Abstract 3323

Baxter is developing a PEGylated recombinant human factor VIII conjugate (BAX 855) based on the modification of full-length FVIII with polyethylene glycol (PEG). The FVIII molecule is derived from a CHO cell line using a plasma-protein-free method.

Any chemical modification of FVIII might alter its immunogenic potential. Therefore, careful preclinical immunogenicity studies of PEGylated FVIII in comparison to unmodified FVIII are important. We evaluated the immunogenicity of BAX 855 in comparison to ADVATE, Baxter‘s unmodified recombinant full-length FVIII concentrate. For this purpose, we used different in vitro and in vivo models to s assess the potential impact of BAX 855 and ADVATE on both the innate and the adaptive immune system.

The assessment of the potential impact of BAX 855 and ADVATE on the human innate immune system was based on their potential to induce the release of pro- inflammatory cytokines (IL-1β, IL-6, IL-8 and TNF-α) in an in vitro human whole blood assay and on its potential to activate the human complement system in human plasma in vitro.

The assessment of the potential impact of BAX 855 and ADVATE on the adaptive immune system was based on their potential to induce antibodies in 3 different hemophilic mouse models and in cynomolgous monkeys. The hemophilic moue models included a hemophilic mouse model with a knockout of the murine FVIII that express a human F8 cDNA as a transgene, a hemophilic mouse model that expresses the human MHC-class II protein HLA-DRB1*1501 on the background of a knockout of the murine MHC-class II complex and a conventional hemophilic mouse model with a knockout of the murine FVIII gene. Hemophilic mice that express a human F8 cDNA as a transgene are immunologically tolerant to human FVIII and develop antibodies against human FVIII only if the immune tolerance breaks down. Using this model, we asked if BAX 855 is able to maintain immune tolerance to human FVIII. The hemophilic mouse models that expresses the human MHC-class II protein HLA-DRB1*1501 mimics the situation in an important fraction of hemophilia A patients with FVIII inhibitors. The human MHC-class II haplotype HLA-DRB1*1501 was previously shown to be associated with an increased risk for patients to develop FVIII inhibitors. Using this model, we asked if BAX 855 expresses an immunogenicity profile similar to ADVATE.

BAX 855 and ADAVTE induced similar low levels of cytokine release (IL-1β, IL-6, IL-8 and TNF-α) that were similar to the cytokine release observed in the buffer control group after incubation with human whole blood in vitro. In addition, BAX 855 and ADVATE induced similar low levels of complement activation that were not different from the buffer control group after incubation with human plasma in vitro.

Importantly, BAX 855 and ADVATE induced similar levels and incidences of antibodies against FVIII and against PEG-FVIII in all mouse models. In addition, immune tolerance to FVIII was maintained in hemophilic mice expressing the human F8 cDNA. Finally, no major differences in antibody titers were seen after treatment of cynomolgus monkeys with BAX 855 and ADVATE.

We conclude that results obtained in comparative immunogenicity studies in vitro and in vivo demonstrate that BAX 855 and ADVATE have a similar immunogenicity profile. Therefore, we expect that BAX 855 will express a similar safety profile as ADVATE in patients.

Disclosures:

Horling:Baxter Innovations GmbH: Employment. Schwele:Baxter Innovations GmbH: Employment. Lubich:Baxter BioScience: Employment. Ahmad:Baxter Innovations GmbH: Employment. Weiller:Baxter BioScience: Employment. Spatzenegger:Baxter Innovation GmbH: Employment. Schwarz:Baxter Innovations GmbH: Employment. Reipert:Baxter Innovations GmbH: Employment.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution