j.jaip.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
During the past 20 years, gene editing has emerged as a novel form of gene therapy. Since the publication of the first potentially therapeutic gene editing platform for genetic disorders, increasingly sophisticated editing technologies have been developed. As with viral vector-mediated gene addition, inborn errors of immunity are excellent candidate diseases for a corrective autologous hematopoietic stem cell gene editing strategy. Research on gene editing for inborn errors of immunity is still entirely preclinical, with no trials yet underway. However, with editing techniques maturing, scientists are investigating this novel form of gene therapy in context of an increasing number of inborn errors of immunity. Here, we present an overview of these studies and the recent progress moving these technologies closer to clinical benefit.
Collapse
Affiliation(s)
- Anne C A Mudde
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Caroline Y Kuo
- Department of Pediatrics, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif
| | - Donald B Kohn
- Department of Pediatrics, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Department of Microbiology, Immunology & Molecular Genetics, UCLA David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif
| | - Claire Booth
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom; Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
2
|
Vamva E, Ozog S, Leaman DP, Yu-Hong Cheng R, Irons NJ, Ott A, Stoffers C, Khan I, Goebrecht GK, Gardner MR, Farzan M, Rawlings DJ, Zwick MB, James RG, Torbett BE. A lentiviral vector B cell gene therapy platform for the delivery of the anti-HIV-1 eCD4-Ig-knob-in-hole-reversed immunoadhesin. Mol Ther Methods Clin Dev 2023; 28:366-384. [PMID: 36879849 PMCID: PMC9984920 DOI: 10.1016/j.omtm.2023.02.004] [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: 03/01/2022] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Barriers to effective gene therapy for many diseases include the number of modified target cells required to achieve therapeutic outcomes and host immune responses to expressed therapeutic proteins. As long-lived cells specialized for protein secretion, antibody-secreting B cells are an attractive target for foreign protein expression in blood and tissue. To neutralize HIV-1, we developed a lentiviral vector (LV) gene therapy platform for delivery of the anti-HIV-1 immunoadhesin, eCD4-Ig, to B cells. The EμB29 enhancer/promoter in the LV limited gene expression in non-B cell lineages. By engineering a knob-in-hole-reversed (KiHR) modification in the CH3-Fc eCD4-Ig domain, we reduced interactions between eCD4-Ig and endogenous B cell immunoglobulin G proteins, which improved HIV-1 neutralization potency. Unlike previous approaches in non-lymphoid cells, eCD4-Ig-KiHR produced in B cells promoted HIV-1 neutralizing protection without requiring exogenous TPST2, a tyrosine sulfation enzyme required for eCD4-Ig-KiHR function. This finding indicated that B cell machinery is well suited to produce therapeutic proteins. Lastly, to overcome the inefficient transduction efficiency associated with VSV-G LV delivery to primary B cells, an optimized measles pseudotyped LV packaging methodology achieved up to 75% transduction efficiency. Overall, our findings support the utility of B cell gene therapy platforms for therapeutic protein delivery.
Collapse
Affiliation(s)
- Eirini Vamva
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Stosh Ozog
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Daniel P. Leaman
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Rene Yu-Hong Cheng
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Nicholas J. Irons
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Andee Ott
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Claire Stoffers
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Iram Khan
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
| | | | - Matthew R. Gardner
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL, USA
| | - Michael Farzan
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL, USA
| | - David J. Rawlings
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Michael B. Zwick
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Richard G. James
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA
| | - Bruce E. Torbett
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA
| |
Collapse
|
3
|
Gray DH, Santos J, Keir AG, Villegas I, Maddock S, Trope EC, Long JD, Kuo CY. A comparison of DNA repair pathways to achieve a site-specific gene modification of the Bruton's tyrosine kinase gene. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:505-516. [PMID: 35036061 PMCID: PMC8728535 DOI: 10.1016/j.omtn.2021.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/09/2021] [Indexed: 01/08/2023]
Abstract
Gene editing utilizing homology-directed repair has advanced significantly for many monogenic diseases of the hematopoietic system in recent years but has also been hindered by decreases between in vitro and in vivo gene integration rates. Homology-directed repair occurs primarily in the S/G2 phases of the cell cycle, whereas long-term engrafting hematopoietic stem cells are typically quiescent. Alternative methods for a targeted integration have been proposed including homology-independent targeted integration and precise integration into target chromosome, which utilize non-homologous end joining and microhomology-mediated end joining, respectively. Non-homologous end joining occurs throughout the cell cycle, while microhomology-mediated end joining occurs predominantly in the S phase. We compared these pathways for the integration of a corrective DNA cassette at the Bruton's tyrosine kinase gene for the treatment of X-linked agammaglobulinemia. Homology-directed repair generated the most integration in K562 cells; however, synchronizing cells into G1 resulted in the highest integration rates with homology-independent targeted integration. Only homology-directed repair produced seamless junctions, making it optimal for targets where insertions and deletions are impermissible. Bulk CD34+ cells were best edited by homology-directed repair and precise integration into the target chromosome, while sorted hematopoietic stem cells contained similar integration rates using all corrective donors.
Collapse
Affiliation(s)
- David H. Gray
- Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, CA 90095, USA
| | - Jasmine Santos
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Alexandra Grace Keir
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Isaac Villegas
- Division of Allergy and Immunology, Department of Pediatrics, David Geffen School of Medicine at the University of California, 10833 Le Conte MDCC 12-430, Los Angeles, CA 90095, USA
| | - Simon Maddock
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Edward C. Trope
- Division of Allergy and Immunology, Department of Pediatrics, David Geffen School of Medicine at the University of California, 10833 Le Conte MDCC 12-430, Los Angeles, CA 90095, USA
| | - Joseph D. Long
- Division of Allergy and Immunology, Department of Pediatrics, David Geffen School of Medicine at the University of California, 10833 Le Conte MDCC 12-430, Los Angeles, CA 90095, USA
| | - Caroline Y. Kuo
- Division of Allergy and Immunology, Department of Pediatrics, David Geffen School of Medicine at the University of California, 10833 Le Conte MDCC 12-430, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Geron I, Savino AM, Fishman H, Tal N, Brown J, Turati VA, James C, Sarno J, Hameiri-Grossman M, Lee YN, Rein A, Maniriho H, Birger Y, Zemlyansky A, Muler I, Davis KL, Marcu-Malina V, Mattson N, Parnas O, Wagener R, Fischer U, Barata JT, Jamieson CHM, Müschen M, Chen CW, Borkhardt A, Kirsch IR, Nagler A, Enver T, Izraeli S. An instructive role for Interleukin-7 receptor α in the development of human B-cell precursor leukemia. Nat Commun 2022; 13:659. [PMID: 35115489 PMCID: PMC8814001 DOI: 10.1038/s41467-022-28218-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Kinase signaling fuels growth of B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Yet its role in leukemia initiation is unclear and has not been shown in primary human hematopoietic cells. We previously described activating mutations in interleukin-7 receptor alpha (IL7RA) in poor-prognosis "ph-like" BCP-ALL. Here we show that expression of activated mutant IL7RA in human CD34+ hematopoietic stem and progenitor cells induces a preleukemic state in transplanted immunodeficient NOD/LtSz-scid IL2Rγnull mice, characterized by persistence of self-renewing Pro-B cells with non-productive V(D)J gene rearrangements. Preleukemic CD34+CD10highCD19+ cells evolve into BCP-ALL with spontaneously acquired Cyclin Dependent Kinase Inhibitor 2 A (CDKN2A) deletions, as commonly observed in primary human BCP-ALL. CRISPR mediated gene silencing of CDKN2A in primary human CD34+ cells transduced with activated IL7RA results in robust development of BCP-ALLs in-vivo. Thus, we demonstrate that constitutive activation of IL7RA can initiate preleukemia in primary human hematopoietic progenitors and cooperates with CDKN2A silencing in progression into BCP-ALL.
