scholarly journals 230. Generation of an Optimized Lentiviral Vector Encoding a High-Expression fVIII Transgene for Gene Therapy of Hemophilia A

2010 ◽  
Vol 18 ◽  
pp. S88
Gene Therapy ◽  
2012 ◽  
Vol 20 (6) ◽  
pp. 607-615 ◽  
Author(s):  
J M Johnston ◽  
G Denning ◽  
C B Doering ◽  
H T Spencer

Blood ◽  
2011 ◽  
Vol 117 (3) ◽  
pp. 798-807 ◽  
Author(s):  
Natalie J. Ward ◽  
Suzanne M. K. Buckley ◽  
Simon N. Waddington ◽  
Thierry VandenDriessche ◽  
Marinee K. L. Chuah ◽  
...  

Abstract Gene therapy for hemophilia A would be facilitated by development of smaller expression cassettes encoding factor VIII (FVIII), which demonstrate improved biosynthesis and/or enhanced biologic properties. B domain deleted (BDD) FVIII retains full procoagulant function and is expressed at higher levels than wild-type FVIII. However, a partial BDD FVIII, leaving an N-terminal 226 amino acid stretch (N6), increases in vitro secretion of FVIII tenfold compared with BDD-FVIII. In this study, we tested various BDD constructs in the context of either wild-type or codon-optimized cDNA sequences expressed under control of the strong, ubiquitous Spleen Focus Forming Virus promoter within a self-inactivating HIV-based lentiviral vector. Transduced 293T cells in vitro demonstrated detectable FVIII activity. Hemophilic mice treated with lentiviral vectors showed expression of FVIII activity and phenotypic correction sustained over 250 days. Importantly, codon-optimized constructs achieved an unprecedented 29- to 44-fold increase in expression, yielding more than 200% normal human FVIII levels. Addition of B domain sequences to BDD-FVIII did not significantly increase in vivo expression. These significant findings demonstrate that shorter FVIII constructs that can be more easily accommodated in viral vectors can result in increased therapeutic efficacy and may deliver effective gene therapy for hemophilia A.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3544-3544
Author(s):  
Nadia Sutherland ◽  
Kerry L Dooriss ◽  
David A McCarty ◽  
Christopher B Doering ◽  
H. Trent Spencer

Abstract Hemophilia A is an X-linked gene disorder that results in a deficiency of circulating coagulation factor VIII (fVIII) and may be ameliorated by only modest amounts of circulating protein, which makes it a logical candidate for gene therapy. Due to the potential risk of insertional mutagenesis from oncoretroviral-mediated gene therapy, cell-specific expression of transgenes using self-inactivating viral vectors may provide a safer gene therapy approach for use in humans. Therefore, we constructed simian immunodeficiency virus (SIV)-based lentiviral vectors containing a 5′ long-terminal repeat (LTR) and 3′ LTR with self-inactivating U3 deletion, the bovine growth hormone polyA signal, a packaging signal (ψ), and a single internal ankyrin-1 or β-globin promoter, designated SIV-Ank and SIV-Bg, respectively. The minimal 314-bp ankyrin-1 promoter and 180-bp β-globin promoter flanked upstream by enhancing sequences, HS2, HS3, and HS4 (Hanawa et al., Hum Gene Ther, 2002) from the locus control region were cloned into the SIV vector backbone upstream from either enhanced green fluorescent protein (eGFP) or B-domain deleted porcine factor VIII (BDDpfVIII). The erythroid-specificity of each promoter was evaluated in vitro by measurement of either eGFP or fVIII expression following transduction of SIV-Ank and SIV-Bg constructs into both K562 myelogenous leukemic cells and 293T human embryonic kidney cells. GFP expression, as measured by flow cytometry, in transduced cells revealed that the ankyrin-1 and β-globin promoters are more active in K562 cells as compared to 293T cells. The β-globin promoter yielded higher mean fluorescent intensity values for GFP compared to the ankyrin-1 promoter at similar MOIs in K562 cells, suggesting stronger β-globin promoter activity in these cells. Transduction of cells with the SIV vector encoding BDDpfVIII driven by the β-globin promoter resulted in a 14-fold higher number of transcripts per DNA copy number in K562 cells compared to 293T cells, while cells transduced with the ankyrin-l promoter had only a 1.4-fold greater number of transcripts per DNA copy number. In addition, SIV-Bg-fVIII-modified K562 cells produced a 5.2-fold greater number of transcripts per DNA copy number than SIV-Ank- fVIII-modified cells. To evaluate the usefulness of these vectors for in vivo expression of BDDpfVIII, hemophilia A mice (exon 16 knockout) were conditioned with 11 Gy total body irradiation and transplanted with gene-modified Sca-1+ cells transduced with either SIV-Ank-fVIII, SIV-Ank-eGFP, SIV-Bg-fVIII, or SIV-Bg-eGFP. The expression of eGFP from donor red blood cells in recipient mice was approximately 8–12% using both the ankyrin-1 and β-globin promoter constructs. Mice that received cells transduced with SIVAnk- fVIII demonstrated therapeutic levels of plasma fVIII up to 0.5 units/mL (i.e. 50% normal human levels). However, fVIII expression decreased over time and real-time PCR analysis of peripheral blood cells confirmed the loss of detectable fVIII transgene by 6 weeks after transplantation, suggesting there was predominantly gene transfer into short-term repopulating hematopoietic cells. Mice transplanted with SIV-Bg-fVIII-modified hematopoietic stem cells demonstrated a similar rise and fall of fVIII expression within the first 4 weeks after transplantation, and showed an increase in fVIII expression by 6 weeks. At 8 weeks post transplantation, fVIII levels greater than 300% normal human levels were observed. Red blood cell count, hemoglobin, and red blood cell morphology were normal despite the high level of expression of fVIII. Overall these data demonstrate the potential for therapeutic expression of factor VIII using a self-inactivating lentiviral vector containing an erythroid-specific internal promoter.


