Evaluation of Antibody Properties and Clinically Relevant Immunogenicity, Anaphylaxis, and Hypersensitivity Reactions in Two Phase III Trials of Tralokinumab in Severe, Uncontrolled Asthma

Mats Carlsson, Martin Braddock, Yuling Li, Jihong Wang, Weichen Xu, Nicholas White, Ayman Megally, Gillian Hunter, Gene Colice, Mats Carlsson, Martin Braddock, Yuling Li, Jihong Wang, Weichen Xu, Nicholas White, Ayman Megally, Gillian Hunter, Gene Colice

Abstract

Introduction: Tralokinumab is a monoclonal antibody (mAb) that neutralizes interleukin (IL)-13, a cytokine involved in the pathogenesis of asthma.

Objective: The objectives of this study were to characterize the potential immunogenic properties of tralokinumab and report data for anti-drug antibodies (ADAs) and hypersensitivity reactions from two phase III clinical trials.

Methods: The oligosaccharide structure of tralokinumab, Fab-arm exchange, and ADAs were characterized by standard techniques. Hypersensitivity adverse events (AEs) were evaluated in two pivotal clinical trials of tralokinumab in severe, uncontrolled asthma: STRATOS 1 and 2 (NCT02161757 and NCT02194699).

Results: No galactose-α-1,3-galactose (α-Gal) epitopes were found in the Fab region of tralokinumab and only 4.5% of glycoforms contained α-Gal in the Fc region. Under non-reducing conditions, Fab-arm exchange did not take place with another immunoglobulin (Ig) G4 mAb (mavrilimumab). However, following glutathione reduction, a hybrid antibody with monovalent bioactivity was detected. ADA incidences (titers) were as follows: STRATOS 1-every 2 weeks (Q2 W) 0.8% (26.0), every 4 weeks (Q4 W) 0.5% (26.0), placebo 0.8% (52.0); STRATOS 2-Q2 W 1.2% (39.0), placebo 0.8% (13.0). Participant-reported hypersensitivity AE rates were as follows: STRATOS 1-Q2 W 25.9%, Q4 W 25.0%, placebo 25.5%; STRATOS 2-Q2 W 13.2%, placebo 9.0%. External evaluation for anaphylaxis by Sampson criteria found no tralokinumab-related severe hypersensitivity or anaphylaxis reactions.

Conclusion: Preclinical assessments suggested a low likelihood of immunogenicity for tralokinumab. In STRATOS 1 and 2, ADA incidence was low, no differences were found between tralokinumab-treated and placebo groups in reporting of hypersensitivity reactions, and there were no Sampson criteria-evaluated anaphylaxis events with tralokinumab treatment. Together, the results suggest that tralokinumab treatment would not increase the risk for severe hypersensitivity or anaphylactic reactions.

Conflict of interest statement

Conflicts of interest

Yuling Li was an employee of MedImmune, the biologics division of AstraZeneca, at the time the study took place; her current affiliation is CBT Pharmaceuticals. Jihong Wang and Weichen Xu are employees of MedImmune, the biologics division of AstraZeneca. Nicholas White is an employee of MedImmune and an AstraZeneca shareholder. Mats Carlsson, Martin Braddock, and Ayman Megally are employees of AstraZeneca, the sponsor of STRATOS 1 and STRATOS 2. Gene Colice is an employee of and holds stock options in AstraZeneca. Gillian Hunter is a consultant employed by AstraZeneca.

Ethical approval and informed consent

STRATOS 1 and STRATOS 2 were performed in accordance with the ethical principles that have their origin in the Declaration of Helsinki and that are consistent with International Conference on Harmonisation (ICH)/Good Clinical Practice (GCP), applicable regulatory requirements, and the AstraZeneca policy on Bioethics. All participating centers obtained approval from an independent ethics committee and all participants provided written informed consent.

