Replacement of the missing factor VIII (FVIII) is the current standard of care for patients with hemophilia A. However, the short half-life of FVIII makes frequent treatment necessary. Current efforts focus on the development of longer-acting FVIII concentrates by introducing chemical and genetic modifications to the protein. Any modification of the FVIII protein, however, risks increasing its immunogenic potential to induce neutralizing antibodies (FVIII inhibitors), and this is one of the major complications in current therapy. It would be highly desirable to identify candidates with a high risk for increased immunogenicity before entering clinical development to minimize the risk of exposing patients to such altered FVIII proteins. In the present study, we describe a transgenic mouse line that expresses a human F8 cDNA. This mouse is immunologically tolerant to therapeutic doses of native human FVIII but is able to mount an antibody response when challenged with a modified FVIII protein that possesses altered immunogenic properties. In this situation, immunologic tolerance breaks down and antibodies develop that recognize both the modified and the native human FVIII. The applicability of this new model for preclinical immunogenicity assessment of new FVIII molecules and its potential use for basic research are discussed.

Hemophilia A is an X-linked bleeding disorder that is caused by reduced function or lack of clotting factor VIII (FVIII).1  Replacement of the missing protein is the current standard of care for patients. However, the short half-life of FVIII, ∼ 7-17 hours,2  makes frequent treatment necessary. Current efforts focus on the development of longer-acting FVIII concentrates, which should decrease the required treatment frequency and therefore improve the quality of life for patients. Recently described approaches in the development of longer-acting concentrates are based on strategies that have been successfully applied to other therapeutic proteins: chemical modifications such as the addition of polyethylene glycol (PEG) polymers, polysialic acids, or hydroxyethyl starch3,4 ; alternative formulations with PEG-modified liposomes3 ; and fusion to the Fc part of human IgG.5  In addition, molecular modifications of the FVIII protein aimed at increasing the duration of its cofactor activity or reducing its clearance in vivo have been reported.3 

Any chemical or molecular modification of the FVIII protein can potentially increase its immunogenic potential. Modifications could generate neo-epitopes for both B and T cells or may induce altered structures that could bind and trigger receptors expressed on cells of the innate immune system, thereby amplifying potential anti-FVIII antibody responses.6  Finally, modifications could generate repetitive epitopes for B cells that might cause the activation and differentiation of B cells and the subsequent production of antibodies without the requirement for T-cell help.7,8  Therefore, the potential impact that any modification of an FVIII concentrate might have on its immunogenic potential requires comprehensive assessment based on immunogenicity data obtained during clinical development. However, it would be highly desirable to identify candidates with a high risk for increased immunogenicity before entering clinical development to minimize the risk of exposing patients to such candidates. So far, assessing the immunogenicity of modified FVIII proteins in preclinical animal models has been difficult because of the xenogenic nature of human FVIII in conventional models. Human FVIII induces antibodies when injected into mice, rats, dogs, or monkeys,9-12  which makes it difficult to distinguish between the immunogenicity of native human FVIII and the immunogenicity of modified human FVIII. Previously, we reported the generation of new transgenic mouse lines that express a human F8 cDNA as a transgene.13,14  The transgene is randomly integrated into the murine genome, and its expression is driven by an albumin promoter and α-fetoprotein enhancer elements. The human F8 transgene should result in the expression of native human FVIII during embryonic development, when the T-cell repertoire in the thymus is shaped and high-avidity “self-reactive” T cells are deleted from the repertoire.15,16  Expression of human FVIII during this period should therefore lead to the depletion of high-avidity human FVIII-specific T cells and result in specific immune tolerance toward human FVIII. Similar genetically engineered mouse models were described for human insulin analogs,17  modified human tissue plasminogen activator variants,18  human IFNs,19,20  human Fas ligand inhibitory protein (decoy receptor 3, DcR3),21  and a mutant of human FVIII.22  Transgenic mouse models for human IFNα2b (hIFNα2b) and human IFNβ (hIFNβ)19,20  were shown to be predictive of the human immune response, because commercially available hIFNβ drugs that were found to induce antibodies in a large number of patients were shown to break immune tolerance in these mice.19 

In the present study, we describe the selection and characterization of a transgenic mouse line that is immunologically tolerant to native human FVIII but is still able to mount an antibody response to human FVIII when challenged with a modified FVIII protein that possesses increased immunogenicity. In this situation, immunologic tolerance breaks down and antibodies develop that recognize both the modified human FVIII and the native human FVIII.

Human F8 transgenic mice

Hemophilic mice that express a transgene of a native human F8 cDNA were generated by random integration of the transgene, as described previously.13,14  Briefly, we used a liver-specific expression vector,23  which was a generous gift from G. Schuetz (German Cancer Research Center, Heidelberg, Germany), to direct F8 expression to the liver. The vector contains an albumin promoter, a poly A signal derived from the human growth hormone, and α-fetoprotein enhancer elements (Figure 1). An Xhol restriction site between the albumin promoter and the poly A tail was used for insertion of the human F8 cDNA, as described previously.13  The vector was linearized and microinjected into the male pronucleus of fertilized oocytes obtained from mated female C57BL/6J mice after superovulation. Founder mice carrying the human F8 cDNA transgene were crossed with hemophilic E17 mice (characterized by a targeted disruption of exon 17 of the murine F8 gene24 ) and bred to homozygosity for the murine hemophilic E17 genotype and the human F8 transgene.

Figure 1

Structure of the linearized human F8 cDNA construct. The human F8 cDNA was expressed under the control of the mouse albumin promoter and enhancer and the mouse α-fetoprotein enhancer. A 2.1-kb fragment of the human growth hormone gene (hGH) was cloned downstream of the human F8 cDNA to ensure correct polyadenylation of the human F8 mRNA.

Figure 1

Structure of the linearized human F8 cDNA construct. The human F8 cDNA was expressed under the control of the mouse albumin promoter and enhancer and the mouse α-fetoprotein enhancer. A 2.1-kb fragment of the human growth hormone gene (hGH) was cloned downstream of the human F8 cDNA to ensure correct polyadenylation of the human F8 mRNA.

Close modal

Confirmation of complete transgene integration

Correct and complete integration of the human F8 cDNA into the mouse genome was confirmed by PCR. For this purpose, genomic DNA was isolated using the nexttec DNA isolation kit (Genomic DNA Isolation Kit; Biotechnologie). The following 9 primer pairs were designed, which produce overlapping F8 DNA products within the human F8 cDNA: pair (P) 1 primer forward (P1for), 5′-TTCTTTCTGTGCCTTTTGCGATTC-3′; P1 primer reverse (P1rev), 5′-TCATTCGTAGTTGGGGTTCCTCTG-3′; P2for, 5′-CAGGCGTCCTTGGAAATCTCGC-3′; P2rev, 5′-TGTGAGGTACCAGCTTCGGTTC-3′; P3for, 5′-CAGTGACTGTAG-AAGATGGGCCAAC-3′; P3rev, 5′-TACCTGCTGCCAAATTGTCTGATG-3′; P4for, 5′-TCCATCAGACAATTTGGCAGCAG-3′; P4rev, 5′-CTTGCCCAGAGTTCAGAGAGTTCT-3′; P5for, 5′-GCCAGAATCAGCAAGGTGGAT-3′; P5rev, 5′-TTGTGGTGCATGCATATTTCTCTAC-3; P6for, 5′-TAGAGAAATATGCATGCACCACAAG-3′; P6rev, 5′-CACAAGCGTTCAGGGACAAAATG-3′; P7for, 5′-TGTTCTCCCGAAACCAGACTTGC-3′; P7rev, 5′-TTCTGCTCCTTGCCTCTGATCTTC-3′; P8for, 5′-AGGAAGATCAGAGGCAAGGAGCAG-3; P8rev, 5′-TCTCTAATGTG-TCCAGAAGCCATTC-3′; P9for, 5′-GGAATGGCTTCTGGACACATTAGAG-3′; and P9rev, 5′-GAACCTCCATCCTCAGGGCAATC-3′. To prove complete integration of the human F8 cDNA into the mouse genome, additional primer pairs were designed to cover the regions flanking the human F8 cDNA: primer pair A (PA, including FVIII ATG) PAfor, 5′-CCAGATGGCAAACATACGCAAG-3′; PArev, 5′- CAGGCAGCTCACCGAGATCAC-3′; primer pair B (PB, including FVIII TGA) PBfor, 5′-CGTTACTGACTCGCTACCTTCGA-3′; and PBrev, 5′-ACTCTCCCTGCTCCAGGAGC-3′. The accuracy of the PCR products was confirmed by sequencing using an ABI Prism 3100 capillary sequencer (Applied Biosystems) and alignment using Vector NTI Advance 11.0.

