Antibiotic use differentially affects the risk of anti-drug antibody formation during anti-TNFα therapy in inflammatory bowel disease patients: a report from the epi-IIRN

Yuri Gorelik, Shay Freilich, Shiran Gerassy-Vainberg, Sigal Pressman, Chagit Friss, Alexandera Blatt, Gili Focht, Yiska Loewenberg Weisband, Shira Greenfeld, Revital Kariv, Nathan Lederman, Iris Dotan, Naama Geva-Zatorsky, Shai Shlomo Shen-Orr, Yechezkel Kashi, Yehuda Chowers, IIRN, Yuri Gorelik, Shay Freilich, Shiran Gerassy-Vainberg, Sigal Pressman, Chagit Friss, Alexandera Blatt, Gili Focht, Yiska Loewenberg Weisband, Shira Greenfeld, Revital Kariv, Nathan Lederman, Iris Dotan, Naama Geva-Zatorsky, Shai Shlomo Shen-Orr, Yechezkel Kashi, Yehuda Chowers, IIRN

Abstract

Objective: Anti-drug antibodies (ADA) to anti-tumour necrosis factor (anti-TNF) therapy drive treatment loss of response. An association between intestinal microbial composition and response to anti-TNF therapy was noted. We therefore aimed to assess the implications of antibiotic treatments on ADA formation in patients with inflammatory bowel disease (IBD).

Design: We analysed data from the epi-IIRN (epidemiology group of the Israeli IBD research nucleus), a nationwide registry of all patients with IBD in Israel. We included all patients treated with anti-TNF who had available ADA levels. Survival analysis with drug use as time varying covariates were used to assess the association between antibiotic use and ADA development. Next, specific pathogen and germ-free C57BL mice were treated with respective antibiotics and challenged with infliximab. ADA were assessed after 14 days.

Results: Among 1946 eligible patients, with a median follow-up of 651 days from initiation of therapy, 363 had positive ADA. Cox proportional hazard model demonstrated an increased risk of ADA development in patients who used cephalosporins (HR=1.97, 95% CI 1.58 to 2.44), or penicillins with β-lactamase inhibitors (penicillin-BLI, HR=1.4, 95% CI 1.13 to 1.74), whereas a reduced risk was noted in patients treated with macrolides (HR=0.38, 95% CI 0.16 to 0.86) or fluoroquinolones (HR=0.20, 95% CI 0.12 to 0.35). In mice exposed to infliximab, significantly increased ADA production was observed in cephalosporin as compared with macrolide pretreated mice. Germ-free mice produced no ADA.

Conclusion: ADA production is associated with the microbial composition. The risk of ADA development during anti-TNF therapy can possibly be reduced by avoidance of cephalosporins and penicillin-BLIs, or by treatment with fluoroquinolones or macrolides.

Keywords: TNF-alpha; antibiotics; inflammatory bowel disease; infliximab; intestinal microbiology.

Conflict of interest statement

Competing interests: YC received research grants, speaker and advisory fees from AbbVie; speaker and advisory fees from Janssen; grant, speaker and advisory fees from Takeda and consultancy fees from CytoReason. SSO received grant funding from Takeda, holds equity and receives consultancy fees from CytoReason. SG-V declares CytoReason advisory fees for last 3 years. RK received consultation fee, research grant, royalties or honorarium from Takeda and Pfizer. ID received consultation fee, research grant or honorarium from Janssen, AbbVie, Takeda, Pfizer, Roche/Genentech, Celgene/BMS, Arena, Neopharm, Gilead, Gallapagos, Celltrion, Ferring, Falk Pharma, MSD, DSM, Cambridge Healthcare, Sublimity, Sangamo, Nestle, Wild bio, Food Industries Association, Integran Holdings, Abbott, Altman Research