Collapse
MESH Headings
- Animals
- Antigens, CD34/genetics
- Antigens, CD34/immunology
- Antigens, CD34/metabolism
- Base Sequence
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Cyclin-Dependent Kinase Inhibitor p16/immunology
- Cyclin-Dependent Kinase Inhibitor p16/metabolism
- Gene Expression/immunology
- Humans
- Interleukin-7 Receptor alpha Subunit/genetics
- Interleukin-7 Receptor alpha Subunit/immunology
- Interleukin-7 Receptor alpha Subunit/metabolism
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cells, B-Lymphoid/immunology
- Precursor Cells, B-Lymphoid/metabolism
- RNA-Seq/methods
- Receptors, Cytokine/genetics
- Receptors, Cytokine/immunology
- Receptors, Cytokine/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- Single-Cell Analysis/methods
- Transplantation, Heterologous
- Mice
Collapse
Affiliation(s)
- Ifat Geron
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Angela Maria Savino
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Hila Fishman
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Noa Tal
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - John Brown
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | | | - Chela James
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Jolanda Sarno
- Department of Pediatrics, Bass Center for Childhood Cancer and Blood Disorders, Stanford University, Stanford, CA, USA
| | - Michal Hameiri-Grossman
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Yu Nee Lee
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Pediatric Department and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital Sheba Medical Center, Tel-Hashomer, Israel
| | - Avigail Rein
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Hillary Maniriho
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Yehudit Birger
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Anna Zemlyansky
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Inna Muler
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Kara L Davis
- Department of Pediatrics, Bass Center for Childhood Cancer and Blood Disorders, Stanford University, Stanford, CA, USA
| | - Victoria Marcu-Malina
- Cytogenetic Unit laboratory of Hematology, Chaim Sheba Medical Center Tel Hashomer, Tel Hashomer, Israel
| | - Nicole Mattson
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Oren Parnas
- The Concern Foundation Laboratories at the Lautenberg Center for immunology and Cancer Research, IMRIC, Hebrew University Faculty of Medicine, Jerusalem, Israel
| | - Rabea Wagener
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Catriona H M Jamieson
- UC San Diego, Moores Cancer Center, Division of Regenerative Medicine, Department of Medicine and Sanford Stem Cell Clinical Center, Ja Jolla, CA, USA
| | - Markus Müschen
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Chun-Wei Chen
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | - Arnon Nagler
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Hematology Division BMT and Cord Blood Bank Chaim Sheba Medical Center Tel-Hashomer, Tel-Hashomer, Israel
| | - Tariq Enver
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Shai Izraeli
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel.
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel.
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA.
| |
Collapse
|
5
|
Gray DH, Villegas I, Long J, Santos J, Keir A, Abele A, Kuo CY, Kohn DB. Optimizing Integration and Expression of Transgenic Bruton's Tyrosine Kinase for CRISPR-Cas9-Mediated Gene Editing of X-Linked Agammaglobulinemia. CRISPR J 2021; 4:191-206. [PMID: 33876953 PMCID: PMC8336228 DOI: 10.1089/crispr.2020.0080] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
X-linked agammaglobulinemia (XLA) is a monogenic primary immune deficiency characterized by very low levels of immunoglobulins and greatly increased risks for recurrent and severe infections. Patients with XLA have a loss-of-function mutation in the Bruton's tyrosine kinase (BTK) gene and fail to produce mature B lymphocytes. Gene editing in the hematopoietic stem cells of XLA patients to correct or replace the defective gene should restore B cell development and the humoral immune response. We used the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 platform to precisely target integration of a corrective, codon-optimized BTK complementary DNA (cDNA) cassette into its endogenous locus. This process is driven by homologous recombination and should place the transgenic BTK under transcriptional control of its endogenous regulatory elements. Each integrated copy of this cDNA in BTK-deficient K562 cells produced only 11% as much BTK protein as the wild-type gene. The donor cDNA was modified to include the terminal intron of the BTK gene. Successful integration of the intron-containing BTK donor led to a nearly twofold increase in BTK expression per cell over the base donor. However, this donor variant was too large to package into an adeno-associated viral vector for delivery into primary cells. Donors containing truncated variants of the terminal intron also produced elevated expression, although to a lesser degree than the full intron. Addition of the Woodchuck hepatitis virus posttranscriptional regulatory element led to a large boost in BTK transgene expression. Combining these modifications led to a BTK donor template that generated nearly physiological levels of BTK expression in cell lines. These reagents were then optimized to maximize integration rates into human hematopoietic stem and progenitor cells, which have reached potentially therapeutic levels in vitro. The novel donor modifications support effective gene therapy for XLA and will likely assist in the development of other gene editing-based therapies for genetic disorders.