Finally, integration of F8 cDNA in full-length was confirmed by long-range PCR (QIAGEN) using the following primer pair: primer forward (Pfor), 5′- GTCATGGGACTATATGCAAAGTGATCTCG-3′, and primer reverse (Prev), 5′- AGAACCTCCATCCTCAGGGCAATCT-3′. The PCR product was partially sequenced to confirm specificity. The consensus of the ∼ 2000 bases that were sequenced was > 99%.

Analysis of F8 gene expression

F8 mRNA levels of various organs were analyzed by a 2-step RT-PCR. RNA was isolated from organ samples using RNeasy Mini Kit (QIAGEN) and then reverse transcribed into cDNA using QuantiTect Reverse Transcription (QIAGEN). This procedure included a DNA wipeout step. TaqMan real-time quantitative PCR for human F8 cDNA was done using TaqMan GeneExpressionMasterMix (Applied Biosystems) and a specific human F8 primer pair and probe: forward primer,13  5′-GGCACTGTACAATCTCTATCCAGGT-3′; reverse primer,13  5′-GATGCTCGCCAATAAGGCAT-3 and the F8-specific dual-labeled Black Hole Quencher probe,13  FAM-5′-CCAAAGCTGGAATTTGGCGGGTG-3′-BHQ. Murine F8 cDNA was analyzed using a set of primers from Applied Biosystems that covers the murine F8 13-14 exon boundary. In addition, as a normalization control, the expression of murine β-actin was determined in all samples using the TaqMan mouse ACTB (actin, beta) endogenous control (VIC/MGB Probe, Primer Limited) from Applied Biosystems.

The expression of full-length human F8 mRNA in liver samples was analyzed using the 9 primer pairs described in “Confirmation of complete transgene integration.”

FISH analysis of the integration sides of human F8 cDNA

FISH analysis was done as described previously.25,26  Briefly, spleen cells obtained from human F8 transgenic mice were stimulated with concanavalin A to induce cell division. Chromosomes were prepared and stained with 4′,6-diamidino-2-phenylindole (DAPI). DAPI-stained metaphases were imaged before hybridization. The human F8-specific probe for hybridization was generated using the 9 human F8-specific primer pairs described in “Confirmation of complete transgene integration.” The probe was labeled with dioxigenin-dUTP and biotin-dUTP. DAPI-stained chromosomes and hybridization signals were analyzed by fluorescence microscopy. Chromosome and FISH signals were displayed in false colors and images were merged on the computer.

Treatment of mice with human FVIII and human VWF

All mouse studies were approved by the local authority in Vienna, Austria in accordance with Austrian federal law (Act BG 501/1989) regulating animal experiments and were approved by the institutional animal care and use committee.

Male and female mice 8-17 weeks of age were treated intravenously with up to 9 weekly doses of 0.2 μg of recombinant human FVIII (∼ 80 IU/kg) or 4 weekly doses of 5 μg of recombinant human VWF (all from Baxter BioScience).

In some experiments, mice were treated with PEG-modified FVIII preparations (mFVIII1 and mFVIII2) from Baxter BioScience. mFVIII1 and mFVIII2 are full-length recombinant FVIII preparations that were chemically modified by coupling 20-kDa PEG reagents using preferential amine coupling to lysine residues at neutral pH, as described by Zaplinski et al.27  The PEG reagents used to prepare mFVIII1 and mFVIII2 were described previously.28-30  mFVIII1 and mFVIII2 mainly differ in their degree of modification: mFVIII1 contains ∼ 2 mol of PEG/mol FVIII and mFVIII2 contains 12 mol of PEG/mol FVIII. Because of the higher degree of modification, mFVIII2 shows reduced procoagulant activity and reduced binding to VWF (Table 1). Analysis of aggregates in native and modified human FVIII by asymmetrical flow field-flow fractionation31  and high-performance size-exclusion chromatography using an Agilent SEC-5 500 Å column (Agilent Technologies) with fluorescence detection (excitation at 280 nm and emission at 340 nm) did not reveal any relevant formation of FVIII protein aggregates in any of the preparations.

Table 1

Comparison of mFVIII1 and mFVIII2 with respect to PEGylation degree, specific activity, and VWF binding

PEGylation degree, mol PEG/mol FVIIISpecific activity, FVIII:C/mg proteinVWF binding, KD, nM
mFVIII1 5773 0.82 
mFVIII2 12 153 7.23 
PEGylation degree, mol PEG/mol FVIIISpecific activity, FVIII:C/mg proteinVWF binding, KD, nM
mFVIII1 5773 0.82 
mFVIII2 12 153 7.23 

Blood samples were taken before the first treatment, 1 week after the fourth treatment, and 1 week after the last treatment. Blood samples were added at a 4:1 (vol/vol) ratio to 0.1M sodium citrate and plasma was separated by centrifugation. Plasma samples were stored at −20°C until further analysis. Samples used for the analysis of FVIII antigen or FVIII activity were kept at −80°C.

Analysis of antibodies and antibody-secreting cells

Binding antibodies against human FVIII and human VWF were measured by ELISA as described previously.32  ELISA plates were coated with 1 μg/mL of one of the following proteins: recombinant human FVIII, PEGylated human FVIII (mFVIII1 or mFVIII2), or recombinant human VWF.

Inhibitory antibodies against FVIII were analyzed by Bethesda assay.33 

Antibody-secreting cells (ASCs) were detected by enzyme-linked immunospot (ELISPOT) assays as described previously.34  ELISPOT plates were coated with 10 μg/mL of either recombinant human FVIII or recombinant human VWF.

Restimulation of FVIII-specific T cells in vitro

Spleens were obtained 7 days after the last dose of FVIII and spleen cell suspensions were prepared and restimulated as described previously.34  Cytokines released into cell culture supernatants were analyzed using a multiplex bead-based assay (Bio-Plex) from Bio-Rad Laboratories. Proliferation of FVIII-specific T cells was analyzed as described previously.34 

Analysis of FVIII antigen, FVIII activity, and binding of FVIII to VWF

Human FVIII antigen in mouse plasma was measured using the human VIIIC:Ag ELISA kit (Diagnostica Stago). FVIII activity in mouse plasma and the potencies of mFVIII1 and mFVIII2 were analyzed using the chromogenic activity assay Immunochrom FVIII (Baxter Diagnostics). The specific activity was determined by calculating the potency of mFVIII1 and mFVIII2 per milligram of protein. Protein was quantified by conventional protein determination using the Bradford method.

