scholarly journals Correction of murine hemophilia A following nonmyeloablative transplantation of hematopoietic stem cells engineered to encode an enhanced human factor VIII variant using a safety-augmented retroviral vector

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.

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 ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 292-292
Author(s):  
Boris Calmels ◽  
Cole Ferguson ◽  
Rima Adler ◽  
Stephanie Sellers ◽  
Mikko Laukkanen ◽  
...  

Abstract Until recently, the risk of insertional mutagenesis using retroviral vectors for gene therapy has been estimated to be low. Owing to reports of proto-oncogenes activation in mice and humans, this estimation is being re-evaluated in regards to the nature of susceptible loci and the number of genetic hits needed for oncogenesis. Separating the impact of overexpressing a growth-altering transgene from the insertional events themselves is particularly important to address in long-term repopulating hematopoietic stem cells. We here report a high frequency of proviral insertions at the MDS1-EVI1 locus in the engrafted gene-modified hematopoiesis of non-human primates. We have recovered vector-genome junction sequences from mature granulocytes and mononuclear cells of 22 rhesus macaques that were transplanted 6 months to 6 years previously with autologous mobilized CD34+ peripheral blood stem cells transduced with an amphotropic Moloney murine leukemia virus-derived vector containing a neomycin-resistance marker gene. Using a modified LAM-PCR method, we have retrieved and analyzed 702 independent integration sites that mapped to a unique genomic location. While several transcription units harbor two or three proviral insertions, we have identified, in 9 animals, an unexpected 13 unique integration events within the two first introns of the MDS1 gene. MDS1 is adjacent to EVI1, a well-known retrovirally-activated zinc finger transcription factor in a number of murine leukemogenesis studies. We used insertion-specific primers to confirm that the fusion sequences between the MDS1 locus and the 5′-LTR of the vector were detectable in four of the animals for which we had the longest follow-up (4 to 6 years). The fusion sequence was present both in purified granulocytes and lymphocytes of one of the animal, and only in granulocytes in the other three animals. In order to determine if the cells carrying proviral insertions at the MDS1-EVI1 locus have a selective growth advantage, we performed quantitative PCR experiments with neomycin and MDS1/5′-LTR-specific probes. The two animals analyzed to date do not have any evidence of clonal expansion of the MDS1-targeted granulocyte population, and the number of retrovirally-transduced circulating cells remains stable 5 and 6 years after transplantation. As retroviral insertion upstream of a proto-oncogene may lead to its activation, we investigated MDS1-EVI1 expression by RT-PCR in neo+ CFU that harbor a proviral insertion at the MDS1 locus. The 8 colonies screened from two animals do not express any of the various MDS1-EVI1 transcripts, suggesting that the transcriptional regulation of the locus has not been altered. Our study suggest that the MDS1-EVI1 locus is particularly susceptible to retroviral integration but the competing hypothesis that proviral insertion within this region favors engraftment and long-term contributions to hematopoiesis will be difficult to eliminate, since specific insertions may have favored engraftment of such clones. It is however important to mention that the long-term follow-up of these primates has revealed completely normal hematopoiesis and lack of any progression towards neoplasia. Systematic analysis of proviral insegration sites in this appropriate pre-clinical model is essential as it provides decisive information for risk assessment in the development of integrating vectors, as well as offering further insights into the mechanisms of retroviral insertion.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2198-2198
Author(s):  
Lily M. Du ◽  
Helen W.G. Franck ◽  
Elizabeth P. Merricks ◽  
Paquita Nurden ◽  
Eric S. Jensen ◽  
...  

