1
|
Montini E, Naldini L, Booth C, Kohn DB, Aiuti A. Balancing efficacy and safety in lentiviral vector-mediated hematopoietic stem cell gene therapy. Mol Ther 2025; 33:6-8. [PMID: 39729983 PMCID: PMC11764623 DOI: 10.1016/j.ymthe.2024.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/29/2024] Open
Affiliation(s)
- Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita - Salute San Raffaele University Medical School, Milan, Italy
| | - Claire Booth
- UCL Great Ormond Street Institute of Child Health, London, UK; Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - Donald B Kohn
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita - Salute San Raffaele University Medical School, Milan, Italy; Pediatric Immunohematology and BMT, San Raffaele Hospital, Milan, Italy
| |
Collapse
|
2
|
Rui X, Calderon FA, Wobma H, Gerdemann U, Albanese A, Cagnin L, McGuckin C, Michaelis KA, Naqvi K, Blazar BR, Tkachev V, Kean LS. Human OX40L-CAR-T regs target activated antigen-presenting cells and control T cell alloreactivity. Sci Transl Med 2024; 16:eadj9331. [PMID: 39413160 PMCID: PMC11789419 DOI: 10.1126/scitranslmed.adj9331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/18/2024]
Abstract
Regulatory T cells (Tregs) make major contributions to immune homeostasis. Because Treg dysfunction can lead to both allo- and autoimmunity, there is interest in correcting these disorders through Treg adoptive transfer. Two of the central challenges in clinically deploying Treg cellular therapies are ensuring phenotypic stability and maximizing potency. Here, we describe an approach to address both issues through the creation of OX40 ligand (OX40L)-specific chimeric antigen receptor (CAR)-Tregs under the control of a synthetic forkhead box P3 (FOXP3) promoter. The creation of these CAR-Tregs enabled selective Treg stimulation by engagement of OX40L, a key activation antigen in alloimmunity, including both graft-versus-host disease and solid organ transplant rejection, and autoimmunity, including rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus. We demonstrated that OX40L-CAR-Tregs were robustly activated in the presence of OX40L-expressing cells, leading to up-regulation of Treg suppressive proteins without induction of proinflammatory cytokine production. Compared with control Tregs, OX40L-CAR-Tregs more potently suppressed alloreactive T cell proliferation in vitro and were directly inhibitory toward activated monocyte-derived dendritic cells (DCs). We identified trogocytosis as one of the central mechanisms by which these CAR-Tregs effectively decrease extracellular display of OX40L, resulting in decreased DC stimulatory capacity. OX40L-CAR-Tregs demonstrated an enhanced ability to control xenogeneic graft-versus-host disease compared with control Tregs without abolishing the graft-versus-leukemia effect. These results suggest that OX40L-CAR-Tregs may have wide applicability as a potent cellular therapy to control both allo- and autoimmune diseases.
Collapse
Affiliation(s)
- Xianliang Rui
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Francesca Alvarez Calderon
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Holly Wobma
- Harvard Medical School, Boston, MA 02115, USA
- Division of Immunology, Boston Children’s Hospital, Boston, MA 02215, USA
| | - Ulrike Gerdemann
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Alexandre Albanese
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Lorenzo Cagnin
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Connor McGuckin
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | - Kisa Naqvi
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Bruce R. Blazar
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Victor Tkachev
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Leslie S. Kean
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Duncan CN, Bledsoe JR, Grzywacz B, Beckman A, Bonner M, Eichler FS, Kühl JS, Harris MH, Slauson S, Colvin RA, Prasad VK, Downey GF, Pierciey FJ, Kinney MA, Foos M, Lodaya A, Floro N, Parsons G, Dietz AC, Gupta AO, Orchard PJ, Thakar HL, Williams DA. Hematologic Cancer after Gene Therapy for Cerebral Adrenoleukodystrophy. N Engl J Med 2024; 391:1287-1301. [PMID: 39383458 PMCID: PMC11846662 DOI: 10.1056/nejmoa2405541] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
BACKGROUND Gene therapy with elivaldogene autotemcel (eli-cel) consisting of autologous CD34+ cells transduced with lentiviral vector containing ABCD1 complementary DNA (Lenti-D) has shown efficacy in clinical studies for the treatment of cerebral adrenoleukodystrophy. However, the risk of oncogenesis with eli-cel is unclear. METHODS We performed integration-site analysis, genetic studies, flow cytometry, and morphologic studies in peripheral-blood and bone marrow samples from patients who received eli-cel therapy in two completed phase 2-3 studies (ALD-102 and ALD-104) and an ongoing follow-up study (LTF-304) involving the patients in both ALD-102 and ALD-104. RESULTS Hematologic cancer developed in 7 of 67 patients after the receipt of eli-cel (1 of 32 patients in the ALD-102 study and 6 of 35 patients in the ALD-104 study): myelodysplastic syndrome (MDS) with unilineage dysplasia in 2 patients at 14 and 26 months; MDS with excess blasts in 3 patients at 28, 42, and 92 months; MDS in 1 patient at 36 months; and acute myeloid leukemia (AML) in 1 patient at 57 months. In the 6 patients with available data, predominant clones contained lentiviral vector insertions at multiple loci, including at either MECOM-EVI1 (MDS and EVI1 complex protein EVI1 [ecotropic virus integration site 1], in 5 patients) or PRDM16 (positive regulatory domain zinc finger protein 16, in 1 patient). Several patients had cytopenias, and most had vector insertions in multiple genes within the same clone; 6 of the 7 patients also had somatic mutations (KRAS, NRAS, WT1, CDKN2A or CDKN2B, or RUNX1), and 1 of the 7 patients had monosomy 7. Of the 5 patients with MDS with excess blasts or MDS with unilineage dysplasia who underwent allogeneic hematopoietic stem-cell transplantation (HSCT), 4 patients remain free of MDS without recurrence of symptoms of cerebral adrenoleukodystrophy, and 1 patient died from presumed graft-versus-host disease 20 months after HSCT (49 months after receiving eli-cel). The patient with AML is alive and had full donor chimerism after HSCT; the patient with the most recent case of MDS is alive and awaiting HSCT. CONCLUSIONS Hematologic cancer developed in a subgroup of patients who were treated with eli-cel; the cases are associated with clonal vector insertions within oncogenes and clonal evolution with acquisition of somatic genetic defects. (Funded by Bluebird Bio; ALD-102, ALD-104, and LTF-304 ClinicalTrials.gov numbers, NCT01896102, NCT03852498, and NCT02698579, respectively.).
Collapse
Affiliation(s)
- Christine N Duncan
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Jacob R Bledsoe
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Bartosz Grzywacz
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Amy Beckman
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Melissa Bonner
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Florian S Eichler
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Jörn-Sven Kühl
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Marian H Harris
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Sarah Slauson
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Richard A Colvin
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Vinod K Prasad
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Gerald F Downey
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Francis J Pierciey
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Melissa A Kinney
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Marianna Foos
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Ankit Lodaya
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Nicole Floro
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Geoffrey Parsons
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Andrew C Dietz
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Ashish O Gupta
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Paul J Orchard
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - Himal L Thakar
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| | - David A Williams
- From Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School (C.N.D., D.A.W.), the Department of Pathology, Boston Children's Hospital (J.R.B., M.H.H.), and Massachusetts General Hospital and Harvard Medical School (F.S.E.) - all in Boston; the Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center (B.G., A.B.), and the Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota (A.O.G., P.J.O.) - both in Minneapolis; Bluebird Bio, Somerville, MA (M.B., S.S., R.A.C., V.K.P., G.F.D., F.J.P., M.A.K., M.F., A.L., N.F., G.P., A.C.D., H.L.T.); the Department of Pediatric Oncology, Hematology and Hemostaseology, Leipzig University Hospital, Leipzig, Germany (J.-S.K.); and the Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC (V.K.P.)
| |
Collapse
|
4
|
Yadav B, Singh D, Mantri S, Rishi V. Genome-wide Methylation Dynamics and Context-dependent Gene Expression Variability in Differentiating Preadipocytes. J Endocr Soc 2024; 8:bvae121. [PMID: 38966711 PMCID: PMC11222978 DOI: 10.1210/jendso/bvae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Indexed: 07/06/2024] Open
Abstract
Obesity, characterized by the accumulation of excess fat, is a complex condition resulting from the combination of genetic and epigenetic factors. Recent studies have found correspondence between DNA methylation and cell differentiation, suggesting a role of the former in cell fate determination. There is a lack of comprehensive understanding concerning the underpinnings of preadipocyte differentiation, specifically when cells are undergoing terminal differentiation (TD). To gain insight into dynamic genome-wide methylation, 3T3 L1 preadipocyte cells were differentiated by a hormone cocktail. The genomic DNA was isolated from undifferentiated cells and 4 hours, 2 days postdifferentiated cells, and 15 days TD cells. We employed whole-genome bisulfite sequencing (WGBS) to ascertain global genomic DNA methylation alterations at single base resolution as preadipocyte cells differentiate. The genome-wide distribution of DNA methylation showed similar overall patterns in pre-, post-, and terminally differentiated adipocytes, according to WGBS analysis. DNA methylation decreases at 4 hours after differentiation initiation, followed by methylation gain as cells approach TD. Studies revealed novel differentially methylated regions (DMRs) associated with adipogenesis. DMR analysis suggested that though DNA methylation is global, noticeable changes are observed at specific sites known as "hotspots." Hotspots are genomic regions rich in transcription factor (TF) binding sites and exhibit methylation-dependent TF binding. Subsequent analysis indicated hotspots as part of DMRs. The gene expression profile of key adipogenic genes in differentiating adipocytes is context-dependent, as we found a direct and inverse relationship between promoter DNA methylation and gene expression.
Collapse
Affiliation(s)
- Binduma Yadav
- Nutritional Biotechnology, National Agri-Food Biotechnology Institute, Mohali, Punjab 140306, India
- Regional Center for Biotechnology, Faridabad, Haryana 160014, India
| | - Dalwinder Singh
- Nutritional Biotechnology, National Agri-Food Biotechnology Institute, Mohali, Punjab 140306, India
- Department of Anatomy and Cell Biology, Western University, London, Ontario N6A 5C1, Canada
| | - Shrikant Mantri
- Nutritional Biotechnology, National Agri-Food Biotechnology Institute, Mohali, Punjab 140306, India
| | - Vikas Rishi
- Nutritional Biotechnology, National Agri-Food Biotechnology Institute, Mohali, Punjab 140306, India
| |
Collapse
|
5
|
Singh S, Pugliano CM, Honaker Y, Laird A, DeGottardi MQ, Lopez E, Lachkar S, Stoffers C, Sommer K, Khan IF, Rawlings DJ. Efficient and sustained FOXP3 locus editing in hematopoietic stem cells as a therapeutic approach for IPEX syndrome. Mol Ther Methods Clin Dev 2024; 32:101183. [PMID: 38282895 PMCID: PMC10818254 DOI: 10.1016/j.omtm.2023.101183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 12/20/2023] [Indexed: 01/30/2024]
Abstract
Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a monogenic disorder caused by mutations in the FOXP3 gene, required for generation of regulatory T (Treg) cells. Loss of Treg cells leads to immune dysregulation characterized by multi-organ autoimmunity and early mortality. Hematopoietic stem cell (HSC) transplantation can be curative, but success is limited by autoimmune complications, donor availability and/or graft-vs.-host disease. Correction of FOXP3 in autologous HSC utilizing a homology-directed repair (HDR)-based platform may provide a safer alternative therapy. Here, we demonstrate efficient editing of FOXP3 utilizing co-delivery of Cas9 ribonucleoprotein complexes and adeno-associated viral vectors to achieve HDR rates of >40% in vitro using mobilized CD34+ cells from multiple donors. Using this approach to deliver either a GFP or a FOXP3 cDNA donor cassette, we demonstrate sustained bone marrow engraftment of approximately 10% of HDR-edited cells in immune-deficient recipient mice at 16 weeks post-transplant. Further, we show targeted integration of FOXP3 cDNA in CD34+ cells from an IPEX patient and expression of the introduced FOXP3 transcript in gene-edited primary T cells from both healthy individuals and IPEX patients. Our combined findings suggest that refinement of this approach is likely to provide future clinical benefit in IPEX.