2009 ◽  
Vol 17 (7) ◽  
pp. 1145-1154 ◽  
Author(s):  
Christopher B Doering ◽  
Gabriela Denning ◽  
Kerry Dooriss ◽  
Bagirath Gangadharan ◽  
Jennifer M Johnston ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3251-3251
Author(s):  
Hideto Matsui ◽  
Masaru Shibata ◽  
Brian Brown ◽  
Andrea Labelle ◽  
Carol Hegadorn ◽  
...  

Abstract Hemophilia is an excellent candidate disorder for the use of gene therapy as a treatment modality. However, significant obstacles have been encountered with systemic delivery of viral vectors that have prevented sustained expression of the therapeutic protein. Investigation of alternative gene therapy strategies for hemophilia that enhance safety and facilitate long-term, therapeutic levels of the transgene product is imperative. In this study, we evaluated an ex vivo gene therapy strategy for hemophilia A. Circulating endothelial cell progenitors (blood outgrowth endothelial cells - BOECs) were isolated from canine and mouse blood and transduced with a third generation self-inactivating lentiviral vector encoding the canine FVIII transgene under the transcriptional control of either the CMV promoter or an endothelial cell-specific regulatory element. Transduced BOECs were injected either intravenously (IV) or subcutaneously mixed with Matrigel (SC+Matrigel) into NOD/SCID mice. Canine FVIII antigen levels were assayed at weekly intervals using an Asserachrom VIII:Ag ELISA that detects canine FVIII against a background of normal murine FVIII levels in the NOD/SCID mice. The mean FVIII antigen levels in mice injected with BOECs at 3 weeks following treatment were 37.5 mU/mL and 105.8mU/mL, for IV and SC+Matrigel administration, respectively. These FVIII antigen levels were sustained up to 12 weeks at therapeutic levels (21.3mU/mL and 21.7mU/mL, for IV and SC+Matrigel administration respectively). To evaluate if the observed loss of FVIII expression by 12 weeks post-treatment resulted from transcriptional silencing of the viral promoter, the CMV promoter was replaced with the endothelial cell-specific thrombomodulin (TM) promoter and transduced BOECs were implanted SC with Matrigel. In contrast to results from the CMV-regulated transgene, sustained therapeutic levels of FVIII have been documented for the duration of the study with the TM-regulated construct (34.3 mU/mL at 3 weeks and 22.5 mU/mL at 20 weeks) Immunostaining at 18 weeks after SC implantation of the transduced BOECs, shows that these cells still express FVIII and von Willebrand Factor. Biodistribution analysis by flow cytometry and quantitative PCR demonstrated that SC-implanted BOECs were retained inside the scaffold and were not detected at any other anatomic site. These results indicate that genetically-modified endothelial progenitors implanted in a SC scaffold can provide sustained therapeutic levels of FVIII and are a promising safe delivery vehicle for gene therapy of hemophilia. Currently, these engineered cells have been implanted into immunocompetant mice and FVIII levels are being assessed.