Binding of FVIII to VWF was analyzed by surface plasmon resonance technology using a Biacore 3000 system. Briefly, a plasma-derived VWF (Diagnostica Stago) was immobilized on the flow cells of a CM5 biosensor chip. The samples to be analyzed were diluted to 20nM FVIII with a 10mM HEPES buffer containing 150mM NaCl and 0.005% surfactant P20, pH 7.4, according to their protein content calculated with a molecular mass of 280 kDa. Diluted samples were then applied to the chip using the “kinject” mode with a constant flow rate of 5 μL/min. Time for association was 10 minutes and time for dissociation was 60 minutes.

Determination of blood loss after tail cut

Blood loss after tail cut was determined using the tail-cut model.35  At the end of the observation period of 60 minutes, mice were killed by cervical dislocation before recovery from anesthesia.

Statistics

Group statistics were performed by 1-way ANOVA with the Dunnett posttest using Prism 5 software (GraphPad). P < .05 was considered statistically significant.

Generation and selection of a human F8 transgenic mouse line that does not develop antibodies against native human FVIII

Previously, we reported the generation of new transgenic mouse lines that express a human full-length F8 cDNA as a transgene on the hemophilic E17 background.13,14  Initially, we obtained 7 transgenic lines. To select a line that does not develop antibodies after repeated intravenous application of therapeutic doses of human native FVIII, we treated all sublines with 8 IV doses of human FVIII at weekly intervals, and analyzed antibody responses against human FVIII. We identified one subline that did not develop antibodies under these conditions. The remaining 6 sublines developed different levels of antibody responses ranging from low-titer to high-titer antibodies. Examples are shown in Figure 2A and B, illustrating results obtained with subline E (no antibody response) and subline G (highest levels of antibody response).

Figure 2

Transgenic mice of sublines E and G differ in their antibody responses to human FVIII. Mice were treated with 8 weekly intravenous doses of human FVIII. Blood for antibody analysis and spleens for the analysis of antibody-secreting cells (ASC) were collected 1 week after the last dose. (A) Titers of anti–human FVIII IgG antibodies were detected by ELISA. Antibody titers were shown for individual mice. Anti-FVIII antibody-secreting cells (ASC) were analyzed by ELISPOT. Results obtained from a representative mouse are shown. Each spot corresponds to 1 ASC. (B) Plasma samples from 3 mice of each subline were analyzed for both titers of anti–human FVIII IgG antibodies by ELISA and FVIII inhibitor titers by Bethesda assay.

Figure 2

Transgenic mice of sublines E and G differ in their antibody responses to human FVIII. Mice were treated with 8 weekly intravenous doses of human FVIII. Blood for antibody analysis and spleens for the analysis of antibody-secreting cells (ASC) were collected 1 week after the last dose. (A) Titers of anti–human FVIII IgG antibodies were detected by ELISA. Antibody titers were shown for individual mice. Anti-FVIII antibody-secreting cells (ASC) were analyzed by ELISPOT. Results obtained from a representative mouse are shown. Each spot corresponds to 1 ASC. (B) Plasma samples from 3 mice of each subline were analyzed for both titers of anti–human FVIII IgG antibodies by ELISA and FVIII inhibitor titers by Bethesda assay.

Close modal

The question arose as to why different transgenic sublines would respond differently to challenges with human FVIII. We chose the 2 sublines presented in Figure 2A and B and investigated whether differences in integration sites of the human F8 cDNA, incomplete integration of the human F8 cDNA, and/or differences in transcription of the human F8 cDNA could explain the differences in the phenotype that we observed.

Using FISH analysis, we specified the integration sites of the human F8 cDNA in sublines E and G. Whereas subline E has the transgene integrated into chromosome 3H3, subline G has it integrated into chromosome XA6 (Figure 3B). We only identified one integration site for each subline.

Figure 3

Differences in integration sites and transcription of human F8 cDNA between sublines E and G. (A) Schematic pictures of 9 overlapping primer pairs spanning the whole human F8 cDNA and of a primer pair designed for long-range PCR. (B) Identification of the integration sites of human F8 cDNA by FISH analysis: chromosomes for FISH analysis were prepared from spleen cells activated with concanavalin A. F8-specific probes were prepared using the 9 overlapping primer pairs shown in panel A and labeled with 2 different fluorochromes: dioxigenin-dUTP and biotin-dUTP. The hybridization signals of the F8-specific probes and the corresponding chromosome mapping for samples obtained from mice of sublines E and G are demonstrated. (C) Detection and sizing of all PCR products obtained from genomic DNA of sublines E and G with the 9 overlapping primer pairs shown in panel A using an Agilent 2100 Bioanalyzer. The molecular size is indicated in the first lane in kilobases. (D) Confirmation of the integration of the full-length human F8 cDNA into the genome of mice of sublines E and G (EM is a male mouse from subline E; EF is a female mouse from subline E; GM is a male mouse from subline G; and GF is a female mouse from subline G) by long-range PCR on genomic DNA using an Agilent 2100 Bioanalyzer. The molecular size is indicated in the first lane in kilobases. (E) Detection and sizing of all PCR products obtained from RNA after reverse transcription into cDNA using an Agilent 2100 Bioanalyzer. The molecular size is indicated in the first lane in kilobases. RNA was prepared from liver samples obtained from mice of sublines E and G.

Figure 3

Differences in integration sites and transcription of human F8 cDNA between sublines E and G. (A) Schematic pictures of 9 overlapping primer pairs spanning the whole human F8 cDNA and of a primer pair designed for long-range PCR. (B) Identification of the integration sites of human F8 cDNA by FISH analysis: chromosomes for FISH analysis were prepared from spleen cells activated with concanavalin A. F8-specific probes were prepared using the 9 overlapping primer pairs shown in panel A and labeled with 2 different fluorochromes: dioxigenin-dUTP and biotin-dUTP. The hybridization signals of the F8-specific probes and the corresponding chromosome mapping for samples obtained from mice of sublines E and G are demonstrated. (C) Detection and sizing of all PCR products obtained from genomic DNA of sublines E and G with the 9 overlapping primer pairs shown in panel A using an Agilent 2100 Bioanalyzer. The molecular size is indicated in the first lane in kilobases. (D) Confirmation of the integration of the full-length human F8 cDNA into the genome of mice of sublines E and G (EM is a male mouse from subline E; EF is a female mouse from subline E; GM is a male mouse from subline G; and GF is a female mouse from subline G) by long-range PCR on genomic DNA using an Agilent 2100 Bioanalyzer. The molecular size is indicated in the first lane in kilobases. (E) Detection and sizing of all PCR products obtained from RNA after reverse transcription into cDNA using an Agilent 2100 Bioanalyzer. The molecular size is indicated in the first lane in kilobases. RNA was prepared from liver samples obtained from mice of sublines E and G.

Close modal

We then investigated whether the human F8 cDNA is completely integrated into the genomes of sublines E and G using 2 different approaches. First, we designed 9 overlapping primer pairs covering the whole F8 cDNA (Figure 3A) and ran PCRs. Results indicated that both sublines have the complete human F8 cDNA integrated into their genome (Figure 3C). To further confirm complete integration, additional primer pairs were designed to cover the regions flanking the human F8 cDNA. The accuracy of the PCR products was confirmed by sequencing.

Our second approach was a long-range PCR that amplified the entire humane F8 cDNA (Figure 3A). The results proved that the complete human F8 cDNA is integrated into the mouse genome of both subline E and G (Figure 3D). Sequencing of ∼ 2000 bases at both the 3′and 5′ ends of the PCR products revealed a homology of > 99% with the original human F8 cDNA for both sublines.

Based on these data, we conclude that the observed differences in the phenotype of sublines E and G were not caused by incomplete integration of the human F8 cDNA into the genome of one of the sublines.

We then investigated whether incomplete transcription of the human F8 cDNA might be associated with differences in the phenotype of sublines E and G. We prepared RNA from the liver, reversely transcribed it into cDNA, and ran PCRs using the 9 overlapping primer pairs that covered the whole human F8 cDNA (Figure 3A). Results obtained with subline E indicated that the complete human F8 cDNA was transcribed into mRNA. However, results obtained with subline G suggested that transcription of F8 cDNA into F8 mRNA was very weak and only partially detectable (Figure 3E).