Collapse
Affiliation(s)
- Swati Singh
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Cole M. Pugliano
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Yuchi Honaker
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Aidan Laird
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - M. Quinn DeGottardi
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Ezra Lopez
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Stefan Lachkar
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Claire Stoffers
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Karen Sommer
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Iram F. Khan
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - David J. Rawlings
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
- Department of Immunology, University of Washington, Seattle, WA 98101, USA
| |
Collapse
|
6
|
Castiello MC, Ferrari S, Villa A. Correcting inborn errors of immunity: From viral mediated gene addition to gene editing. Semin Immunol 2023; 66:101731. [PMID: 36863140 PMCID: PMC10109147 DOI: 10.1016/j.smim.2023.101731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/25/2023] [Accepted: 02/14/2023] [Indexed: 03/04/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation is an effective treatment to cure inborn errors of immunity. Remarkable progress has been achieved thanks to the development and optimization of effective combination of advanced conditioning regimens and use of immunoablative/suppressive agents preventing rejection as well as graft versus host disease. Despite these tremendous advances, autologous hematopoietic stem/progenitor cell therapy based on ex vivo gene addition exploiting integrating γ-retro- or lenti-viral vectors, has demonstrated to be an innovative and safe therapeutic strategy providing proof of correction without the complications of the allogeneic approach. The recent advent of targeted gene editing able to precisely correct genomic variants in an intended locus of the genome, by introducing deletions, insertions, nucleotide substitutions or introducing a corrective cassette, is emerging in the clinical setting, further extending the therapeutic armamentarium and offering a cure to inherited immune defects not approachable by conventional gene addition. In this review, we will analyze the current state-of-the art of conventional gene therapy and innovative protocols of genome editing in various primary immunodeficiencies, describing preclinical models and clinical data obtained from different trials, highlighting potential advantages and limits of gene correction.
Collapse
Affiliation(s)
- Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy.
| |
Collapse
|
7
|
Smith MC, Belur LR, Karlen AD, Erlanson O, Podetz-Pedersen KM, McKenzie J, Detellis J, Gagnidze K, Parsons G, Robinson N, Labarre S, Shah S, Furcich J, Lund TC, Tsai HC, McIvor RS, Bonner M. Phenotypic Correction of Murine Mucopolysaccharidosis Type II by Engraftment of Ex Vivo Lentiviral Vector-Transduced Hematopoietic Stem and Progenitor Cells. Hum Gene Ther 2022; 33:1279-1292. [PMID: 36226412 PMCID: PMC9808798 DOI: 10.1089/hum.2022.141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mucopolysaccharidosis type II (MPS II, Hunter syndrome) is an X-linked recessive lysosomal disease caused by deficiency of iduronate-2-sulfatase (IDS). The absence of IDS results in the accumulation of the glycosaminoglycans (GAGs) heparan sulfate and dermatan sulfate. Currently, the only approved treatment option for MPS II is enzyme replacement therapy (ERT), Elaprase. However, ERT is demanding for the patient and does not ameliorate neurological manifestations of the disease. Using an IDS-deficient mouse model that phenocopies the human disease, we evaluated hematopoietic stem and progenitor cells (HSPCs) transduced with a lentiviral vector (LVV) carrying a codon-optimized human IDS coding sequence regulated by a ubiquitous MNDU3 promoter (MNDU3-IDS). Mice treated with MNDU3-IDS LVV-transduced cells showed supraphysiological levels of IDS enzyme activity in plasma, peripheral blood mononuclear cells, and in most analyzed tissues. These enzyme levels were sufficient to normalize GAG storage in analyzed tissues. Importantly, IDS levels in the brains of MNDU3-IDS-engrafted animals were restored to 10-20% than that of wild-type mice, sufficient to normalize GAG content and prevent emergence of cognitive deficit as evaluated by neurobehavioral testing. These results demonstrate the potential effectiveness of ex vivo MNDU3-IDS LVV-transduced HSPCs for treatment of MPS II.
Collapse
Affiliation(s)
- Miles C. Smith
- Center for Genome Engineering, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lalitha R. Belur
- Center for Genome Engineering, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrea D. Karlen
- Center for Genome Engineering, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Olivia Erlanson
- Center for Genome Engineering, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kelly M. Podetz-Pedersen
- Center for Genome Engineering, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | | | | | | | - Saumil Shah
- bluebird bio, Inc., Cambridge, Massachusetts, USA
| | - Justin Furcich
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Troy C. Lund
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - R. Scott McIvor
- Center for Genome Engineering, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA,Correspondence: Dr. R. Scott McIvor, Department of Genetics, Cell Biology and Development, University of Minnesota, 6-160 Jackson Hall, 321 Church St. S.E., Minneapolis, MN 55455, USA.
| | | |
Collapse
|
8
|
Gupta AO, Raymond G, Pierpont RI, Kemp S, McIvor RS, Rayannavar A, Miller B, Lund TC, Orchard PJ. Treatment of cerebral adrenoleukodystrophy: allogeneic transplantation and lentiviral gene therapy. Expert Opin Biol Ther 2022; 22:1151-1162. [DOI: 10.1080/14712598.2022.2124857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Ashish O Gupta
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| | - Gerald Raymond
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rene I Pierpont
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, University of Minnesota
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC - University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Neuroscience, 1105 AZ Amsterdam, The Netherlands
| | - R Scott McIvor
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota
| | | | - Bradley Miller
- Division of Pediatric Endocrinology, University of Minnesota
| | - Troy C Lund
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| |
Collapse
|
9
|
Laoharawee K, Johnson MJ, Lahr WS, Sipe CJ, Kleinboehl E, Peterson JJ, Lonetree CL, Bell JB, Slipek NJ, Crane AT, Webber BR, Moriarity BS. A Pan-RNase Inhibitor Enabling CRISPR-mRNA Platforms for Engineering of Primary Human Monocytes. Int J Mol Sci 2022; 23:9749. [PMID: 36077152 PMCID: PMC9456164 DOI: 10.3390/ijms23179749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/16/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
Monocytes and their downstream effectors are critical components of the innate immune system. Monocytes are equipped with chemokine receptors, allowing them to migrate to various tissues, where they can differentiate into macrophage and dendritic cell subsets and participate in tissue homeostasis, infection, autoimmune disease, and cancer. Enabling genome engineering in monocytes and their effector cells will facilitate a myriad of applications for basic and translational research. Here, we demonstrate that CRISPR-Cas9 RNPs can be used for efficient gene knockout in primary human monocytes. In addition, we demonstrate that intracellular RNases are likely responsible for poor and heterogenous mRNA expression as incorporation of pan-RNase inhibitor allows efficient genome engineering following mRNA-based delivery of Cas9 and base editor enzymes. Moreover, we demonstrate that CRISPR-Cas9 combined with an rAAV vector DNA donor template mediates site-specific insertion and expression of a transgene in primary human monocytes. Finally, we demonstrate that SIRPa knock-out monocyte-derived macrophages have enhanced activity against cancer cells, highlighting the potential for application in cellular immunotherapies.
Collapse
Affiliation(s)
- Kanut Laoharawee
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matthew J. Johnson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Walker S. Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher J. Sipe
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Evan Kleinboehl
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph J. Peterson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Cara-lin Lonetree
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jason B. Bell
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicholas J. Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Andrew T. Crane
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Beau R. Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Branden S. Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
10
|
Sonzogni O, Zak DE, Sasso MS, Lear R, Muntzer A, Zonca M, West K, Champion BR, Rottman JB. T-SIGn tumor reengineering therapy and CAR T cells synergize in combination therapy to clear human lung tumor xenografts and lung metastases in NSG mice. Oncoimmunology 2022; 11:2029070. [PMID: 35154906 PMCID: PMC8837249 DOI: 10.1080/2162402x.2022.2029070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although chimeric antigen receptor (CAR) T cells have emerged as highly effective treatments for patients with hematologic malignancies, similar efficacy has not been achieved in the context of solid tumors. There are several reasons for this disparity including a) fewer solid tumor target antigens, b) heterogenous target expression amongst tumor cells, c) poor trafficking of CAR T cells to the solid tumor and d) an immunosuppressive tumor microenvironment (TME). Oncolytic viruses have the potential to change this paradigm by a) directly lysing tumor cells and releasing tumor neoantigens, b) stimulating the local host innate immune response to release cytokines and recruit additional innate and adaptive immune cells, c) carrying virus-encoded transgenes to “re-program” the TME to a pro-inflammatory environment and d) promoting an adaptive immune response to the neoantigens in this newly permissive TME. Here we show that the Tumor-Specific Immuno-Gene (T-SIGn) virus NG-347 which encodes IFNα, MIP1α and CD80 synergizes with anti-EGFR CAR T cells as well as anti-HER-2 CAR T cells to clear A549 human tumor xenografts and their pulmonary metastases at doses which are subtherapeutic when each is used as a sole treatment. We show that NG-347 changes the TME to a pro-inflammatory environment resulting in the recruitment and activation of both CAR T cells and mouse innate immune cells. We also show that the transgenes encoded by the virus are critical as synergy is lost in their absence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Katy West
- PsiOxus Therapeutics Limited, Abingdon, UK
| | | | | |
Collapse
|
11
|
Takushi SE, Paik NY, Fedanov A, Prince C, Doering CB, Spencer HT, Chandrakasan S. Lentiviral Gene Therapy for Familial Hemophagocytic Lymphohistiocytosis Type 3, Caused by UNC13D Genetic Defects. Hum Gene Ther 2021; 31:626-638. [PMID: 32253931 DOI: 10.1089/hum.2019.329] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Familial hemophagocytic lymphohistiocytosis type 3 (FHL3) is a rare disease caused by mutations to the UNC13D gene and the subsequent absence or decreased activity of the Munc13-4 protein. Munc13-4 is essential for the exocytosis of perforin and granzyme containing granules from cytotoxic cells. Without it, these cells are able to recognize an immunological insult but are unable to execute their cytotoxic functions. The result is a hyperinflammatory state that, if left untreated, is fatal. At present, the only curative treatment is hematopoietic stem cell transplantation (HSCT), but eligibility and response to this treatment are largely dependent on the ability to control inflammation before HSCT. In this study, we describe an optimized lentiviral vector that can restore Munc13-4 expression and degranulation capacity in both transduced FHL3 patient T cells and transduced hematopoietic stem cells from the FHL3 (Jinx) disease model.
Collapse
Affiliation(s)
- Sarah E Takushi
- Department of Immunology and Molecular Pathogenesis, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, Georgia, USA.,Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Na Yoon Paik
- Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Andrew Fedanov
- Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Chengyu Prince
- Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Christopher B Doering
- Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Emory University, Atlanta, Georgia, USA.,Department of Molecular and Systems Pharmacology, Graduate Division of Biological and Biomedical Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - H Trent Spencer
- Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Emory University, Atlanta, Georgia, USA.,Department of Molecular and Systems Pharmacology, Graduate Division of Biological and Biomedical Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shanmuganathan Chandrakasan
- Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Emory University, Atlanta, Georgia, USA.,Bone Marrow Transplant Program, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
12
|
Wang X, Fu RY, Li C, Chen CY, Firrman J, Konkle BA, Zhang J, Li L, Xiao W, Poncz M, Miao CH. Enhancing therapeutic efficacy of in vivo platelet-targeted gene therapy in hemophilia A mice. Blood Adv 2020; 4:5722-5734. [PMID: 33216891 PMCID: PMC7686911 DOI: 10.1182/bloodadvances.2020002479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/15/2020] [Indexed: 11/20/2022] Open
Abstract
Our previous studies demonstrated that intraosseous (IO) infusion of lentiviral vectors (LVs) carrying a modified B domain-deleted factor VIII (FVIII) transgene driven by a megakaryocyte-specific promoter (GP1Bα promoter; G-F8/N6-LV) successfully transduced hematopoietic stem cells (HSCs) to produce FVIII stored in the platelet α-granules. Platelet FVIII corrected the bleeding phenotype with limited efficacy in hemophilia A (HemA) mice with and without preexisting anti-FVIII inhibitors. The present study sought to further enhance the therapeutic efficacy of this treatment protocol by increasing both the efficiency of LV transduction and the functional activity of platelet FVIII. A combined drug regimen of dexamethasone and anti-CD8α monoclonal antibody enhanced the percentage of transduced bone marrow and HSCs over time. In G-F8/N6-LV-treated HemA mice, significant improvement in phenotypic correction was observed on day 84. To improve platelet FVIII functionality, genes encoding FVIII variant F8X10K12 with increased expression or F8N6K12RH with increased functional activity compared with F8/N6 were incorporated into LVs. Treatment with G-F8X10K12-LV in HemA mice produced a higher level of platelet FVIII but induced anti-FVIII inhibitors. After treatment with combined drugs and IO infusion of G-F8/N6K12RH-LV, HemA mice showed significant phenotypic correction without anti-FVIII inhibitor formation. These results indicate that new human FVIII variant F8/N6K12RH combined with immune suppression could significantly enhance the therapeutic efficacy of in vivo platelet-targeted gene therapy for murine HemA via IO delivery. This protocol provides a safe and effective treatment for hemophilia that may be translatable to and particularly beneficial for patients with preexisting inhibitory antibodies to FVIII.
