Discovery and characterization of COVA322, a clinical-stage bispecific TNF/IL-17A inhibitor for the treatment of inflammatory diseases

Michela Silacci, Wibke Lembke, Richard Woods, Isabella Attinger-Toller, Nadja Baenziger-Tobler, Sarah Batey, Roger Santimaria, Ulrike von der Bey, Susann Koenig-Friedrich, Wenjuan Zha, Bernd Schlereth, Mathias Locher, Julian Bertschinger, Dragan Grabulovski, Michela Silacci, Wibke Lembke, Richard Woods, Isabella Attinger-Toller, Nadja Baenziger-Tobler, Sarah Batey, Roger Santimaria, Ulrike von der Bey, Susann Koenig-Friedrich, Wenjuan Zha, Bernd Schlereth, Mathias Locher, Julian Bertschinger, Dragan Grabulovski

Abstract

Biologic treatment options such as tumor necrosis factor (TNF) inhibitors have revolutionized the treatment of inflammatory diseases, including rheumatoid arthritis. Recent data suggest, however, that full and long-lasting responses to TNF inhibitors are limited because of the activation of the pro-inflammatory TH17/interleukin (IL)-17 pathway in patients. Therefore, dual TNF/IL-17A inhibition is an attractive avenue to achieve superior efficacy levels in such diseases. Based on the marketed anti-TNF antibody adalimumab, we generated the bispecific TNF/IL-17A-binding FynomAb COVA322. FynomAbs are fusion proteins of an antibody and a Fyn SH3-derived binding protein. COVA322 was characterized in detail and showed a remarkable ability to inhibit TNF and IL-17A in vitro and in vivo. Through its unique mode-of-action of inhibiting simultaneously TNF and the IL-17A homodimer, COVA322 represents a promising drug candidate for the treatment of inflammatory diseases. COVA322 is currently being tested in a Phase 1b/2a study in psoriasis ( ClinicalTrials.gov Identifier: NCT02243787).

Keywords: FynomAb; IL-17; TNF; bispecific antibody; inflammatory diseases.

Figures

Figure 1.
Figure 1.
Characterization of COVA322 (A) Schematic picture of COVA322 showing that the anti-IL-17A Fynomer (orange circle) was genetically fused to the C terminus of the light chain of the anti-TNF antibody adalimumab. (B) COVA322 was expressed transiently in CHO cells and purified using protein A. The resulting protein was >95 % pure and monomeric, as determined by size exclusion chromatography over a period of at least 2 months in a non-optimized phosphate buffered saline buffer. (C) Dual TNF and IL-17A binding of COVA322 is shown using immobilized IL-17A on a BIAcore chip with subsequent injection of COVA322 and TNF (red) or COVA322 only (black) (D) COVA322 and the control anti-IL-17A antibody secukinumab were tested in a cell assay after stimulation with IL-17A and IL-1β. COVA322 and the control antibody specifically inhibited IL-17A with IC50 values of 121 pM and 470 pM, respectively. Mean values of triplicates are shown, error bars represent standard deviations (SD) (E) Gro-α ELISA levels in the supernatants are shown after the stimulation of HT-29 cells with 200 pM TNF and 400 pM IL-17A. After addition of the indicated concentrations of COVA322, a dose-dependent inhibition of IL-17A and TNF can be observed because the Gro-α levels decline with higher concentrations of the inhibitors. As controls, cells were treated with the single cytokines (IL-17A or TNF) or with medium only. Mean values of triplicates are shown, error bars represent standard deviations (SD). (F) COVA322 mediated inhibition of T-cell derived IL-17A induced IL-6 release from HT-1080 cells, comparable to the control anti-IL-17A antibody secukinumab in the presence of excess of anti-TNF antibody adalimumab (Adal.) and anti-IL-1β antibody canakinumab (Cana.). Mean values of quadruplicates are shown, error bars represent standard deviations (SD).
Figure 2.
Figure 2.
Plasma concentration of COVA322 in cynomolgus monkeys. (A) Bifunctional ELISA was performed to detect intact COVA322. Biotinylated TNF was immobilized in the wells of neutravidin-coated microtiter 96-well plates. Plasma containing COVA322 was added to the wells. For detection, digoxigenin-labeled IL-17A was used, followed by an anti-digoxigenin antibody-HRP conjugate for substrate processing and color development. Monofunctional ELISA was performed to detect specific TNF binding of COVA322, adalimumab or golimumab. Biotinylated TNF was immobilized in the wells of neutravidin-coated microtiter 96-well plates. Plasma containing COVA322 or anti-TNF antibody was added to the wells. TNF binding compound was detected using an anti-human IgG antibody-HRP-conjugate for substrate processing and color development. (B) Plasma concentrations of COVA322 and the anti-TNF antibodies adalimumab and golimumab at different time-points after a single iv injection into cynomolgus monkeys. The COVA322 or anti-TNF antibody concentration in plasma was determined by Bifunctional and Monofunctional ELISA, respectively. Mean plasma concentrations of 3 cynomolgus monkeys are plotted versus time, error bars represent standard deviations (SD). (C) Comparison of the plasma concentrations of COVA322 in cynomolgus monkeys determined by Bifunctional and Monofunctional ELISA. The plasma concentrations obtained from both assays are comparable, indicating that COVA322 is stable in cynomolgus monkeys for at least 220 hours.
Figure 3.
Figure 3.
Inhibition of human IL-17A and TNF in vivo. Mice were injected intravenously (iv) with COVA322, anti-IL-17A antibody secukinumab or anti-TNF antibody adalimumab followed by subcutaneous injection of human TNF (A) or human IL-17A (B). Two hours after the administration of the indicated cytokine, blood samples were taken from the mice and KC levels were determined by ELISA. As a control, basal KC levels are shown (mice treated with PBS only (iv), without cytokine stimulation). Mean KC levels of 5 mice per group are shown (± SEM).

