Complete correction of murine Artemis immunodeficiency by lentiviral vector-mediated gene transfer

G Mostoslavsky, AJ Fabian, S Rooney… - Proceedings of the …, 2006 - National Acad Sciences
G Mostoslavsky, AJ Fabian, S Rooney, FW Alt, RC Mulligan
Proceedings of the National Academy of Sciences, 2006National Acad Sciences
Artemis gene mutations are responsible for the development of a severe combined
immunodeficiency [radiation-sensitive (RS) SCID] characterized by a severe B and T cell
deficiency and a normal natural killer cell population. To establish the feasibility of a gene
therapy approach to the treatment of RS-SCID, we generated a series of lentiviral vectors
expressing human Artemis from different promoters and used them to transduce highly
purified hematopoietic stem cells (HSCs) from Artemis knockout mice. HSCs transduced by …
Artemis gene mutations are responsible for the development of a severe combined immunodeficiency [radiation-sensitive (RS) SCID] characterized by a severe B and T cell deficiency and a normal natural killer cell population. To establish the feasibility of a gene therapy approach to the treatment of RS-SCID, we generated a series of lentiviral vectors expressing human Artemis from different promoters and used them to transduce highly purified hematopoietic stem cells (HSCs) from Artemis knockout mice. HSCs transduced by the different viruses were transplanted into either lethally irradiated Rag-1-deficient animals or Artemis knockout mice treated with a nonmyeloablative dose of Busulfan. In both models, transplantation of HSCs transduced by a vector that used a murine phosphoglycerate kinase (PGK) promoter led to a complete functional correction of the immunodeficiency. Corrected animals displayed rescue of mature B cells with normal levels of serum immunoglobulins, together with complete rescue of the T cell compartment as evidenced by the presence of mature T lymphocytes in peripheral blood as well as normal values of thymocytes in thymus. Those B and T cells were capable of activation, as shown both by in vitro stimulation responses and in vivo after immune challenge. Overall, the results indicate that a gene therapy approach for RS-SCID involving the transplantation of genetically modified HSCs is indeed feasible. Furthermore, our studies suggest the possibility that nonmyeloablative conditioning regimens might be effectively used to promote engraftment of genetically modified cells in the case of diseases where standard irradiation-based myeloablative bone marrow transplantation protocols may prove problematic.
National Acad Sciences