Collapse
Affiliation(s)
- Xuefeng Wang
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Richard Y Fu
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Chong Li
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Chun-Yu Chen
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Jenni Firrman
- Department of Pediatrics, Indiana University, Indianapolis, IN
| | | | - Junping Zhang
- Department of Pediatrics, Indiana University, Indianapolis, IN
| | - Lei Li
- Department of Chemistry, Georgia State University, Atlanta, GA; and
| | - Weidong Xiao
- Department of Pediatrics, Indiana University, Indianapolis, IN
| | - Mortimer Poncz
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Carol H Miao
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| |
Collapse
|
13
|
Magnani CF, Gaipa G, Lussana F, Belotti D, Gritti G, Napolitano S, Matera G, Cabiati B, Buracchi C, Borleri G, Fazio G, Zaninelli S, Tettamanti S, Cesana S, Colombo V, Quaroni M, Cazzaniga G, Rovelli A, Biagi E, Galimberti S, Calabria A, Benedicenti F, Montini E, Ferrari S, Introna M, Balduzzi A, Valsecchi MG, Dastoli G, Rambaldi A, Biondi A. Sleeping Beauty-engineered CAR T cells achieve antileukemic activity without severe toxicities. J Clin Invest 2020; 130:6021-6033. [PMID: 32780725 PMCID: PMC7598053 DOI: 10.1172/jci138473] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/29/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUNDChimeric antigen receptor (CAR) T cell immunotherapy has resulted in complete remission (CR) and durable response in highly refractory patients. However, logistical complexity and high costs of manufacturing autologous viral products limit CAR T cell availability.METHODSWe report the early results of a phase I/II trial in B cell acute lymphoblastic leukemia (B-ALL) patients relapsed after allogeneic hematopoietic stem cell transplantation (HSCT) using donor-derived CD19 CAR T cells generated with the Sleeping Beauty (SB) transposon and differentiated into cytokine-induced killer (CIK) cells.RESULTSThe cellular product was produced successfully for all patients from the donor peripheral blood (PB) and consisted mostly of CD3+ lymphocytes with 43% CAR expression. Four pediatric and 9 adult patients were infused with a single dose of CAR T cells. Toxicities reported were 2 grade I and 1 grade II cytokine-release syndrome (CRS) cases at the highest dose in the absence of graft-versus-host disease (GVHD), neurotoxicity, or dose-limiting toxicities. Six out of 7 patients receiving the highest doses achieved CR and CR with incomplete blood count recovery (CRi) at day 28. Five out of 6 patients in CR were also minimal residual disease negative (MRD-). Robust expansion was achieved in the majority of the patients. CAR T cells were measurable by transgene copy PCR up to 10 months. Integration site analysis showed a positive safety profile and highly polyclonal repertoire in vitro and at early time points after infusion.CONCLUSIONSB-engineered CAR T cells expand and persist in pediatric and adult B-ALL patients relapsed after HSCT. Antileukemic activity was achieved without severe toxicities.TRIAL REGISTRATIONClinicalTrials.gov NCT03389035.FUNDINGThis study was supported by grants from the Fondazione AIRC per la Ricerca sul Cancro (AIRC); Cancer Research UK (CRUK); the Fundación Científica de la Asociación Española Contra el Cáncer (FC AECC); Ministero Della Salute; Fondazione Regionale per la Ricerca Biomedica (FRRB).
Collapse
Affiliation(s)
- Chiara F. Magnani
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Giuseppe Gaipa
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Federico Lussana
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Daniela Belotti
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
- Department of Pediatrics, University of Milano–Bicocca, Milan, Italy
| | - Giuseppe Gritti
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Sara Napolitano
- Clinica Pediatrica, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Giada Matera
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Benedetta Cabiati
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Chiara Buracchi
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Gianmaria Borleri
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Grazia Fazio
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | | | - Sarah Tettamanti
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Stefania Cesana
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Valentina Colombo
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Michele Quaroni
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Giovanni Cazzaniga
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Attilio Rovelli
- Clinica Pediatrica, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Ettore Biagi
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
- Clinica Pediatrica, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Stefania Galimberti
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre, Department of Medicine and Surgery, University of Milano–Bicocca, Milan, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET)/IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET)/IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET)/IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Ferrari
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Martino Introna
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
- USS Centro di Terapia Cellulare “G. Lanzani,” Bergamo, Italy
| | - Adriana Balduzzi
- Department of Pediatrics, University of Milano–Bicocca, Milan, Italy
- Clinica Pediatrica, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Maria Grazia Valsecchi
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre, Department of Medicine and Surgery, University of Milano–Bicocca, Milan, Italy
| | - Giuseppe Dastoli
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Andrea Biondi
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
- Clinica Pediatrica, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| |
Collapse
|
14
|
Stone D, Kenkel EJ, Loprieno MA, Tanaka M, De Silva Feelixge HS, Kumar AJ, Stensland L, Obenza WM, Wangari S, Ahrens CY, Murnane RD, Peterson CW, Kiem HP, Huang ML, Aubert M, Hu SL, Jerome KR. Gene Transfer in Adeno-Associated Virus Seropositive Rhesus Macaques Following Rapamycin Treatment and Subcutaneous Delivery of AAV6, but Not Retargeted AAV6 Vectors. Hum Gene Ther 2020; 32:96-112. [PMID: 32998579 DOI: 10.1089/hum.2020.113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Adeno-associated virus (AAV) vectors such as AAV6, which shows tropism for primary human CD4+ T cells in vitro, are being explored for delivery of anti-HIV therapeutic modalities in vivo. However, pre-existing immunity and sequestration in nontarget organs can significantly hinder their performance. To overcome these challenges, we investigated whether immunosuppression would allow gene delivery by AAV6 or targeted AAV6 derivatives in seropositive rhesus macaques. Animals were immune suppressed with rapamycin before intravenous (IV) or subcutaneous (SC) delivery of AAV, and we monitored vector biodistribution, gene transfer, and safety. Macaques received phosphate-buffered saline, AAV6 alone, or an equal dose of AAV6 and an AAV6-55.2 vector retargeted to CD4 through a direct ankyrin repeat protein (DARPin). AAV6 and AAV6-55.2 vector genomes were found in peripheral blood mononuclear cells and most organs up to 28 days postadministration, with the highest levels seen in liver, spleen, lymph nodes (LNs), and muscle, suggesting that retargeting did not prevent vector sequestration. Despite vector genome detection, gene expression from AAV6-55.2 was not detected in any tissue. SC injection of AAV6 facilitated efficient gene expression in muscle adjacent to the injection site, plus low-level gene expression in spleen, LNs, and liver, whereas gene expression following IV injection of AAV6 was predominantly seen in the spleen. AAV vectors were well tolerated, although elevated liver enzymes were detected in three of four AAV-treated animals 14 days after rapamycin withdrawal. One SC-injected animal had muscle inflammation proximal to the injection site, plus detectable T cell responses against transgene and AAV6 capsid at study finish. Overall, our data suggest that rapamycin treatment may offer a possible strategy to express anti-HIV therapeutics such as broadly neutralizing antibodies from muscle. This study provides important safety and efficacy data that will aid study design for future anti-HIV gene therapies.
Collapse
Affiliation(s)
- Daniel Stone
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Elizabeth J Kenkel
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Michelle A Loprieno
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Motoko Tanaka
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Arjun J Kumar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Laurence Stensland
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Willimark M Obenza
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Solomon Wangari
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Chul Y Ahrens
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Robert D Murnane
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Christopher W Peterson
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Hans-Peter Kiem
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Meei-Li Huang
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Martine Aubert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Shiu-Lok Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.,Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Keith R Jerome
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
15
|
Honaker Y, Hubbard N, Xiang Y, Fisher L, Hagin D, Sommer K, Song Y, Yang SJ, Lopez C, Tappen T, Dam EM, Khan I, Hale M, Buckner JH, Scharenberg AM, Torgerson TR, Rawlings DJ. Gene editing to induce FOXP3 expression in human CD4+ T cells leads to a stable regulatory phenotype and function. Sci Transl Med 2020; 12:12/546/eaay6422. [DOI: 10.1126/scitranslmed.aay6422] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 12/09/2019] [Accepted: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Thymic regulatory T cells (tTregs) are potent inhibitors of autoreactive immune responses, and loss of tTreg function results in fatal autoimmune disease. Defects in tTreg number or function are also implicated in multiple autoimmune diseases, leading to growing interest in use of Treg as cell therapies to establish immune tolerance. Because tTregs are present at low numbers in circulating blood and may be challenging to purify and expand and also inherently defective in some subjects, we designed an alternative strategy to create autologous Treg-like cells from bulk CD4+ T cells. We used homology-directed repair (HDR)–based gene editing to enforce expression of FOXP3, the master transcription factor for tTreg. Targeted insertion of a robust enhancer/promoter proximal to the first coding exon bypassed epigenetic silencing, permitting stable and robust expression of endogenous FOXP3. HDR-edited T cells, edTregs, manifested a transcriptional program leading to sustained expression of canonical markers and suppressive activity of tTreg. Both human and murine edTregs mediated immunosuppression in vivo in models of inflammatory disease. Further, this engineering strategy permitted generation of antigen-specific edTreg with robust in vitro and in vivo functional activity. Last, edTreg could be enriched and expanded at scale using clinically relevant methods. Together, these findings suggest that edTreg production may permit broad future clinical application.
Collapse
Affiliation(s)
- Yuchi Honaker
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Nicholas Hubbard
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Yufei Xiang
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Logan Fisher
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - David Hagin
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Karen Sommer
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Yumei Song
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | | | - Christina Lopez
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Tori Tappen
- Benaroya Research Institute, Seattle, WA 98101, USA
| | | | - Iram Khan
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Malika Hale
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Jane H. Buckner
- Benaroya Research Institute, Seattle, WA 98101, USA
- Department of Medicine, University of Washington, Seattle, WA 98101, USA
- Department of Immunology, University of Washington, Seattle, WA 98101, USA
| | - Andrew M. Scharenberg
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Immunology, University of Washington, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
| | - Troy R. Torgerson
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Immunology, University of Washington, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
| | - David J. Rawlings
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Immunology, University of Washington, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
| |
Collapse
|
16
|
Young PA, Yamada RE, Trinh KR, Vasuthasawat A, De Oliveira S, Yamada DH, Morrison SL, Timmerman JM. Activity of Anti-CD19 Chimeric Antigen Receptor T Cells Against B Cell Lymphoma Is Enhanced by Antibody-Targeted Interferon-Alpha. J Interferon Cytokine Res 2019; 38:239-254. [PMID: 29920129 DOI: 10.1089/jir.2018.0030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
An important emerging form of immunotherapy targeting B cell malignancies is chimeric antigen receptor (CAR) T cell therapy. Despite encouraging response rates of anti-CD19 CAR T cell therapy in B cell lymphomas, limited durability of response necessitates further study to potentiate CAR T cell efficacy. Antibody-targeted interferon (IFN) therapy is a novel approach in immunotherapy. Given the ability of IFNs to promote T cell activation and survival, target cell recognition, and cytotoxicity, we asked whether antibody-targeted IFN could enhance the antitumor effects of anti-CD19 CAR T cells. We produced an anti-CD20-IFN fusion protein containing the potent type 1 IFN isoform alpha14 (α14), and demonstrated its ability to suppress proliferation and induce apoptosis of human B cell lymphomas. Indeed, with the combination of anti-CD20-hIFNα14 and CAR T cells, we found enhanced cell killing among B cell lymphoma lines. Importantly, for all cell lines pretreated with anti-CD20-hIFNα14, the subsequent cytokine production by CAR T cells was markedly increased regardless of the degree of cell killing. Thus, several activities of CD19 CAR T cells were enhanced in the presence of anti-CD20-hIFNα14. These data suggest that antibody-targeted IFN may be an important novel approach to improving the efficacy of CAR T cell therapy.
Collapse
Affiliation(s)
- Patricia A Young
- 1 Division of Hematology & Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Reiko E Yamada
- 1 Division of Hematology & Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Kham R Trinh
- 2 Department of Microbiology, Immunology, and Molecular Genetics, University of California , Los Angeles, Los Angeles, California
| | - Alex Vasuthasawat
- 2 Department of Microbiology, Immunology, and Molecular Genetics, University of California , Los Angeles, Los Angeles, California
| | - Satiro De Oliveira
- 3 Division of Pediatric Hematology & Oncology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, California
| | - Douglas H Yamada
- 2 Department of Microbiology, Immunology, and Molecular Genetics, University of California , Los Angeles, Los Angeles, California
| | - Sherie L Morrison
- 2 Department of Microbiology, Immunology, and Molecular Genetics, University of California , Los Angeles, Los Angeles, California
| | - John M Timmerman
- 1 Division of Hematology & Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
17
|
Espinoza DA, Fan X, Yang D, Cordes SF, Truitt LL, Calvo KR, Yabe IM, Demirci S, Hope KJ, Hong SG, Krouse A, Metzger M, Bonifacino A, Lu R, Uchida N, Tisdale JF, Wu X, DeRavin SS, Malech HL, Donahue RE, Wu C, Dunbar CE. Aberrant Clonal Hematopoiesis following Lentiviral Vector Transduction of HSPCs in a Rhesus Macaque. Mol Ther 2019; 27:1074-1086. [PMID: 31023523 PMCID: PMC6554657 DOI: 10.1016/j.ymthe.2019.04.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 01/21/2023] Open
Abstract
Lentiviral vectors (LVs) are used for delivery of genes into hematopoietic stem and progenitor cells (HSPCs) in clinical trials worldwide. LVs, in contrast to retroviral vectors, are not associated with insertion site-associated malignant clonal expansions and, thus, are considered safer. Here, however, we present a case of markedly abnormal dysplastic clonal hematopoiesis affecting the erythroid, myeloid, and megakaryocytic lineages in a rhesus macaque transplanted with HSPCs that were transduced with a LV containing a strong retroviral murine stem cell virus (MSCV) constitutive promoter-enhancer in the LTR. Nine insertions were mapped in the abnormal clone, resulting in overexpression and aberrant splicing of several genes of interest, including the cytokine stem cell factor and the transcription factor PLAG1. This case represents the first clear link between lentiviral insertion-induced clonal expansion and a clinically abnormal transformed phenotype following transduction of normal primate or human HSPCs, which is concerning, and suggests that strong constitutive promoters should not be included in LVs.
