Hemophilia A is caused by a deficiency of blood coagulation factor VIII (FVIII) and has been widely discussed as a candidate for gene therapy. While the natural canine model of hemophilia A has been valuable for the development of FVIII pharmaceutical products, the use of hemophiliac dogs for gene therapy studies has several limitations such as expense and the long canine generation time. The recent creation of two strains of FVIII-deficient mice provides the first small animal model of hemophilia A. Treatment of hemophiliac mice of both genotypes with potent, human FVIII-encoding adenoviral vectors resulted in expression of biologically active human FVIII at levels, which declined, but remained above the human therapeutic range for over 9 months. The duration of expression and FVIII plasma levels achieved were similar in both hemophiliac mouse strains. Treated mice readily survived tail clipping with minimal blood loss, thus showing phenotypic correction of murine hemophilia A by in vivo gene therapy.

HEMOPHILIA A, A COMMON bleeding disorder affecting one in 5 to 10,000 males in all populations,1 is caused by a deficiency of blood coagulation factor VIII (FVIII). Hemophilia A is categorized into severe, moderate, or mild forms, with over half of the patients manifesting the severe disease.1Severe hemophiliacs, defined as having less than 1% of normal FVIII levels, suffer from episodes of spontaneous and prolonged bleeding into muscles, internal organs, and joints and frequently develop a disabling arthropathy.1 Current treatment is directed toward replacing the missing clotting factor in response to bleeding crises with infusions of plasma-derived or recombinant FVIII.2While prophylactic treatment of hemophilia A has been shown to reduce the frequency and severity of bleeding, such therapy is limited by the availability and high cost of purified FVIII, and the short half-life of FVIII in vivo.2 

Somatic cell gene therapy, which would provide constant blood levels of FVIII, would be a significant treatment improvement.3However, expression of FVIII is problematic. The accumulation of FVIII mRNA is inhibited by sequences present in the coding region,4-8 and secretion of FVIII is inefficient.9-11 In addition, the FVIII protein must be secreted directly into the vasculature4,12,13 where the protein is stabilized by formation of a complex with von Willebrand factor (vWF).4 

Previous studies in gene therapy for hemophilia A had used ex vivo retroviral transduction approaches. In general, expression of FVIII protein in genetically modified cells in vitro was low5,8,14-17 and undetectable in vivo.15,16Short-term expression of human FVIII in mice was achieved via ex vivo gene transfer strategies,12,13 although the necessity for ex vivo cell manipulation and reimplantation is a limitation to the application of gene therapy in large animal models and humans.

Considerable progress has been made recently in the development of adenoviral-mediated in vivo gene therapy of hemophilia A.18-21 Adenoviral vectors are an efficient system for in vivo FVIII gene delivery because a peripheral vein injection in mice18,22,23 and dogs21 results in efficient transduction of hepatocytes, cells capable of secreting FVIII directly into the blood.4 A vector encoding an albumin-promoted FVIII cDNA resulted in high-level, liver-specific expression of human FVIII in normal adult mice,19 which was sustained for over 5 months at levels fourfold above the human therapeutic range.20 Furthermore, administration of a potent FVIII-encoding adenoviral vector to FVIII-deficient hemophiliac dogs, a well characterized, large hemophilia A animal model,24,25resulted in high-level human FVIII expression and complete correction of the coagulation deficiency.21 However, phenotypic correction in the treated dogs was transient, as the animals developed a strong antibody response directed to the human protein.21Therefore, the canine hemophilia A model is not amenable to long-term expression of human FVIII.

The recent generation of FVIII-deficient mice, by gene disruption techniques, provides the first small animal model of hemophilia A.26 Two distinct hemophiliac genotypes were developed by insertion of a neomycin expression cassette into exon 16 or exon 17 of the murine FVIII gene.26 Similar mutations in humans are known to cause severe hemophilia A.27 Affected mice of both genotypes have FVIII activity levels less than 1% of normal and display lethal bleeding after trauma.26 Therefore, the phenotype of these mice is similar to that of human hemophiliacs.1 The murine hemophilia A model provides a novel tool for the development of hemophilia gene therapy.

In this work, we evaluated adenoviral vectors for the treatment of murine hemophilia A. Studies were conducted in both hemophiliac mouse strains and used vectors prepared in two distinct adenoviral vector backbones. The level and duration of FVIII expression, phenotypic correction, and the antigenicity of human FVIII in the mouse model was assessed. We showed expression of functional human FVIII with levels, which declined, but remained above therapeutic for at least 9 months and achieved complete phenotypic correction.

Construction of recombinant adenoviruses.

The recombinant adenovirus encoding human FVIII, Av1H8101 (previously named Av1ALAPH81)19 has been described.19 The vector was checked for the presence of replication-competent adenovirus contamination by polymerase chain reaction (PCR) directed at E1a sequences,28 and all vector preparations contained less than 10 plaque-forming units (pfu) of E1a-containing vector per 108 pfu. The β-galactosidase-encoding vector, Av3nBg, has been described.29 Av3H8101 was generated by cotransfection of the plasmid, pAvALAPH81,19 and viral DNA from the vector, Av3nBg,29 into AE1-2a cells29 as described.29 The Av1H8101 vector concentration was determined by spectrophotometric analysis30 and by plaque assay31 on 293 cells. The particle to pfu ratio was 80. Av3H8101 vector concentrations were determined by spectrophotometric analysis.30 Titers are given as particles per milliliter.

Murine hemophilia A pathophysiology, animal breeding, genotyping, and manipulations.

Breeding colonies of both genotypes of FVIII knockout mice have been established.26 Initially, carrier females were bred with normal C57BL/6 male mice. The genotypes of 4-week-old pups were determined by PCR analysis of genomic DNA isolated from tail clips. Briefly, mice were lightly anesthetized with isofluorane, and 1-cm sections of the tail were collected and frozen on dry ice. The tails were immediately cauterized to stop bleeding. Notably, approximately 30% of affected mice did not survive tail clipping with cautery. DNA was isolated from the tail clips using the QIAmp Tissue Kit (Qiagen, Chatsworth, CA) following the suggested protocol. PCR analysis was performed as described26,32 using 400 ng of each genomic DNA and primers specific for the inserted neo gene, exon 16 or exon 17.32 Subsequently, hemizygous affected males and homozygous affected females were mated.

The hemophiliac mice are occasionally anemic and bleed severely from scratches and routine procedures such as ear tagging. Furthermore, the knockout mice suffer from joint bleeds, subcutaneous bleeding, and spontaneous death indicating a similarity to the pathophysiology of human hemophilia A.1 Surprisingly, however, hemophiliac females of both genotypes survive pregnancy, birth, and the nursing of pups32 (S.C. unpublished). Breeding of affected males and females has eliminated the need to genotype the pups by PCR analysis of genomic DNA.26,32 All mice were confirmed FVIII-deficient by analysis of plasma levels of functional FVIII using the Coatest bioassay (see below).