Figures

Fig. 1
Fig. 1
Oligosaccharide profile of tralokinumab. Separation of 2-aminobenzamide labeled glycans from tralokinumab by hydrophilic interaction liquid chromatography. f fucose, G galactose, GN N-acetyl-glucosamine, M mannose, NGc N-glycolylneuraminic acid
Fig. 2
Fig. 2
Structure and nomenclature of oligosaccharides in the tralokinumab Fab region. f fucose, G galactose, GlcNAc N-acetylglucosamine, Man mannose, NGc N-glycolylneuraminic acid
Fig. 3
Fig. 3
Characterization of tralokinumab with different degrees of reduction by non-reducing gel electrophoresis: (a) non-reduced; (b) reduced; and (c) partially reduced. HHL two heavy chains + one light chain, IgG immunoglobulin G
Fig. 4
Fig. 4
Ion exchange chromatography profiles of tralokinumab and mavrilimumab Fab-arm exchange under (a) physiological conditions and (b) non-physiological conditions
Fig. 5
Fig. 5
Patient flow diagram for (a) STRATOS 1 and (b) STRATOS 2. aThe STRATOS 1 placebo treatment group is a combined treatment group (placebo every 2 weeks plus placebo every 4 weeks), where the two placebo cohorts were given weights proportional to the number of participants in each cohort. For STRATOS 2, eight participants on tralokinumab every 2 weeks and 11 on placebo were excluded from the full analysis set. bTwo participants were duplicates and were not included in the safety set; these two participants and five who did not have the potential to receive 52 weeks of treatment were not included in the full analysis set. cFive participants did not have the potential to receive 52 weeks of treatment, so were not included in the full analysis set. dOf participants randomly assigned. Republished with permission of Elsevier, from Panettieri et al. [7]; permission conveyed through the Copyright Clearance Center, Inc

