The development of inhibitory antibodies to exogenous FVIII is an important complication of FVIII infusion in hemophilia A patients. Generation of such inhibitors might have the potential to preclude gene therapy for hemophilia A. In this project, we propose to investigate a novel approach for gene therapy of hemophilia A and hemophilia A with inhibitors based on the hypothesis that targeting the production of FVIII to a regulated secretory cell type (platelet) that activates at the site where FVIII is needed could overcome the presence of inhibitory antibodies. We have recently developed a vector (2bF8) that targets human FVIII expression to platelets under control of the platelet-specific 1IIb gene promoter. Using this construct, we can correct the murine hemophilia A phenotype even in the presence of high titer inhibitory antibodies in a transgenic mouse model. Another study using 2bF8 lentivirus-mediated bone marrow (BM) transduction and syngeneic transplantation resulted in therapeutic levels of FVIII expression in platelets in hemophilic mice with no antibody development. Our most recent studies have demonstrated that syngeneic transplantation of hematopoietic stem cells (HSC) from 2bF8 transgenic mice can efficiently restore hemostasis to hemophilic mice with inhibitors. In this R01 grant application, we propose to further explore whether 2bF8 lentiviral-mediated gene transfer can introduce sufficient platelet-FVIII expression and rescue the hemorrhagic phenotype in hemophilic mice and hemophilic sheep with inhibitors. To accomplish these, we first propose to transduce HSC from immunized FVIII null mice with 2bF8 lentivirus and transplant into immunized littermates. After BM reconstitution, the recipients will be analyzed by PCR, real-time PCR, LAM-PCR, confocal microscopy, FVIII activity assays, and inhibitor assays. Phenotypic correction will be assessed by tail clip survival test and in vivo thrombus formation. Secondly, we will transduce HSC from hemophilic sheep with or without inhibitors using 2bF8 lentivirus and then transplant the 2bF8-transduced autologous cells back into the animals. The treated animals will be analyzed by the assays as described above, plus whole blood clotting time, thrombin generation assay and thromboelastography assay. Treated animals will be closely monitored to determine if the hemorrhagic phenotype has been improved. Lastly, we will transplant the 2bF8-transduced sheep HSC into hemophilic sheep fetuses to investigate the efficacy and safety of fetal gene therapy using 2bF8 lentiviral vector. These studies should allow us to address the efficacy and safety of our lentivirus-mediated platelet-specific gene therapy approach in animal models with anti-FVIII specific immunity, with the potential to develop a long- term strategy for gene therapy of hemophilia A patients and patients with inhibitors.

Public Health Relevance

Hemophilia A is the most common severe bleeding disorder resulting from hereditary deficiency of the blood clotting protein, factor VIII (FVIII). The development of inhibitory antibodies to exogenous FVIII is considered a severe and important complication of FVIII infusion in hemophilia A patients. In this project, we will develop a gene therapy approach by targeting the synthesis of FVIII to blood platelets, that might cure hemophilia A patients as well as patients with inhibitors.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL102035-03
Application #
8267673
Study Section
Hemostasis and Thrombosis Study Section (HT)
Program Officer
Link, Rebecca P
Project Start
2010-07-01
Project End
2015-04-30
Budget Start
2012-05-01
Budget End
2013-04-30
Support Year
3
Fiscal Year
2012
Total Cost
$355,610
Indirect Cost
$25,182
Name
Medical College of Wisconsin
Department
Pediatrics
Type
Schools of Medicine
DUNS #
937639060
City
Milwaukee
State
WI
Country
United States
Zip Code
53226
Shi, Qizhen (2018) Platelet-Targeted Gene Therapy for Hemophilia. Mol Ther Methods Clin Dev 9:100-108
Luo, Xiaofeng; Chen, Juan; Schroeder, Jocelyn A et al. (2018) Platelet Gene Therapy Promotes Targeted Peripheral Tolerance by Clonal Deletion and Induction of Antigen-Specific Regulatory T Cells. Front Immunol 9:1950
Baumgartner, C K; Mattson, J G; Weiler, H et al. (2017) Targeting factor VIII expression to platelets for hemophilia A gene therapy does not induce an apparent thrombotic risk in mice. J Thromb Haemost 15:98-109
Chen, Juan; Schroeder, Jocelyn A; Luo, Xiaofeng et al. (2017) The impact of von Willebrand factor on factor VIII memory immune responses. Blood Adv 1:1565-1574
Chen, Y; Luo, X; Schroeder, J A et al. (2017) Immune tolerance induced by platelet-targeted factor VIII gene therapy in hemophilia A mice is CD4 T cell mediated. J Thromb Haemost 15:1994-2004
Chen, Yingyu; Schroeder, Jocelyn A; Chen, Juan et al. (2016) The immunogenicity of platelet-derived FVIII in hemophilia A mice with or without preexisting anti-FVIII immunity. Blood 127:1346-54
Haribhai, Dipica; Luo, Xiaofeng; Chen, Juan et al. (2016) TGF-?1 along with other platelet contents augments Treg cells to suppress anti-FVIII immune responses in hemophilia A mice. Blood Adv 1:139-151
Baumgartner, C K; Zhang, G; Kuether, E L et al. (2015) Comparison of platelet-derived and plasma factor VIII efficacy using a novel native whole blood thrombin generation assay. J Thromb Haemost 13:2210-9
Shi, Q; Schroeder, J A; Kuether, E L et al. (2015) The important role of von Willebrand factor in platelet-derived FVIII gene therapy for murine hemophilia A in the presence of inhibitory antibodies. J Thromb Haemost 13:1301-9
Chen, Yingyu; Schroeder, Jocelyn A; Kuether, Erin L et al. (2014) Platelet gene therapy by lentiviral gene delivery to hematopoietic stem cells restores hemostasis and induces humoral immune tolerance in FIX(null) mice. Mol Ther 22:169-77

Showing the most recent 10 out of 24 publications