Mice were ear tagged for identification and housed in cages of five to six each. Occasionally, mice would bleed profusely from ear tagging, scratches, and injuries incurred from fighting. Topical thrombin (Thrombostat, Parke-Davis, Morris Plains, NJ) was applied to halt the bleeding. Approximately 10% of the affected mice died spontaneously.

The tail clip survival study involved lightly anesthetizing the mice and clipping 2-cm sections of the tail, without subsequent cauterization. After the procedure, mice were checked every 4 hours. The surviving mice at 24 hours after the procedure were recorded. Alternative assays to assess bleeding time and/or blood loss over time were performed before the tail clip survival study. However, such assays did not distinguish definitively between normal and hemophiliac mice. Similarly, assessment of the activated partial thromboplastin time (APTT) of mouse plasma samples did not distinguish normal from hemophiliac mice (unpublished data). All experiments involving mice adhered to protocols approved by the Institutional Animal Care and Use Committee in accordance with the Animal Welfare Act.

Adenoviral vector administration via tail vein injections and retroobital phlebotomy were performed as described.18 Mice injected with Av1H8101 received a dose of 4 × 1010particles per mouse, and mice injected with the Av3 vectors, Av3H8101 and Av3nBg received a dose of 6 × 1010 particles per mouse. These vector doses yielded equal liver transduction as determined by Southern analysis (data not shown). Topical thrombin (Thrombostat, Parke-Davis) was applied to the injection site to halt the bleeding after vector administration. Notably, the hemophiliac mice did not show any adverse effects from retroorbital bleeding.

FVIII assays, Southern blot, and RNAse protection analyses.

Biologically active human FVIII was measured using the Coatest chromogenic bioassay (Chromogenix, Mölndal, Sweden) as directed. Coatest measures the FVIII-dependent generation of factor Xa from factor X, with one unit defined as the amount of FVIII activity in 1 mL of pooled human plasma, 100 to 200 ng/mL.33 Pooled human plasma (George King Bio-Medical, Inc, Overland Park, KS) was used as the FVIII activity standard to generate a standard curve. Pooled mouse plasma collected from normal C57BL/6 mice served as the normal mouse FVIII activity positive control. When compared with human plasma, normal C57BL/6 plasma FVIII levels showed a median value of 2,500 mU/mL. Pooled plasma isolated from exon 17-disrupted affected mice served as the negative control (<1% of normal mouse levels) in each assay performed. FVIII activity values are reported as the mean value and the standard error of the mean.

The enzyme-linked immunosorbent assay (ELISA) assay, designed to measure human FVIII-specific antibodies, was performed as follows. Recombinant human FVIII (Hyland Division of Baxter Healthcare, Glendale, CA), 100 μL/well at 3 U/mL, in 0.05 mol/L carbonate-bicarbonate buffer pH 9.0, was incubated on 96-well Immunlon 1 pates (Dynatech, Chantilly, VA) at 4°C, overnight. The plate was washed one time with TBS (20 mmol/L Tris-Cl in 0.9% saline) and 200 μL of blocking buffer (0.17 mol/L H3BO3, 0.12 mol/L NaCl, and 0.5% bovine serum albumin [BSA]) was added for 5 hours at room temperature (RT). Plates were washed three times with TBS, and 100 μL mouse plasma samples, appropriately diluted in TBS plus 0.5% BSA, were added to each well and incubated overnight at 4°C. Plates were washed three times with TBS, and the detection antibody (100 μL), alkaline-phosphatase conjugated goat antimouse IgG (Southern Biotechnology Associates, Inc, Birmingham AL) diluted 1:3,000 in TBS with 0.5% BSA, was added to each well and incubated at RT for 2 hours. p-Nitrophenyl phosphate (P-NPP; Calbiochem, La Jolla, CA) 2 mg/mL, in buffer containing 0.1 mol/L glycine, 1 mmol/L MgCl2, 2 mmol/L ZnCl, pH 10.4, was added to each well (100 μL) and the absorbance was read at 405 nm using the Dynatech MR5000 (Dynatech) automated microplate ELISA reader. The concentration of anti-FVIII antibody was calculated from a standard curve using a monoclonal mouse antihuman FVIII antibody (MoAb) 413.34 The limit of sensitivity of the ELISA was 50 ng/mL for mouse plasma samples diluted 1:10.

DNA was isolated from mouse livers using the QIAmp Tissue Kit (Qiagen). A total of 10 μg of each DNA sample was digested with BamHI and subjected to Southern analysis.18 The probe, prepared by random oligonucleotide priming, contained FVIII cDNA sequences from +73 to +1,345.35,36 The copy number control standards were prepared by adding 600 pg and 60 pg of viral DNA, equivalent to 10 and 1 vector copies per cell, respectively, to 10 μg of control mouse liver genomic DNA and digesting with BamHI. No vector was detected in uninjected control mouse liver DNA (data not shown). The band intensities were quantitated with a Molecular Dynamics' PhosphorImager SF (Sunnydale, CA).

RNA was isolated from mouse livers using the RNAzole B (Tel-Test, Friendswood, TX) extraction method. RNAse protection analyses were performed using the RNAse Protection Kit II (Ambion, Austin, TX). For each sample, 20 to 50 μg of total cellular RNA were hybridized with an excess of a gel-purified RNA probe (see below), digested with the RNAse A/T1 solution provided with the kit diluted 1:100, processed as directed, and analyzed on an 8% polyacrylamide-8 mol/L urea gel (SequaGel, National Diagnostics, Atlanta, GA). 32P-labeled fragments from HpaII-digested pBR322 were used as the DNA size markers. The FVIII probe template, pGemSRpr,19 contains FVIII coding region sequences from (+9) to (+214) and was linearized with HindIII. The mouse glyceraldehyde-3-phosphodehydrogenase (GAPDH)-specific probe template was generated from the pTRI-GAPDH mouse plasmid (Ambion, Austin, TX) digested with StyI. All antisense RNA probes were synthesized with SP6 polymerase and α-32P-CTP (3,000 Ci/mmole, Amersham, Arlington Heights, IL).

Expression of functional human FVIII in hemophiliac mice.