Collapse
Affiliation(s)
- David H. Gray
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA; University of California, Los Angeles, Los Angeles, California, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA; University of California, Los Angeles, Los Angeles, California, USA
| | - Isaac Villegas
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA; Departments of University of California, Los Angeles, Los Angeles, California, USA
| | - Joseph Long
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA; Departments of University of California, Los Angeles, Los Angeles, California, USA
| | - Jasmine Santos
- Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Alexandra Keir
- Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA
| | - Alison Abele
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Caroline Y. Kuo
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA; Departments of University of California, Los Angeles, Los Angeles, California, USA
| | - Donald B. Kohn
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA; Departments of University of California, Los Angeles, Los Angeles, California, USA
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
- Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA; University of California, Los Angeles, Los Angeles, California, USA
- The Eli & Edith Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
6
|
Seymour BJ, Singh S, Certo HM, Sommer K, Sather BD, Khim S, Clough C, Hale M, Pangallo J, Ryu BY, Khan IF, Adair JE, Rawlings DJ. Effective, safe, and sustained correction of murine XLA using a UCOE-BTK promoter-based lentiviral vector. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:635-651. [PMID: 33718514 PMCID: PMC7907679 DOI: 10.1016/j.omtm.2021.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
X-linked agammaglobulinemia (XLA) is an immune disorder caused by mutations in Bruton’s tyrosine kinase (BTK). BTK is expressed in B and myeloid cells, and its deficiency results in a lack of mature B cells and protective antibodies. We previously reported a lentivirus (LV) BTK replacement therapy that restored B cell development and function in Btk and Tec double knockout mice (a phenocopy of human XLA). In this study, with the goal of optimizing both the level and lineage specificity of BTK expression, we generated LV incorporating the proximal human BTK promoter. Hematopoietic stem cells from Btk−/−Tec−/− mice transduced with this vector rescued lineage-specific expression and restored B cell function in Btk−/−Tec−/− recipients. Next, we tested addition of candidate enhancers and/or ubiquitous chromatin opening elements (UCOEs), as well as codon optimization to improve BTK expression. An Eμ enhancer improved B cell rescue, but increased immunoglobulin G (IgG) autoantibodies. Addition of the UCOE avoided autoantibody generation while improving B cell development and function and reducing vector silencing. An optimized vector containing a truncated UCOE upstream of the BTK promoter and codon-optimized BTK cDNA resulted in stable, lineage-regulated BTK expression that mirrored endogenous BTK, making it a strong candidate for XLA therapy.
Collapse
Affiliation(s)
- Brenda J Seymour
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Swati Singh
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Hannah M Certo
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Karen Sommer
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Blythe D Sather
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Socheath Khim
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Courtnee Clough
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Malika Hale
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Joseph Pangallo
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Byoung Y Ryu
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Iram F Khan
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Jennifer E Adair
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Medical Oncology, University of Washington, Seattle, WA 98195, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Departments of Pediatrics and Immunology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
7
|
Houghton BC, Booth C. Gene Therapy for Primary Immunodeficiency. Hemasphere 2021; 5:e509. [PMID: 33403354 PMCID: PMC7773329 DOI: 10.1097/hs9.0000000000000509] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/21/2020] [Indexed: 12/27/2022] Open
Abstract
Over the past 3 decades, there has been significant progress in refining gene therapy technologies and procedures. Transduction of hematopoietic stem cells ex vivo using lentiviral vectors can now create a highly effective therapeutic product, capable of reconstituting many different immune system dysfunctions when reinfused into patients. Here, we review the key developments in the gene therapy landscape for primary immune deficiency, from an experimental therapy where clinical efficacy was marred by adverse events, to a commercialized product with enhanced safety and efficacy. We also discuss progress being made in preclinical studies for challenging disease targets and emerging gene editing technologies that are showing promising results, particularly for conditions where gene regulation is important for efficacy.
Collapse
Affiliation(s)
- Benjamin C. Houghton
- Molecular and Cellular Immunology, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Claire Booth
- Molecular and Cellular Immunology, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Paediatric Immunology, Great Ormond Street NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
8
|
Whaley RE, Ameny S, Arkatkar T, Seese A, Wall A, Khan I, Carter JJ, Scherer EM, Rawlings DJ, Galloway DA, McElrath MJ, Cohen KW, McGuire AT. Generation of a cost-effective cell line for support of high-throughput isolation of primary human B cells and monoclonal neutralizing antibodies. J Immunol Methods 2020; 488:112901. [PMID: 33069767 PMCID: PMC7560121 DOI: 10.1016/j.jim.2020.112901] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 01/11/2023]
Abstract
The isolation of human monoclonal antibodies (mAbs) arising from natural infection with human pathogens has proven to be a powerful technology, facilitating the understanding of the host response to infection at a molecular level. mAbs can reveal sites of vulnerability on pathogens and illuminate the biological function of the antigenic targets. Moreover, mAbs have the potential to be used directly for therapeutic applications such as passive delivery to prevent infection in susceptible target populations, and as treatment of established infection. The isolation of antigen-specific B cells from vaccine trials can also assist in deciphering whether the desired B cells are being targeted by a given vaccine. Several different processes have been developed to isolate mAbs, but all are generally labor-intensive and result in varying degrees of efficiency. Here, we describe the development of a cost-effective feeder cell line that stably expresses CD40-ligand, interleukin-2 and interleukin-21. Sorting of single B cells onto a layer of irradiated feeder cells sustained antibody production that permits functional screening of secreted antibodies in a manner that enables subsequent recovery of B cells for recombinant antibody cloning. As a proof of concept, we show that this approach can be used to isolate B cells that secrete antibodies that neutralize human papilloma virus (HPV) from participants of an HPV vaccine study. Development of a cell line that provides signals for human B cell growth and antibody secretion. 50% cultures seeded from single B cells produce detectable IgG. B cells secreting HPV-neutralizing antibodies from a vaccine study are identified.
Collapse
Affiliation(s)
- Rachael E Whaley
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sarah Ameny
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tanvi Arkatkar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Aaron Seese
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Abigail Wall
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Iram Khan
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA, USA
| | - Joseph J Carter
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Erin M Scherer
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA, USA; Departments of Pediatrics and Immunology, University of Washington, Seattle, WA, USA
| | - Denise A Galloway
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kristen W Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Andrew T McGuire
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Deparment of Laboratory Medicine and Pahthology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
9
|
Advances in site-specific gene editing for primary immune deficiencies. Curr Opin Allergy Clin Immunol 2019; 18:453-458. [PMID: 30299399 DOI: 10.1097/aci.0000000000000483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE OF REVIEW Conventional gene therapy has been a successful, curative treatment modality for many primary immune deficiencies with significant improvements in the last decade. However, the risk of leukemic transformation with viral-mediated gene addition still remains, and unregulated gene addition is not an option for certain diseases in which the target gene is closely controlled. The recent bloom in genome modification platforms has created the opportunity to site-specifically correct mutated DNA base pairs or insert a corrective cDNA minigene while maintaining gene expression under control of endogenous regulatory elements. RECENT FINDINGS There is an abundance of ongoing research utilizing programmable nucleases to facilitate site-specific gene correction of many primary immune deficiencies including X-linked severe combined immune deficiency, X-linked chronic granulomatous disease, Wiskott-Aldrich syndrome, X-linked hyper-IgM syndrome, X-linked agammaglobulinemia, and immune dysregulation, polyendocrinopathy, enteropathy, X-linked. In all, these studies have demonstrated the ability to integrate corrective DNA sequences at a precise location in the genome at rates likely to either cure or ameliorate disease. SUMMARY Gene editing for primary immune deficiency (PID) has advanced to the point to that translation to clinical trials is likely to occur in the next several years. At the current pace of research in DNA repair mechanisms, stem cell biology, and genome-editing technology, targeted genome modification represents the next chapter of gene therapy for PID.