Collapse
Affiliation(s)
- Diego A Espinoza
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xing Fan
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Di Yang
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Stefan F Cordes
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Lauren L Truitt
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Katherine R Calvo
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Idalia M Yabe
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Selami Demirci
- Sickle Cell and Vascular Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Kristin J Hope
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - So Gun Hong
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Allen Krouse
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Mark Metzger
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Aylin Bonifacino
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Rong Lu
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Naoya Uchida
- Sickle Cell and Vascular Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - John F Tisdale
- Sickle Cell and Vascular Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Xiaolin Wu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Suk See DeRavin
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Harry L Malech
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Robert E Donahue
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Chuanfeng Wu
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA.
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
18
|
Friedman KM, Garrett TE, Evans JW, Horton HM, Latimer HJ, Seidel SL, Horvath CJ, Morgan RA. Effective Targeting of Multiple B-Cell Maturation Antigen-Expressing Hematological Malignances by Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor T Cells. Hum Gene Ther 2019; 29:585-601. [PMID: 29641319 DOI: 10.1089/hum.2018.001] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
B-cell maturation antigen (BCMA) expression has been proposed as a marker for the identification of malignant plasma cells in patients with multiple myeloma (MM). Nearly all MM tumor cells express BCMA, while normal tissue expression is restricted to plasma cells and a subset of mature B cells. Consistent BCMA expression was confirmed on MM biopsies (29/29 BCMA+), and it was further demonstrated that BCMA is expressed in a substantial number of lymphoma samples, as well as primary chronic lymphocytic leukemia B cells. To target BCMA using redirected autologous T cells, lentiviral vectors (LVV) encoding chimeric antigen receptors (CARs) were constructed with four unique anti-BCMA single-chain variable fragments, fused to the CD137 (4-1BB) co-stimulatory and CD3ζ signaling domains. One LVV, BB2121, was studied in detail, and BB2121 CAR-transduced T cells (bb2121) exhibited a high frequency of CAR + T cells and robust in vitro activity against MM cell lines, lymphoma cell lines, and primary chronic lymphocytic leukemia peripheral blood. Based on receptor quantification, bb2121 recognized tumor cells expressing as little as 222 BCMA molecules per cell. The in vivo pharmacology of anti-BCMA CAR T cells was studied in NSG mouse models of human MM, Burkitt lymphoma, and mantle cell lymphoma, where mice received a single intravenous administration of vehicle, control vector-transduced T cells, or anti-BCMA CAR-transduced T cells. In all models, the vehicle and control CAR T cells failed to inhibit tumor growth. In contrast, treatment with bb2121 resulted in rapid and sustained elimination of the tumors and 100% survival in all treatment models. Together, these data support the further development of anti-BCMA CAR T cells as a potential treatment for not only MM but also some lymphomas.
Collapse
|
19
|
Puig-Saus C, Parisi G, Garcia-Diaz A, Krystofinski PE, Sandoval S, Zhang R, Champhekar AS, McCabe J, Cheung-Lau GC, Truong NA, Vega-Crespo A, Komenan MDS, Pang J, Macabali MH, Saco JD, Goodwin JL, Bolon B, Seet CS, Montel-Hagen A, Crooks GM, Hollis RP, Campo-Fernandez B, Bischof D, Cornetta K, Gschweng EH, Adelson C, Nguyen A, Yang L, Witte ON, Baltimore D, Comin-Anduix B, Kohn DB, Wang X, Cabrera P, Kaplan-Lefko PJ, Berent-Maoz B, Ribas A. IND-Enabling Studies for a Clinical Trial to Genetically Program a Persistent Cancer-Targeted Immune System. Clin Cancer Res 2018; 25:1000-1011. [PMID: 30409823 DOI: 10.1158/1078-0432.ccr-18-0963] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/21/2018] [Accepted: 11/05/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE To improve persistence of adoptively transferred T-cell receptor (TCR)-engineered T cells and durable clinical responses, we designed a clinical trial to transplant genetically-modified hematopoietic stem cells (HSCs) together with adoptive cell transfer of T cells both engineered to express an NY-ESO-1 TCR. Here, we report the preclinical studies performed to enable an investigational new drug (IND) application. EXPERIMENTAL DESIGN HSCs transduced with a lentiviral vector expressing NY-ESO-1 TCR and the PET reporter/suicide gene HSV1-sr39TK and T cells transduced with a retroviral vector expressing NY-ESO-1 TCR were coadministered to myelodepleted HLA-A2/Kb mice within a formal Good Laboratory Practice (GLP)-compliant study to demonstrate safety, persistence, and HSC differentiation into all blood lineages. Non-GLP experiments included assessment of transgene immunogenicity and in vitro viral insertion safety studies. Furthermore, Good Manufacturing Practice (GMP)-compliant cell production qualification runs were performed to establish the manufacturing protocols for clinical use. RESULTS TCR genetically modified and ex vivo-cultured HSCs differentiated into all blood subsets in vivo after HSC transplantation, and coadministration of TCR-transduced T cells did not result in increased toxicity. The expression of NY-ESO-1 TCR and sr39TK transgenes did not have a detrimental effect on gene-modified HSC's differentiation to all blood cell lineages. There was no evidence of genotoxicity induced by the lentiviral vector. GMP batches of clinical-grade transgenic cells produced during qualification runs had adequate stability and functionality. CONCLUSIONS Coadministration of HSCs and T cells expressing an NY-ESO-1 TCR is safe in preclinical models. The results presented in this article led to the FDA approval of IND 17471.
Collapse
Affiliation(s)
- Cristina Puig-Saus
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California. .,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Giulia Parisi
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Angel Garcia-Diaz
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Paige E Krystofinski
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Salemiz Sandoval
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Ruixue Zhang
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Ameya S Champhekar
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - James McCabe
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Gardenia C Cheung-Lau
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Nhat A Truong
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Agustin Vega-Crespo
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Marie Desiles S Komenan
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Jia Pang
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Mignonette H Macabali
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Justin D Saco
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Jeffrey L Goodwin
- Division of Laboratory Animal Medicine (DLAM), Department of Medicine, DGSOM, UCLA, Los Angeles, California
| | | | - Christopher S Seet
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California
| | - Amelie Montel-Hagen
- Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California
| | - Gay M Crooks
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.,Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, DGSOM, UCLA, Los Angeles, California.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California
| | - Roger P Hollis
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California
| | - Beatriz Campo-Fernandez
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California
| | - Daniela Bischof
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indiana University Vector Production Facility, Indianapolis, Indiana
| | - Kenneth Cornetta
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indiana University Vector Production Facility, Indianapolis, Indiana
| | - Eric H Gschweng
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California
| | - Celia Adelson
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California
| | - Alexander Nguyen
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Lili Yang
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California.,Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California
| | - Owen N Witte
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California.,Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
| | - David Baltimore
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| | - Begonya Comin-Anduix
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.,Division of Surgical Oncology, Department of Surgery, UCLA, Los Angeles, California
| | - Donald B Kohn
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California.,Department of Pediatrics, UCLA Children's Discovery and Innovation Institute, DGSOM, University of California, Los Angeles, California
| | - Xiaoyan Wang
- Statistics Core, Department of Medicine, UCLA, Los Angeles, California
| | - Paula Cabrera
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Paula J Kaplan-Lefko
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Beata Berent-Maoz
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Antoni Ribas
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California. .,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,Division of Surgical Oncology, Department of Surgery, UCLA, Los Angeles, California
| |
Collapse
|
20
|
Hung KL, Meitlis I, Hale M, Chen CY, Singh S, Jackson SW, Miao CH, Khan IF, Rawlings DJ, James RG. Engineering Protein-Secreting Plasma Cells by Homology-Directed Repair in Primary Human B Cells. Mol Ther 2018; 26:456-467. [PMID: 29273498 PMCID: PMC5835153 DOI: 10.1016/j.ymthe.2017.11.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/09/2017] [Accepted: 11/11/2017] [Indexed: 12/15/2022] Open
Abstract
The ability to engineer primary human B cells to differentiate into long-lived plasma cells and secrete a de novo protein may allow the creation of novel plasma cell therapies for protein deficiency diseases and other clinical applications. We initially developed methods for efficient genome editing of primary B cells isolated from peripheral blood. By delivering CRISPR/CRISPR-associated protein 9 (Cas9) ribonucleoprotein (RNP) complexes under conditions of rapid B cell expansion, we achieved site-specific gene disruption at multiple loci in primary human B cells (with editing rates of up to 94%). We used this method to alter ex vivo plasma cell differentiation by disrupting developmental regulatory genes. Next, we co-delivered RNPs with either a single-stranded DNA oligonucleotide or adeno-associated viruses containing homologous repair templates. Using either delivery method, we achieved targeted sequence integration at high efficiency (up to 40%) via homology-directed repair. This method enabled us to engineer plasma cells to secrete factor IX (FIX) or B cell activating factor (BAFF) at high levels. Finally, we show that introduction of BAFF into plasma cells promotes their engraftment into immunodeficient mice. Our results highlight the utility of genome editing in studying human B cell biology and demonstrate a novel strategy for modifying human plasma cells to secrete therapeutic proteins.
Collapse
Affiliation(s)
- King L Hung
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Iana Meitlis
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Malika Hale
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Chun-Yu Chen
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Swati Singh
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Shaun W Jackson
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Carol H Miao
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Iram F Khan
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA.
| | - Richard G James
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
21
|
Šenigl F, Miklík D, Auxt M, Hejnar J. Accumulation of long-term transcriptionally active integrated retroviral vectors in active promoters and enhancers. Nucleic Acids Res 2018; 45:12752-12765. [PMID: 29244184 PMCID: PMC5727404 DOI: 10.1093/nar/gkx889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/26/2017] [Indexed: 01/13/2023] Open
Abstract
Most retroviruses preferentially integrate into certain genomic locations and, as a result, their genome-wide integration patterns are non-random. We investigate the epigenetic landscape of integrated retroviral vectors and correlate it with the long-term stability of proviral transcription. Retroviral vectors derived from the avian sarcoma/leukosis virus expressing the GFP reporter were used to transduce the human myeloid lymphoblastoma cell line K562. Because of efficient silencing of avian retrovirus in mammalian cells, only ∼3% of established clones displayed stable proviral expression. We analyzed the vector integration sites in non-selected cells and in clones selected for the GFP expression. This selection led to overrepresentation of proviruses integrated in active transcription units, with particular accumulation in promoter-proximal areas. In parallel, we investigated the integration of vectors equipped with an anti-silencing CpG island core sequence. Such modification increased the frequency of stably expressing proviruses by one order. The modified vectors are also overrepresented in active transcription units, but stably expressed in distal parts of transcriptional units further away from promoters with marked accumulation in enhancers. These results suggest that integrated retroviruses subject to gradual epigenetic silencing during long-term cultivation. Among most genomic compartments, however, active promoters and enhancers protect the adjacent retroviruses from transcriptional silencing.
Collapse
Affiliation(s)
- Filip Šenigl
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, CZ-14220 Prague 4, Czech Republic
| | - Dalibor Miklík
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, CZ-14220 Prague 4, Czech Republic
| | - Miroslav Auxt
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, CZ-14220 Prague 4, Czech Republic
| | - Jirí Hejnar
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, CZ-14220 Prague 4, Czech Republic
| |
Collapse
|
22
|
Keriquel V, Oliveira H, Rémy M, Ziane S, Delmond S, Rousseau B, Rey S, Catros S, Amédée J, Guillemot F, Fricain JC. In situ printing of mesenchymal stromal cells, by laser-assisted bioprinting, for in vivo bone regeneration applications. Sci Rep 2017; 7:1778. [PMID: 28496103 PMCID: PMC5431768 DOI: 10.1038/s41598-017-01914-x] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023] Open
Abstract
Bioprinting has emerged as a novel technological approach with the potential to address unsolved questions in the field of tissue engineering. We have recently shown that Laser Assisted Bioprinting (LAB), due to its unprecedented cell printing resolution and precision, is an attractive tool for the in situ printing of a bone substitute. Here, we show that LAB can be used for the in situ printing of mesenchymal stromal cells, associated with collagen and nano-hydroxyapatite, in order to favor bone regeneration, in a calvaria defect model in mice. Also, by testing different cell printing geometries, we show that different cellular arrangements impact on bone tissue regeneration. This work opens new avenues on the development of novel strategies, using in situ bioprinting, for the building of tissues, from the ground up.