The first generation, recombinant human FVIII adenoviral vector, Av1H8101 (previously referred to as Av1ALAPH81)19 contains a mouse albumin promoter, an intron from the human apoliprotein A1 gene, and a human B-domain deleted (BDD) FVIII cDNA. Absence of the B-domain has no effect on FVIII function, activity, or immunogenicity.11,37,38 The vector backbone was derived from adenovirus serotype 5 (Ad5) and is devoid of the E1 and E3 regions. A second human FVIII-encoding adenoviral vector, Av3H8101, was generated with the identical FVIII expression cassette and an Ad5 viral backbone with an additional deletion of the E2a region.29Attenuation of E2a gene expression has been reported to prolong transgene expression in some model systems.39-41 

A breeder colony of FVIII knockout mice has been established, and we confirmed that the affected mice exhibit FVIII activity levels less than 1% of normal and bleed acutely in response to trauma.26 To assess human FVIII expression in the hemophiliac mice, Av1H8101 and Av3H8101 were administered via tail vein injection to groups of five exon 17–disrupted mice (see Materials and Methods). Plasma levels of biologically active human FVIII were measured using the Coatest chromogenic bioassay. Before vector treatment, all mice showed FVIII plasma levels at less than 1% the amount detected in a normal, C57BL/6 mouse plasma sample, 2500 mU/mL. Two weeks after vector treatment, FVIII plasma levels were increased to mean values of 960 ± 68 and 946 ± 170 mU/mL in the Av1 and Av3 vector-treated groups, respectively. Human physiologic levels of FVIII are defined as 1,000 mU/mL, and therapeutic levels, the amount of FVIII necessary to convert a severe hemophiliac to a mild or moderate hemophiliac condition, are 50 mU/mL.33 Therefore, the mice treated with either the Av1 and Av3 vectors expressed human physiologic levels of biologically active FVIII, showing directly that adenoviral vector-mediated expression of FVIII resulted in secretion of functional FVIII protein. Furthermore, removal of the E2a region had no effect on vector transduction efficiency or transgene transcriptional activity (data not shown), indicating that the Av1 and Av3 vectors were equally efficacious.

Time course of human FVIII expression in the hemophiliac mice.

To assess the time course of human FVIII expression with Av1 and Av3 vectors in exon 17–disrupted hemophiliac mice, four animals were treated with Av1H8101 and eight were treated with Av3H8101. Plasma was obtained at the indicated times and assayed for FVIII activity (Fig 1A). The Av1 and Av3 vectors mediated expression of supraphysiologic levels of FVIII with peaks of 2,100 and 2,550 mU/mL, respectively, at 1 to 2 weeks postinjection. FVIII expression was maintained at levels above the human therapeutic range for at least 40 weeks. With both vectors, the decline in FVIII expression was greatest between 10 to 20 weeks with more stable expression observed in some animals thereafter (Fig 1A).

Fig. 1.

Time course of FVIII expression in hemophiliac mice. The adenoviral vectors Av1H8101 (4 × 1010 particles/mouse) or Av3H8101 (6 × 1010 particles/mouse) were administered via tail vein injection to groups of 4 or 8 exon 17–disrupted hemophiliac mice, respectively. These vector doses yielded equal liver transduction as determined by Southern analysis (data not shown). At the indicated time points, plasma samples were collected and FVIII biological activity was quantitated. (A) Mean plasma levels of biologically active FVIII. (•) Mice that received Av1H8101. (▪) Mice that received Av3H8101. Data are plotted as a mean value and the standard error of the mean at each time point. The dotted line represents the human therapeutic level of FVIII, 50 mU/mL.33 (B) FVIII plasma levels of individual Av3 vector-treated mice. One mouse (▪) died between 34 and 40 weeks.

Fig. 1.

Time course of FVIII expression in hemophiliac mice. The adenoviral vectors Av1H8101 (4 × 1010 particles/mouse) or Av3H8101 (6 × 1010 particles/mouse) were administered via tail vein injection to groups of 4 or 8 exon 17–disrupted hemophiliac mice, respectively. These vector doses yielded equal liver transduction as determined by Southern analysis (data not shown). At the indicated time points, plasma samples were collected and FVIII biological activity was quantitated. (A) Mean plasma levels of biologically active FVIII. (•) Mice that received Av1H8101. (▪) Mice that received Av3H8101. Data are plotted as a mean value and the standard error of the mean at each time point. The dotted line represents the human therapeutic level of FVIII, 50 mU/mL.33 (B) FVIII plasma levels of individual Av3 vector-treated mice. One mouse (▪) died between 34 and 40 weeks.

Close modal

Examination of the time course data from individual mice showed an initial decline between 10 to 20 weeks. Subsequently, FVIII levels plateaued in some animals for at least 40 weeks. Interestingly, some mice showed a sudden, sporadic decline in FVIII expression. To illustrate this observation, the FVIII expression levels of the individual mice that received the Av3 vector are plotted from weeks 12 to 40 (Fig 1B). One mouse of the four that received Av1 (data not shown) and three of the eight that received Av3 expressed constant levels of FVIII from weeks 12 to 40 (Fig 1B).

Comparison of FVIII expression in exon 16 and exon 17–disrupted hemophiliac mice.

Two separate genotypes of the FVIII knockout mice were generated by disruption of either exon 16 or exon 17.26,32 To show that long-term expression of human FVIII in the mice was not dependent on a specific murine FVIII mutation, groups of 12 exon 16– and 11 exon 17–disrupted mice were treated with Av3H8101. High level expression of biologically active FVIII was detected in both mouse genotypes with expression sustained for at least 24 weeks at levels well above the human therapeutic range (Fig 2). There was no difference in the level or duration of FVIII expression between the two genotypes.

Fig. 2.

Comparison of human FVIII expression in exon 16 and exon 17–disrupted hemophiliac mice. The adenoviral vector, Av3H8101 (6 × 1010 particles/mouse) was administered via tail vein injection to groups of 12 exon 16– or 11 exon 17–disrupted hemophiliac mice. At the indicated time points, plasma samples were collected and FVIII biological activity was quantitated. Mice that were expressing below less than 1% of normal FVIII levels (<25 mU/mL) were killed at 24 weeks. The FVIII levels in the remaining mice were assayed for an additional 16 weeks. (A) Mean plasma levels of biologically active FVIII. (•) Exon 16-disrupted mice. (▴) Exon 17–disrupted mice. Data are plotted as a mean value and the standard error of the mean at each time point. The dotted line represents the human therapeutic level of FVIII, 50 mU/mL.33 

Fig. 2.