References

    1. May RD, Monk PD, Cohen ES, Manuel D, Dempsey F, Davis NH, et al. Preclinical development of CAT-354, an IL-13 neutralizing antibody, for the treatment of severe uncontrolled asthma. Br J Pharmacol. 2012;166(1):177–193. doi: 10.1111/j.1476-5381.2011.01659.x.
    1. Popovic B, Breed J, Rees DG, Gardender MJ, Vinall LMK, Kemp B, et al. Structural characterisation reveals mechanism of IL-13 neutralising monoclonal antibody tralokinumab as inhibition of binding to IL-13Rα1 and IL-13Rα2. J Mol Biol. 2017;429:208–219. doi: 10.1016/j.jmb.2016.12.005.
    1. Corren J. Role of interleukin-13 in asthma. Curr Allergy Asthma Rep. 2013;13(5):415–420. doi: 10.1007/s11882-013-0373-9.
    1. Wills-Karp M. Interleukin-13 in asthma pathogenesis. Curr Allergy Asthma Rep. 2004;4(2):123–131. doi: 10.1007/s11882-004-0057-6.
    1. Panettieri R, Wang M, Braddock M, Bowen K, Colice G. Tralokinumab for the treatment of severe, uncontrolled asthma: the ATMOSPHERE clinical development program. Immunotherapy. 2018;10(6):473–490. doi: 10.2217/imt-2017-0191.
    1. Panettieri RA, Jr, Brightling C, Sjobring U, Péterffy A, Tornling G, Daoud SZ, et al. STRATOS 1 and 2: considerations in clinical trial design for a fully human monoclonal antibody in severe asthma. Clin Invest (Lond). 2015;5(8):701–711. doi: 10.4155/cli.15.38.
    1. Panettieri R, Sjobring U, Péterffy A, Wessman P, Bowen K, Piper E, et al. Tralokinumab for severe, uncontrolled asthma (STRATOS 1 and STRATOS 2): two randomised, double-blind, placebo-controlled, phase 3 clinical trials. Lancet Respir Med. 2018;6(7):511–525. doi: 10.1016/S2213-2600(18)30184-X.
    1. Harding FA, Stickler MM, Razo J, DuBridge RB. The immunogenicity of humanized and fully human antibodies: residual immunogenicity resides in the CDR regions. MAbs. 2010;2(3):256–265. doi: 10.4161/mabs.2.3.11641.
    1. Singh SK. Impact of product-related factors on immunogenicity of biotherapeutics. J Pharm Sci. 2011;100(2):354–387. doi: 10.1002/jps.22276.
    1. Chung CH, Mirakhur B, Chan E, Le QT, Berlin J, Morse M, et al. Cetuximab-induced anaphylaxis and IgE specific for galactose-alpha-1,3-galactose. N Engl J Med. 2008;358(11):1109–1117. doi: 10.1056/NEJMoa074943.
    1. Steinke JW, Platts-Mills TA, Commins SP. The alpha-gal story: lessons learned from connecting the dots. J Allergy Clin Immunol. 2015;135(3):589–596. doi: 10.1016/j.jaci.2014.12.1947.
    1. Eli Lilly. ERBITUX® (cetuximab): prescribing information. May 2018. . Accessed 4 Jun 2018.
    1. Bosques CJ, Collins BE, Meador JW, 3rd, Sarvaiya H, Murphy JL, Dellorusso G, et al. Chinese hamster ovary cells can produce galactose-alpha-1,3-galactose antigens on proteins. Nat Biotechnol. 2010;28(11):1153–1156. doi: 10.1038/nbt1110-1153.
    1. Burton DR, Wilson IA. Immunology. Square-dancing antibodies. Science. 2007;317(5844):1507–1508.
    1. Salfeld JG. Isotype selection in antibody engineering. Nat Biotechnol. 2007;25:1369. doi: 10.1038/nbt1207-1369.
    1. Jefferis R, Lefranc MP. Human immunoglobulin allotypes: possible implications for immunogenicity. MAbs. 2009;1(4):332–338. doi: 10.4161/mabs.1.4.9122.
    1. Angal S, King DJ, Bodmer MW, Turner A, Lawson AD, Roberts G, et al. A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (IgG4) antibody. Mol Immunol. 1993;30(1):105–108. doi: 10.1016/0161-5890(93)90432-B.
    1. Aalberse RC, Schuurman J. IgG4 breaking the rules. Immunology. 2002;105(1):9–19. doi: 10.1046/j.0019-2805.2001.01341.x.
    1. Labrijn AF, Buijsse AO, van den Bremer ET, Verwilligen AY, Bleeker WK, Thorpe SJ, et al. Therapeutic IgG4 antibodies engage in Fab-arm exchange with endogenous human IgG4 in vivo. Nat Biotechnol. 2009;27(8):767–771. doi: 10.1038/nbt.1553.
    1. van der Neut Kolfschoten M, Schuurman J, Losen M, Bleeker WK, Martinez-Martinez P, Vermeulen E, et al. Anti-inflammatory activity of human IgG4 antibodies by dynamic Fab arm exchange. Science. 2007;317(5844):1554–1557. doi: 10.1126/science.1144603.