Based on these results, we selected subline E for all subsequent studies.

Lack of antibody development against FVIII in subline E is caused by specific immune tolerance

To exclude the possibility that the human F8 cDNA transgene resulted in some form of general immune suppression, we investigated whether mice of subline E could still respond to an unrelated human protein. We treated mice with 8 weekly IV doses of native human FVIII and subsequently with 4 weekly IV doses of human VWF, which was shown to induce high titer antibodies in hemophilic E17 mice.36  We included conventional hemophilic E17 mice as controls.

All conventional hemophilic E17 mice developed high-titer anti-FVIII antibodies that were correlated with a high frequency of anti-FVIII ASCs in the spleen (Figure 4A). In contrast, human F8 transgenic mice did not develop detectable antibodies in the circulation, which correlated with the absence of anti-FVIII ASCs in the spleen (Figure 4A). However, both conventional E17 mice and human F8 transgenic mice developed high-titer anti-VWF antibodies in the circulation and high frequencies of anti-VWF ASCs in the spleen after treatment with human VWF (Figure 4B).

Figure 4

Lack of antibody development against human FVIII in mice of subline E is caused by specific immune tolerance. Mice were treated with 8 weekly IV doses of human FVIII (A), followed by 4 weekly IV doses of human VWF (B). Blood for antibody analysis was drawn 1 week after the fourth (4xFVIII, A) and the eighth (8xFVIII, A) dose of FVIII, as well as 1 week after the fourth dose of VWF (4xVWF, B). Spleens for the analysis of ASCs were collected from 3 mice 1 week after the eighth dose of FVIII (A) and from the remaining mice 1 week after the fourth dose of VWF (B). Titers of anti–human FVIII IgG antibodies (A) and anti–human VWF IgG antibodies (B) were detected by ELISA. Antibody titers are shown for individual mice as follows: human F8 transgenic mice, male (▴) and female (●); conventional E17 F8-knockout mice: male (♢). Anti–human FVIII ASCs (A) and anti–human VWF ASCs (B) in the spleens of human F8 transgenic mice (huF8) and conventional E17 F8-knockout mice (E17F8ko) were analyzed by ELISPOT. Results obtained from a representative animal are shown. Each spot represents 1 ASC.

Figure 4

Lack of antibody development against human FVIII in mice of subline E is caused by specific immune tolerance. Mice were treated with 8 weekly IV doses of human FVIII (A), followed by 4 weekly IV doses of human VWF (B). Blood for antibody analysis was drawn 1 week after the fourth (4xFVIII, A) and the eighth (8xFVIII, A) dose of FVIII, as well as 1 week after the fourth dose of VWF (4xVWF, B). Spleens for the analysis of ASCs were collected from 3 mice 1 week after the eighth dose of FVIII (A) and from the remaining mice 1 week after the fourth dose of VWF (B). Titers of anti–human FVIII IgG antibodies (A) and anti–human VWF IgG antibodies (B) were detected by ELISA. Antibody titers are shown for individual mice as follows: human F8 transgenic mice, male (▴) and female (●); conventional E17 F8-knockout mice: male (♢). Anti–human FVIII ASCs (A) and anti–human VWF ASCs (B) in the spleens of human F8 transgenic mice (huF8) and conventional E17 F8-knockout mice (E17F8ko) were analyzed by ELISPOT. Results obtained from a representative animal are shown. Each spot represents 1 ASC.

Close modal

Based on these data, we conclude that transgenic mice of subline E express specific immune tolerance toward native human FVIII.

Mice of subline E express human F8 mRNA in a range of different organs but do not express FVIII protein in the circulation

We then assessed the transcription of human F8 cDNA in various murine tissues, including liver, brain, reproductive organs, kidneys, bone marrow, muscle, heart, thymus, lymph nodes, spleens, and lungs, and compared the expression of the human F8 cDNA with the expression of the murine F8 gene in the same organs in conventional C57BL/6 mice. Expression of both the human F8 cDNA and the murine F8 gene was normalized to the expression of the murine β-actin gene. Because we observed differences in the transcription of the murine β-actin gene between different organs, we compared levels of F8 mRNA only within the same organ. Human F8 mRNA was detected in all tissues studied, predominantly in the liver (Figure 5A). In comparison, expression of the murine F8 gene was also found in all tissues obtained from conventional C57BL/6 mice (Figure 5A). Analysis of liver and brain revealed similar expression levels for the human F8 cDNA in transgenic mice and the murine F8 gene in C57BL/6 mice. All other organs studied showed higher expression levels of the murine F8 gene in C57BL/6 mice compared with the expression levels of the human F8 cDNA in transgenic mice of subline E.

Figure 5

Transgenic mice of subline E express human F8 mRNA in a variety of different organs but express a hemophilic phenotype. (A) Expression of F8 mRNA in transgenic male (▴) and female (●) mice compared with the expression of murine F8 mRNA in wild-type C57BL/6 male (▵) and female mice (○). Total RNA was isolated from liver (li), brain (br), reproductive organs (ro), kidneys (ki), bone marrow (bm), muscle (mu), heart (he), thymus (th), lymph nodes (ln), spleen (sp), and lungs (lu) and reverse transcribed into cDNA. Expression of human and murine F8 mRNA was analyzed in relation to murine β-actin by TaqMan real-time quantitative PCR. Each data point represents results obtained from an individual mouse. (B) Blood loss after tail cut was determined over a period of 60 minutes using the tail-cut model.35  Presented are box plots representing the results obtained with groups of 16 mice. Compared are transgenic mice expressing the human F8 cDNA (huF8), conventional hemophilic E17 F8-knockout mice (E17 F8 ko), and wild-type mice (C57BL/6). ns indicates that the difference was not significant. *P < .001.

Figure 5

Transgenic mice of subline E express human F8 mRNA in a variety of different organs but express a hemophilic phenotype. (A) Expression of F8 mRNA in transgenic male (▴) and female (●) mice compared with the expression of murine F8 mRNA in wild-type C57BL/6 male (▵) and female mice (○). Total RNA was isolated from liver (li), brain (br), reproductive organs (ro), kidneys (ki), bone marrow (bm), muscle (mu), heart (he), thymus (th), lymph nodes (ln), spleen (sp), and lungs (lu) and reverse transcribed into cDNA. Expression of human and murine F8 mRNA was analyzed in relation to murine β-actin by TaqMan real-time quantitative PCR. Each data point represents results obtained from an individual mouse. (B) Blood loss after tail cut was determined over a period of 60 minutes using the tail-cut model.35  Presented are box plots representing the results obtained with groups of 16 mice. Compared are transgenic mice expressing the human F8 cDNA (huF8), conventional hemophilic E17 F8-knockout mice (E17 F8 ko), and wild-type mice (C57BL/6). ns indicates that the difference was not significant. *P < .001.

Close modal

Based on these data, we investigated whether the expression of human F8 mRNA results in the expression of human FVIII protein in the circulation of transgenic mice. We analyzed plasma samples for human FVIII antigen and human FVIII activity. The levels of human FVIII were below the detection limits for both assays (below 0.005 IU/mL and 0.006 IU/mL, respectively) indicating a lack of circulating FVIII protein.

We next investigated whether the lack of circulating human FVIII protein would correspond to a hemophilic phenotype in transgenic mice. We used the tail-cut model35  and determined the resulting amount of blood loss. Both conventional hemophilic E17 mice and human F8 transgenic mice of subline E expressed a hemophilic phenotype. In contrast, normal wild-type C57BL/6 mice expressed normal hemostasis (Figure 5B).