Comparison of human FVIII expression in exon 16 and exon 17–disrupted hemophiliac mice. The adenoviral vector, Av3H8101 (6 × 1010 particles/mouse) was administered via tail vein injection to groups of 12 exon 16– or 11 exon 17–disrupted hemophiliac mice. At the indicated time points, plasma samples were collected and FVIII biological activity was quantitated. Mice that were expressing below less than 1% of normal FVIII levels (<25 mU/mL) were killed at 24 weeks. The FVIII levels in the remaining mice were assayed for an additional 16 weeks. (A) Mean plasma levels of biologically active FVIII. (•) Exon 16-disrupted mice. (▴) Exon 17–disrupted mice. Data are plotted as a mean value and the standard error of the mean at each time point. The dotted line represents the human therapeutic level of FVIII, 50 mU/mL.33 

Close modal

Examination of the time courses of FVIII expression for the individual mice showed the same pattern of stable expression previously shown in Fig 1B. At 40 weeks, four of the exon 16 mice, and four of the exon 17 mice displayed stable, sustained FVIII expression (data not shown). Notably, by 52 weeks, three exon 16 mice and one exon 17 mouse continued to display constant, stable FVIII plasma levels (data not shown).

Phenotypic correction of murine hemophilia A.

Mice expressing therapeutic levels of FVIII appeared phenotypically normal, as they were no longer anemic, did not die spontaneously, and did not bleed profusely from scratches or ear tagging. To show directly that expression of biologically active human FVIII in the treated mice resulted in correction of the bleeding defect, a tail clip survival study was performed (Table 1). Groups of exon 16– and exon 17–disrupted hemophiliac mice were treated with Av3H8101, a β-galactosidase-encoding adenoviral vector, Av3nBg,29 or were untreated. All of the Av3H8101-treated animals expressed therapeutic levels of FVIII 5 days after vector administration. On day 6, tail clips were performed on the treated and untreated hemophiliac mice, as well as normal, age-matched C57BL/6 mice. All of the normal C57BL/6 mice readily survived tail clipping with no evidence of distress. Only 5% to 30% of the untreated and Av3nBg-treated hemophiliac mice survived tail clipping and the survivors were moribund at 24 hours. In contrast, all mice that received Av3H8101 readily survived tail clipping with no evidence of distress. These data show phenotypic correction of murine hemophilia A.

Table 1.

Phenotypic Correction of Murine Hemophilia A

Mouse Strain Vector Survivors
C57BL/6  None 20/20  
Ex 16 KO   2/10  
 None  
Ex 17 KO  4/10-150 
Ex 16 KO   1/10-151 
 Av3nBg  
Ex 17 KO  0/10-150 
Ex 16 KO   11/11  
 Av3H8101  
Ex 17 KO  12/12 
Mouse Strain Vector Survivors
C57BL/6  None 20/20  
Ex 16 KO   2/10  
 None  
Ex 17 KO  4/10-150 
Ex 16 KO   1/10-151 
 Av3nBg  
Ex 17 KO  0/10-150 
Ex 16 KO   11/11  
 Av3H8101  
Ex 17 KO  12/12 

A group of 20 age-matched, normal C57BL/6 mice and groups of 10-12 exon 16–(Ex 16 KO) or exon 17–disrupted (Ex 17 KO) hemophiliac mice were treated with the indicated vectors (6 × 1010particles per mouse) or were untreated. FVIII plasma levels were measured before vector administration and 5 days after treatment. All mice treated with Av3H8101 showed expression of functional FVIII at levels well above therapeutic. On day 6, a 2-cm section of the mouse tail was clipped and the mouse survival rate recorded 24 hours later.

F0-150

One mouse died before tail clipping.

F0-151

Two mice died before tail clipping.

Assessment of vector persistence and antihuman FVIII antibody response.

To evaluate the mechanism by which FVIII expression was attenuated in some mice (see Fig 1), Av1H8101-treated exon 17–disrupted mice were killed 2 and 16 weeks after vector administration and liver DNA and RNA were analyzed (Fig 3). Southern analysis showed an average of six vector copies per cell at 2 weeks and 0.8 vector copies per cell at 16 weeks (Fig 3A). Despite the substantial drop in vector copy number, FVIII RNA levels decreased only twofold to threefold (Fig 3B). Furthermore, FVIII RNA levels did not parallel or predict FVIII protein expression in the plasma (Fig 3B). Therefore, loss of FVIII expression at 16 weeks in three of the four mice analyzed was not due to a complete loss of vector DNA or to complete inactivation of the albumin promoter.

Fig. 3.

Time course of vector persistence in exon 17–disrupted hemophiliac mice. Av1H8101 (4 × 1010 particles/mouse) was administered via tail vein injection to a group of 8 exon 17–disrupted hemophiliac mice. FVIII biological activity was measured before and at 2 and 16 weeks after vector administration. Groups of four mice each were killed at 2 or 16 weeks, and DNA and RNA were isolated from each mouse liver. (A) Southern analysis. Each DNA sample (10 μg) was digested with BamHI. The arrow designates a 3.4-kb fragment containing the vector-derived FVIII sequence. The standards (lanes 1 and 2) were generated by digesting purified Av1H8101 viral DNA in amounts equivalent to 10 and 1 vector copies per cell. Lanes 3 through 6 and 7 through 10 represent liver DNA from mice treated with Av1H8101 or Av3H8101, respectively. No vector was detected in uninjected control mouse liver DNA (data not shown). (B) FVIII protein expression and RNAse protection analysis. Plasma levels of biologically active FVIII protein measured in each mouse are displayed above the lanes. FVIII levels at 2 weeks and 16 weeks are displayed above lanes 4 through 7, and 8 through 11, respectively. For the RNAse protection analysis, 50 μgs of total cellular RNA isolated from the mouse livers were used in each reaction. The arrow labeled FVIII designates the 212-nt human FVIII-specific protected probe fragment. Lane 2 contains undigested full-length probe. Lane 3 contains liver RNA isolated from an uninjected control exon 17–disrupted hemophiliac mouse. Lanes 4 through 7 and 8 through 11 represent RNA from mice at 2 or 16 weeks after vector treatment, respectively. Lanes 1 and 12 contain32P-labeled DNA molecular-weight markers. The lower panel displays a separate RNAse protection assay using 20 μg of total cellular mouse liver RNA and an antisense RNA probe encoding a portion of the mouse glyceraldehyde 3-phosphodehydrogenase (GAPDH) cDNA. The arrow labeled GAPDH designates the 134-nt mouse GAPDH-specific protected probe fragment.

Fig. 3.