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Kaplan-Meier curves of cumulative risk of ADA development for prior use of different antibiotic classes. Tick marks represent censoring and shaded area represent 95% CI. Plot is limited to first 2000 days of follow-up. A p value for the log-rank test is presented in each plot. ADA, anti-drug antibodies; BLI, β-lactamase inhibitors.
Figure 2
Figure 2
Multivariable-adjusted HRs (represented by blue points) for anti-drug antibodies development for use of various antibiotic classes during anti-tumour necrosis factor therapy. HRs are presented on a log10 scale. Black lines represent 95% CIs. BLI, β-lactamase inhibitors.
Figure 3
Figure 3
Estimated adjusted regression coefficients for a range of number of dispensations for cephalosporins (A); penicillin-BLIs (B); macrolides (C); and fluoroquinolones (D). Shaded areas represent 95% CIs. BLIs, β-lactamase inhibitors.
Figure 4
Figure 4
Kaplan-Meier curves with risk tables of cumulative risk of ADA development for prior use of combinations of pairs from cephalosporins, penicillin-BLIs, macrolides and fluoroquinolones. Tick marks represent censoring and shaded areas represent the 95% CI. The plot is limited to the first 2000 days of follow-up. A p value for the log-rank test is presented in each plot. ADA, anti-drug antibodies; BLI, β-lactamase inhibitors.
Figure 5
Figure 5
Mean anti-drug antibodies levels in mice treated with anti-tumour necrosis factor and prior cefuroxime (n=20), azithromycin (n=21) or no antibiotic (regular control, n=25, or germ-free, n=5). ADA, anti-drug antibodies.

