scholarly journals Hematopoietic Stem Cells Encoding Porcine Factor VIII Induce Pro-coagulant Activity in Hemophilia A Mice with Pre-existing Factor VIII Immunity

2007 ◽  
Vol 15 (6) ◽  
pp. 1093-1099 ◽  
Author(s):  
Christopher B Doering ◽  
Bagirath Gangadharan ◽  
Hillary Z Dukart ◽  
H Trent Spencer
Blood ◽  
2008 ◽  
Vol 112 (7) ◽  
pp. 2713-2721 ◽  
Author(s):  
Qizhen Shi ◽  
Scot A. Fahs ◽  
David A. Wilcox ◽  
Erin L. Kuether ◽  
Patricia A. Morateck ◽  
...  

Abstract Although genetic induction of factor VIII (FVIII) expression in platelets can restore hemostasis in hemophilia A mice, this approach has not been studied in the clinical setting of preexisting FVIII inhibitory antibodies to determine whether such antibodies would affect therapeutic engraftment. We generated a line of transgenic mice (2bF8) that express FVIII only in platelets using the platelet-specific αIIb promoter and bred this 2bF8 transgene into a FVIIInull background. Bone marrow (BM) from heterozygous 2bF8 transgenic (2bF8tg+/−) mice was transplanted into immunized FVIIInull mice after lethal or sublethal irradiation. After BM reconstitution, 85% of recipients survived tail clipping when the 1100-cGy (myeloablative) regimen was used, 85.7% of recipients survived when 660-cGy (nonmyeloablative) regimens were used, and 60% of recipients survived when the recipients were conditioned with 440 cGy. Our further studies showed that transplantation with 1% to 5% 2bF8tg+/− BM cells still improved hemostasis in hemophilia A mice with inhibitors. These results demonstrate that the presence of FVIII-specific immunity in recipients does not negate engraftment of 2bF8 genetically modified hematopoietic stem cells, and transplantation of these hematopoietic stem cells can efficiently restore hemostasis to hemophilic mice with preexisting inhibitory antibodies under either myeloablative or nonmyeloablative regimens.


Blood ◽  
2009 ◽  
Vol 114 (3) ◽  
pp. 526-534 ◽  
Author(s):  
Ali Ramezani ◽  
Robert G. Hawley

Abstract Insertional mutagenesis by retroviral vectors is a major impediment to the clinical application of hematopoietic stem cell gene transfer for the treatment of hematologic disorders. We recently developed an insulated self-inactivating gammaretroviral vector, RMSinOFB, which uses a novel enhancer-blocking element that significantly decreases genotoxicity of retroviral integration. In this study, we used the RMSinOFB vector to evaluate the efficacy of a newly bioengineered factor VIII (fVIII) variant (efVIII)—containing a combination of A1 domain point mutations (L303E/F309S) and an extended partial B domain for improved secretion plus A2 domain mutations (R484A/R489A/P492A) for reduced immunogenicity—toward successful treatment of murine hemophilia A. In cell lines, efVIII was secreted at up to 6-fold higher levels than an L303E/F309S A1 domain–only fVIII variant (sfVIIIΔB). Most important, when compared with a conventional gammaretroviral vector expressing sfVIIIΔB, lower doses of RMSin-efVIII-OFB–transduced hematopoietic stem cells were needed to generate comparable curative fVIII levels in hemophilia A BALB/c mice after reduced-intensity total body irradiation or nonmyeloablative chemotherapy conditioning regimens. These data suggest that the safety-augmented RMSin-efVIII-OFB platform represents an encouraging step in the development of a clinically appropriate gene addition therapy for hemophilia A.


2010 ◽  
Vol 12 (4) ◽  
pp. 333-344 ◽  
Author(s):  
Lucienne M. Ide ◽  
Neal N. Iwakoshi ◽  
Bagirath Gangadharan ◽  
Shawn Jobe ◽  
Robert Moot ◽  
...  

2012 ◽  
Vol 01 (S1) ◽  
Author(s):  
Philip M. Zakas ◽  
H. Trent Spencer ◽  
Christopher B. Doering

Author(s):  
T. Tonn ◽  
S. Becker ◽  
C. Herder ◽  
M. Grez ◽  
E. Seifried

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 691-691 ◽  
Author(s):  
Lily M. Du ◽  
Timothy C. Nichols ◽  
Sandra L. Haberichter ◽  
Paula M. Jacobi ◽  
Eric S. Jensen ◽  
...  

Abstract Abstract 691 Introduction: The goal of our study was to develop a clinically relevant strategy for platelet-targeted gene therapy of the commonly inherited bleeding disorder, Hemophilia A. We hypothesized that adult dogs (25 kg) affected with Hemophilia A could serve as a relevant “large animal” model to test if FVIII could be synthesized and sequestered within platelets derived from lentivirus-transduced hematopoietic stem cells. This approach is novel because it should permit the regulated release of FVIII from activated platelet progeny directly at the wound site as a physiological hemostatic response to bleeding challenges. Methods: cG-CSF/cSCF mobilized peripheral blood stem cells (PBSC) were immuno-selected for CD34 antigen from an apheresis product, transduced with a lentivirus vector under the transcriptional control of platelet-specific integrin αaIIb gene promoter driving expression of human BDD-FVIII. The PBSC (3 × 10 6/kg) were then autologously transplanted into animals that were preconditioned with Bulsulfan (5-10 mg/kg i.v.). After transplant, the dogs received oral cyclosporine to maintain levels at 200–400 ng/ml for 90 days and MMF at 8mg/kg for 35 days. Results: Two transplant recipients underwent periodic testing for incorporation and expression of the FVIII transgene as well as immune tolerance and phenotypic correction of Hemophilia A. PCR analysis detected the lentivirus vector within genomic DNA isolated from circulating peripheral blood leukocytes for more than one year after transplant. Immunofluorescence confocal microscopy showed synthesis of FVIII within tissue cultured canine megakaryocytes and circulating peripheral blood platelets. Chromogenic analysis of platelets isolated from transplanted dogs demonstrated the presence of a biologically active form of FVIII (FVIII:C) at approximately 5 mU/ml/1×108 platelets from 20 weeks through greater than one year after PBSC transplant. In contrast, FVIII:C was not detected within the plasma of these animals. This result coupled with the immunomodulation drugs may help to explain why the dogs failed to develop inhibitory antibodies to human FVIII. Following successful gene transfer and engraftment, both animals showed signs of clinical improvement of Hemophilia A: one dog has had one bleed per year for 2 years (vs expected 5–6/year) and the second dog has had no bleeds for 8 months following transplantation. In addition, one dog showed significant recovery from prolonged (months) history of gastrointestinal bleeding. These data are consistent with our previous results demonstrating synthesis, trafficking and storage of FVIII within αa-granules of human megakaryoyctes in vitro and platelets of the murine “small animal” model for Hemophilia A. Conclusions: This outcome raises the possibility of developing a protocol for delivering a locally inducible secretory pool of FVIII in platelets of patients with Hemophilia A following autologous transplantation of FVIII-transduced hematopoietic stem cells. Disclosures: Montgomery: GTI Diagnostics: Consultancy; Baxter: Consultancy; AstraZeneca: Consultancy; Bayer: Research Funding; CSL Behring: Membership on an entity's Board of Directors or advisory committees. Wilcox:Amgen, INC: Research Reagents, canine growth factors: cG-CSF and cSCF.


Sign in / Sign up

Export Citation Format

Share Document