Collapse
Affiliation(s)
- Virginie Keriquel
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Hugo Oliveira
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France. .,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France. .,ART BioPrint, Inserm, U1026, F-33076, Bordeaux, France.
| | - Murielle Rémy
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Sophia Ziane
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Samantha Delmond
- Clinical Research Center - Technological Innovation, Inserm, Bordeaux University Hospital, Pessac, 33600, France
| | - Benoit Rousseau
- Animalerie A2, Université Bordeaux Segalen, Bordeaux, France
| | - Sylvie Rey
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Sylvain Catros
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,CHU Bordeaux, Services d'Odontologie et de Santé Buccale, F-33076, Bordeaux, France
| | - Joelle Amédée
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Fabien Guillemot
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France
| | - Jean-Christophe Fricain
- University of Bordeaux, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,Inserm, Tissue Bioengineering, U1026, F-33076, Bordeaux, France.,CHU Bordeaux, Services d'Odontologie et de Santé Buccale, F-33076, Bordeaux, France
| |
Collapse
|
23
|
Shaw KL, Garabedian E, Mishra S, Barman P, Davila A, Carbonaro D, Shupien S, Silvin C, Geiger S, Nowicki B, Smogorzewska EM, Brown B, Wang X, de Oliveira S, Choi Y, Ikeda A, Terrazas D, Fu PY, Yu A, Fernandez BC, Cooper AR, Engel B, Podsakoff G, Balamurugan A, Anderson S, Muul L, Jagadeesh GJ, Kapoor N, Tse J, Moore TB, Purdy K, Rishi R, Mohan K, Skoda-Smith S, Buchbinder D, Abraham RS, Scharenberg A, Yang OO, Cornetta K, Gjertson D, Hershfield M, Sokolic R, Candotti F, Kohn DB. Clinical efficacy of gene-modified stem cells in adenosine deaminase-deficient immunodeficiency. J Clin Invest 2017; 127:1689-1699. [PMID: 28346229 DOI: 10.1172/jci90367] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/24/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Autologous hematopoietic stem cell transplantation (HSCT) of gene-modified cells is an alternative to enzyme replacement therapy (ERT) and allogeneic HSCT that has shown clinical benefit for adenosine deaminase-deficient (ADA-deficient) SCID when combined with reduced intensity conditioning (RIC) and ERT cessation. Clinical safety and therapeutic efficacy were evaluated in a phase II study. METHODS Ten subjects with confirmed ADA-deficient SCID and no available matched sibling or family donor were enrolled between 2009 and 2012 and received transplantation with autologous hematopoietic CD34+ cells that were modified with the human ADA cDNA (MND-ADA) γ-retroviral vector after conditioning with busulfan (90 mg/m2) and ERT cessation. Subjects were followed from 33 to 84 months at the time of data analysis. Safety of the procedure was assessed by recording the number of adverse events. Efficacy was assessed by measuring engraftment of gene-modified hematopoietic stem/progenitor cells, ADA gene expression, and immune reconstitution. RESULTS With the exception of the oldest subject (15 years old at enrollment), all subjects remained off ERT with normalized peripheral blood mononuclear cell (PBMC) ADA activity, improved lymphocyte numbers, and normal proliferative responses to mitogens. Three of nine subjects were able to discontinue intravenous immunoglobulin replacement therapy. The MND-ADA vector was persistently detected in PBMCs (vector copy number [VCN] = 0.1-2.6) and granulocytes (VCN = 0.01-0.3) through the most recent visits at the time of this writing. No patient has developed a leukoproliferative disorder or other vector-related clinical complication since transplant. CONCLUSION These results demonstrate clinical therapeutic efficacy from gene therapy for ADA-deficient SCID, with an excellent clinical safety profile. TRIAL REGISTRATION ClinicalTrials.gov NCT00794508. FUNDING Food and Drug Administration Office of Orphan Product Development award, RO1 FD003005; NHLBI awards, PO1 HL73104 and Z01 HG000122; UCLA Clinical and Translational Science Institute awards, UL1RR033176 and UL1TR000124.
Collapse
|
24
|
Hale M, Mesojednik T, Romano Ibarra GS, Sahni J, Bernard A, Sommer K, Scharenberg AM, Rawlings DJ, Wagner TA. Engineering HIV-Resistant, Anti-HIV Chimeric Antigen Receptor T Cells. Mol Ther 2017; 25:570-579. [PMID: 28143740 DOI: 10.1016/j.ymthe.2016.12.023] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/27/2016] [Accepted: 12/27/2016] [Indexed: 12/28/2022] Open
Abstract
The treatment or cure of HIV infection by cell and gene therapy has been a goal for decades. Recent advances in both gene editing and chimeric antigen receptor (CAR) technology have created new therapeutic possibilities for a variety of diseases. Broadly neutralizing monoclonal antibodies (bNAbs) with specificity for the HIV envelope glycoprotein provide a promising means of targeting HIV-infected cells. Here we show that primary human T cells engineered to express anti-HIV CARs based on bNAbs (HIVCAR) show specific activation and killing of HIV-infected versus uninfected cells in the absence of HIV replication. We also show that homology-directed recombination of the HIVCAR gene expression cassette into the CCR5 locus enhances suppression of replicating virus compared with HIVCAR expression alone. This work demonstrates that HIV immunotherapy utilizing potent bNAb-based single-chain variable fragments fused to second-generation CAR signaling domains, delivered directly into the CCR5 locus of T cells by homology-directed gene editing, is feasible and effective. This strategy has the potential to target HIV-infected cells in HIV-infected individuals, which might help in the effort to cure HIV.
Collapse
Affiliation(s)
- Malika Hale
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Taylor Mesojednik
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Guillermo S Romano Ibarra
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Jaya Sahni
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Alison Bernard
- Center for Global Infectious Disease Research, 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
| | - Andrew M Scharenberg
- 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 98101, USA; Department of Immunology, University of Washington, Seattle, WA 98101, 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 98101, USA; Department of Immunology, University of Washington, Seattle, WA 98101, USA.
| | - Thor A Wagner
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98101, USA.
| |
Collapse
|
25
|
Hale M, Lee B, Honaker Y, Leung WH, Grier AE, Jacobs HM, Sommer K, Sahni J, Jackson SW, Scharenberg AM, Astrakhan A, Rawlings DJ. Homology-Directed Recombination for Enhanced Engineering of Chimeric Antigen Receptor T Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 4:192-203. [PMID: 28345004 PMCID: PMC5363294 DOI: 10.1016/j.omtm.2016.12.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/28/2016] [Indexed: 12/17/2022]
Abstract
Gene editing by homology-directed recombination (HDR) can be used to couple delivery of a therapeutic gene cassette with targeted genomic modifications to generate engineered human T cells with clinically useful profiles. Here, we explore the functionality of therapeutic cassettes delivered by these means and test the flexibility of this approach to clinically relevant alleles. Because CCR5-negative T cells are resistant to HIV-1 infection, CCR5-negative anti-CD19 chimeric antigen receptor (CAR) T cells could be used to treat patients with HIV-associated B cell malignancies. We show that targeted delivery of an anti-CD19 CAR cassette to the CCR5 locus using a recombinant AAV homology template and an engineered megaTAL nuclease results in T cells that are functionally equivalent, in both in vitro and in vivo tumor models, to CAR T cells generated by random integration using lentiviral delivery. With the goal of developing off-the-shelf CAR T cell therapies, we next targeted CARs to the T cell receptor alpha constant (TRAC) locus by HDR, producing TCR-negative anti-CD19 CAR and anti-B cell maturation antigen (BCMA) CAR T cells. These novel cell products exhibited in vitro cytolytic activity against both tumor cell lines and primary cell targets. Our combined results indicate that high-efficiency HDR delivery of therapeutic genes may provide a flexible and robust method that can extend the clinical utility of cell therapeutics.
Collapse
Affiliation(s)
- Malika Hale
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Yuchi Honaker
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Alexandra E Grier
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Immunology, University of Washington, Seattle, WA 98101, USA
| | - Holly M Jacobs
- 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; Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Jaya Sahni
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Shaun W Jackson
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
| | - Andrew M Scharenberg
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98101, USA; Department of Immunology, University of Washington, Seattle, WA 98101, USA
| | | | - David J Rawlings
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98101, USA; Department of Immunology, University of Washington, Seattle, WA 98101, USA
| |
Collapse
|
26
|
Larson SM, Truscott LC, Chiou TT, Patel A, Kao R, Tu A, Tyagi T, Lu X, Elashoff D, De Oliveira SN. Pre-clinical development of gene modification of haematopoietic stem cells with chimeric antigen receptors for cancer immunotherapy. Hum Vaccin Immunother 2017; 13:1094-1104. [PMID: 28059624 DOI: 10.1080/21645515.2016.1268745] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Patients with refractory or recurrent B-lineage hematologic malignancies have less than 50% of chance of cure despite intensive therapy and innovative approaches are needed. We hypothesize that gene modification of haematopoietic stem cells (HSC) with an anti-CD19 chimeric antigen receptor (CAR) will produce a multi-lineage, persistent immunotherapy against B-lineage malignancies that can be controlled by the HSVsr39TK suicide gene. High-titer third-generation self-inactivating lentiviral constructs were developed to deliver a second-generation CD19-specific CAR and the herpes simplex virus thymidine kinase HSVsr39TK to provide a suicide gene to allow ablation of gene-modified cells if necessary. Human HSC were transduced with such lentiviral vectors and evaluated for function of both CAR and HSVsr39TK. Satisfactory transduction efficiency was achieved; the addition of the suicide gene did not impair CAR expression or antigen-specific cytotoxicity, and determined marked cytotoxicity to ganciclovir. NSG mice transplanted with gene-modified human HSC showed CAR expression not significantly different between transduced cells with or without HSVsr39TK, and expression of anti-CD19 CAR conferred anti-tumor survival advantage. Treatment with ganciclovir led to significant ablation of gene-modified cells in mouse tissues. Haematopoietic stem cell transplantation is frequently part of the standard of care for patients with relapsed and refractory B cell malignancies; following HSC collection, a portion of the cells could be modified to express the CD19-specific CAR and give rise to a persistent, multi-cell lineage, HLA-independent immunotherapy, enhancing the graft-versus-malignancy activity.
Collapse
Affiliation(s)
- Sarah M Larson
- a Department of Internal Medicine , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Laurel C Truscott
- b Department of Pediatrics , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Tzu-Ting Chiou
- b Department of Pediatrics , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Amie Patel
- c Western University of Health Sciences , Pomona , CA , USA
| | - Roy Kao
- b Department of Pediatrics , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Andy Tu
- b Department of Pediatrics , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Tulika Tyagi
- b Department of Pediatrics , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Xiang Lu
- a Department of Internal Medicine , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA.,d Clinical Translational Science Institute (CTSI), David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - David Elashoff
- a Department of Internal Medicine , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA.,d Clinical Translational Science Institute (CTSI), David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Satiro N De Oliveira
- b Department of Pediatrics , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| |
Collapse
|
27
|
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
|
28
|
Tiwari S, Hontz A, Terrell CE, Arumugam P, Carmo M, Risma K, Jordan M, Malik P. High Level of Perforin Expression Is Required for Effective Correction of Hemophagocytic Lymphohistiocytosis. Hum Gene Ther 2016; 27:847-859. [PMID: 27471778 PMCID: PMC5749578 DOI: 10.1089/hum.2016.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/26/2016] [Indexed: 02/06/2023] Open
Abstract
Perforin-1 mutations result in a potentially fatal hemophagocytic lymphohistiocytosis (HLH) with heightened immune activation, hypercytokinemia, pancytopenia, and end-organ damage. At present, hematopoietic stem cell (HSC) transplantation is curative, but limited by donor availability and associated mortality, making gene therapy an attractive alternative approach for HLH. We reported that perforin expression driven by cellular promoters in lentiviral (LV) vectors resulted in significant, albeit partial, correction of the inflammatory features in a murine model of HLH. We hypothesized that the level of perforin expression achieved per cell from ectopic moderate-strength cellular promoters (phosphoglycerate kinase gene/perforin-1 gene) is inadequate and thus engineered an LV vector using a viral promoter (MND; a modified Moloney murine leukemia virus long terminal repeat with myeloproliferative sarcoma virus enhancer) containing microRNA126 target sequences to restrict perforin expression in HSCs. We show here that the MND-LV vector restored perforin expression to normal levels in a perforin-deficient human natural killer cell line and perforin gene-corrected Perforin1-/- transplant recipients, whereas cellular promoters drove only partial correction. On lymphocytic choriomeningitis virus challenge, the clinical scores and survival improved only with the MND-LV vector, but inflammatory markers and cytotoxicity were improved with all LV vectors. Our studies suggest that although moderate levels of expression can result in partial amelioration of the HLH phenotype, high levels of perforin expression per cell are required for complete correction of HLH.