    1. Liu H, May K. Disulfide bond structures of IgG molecules: structural variations, chemical modifications and possible impacts to stability and biological function. MAbs. 2012;4(1):17–23. doi: 10.4161/mabs.4.1.18347.
    1. Krishna M, Nadler SG. Immunogenicity to biotherapeutics—the role of anti-drug immune complexes. Front Immunol. 2016;7:21. doi: 10.3389/fimmu.2016.00021.
    1. Ruhaak LR, Zauner G, Huhn C, Bruggink C, Deelder AM, Wuhrer M. Glycan labeling strategies and their use in identification and quantification. Anal Bioanal Chem. 2010;397(8):3457–3481. doi: 10.1007/s00216-010-3532-z.
    1. Food and Drug Administration. Assay Development and Validation for Immunogenicity Testing of Therapeutic Protein Products. 2016. . Accessed 30 Jul 2018.
    1. European Medicines Agency. Guideline on Immunogenicity assessment of therapeutic proteins. 2017. . Accessed 21 Aug 2018.
    1. Sampson HA, Munoz-Furlong A, Campbell RL, Adkinson NF, Jr, Bock SA, Branum A, et al. Second symposium on the definition and management of anaphylaxis: summary report–Second National Institute of Allergy and Infectious Disease/Food Allergy and Anaphylaxis Network symposium. J Allergy Clin Immunol. 2006;117(2):391–397. doi: 10.1016/j.jaci.2005.12.1303.
    1. Qian J, Liu T, Yang L, Daus A, Crowley R, Zhou Q. Structural characterization of N-linked oligosaccharides on monoclonal antibody cetuximab by the combination of orthogonal matrix-assisted laser desorption/ionization hybrid quadrupole-quadrupole time-of-flight tandem mass spectrometry and sequential enzymatic digestion. Anal Biochem. 2007;364(1):8–18. doi: 10.1016/j.ab.2007.01.023.
    1. van Bueren JJL, Rispens T, Verploegen S, van der Palen-Merkus T, Stapel S, Workman LJ, et al. Anti-galactose-α-1,3-galactose IgE from allergic patients does not bind α-galactosylated glycans on intact therapeutic antibody Fc domains. Nat Biotechnol. 2011;29:574.
    1. Janin-Bussat MC, Tonini L, Huillet C, Colas O, Klinguer-Hamour C, Corvaia N, et al. Cetuximab Fab and Fc N-glycan fast characterization using IdeS digestion and liquid chromatography coupled to electrospray ionization mass spectrometry. Methods Mol Biol. 2013;988:93–113. doi: 10.1007/978-1-62703-327-5_7.
    1. Gao B, Long C, Lee W, Zhang Z, Gao X, Landsittel D, et al. Anti-Neu5Gc and anti-non-Neu5Gc antibodies in healthy humans. PLoS One. 2017;12(7):e0180768. doi: 10.1371/journal.pone.0180768.
    1. Zhu A, Hurst R. Anti-N-glycolylneuraminic acid antibodies identified in healthy human serum. Xenotransplantation. 2002;9(6):376–381. doi: 10.1034/j.1399-3089.2002.02138.x.
    1. Kanda Y, Yamada T, Mori K, Okazaki A, Inoue M, Kitajima-Miyama K, et al. Comparison of biological activity among nonfucosylated therapeutic IgG1 antibodies with three different N-linked Fc oligosaccharides: the high-mannose, hybrid, and complex types. Glycobiology. 2007;17(1):104–118. doi: 10.1093/glycob/cwl057.
    1. Wright A, Morrison SL. Effect of altered CH2-associated carbohydrate structure on the functional properties and in vivo fate of chimeric mouse-human immunoglobulin G1. J Exp Med. 1994;180(3):1087–1096. doi: 10.1084/jem.180.3.1087.
    1. Wright A, Sato Y, Okada T, Chang K, Endo T, Morrison S. In vivo trafficking and catabolism of IgG1 antibodies with Fc associated carbohydrates of differing structure. Glycobiology. 2000;10(12):1347–1355. doi: 10.1093/glycob/10.12.1347.
    1. Yu M, Brown D, Reed C, Chung S, Lutman J, Stefanich E, et al. Production, characterization, and pharmacokinetic properties of antibodies with N-linked mannose-5 glycans. MAbs. 2012;4(4):475–487. doi: 10.4161/mabs.20737.
    1. Zhou Q, Shankara S, Roy A, Qiu H, Estes S, McVie-Wylie A, et al. Development of a simple and rapid method for producing non-fucosylated oligomannose containing antibodies with increased effector function. Biotechnol Bioeng. 2008;99(3):652–65.