Collapse
|
10
|
Singh S, Khan I, Khim S, Seymour B, Sommer K, Wielgosz M, Norgaard Z, Kiem HP, Adair J, Liggitt D, Nienhuis A, Rawlings DJ. Safe and Effective Gene Therapy for Murine Wiskott-Aldrich Syndrome Using an Insulated Lentiviral Vector. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 4:1-16. [PMID: 28344987 PMCID: PMC5363182 DOI: 10.1016/j.omtm.2016.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022]
Abstract
Wiskott-Aldrich syndrome (WAS) is a life-threatening immunodeficiency caused by mutations within the WAS gene. Viral gene therapy to restore WAS protein (WASp) expression in hematopoietic cells of patients with WAS has the potential to improve outcomes relative to the current standard of care, allogeneic bone marrow transplantation. However, the development of viral vectors that are both safe and effective has been problematic. While use of viral transcriptional promoters may increase the risk of insertional mutagenesis, cellular promoters may not achieve WASp expression levels necessary for optimal therapeutic effect. Here we evaluate a self-inactivating (SIN) lentiviral vector combining a chromatin insulator upstream of a viral MND (MPSV LTR, NCR deleted, dl587 PBS) promoter driving WASp expression. Used as a gene therapeutic in Was−/− mice, this vector resulted in stable WASp+ cells in all hematopoietic lineages and rescue of T and B cell defects with a low number of viral integrations per cell, without evidence of insertional mutagenesis in serial bone marrow transplants. In a gene transfer experiment in non-human primates, the insulated MND promoter (driving GFP expression) demonstrated long-term polyclonal engraftment of GFP+ cells. These observations demonstrate that the insulated MND promoter safely and efficiently reconstitutes clinically effective WASp expression and should be considered for future WAS therapy.
Collapse
Affiliation(s)
- Swati Singh
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Iram Khan
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Socheath Khim
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Brenda Seymour
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Karen Sommer
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Matthew Wielgosz
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zachary Norgaard
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Pathology, University of Washington, Seattle, WA 98105, USA
| | - Jennifer Adair
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Medical Oncology, University of Washington, Seattle, WA 98105, USA
| | - Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, WA 98105, USA
| | - Arthur Nienhuis
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Immunology, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
11
|
Dam EM, Habib T, Chen J, Funk A, Glukhova V, Davis-Pickett M, Wei S, James R, Buckner JH, Cerosaletti K. The BANK1 SLE-risk variants are associated with alterations in peripheral B cell signaling and development in humans. Clin Immunol 2016; 173:171-180. [PMID: 27816669 DOI: 10.1016/j.clim.2016.10.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/11/2016] [Accepted: 10/30/2016] [Indexed: 02/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the development of autoantibodies that drive disease pathogenesis. Genetic studies have associated nonsynonymous variants in the BANK1 B cell scaffolding gene with susceptibility to SLE and autoantibodies in lupus. To determine how the BANK1 SLE-risk variants contribute to the dysregulated B cell program in lupus, we performed genotype/phenotype studies in human B cells. Targeted phospho-proteomics were used to evaluate BCR/CD40 signaling in human B cell lines engineered to express the BANK1 risk or non-risk variant proteins. We found that phosphorylation of proximal BCR signaling molecules was reduced in B cells expressing the BANK1 risk protein compared to the non-risk protein. Similar to these findings, we observed decreased B cell signaling in primary B cells from genotyped healthy control subjects carrying the BANK1 risk haplotype, including blunted BCR- and CD40-dependent AKT activation. Consistent with decreased AKT activation, we found that BANK1 risk B cells expressed increased basal levels of FOXO1 protein and increased expression of FOXO1 target genes upon stimulation compared to non-risk B cells. Healthy subjects carrying the BANK1 risk haplotype were also characterized by an expansion of memory B cells. Taken together, our results suggest that the SLE susceptibility variants in the BANK1 gene may contribute to lupus by altering B cell signaling, increasing FOXO1 levels, and enhancing memory B cell development.
Collapse
Affiliation(s)
- Elizabeth M Dam
- Translational Research Program, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101
| | - Tania Habib
- Translational Research Program, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101
| | - Janice Chen
- Translational Research Program, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101
| | - Andrew Funk
- Translational Research Program, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101
| | - Veronika Glukhova
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101
| | - Mel Davis-Pickett
- Translational Research Program, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101
| | - Shan Wei
- Translational Research Program, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101
| | - Richard James
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101
- Department of Pediatrics and Pharmacology, University of Washington School of Medicine
| | - Jane H Buckner
- Translational Research Program, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101
| | - Karen Cerosaletti
- Translational Research Program, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101
| |
Collapse
|
12
|
Booth C, Gaspar HB, Thrasher AJ. Treating Immunodeficiency through HSC Gene Therapy. Trends Mol Med 2016; 22:317-327. [PMID: 26993219 DOI: 10.1016/j.molmed.2016.02.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 11/19/2022]
Abstract
Haematopoietic stem cell (HSC) gene therapy has been successfully employed as a therapeutic option to treat specific inherited immune deficiencies, including severe combined immune deficiencies (SCID) over the past two decades. Initial clinical trials using first-generation gamma-retroviral vectors to transfer corrective DNA demonstrated clinical benefit for patients, but were associated with leukemogenesis in a number of cases. Safer vectors have since been developed, affording comparable efficacy with an improved biosafety profile. These vectors are now in Phase I/II clinical trials for a number of immune disorders with more preclinical studies underway. Targeted gene editing allowing precise DNA correction via platforms such as ZFNs, TALENs and CRISPR/Cas9 may now offer promising strategies to improve the safety and efficacy of gene therapy in the future.