References

    1. Radner H, Smolen JS, Aletaha D. Remission in rheumatoid arthritis: benefit over low disease activity in patient-reported outcomes and costs. Arthritis Res Therapy 2014; 16:R56;
    1. Rubbert-Roth A, Finckh A. Treatment options in patients with rheumatoid arthritis failing initial TNF inhibitor therapy: a critical review. Arthritis Res Therapy 2009; 11 Suppl 1:S1;
    1. Smolen JS, Landewe R, Breedveld FC, Buch M, Burmester G, Dougados M, Emery P, Gaujoux-Viala C, Gossec L, Nam J, et al.. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis 2014; 73:492-509; PMID:24161836;
    1. Geiler J, Buch M, McDermott MF. Anti-TNF treatment in rheumatoid arthritis. Curr Pharmaceutical Design 2011; 17:3141-54;
    1. Villeneuve E, Haraoui B. To switch or to change class-the biologic dilemma in rheumatoid arthritis. Nat Rev Rheumatol 2010; 6:301-5; PMID:20386564;
    1. Benedetti G, Miossec P. Interleukin 17 contributes to the chronicity of inflammatory diseases such as rheumatoid arthritis. Eur J Immunol 2014; 44:339-47; PMID:24310226;
    1. Koenders MI, Marijnissen RJ, Devesa I, Lubberts E, Joosten LA, Roth J, van Lent PL, van de Loo FA, van den Berg WB. Tumor necrosis factor-interleukin-17 interplay induces S100A8, interleukin-1beta, and matrix metalloproteinases, and drives irreversible cartilage destruction in murine arthritis: rationale for combination treatment during arthritis. Arthritis Rheum 2011; 63:2329-39; PMID:21520013;
    1. Alzabin S, Abraham SM, Taher TE, Palfreeman A, Hull D, McNamee K, Jawad A, Pathan E, Kinderlerer A, Taylor PC, et al.. Incomplete response of inflammatory arthritis to TNFalpha blockade is associated with the Th17 pathway. Ann Rheum Dis 2012; 71:1741-8; PMID:22550316;
    1. Grabulovski D, Kaspar M, Neri D. A novel, non-immunogenic Fyn SH3-derived binding protein with tumor vascular targeting properties. J Biol Chem 2007; 282:3196-204; PMID:17130124;
    1. Bertschinger J, Grabulovski D, Neri D. Selection of single domain binding proteins by covalent DNA display. Protein Eng Des Sel 2007; 20:57-68; PMID:17242027;
    1. Schlatter D, Brack S, Banner DW, Batey S, Benz J, Bertschinger J, Huber W, Joseph C, Rufer A, van der Klooster A, et al.. Generation, characterization and structural data of chymase binding proteins based on the human Fyn kinase SH3 domain. MAbs 2012; 4:497-508; PMID:22653218;
    1. Banner DW, Gsell B, Benz J, Bertschinger J, Burger D, Brack S, Cuppuleri S, Debulpaep M, Gast A, Grabulovski D, et al.. Mapping the conformational space accessible to BACE2 using surface mutants and cocrystals with Fab fragments, Fynomers and Xaperones. Acta Crystallogr D Biol Crystallogr 2013; 69:1124-37; PMID:23695257;
    1. Brack S, Attinger-Toller I, Schade B, Mourlane F, Klupsch K, Woods R, Hachemi H, von der Bey U, Koenig-Friedrich S, Bertschinger J, et al.. A bispecific HER2-targeting FynomAb with superior antitumor activity and novel mode of action. Mol Cancer Therapeutics 2014; 13:2030-9;