We conclude that transgenic mice of subline E express human F8 mRNA in a variety of different organs but do not express detectable levels of human FVIII protein in the circulation. This lack of circulating human FVIII protein corresponds to a hemophilic phenotype.

Mice of subline E develop antibodies against FVIII when treated with a modified FVIII that possesses increased immunogenicity

After demonstrating specific immune tolerance to native human FVIII, we investigated whether transgenic mice of subline E would still be able to develop antibodies against human FVIII when treated with a modified human FVIII protein that exhibits increased immunogenicity. We studied 2 PEG-FVIII proteins that contained different amounts of PEG residues (mFVIII1 and mFVIII2). From earlier studies, we already knew that mFVIII2 expresses increased immunogenicity compared with native human FVIII in conventional hemophilic E17 mice. We treated mice with 8 IV doses of either human native FVIII or one of the PEGylated FVIII preparations, and analyzed antibodies against FVIII after 4 and 8 doses. We included conventional hemophilic E17 mice as controls. Conventional hemophilic mice developed anti-FVIII antibodies after treatment with each of the 3 FVIII preparations tested (Figure 6). Native human FVIII and mFVIII1 induced similar titers of anti-FVIII antibodies, whereas mFVIII2 induced significantly higher titers of anti-FVIII antibodies, confirming our earlier observation. Anti-FVIII antibodies induced by mFVIII1 and mFVIII2 recognized both native human FVIII and the PEGylated FVIII that was used for treatment. Titers measured against native human FVIII and against the respective PEGylated FVIII were comparable. For simplicity, we only show antibody titers against native human FVIII in Figure 6.

Figure 6

Transgenic mice of subline E develop antibodies against human FVIII when treated with a modified human FVIII with high immunogenic potential. Transgenic mice (huF8) and conventional hemophilic E17 F8-knockout mice (E17F8 ko) were treated with 8 doses of either native human FVIII (FVIII) or 1 of 2 PEGylated human FVIII proteins (mFVIII1 and mFVIII2). Titers of antibodies against native human FVIII were determined by ELISA 1 week after the eighth dose. Results for individual mice are shown. ns indicates that the difference was not significant. *P < .001.

Figure 6

Transgenic mice of subline E develop antibodies against human FVIII when treated with a modified human FVIII with high immunogenic potential. Transgenic mice (huF8) and conventional hemophilic E17 F8-knockout mice (E17F8 ko) were treated with 8 doses of either native human FVIII (FVIII) or 1 of 2 PEGylated human FVIII proteins (mFVIII1 and mFVIII2). Titers of antibodies against native human FVIII were determined by ELISA 1 week after the eighth dose. Results for individual mice are shown. ns indicates that the difference was not significant. *P < .001.

Close modal

In contrast to conventional hemophilic E17 mice, human F8 transgenic mice of subline E did not develop antibodies against human FVIII when treated with either native human FVIII or mFVIII1 (Figure 6). However, transgenic mice treated with mFVIII2 developed antibodies against human FVIII that recognized both native human FVIII (Figure 6) and mFVIII2 (results not shown). These findings indicate a break of immune tolerance in mice of subline E by mFVIII2.

We then investigated whether the isotype and subclass distribution of anti-FVIII antibodies seen in transgenic mice after break of immune tolerance by mFVIII2 would be similar or different to the isotype and subclass distribution of anti-FVIII antibodies induced by native human FVIII, mFVIII1, or mFVIII2 in conventional hemophilic E17 mice. Results presented in Figure 7 indicate that the isotype and subclass distribution of anti-FVIII antibodies induced in conventional hemophilic E17 mice were similar for FVIII, mFVIII1, and mFVIII2 (Figure 7D-F). All antibody titers in conventional hemophilic E17 mice revealed a clear ranking of IgG1 → IgG2a → IgG2b → IgG2c (Figure 7D-F). However, the subclass distribution after break of immune tolerance induced by mFVIII2 in human F8 transgenic mice was different (Figure 7C). The major IgG subclass was IgG1, whereas there was no difference in titers between IgG2a, IgG2b, and IgG2c.

Figure 7

Break of immune tolerance against human FVIII in mice of subline E is not isotypically restricted. Transgenic mice expressing human F8 cDNA (huF8, A-C) and conventional hemophilic E17 F8-knockout mice (E17F8ko, D-F) were treated weekly with 9 doses of either native human FVIII (FVIII, A and D) or 1 of 2 PEGylated human FVIII proteins (mFVIII1, B and E; mFVIII2, C and F). Subclasses of IgG, IgA, and IgM antibodies specific for native human FVIII were analyzed by ELISA 1 week after the last treatment. Results of individual mice are shown.

Figure 7

Break of immune tolerance against human FVIII in mice of subline E is not isotypically restricted. Transgenic mice expressing human F8 cDNA (huF8, A-C) and conventional hemophilic E17 F8-knockout mice (E17F8ko, D-F) were treated weekly with 9 doses of either native human FVIII (FVIII, A and D) or 1 of 2 PEGylated human FVIII proteins (mFVIII1, B and E; mFVIII2, C and F). Subclasses of IgG, IgA, and IgM antibodies specific for native human FVIII were analyzed by ELISA 1 week after the last treatment. Results of individual mice are shown.

Close modal

Both human F8 transgenic mice and conventional hemophilic E17 mice developed similar levels of low-titer IgM antibodies against FVIII when treated with human native FVIII or mFVIII1, which might indicate a T cell–independent, low-titer, low-affinity antibody response (Figure 7A-F).

We also investigated whether break of immune tolerance by mFVIII2 in human F8 transgenic mice is associated with an altered pattern of cytokines released into cell-culture supernatants or with an increased proliferation after in vitro restimulation of FVIIII-specific T cells. For this purpose, we isolated spleen cells from mice treated with the different FVIII preparations and restimulated them in vitro for up to 6 days with either native human FVIII or the modified FVIII preparation that was used for treating the mice. We did not observe significant differences in cytokines (IL-4, IL-5, IL-6, IL-10, IL-17, and IFN-γ) released into culture supernatants from spleen cells obtained from human F8 transgenic mice treated with native human FVIII, mFVIII1, or mFVIII2. A similar lack of differences was seen when spleen cell proliferation was compared between the groups (Table 2). The situation was different in conventional hemophilic E17 mice, in which the results revealed a significant increase in proliferation when spleen cells obtained from mice treated with mFVIII2 were in vitro restimulated with native human FVIII (Table 2).

Table 2

Incorporation of 3H-thymidin in spleen cells restimulated for 3 days with human FVIII in vitro

Mouse strainTreatmentFVIII in culture, μg/mL3H incorporation, cpm
Stimulation index
mean (n = 3)SD
E17F8ko FVIII 5.020 460 
  8.586 2.075 1.70 
  10 9.252 2.333 1.84 
E17F8ko mFVIII1 4.758 401 
  6.272 2.210 1.32 
  10 5.518 999 1.16 
E17F8ko mFVIII2 5.666 939 
  34.257 2.788 6.05* 
  10 31.402 2.170 5.54* 
huF8 FVIII 3.647 671 
  4.002 452 1.10 
  10 4.297 1.721 1.18 
huF8 mFVIII1 5.041 505 
  6.215 502 1.23 
  10 4.714 530 0.94 
huF8 mFVIII2 4.757 268 
  4.723 466 0.99 
  10 5.767 1.167 1.21 
Mouse strainTreatmentFVIII in culture, μg/mL3H incorporation, cpm
Stimulation index
mean (n = 3)SD
E17F8ko FVIII 5.020 460 
  8.586 2.075 1.70 
  10 9.252 2.333 1.84 
E17F8ko mFVIII1 4.758 401 
  6.272 2.210 1.32 
  10 5.518 999 1.16 
E17F8ko mFVIII2 5.666 939 
  34.257 2.788 6.05* 
  10 31.402 2.170 5.54* 
huF8 FVIII 3.647 671 
  4.002 452 1.10 
  10 4.297 1.721 1.18 
huF8 mFVIII1 5.041 505 
  6.215 502 1.23 
  10 4.714 530 0.94 
huF8 mFVIII2 4.757 268 
  4.723 466 0.99 
  10 5.767 1.167 1.21 

Spleen cells were obtained from conventional hemophilic E17 F8-knockout mice or human F8-transgenic mice at 1 week after the last dose of 9 weekly treatments with either human FVIII or 1 of 2 PEGylated FVIII preparations (mFVIII1 and mFVIII2).

