Several recent technical advances in gene engineering technologies are improving both the safety and efficiency with which human hematopoietic stem cells (HSC) can be engineered. In particular, the new class of targeted nucleases, which includes zinc finger nucleases (ZFNs), TALENs and the CRISPR/Cas9 system, are allowing the precise modification of genetic loci and are therefore being embraced for human gene therapy applications. For HIV/AIDS, the first-in-man use of ZFNs is being explored in a strategy based on disruption of the CCR5 co-receptor gene in patients' own T cells. A similar HSC based trial is expected to follow shortly, providing the possibility of more long-term effects from this therapy. However, CCR5 disruption strategies alone may be limited by the fact that (i) cells with only a single CCR5 allele disrupted by the ZFNs will not become resistant to HIV, and (ii) there will be no resistance to dual or CXCR4-tropic viruses. To address these issues and to develop the next generation of therapies for HIV/AIDS based on these remarkable gene editing tools, we are now exploiting their abilities to promote precise gene editing or gene insertion events. This is possible because nuclease-induced DNA breaks can be repaired by homology directed repair (HDR) pathways, where genetic information is copied from a homologous `donor sequence'. Such donors can be designed to result in gene editing events, or to promote the insertion of new genetic material at a highly- specific location. We have recently bypassed a significant bottleneck that had limited the use of this technology in human HSC and can now achieve HDR-mediated gene editing at high levels. We will use this capability to expand the use of targeted nucleases beyond CCR5 gene knockout, by developing new approaches for HIV gene therapy based on either the in situ editing of endogenous human restriction factors, or the precise knock- in of broad spectrum anti-HIV genes at defined loci.

Public Health Relevance

The inability of antiretroviral drugs to cure HIV infection compounds the public health impact of this devastating pathogen and warrants investigations of alternate treatment approaches Anti-HIV gene therapies based on CCR5 gene disruption by zinc finger nucleases are already in clinical trials. We will develop a next generation of such treatments based on either the in situ editing of restriction factor genes, or the precise knock-in of broad spectrum anti-HIV genes at defined loci.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Program--Cooperative Agreements (U19)
Project #
5U19HL129902-04
Application #
9462850
Study Section
Special Emphasis Panel (ZAI1)
Program Officer
Welniak, Lisbeth A
Project Start
Project End
Budget Start
2018-04-01
Budget End
2019-03-31
Support Year
4
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of Southern California
Department
Type
DUNS #
072933393
City
Los Angeles
State
CA
Country
United States
Zip Code
90089
Canny, Marella D; Moatti, Nathalie; Wan, Leo C K et al. (2018) Inhibition of 53BP1 favors homology-dependent DNA repair and increases CRISPR-Cas9 genome-editing efficiency. Nat Biotechnol 36:95-102
Colonna, Lucrezia; Peterson, Christopher W; Schell, John B et al. (2018) Evidence for persistence of the SHIV reservoir early after MHC haploidentical hematopoietic stem cell transplantation. Nat Commun 9:4438
Paul, Biswajit; Ibarra, Guillermo S Romano; Hubbard, Nicholas et al. (2018) Efficient Enrichment of Gene-Modified Primary T Cells via CCR5-Targeted Integration of Mutant Dihydrofolate Reductase. Mol Ther Methods Clin Dev 9:347-357
Reeves, Daniel B; Peterson, Christopher W; Kiem, Hans-Peter et al. (2017) Autologous Stem Cell Transplantation Disrupts Adaptive Immune Responses during Rebound Simian/Human Immunodeficiency Virus Viremia. J Virol 91:
Peterson, Christopher W; Benne, Clarisse; Polacino, Patricia et al. (2017) Loss of immune homeostasis dictates SHIV rebound after stem-cell transplantation. JCI Insight 2:e91230
Peterson, Christopher W; Wang, Jianbin; Norman, Krystin K et al. (2016) Long-term multilineage engraftment of autologous genome-edited hematopoietic stem cells in nonhuman primates. Blood 127:2416-26
Wang, Cathy X; Cannon, Paula M (2016) The clinical applications of genome editing in HIV. Blood 127:2546-52
Wang, Jianbin; Exline, Colin M; DeClercq, Joshua J et al. (2015) Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors. Nat Biotechnol 33:1256-1263