    1. Silacci M, Baenziger-Tobler N, Lembke W, Zha W, Batey S, Bertschinger J, Grabulovski D. Linker length matters, fynomer-Fc fusion with an optimized linker displaying picomolar IL-17A inhibition potency. J Biol Chem 2014; 289:14392-8; PMID:24692552;
    1. Urech DM, Feige U, Ewert S, Schlosser V, Ottiger M, Polzer K, Schett G, Lichtlen P. Anti-inflammatory and cartilage-protecting effects of an intra-articularly injected anti-TNF{α} single-chain Fv antibody (ESBA105) designed for local therapeutic use. Ann Rheum Dis 2010; 69:443-9; PMID:19293161;
    1. Gerhardt S, Abbott WM, Hargreaves D, Pauptit RA, Davies RA, Needham MR, Langham C, Barker W, Aziz A, Snow MJ, et al.. Structure of IL-17A in complex with a potent, fully human neutralizing antibody. J Mol Biol 2009; 394:905-21; PMID:19835883;
    1. Deng R, Loyet KM, Lien S, Iyer S, DeForge LE, Theil FP, Lowman HB, Fielder PJ, Prabhu S. Pharmacokinetics of humanized monoclonal anti-tumor necrosis factor-{α} antibody and its neonatal Fc receptor variants in mice and cynomolgus monkeys. Drug Metab Dispos 2010; 38:600-5; PMID:20071453;
    1. Fischer JA, Hueber AJ, Wilson S, Galm M, Baum W, Kitson C, Auer J, Lorenz SH, Moelleken J, Bader M, et al.. Combined Inhibition of Tumor Necrosis Factor α and Interleukin-17 As a Therapeutic Opportunity in Rheumatoid Arthritis: Development and Characterization of a Novel Bispecific Antibody. Arthritis Rheumatol 2015; 67:51-62; PMID:25303306;
    1. Genovese MC, Cohen S, Moreland L, Lium D, Robbins S, Newmark R, Bekker P, Study G. Combination therapy with etanercept and anakinra in the treatment of patients with rheumatoid arthritis who have been treated unsuccessfully with methotrexate. Arthritis Rheum 2004; 50:1412-9; PMID:15146410;
    1. Weinblatt M, Schiff M, Goldman A, Kremer J, Luggen M, Li T, Chen D, Becker JC. Selective costimulation modulation using abatacept in patients with active rheumatoid arthritis while receiving etanercept: a randomised clinical trial. Ann Rheum Dis 2007; 66:228-34; PMID:16935912;
    1. Thomaidis T, Worns MA, Galle PR, Mohler M, Schattenberg JM. Treatment of malignant ascites with a second cycle of catumaxomab in gastric signet cell carcinoma - a report of 2 cases. Oncology Res Treatment 2014; 37:674-7;
    1. Topp MS, Gokbuget N, Stein AS, Zugmaier G, O'Brien S, Bargou RC, Dombret H, Fielding AK, Heffner L, Larson RA, et al.. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol 2014; 16(1):57-66; PMID:25524800;
    1. Muller D, Kontermann RE. Bispecific antibodies for cancer immunotherapy: Current perspectives. BioDrugs 2010; 24:89-98; PMID:20199124;
    1. Wu C, Ying H, Grinnell C, Bryant S, Miller R, Clabbers A, Bose S, McCarthy D, Zhu RR, Santora L, et al.. Simultaneous targeting of multiple disease mediators by a dual-variable-domain immunoglobulin. Nat Biotechnol 2007; 25:1290-7; PMID:17934452;
    1. Viti F, Nilsson F, Demartis S, Huber A, Neri D. Design and use of phage display libraries for the selection of antibodies and enzymes. Methods Enzymol 2000; 326:480-505; PMID:11036659;

Source: PubMed

3
Se inscrever