    1. Luong M, Lam JS, Chen J, Levitz SM. Effects of fungal N- and O-linked mannosylation on the immunogenicity of model vaccines. Vaccine. 2007;25(22):4340–4344. doi: 10.1016/j.vaccine.2007.03.027.
    1. Almond RJ, Flanagan BF, Kimber I, Dearman RJ. Influence of protein expression system on elicitation of IgE antibody responses: experience with lactoferrin. Toxicology. 2012;301(1–3):50–57. doi: 10.1016/j.tox.2012.06.018.
    1. Tatsumi Y, Sasahara Y, Kohyama N, Ayano S, Endo M, Yoshida T, et al. Introducing site-specific glycosylation using protein engineering techniques reduces the immunogenicity of beta-lactoglobulin. Biosci Biotechnol Biochem. 2012;76(3):478–485. doi: 10.1271/bbb.110753.
    1. Jefferis R. Posttranslational modifications and the immunogenicity of biotherapeutics. J Immunol Res. 2016;2016:5358272. doi: 10.1155/2016/5358272.
    1. Schuurman J, Van Ree R, Perdok GJ, Van Doorn HR, Tan KY, Aalberse RC. Normal human immunoglobulin G4 is bispecific: it has two different antigen-combining sites. Immunology. 1999;97(4):693–698. doi: 10.1046/j.1365-2567.1999.00845.x.
    1. Koneczny I, Stevens JA, De Rosa A, Huda S, Huijbers MG, Saxena A, et al. IgG4 autoantibodies against muscle-specific kinase undergo Fab-arm exchange in myasthenia gravis patients. J Autoimmun. 2017;77:104–115. doi: 10.1016/j.jaut.2016.11.005.
    1. Broug E, Bland-Ward PA, Powell J, Johnson KS. Fab-arm exchange. Nat Biotechnol. 2010;28(2):123–126. doi: 10.1038/nbt0210-123.
    1. Mullins RJ, Wainstein BK, Barnes EH, Liew WK, Campbell DE. Increases in anaphylaxis fatalities in Australia from 1997 to 2013. Clin Exp Allergy. 2016;46(8):1099–1110. doi: 10.1111/cea.12748.
    1. Turner PJ, Gowland MH, Sharma V, Ierodiakonou D, Harper N, Garcez T, et al. Increase in anaphylaxis-related hospitalizations but no increase in fatalities: an analysis of United Kingdom national anaphylaxis data, 1992–2012. J Allergy Clin Immunol. 2015;135(4):956–963. doi: 10.1016/j.jaci.2014.10.021.
    1. Hanania NA, Korenblat P, Chapman KR, Bateman ED, Kopecky P, Paggiaro P, et al. Efficacy and safety of lebrikizumab in patients with uncontrolled asthma (LAVOLTA I and LAVOLTA II): replicate, Phase 3, randomised, double-blind, placebo-controlled trials. Lancet Respir Med. 2016;4(10):781–796. doi: 10.1016/S2213-2600(16)30265-X.
    1. O’Neil BH, Allen R, Spigel DR, Stinchcombe TE, Moore DT, Berlin JD, et al. High incidence of cetuximab-related infusion reactions in Tennessee and North Carolina and the association with atopic history. J Clin Oncol. 2007;25(24):3644–3648. doi: 10.1200/JCO.2007.11.7812.
    1. Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351(4):337–345. doi: 10.1056/NEJMoa033025.
    1. Merck. ERBITUX® (cetuximab): summary of product characteristics. 19 Jul 2017. . Accessed 4 Jun 2018.
    1. Merck SDL. SIMPONI® (golimumab): summary of product characteristics. 19 Apr 2018. . Accessed 25 May 2018.
    1. Biogen Abbevie. ZINBRYTA® (daclizumab) prescribing information. 2017. . Accessed 16 Jul 2018.
    1. Janssen-Cilag Ltd. STELARA® (ustekinumab): summay of product characteristics. 22 Mar 2018. . Accessed 25 May 2018.
    1. Novartis. ILARIS® (canakinumab): prescribing information. 2013. . Accessed 20 Nov 2017.
    1. Janssen Biotech, Inc. Golimumab prescribing information. 2018. . Accessed 13 Jun 2018.
    1. EMA. Ilaris-H-C-1109-II-0010: EPAR—assessment report—variation. 2013. . Accessed 13 Jun 2018.
    1. Zhang J, Li W, Roskos LK, Yang H. Immunogenicity assay cut point determination using nonparametric tolerance limit. J Immunol Methods. 2017;442:29–34. doi: 10.1016/j.jim.2017.01.001.
    1. European Medicines Agency. Guideline on similar biological medicinal products containing monoclonal antibodies—non-clinical and clinical issues. 30 May 2012. . Accessed 6 Dec 2018.

Source: PubMed

3
Prenumerera