Collapse
Affiliation(s)
- Swati Tiwari
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, Ohio
- Division of Experimental Hematology and Oncology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio
| | - Adrianne Hontz
- Division of Allergy/Immunology, Department of Pediatrics, CCHMC, Cincinnati, Ohio
| | - Catherine E. Terrell
- Division of Experimental Hematology and Oncology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio
| | - Paritha Arumugam
- Division of Experimental Hematology and Oncology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio
| | - Marlene Carmo
- Infection, Immunity, Inflammation, and Physiological Medicine Programme, Molecular and Cellular Immunology Section, UCL Institute of Child Health, London, United Kingdom
| | - Kimberly Risma
- Division of Allergy/Immunology, Department of Pediatrics, CCHMC, Cincinnati, Ohio
| | - Michael Jordan
- Division of Immunobiology, CCHMC, Cincinnati, Ohio
- Department of Bone Marrow Transplantation and Immune Deficiency, CCHMC, Cincinnati, Ohio
| | - Punam Malik
- Division of Experimental Hematology and Oncology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio
| |
Collapse
|
29
|
Sather BD, Romano Ibarra GS, Sommer K, Curinga G, Hale M, Khan IF, Singh S, Song Y, Gwiazda K, Sahni J, Jarjour J, Astrakhan A, Wagner TA, Scharenberg AM, Rawlings DJ. Efficient modification of CCR5 in primary human hematopoietic cells using a megaTAL nuclease and AAV donor template. Sci Transl Med 2016; 7:307ra156. [PMID: 26424571 DOI: 10.1126/scitranslmed.aac5530] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Genetic mutations or engineered nucleases that disrupt the HIV co-receptor CCR5 block HIV infection of CD4(+) T cells. These findings have motivated the engineering of CCR5-specific nucleases for application as HIV therapies. The efficacy of this approach relies on efficient biallelic disruption of CCR5, and the ability to efficiently target sequences that confer HIV resistance to the CCR5 locus has the potential to further improve clinical outcomes. We used RNA-based nuclease expression paired with adeno-associated virus (AAV)-mediated delivery of a CCR5-targeting donor template to achieve highly efficient targeted recombination in primary human T cells. This method consistently achieved 8 to 60% rates of homology-directed recombination into the CCR5 locus in T cells, with over 80% of cells modified with an MND-GFP expression cassette exhibiting biallelic modification. MND-GFP-modified T cells maintained a diverse repertoire and engrafted in immune-deficient mice as efficiently as unmodified cells. Using this method, we integrated sequences coding chimeric antigen receptors (CARs) into the CCR5 locus, and the resulting targeted CAR T cells exhibited antitumor or anti-HIV activity. Alternatively, we introduced the C46 HIV fusion inhibitor, generating T cell populations with high rates of biallelic CCR5 disruption paired with potential protection from HIV with CXCR4 co-receptor tropism. Finally, this protocol was applied to adult human mobilized CD34(+) cells, resulting in 15 to 20% homologous gene targeting. Our results demonstrate that high-efficiency targeted integration is feasible in primary human hematopoietic cells and highlight the potential of gene editing to engineer T cell products with myriad functional properties.
Collapse
Affiliation(s)
- Blythe D Sather
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Guillermo S Romano Ibarra
- 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
| | - Gabrielle Curinga
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Malika Hale
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Iram F Khan
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Swati Singh
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Yumei Song
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Kamila Gwiazda
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Jaya Sahni
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | | | - Thor A Wagner
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98101, USA. Department of Pediatrics, University of Washington, Seattle, WA 98101, USA
| | - Andrew M Scharenberg
- 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 98101, USA. Department of Immunology, University of Washington, Seattle, WA 98101, 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 98101, USA. Department of Immunology, University of Washington, Seattle, WA 98101, USA
| |
Collapse
|
30
|
Zhou S, Fatima S, Ma Z, Wang YD, Lu T, Janke LJ, Du Y, Sorrentino BP. Evaluating the Safety of Retroviral Vectors Based on Insertional Oncogene Activation and Blocked Differentiation in Cultured Thymocytes. Mol Ther 2016; 24:1090-1099. [PMID: 26957223 PMCID: PMC4923324 DOI: 10.1038/mt.2016.55] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/24/2016] [Indexed: 12/20/2022] Open
Abstract
Insertional oncogenesis due to retroviral (RV) vector integration has caused recurrent leukemia in multiple gene therapy trials, predominantly due to vector integration effects at the LMO2 locus. While currently available preclinical safety models have been used for evaluating vector safety, none have predicted or reproduced the recurrent LMO2 integrations seen in previous X-linked severe combined immunodeficiency (X-SCID) and Wiskott-Aldrich clinical gene therapy trials. We now describe a new assay for assessing vector safety that recapitulates naturally occurring insertions into Lmo2 and other T-cell proto-oncogenes leading to a preleukemic developmental arrest in primary murine thymocytes cultured in vitro. This assay was used to compare the relative oncogenic potential of a variety of gamma-RV and lentiviral vectors and to assess the risk conferred by various transcriptional elements contained in these genomes. Gamma-RV vectors that contained full viral long-terminal repeats were most prone to causing double negative 2 (DN2) arrest and led to repeated cases of Lmo2 pathway activation, while lentiviral vectors containing these same elements were significantly less prone to activate proto-oncogenes or cause DN2 arrest. This work provides a new preclinical assay that is especially relevant for assessing safety in SCID disorders and provides a new tool for designing safer RV vectors.
Collapse
Affiliation(s)
- Sheng Zhou
- Division of Experimental Hematology, Department of Hematology, Memphis, Tennessee, USA
| | - Soghra Fatima
- Division of Experimental Hematology, Department of Hematology, Memphis, Tennessee, USA
| | - Zhijun Ma
- Division of Experimental Hematology, Department of Hematology, Memphis, Tennessee, USA
| | - Yong-Dong Wang
- Department of Computational Biology, Memphis, Tennessee, USA
| | - Taihe Lu
- Division of Experimental Hematology, Department of Hematology, Memphis, Tennessee, USA
| | - Laura J Janke
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yang Du
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Brian P Sorrentino
- Division of Experimental Hematology, Department of Hematology, Memphis, Tennessee, USA.
| |
Collapse
|
31
|
Pollock K, Dahlenburg H, Nelson H, Fink KD, Cary W, Hendrix K, Annett G, Torrest A, Deng P, Gutierrez J, Nacey C, Pepper K, Kalomoiris S, D Anderson J, McGee J, Gruenloh W, Fury B, Bauer G, Duffy A, Tempkin T, Wheelock V, Nolta JA. Human Mesenchymal Stem Cells Genetically Engineered to Overexpress Brain-derived Neurotrophic Factor Improve Outcomes in Huntington's Disease Mouse Models. Mol Ther 2016; 24:965-77. [PMID: 26765769 PMCID: PMC4881765 DOI: 10.1038/mt.2016.12] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 12/05/2015] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a fatal degenerative autosomal dominant neuropsychiatric disease that causes neuronal death and is characterized by progressive striatal and then widespread brain atrophy. Brain-derived neurotrophic factor (BDNF) is a lead candidate for the treatment of HD, as it has been shown to prevent cell death and to stimulate the growth and migration of new neurons in the brain in transgenic mouse models. BDNF levels are reduced in HD postmortem human brain. Previous studies have shown efficacy of mesenchymal stem/stromal cells (MSC)/BDNF using murine MSCs, and the present study used human MSCs to advance the therapeutic potential of the MSC/BDNF platform for clinical application. Double-blinded studies were performed to examine the effects of intrastriatally transplanted human MSC/BDNF on disease progression in two strains of immune-suppressed HD transgenic mice: YAC128 and R6/2. MSC/BDNF treatment decreased striatal atrophy in YAC128 mice. MSC/BDNF treatment also significantly reduced anxiety as measured in the open-field assay. Both MSC and MSC/BDNF treatments induced a significant increase in neurogenesis-like activity in R6/2 mice. MSC/BDNF treatment also increased the mean lifespan of the R6/2 mice. Our genetically modified MSC/BDNF cells set a precedent for stem cell-based neurotherapeutics and could potentially be modified for other neurodegenerative disorders such as amyotrophic lateral sclerosis, Alzheimer's disease, and some forms of Parkinson's disease. These cells provide a platform delivery system for future studies involving corrective gene-editing strategies.
Collapse
Affiliation(s)
- Kari Pollock
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Heather Dahlenburg
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Haley Nelson
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Kyle D Fink
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Whitney Cary
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Kyle Hendrix
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Geralyn Annett
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Audrey Torrest
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Peter Deng
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Joshua Gutierrez
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Catherine Nacey
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Karen Pepper
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Stefanos Kalomoiris
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Johnathon D Anderson
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Jeannine McGee
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - William Gruenloh
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Brian Fury
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Gerhard Bauer
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Alexandria Duffy
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Theresa Tempkin
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Vicki Wheelock
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Jan A Nolta
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| |
Collapse
|
32
|
De Ravin SS, Reik A, Liu PQ, Li L, Wu X, Su L, Raley C, Theobald N, Choi U, Song AH, Chan A, Pearl JR, Paschon DE, Lee J, Newcombe H, Koontz S, Sweeney C, Shivak DA, Zarember KA, Peshwa MV, Gregory PD, Urnov FD, Malech HL. Targeted gene addition in human CD34(+) hematopoietic cells for correction of X-linked chronic granulomatous disease. Nat Biotechnol 2016; 34:424-9. [PMID: 26950749 PMCID: PMC4824656 DOI: 10.1038/nbt.3513] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 02/16/2016] [Indexed: 01/08/2023]
Abstract
Gene therapy with genetically modified human CD34(+) hematopoietic stem and progenitor cells (HSPCs) may be safer using targeted integration (TI) of transgenes into a genomic 'safe harbor' site rather than random viral integration. We demonstrate that temporally optimized delivery of zinc finger nuclease mRNA via electroporation and adeno-associated virus (AAV) 6 delivery of donor constructs in human HSPCs approaches clinically relevant levels of TI into the AAVS1 safe harbor locus. Up to 58% Venus(+) HSPCs with 6-16% human cell marking were observed following engraftment into mice. In HSPCs from patients with X-linked chronic granulomatous disease (X-CGD), caused by mutations in the gp91phox subunit of the NADPH oxidase, TI of a gp91phox transgene into AAVS1 resulted in ∼15% gp91phox expression and increased NADPH oxidase activity in ex vivo-derived neutrophils. In mice transplanted with corrected HSPCs, 4-11% of human cells in the bone marrow expressed gp91phox. This method for TI into AAVS1 may be broadly applicable to correction of other monogenic diseases.
Collapse
Affiliation(s)
- Suk See De Ravin
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | | | | | - Xiaolin Wu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD
| | - Ling Su
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD
| | - Castle Raley
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD
| | - Narda Theobald
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Uimook Choi
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | - Andy Chan
- Sangamo BioSciences, Inc., Richmond, CA
| | | | | | - Janet Lee
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Hannah Newcombe
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Sherry Koontz
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Colin Sweeney
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | - Kol A. Zarember
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Madhusudan V. Peshwa
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD
| | | | | | - Harry L. Malech
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
33
|
Targeted gene editing restores regulated CD40L function in X-linked hyper-IgM syndrome. Blood 2016; 127:2513-22. [PMID: 26903548 DOI: 10.1182/blood-2015-11-683235] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/10/2016] [Indexed: 11/20/2022] Open
Abstract
Loss of CD40 ligand (CD40L) expression or function results in X-linked hyper-immunoglobulin (Ig)M syndrome (X-HIGM), characterized by recurrent infections due to impaired immunoglobulin class-switching and somatic hypermutation. Previous attempts using retroviral gene transfer to correct murine CD40L expression restored immune function; however, treated mice developed lymphoproliferative disease, likely due to viral-promoter-dependent constitutive CD40L expression. These observations highlight the importance of preserving endogenous gene regulation in order to safely correct this disorder. Here, we report efficient, on-target, homology-directed repair (HDR) editing of the CD40LG locus in primary human T cells using a combination of a transcription activator-like effector nuclease-induced double-strand break and a donor template delivered by recombinant adeno-associated virus. HDR-mediated insertion of a coding sequence (green fluorescent protein or CD40L) upstream of the translation start site within exon 1 allowed transgene expression to be regulated by endogenous CD40LG promoter/enhancer elements. Additionally, inclusion of the CD40LG 3'-untranslated region in the transgene preserved posttranscriptional regulation. Expression kinetics of the transgene paralleled that of endogenous CD40L in unedited T cells, both at rest and in response to T-cell stimulation. The use of this method to edit X-HIGM patient T cells restored normal expression of CD40L and CD40-murine IgG Fc fusion protein (CD40-muIg) binding, and rescued IgG class switching of naive B cells in vitro. These results demonstrate the feasibility of engineered nuclease-directed gene repair to restore endogenously regulated CD40L, and the potential for its use in T-cell therapy for X-HIGM syndrome.