Abstract Abstract 2198 Introduction: The goal of our study was to develop a clinically relevant strategy for platelet-targeted gene therapy of the 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 Factor VIII (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 a site of vascular injury as a physiological hemostatic response to a severe intermittent bleeding challenge. 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 αIIb gene promoter driving expression of human BDD-FVIII. The PBSC (2 × 106/kg) were then autologously transplanted (Tx) 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 8 mg/kg for 35 days. Results: Three Tx recipients underwent periodic testing for incorporation and expression of the FVIII transgene as well as immune tolerance and phenotypic correction of Hemophilia A. LAM PCR analysis detected and localized the lentivirus vector within genomic DNA isolated from circulating peripheral blood leukocytes from each animal. Immunofluorescence confocal microscopy detected FVIII within a subset of circulating peripheral blood platelets. Immune electron microscopic analysis revealed that the FVIII had been trafficked and stored within the platelet secretory α-granules. Chromogenic analysis of platelets isolated from dogs demonstrated that FVIII was present in its biologically active form (FVIII:C) at approximately 5 mU/ml/1×108 platelets from 20 weeks through greater than two years after PBSC Tx. In contrast, FVIII:C was not detected within the plasma of these animals. This result coupled with the use immunomodulation drugs may help to explain why the dogs remained tolerant of human FVIII as indicated by our inability to detect inhibitory antibodies to FVIII. Following successful gene transfer and PBSC engraftment, all animals showed signs of clinical improvement of Hemophilia A: the longest Tx recipient had one bleed per year for 2 years (vs expected 5 – 6/year), the second dog has had no bleeds for twenty months following PBSC Tx and the third animal had no bleeding episodes for first seven months post Tx. In addition, the first Tx dog showed significant recovery from prolonged (months) history of chronic gastrointestinal bleeding. Remarkably, all three animals displayed improved whole blood clotting times. These data are consistent with our previous results demonstrating synthesis, trafficking, storage and regulated release of FVIII from α-granules of human megakaryoyctes in vitro and platelets of the murine “small animal” model for Hemophilia A. Conclusions: The outcome of this work demonstrates a feasible strategy for controlling severe bleeding episodes within patients with Hemophilia A by providing a locally inducible secretory pool of FVIII in platelets derived from an autologous Tx of FVIII-transduced PBSC. Disclosures: No relevant conflicts of interest to declare.


2007 ◽  
Vol 15 (6) ◽  
pp. 1093-1099 ◽  
Author(s):  
Christopher B Doering ◽  
Bagirath Gangadharan ◽  
Hillary Z Dukart ◽  
H Trent Spencer

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

Blood ◽  
2006 ◽  
Vol 107 (9) ◽  
pp. 3772-3778 ◽  
Author(s):  
André Larochelle ◽  
Allen Krouse ◽  
Mark Metzger ◽  
Donald Orlic ◽  
Robert E. Donahue ◽  
...  

AMD3100, a bicyclam antagonist of the chemokine receptor CXCR4, has been shown to induce rapid mobilization of CD34+ hematopoietic cells in mice, dogs, and humans, offering an alternative to G-CSF mobilization of peripheral-blood hematopoietic stem cells. In this study, AMD3100-mobilized CD34+ cells were phenotypically analyzed, marked with NeoR-containing retroviral vectors, and subsequently transplanted into myeloablated rhesus macaques. We show engraftment of transduced AMD3100-mobilized CD34+ cells with NeoR gene marked myeloid and lymphoid cells up to 32 months after transplantation, demonstrating the ability of AMD3100 to mobilize true long-term repopulating hematopoietic stem cells. More AMD3100-mobilized CD34+ cells are in the G1 phase of the cell cycle and more cells express CXCR4 and VLA-4 compared with G-CSF-mobilized CD34+ cells. In vivo gene marking levels obtained with AMD3100-mobilized CD34+ cells were better than those obtained using CD34+ cells mobilized with G-CSF alone. Overall, these results indicate that AMD3100 mobilizes a population of hematopoietic stem cells with intrinsic characteristics different from those of hematopoietic stem cells mobilized with G-CSF, suggesting fundamental differences in the mechanism of AMD3100-mediated and G-CSF-mediated hematopoietic stem cell mobilization. Thus, AMD3100-mobilized CD34+ cells represent an alternative source of hematopoietic stem cells for clinical stem cell transplantation and genetic manipulation with integrating retroviral vectors.


Sign in / Sign up

Export Citation Format

Share Document