The goal of this study is to develop strategies to reduce/eliminate inhibitory antibodies in hemophilia A patients following repeated factor VIII infusion or gene therapy. Formation of inhibitory antibodies is a major problem in protein replacement therapy and expected to be a problem in gene therapy for hemophilia A patients. Approximately 25-50% of hemophilic patients develop anti-FVIII antibodies after repeated infusion of recombinant or pooled hFVIII protein. Following successful transfer of the factor VIII gene into the livers of hemophilia A mice by hydrodynamic-based nonviral gene transfer of a liver-specific, high-expressing FVIII plasmid, resulting in Supra-physiological levels of FVIII. We observed a robust immune response against FVIII two weeks post-treatment. This led to complete inhibition of circulating FVIII activity. This inhibitory response was mediated by a Th2-dominant human immune response. This inhibitory response completely ablated FVIII activity despite continued (essentially lifelong) FVIII production. This animal model is an ideal system for developing strategies to ameliorate the immune responses against FVIII. Recent data have demonstrated a critical role that CD4+CD25* regulatory T (Treg) cells in the regulation and suppression of autoimmune and alloimmune responses. Clinical trials of Treg infusion in BMT patients to prevent graft-versus-host disease (GVHD) are under way. Furthermore, Foxp3 which encodes the transcription factor Scurfin, is a master regulatory gene for the development and function of CD4+CD25+ regulatory T cells. In mice, ex vivo retroviral gene transfer of Foxp3 can convert peripheral CD25-CD45RO-CD4+ naive T cells into a regulatory T cell phenotype similar to CD4+CD25+ regulatory T cells. In addition, Treg type 1 cells (Tr1 cells) which can be induced by specific antigen and IL-10 have also been shown to maintain peripheral tolerance. We anticipate that an effective method to produce FVIII-specific CD4+CD25+ Treg cells (and/or Tr1) cells could provide a novel strategy to eliminate existing inhibitory antibodies and potentially induce long-term tolerance for hemophilia A patients with inhibitors. In this proposal we will test the hypotheses that: 1) Overexpression of Foxp3 in hemophilia A mice will modulate the immune responses against FVIII following gene transfer; 2) Lentiviral-based Foxp3 expression within FVIII-specific CD4+ T cells will induce CD4+CD25+Foxp3+ Treg cells. Further we predict that transplantation of such FVIII-specific Treg cells into hemophilia A mice will induce tolerance against FVIII; and 3) Lentiviral-based Foxp3 expression within FVIII-specific CD4+ T cells isolated from human hemophilia A patients with inhibitors can be used to generate FVIII-specific CD4+CD25+Foxp3+ Treg cells. This cell population could potentially be utilized in the future for cellular therapy in this disease setting.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL082600-03
Application #
7277653
Study Section
Special Emphasis Panel (ZHL1-CSR-D (S1))
Program Officer
Link, Rebecca P
Project Start
2005-09-27
Project End
2009-08-31
Budget Start
2007-09-01
Budget End
2008-08-31
Support Year
3
Fiscal Year
2007
Total Cost
$428,084
Indirect Cost
Name
Seattle Children's Hospital
Department
Type
DUNS #
048682157
City
Seattle
State
WA
Country
United States
Zip Code
98105
Liu, Chao Lien; Lyle, Meghan J; Shin, Simon C et al. (2016) The dataset from administration of single or combined immunomodulation agents to modulate anti-FVIII antibody responses in FVIII plasmid or protein primed hemophilia A mice. Data Brief 7:973-80
Liu, C L; Ye, P; Lin, J et al. (2014) Long-term tolerance to factor VIII is achieved by administration of interleukin-2/interleukin-2 monoclonal antibody complexes and low dosages of factor VIII. J Thromb Haemost 12:921-31
Liu, Chao-Lien; Ye, Peiqing; Yen, Benjamin C et al. (2011) In vivo expansion of regulatory T cells with IL-2/IL-2 mAb complexes prevents anti-factor VIII immune responses in hemophilia A mice treated with factor VIII plasmid-mediated gene therapy. Mol Ther 19:1511-20
Miao, C H (2011) Tilt balance towards regulation: evolving new strategy for treatment of hemophilia inhibitors. J Thromb Haemost 9:1521-3
Lin, C N; Kao, C Y; Miao, C H et al. (2010) Generation of a novel factor IX with augmented clotting activities in vitro and in vivo. J Thromb Haemost 8:1773-83
Miao, Carol H (2010) Immunomodulation for inhibitors in hemophilia A: the important role of Treg cells. Expert Rev Hematol 3:469-83
Peng, Baowei; Ye, Peiqing; Rawlings, David J et al. (2009) Anti-CD3 antibodies modulate anti-factor VIII immune responses in hemophilia A mice after factor VIII plasmid-mediated gene therapy. Blood 114:4373-82
Miao, Carol H; Harmeling, Benjamin R; Ziegler, Steven F et al. (2009) CD4+FOXP3+ regulatory T cells confer long-term regulation of factor VIII-specific immune responses in plasmid-mediated gene therapy-treated hemophilia mice. Blood 114:4034-44
Peng, Baowei; Ye, Peiqing; Blazar, Bruce R et al. (2008) Transient blockade of the inducible costimulator pathway generates long-term tolerance to factor VIII after nonviral gene transfer into hemophilia A mice. Blood 112:1662-72
Shen, Z P; Brayman, A A; Chen, L et al. (2008) Ultrasound with microbubbles enhances gene expression of plasmid DNA in the liver via intraportal delivery. Gene Ther 15:1147-55

Showing the most recent 10 out of 11 publications