Time course of vector persistence in exon 17–disrupted hemophiliac mice. Av1H8101 (4 × 1010 particles/mouse) was administered via tail vein injection to a group of 8 exon 17–disrupted hemophiliac mice. FVIII biological activity was measured before and at 2 and 16 weeks after vector administration. Groups of four mice each were killed at 2 or 16 weeks, and DNA and RNA were isolated from each mouse liver. (A) Southern analysis. Each DNA sample (10 μg) was digested with BamHI. The arrow designates a 3.4-kb fragment containing the vector-derived FVIII sequence. The standards (lanes 1 and 2) were generated by digesting purified Av1H8101 viral DNA in amounts equivalent to 10 and 1 vector copies per cell. Lanes 3 through 6 and 7 through 10 represent liver DNA from mice treated with Av1H8101 or Av3H8101, respectively. No vector was detected in uninjected control mouse liver DNA (data not shown). (B) FVIII protein expression and RNAse protection analysis. Plasma levels of biologically active FVIII protein measured in each mouse are displayed above the lanes. FVIII levels at 2 weeks and 16 weeks are displayed above lanes 4 through 7, and 8 through 11, respectively. For the RNAse protection analysis, 50 μgs of total cellular RNA isolated from the mouse livers were used in each reaction. The arrow labeled FVIII designates the 212-nt human FVIII-specific protected probe fragment. Lane 2 contains undigested full-length probe. Lane 3 contains liver RNA isolated from an uninjected control exon 17–disrupted hemophiliac mouse. Lanes 4 through 7 and 8 through 11 represent RNA from mice at 2 or 16 weeks after vector treatment, respectively. Lanes 1 and 12 contain32P-labeled DNA molecular-weight markers. The lower panel displays a separate RNAse protection assay using 20 μg of total cellular mouse liver RNA and an antisense RNA probe encoding a portion of the mouse glyceraldehyde 3-phosphodehydrogenase (GAPDH) cDNA. The arrow labeled GAPDH designates the 134-nt mouse GAPDH-specific protected probe fragment.

Close modal

A second hypothesis to explain the sudden, sporadic decline in FVIII plasma levels observed in some mice (see Fig 1) was the development of a humoral immune response directed against the human FVIII protein. An ELISA designed to detect antihuman FVIII antibodies was used to assay plasma from the exon 16 and exon 17 mice represented in Fig 2 before vector administration and at various time points thereafter (Table 2). In all cases, antibody levels were either undetectable or low, ranging from below the limit of sensitivity, 50 ng/mL, up to 97 ng/mL. There was no case in which the drop in FVIII plasma levels could be attributed to the generation of a strong antibody response. Interestingly, Qian et al42showed that hemophiliac mice of both genotypes treated with a single intravenous administration of purified full-length FVIII protein rapidly developed a strong immune response (approximately 40,000 ng/mL) to the human protein.

Table 2.

Hemophiliac Mouse Plasma Levels of Antihuman FVIII Antibodies

Genotype Mouse No. Time of Sample Collection (wk)Factor VIII Levels (mU/mL) Antihuman FVIII Antibody Levels (ng/mL)
Ex 16 KO   Prebleed  (−) <50  
 1  12  277  52  
  16 (−)  54  
  Prebleed  (−) <50  
 2  12  245  51  
  16  26  60 
  Prebleed  (−)  <50  
 3  16 208  <50  
  20  33  62  
  Prebleed (−)  <50  
 4   8  1983  53 
  24  300  79  
  Prebleed  (−) <50  
 5  12  184  62  
  16 (−)  93  
Ex 17 KO   Prebleed (−)  <50  
 6   2  136  53 
   6  33  51  
  Prebleed  (−) <50  
 7  12  240  97  
  16 (−)  76  
  Prebleed  (−) <50  
 8  12  554  58  
  24  318  84 
  Prebleed  (−)  62  
 9  20  242 <50  
  24  24  87 
Genotype Mouse No. Time of Sample Collection (wk)Factor VIII Levels (mU/mL) Antihuman FVIII Antibody Levels (ng/mL)
Ex 16 KO   Prebleed  (−) <50  
 1  12  277  52  
  16 (−)  54  
  Prebleed  (−) <50  
 2  12  245  51  
  16  26  60 
  Prebleed  (−)  <50  
 3  16 208  <50  
  20  33  62  
  Prebleed (−)  <50  
 4   8  1983  53 
  24  300  79  
  Prebleed  (−) <50  
 5  12  184  62  
  16 (−)  93  
Ex 17 KO   Prebleed (−)  <50  
 6   2  136  53 
   6  33  51  
  Prebleed  (−) <50  
 7  12  240  97  
  16 (−)  76  
  Prebleed  (−) <50  
 8  12  554  58  
  24  318  84 
  Prebleed  (−)  62  
 9  20  242 <50  
  24  24  87 

Plasma was collected at the indicated times from the exon 16–(Ex 16 KO) and exon 17–disrupted (Ex 17 KO) mice treated with Av3H8101 (6 × 1010 particles/mouse) represented in Fig 3. Plasma was analyzed for FVIII biological activity and for the presence of antihuman FVIII antibodies by ELISA. For the antibody analysis, plasma samples were diluted 1:10, and the limit of sensitivity of the assay was 50 ng/mL. (−) represents a FVIII activity level of less than 1% of that detected in a normal mouse plasma sample analyzed in parallel in each assay.

In this work, we showed long-term phenotypic correction of hemophilia A by in vivo gene therapy in a clinically relevant small hemophilia A animal model. Whereas sustained human FVIII expression was demonstrated previously in normal mice20 and short-term phenotypic correction was achieved in hemophiliac dogs,21 the FVIII-deficient hemophiliac mouse26 has several advantages over these animal models for the study of hemophilia A gene therapy and FVIII in vivo function. In contrast to normal mice, the biologic activity and function of the human FVIII protein expressed in hemophiliac mice can be measured directly, and phenotypic correction of the bleeding defect assessed. In addition, both FVIII-deficient mouse genotypes were developed as a mixture of two mouse strains, C57BL/6 and 129SV,26 providing a more diverse genetic background than inbred strains for gene therapy studies. Compared with the canine hemophilia A model, the knockout mice provide the only available small animal model of hemophilia A. Small animal models facilitate the performance of multiple studies with large cohorts to better characterize vector function and FVIII protein expression.

An important attribute that distinguishes the hemophiliac mouse model from that of normal mice is the ability to show phenotypic correction. The murine hemophiliac phenotype is characterized by subcutaneous, intraperitoneal, intrathoracic, and joint bleeds. The mice bleed extensively after minor lacerations, are occasionally anemic, and exhibit a high spontaneous mortality rate. We initially observed that, after vector treatment, the mice appeared normal and the bleeding phenotype resolved. To obtain an objective assessment of phenotypic correction, a tail clip survival study, which clearly distinguished hemophiliac from normal mice, was used. This assay showed that human FVIII expressed by an intravenously administered adenoviral vector is biologically active, functional in mice, and therapeutic.