Collapse
Affiliation(s)
- Claire Booth
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, London, UK; Department of Paediatric Immunology, Great Ormond Street Hospital, London, UK
| | - H Bobby Gaspar
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, London, UK; Department of Paediatric Immunology, Great Ormond Street Hospital, London, UK
| | - Adrian J Thrasher
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, London, UK; Department of Paediatric Immunology, Great Ormond Street Hospital, London, UK.
| |
Collapse
|
13
|
Wang X, Shin SC, Chiang AFJ, Khan I, Pan D, Rawlings DJ, Miao CH. Intraosseous delivery of lentiviral vectors targeting factor VIII expression in platelets corrects murine hemophilia A. Mol Ther 2015; 23:617-26. [PMID: 25655313 DOI: 10.1038/mt.2015.20] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 01/20/2015] [Indexed: 12/11/2022] Open
Abstract
Intraosseous (IO) infusion of lentiviral vectors (LVs) for in situ gene transfer into bone marrow may avoid specific challenges posed by ex vivo gene delivery, including, in particular, the requirement of preconditioning. We utilized IO delivery of LVs encoding a GFP or factor VIII (FVIII) transgene directed by ubiquitous promoters (a MND or EF-1α-short element; M-GFP-LV, E-F8-LV) or a platelet-specific, glycoprotein-1bα promoter (G-GFP-LV, G-F8-LV). A single IO infusion of M-GFP-LV or G-GFP-LV achieved long-term and efficient GFP expression in Lineage(-)Sca1(+)c-Kit(+) hematopoietic stem cells and platelets, respectively. While E-F8-LV produced initially high-level FVIII expression, robust anti-FVIII immune responses eliminated functional FVIII in circulation. In contrast, IO delivery of G-F8-LV achieved long-term platelet-specific expression of FVIII, resulting in partial correction of hemophilia A. Furthermore, similar clinical benefit with G-F8-LV was achieved in animals with pre-existing anti-FVIII inhibitors. These findings further support platelets as an ideal FVIII delivery vehicle, as FVIII, stored in α-granules, is protected from neutralizing antibodies and, during bleeding, activated platelets locally excrete FVIII to promote clot formation. Overall, a single IO infusion of G-F8-LV was sufficient to correct hemophilia phenotype for long term, indicating that this approach may provide an effective means to permanently treat FVIII deficiency.
Collapse
Affiliation(s)
- Xuefeng Wang
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Simon C Shin
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Andy F J Chiang
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Iram Khan
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Dao Pan
- Division of Experimental Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - David J Rawlings
- 1] Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA [2] Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Carol H Miao
- 1] Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA [2] Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
14
|
McGuire AT, Dreyer AM, Carbonetti S, Lippy A, Glenn J, Scheid JF, Mouquet H, Stamatatos L. HIV antibodies. Antigen modification regulates competition of broad and narrow neutralizing HIV antibodies. Science 2014; 346:1380-1383. [PMID: 25504724 DOI: 10.1126/science.1259206] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Some HIV-infected individuals develop broadly neutralizing antibodies (bNAbs), whereas most develop antibodies that neutralize only a narrow range of viruses (nNAbs). bNAbs, but not nNAbs, protect animals from experimental infection and are likely a key component of an effective vaccine. nNAbs and bNAbs target the same regions of the viral envelope glycoprotein (Env), but for reasons that remain unclear only nNAbs are elicited by Env immunization. We show that in contrast to germline-reverted (gl) bNAbs, glnNAbs recognized diverse recombinant Envs. Moreover, owing to binding affinity differences, nNAb B cell progenitors had an advantage in becoming activated and internalizing Env compared with bNAb B cell progenitors. We then identified an Env modification strategy that minimized the activation of nNAb B cells targeting epitopes that overlap those of bNAbs.
Collapse
Affiliation(s)
| | - Anita M Dreyer
- Seattle Biomedical Research Institute, Seattle, WA 98109, USA
| | - Sara Carbonetti
- Seattle Biomedical Research Institute, Seattle, WA 98109, USA
| | - Adriana Lippy
- Seattle Biomedical Research Institute, Seattle, WA 98109, USA
| | - Jolene Glenn
- Seattle Biomedical Research Institute, Seattle, WA 98109, USA
| | - Johannes F Scheid
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Hugo Mouquet
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur and CNRS-URA 1961, 75015 Paris, France
| | - Leonidas Stamatatos
- Seattle Biomedical Research Institute, Seattle, WA 98109, USA.,Department of Global Health, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
15
|
Leoh LS, Morizono K, Kershaw KM, Chen ISY, Penichet ML, Daniels-Wells TR. Gene delivery in malignant B cells using the combination of lentiviruses conjugated to anti-transferrin receptor antibodies and an immunoglobulin promoter. J Gene Med 2014; 16:11-27. [PMID: 24436117 DOI: 10.1002/jgm.2754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 12/05/2013] [Accepted: 01/09/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND We previously developed an antibody-avidin fusion protein (ch128.1Av) specific for the human transferrin receptor 1 (TfR1; CD71) to be used as a delivery vector for cancer therapy and showed that ch128.1Av delivers the biotinylated plant toxin saporin-6 into malignant B cells. However, as a result of widespread expression of TfR1, delivery of the toxin to normal cells is a concern. Therefore, we explored the potential of a dual targeted lentiviral-mediated gene therapy strategy to restrict gene expression to malignant B cells. Targeting occurs through the use of ch128.1Av or its parental antibody without avidin (ch128.1) and through transcriptional regulation using an immunoglobulin promoter. METHODS Flow cytometry was used to detect the expression of enhanced green fluorescent protein (EGFP) in a panel of cell lines. Cell viability after specific delivery of the therapeutic gene FCU1, a chimeric enzyme consisting of cytosine deaminase genetically fused to uracil phosphoribosyltransferse that converts the 5-fluorocytosine (5-FC) prodrug into toxic metabolites, was monitored using the MTS or WST-1 viability assay. RESULTS We found that EGFP was specifically expressed in a panel of human malignant B-cell lines, but not in human malignant T-cell lines. EGFP expression was observed in all cell lines when a ubiquitous promoter was used. Furthermore, we show the decrease of cell viability in malignant plasma cells in the presence of 5-FC and the FCU1 gene. CONCLUSIONS The present study demonstrates that gene expression can be restricted to malignant B cells and suggests that this dual targeted gene therapy strategy may help to circumvent the potential side effects of certain TfR1-targeted protein delivery approaches.