References

    1. Hanauer SB, Feagan BG, Lichtenstein GR, et al. . Maintenance infliximab for Crohn's disease: the accent I randomised trial. Lancet 2002;359:1541–9. 10.1016/S0140-6736(02)08512-4
    1. Rutgeerts P, Sandborn WJ, Feagan BG, et al. . Infliximab for induction and maintenance therapy for ulcerative colitis. N Engl J Med 2005;353:2462–76. 10.1056/NEJMoa050516
    1. Sandborn WJ, Hanauer SB, Rutgeerts P, et al. . Adalimumab for maintenance treatment of Crohn's disease: results of the classic II trial. Gut 2007;56:1232–9. 10.1136/gut.2006.106781
    1. Reinisch W, Sandborn WJ, Hommes DW, et al. . Adalimumab for induction of clinical remission in moderately to severely active ulcerative colitis: results of a randomised controlled trial. Gut 2011;60:780–7. 10.1136/gut.2010.221127
    1. Hazlewood GS, Rezaie A, Borman M, et al. . Comparative effectiveness of immunosuppressants and biologics for inducing and maintaining remission in Crohn's disease: a network meta-analysis. Gastroenterology 2015;148:344–54. 10.1053/j.gastro.2014.10.011
    1. Ben-Horin S, Chowers Y. Tailoring anti-TNF therapy in IBD: drug levels and disease activity. Nat Rev Gastroenterol Hepatol 2014;11:243–55. 10.1038/nrgastro.2013.253
    1. Papamichael K, Gils A, Rutgeerts P, et al. . Role for therapeutic drug monitoring during induction therapy with TNF antagonists in IBD: evolution in the definition and management of primary nonresponse. Inflamm Bowel Dis 2015;21:182–97. 10.1097/MIB.0000000000000202
    1. Gisbert JP, Panés J. Loss of response and requirement of infliximab dose intensification in Crohn's disease: a review. Am J Gastroenterol 2009;104:760–7. 10.1038/ajg.2008.88
    1. Ben-Horin S, Chowers Y. Review article: loss of response to anti-TNF treatments in Crohn's disease. Aliment Pharmacol Ther 2011;33:987–95. 10.1111/j.1365-2036.2011.04612.x
    1. Ding NS, Hart A, De Cruz P. Systematic review: predicting and optimising response to anti-TNF therapy in Crohn's disease - algorithm for practical management. Aliment Pharmacol Ther 2016;43:30–51. 10.1111/apt.13445
    1. Bar-Yoseph H, Levhar N, Selinger L, et al. . Early drug and anti-infliximab antibody levels for prediction of primary nonresponse to infliximab therapy. Aliment Pharmacol Ther 2018;47:212–8. 10.1111/apt.14410
    1. Moss AC, Brinks V, Carpenter JF. Review article: immunogenicity of anti-TNF biologics in IBD - the role of patient, product and prescriber factors. Aliment Pharmacol Ther 2013;38:1188–97. 10.1111/apt.12507
    1. Sazonovs A, Kennedy NA, Moutsianas L, et al. . HLA-DQA1*05 carriage associated with development of anti-drug antibodies to infliximab and adalimumab in patients with Crohn's disease. Gastroenterology 2020;158:189–99. 10.1053/j.gastro.2019.09.041
    1. Vermeire S, Gils A, Accossato P, et al. . Immunogenicity of biologics in inflammatory bowel disease. Therap Adv Gastroenterol 2018;11:1756283X17750355. 10.1177/1756283X17750355
    1. Bar-Yoseph H, Pressman S, Blatt A, et al. . Infliximab-Tumor necrosis factor complexes elicit formation of anti-drug antibodies. Gastroenterology 2019;157:1338–51. 10.1053/j.gastro.2019.08.009
    1. Colombel JF, Sandborn WJ, Reinisch W, et al. . Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med 2010;362:1383–95. 10.1056/NEJMoa0904492
    1. Ungar B, Chowers Y, Yavzori M, et al. . The temporal evolution of antidrug antibodies in patients with inflammatory bowel disease treated with infliximab. Gut 2014;63:1258–64. 10.1136/gutjnl-2013-305259
    1. Roblin X, Williet N, Boschetti G, et al. . Addition of azathioprine to the switch of anti-TNF in patients with IBD in clinical relapse with undetectable anti-TNF Trough levels and antidrug antibodies: a prospective randomised trial. Gut 2020;69:1206–12. 10.1136/gutjnl-2019-319758
    1. Ben-Horin S, Waterman M, Kopylov U, et al. . Addition of an immunomodulator to infliximab therapy eliminates antidrug antibodies in serum and restores clinical response of patients with inflammatory bowel disease. Clin Gastroenterol Hepatol 2013;11:444–7. 10.1016/j.cgh.2012.10.020
    1. Ungar B, Kopylov U, Engel T, et al. . Addition of an immunomodulator can reverse antibody formation and loss of response in patients treated with adalimumab. Aliment Pharmacol Ther 2017;45:276–82. 10.1111/apt.13862
    1. Kennedy NA, Heap GA, Green HD, et al. . Predictors of anti-TNF treatment failure in anti-TNF-naive patients with active luminal Crohn's disease: a prospective, multicentre, cohort study. Lancet Gastroenterol Hepatol 2019;4:341–53. 10.1016/S2468-1253(19)30012-3
    1. Lemaitre M, Kirchgesner J, Rudnichi A, et al. . Association between use of thiopurines or tumor necrosis factor antagonists alone or in combination and risk of lymphoma in patients with inflammatory bowel disease. JAMA 2017;318:1679–86. 10.1001/jama.2017.16071