*

P < .01.

Summarizing our data, we conclude that human F8 transgenic mice that are immunologically tolerant to native human FVIII are still able to develop antibodies against human FVIII when treated with a modified FVIII protein that exhibits increased immunogenicity. The break of immune tolerance involves the development of different IgG subclasses with a dominance of IgG1.

Previously, we reported the development of new transgenic mouse lines that express a human F8 cDNA as a transgene.13,14  In the present study, we describe the selection of one particular transgenic mouse line that does not develop antibodies against native human FVIII given IV in therapeutic doses, but is still able to respond to human FVIII when treated with a modified FVIII preparation expressing increased immunogenicity. The lack of antibody response against native human FVIII was caused by specific immune tolerance and was not the result of a more general immune suppression. The development of specific immune tolerance is most likely caused by the deletion of high-avidity human FVIII-reactive CD4+ T cells during embryonic development, when the T-cell repertoire in the thymus is shaped and central immune tolerance is established.15,16  In addition, peripheral tolerance mechanisms such as anergy, clonal deletion, and active suppression by regulatory T cells37,38  could contribute to the immune tolerance. Both pathways of tolerance induction are initiated by the presentation of peptides through MHC class II expressed on antigen-presenting cells that requires intracellular protein or peptides. Therefore, it seems clear that human F8 mRNA is translated into FVIII protein, even though human FVIII protein cannot be detected in the circulation of transgenic mice. The question arises as to why human FVIII protein is present intracellularly but is not found in the circulation. We were able to show that human F8 cDNA is completely integrated into the mouse genome and is completely transcribed into F8 mRNA. However, the integration site of the human F8 cDNA is different from the location of the murine F8 gene, which could influence the function of the transgene and might suppress its effective transcription and translation.39  Furthermore, we used a cDNA for the construction of the transgene because the genomic sequence of the human F8 locus is very large and difficult to handle. The artificial structure of the cDNA construct could have a strong impact on its function. The loss of introns in the transgenic sequence can result in disturbances of the nucleosome alignment, which is important for the transcription of the transgene. Therefore, the expression of cDNA constructs is usually rather low.40-42  Although we observed levels of human F8 mRNA in the liver that were similar to levels of murine F8 mRNA in the livers of wild-type mice, these levels might not be sufficient to produce enough human protein to achieve detectable levels in the circulation. Furthermore, it is most likely that the human FVIII protein is mainly expressed in hepatocytes because its expression is driven by a murine albumin promoter. Hepatocytes do not produce VWF, which could facilitate a rapid degradation of FVIII after the secretion or the clearance of FVIII by clearance receptors such as members of the low-density lipoprotein receptor family.43  It has been shown previously that FVIII requires stabilization by VWF after secretion.44,45  Successful gene therapy approaches using viral vectors containing a full-length human F8 cDNA driven by an albumin promoter to achieve detectable levels of circulating human FVIII protein in hemophilic mice were described previously.46  However, stable integration of human F8 cDNA into the mouse genome cannot be compared with gene-therapy approaches that achieve reasonable levels of circulating human FVIII. Stably integrated transgenes are transmitted to the next generation via the germline. This process includes DNA demethylation and de novo methylation. Transgenic constructs are often silenced by cytosine methylation.47  This phenomenon is especially true for cDNA constructs.39  Therefore, the difference in the levels of human F8 cDNA and human F8 mRNA found after successful gene-therapy approaches and after transgenic expression of human F8 cDNA are likely to be substantial. There are several other examples of transgenic mouse models that are immunologically tolerant to specific human proteins such as insulin,17  IFNs,19,20  or factor IX,48  but lack detectable human protein in the circulation. Moreover, Connely et al49  demonstrated that the lack of circulating FVIII protein observed after gene therapy using a vector containing a human F8 cDNA driven by an albumin promoter was not necessarily associated with a lack of the production of functional human FVIII protein. Therefore, we believe that the transgenic mouse line that is immunotolerant to human FVIII does produce human FVIII protein but does not produce sufficient quantities of protein to be detectable in the circulation.

An important objective in the selection of the transgenic mouse line was the requirement that mice should still be able to develop antibodies against human FVIII when challenged with a modified human FVIII protein expressing increased immunogenicity. We tested 2 different PEGylated FVIII preparations, mFVIII1 and mFVIII2, which showed different immunogenicity profiles in conventional hemophilic E17 mice. mFVII1 and mFVIII2 mainly differed in their degree of modification. An increase in the formation of protein aggregates as the cause for the different immunogenicity profiles can be excluded. When we tested mFVIII1, mFVIII2, and native human FVIII in our new transgenic mice, mFVIII1 and native FVIII did not induce anti-FVIII antibodies, whereas mFVIII2 induced antibodies that recognized both native human FVIII and mFVIII2. mFVIII1 and mFVIII2 differed substantially in their specific activity and in their binding to VWF, which might be explained by the difference in their degree of modification. PEGylation could cause a reduced ability of mFVIII2 to function as a cofactor in the “tenase” complex and could block binding sites for VWF. It remains to be determined whether these 2 modifications to FVIII function caused mFVIII2 to break tolerance in the transgenic mouse model. Several other mechanisms could explain the break of immune tolerance by mFVIII2. Protein modification could induce new epitopes (neo-epitopes) for T cells, possibly through different processing of mFVIII2 in endo-lysosomal compartments of antigen-presenting cells. PEGylation could shield certain cleavage sites of proteolytic enzymes responsible for the generation of peptides within endo-lysosomal compartments, resulting in altered peptide patterns, which could eventually lead to the presentation of new FVIII peptide epitopes to CD4+ T cells. Alternatively, PEGylation could generate altered structures that trigger certain pattern recognition receptors expressed on innate immune cells,50  thereby activating these cells and disturbing peripheral mechanisms of immune tolerance. Our results demonstrate that treatment with mFVIII2 caused an amplification of the anti-FVIII antibody response in conventional hemophilic E17 mice, which could indicate a stimulation of the innate immune system. Break of immune tolerance by mFVIII2 could also be driven by B cells if PEGylation would induce structural changes giving rise to novel B-cell epitopes. In this case, however, some T-cell help would probably be required. Antibody production without T-cell help would only be expected if PEGylation would induce repetitive B-cell epitopes that could induce T-cell–independent antibody responses by cross-linking and triggering B-cell receptors.7,8  The induction of several subclasses of IgG antibodies, however, would suggest that the break of tolerance involved antibody class switch, which would be expected to require T-cell help.7,8  The exact mechanisms for the break of immune tolerance by mFVIII2 are currently being studied.

In summary, in the present study, we created a new transgenic mouse line with a hemophilic phenotype in which specific immune tolerance towards native human FVIII is achieved by transgenic expression of a full-length human F8 cDNA. Immune tolerance breaks down when mice are challenged with a modified human FVIII molecule that expresses increased immunogenicity. We believe that this new mouse line provides an important advantage over existing hemophilic mouse models in the preclinical immunogenicity assessment of new FVIII molecules with chemical or molecular modifications. Furthermore, this model seems suitable to study mechanisms causing break of immune tolerance against human FVIII.