2018 ◽  
Vol 29 (10) ◽  
pp. 1183-1201 ◽  
Author(s):  
Christopher B. Doering ◽  
Gabriela Denning ◽  
Jordan E. Shields ◽  
Eli J. Fine ◽  
Ernest T. Parker ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4210-4210
Author(s):  
Harrison C. Brown ◽  
Jordan E Shields ◽  
Shangzhen Zhou ◽  
J. Fraser Wright ◽  
H. Trent Spencer ◽  
...  

Abstract Preclinical and, more recently, clinical data support the feasibility and safety of recombinant adeno-associated viral vectors (rAAV) in gene therapy applications. Despite several clinical trials of rAAV-based gene transfer for hemophilia B, a unique set of obstacles impede the development of a similar approach for hemophilia A. These include 1) inefficient biosynthesis of human coagulation factor VIII (fVIII), 2) limited packaging capacity of rAAV (4.9kb) for the large B domain-deleted (BDD) fVIII transgene (4.5 kb), 3) humoral immune responses to the transgene product and 4) dose limitations imposed by capsid mediated cytotoxic immunity. Our laboratory has developed and validated bioengineered fVIII molecules that are biosynthesized more efficiently than similar BDD human fVIII molecules due to superior post-translational transit through the ER/golgi/secretory pathway. In the current study, we incorporated one of these constructs, previously designated HP47 (Doering et al. 2004 J Biol Chem. 279:8) and now termed ET-3, into an AAV-based gene transfer approach similar to that previous tested for patients with hemophilia B. Briefly, a rAAV2/8 vector encoding the ET-3 transgene under the control of a liver-specific promoter comprised of an Apo E hepatic control region (HCR) and the α-1 antitrypsin enhancer/promoter (HAAT), flanked by AAV inverted terminal repeats (ITRs), was constructed and designated rAAV-HCR-ET3. In prior studies, we demonstrated that recombinant ET-3 displays i) 100-fold more efficient biosynthesis, ii) 2 – 3-fold higher specific activity, and iii) 3-fold slower decay compared to BDD human fVIII. However, the vector genome size of rAAV-HCR-ET3, 5.9kb, exceeds the packaging capacity of rAAV by 20%. To address this concern, molecular analysis of rAAV-HCR-ET3 was performed and showed packaging primarily of 5' or 3' truncated vector genomes (vg). Further quantitative (q) PCR of vg using a series of primers pairs spanning the vector sequence showed 5-fold fewer copies of the termini than the center, which is indicative of terminally truncated vg. Thus, special consideration must be taken when using qPCR-based methods alone to titer vector preparations. Our data support a model whereby cells are infected by multiple truncated but overlapping vg, which then reassemble at some frequency to form complete, functional fVIII transgenes and/or RNA transcripts. In the present study, adult hemophilia A mice were administered a single peripheral vein injection of rAAV-HCR-ET3 at doses ranging from 3.9e10 vector particles (vp) /kg to 2.0e13 vp/kg as determined by viral protein concentration. These stated doses are 5 – 25-fold higher than would have been assigned using qPCR and expressed as vg/kg as opposed to vp/kg. At the conclusion of the study (11 - 50 weeks post-infusion), correction of fVIII deficiency to curative levels of fVIII (>0.4 U/mL in circulating plasma) was achieved at vector doses as low as 6.25e11 vp/kg, and partial correction (>0.01 U/mL) was seen at doses as low as 1.6e11 vp/kg. Tail transection bleeding assay demonstrated correction of the bleeding phenotype out to as long as 50 weeks after rAAV infusion. While gene transfer and phenotypic correction was achieved in both male and female mice, male mice receiving 2.5e12 vp/kg showed 9-fold greater circulating fVIII levels than female mice receiving the same dose, which is consistent with previous reports. Taken together, these data suggest significant benefit of the bioengineered high expression fVIII transgene (ET-3) in the context of liver-directed rAAV delivery at low viral vector doses, which if based on qPCR, could be stated as low as 1e10 vg/kg and even that includes mostly incomplete vg. Recent data suggest that reducing the size of the rAAV-fVIII vg through the incorporation of shorter DNA control elements and smaller fVIII transgenes can increase the vector potency (McIntosh et al. 2013 Blood 121:17). Despite its large vg size, rAAV-HCR-ET3 achieved curative levels of fVIII activity at vector doses comparable to those recently reported for smaller vectors. Therefore, it appears likely that further enhancement of the rAAV-bioengineered high expression fVIII gene transfer approach will be possible through incorporation of smaller genetic elements that increase the frequency of functional ET-3 gene transfer in the context of hemophilia A gene therapy. Disclosures: Spencer: Expression Therapeutics, LLC: Equity Ownership. Doering:Expression Therapeutics, LLC: Equity Ownership.


Sign in / Sign up

Export Citation Format

Share Document