    1. Kirchgesner J, Lemaitre M, Carrat F, et al. . Risk of serious and opportunistic infections associated with treatment of inflammatory bowel diseases. Gastroenterology 2018;155:337–46. 10.1053/j.gastro.2018.04.012
    1. Singh S, Facciorusso A, Dulai PS, et al. . Comparative risk of serious infections with biologic and/or immunosuppressive therapy in patients with inflammatory bowel diseases: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 2020;18:69–81. 10.1016/j.cgh.2019.02.044
    1. Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014;146:1489–99. 10.1053/j.gastro.2014.02.009
    1. Bazin T, Hooks KB, Barnetche T, et al. . Microbiota composition may predict anti-TNF alpha response in spondyloarthritis patients: an exploratory study. Sci Rep 2018;8:1–11. 10.1038/s41598-018-23571-4
    1. Zhou Y, Xu ZZ, He Y, et al. . Gut microbiota offers universal biomarkers across ethnicity in inflammatory bowel disease diagnosis and infliximab response prediction. mSystems 2018;3:188–205. 10.1128/mSystems.00188-17
    1. Dovrolis N, Michalopoulos G, Theodoropoulos GE, et al. . The interplay between mucosal microbiota composition and host gene-expression is linked with infliximab response in inflammatory bowel diseases. Microorganisms 2020;8:438. 10.3390/microorganisms8030438
    1. Estevinho MM, Rocha C, Correia L, et al. . Features of fecal and colon Microbiomes associate with responses to biologic therapies for inflammatory bowel diseases: a systematic review. Clin Gastroenterol Hepatol 2020;18:1054–69. 10.1016/j.cgh.2019.08.063
    1. Hagan T, Cortese M, Rouphael N, et al. . Antibiotics-Driven gut microbiome perturbation alters immunity to vaccines in humans. Cell 2019;178:1313–28. 10.1016/j.cell.2019.08.010
    1. Friedman MY, Leventer-Roberts M, Rosenblum J, et al. . Development and validation of novel algorithms to identify patients with inflammatory bowel diseases in Israel: an epi-IIRN group study. Clin Epidemiol 2018;10:671–81. 10.2147/CLEP.S151339
    1. FDA . Guidance for industry estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers pharmacology and toxicology guidance for industry estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers, 2005. Available:
    1. Kopylov U, Mazor Y, Yavzori M, et al. . Clinical utility of antihuman lambda chain-based enzyme-linked immunosorbent assay (ELISA) versus double antigen ELISA for the detection of anti-infliximab antibodies. Inflamm Bowel Dis 2012;18:1628–33. 10.1002/ibd.21919
    1. Helsel D. Much ado about next to nothing: incorporating nondetects in science. Ann Occup Hyg 2010;54:257–62. 10.1093/annhyg/mep092
    1. Hässler S, Bachelet D, Duhaze J, et al. . Clinicogenomic factors of biotherapy immunogenicity in autoimmune disease: a prospective multicohort study of the ABIRISK Consortium. PLoS Med 2020;17:e1003348. 10.1371/journal.pmed.1003348
    1. Ruiz VE, Battaglia T, Kurtz ZD, et al. . A single early-in-life macrolide course has lasting effects on murine microbial network topology and immunity. Nat Commun 2017;8:1–14. 10.1038/s41467-017-00531-6
    1. Kolho K-L, Korpela K, Jaakkola T, et al. . Fecal microbiota in pediatric inflammatory bowel disease and its relation to inflammation. Am J Gastroenterol 2015;110:921–30. 10.1038/ajg.2015.149
    1. Magnusson MK, Strid H, Sapnara M, et al. . Anti-Tnf therapy response in patients with ulcerative colitis is associated with colonic antimicrobial peptide expression and microbiota composition. J Crohns Colitis 2016;10:943–52. 10.1093/ecco-jcc/jjw051
    1. Zimmermann P, Curtis N. The effect of antibiotics on the composition of the intestinal microbiota - a systematic review. J Infect 2019;79:471–89. 10.1016/j.jinf.2019.10.008
    1. Zimmermann P, Ziesenitz VC, Curtis N, et al. . The immunomodulatory effects of macrolides-A systematic review of the underlying mechanisms. Front Immunol 2018;9:302. 10.3389/fimmu.2018.00302
    1. Mencarelli A, Distrutti E, Renga B, et al. . Development of non-antibiotic macrolide that corrects inflammation-driven immune dysfunction in models of inflammatory bowel diseases and arthritis. Eur J Pharmacol 2011;665:29–39. 10.1016/j.ejphar.2011.04.036
    1. Fernandez AD, Elmore MK, Metzger DW. Azithromycin modulates murine immune responses to pneumococcal conjugate vaccine and inhibits nasal clearance of bacteria. J Infect Dis 2004;190:1762–6. 10.1086/425038
    1. Ogino H, Fujii M, Ono M, et al. . In vivo and in vitro effects of fluoroquinolones on lipopolysaccharide-induced pro-inflammatory cytokine production. J Infect Chemother 2009;15:168–73. 10.1007/s10156-009-0680-1

Source: PubMed

3
Subscribe