This project studies peripheral blood hematopoietic progenitors (PBHP) as a target for gene therapy of inherited diseases affecting cells of the immune system. This project also studies the pathophysiology of inherited immune deficiencies with the ultimate goal of developing hematopoietic stem cell gene therapy for these disorders. We have developed new methods and materials which improve our ability to get new genes into human blood stem cells. The specific goals were to develop a system of gene therapy to correct the genetic defect in the X-linked genetic form of chronic granulomatous disease (CGD) and the X-linked form of severe combined immune deficiency (XSCID). Earlier results of this development program have been used in an ongoing clinical trial of gene therapy for CGD and in the pre-clinical work required to set up a clinical trial for XSCID. That clinical trial for X-CGD and the results from that clinical trial will be discussed in the report for Project Z01-AI-00645. Specifically, we developed a retrovirus vector producer cell line that secretes high titers of the MFGS vectors containing the gp91phox cDNA that will correct the functional defect in X-linked CGD neutrophils. We demonstrated that the fibronectin fragment CH-296 coated on culture vessel surfaces will greatly augment the gene transfer correction of stem cells from patients with CGD. More recently in collaboration with a group at St. Jude's Hospital Medical Center in Memphis we have developed a version of our CGD corrective vector that is pseudotyped with the FLYDR (RD114) retrovirus envelope resulting in extraordinarily high level of gene transfer into hematopoietic stem cells. This vector will serve as the basis for the next generation clinical trial of gene therapy in the planning stage. We have also begun to explore the use of highly modified lentivirus vectors in the laboratory. These vectors may have greater potential to target the most primitive stem cells which are not dividing. In other studies we are examining the role of different growth factors in stimulating CD34+ stem cells to divide and to determine the relationship between entry into the cell cycle, ability to transduce with retrovirus vectors, and the maintenance or loss of long term engrafting potential. These studies are essential to achieving our goal of high levels of gene transfer into long term engrafting stem cell. In other studies we have studied the effects of low dose radiation on the engraftment of stem cells in animal models. Our initial studies were in mice, but in collaboration with investigators at the University of Maryland and the New England Primate Center (Harvard University, we have demonstrated high levels of engraftment of gene marked cells in primate models using low intensity conditioning regimens consisting of non-ablative levels of total body radiation. Follow up studies are in progress looking at non-ablative chemotherapy regimens instead of using radiation. In other studies we have contributed to genetic studies of patients with the inherited disorder known as Hyper IgE-Recurrent Infection Syndrome. These studies involved the delineation of the clinical features and the delineation of potential sites of the gene(s) causing this disorder.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Intramural Research (Z01)
Project #
1Z01AI000644-09
Application #
6431616
Study Section
(LHD)
Project Start
Project End
Budget Start
Budget End
Support Year
9
Fiscal Year
2000
Total Cost
Indirect Cost
Name
Niaid Extramural Activities
Department
Type
DUNS #
City
State
Country
United States
Zip Code
De Ravin, Suk See; Li, Linhong; Wu, Xiaolin et al. (2017) CRISPR-Cas9 gene repair of hematopoietic stem cells from patients with X-linked chronic granulomatous disease. Sci Transl Med 9:
Sweeney, Colin L; Teng, Ruifeng; Wang, Hongmei et al. (2016) Molecular Analysis of Neutrophil Differentiation from Human Induced Pluripotent Stem Cells Delineates the Kinetics of Key Regulators of Hematopoiesis. Stem Cells 34:1513-26
De Ravin, Suk See; Reik, Andreas; Liu, Pei-Qi et al. (2016) Targeted gene addition in human CD34(+) hematopoietic cells for correction of X-linked chronic granulomatous disease. Nat Biotechnol 34:424-9
Merling, Randall K; Sweeney, Colin L; Chu, Jessica et al. (2015) An AAVS1-targeted minigene platform for correction of iPSCs from all five types of chronic granulomatous disease. Mol Ther 23:147-57
Zou, Jizhong; Sweeney, Colin L; Chou, Bin-Kuan et al. (2011) Oxidase-deficient neutrophils from X-linked chronic granulomatous disease iPS cells: functional correction by zinc finger nuclease-mediated safe harbor targeting. Blood 117:5561-72
Kang, Elizabeth M; Malech, Harry L (2009) Advances in treatment for chronic granulomatous disease. Immunol Res 43:77-84
Sweeney, Colin L; Malech, Harry L (2009) Functional neutrophils from human ES cells. Blood 113:6503-5
De Ravin, Suk See; Malech, Harry L (2009) Partially corrected X-linked severe combined immunodeficiency: long-term problems and treatment options. Immunol Res 43:223-42
Kawai, Toshinao; Choi, Uimook; Cardwell, Lanise et al. (2007) WHIM syndrome myelokathexis reproduced in the NOD/SCID mouse xenotransplant model engrafted with healthy human stem cells transduced with C-terminus-truncated CXCR4. Blood 109:78-84
Naumann, N; De Ravin, S S; Choi, U et al. (2007) Simian immunodeficiency virus lentivector corrects human X-linked chronic granulomatous disease in the NOD/SCID mouse xenograft. Gene Ther 14:1513-24

Showing the most recent 10 out of 67 publications