An Inside Blood analysis of this article appears at the front of this issue.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors are grateful to Elisabeth Hopfner, Ingrid Neunteufl, Katharina Steinitz, Azra Trbic, Nicole Pfeffer, Thomas Wurz, Fatima Al-Awadi, Nidha Abrar, and Monika Grewal for technical assistance; to Maurus de la Rosa for critical discussion; and to John-Philip Lawo for statistical analysis. Furthermore, they acknowledge the contribution of Maria Sasgary to the development of the construct containing the human F8 cDNA and the contribution of Thomas Rülicke, University of Veterinary Medicine Vienna, who generated the first transgenic founder mouse. They also thank Elise Langdon-Neuner for editing the manuscript.

This study was supported by Baxter BioScience.

Contribution: P.M.v.H. designed research, performed the immunologic in vitro analysis, analyzed and interpreted the data, and wrote the manuscript; S.U. designed the research, did the molecular biological in vitro analysis, and analyzed and interpreted the data; C.H. designed the research and analyzed and interpreted the molecular biological data; M.S. designed and supervised the breeding and cross-breeding of transgenic mice; R.U.A. designed, supervised, and performed the animal experiments; A.N.S. designed, performed, and interpreted the tail-cut experiments; M.W. designed and performed the animal experiments; G.A. designed the molecular biological studies and interpreted these data; P.L.T. supervised the generation and analysis of mFVIII1 and mFVIII2; E.M.M. supervised the breeding of mice and the animal experiments; H.P.W. interpreted the data; and B.M.P. designed the research, analyzed and interpreted the data, and wrote the manuscript.

Conflict-of-interest disclosure: The authors are employees of Baxter BioScience.

Correspondence: Birgit M. Reipert, Baxter BioScience, Industriestrasse 72, A-1220 Vienna, Austria; e-mail: birgit_reipert@baxter.com.

1
Mannucci
 
PM
Tuddenham
 
EG
The hemophilias-from royal genes to gene therapy.
N Engl J Med
2001
, vol. 
344
 
23
(pg. 
1773
-
1779
)
2
Collins
 
PW
Bjorkman
 
S
Fischer
 
K
, et al. 
Factor VIII requirement to maintain a target plasma level in the prophylactic treatment of severe hemophilia A: influences of variance in pharmacokinetics and treatment regimens.
J Thromb Haemost
2010
, vol. 
8
 
2
(pg. 
269
-
275
)
3
Saenko
 
EL
Pipe
 
SW
Strategies towards a longer acting factor VIII.
Haemophilia
2006
, vol. 
12
 
suppl 3
(pg. 
42
-
51
)
4
Sommer
 
J
Sim
 
D
Leong
 
L
Hacket
 
F
Subramanyam
 
B
Haaning
 
J
Preservation of activity and prolonged efficacy of rFVIII after site-specific modification with high molecular weight hydroxyethyl starch.
Haemophilia
2010
, vol. 
16
 
suppl 4
pg. 
39
 
5
Dumont
 
JA
Kamphaus
 
GD
Fraley
 
C
, et al. 
Factor VIII-Fc fusion protein shows extended half-life and hemostatic activity in hemophilia A dogs [Abstract].
Blood
2009
, vol. 
114
 
11
pg. 
545
 
6
Matzinger
 
P
The danger model: a renewed sense of self.
Science
2002
, vol. 
296
 
5566
(pg. 
301
-
305
)
7
Fehr
 
T
Bachmann
 
MF
Bucher
 
E
, et al. 
Role of repetitive antigen patterns for induction of antibodies against antibodies.
J Exp Med
1997
, vol. 
185
 
10
(pg. 
1785
-
1792
)
8
Vos
 
Q
Lees
 
A
Wu
 
ZQ
Snapper
 
CM
Mond
 
JJ
B-cell activation by T-cell-independent type 2 antigens as an integral part of the humoral immune response to pathogenic microorganisms.
Immunol Rev
2000
, vol. 
176
 (pg. 
154
-
170
)
9
Levin
 
LG
Jarvis
 
M
Powell
 
J
Harrison
 
JA
Reisner
 
HM
Induction of human factor VIII inhibitors in rats 2: Fine mapping of rat anti-human rFVIII antibodies.
Thromb Haemost
1996
, vol. 
76
 
6
(pg. 
998
-
1003
)
10
Reipert
 
BM
Ahmad
 
RU
Turecek
 
PL
Schwarz
 
HP
Characterization of antibodies induced by human factor VIII in a murine knockout model of hemophilia A.
Thromb Haemost
2000
, vol. 
84
 
5
(pg. 
826
-
832
)
11
Littlewood
 
JD
Barrowcliffe
 
TW
The development and characterisation of antibodies to human factor VIII in haemophilic dogs.
Thromb Haemost
1987
, vol. 
57
 
3
(pg. 
314
-
321
)
12
Andrews
 
JL
Shirley
 
PS
Iverson
 
WO
, et al. 
Evaluation of the duration of human factor VIII expression in nonhuman primates after systemic delivery of an adenoviral vector.
Hum Gene Ther
2002
, vol. 
13
 
11
(pg. 
1331
-
1336
)
13
Sasgary
 
M
Schuster
 
M
Schwarz
 
HP
Reipert
 
BM
Antoine
 
G
Ehrlich
 
HJ
Transgenic non-human animals expressing human blood clotting factors.
Accessed July 27, 2011 
14
van Helden
 
PM
Sasgary
 
M
Unterthurner
 
S
, et al. 
immune tolerance to human factor VIII induced by liver-specific expression of a human factor VIII le [Abstract].
Blood
2008
, vol. 
112
 
11
pg. 
3387
 
15
Starr
 
TK
Jameson
 
SC
Hogquist
 
KA
Positive and negative selection of T cells.
Annu Rev Immunol
2003
, vol. 
21
 (pg. 
139
-
176
)
16
von Boehmer
 
H
Melchers
 
F
Checkpoints in lymphocyte development and autoimmune disease.
Nat Immunol
2010
, vol. 
11
 
1
(pg. 
14
-
20
)
17
Ottesen
 
JL
Nilsson
 
P
Jami
 
J
, et al. 
The potential immunogenicity of human insulin and insulin analogues evaluated in a transgenic mouse model.
Diabetologia
1994
, vol. 
37
 
12
(pg. 
1178
-
1185
)
18
Stewart
 
TA
Hollingshead
 
PG
Pitts
 
SL
Chang
 
R
Martin
 
LE
Oakley
 
H
Transgenic mice as a model to test the immunogenicity of proteins altered by site-specific mutagenesis.
Mol Biol Med
1989
, vol. 
6
 
4
(pg. 
275
-
281
)
19
Hermeling
 
S
Jiskoot
 
W
Crommelin
 
D
Bornaes
 
C
Schellekens
 
H
Development of a transgenic mouse model immune tolerant for human interferon beta.
Pharm Res
2005
, vol. 
22
 
6
(pg. 
847
-
851
)
20
Hermeling
 
S
Aranha
 
L
Damen
 
JM
, et al. 
Structural characterization and immunogenicity in wild-type and immune tolerant mice of degraded recombinant human interferon alpha2b.
Pharm Res
2005
, vol. 
22
 
12
(pg. 
1997
-
2006
)
21
Smith
 
HW
Wierda
 
D
Preclinical immunogenicity testing for recombinant therapeutic proteins.
J Immunotoxicol
2005
, vol. 
2
 
4
(pg. 
203
-
210
)
22
Bril
 
WS
van Helden
 
PM
Hausl
 
C
, et al. 
Tolerance to factor VIII in a transgenic mouse expressing human factor VIII cDNA carrying an Arg(593) to Cys substitution.
Thromb Haemost
2006
, vol. 
95
 
