The potential for hematopoietic stem cell therapy to cure HIV disease gains credence from the experience of the `Berlin patient' and is made more feasible by emerging developments in gene editing, particularly when coupled with non-mutating RNA based transduction strategies: the strength of Project 1. It is also now possible to credibly test anti-HIV cell therapies pre-clinically using humanized mouse models: the strength of Project 3 and Core B. What remains a challenge to reduce the complexity of hematopoietic stem cell transplant so that it may be more readily adopted in settings that are not acutely life threatening such as chronic HIV disease. Gene editing will make possible autologous cell transplant thereby removing the devastating complication of graft versus host disease. However, `conditioning' to enable stem cells to engraft the marrow is toxic and requires resource intensive hospitalization as currently practiced. We propose to use methods of modifying hematopoietic stem/progenitor cells' (HSPC) interactions with their bone marrow microenvironment to test novel, niche sparing methods to enable HSPC engraftment with reduced toxicity and potentially less cost, while assessing the impact on HIV-1 reservoirs and generation of T lymphoid reconstitution.
The specific aims of this project are:
Specific aim 1. Competitively advantage donor cells over endogenous stem cells through selective inhibition of eicosanoid signaling.
Specific aim 2. Enhance niche vacancy without niche toxicity through manipulation of CXCR2 and heparan sulfate proteoglycan interactions between stem cells and the bone marrow niche.
Specific aim 3. Selectively deplete endogenous hematopoietic cells using a novel saporin-antibody hybrid based targeting system.
Specific aim 4. Test the role of conditioning in reducing the `reservoir' of HIV infected cells by comparative analysis of cytotoxic and non-cytotoxic methods of conditioning in HIV infected xenogeneic models. Test methods of enhancing T cell reconstitution to improve generation of anti-HIV-1 immunity. If successful, this project will provide specific interventions that can lower the barrier for gene edited HSPC transplantation as an approach to cure HIV/AIDS.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Program--Cooperative Agreements (U19)
Project #
5U19HL129903-04
Application #
9462623
Study Section
Special Emphasis Panel (ZAI1)
Program Officer
Thomas, John
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
Harvard University
Department
Type
DUNS #
082359691
City
Cambridge
State
MA
Country
United States
Zip Code
Hoggatt, Jonathan; Singh, Pratibha; Tate, Tiffany A et al. (2018) Rapid Mobilization Reveals a Highly Engraftable Hematopoietic Stem Cell. Cell 172:191-204.e10
Mondal, Nandini; Dykstra, Brad; Lee, Jungmin et al. (2018) Distinct human ?(1,3)-fucosyltransferases drive Lewis-X/sialyl Lewis-X assembly in human cells. J Biol Chem 293:7300-7314
Garrison, Brian S; Rybak, Adrian P; Beerman, Isabel et al. (2017) ZFP521 regulates murine hematopoietic stem cell function and facilitates MLL-AF9 leukemogenesis in mouse and human cells. Blood 130:619-624
Lee, Jungmin; Dykstra, Brad; Spencer, Joel A et al. (2017) mRNA-mediated glycoengineering ameliorates deficient homing of human stem cell-derived hematopoietic progenitors. J Clin Invest 127:2433-2437
Gutierrez-Martinez, Paula; Rossi, Derrick J; Beerman, Isabel (2016) DNA Damage and Aging Around the Clock. Trends Mol Med 22:635-637
Palchaudhuri, Rahul; Saez, Borja; Hoggatt, Jonathan et al. (2016) Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin. Nat Biotechnol 34:738-45
Dykstra, Brad; Lee, Jungmin; Mortensen, Luke J et al. (2016) Glycoengineering of E-Selectin Ligands by Intracellular versus Extracellular Fucosylation Differentially Affects Osteotropism of Human Mesenchymal Stem Cells. Stem Cells 34:2501-2511