Collapse
|
34
|
von Palffy S, Bulaeva E, Babovic S, Kannan N, Knapp DJ, Wei L, Eaves CJ, Beer PA. Dominant-negative IKAROS enhances IL-3-stimulated signaling in wild-type but not BCR-ABL1+ mouse BA/F3 cells. Exp Hematol 2015; 43:514-23.e1-2. [DOI: 10.1016/j.exphem.2015.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 02/08/2023]
|
35
|
DNA methylation and histone modifications are the molecular lock in lentivirally transduced hematopoietic progenitor cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:346134. [PMID: 25961011 PMCID: PMC4417590 DOI: 10.1155/2015/346134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 01/12/2023]
Abstract
Stable introduction of a functional gene in hematopoietic progenitor cells (HPCs) has appeared to be an alternative approach to correct genetically linked blood diseases. However, it is still unclear whether lentiviral vector (LV) is subjected to gene silencing in HPCs. Here, we show that LV carrying green fluorescent protein (GFP) reporter gene driven by cytomegalovirus (CMV) promoter was subjected to transgene silencing after transduction into HPCs. This phenomenon was not due to the deletion of proviral copy number. Study using DNA demethylating agent and histone deacetylase (HDAC) inhibitor showed that the drugs could either prevent or reverse the silencing effect. Using sodium bisulfite sequencing and chromatin immunoprecipitation (ChIP) assay, we demonstrated that DNA methylation occurred soon after LV transduction. At the highest level of gene expression, CMV promoter was acetylated and was in a euchromatin state, while GFP reporter gene was acetylated but was strangely in a heterochromatin state. When the expression declined, CMV promoter underwent transition from acetylated and euchromatic state to a heterochromatic state, while the GFP reporter gene was in deacetylated and heterochromatic state. With these, we verify that DNA methylation and dynamic histone modifications lead to transgene silencing in HPCs transduced with LV.
Collapse
|
36
|
Disruption of IKAROS activity in primitive chronic-phase CML cells mimics myeloid disease progression. Blood 2014; 125:504-15. [PMID: 25370416 DOI: 10.1182/blood-2014-06-581173] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Without effective therapy, chronic-phase chronic myeloid leukemia (CP-CML) evolves into an acute leukemia (blast crisis [BC]) that displays either myeloid or B-lymphoid characteristics. This transition is often preceded by a clinically recognized, but biologically poorly characterized, accelerated phase (AP). Here, we report that IKAROS protein is absent or reduced in bone marrow blasts from most CML patients with advanced myeloid disease (AP or BC). This contrasts with primitive CP-CML cells and BCR-ABL1-negative acute myeloid leukemia blasts, which express readily detectable IKAROS. To investigate whether loss of IKAROS contributes to myeloid disease progression in CP-CML, we examined the effects of forced expression of a dominant-negative isoform of IKAROS (IK6) in CP-CML patients' CD34(+) cells. We confirmed that IK6 disrupts IKAROS activity in transduced CP-CML cells and showed that it confers on them features of AP-CML, including a prolonged increased output in vitro and in xenografted mice of primitive cells with an enhanced ability to differentiate into basophils. Expression of IK6 in CD34(+) CP-CML cells also led to activation of signal transducer and activator of transcription 5 and transcriptional repression of its negative regulators. These findings implicate loss of IKAROS as a frequent step and potential diagnostic harbinger of progressive myeloid disease in CML patients.
Collapse
|
37
|
Beer PA, Knapp DJHF, Kannan N, Miller PH, Babovic S, Bulaeva E, Aghaeepour N, Rabu G, Rostamirad S, Shih K, Wei L, Eaves CJ. A dominant-negative isoform of IKAROS expands primitive normal human hematopoietic cells. Stem Cell Reports 2014; 3:841-57. [PMID: 25418728 PMCID: PMC4235152 DOI: 10.1016/j.stemcr.2014.09.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 09/08/2014] [Accepted: 09/08/2014] [Indexed: 12/11/2022] Open
Abstract
Disrupted IKAROS activity is a recurrent feature of some human leukemias, but effects on normal human hematopoietic cells are largely unknown. Here, we used lentivirally mediated expression of a dominant-negative isoform of IKAROS (IK6) to block normal IKAROS activity in primitive human cord blood cells and their progeny. This produced a marked (10-fold) increase in serially transplantable multipotent IK6+ cells as well as increased outputs of normally differentiating B cells and granulocytes in transplanted immunodeficient mice, without producing leukemia. Accompanying T/natural killer (NK) cell outputs were unaltered, and erythroid and platelet production was reduced. Mechanistically, IK6 specifically increased human granulopoietic progenitor sensitivity to two growth factors and activated CREB and its targets (c-FOS and Cyclin B1). In more primitive human cells, IK6 prematurely initiated a B cell transcriptional program without affecting the hematopoietic stem cell-associated gene expression profile. Some of these effects were species specific, thus identifying novel roles of IKAROS in regulating normal human hematopoietic cells. IKAROS protein is abundantly expressed in primitive human hematopoietic cells IK6 enhances human blood stem cell expansion in vivo without causing leukemia IK6 has a unique profile of lineage-specific effects on human hematopoietic cells IK6 activates B-lineage transcripts prematurely in human blood stem cells
Collapse
Affiliation(s)
- Philip A Beer
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - David J H F Knapp
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Nagarajan Kannan
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Paul H Miller
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Sonja Babovic
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Elizabeth Bulaeva
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Nima Aghaeepour
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Gabrielle Rabu
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Shabnam Rostamirad
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Kingsley Shih
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Lisa Wei
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|
38
|
Bosch MK, Nerbonne JM, Ornitz DM. Dual transgene expression in murine cerebellar Purkinje neurons by viral transduction in vivo. PLoS One 2014; 9:e104062. [PMID: 25093726 PMCID: PMC4122438 DOI: 10.1371/journal.pone.0104062] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/08/2014] [Indexed: 12/19/2022] Open
Abstract
Viral-vector mediated gene transfer to cerebellar Purkinje neurons in vivo is a promising avenue for gene therapy of cerebellar ataxias and for genetic manipulation in functional studies of animal models of cerebellar disease. Here, we report the results of experiments designed to identify efficient methods for viral transduction of adult murine Purkinje neurons in vivo. For these analyses, several lentiviral and an adeno-associated virus (AAV), serotype 1, vector with various promoter combinations were generated and compared for in situ transduction efficiency, assayed by fluorescent reporter protein expression in Purkinje neurons. Additional experiments were also conducted to identify the optimal experimental strategy for co-expression of two proteins in individual Purkinje neurons. Of the viruses tested, AAV1 with a CAG promoter exhibited the highest specificity for Purkinje neurons. To deliver two proteins to the same Purkinje neuron, several methods were tested, including: an internal ribosome entry site (IRES), a 2A sequence, a dual promoter vector, and co-injection of two viruses. Efficient expression of both proteins in the same Purkinje neuron was only achieved by co-injecting two AAV1-CAG viruses. We found that use of an AAV1-CAG virus outperformed similar lentivirus vectors and that co-injection of two AAV1-CAG viruses could be used to efficiently deliver two proteins to the same Purkinje neuron in adult mice. AAV1 with a CAG promoter is highly efficient and selective at transducing adult cerebellar Purkinje neurons and two AAV-CAG viruses can be used to efficiently express two proteins in the same neuron in vivo.
Collapse
Affiliation(s)
- Marie K. Bosch
- Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jeanne M. Nerbonne
- Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - David M. Ornitz
- Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
39
|
Zhou X, Florian MC, Arumugam P, Chen X, Cancelas JA, Lang R, Malik P, Geiger H, Zheng Y. RhoA GTPase controls cytokinesis and programmed necrosis of hematopoietic progenitors. ACTA ACUST UNITED AC 2013; 210:2371-85. [PMID: 24101377 PMCID: PMC3804933 DOI: 10.1084/jem.20122348] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The GTPase RhoA is required for the appropriate division and survival of hematopoietic progenitor cells. Hematopoietic progenitor cells (HPCs) are central to hematopoiesis as they provide large numbers of lineage-defined blood cells necessary to sustain blood homeostasis. They are one of the most actively cycling somatic cells, and their precise control is critical for hematopoietic homeostasis. The small GTPase RhoA is an intracellular molecular switch that integrates cytokine, chemokine, and adhesion signals to coordinate multiple context-dependent cellular processes. By using a RhoA conditional knockout mouse model, we show that RhoA deficiency causes a multilineage hematopoietic failure that is associated with defective multipotent HPCs. Interestingly, RhoA−/− hematopoietic stem cells retained long-term engraftment potential but failed to produce multipotent HPCs and lineage-defined blood cells. This multilineage hematopoietic failure was rescued by reconstituting wild-type RhoA into the RhoA−/− Lin−Sca-1+c-Kit+ compartment. Mechanistically, RhoA regulates actomyosin signaling, cytokinesis, and programmed necrosis of the HPCs, and loss of RhoA results in a cytokinesis failure of HPCs manifested by an accumulation of multinucleated cells caused by failed abscission of the cleavage furrow after telophase. Concomitantly, the HPCs show a drastically increased death associated with increased TNF–RIP-mediated necrosis. These results show that RhoA is a critical and specific regulator of multipotent HPCs during cytokinesis and thus essential for multilineage hematopoiesis.
Collapse
Affiliation(s)
- Xuan Zhou
- Division of Experimental Hematology and Cancer Biology, 2 Molecular and Development Biology Graduate Program, 3 Division of Ophthalmology, and 4 Division of Developmental Biology, Cincinnati Children's Research Foundation, University of Cincinnati, Cincinnati, OH 45229
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
The Lin28b-let-7-Hmga2 axis determines the higher self-renewal potential of fetal haematopoietic stem cells. Nat Cell Biol 2013; 15:916-25. [PMID: 23811688 DOI: 10.1038/ncb2783] [Citation(s) in RCA: 276] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 05/13/2013] [Indexed: 12/14/2022]
Abstract
Mouse haematopoietic stem cells (HSCs) undergo a postnatal transition in several properties, including a marked reduction in their self-renewal activity. We now show that the developmentally timed change in this key function of HSCs is associated with their decreased expression of Lin28b and an accompanying increase in their let-7 microRNA levels. Lentivirus-mediated overexpression of Lin28 in adult HSCs elevates their self-renewal activity in transplanted irradiated hosts, as does overexpression of Hmga2, a well-established let-7 target that is upregulated in fetal HSCs. Conversely, HSCs from fetal Hmga2(-/-) mice do not exhibit the heightened self-renewal activity that is characteristic of wild-type fetal HSCs. Interestingly, overexpression of Hmga2 in adult HSCs does not mimic the ability of elevated Lin28 to activate a fetal lymphoid differentiation program. Thus, Lin28b may act as a master regulator of developmentally timed changes in HSC programs with Hmga2 serving as its specific downstream modulator of HSC self-renewal potential.
Collapse
|
41
|
Koldej RM, Carney G, Wielgosz MM, Zhou S, Zhan J, Sorrentino BP, Nienhuis AW. Comparison of insulators and promoters for expression of the Wiskott-Aldrich syndrome protein using lentiviral vectors. HUM GENE THER CL DEV 2013; 24:77-85. [PMID: 23786330 DOI: 10.1089/humc.2012.244] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Gene therapy for the treatment of Wiskott-Aldrich syndrome (WAS) presents an alternative to the current use of allogeneic bone marrow transplantation. We describe the development of a self-inactivating lentiviral vector containing chromatin insulators for treatment of WAS and compare a gammaretroviral (MND), human cellular (EF1α), and the human WASp gene promoter for expression patterns in vivo during murine hematopoiesis using the green fluorescent protein (GFP) marker. Compared with the EF1α and the WASp promoters, expression from the MND promoter in mouse transplant recipients was much higher in all lineages examined. Importantly, there was sustained expression in the platelets of secondary recipient animals, necessary to correct the thrombocytopenia defect in WAS patients. Analysis of WAS protein expression in transduced human EBV-immortalized B-cells and transduced patient peripheral blood mononuclear cells also demonstrated stronger expression per copy from the MND promoter compared with the other promoters. In addition, when analyzed in an LM02 activation assay, the addition of an insulator to MND-promoter-containing constructs reduced transactivation of the LM02 gene. We propose a clinical trial design in which cytokine-mobilized, autologous, transduced CD34(+) cells are administered after myelosuppression.
