[HTML][HTML] Preclinical characterization of DEKAVIL (F8-IL10), a novel clinical-stage immunocytokine which inhibits the progression of collagen-induced arthritis

K Schwager, M Kaspar, F Bootz, R Marcolongo… - Arthritis research & …, 2009 - Springer
K Schwager, M Kaspar, F Bootz, R Marcolongo, E Paresce, D Neri, E Trachsel
Arthritis research & therapy, 2009Springer
Introduction In this article, we present a comparative immunohistochemical evaluation of four
clinical-stage antibodies (L19, F16, G11 and F8) directed against splice isoforms of
fibronectin and of tenascin-C for their ability to stain synovial tissue alterations in rheumatoid
arthritis patients. Furthermore we have evaluated the therapeutic potential of the most
promising antibody, F8, fused to the anti-inflammatory cytokine interleukin (IL) 10. Methods
F8-IL10 was produced and purified to homogeneity in CHO cells and shown to comprise …
Introduction
In this article, we present a comparative immunohistochemical evaluation of four clinical-stage antibodies (L19, F16, G11 and F8) directed against splice isoforms of fibronectin and of tenascin-C for their ability to stain synovial tissue alterations in rheumatoid arthritis patients. Furthermore we have evaluated the therapeutic potential of the most promising antibody, F8, fused to the anti-inflammatory cytokine interleukin (IL) 10.
Methods
F8-IL10 was produced and purified to homogeneity in CHO cells and shown to comprise biological active antibody and cytokine moieties by binding assays on recombinant antigen and by MC/9 cell proliferation assays. We have also characterized the ability of F8-IL10 to inhibit arthritis progression in the collagen-induced arthritis mouse model.
Results
The human antibody F8, specific to the extra-domain A of fibronectin, exhibited the strongest and most homogenous staining pattern in synovial biopsies and was thus selected for the development of a fully human fusion protein with IL10 (F8-IL10, also named DEKAVIL). Following radioiodination, F8-IL10 was able to selectively target arthritic lesions and tumor neo-vascular structures in mice, as evidenced by autoradiographic analysis and quantitative biodistribution studies. The subcutaneous administration route led to equivalent targeting results when compared with intravenous administration and was thus selected for the clinical development of the product. F8-IL10 potently inhibited progression of established arthritis in the collagen-induced mouse model when tested alone and in combination with methotrexate. In preparation for clinical trials in patients with rheumatoid arthritis, F8-IL10 was studied in rodents and in cynomolgus monkeys, revealing an excellent safety profile at doses tenfold higher than the planned starting dose for clinical phase I trials.
Conclusions
Following the encouraging preclinical results presented in this paper, clinical trials with F8-IL10 will now elucidate the therapeutic potential of this product and whether the targeted delivery of IL10 potentiates the anti-arthritic action of the cytokine in rheumatoid arthritis patients.
Springer