The time course of FVIII expression in individual mice showed an intriguing pattern. With both the Av1 and Av3 vectors, treated mice displayed an initial drop in FVIII plasma levels, probably due to a low level of vector-mediated toxicity as previously described with normal mice.20 FVIII expression in several of the mice subsequently stabilized, showing that a single adenoviral vector administration has the potential to achieve long-term, constant-level expression. Furthermore, long-term FVIII expression was not dependent on a specific murine FVIII mutation, as a similar expression pattern was observed in both hemophiliac mouse genotypes. However, sustained expression was not uniformly achieved, as FVIII levels rapidly and sporadically declined in some mice.

The mechanism by which FVIII expression levels declined was investigated by DNA, RNA, and human FVIII-specific antibody analyses. Over time, vector DNA levels declined substantially, while FVIII RNA levels showed only a minimal decrease. Furthermore, FVIII RNA levels were similar in mice with widely divergent FVIII plasma levels. Thus, the decline in plasma levels could not be completely attributed to either vector loss or transcriptional inactivation. The development of an immune response to human FVIII seemed a likely explanation for the loss of FVIII expression. Little or no human FVIII-specific antibody response was detected in either strain of hemophiliac mice before or after attenuation of expression, suggesting that a humoral immune response was not responsible for the loss of FVIII. However, we cannot rule out the possibility that loss of expression resulted from a shift in the subclass or quality of a low titer antibody. A cell-mediated immune response directed toward hepatocytes expressing human FVIII could also explain the decline in FVIII expression, but the elimination of hepatocytes would have resulted in a parallel loss of FVIII RNA, which was not observed. Presently, the explanation for the rapid loss of expression in some mice remains unclear. Further investigation of this phenomenon may lead to a better understanding of vector and FVIII physiology and to the development of vectors which more uniformly remain efficacious.

A major issue broadly relevant to the field of gene therapy is the potential immunogenicity of endogenously expressed foreign proteins. Ten percent to 30% of human hemophiliacs treated with intermittent, intravenous FVIII protein administration develop FVIII inhibitory antibodies.1,43 Hemophiliac mice injected intravenously with human full-length FVIII develop a potent anti-FVIII antibody response,42 and a T-cell response directed to the full-length protein.44 However, hemophiliac mice treated with an adenoviral vector encoding human BDD FVIII generated little or no antibody response to human BDD FVIII. The induction of a cytotoxic T-lymphocyte (CTL) response was not investigated. Furthermore, the low antibody levels detected in either strain of hemophiliac mice indicate that the lack of FVIII immunogenicity was not dependent on the specific FVIII mutation. These observations represent preliminary evidence to suggest that constant level, endogenous expression of human FVIII may be less immunogenic than intermittent, intravenous protein administration. Verification of this hypothesis will require a controlled study in which the immunogenicity of BDD FVIII protein is compared directly with vector-mediated expression of BDD FVIII. Notably, vector-mediated expression of BDD FVIII can result in the generation of antibodies against the human protein, as was observed in vector-treated hemophiliac dogs.21 Because the murine immune system is well studied and easily manipulated, hemophiliac mice represent an excellent model for characterizing the immune response to FVIII. Furthermore, this model can be used for evaluating gene therapy strategies to treat FVIII inhibitory antibodies, a significant clinical problem in hemophilia.

Two human BDD FVIII-encoding vectors were evaluated in this study. The first generation Av1 vector is composed of an adenoviral backbone from which the E1 and E3 regions were removed. The third generation Av3 vector contains, in addition, the deletion of the E2a region. Av3 vectors are completely replication defective in vitro and do not express detectable levels of hexon capsid protein.29 In some animal models, attenuation of E2a gene expression results in prolonged transgene expression and a reduction in the host immune response to the vector.39-41 However, in other mouse strains and hemophilia B dogs, the inclusion of a temperature sensitive mutation or removal of the E2a gene had no effect on vector persistence.45,46 In this work, treatment of hemophiliac mice with either the Av1 or the Av3 FVIII vectors resulted in similar liver transduction efficiency, transgene transcriptional activity, and FVIII expression levels. Both Av1 and Av3 vectors achieved sustained expression with attenuation in the first 20 weeks followed by stable FVIII plasma levels in some animals for over 9 months. The finding that Av3 vectors did not provide for greater efficacy in the hemophiliac mice is consistent with the hypothesis that the immune system did not play a major role in limiting the duration of expression in this model. Furthermore, the attenuation of FVIII expression in the first 20 weeks was probably due to direct vector-mediated liver toxicity,20,46 which is similar with both Av1 and Av3 vectors (T.A.G. Smith, personal communication, August 1997) and may be related to the E4 gene,47-50which remains in both backbones. The removal of additional backbone genes may decrease toxicity, minimize the attenuation of expression in the first 20 weeks, and enable more sustained expression.

The data presented in this work support the use of recombinant adenoviruses for the treatment of hemophilia A. Prolonged curative therapy was achieved with a single vector administration in a hemophiliac animal model. However, clinical benefit in humans will require therapy that extends for years, necessitating repeated treatments or more sustained expression. Current studies to allow vector readministration51-53 together with the development of improved, more attenuated adenoviral vectors54-59 may soon provide long-term correction of hemophilia A.

We thank Drs Martin Woodle and Theodore A.G. Smith for critical review of the manuscript, Dr Theodore A.G. Smith for communication of data before publication, Dr Russette Lyons and Christoph Wey for assistance with the animal procedures, and Adam Shoemaker for vector production and quality control.

Address reprint requests to Michael Kaleko, MD, PhD, Genetic Therapy, Inc, 938 Clopper Rd, Gaithersburg, MD 20878.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Sadler JE, Davie EW: Hemophilia A, hemophilia B, and von Willebrand's disease, in Nienhuis AW, Leder P, Majerus PW (eds): The Molecular Basis of Blood Diseases. Philadelphia, PA, Saunders, 1987, p 575
2
Rosendaal
FR
Smit
C
Briët
E
Hemophilia treatment in historical perspective: A review of medical and social developments.
Ann Hematol
62
1991
5
3
Connelly
S
Kaleko
M
Gene therapy for hemophilia A.
Thromb Haemost
78
1997
31
4
Kaufman
RJ
Biological regulation of factor VIII activity.
Annu Rev Med
43
1992
325
5
Lynch
CM
Israel
DI
Kaufman
RJ
Miller
AD
Sequences in the coding region of clotting factor VIII act as dominant inhibitors of RNA accumulation and protein production.
Hum Gene Ther
4
1993
259
6
Hoeben
RC
Fallaux
FJ
Cramer
SJ
van den Wollenberg
JM
Van Ormondt
H
Briet
E
Van Der Eb
AJ
Expression of the blood-clotting factor-VIII cDNA is repressed by a transcriptional silencer located in its coding region.
Blood
85
1995
2447
7
Koeberl
DD
Halbert
CL
Krumm
A
Miller
AD
Sequences within the coding regions of clotting factor VIII and CFTR block transcriptional elongation.
Hum Gene Ther
6
1995
469
8
Chuah
MKL
Vandendriessche
T
Morgan
RA
Development and analysis of retroviral vectors expressing human factor VIII as a potential gene therapy for hemophilia A.
Hum Gene Ther
6
1995
1363
9
Kaufman
RJ
Wasley
LC
Dorner
AJ
Synthesis, processing, and secretion of recombinant human factor VIII expressed in mammalian cells.
J Biol Chem
263
1988
6352
10
Kaufman
RJ
Wasley
LC
Davies
MV
Wise
RJ
Israel
DI
Dorner
AJ
Effect of von Willebrand factor coexpression on the synthesis and secretion of factor VIII in Chinese hamster ovary cells.
Mol Cell Biol
9
1989
1233
11
Pittman
DD
Alderman
EM
Tomkinson
KN
Wang
JH
Giles
AR
Kaufman
RJ
Biochemical, immunological, and in vivo functional characterization of B-domain-deleted factor VIII.
Blood
81
1993
2925
12
Zatloukal
K
Cotten
M
Berger
M
Schmidt
W
Wagner
E
Birnstiel
ML
In vivo production of human factor VIII in mice after intrasplenic implantation of primary fibroblasts transfected by receptor-mediated, adenovirus-augmented gene delivery.
Proc Natl Acad Sci USA
91
1994
5148
13
Dwarki
VJ
Belloni
P
Nijjar
T
Smith
J
Couto
L
Rabier
M
Clift
S
Berns
A
Cohen
LK
Gene therapy for hemophilia A: Production of therapeutic levels of human factor VIII in vivo in mice.
Proc Natl Acad Sci USA
92
1995
1023
14
Hoeben
RC
van der Jagt
RC
Schoute
F
Van Tilburg
NH
Verbeet
MP
Briet
E
Van Ormondt
H
Van Der Eb
AJ
Expression of functional factor VIII in primary human skin fibroblasts after retrovirus-mediated gene transfer.
J Biol Chem
265
1990
7318
15
Hoeben
RC
Einerhand
MP
Briet
E
Van Ormondt
H
Valerio
D
Van Der Eb
AJ
Toward gene therapy in haemophilia A: Retrovirus-mediated transfer of a factor VIII gene into murine haematopoietic progenitor cells.
Thromb Haemost
67
1992
341
16
Hoeben
RC
Fallaux
FJ
Van Tilburg
NH
Cramer
SJ
Van Ormondt
H
Briet
E
Van Der Eb
AJ
Toward gene therapy for hemophilia A: Long-term persistence of factor VIII-secreting fibroblasts after transplantation into immunodeficient mice.
Hum Gene Ther
4
1993
179
17
Israel
DI
Kaufman
RJ
Retroviral-mediated transfer and amplification of a functional human factor VIII gene.
Blood
75
1990
1074
18
Connelly
S
Smith
TAG
Dhir
G
Gardner
JM
Mehaffey
MG
Zaret
KS
McClelland
A
Kaleko
M
In vivo gene delivery and expression of physiological levels of functional human factor VIII in mice.
Hum Gene Ther
6
1995
185
19
Connelly
S
Gardner
JM
McClelland
A
Kaleko
M
High level tissue-specific expression of functional human FVIII in mice.
Hum Gene Ther
7
1996
183
20
Connelly
S
Gardner
JM
Lyons
RM
McClelland
A
Kaleko
M
Sustained expression of therapeutic levels of human factor VIII in mice.
Blood
87
1996
4671
21
Connelly
S
Mount
J
Mauser
A
Gardner
JM
Kaleko
M
McClelland
A
Lothrop
CD
Complete short-term correction of canine hemophilia A by in vivo gene therapy.
Blood
88
1996
3846
22
Smith
TAG
Mehaffey
MG
Kayda
DB
Saunders
JM
Yei
S
Trapnell
BC
McClelland
A
Kaleko
M
Adenovirus-mediated expression of therapeutic plasma levels of human factor IX in mice.
Nat Genet
5
1993
397
23
Kozarsky
KF
Wilson
JM
Gene therapy: Adenovirus vectors.
Curr Opin Genet Dev
3
1993
499
24
Graham
JB
Buckwalter
JA
Hartley
LJ
Brinkhous
KM
Canine hemophilia: Observations on the course, the clotting anomaly, and the effects of blood transfusion.
J Exp Med
90
1949
97
25
Giles
AR
Tinlin
S
Greenwood
R
A canine model of hemophilic (factor VIII:C deficiency) bleeding.
Blood
60
1982
727
26
Bi
L
Lawler
AM
Antonarakis
SE
High
KA
Gerhart
JD
Kazazian
HH
Targeted disruption of the mouse factor VIII gene produces a model of haemophilia A.
Nat Genet
10
1995
119
27
Antonarakis
SE
Kazazian
HH
Tuddenham
EGD
Molecular etiology of factor VIII deficiency in hemophilia A.
Hum Mutat
5
1995
1
28
Tolstoshev P, Mittereder N, Yei S, Trapnell BC: Adenovirus vectors for gene therapy, in: Cancer Chemotherapy: Challenges for the Future. Tokyo, Japan, Excerpta Medica, 1994, p 23
29
Gorziglia
MI
Kadan
MJ
Yei
S
Lim
J
Lee
GM
Luthra
R
Trapnell
BC
Elimination of both E1 and E2a from adenovirus vectors further improves prospects for in vivo human gene therapy.
J Virol
6
1996
4173
30
Mittereder
N
March
KL
Trapnell
BC
Evaluation of the concentration and bioactivity of adenovirus vectors for gene therapy.
J Virol
70
1996
7498
31
Rosenfeld
MA
Yoshimura
K
Trapnell
BC
Yoneyama
K
Rosenthal
ER
Dalemans
W
Fukayama
M
Bargon
J
Stier
LE