Collapse
Affiliation(s)
- Lai Sum Leoh
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
16
|
van Til NP, Sarwari R, Visser TP, Hauer J, Lagresle-Peyrou C, van der Velden G, Malshetty V, Cortes P, Jollet A, Danos O, Cassani B, Zhang F, Thrasher AJ, Fontana E, Poliani PL, Cavazzana M, Verstegen MM, Villa A, Wagemaker G. Recombination-activating gene 1 (Rag1)–deficient mice with severe combined immunodeficiency treated with lentiviral gene therapy demonstrate autoimmune Omenn-like syndrome. J Allergy Clin Immunol 2014; 133:1116-23. [DOI: 10.1016/j.jaci.2013.10.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 10/04/2013] [Accepted: 10/09/2013] [Indexed: 12/20/2022]
|
17
|
Wang Y, Khan IF, Boissel S, Jarjour J, Pangallo J, Thyme S, Baker D, Scharenberg AM, Rawlings DJ. Progressive engineering of a homing endonuclease genome editing reagent for the murine X-linked immunodeficiency locus. Nucleic Acids Res 2014; 42:6463-75. [PMID: 24682825 PMCID: PMC4041414 DOI: 10.1093/nar/gku224] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
LAGLIDADG homing endonucleases (LHEs) are compact endonucleases with 20–22 bp recognition sites, and thus are ideal scaffolds for engineering site-specific DNA cleavage enzymes for genome editing applications. Here, we describe a general approach to LHE engineering that combines rational design with directed evolution, using a yeast surface display high-throughput cleavage selection. This approach was employed to alter the binding and cleavage specificity of the I-Anil LHE to recognize a mutation in the mouse Bruton tyrosine kinase (Btk) gene causative for mouse X-linked immunodeficiency (XID)—a model of human X-linked agammaglobulinemia (XLA). The required re-targeting of I-AniI involved progressive resculpting of the DNA contact interface to accommodate nine base differences from the native cleavage sequence. The enzyme emerging from the progressive engineering process was specific for the XID mutant allele versus the wild-type (WT) allele, and exhibited activity equivalent to WT I-AniI in vitro and in cellulo reporter assays. Fusion of the enzyme to a site-specific DNA binding domain of transcription activator-like effector (TALE) resulted in a further enhancement of gene editing efficiency. These results illustrate the potential of LHE enzymes as specific and efficient tools for therapeutic genome engineering.
Collapse
Affiliation(s)
- Yupeng Wang
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Iram F Khan
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Sandrine Boissel
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | - Joseph Pangallo
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Summer Thyme
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Andrew M Scharenberg
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA Departments of Pediatrics and Immunology, University of Washington, Seattle, WA 98195, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA Departments of Pediatrics and Immunology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
18
|
Epstein-Barr virus utilizes Ikaros in regulating its latent-lytic switch in B cells. J Virol 2014; 88:4811-27. [PMID: 24522918 DOI: 10.1128/jvi.03706-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Ikaros is a zinc finger DNA-binding protein that regulates chromatin remodeling and the expression of genes involved in the cell cycle, apoptosis, and Notch signaling. It is a master regulator of lymphocyte differentiation and functions as a tumor suppressor in acute lymphoblastic leukemia. Nevertheless, no previous reports described effects of Ikaros on the life cycle of any human lymphotropic virus. Here, we demonstrate that full-length Ikaros (IK-1) functions as a major factor in the maintenance of viral latency in Epstein-Barr virus (EBV)-positive Burkitt's lymphoma Sal and MutuI cell lines. Either silencing of Ikaros expression by small hairpin RNA (shRNA) knockdown or ectopic expression of a non-DNA-binding isoform induced lytic gene expression. These effects synergized with other lytic inducers of EBV, including transforming growth factor β (TGF-β) and the hypoxia mimic desferrioxamine. Data from chromatin immunoprecipitation (ChIP)-quantitative PCR (qPCR) and ChIP-sequencing (ChIP-seq) analyses indicated that Ikaros did not bind to either of the EBV immediate early genes BZLF1 and BRLF1. Rather, Ikaros affected the expression of Oct-2 and Bcl-6, other transcription factors that directly inhibit EBV reactivation and plasma cell differentiation, respectively. IK-1 also complexed with the EBV immediate early R protein in coimmunoprecipitation assays and partially colocalized with R within cells. The presence of R alleviated IK-1-mediated transcriptional repression, with IK-1 then cooperating with Z and R to enhance lytic gene expression. Thus, we conclude that Ikaros plays distinct roles at different stages of EBV's life cycle: it contributes to maintaining latency via indirect mechanisms, and it may also synergize with Z and R to enhance lytic replication through direct association with R and/or R-induced alterations in Ikaros' functional activities via cellular signaling pathways. IMPORTANCE This is the first report showing that the cellular protein Ikaros, a known master regulator of hematopoiesis and critical tumor suppressor in acute lymphoblastic leukemia, also plays important roles in the life cycle of Epstein-Barr virus in B cells.
Collapse
|
19
|
Boissel S, Jarjour J, Astrakhan A, Adey A, Gouble A, Duchateau P, Shendure J, Stoddard BL, Certo MT, Baker D, Scharenberg AM. megaTALs: a rare-cleaving nuclease architecture for therapeutic genome engineering. Nucleic Acids Res 2014; 42:2591-601. [PMID: 24285304 PMCID: PMC3936731 DOI: 10.1093/nar/gkt1224] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/04/2013] [Accepted: 11/05/2013] [Indexed: 01/13/2023] Open
Abstract
Rare-cleaving endonucleases have emerged as important tools for making targeted genome modifications. While multiple platforms are now available to generate reagents for research applications, each existing platform has significant limitations in one or more of three key properties necessary for therapeutic application: efficiency of cleavage at the desired target site, specificity of cleavage (i.e. rate of cleavage at 'off-target' sites), and efficient/facile means for delivery to desired target cells. Here, we describe the development of a single-chain rare-cleaving nuclease architecture, which we designate 'megaTAL', in which the DNA binding region of a transcription activator-like (TAL) effector is used to 'address' a site-specific meganuclease adjacent to a single desired genomic target site. This architecture allows the generation of extremely active and hyper-specific compact nucleases that are compatible with all current viral and nonviral cell delivery methods.