2
(pg. 
341
-
347
)
23
Kellendonk
 
C
Opherk
 
C
Anlag
 
K
Schutz
 
G
Tronche
 
F
Hepatocyte-specific expression of Cre recombinase.
Genesis
2000
, vol. 
26
 
2
(pg. 
151
-
153
)
24
Bi
 
L
Lawler
 
AM
Antonarakis
 
SE
, et al. 
Targeted disruption of the mouse factor VIII gene produces a model of haemophilia A.
Nat Genet
1995
, vol. 
10
 
1
(pg. 
119
-
121
)
25
Müller
 
S
Neusser
 
M
Wienberg
 
J
Towards unlimited colors for fluorescence in-situ hybridization (FISH).
Chromosome Res
2002
, vol. 
10
 
3
(pg. 
223
-
232
)
26
Pinkel
 
D
Landegent
 
J
Collins
 
C
, et al. 
Fluorescence in situ hybridization with human chromosome-specific libraries: detection of trisomy 21 and translocations of chromosome 4.
Proc Natl Acad Sci U S A
1988
, vol. 
85
 
23
(pg. 
9138
-
9142
)
27
Zalipsky
 
S
Functionalized poly(ethylene glycol) for preparation of biologically relevant conjugates.
Bioconjug Chem
1995
, vol. 
6
 
2
(pg. 
150
-
165
)
28
Kemptner
 
J
Marchetti-Deschmann
 
M
Siekmann
 
J
Turecek
 
PL
Schwarz
 
HP
Allmaier
 
G
GEMMA and MALDI-TOF MS of reactive PEGs for pharmaceutical applications.
J Pharm Biomed Anal
2010
, vol. 
52
 
4
(pg. 
432
-
437
)
29
Rottensteiner
 
H
Turecek
 
PL
Meijer
 
AB
, et al. 
The prolonged half-life of PEGylated and polysialylated FVIII in murine models might be caused by a reduced binding to the LRP1 receptor [abstract].
Hämostaseologie
2008
28
pg. 
P-02A-03
 
30
Monetti
 
C
Rottensteiner
 
H
Gritsch
 
H
Weber
 
A
Scheiflinger
 
F
Turecek
 
PL
Structural analysis of the recombinant therapeutic product rFVIII and its PEGylated variants using 2-D DIGE.
Electrophoresis
2011
, vol. 
32
 
11
(pg. 
1292
-
1301
)
31
Fraunhofer
 
W
Winter
 
G
The use of asymmetrical flow field-flow fractionation in pharmaceutics and biopharmaceutics.
Eur J Pharm Biopharm
2004
, vol. 
58
 
2
(pg. 
369
-
383
)
32
Hausl
 
C
Maier
 
E
Schwarz
 
HP
, et al. 
Long-term persistence of anti-factor VIII antibody-secreting cells in hemophilic mice after treatment with human factor VIII.
Thromb Haemost
2002
, vol. 
87
 
5
(pg. 
840
-
845
)
33
Kasper
 
CK
Aledort
 
L
Aronson
 
D
, et al. 
Proceedings: A more uniform measurement of factor VIII inhibitors.
Thromb Diath Haemorrh
1975
, vol. 
34
 
2
pg. 
612
 
34
Hausl
 
C
Ahmad
 
RU
Sasgary
 
M
, et al. 
High-dose factor VIII inhibits factor VIII-specific memory B cells in hemophilia A with factor VIII inhibitors.
Blood
2005
, vol. 
106
 
10
(pg. 
3415
-
3422
)
35
Baumgartner
 
B
Jaki
 
T
Wolfsegger
 
MJ
, et al. 
Optimization, refinement and reduction of murine in vivo experiments to assess therapeutic approaches for haemophilia A.
Lab Anim
2010
, vol. 
44
 
3
(pg. 
211
-
217
)
36
Qadura
 
M
Waters
 
B
Burnett
 
E
, et al. 
Recombinant and plasma-derived factor VIII products induce distinct splenic cytokine microenvironments in hemophilia A mice.
Blood
2009
, vol. 
114
 
4
(pg. 
871
-
880
)
37
Mueller
 
DL
Mechanisms maintaining peripheral tolerance.
Nat Immunol
2010
, vol. 
11
 
1
(pg. 
21
-
27
)
38
Wing
 
K
Sakaguchi
 
S
Regulatory T cells exert checks and balances on self tolerance and autoimmunity.
Nat Immunol
2010
, vol. 
11
 
1
(pg. 
7
-
13
)
39
Clark
 
AJ
Harold
 
G
Yull
 
FE
Mammalian cDNA and prokaryotic reporter sequences silence adjacent transgenes in transgenic mice.
Nucleic Acids Res
1997
, vol. 
25
 
5
(pg. 
1009
-
1014
)
40
Brinster
 
RL
Sandgren
 
EP
Behringer
 
RR
Palmiter
 
RD
No simple solution for making transgenic mice.
Cell
1989
, vol. 
59
 
2
(pg. 
239
-
241
)
41
Palmiter
 
RD
Sandgren
 
EP
Avarbock
 
MR
Allen
 
DD
Brinster
 
RL
Heterologous introns can enhance expression of transgenes in mice.
Proc Natl Acad Sci U S A
1991
, vol. 
88
 
2
(pg. 
478
-
482
)
42
Liu
 
K
Sandgren
 
EP
Palmiter
 
RD
Stein
 
A
Rat growth hormone gene introns stimulate nucleosome alignment in vitro and in transgenic mice.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 
17
(pg. 
7724
-
7728
)
43
Bovenschen
 
N
Mertens
 
K
Hu
 
L
Havekes
 
LM
van Vlijmen
 
BJ
LDL receptor cooperates with LDL receptor-related protein in regulating plasma levels of coagulation factor VIII in vivo.
Blood
2005
, vol. 
106
 
3
(pg. 
906
-
912
)
44
Kaufman
 
RJ
Wasley
 
LC
Dorner
 
AJ
Synthesis, processing, and secretion of recombinant human factor VIII expressed in mammalian cells.
J Biol Chem
1988
, vol. 
263
 
13
(pg. 
6352
-
6362
)
45
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
1989
, vol. 
9
 
3
(pg. 
1233
-
1242
)
46
Balagué
 
C
Zhou
 
J
Dai
 
Y
, et al. 
Sustained high-level expression of full-length human factor VIII and restoration of clotting activity in hemophilic mice using a minimal adenovirus vector.
Blood
2000
, vol. 
95
 
3
(pg. 
820
-
828
)
47
Garrick
 
D
Fiering
 
S
Martin
 
DI
Whitelaw
 
E
Repeat-induced gene silencing in mammals.
Nat Genet
1998
, vol. 
18
 
1
(pg. 
56
-
59
)
48
Sabatino
 
DE
Armstrong
 
E
Edmonson
 
S
, et al. 
Novel hemophilia B mouse models exhibiting a range of mutations in the Factor IX gene.
Blood
2004
, vol. 
104
 
9
(pg. 
2767
-
2774
)
49
Connelly
 
S
Andrews
 
JL
Gallo-Penn
 
AM
Tagliavacca
 
L
Kaufman
 
RJ
Kaleko
 
M
Evaluation of an adenoviral vector encoding full-length human factor VIII in hemophiliac mice.
Thromb Haemost
1999
, vol. 
81
 
2
(pg. 
234
-
239
)
50
Mukhopadhyay
 
S
Pluddemann
 
A
Gordon
 
S
Macrophage pattern recognition receptors in immunity, homeostasis and self-tolerance.
Adv Exp Med Biol
2009
, vol. 
653
 (pg. 
1
-
14
)
Sign in via your Institution