Collapse
Affiliation(s)
- Rachel M Koldej
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Allelic exclusion and peripheral reconstitution by TCR transgenic T cells arising from transduced human hematopoietic stem/progenitor cells. Mol Ther 2013; 21:1044-54. [PMID: 23380815 DOI: 10.1038/mt.2013.8] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Transduction and transplantation of human hematopoietic stem/progenitor cells (HSPC) with the genes for a T-cell receptor (TCR) that recognizes a tumor-associated antigen may lead to sustained long-term production of T cells expressing the TCR and confer specific antitumor activity. We evaluated this using a lentiviral vector (CCLc-MND-F5) carrying cDNA for a human TCR specific for an HLA-A*0201-restricted peptide of Melanoma Antigen Recognized by T cells (MART-1). CD34(+) HSPC were transduced with the F5 TCR lentiviral vector or mock transduced and transplanted into neonatal NSG mice or NSG mice transgenic for human HLA-A*0201 (NSG-A2). Human CD8(+) and CD4(+) T cells expressing the human F5 TCR were present in the thymus, spleen, and peripheral blood after 4-5 months. Expression of human HLA-A*0201 in NSG-A2 recipient mice led to significantly increased numbers of human CD8(+) and CD4(+) T cells expressing the F5 TCR, compared with control NSG recipients. Transduction of the human CD34(+) HSPC by the F5 TCR transgene caused a high degree of allelic exclusion, potently suppressing rearrangement of endogenous human TCR-β genes during thymopoiesis. In summary, we demonstrated the feasibility of engineering human HSPC to express a tumor-specific TCR to serve as a long-term source of tumor-targeted mature T cells for immunotherapy of melanoma.
Collapse
|
43
|
Gene therapy for adenosine deaminase-deficient severe combined immune deficiency: clinical comparison of retroviral vectors and treatment plans. Blood 2012; 120:3635-46. [PMID: 22968453 DOI: 10.1182/blood-2012-02-400937] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We conducted a gene therapy trial in 10 patients with adenosine deaminase (ADA)-deficient severe combined immunodeficiency using 2 slightly different retroviral vectors for the transduction of patients' bone marrow CD34(+) cells. Four subjects were treated without pretransplantation cytoreduction and remained on ADA enzyme-replacement therapy (ERT) throughout the procedure. Only transient (months), low-level (< 0.01%) gene marking was observed in PBMCs of 2 older subjects (15 and 20 years of age), whereas some gene marking of PBMC has persisted for the past 9 years in 2 younger subjects (4 and 6 years). Six additional subjects were treated using the same gene transfer protocol, but after withdrawal of ERT and administration of low-dose busulfan (65-90 mg/m(2)). Three of these remain well, off ERT (5, 4, and 3 years postprocedure), with gene marking in PBMC of 1%-10%, and ADA enzyme expression in PBMC near or in the normal range. Two subjects were restarted on ERT because of poor gene marking and immune recovery, and one had a subsequent allogeneic hematopoietic stem cell transplantation. These studies directly demonstrate the importance of providing nonmyeloablative pretransplantation conditioning to achieve therapeutic benefits with gene therapy for ADA-deficient severe combined immunodeficiency.
Collapse
|
44
|
Gene therapy/bone marrow transplantation in ADA-deficient mice: roles of enzyme-replacement therapy and cytoreduction. Blood 2012; 120:3677-87. [PMID: 22833548 DOI: 10.1182/blood-2012-02-408591] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Gene therapy (GT) for adenosine deaminase-deficient severe combined immune deficiency (ADA-SCID) can provide significant long-term benefit when patients are given nonmyeloablative conditioning and ADA enzyme-replacement therapy (ERT) is withheld before autologous transplantation of γ-retroviral vector-transduced BM CD34+ cells. To determine the contributions of conditioning and discontinuation of ERT to the therapeutic effects, we analyzed these factors in Ada gene knockout mice (Ada(-/-)). Mice were transplanted with ADA-deficient marrow transduced with an ADA-expressing γ-retroviral vector without preconditioning or after 200 cGy or 900 cGy total-body irradiation and evaluated after 4 months. In all tissues analyzed, vector copy numbers (VCNs) were 100- to 1000-fold greater in mice receiving 900 cGy compared with 200 cGy (P < .05). In mice receiving 200 cGy, VCN was similar whether ERT was stopped or given for 1 or 4 months after GT. In unconditioned mice, there was decreased survival with and without ERT, and VCN was very low to undetectable. When recipients were conditioned with 200 cGy and received transduced lineage-depleted marrow, only recipients receiving ERT (1 or 4 months) had detectable vector sequences in thymocytes. In conclusion, cytoreduction is important for the engraftment of gene-transduced HSC, and short-term ERT after GT did not diminish the capacity of gene-corrected cells to engraft and persist.
Collapse
|
45
|
Šenigl F, Auxt M, Hejnar J. Transcriptional provirus silencing as a crosstalk of de novo DNA methylation and epigenomic features at the integration site. Nucleic Acids Res 2012; 40:5298-312. [PMID: 22379139 PMCID: PMC3384319 DOI: 10.1093/nar/gks197] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 02/12/2012] [Accepted: 02/13/2012] [Indexed: 12/03/2022] Open
Abstract
The autonomous transcription of integrated retroviruses strongly depends on genetic and epigenetic effects of the chromatin at the site of integration. These effects are mostly suppressive and proviral activity can be finally silenced by mechanisms, such as DNA methylation and histone modifications. To address the role of the integration site at the whole-genome-scale, we performed clonal analysis of provirus silencing with an avian leucosis/sarcoma virus-based reporter vector and correlated the transcriptional silencing with the epigenomic landscape of respective integrations. We demonstrate efficient provirus silencing in human HCT116 cell line, which is strongly but not absolutely dependent on the de novo DNA methyltransferase activity, particularly of Dnmt3b. Proviruses integrated close to the transcription start sites of active genes into the regions enriched in H3K4 trimethylation display long-term stability of expression and are resistant to the transcriptional silencing after over-expression of Dnmt3a or Dnmt3b. In contrast, proviruses in the intergenic regions tend to spontaneous transcriptional silencing even in Dnmt3a(-/-) Dnmt3b(-/-) cells. The silencing of proviruses within genes is accompanied with DNA methylation of long terminal repeats, whereas silencing in intergenic regions is DNA methylation-independent. These findings indicate that the epigenomic features of integration sites are crucial for their permissivity to the proviral expression.
Collapse
Affiliation(s)
- Filip Šenigl
- Department of Cellular and Viral Genetics, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220 Prague, Czech Republic
| | | | - Jiří Hejnar
- Department of Cellular and Viral Genetics, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220 Prague, Czech Republic
| |
Collapse
|
46
|
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
|
47
|
Cao H, Molday RS, Hu J. Gene therapy: light is finally in the tunnel. Protein Cell 2012; 2:973-89. [PMID: 22231356 DOI: 10.1007/s13238-011-1126-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 11/27/2011] [Indexed: 01/23/2023] Open
Abstract
After two decades of ups and downs, gene therapy has recently achieved a milestone in treating patients with Leber's congenital amaurosis (LCA). LCA is a group of inherited blinding diseases with retinal degeneration and severe vision loss in early infancy. Mutations in several genes, including RPE65, cause the disease. Using adeno-associated virus as a vector, three independent teams of investigators have recently shown that RPE65 can be delivered to retinal pigment epithelial cells of LCA patients by subretinal injections resulting in clinical benefits without side effects. However, considering the whole field of gene therapy, there are still major obstacles to clinical applications for other diseases. These obstacles include innate and immune barriers to vector delivery, toxicity of vectors and the lack of sustained therapeutic gene expression. Therefore, new strategies are needed to overcome these hurdles for achieving safe and effective gene therapy. In this article, we shall review the major advancements over the past two decades and, using lung gene therapy as an example, discuss the current obstacles and possible solutions to provide a roadmap for future gene therapy research.
Collapse
Affiliation(s)
- Huibi Cao
- Programme in Physiology and Experimental Medicine, Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5G, 1X8, Canada
| | | | | |
Collapse
|
48
|
Catros S, Guillemot F, Nandakumar A, Ziane S, Moroni L, Habibovic P, van Blitterswijk C, Rousseau B, Chassande O, Amédée J, Fricain JC. Layer-by-layer tissue microfabrication supports cell proliferation in vitro and in vivo. Tissue Eng Part C Methods 2011; 18:62-70. [PMID: 21895563 DOI: 10.1089/ten.tec.2011.0382] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Layer-by-layer biofabrication represents a novel strategy to create three-dimensional living structures with a controlled internal architecture, using cell micromanipulation technologies. Laser assisted bioprinting (LAB) is an effective printing method for patterning cells, biomolecules, and biomaterials in two dimensions. "Biopapers," made of thin polymer scaffolds, may be appropriate to achieve three-dimensional constructs and to reinforce mechanical properties of printed materials. The aim of this work was to evaluate the effect of the tridimensional organization of cells and biomaterials on cell proliferation in vitro and in vivo. The experimental LAB setup was comprised of an infrared laser, focused onto a glass ribbon coated with an absorbing layer of gold. The cell bioink was made of MG63 cells (50 millions cells/mL in culture medium and 1% alginate), transduced with Luciferase gene for tracking and quantification. The printing substrate was a 100-μm-thick polycaprolacton (PCL) electrospun scaffold. The building sequence comprised sequential layers of cells and PCL scaffolds stacked using two different tridimensional arrangements, which were compared in this study (layer-by-layer vs. seeding on a single locus of the scaffolds). Then the cell-seeded materials were cultured in vitro or implanted in vivo in NOD-SCID mice. The qualitative follow-up involved scanning electron microscopy (SEM) observations, live-dead assays, and histology. The cell amount was quantified by photon imager during 21 days in vitro and 2 months in vivo. Live- dead assay and SEM revealed that the cells survived after printing and spread onto PCL membranes. Circle-shaped patterns were maintained in vitro during the first week but they were no longer observable after 2 weeks, due to cell proliferation. Luciferase tracking displayed that the cell amount was increased in vitro and in vivo when the materials and the cells where stacked layer by layer. Histological sections of the in vivo samples revealed a thicker fibrous tissue in the layer-by-layer samples. We have demonstrated in this study that PCL electrospun biopapers can act as a shock-absorbing mattress for cell printing and could further support cell proliferation. The layer-by-layer printing provided an appropriate 3D environment for cell survival and enhanced cell proliferation in vitro and in vivo.
Collapse
Affiliation(s)
- Sylvain Catros
- Inserm U 1026, Tissue BioEnginnering, Bordeaux Segalen University, Bordeaux, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gammaretroviral vectors: biology, technology and application. Viruses 2011; 3:677-713. [PMID: 21994751 PMCID: PMC3185771 DOI: 10.3390/v3060677] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 05/03/2011] [Accepted: 05/09/2011] [Indexed: 12/11/2022] Open
Abstract
Retroviruses are evolutionary optimized gene carriers that have naturally adapted to their hosts to efficiently deliver their nucleic acids into the target cell chromatin, thereby overcoming natural cellular barriers. Here we will review—starting with a deeper look into retroviral biology—how Murine Leukemia Virus (MLV), a simple gammaretrovirus, can be converted into an efficient vehicle of genetic therapeutics. Furthermore, we will describe how more rational vector backbones can be designed and how these so-called self-inactivating vectors can be pseudotyped and produced. Finally, we will provide an overview on existing clinical trials and how biosafety can be improved.
Collapse
|
50
|
Linterman KS, Palmer DN, Kay GW, Barry LA, Mitchell NL, McFarlane RG, Black MA, Sands MS, Hughes SM. Lentiviral-mediated gene transfer to the sheep brain: implications for gene therapy in Batten disease. Hum Gene Ther 2011; 22:1011-20. [PMID: 21595499 DOI: 10.1089/hum.2011.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs; Batten disease) are inherited neurodegenerative lysosomal storage diseases with common clinical features of blindness and seizures culminating in premature death. Gene-therapy strategies for these diseases depend on whether the missing activity is a secreted lysosomal protein taken up by neighboring cells, or an intramembrane protein that requires careful targeting. Therapies are best developed in animal models with large complex human-like brains. Lentiviral-mediated gene delivery to neural cell cultures from normal sheep and sheep affected with an NCL resulted in green fluorescent protein (GFP) expression in neurons and neuroblasts, more efficiently than in astrocytes. Similar transgene expression was obtained from two constitutive promoters, the viral MND promoter and the human EF1α promoter. In vivo studies showed stable and persistent GFP expression throughout the cell bodies, axons, and dendrites from intracortical injections and indicated ependymal and subependymal transduction. The sheep showed no ill effects from the injections. These data support continuing gene-therapy trials in the sheep models of Batten disease.
Collapse
Affiliation(s)
- Kathryn S Linterman
- Department of Biochemistry, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| | | | | | | | | | | | | | | | | |
Collapse
|