Collapse
Affiliation(s)
- Sandrine Boissel
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Jordan Jarjour
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Alexander Astrakhan
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Andrew Adey
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Agnès Gouble
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Philippe Duchateau
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Jay Shendure
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Barry L. Stoddard
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Michael T. Certo
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - David Baker
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Andrew M. Scharenberg
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA 98101, USA, Pregenen, Inc., Seattle, WA 98103, USA, Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA, Cellectis S.A., Paris, 75013, France, Division of Basic Sciences, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA and Department of Immunology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
20
|
Berglöf A, Turunen JJ, Gissberg O, Bestas B, Blomberg KEM, Smith CIE. Agammaglobulinemia: causative mutations and their implications for novel therapies. Expert Rev Clin Immunol 2014; 9:1205-21. [DOI: 10.1586/1744666x.2013.850030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
21
|
Kinase-independent feedback of the TAK1/TAB1 complex on BCL10 turnover and NF-κB activation. Mol Cell Biol 2013; 33:1149-63. [PMID: 23297344 DOI: 10.1128/mcb.06407-11] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Antigen receptors activate pathways that control cell survival, proliferation, and differentiation. Two important targets of antigen receptors, NF-κB and Jun N-terminal kinase (JNK), are activated downstream of CARMA1, a scaffolding protein that nucleates a complex including BCL10, MALT1, and other IκB kinase (IKK)-signalosome components. Somatic mutations that constitutively activate CARMA1 occur frequently in diffuse large B cell lymphoma (DLBCL) and mediate essential survival signals. Mechanisms that downregulate this pathway might thus yield important therapeutic targets. Stimulation of antigen receptors induces not only BCL10 activation but also its degradation downstream of CARMA1, thereby ultimately limiting signals to its downstream targets. Here, using lymphocyte cell models, we identify a kinase-independent requirement for TAK1 and its adaptor, TAB1, in antigen receptor-induced BCL10 degradation. We show that TAK1 acts as an adaptor for E3 ubiquitin ligases that target BCL10 for degradation. Functionally, TAK1 overexpression restrains CARMA1-dependent activation of NF-κB by reducing BCL10 levels. TAK1 also promotes counterselection of NF-κB-addicted DLBCL lines by a dual mechanism involving kinase-independent degradation of BCL10 and kinase-dependent activation of JNK. Thus, by directly promoting BCL10 degradation, TAK1 counterbalances NF-κB and JNK signals essential for the activation and survival of lymphocytes and CARMA1-addicted lymphoma types.
Collapse
|
22
|
Suerth JD, Schambach A, Baum C. Genetic modification of lymphocytes by retrovirus-based vectors. Curr Opin Immunol 2012; 24:598-608. [PMID: 22995202 DOI: 10.1016/j.coi.2012.08.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 08/23/2012] [Indexed: 01/02/2023]
Abstract
The genetic modification of lymphocytes is an important topic in the emerging field of gene therapy. Many clinical trials targeting immunodeficiency syndromes or cancer have shown therapeutic benefit; further applications address inflammatory and infectious disorders. Retroviral vector development requires a detailed understanding of the interactions with the host. Most researchers have used simple gammaretroviral vectors to modify lymphocytes, either directly or via hematopoietic stem and progenitor cells. Lentiviral, spumaviral (foamyviral) and alpharetroviral vectors were designed to reduce the necessity for cell stimulation and to utilize potentially safer integration properties. Novel surface modifications (pseudotyping) and transgenes, built using synthetic components, expand the retroviral toolbox, altogether promising increased specificity and potency. Product consistency will be an important criterion for routine clinical use.
Collapse
Affiliation(s)
- Julia D Suerth
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße 1, D-30625 Hannover, Germany
| | | | | |
Collapse
|
23
|
Coupling endonucleases with DNA end-processing enzymes to drive gene disruption. Nat Methods 2012; 9:973-5. [PMID: 22941364 DOI: 10.1038/nmeth.2177] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/27/2012] [Indexed: 11/08/2022]
Abstract
Targeted DNA double-strand breaks introduced by rare-cleaving designer endonucleases can be harnessed for gene disruption applications by engaging mutagenic nonhomologous end-joining DNA repair pathways. However, endonuclease-mediated DNA breaks are often subject to precise repair, which limits the efficiency of targeted genome editing. To address this issue, we coupled designer endonucleases to DNA end-processing enzymes to drive mutagenic break resolution, achieving up to 25-fold enhancements in gene disruption rates.
Collapse
|
24
|
Aiuti A, Bacchetta R, Seger R, Villa A, Cavazzana-Calvo M. Gene therapy for primary immunodeficiencies: Part 2. Curr Opin Immunol 2012; 24:585-91. [PMID: 22909900 DOI: 10.1016/j.coi.2012.07.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 07/31/2012] [Indexed: 12/21/2022]
Abstract
Gene therapy has become an attractive alternative therapeutic strategy to allogeneic transplant for primary immunodeficiencies (PIDs) owing to known genetic defects. Clinical trials using gammaretroviral vectors have demonstrated the proof of principle of gene therapy for Wiskott-Aldrich syndrome (WAS) and chronic granulomatous disease (CGD), but have also highlighted limitations of the technology. New strategies based on vectors that can achieve more robust correction with less risk of insertional mutagenesis are being developed. In this review we present the status of gene therapy for WAS and CGD, and discuss the emerging application of similar strategies to a broader range of PIDs, such as IPEX syndrome.
Collapse
Affiliation(s)
- Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Division of Regenerative Medicine, San Raffaele Scientific Institute, Milan, Italy.
| | | | | | | | | |
Collapse
|
25
|
Andrews SF, Dai X, Ryu BY, Gulick T, Ramachandran B, Rawlings DJ. Developmentally regulated expression of MEF2C limits the response to BCR engagement in transitional B cells. Eur J Immunol 2012; 42:1327-36. [PMID: 22311635 DOI: 10.1002/eji.201142226] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Transitional and naïve mature peripheral B cells respond very differently to B-cell receptor (BCR) cross-linking. While transitional B cells undergo apoptosis upon BCR engagement, mature B cells survive and proliferate. This differential response correlates with the capacity of mature, but not transitional B cells to transcribe genes that promote cell survival and proliferation, including those encoding c-Myc and the Bcl-2 family members Bcl-xL and A1. We recently demonstrated that transitional B cells fail to assemble transcriptional machinery at the promoter region of these target genes despite equivalent cytoplasmic signaling and nuclear translocation of key transcription factors including NF-κB and nuclear factor of activated T cells (NFAT). The transcription factor myocyte enhancer factor-2C (MEF2C) is regulated by both calcineurin and mitogen-activated protein kinase signaling pathways, and is essential for proliferation and survival downstream of BCR engagement in mature B cells. In this work, we demonstrate that transitional B cells have intrinsically low levels of MEF2C protein and DNA-binding activity, and that this developmental difference in MEF2C expression is functionally significant. Forced expression of MEF2C in transitional B cells promoted cell survival, proliferation, and upregulation of pro-survival genes. Thus, low MEF2C expression limits transitional B-cell responsiveness to BCR engagement before these cells reach maturity.