Stratford Perricaudet
L
Perricaudet
M
Pavicani
A
Lecocq
JP
Crystal
RG
In vivo transfer of the human cystic fibrosis transmembrane conductance regulator gene to the airway epithelium.
Cell
68
1992
143
32
Bi
L
Sarkar
R
Naas
T
Lawler
AM
Pain
J
Shumaker
S
Bedian
V
Kazazian
HH
Further characterization of factor VIII-deficient mice created by gene targeting: RNA and protein studies.
Blood
88
1996
3446
33
Hoyer
LW
Hemophilia A.
N Engl J Med
330
1994
38
34
Lollar
P
Parker
ET
Curtis
JE
Helgerson
SL
Hoyer
LW
Scott
ME
Scandella
D
Inhibition of human factor VIIIa by anti-A2 subunit antibodies.
J Clin Invest
93
1994
2497
35
Toole
JJ
Knopf
JL
Wozney
JM
Sultzman
PA
Buecker
J
Pittman
DD
Kaufman
RJ
Brown
E
Shoemaker
C
Orr
EC
Amphlett
GW
Foster
WB
Coe
ML
Knutson
GJ
Fass
DN
Hewick
RM
Molecular cloning of a cDNA encoding antihemophiliac factor.
Nature
312
1984
342
36
Wood
WI
Capon
DJ
Simonsen
CC
Eaton
DL
Gitschier
J
Keyt
B
Seeburg
PH
Smith
DH
Hollingshead
P
Wion
KL
Delwart
E
Tuddenham
EGD
Vehar
GA
Lawn
RM
Expression of active human factor VIII from recombinant DNA cloned.
Nature
312
1984
330
37
Toole
JJ
Pittman
DD
Orr
EC
Murtha
P
Wasley
LC
Kaufman
RJ
A large region (∼95 kDa) of human factor VIII is dispensable for in vitro procoagulant activity.
Proc Natl Acad Sci USA
83
1986
5939
38
Eaton
DL
Wood
WI
Eaton
D
Hass
PE
Hollingshead
P
Wion
K
Mather
J
Lawn
RM
Vehar
GA
Gorman
C
Construction and characterization of an active factor VIII variant lacking the central one-third of the molecule.
Biochemistry
25
1986
1
39
Engelhardt
JF
Ye
X
Doranz
B
Wilson
JM
Ablation of E2A in recombinant adenoviruses improves transgene persistence and decreases inflammatory response in mouse liver.
Proc Natl Acad Sci USA
91
1994
6196
40
Engelhardt
JF
Litzky
L
Wilson
JM
Prolonged transgene expression in cotton rat lung with recombinant adenoviruses defective in E2a.
Hum Gene Ther
5
1994
1217
41
Yang
Y
Nunes
FA
Berencsi
K
Gonczol
E
Engelhardt
JF
Wilson
JM
Inactivation of E2a in recombinant adenoviruses improves the prospect for gene therapy in cystic fibrosis.
Nat Genet
7
1994
362
42
(suppl, abstr)
Qian
J
Bi
L
Kazazian
HH
Hoyer
LW
Inhibitor development and T cell response to human factor VIII in murine hemophilia A.
Blood
88
1996
656a
43
(suppl 1)
White
GC
Roberts
HR
The treatment of factor VIII inhibitors — a general overview.
Vox Sang
70
1996
19
44
(abstr, suppl 1)
Qian
J
Scott
DE
Hoyer
LW
T cell response to human factor VIII in murine hemophilia A.
Blood
90
1997
599a
45
Fang
B
Wang
H
Gordon
G
Bellinger
DA
Read
MS
Brinkhous
KM
Woo
SL
Eisensmith
RC
Lack of persistence of E1-recombinant adenoviral vectors containing a temperature-sensitive E2A mutation in immunocompetent mice and hemophilia B dogs.
Gene Ther
3
1996
217
46
Morral
N
O'Neal
W
Zhou
H
Langston
C
Beaudet
A
Immune responses to reporter proteins and high viral dose limit duration of expression with adenoviral vectors: Comparison of E2a wild type and E2a deleted vectors.
Hum Gene Ther
8
1997
1275
47
Gao
G
Yang
Y
Wilson
JM
Biology of adenovirus vectors with E1 and E4 deletions for liver-directed gene therapy.
J Virol
70
1996
8934
48
Armentano
D
Zabner
J
Sacks
C
Sookdeo
CC
Smith
MP
St. George
JA
Wadsworth
SC
Smith
AE
Gregory
RJ
Effect of the E4 region on the persistence of transgene expression from adenovirus vectors.
J Virol
71
1997
2408
49
Dedieu
JF
Vigne
E
Torrent
C
Jullien
C
Mahfouz
I
Caillaud
JM
Aubailly
N
Orsini
C
Guillaume
JM
Opolon
P
Delaere
P
Perricaudet
M
Yeh
P
Long-term gene delivery into the livers of immunocompetent mice with E1/E4-defective adenoviruses.
J Virol
71
1997
4626
50
Wang
Q
Greenburg
G
Bunch
D
Farson
D
Finer
MH
Persistent transgene expression in mouse liver following in vivo gene transfer with a ΔE1/ΔE4 adenovirus vector.
Gene Ther
4
1997
393
51
Smith
TAG
White
BD
Gardner
JM
Kaleko
M
McClelland
A
Transient immunosuppression permits successful repetitive intravenous administration of an adenovirus vector.
Gene Ther
3
1996
496
52
Yang
Y
Trinchieri
G
Wilson
JM
Recombinant IL-12 prevents formation of blocking IgA antibodies to recombinant adenovirus and allows repeated gene therapy to mouse lung.
Nat Med
1
1995
890
53
Yang
Y
Greenough
K
Wilson
JM
Transient immune blockade prevents formation of neutralizing antibody to recombinant adenovirus and allows repeated gene transfer to mouse liver.
Gene Ther
5
1996
412
54
Clemens
PR
Kochanek
S
Sunada
Y
Chan
S
Chen
H
Campbell
KP
Caskey
CT
In vivo muscle gene transfer of full-length dystrophin with an adenoviral vector that lacks all viral genes.
Gene Ther
3
1996
965
55
Fisher
KJ
Choi
H
Bruda
J
Chen
S
Wilson
JM
Recombinant adenovirus deleted of all viral genes for gene therapy of cystic fibrosis.
Virology
217
1996
11
56
Haecker
SE
Stedman
HH
Balice-Gordon
RJ
Smith
DBJ
Greelish
JP
Mitchell
MA
Wells
A
Sweeney
HL
Wilson
JM
In vivo expression of full-length human dystrophin from adenoviral vectors deleted of all viral genes.
Hum Gene Ther
7
1996
1907
57
Kochanek
S
Clemens
PR
Mitani
K
Chen
H
Chan
S
Caskey
CT
A new adenoviral vector: Replacement of all viral coding sequences with 28 kb of DNA independently expressing both full-length dystrophin and beta-galactosidase.
Proc Natl Acad Sci USA
93
1996
5731
58
Lieber
A
He
C
Kirillova
I
Kay
MA
Recombinant adenoviruses with large deletions generated by cre-mediated excision exhibit different biological properties compared with first-generation vectors in vitro and in vivo.
J Virol
70
1996
8944
59
Hardy
S
Kitamura
M
Harris-Stansil
T
Dai
Y
Phipps
ML
Construction of adenovirus vectors through Cre-lox recombination.
J Virol
71
1997
1842
Sign in via your Institution