Collapse
Affiliation(s)
- Sarah F Andrews
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | | | | | | |
Collapse
|
26
|
Baxter S, Lambert AR, Kuhar R, Jarjour J, Kulshina N, Parmeggiani F, Danaher P, Gano J, Baker D, Stoddard BL, Scharenberg AM. Engineering domain fusion chimeras from I-OnuI family LAGLIDADG homing endonucleases. Nucleic Acids Res 2012; 40:7985-8000. [PMID: 22684507 PMCID: PMC3439895 DOI: 10.1093/nar/gks502] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although engineered LAGLIDADG homing endonucleases (LHEs) are finding increasing applications in biotechnology, their generation remains a challenging, industrial-scale process. As new single-chain LAGLIDADG nuclease scaffolds are identified, however, an alternative paradigm is emerging: identification of an LHE scaffold whose native cleavage site is a close match to a desired target sequence, followed by small-scale engineering to modestly refine recognition specificity. The application of this paradigm could be accelerated if methods were available for fusing N- and C-terminal domains from newly identified LHEs into chimeric enzymes with hybrid cleavage sites. Here we have analyzed the structural requirements for fusion of domains extracted from six single-chain I-OnuI family LHEs, spanning 40–70% amino acid identity. Our analyses demonstrate that both the LAGLIDADG helical interface residues and the linker peptide composition have important effects on the stability and activity of chimeric enzymes. Using a simple domain fusion method in which linker peptide residues predicted to contact their respective domains are retained, and in which limited variation is introduced into the LAGLIDADG helix and nearby interface residues, catalytically active enzymes were recoverable for ∼70% of domain chimeras. This method will be useful for creating large numbers of chimeric LHEs for genome engineering applications.
Collapse
Affiliation(s)
- Sarah Baxter
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ubiquitous high-level gene expression in hematopoietic lineages provides effective lentiviral gene therapy of murine Wiskott-Aldrich syndrome. Blood 2012; 119:4395-407. [PMID: 22431569 DOI: 10.1182/blood-2011-03-340711] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The immunodeficiency disorder Wiskott-Aldrich syndrome (WAS) leads to life-threatening hematopoietic cell dysfunction. We used WAS protein (WASp)-deficient mice to analyze the in vivo efficacy of lentiviral (LV) vectors using either a viral-derived promoter, MND, or the human proximal WAS promoter (WS1.6) for human WASp expression. Transplantation of stem cells transduced with MND-huWASp LV resulted in sustained, endogenous levels of WASp in all hematopoietic lineages, progressive selection for WASp+ T, natural killer T and B cells, rescue of T-cell proliferation and cytokine production, and substantial restoration of marginal zone (MZ) B cells. In contrast, WS1.6-huWASp LV recipients exhibited subendogenous WASp expression in all cell types with only partial selection of WASp+ T cells and limited correction in MZ B-cell numbers. In parallel, WS1.6-huWASp LV recipients exhibited an altered B-cell compartment, including higher numbers of λ-light-chain+ naive B cells, development of self-reactive CD11c+FAS+ B cells, and evidence for spontaneous germinal center (GC) responses. These observations correlated with B-cell hyperactivity and increased titers of immunoglobulin (Ig)G2c autoantibodies, suggesting that partial gene correction may predispose toward autoimmunity. Our findings identify the advantages and disadvantages associated with each vector and suggest further clinical development of the MND-huWASp LV for a future clinical trial for WAS.
Collapse
|
28
|
Drake AC, Chen Q, Chen J. Engineering humanized mice for improved hematopoietic reconstitution. Cell Mol Immunol 2012; 9:215-24. [PMID: 22425741 DOI: 10.1038/cmi.2012.6] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Humanized mice are immunodeficient animals engrafted with human hematopoietic stem cells that give rise to various lineages of human blood cells throughout the life of the mouse. This article reviews recent advances in the generation of humanized mice, focusing on practical considerations. We discuss features of different immunodeficient recipient mouse strains, sources of human hematopoietic stem cells, advances in expansion and genetic modification of hematopoietic stem cells, and techniques to modulate the cytokine environment of recipient mice, in order to enhance reconstitution of specific human blood lineage cells. We highlight the opportunities created by new technologies and discuss practical considerations on how to make best use of the widening array of basic models for specific research applications.
Collapse
Affiliation(s)
- Adam C Drake
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | |
Collapse
|
29
|
Bandaranayake AD, Correnti C, Ryu BY, Brault M, Strong RK, Rawlings DJ. Daedalus: a robust, turnkey platform for rapid production of decigram quantities of active recombinant proteins in human cell lines using novel lentiviral vectors. Nucleic Acids Res 2011; 39:e143. [PMID: 21911364 PMCID: PMC3241668 DOI: 10.1093/nar/gkr706] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
A key challenge for the academic and biopharmaceutical communities is the rapid and scalable production of recombinant proteins for supporting downstream applications ranging from therapeutic trials to structural genomics efforts. Here, we describe a novel system for the production of recombinant mammalian proteins, including immune receptors, cytokines and antibodies, in a human cell line culture system, often requiring <3 weeks to achieve stable, high-level expression: Daedalus. The inclusion of minimized ubiquitous chromatin opening elements in the transduction vectors is key for preventing genomic silencing and maintaining the stability of decigram levels of expression. This system can bypass the tedious and time-consuming steps of conventional protein production methods by employing the secretion pathway of serum-free adapted human suspension cell lines, such as 293 Freestyle. Using optimized lentiviral vectors, yields of 20–100 mg/l of correctly folded and post-translationally modified, endotoxin-free protein of up to ~70 kDa in size, can be achieved in conventional, small-scale (100 ml) culture. At these yields, most proteins can be purified using a single size-exclusion chromatography step, immediately appropriate for use in structural, biophysical or therapeutic applications.
Collapse
|
30
|
Abstract
INTRODUCTION Retroviral vectors have been developed for hematopoietic stem cell (HSC) gene therapy and have successfully cured X-linked severe combined immunodeficiency (SCID-X1), adenosine deaminase deficiency (ADA-SCID), adrenoleukodystrophy, and Wiskott-Aldrich syndrome. However, in HSC gene therapy clinical trials, genotoxicity mediated by integrated vector proviruses has led to clonal expansion, and in some cases frank leukemia. Numerous studies have been performed to understand the molecular basis of vector-mediated genotoxicity with the aim of developing safer vectors and safer gene therapy protocols. These genotoxicity studies are critical to advancing HSC gene therapy. AREAS COVERED This review provides an introduction to the mechanisms of retroviral vector genotoxicity. It also covers advances over the last 20 years in designing safer gene therapy vectors, and in integration site analysis in clinical trials and large animal models. Mechanisms of retroviral-mediated genotoxicity, and the risk factors that contribute to clonal expansion and leukemia in HSC gene therapy are introduced. EXPERT OPINION Continued research on virus-host interactions and next-generation vectors should further improve the safety of future HSC gene therapy vectors and protocols.
Collapse
Affiliation(s)
- Grant D Trobridge
- Washington State University, Department of Pharmaceutical Sciences and School of Molecular Biosciences, P.O. Box 646534, Pullman, WA 99164-6534, USA.
| |
Collapse
|