Lack of placental transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing, pharmacokinetic study

Xavier Mariette, Frauke Förger, Bincy Abraham, Ann D Flynn, Anna Moltó, René-Marc Flipo, Astrid van Tubergen, Laura Shaughnessy, Jeff Simpson, Marie Teil, Eric Helmer, Maggie Wang, Eliza F Chakravarty, Xavier Mariette, Frauke Förger, Bincy Abraham, Ann D Flynn, Anna Moltó, René-Marc Flipo, Astrid van Tubergen, Laura Shaughnessy, Jeff Simpson, Marie Teil, Eric Helmer, Maggie Wang, Eliza F Chakravarty

Abstract

Objectives: There is a need for effective and safe treatment during pregnancy in women with chronic inflammatory diseases. This study evaluated placental transfer of certolizumab pegol (CZP), an Fc-free anti-tumour necrosis factor drug, from CZP-treated pregnant women to their infants.

Methods: CRIB was a pharmacokinetic (PK) study of women ≥30 weeks pregnant receiving commercial CZP for a locally approved indication (last dose ≤35 days prior to delivery). Blood samples were collected from mothers, umbilical cords and infants at delivery, and infants again at weeks 4 and 8 post-delivery. CZP plasma concentrations were measured with a highly sensitive and CZP-specific electrochemiluminescence immunoassay (lower limit of quantification 0.032 μg/mL).

Results: Sixteen women entered and completed the study. Maternal CZP plasma levels at delivery were within the expected therapeutic range (median [range] 24.4 [5.0-49.4] μg/mL). Of the 16 infants, 2 were excluded from the per-protocol set: 1 due to missing data at birth and 1 due to implausible PK data. Of the remaining 14 infants, 13 had no quantifiable CZP levels at birth (<0.032 μg/mL), and 1 had a minimal CZP level of 0.042 μg/mL (infant/mother plasma ratio 0.0009); no infants had quantifiable CZP levels at weeks 4 and 8. Of 16 umbilical cord samples, 1 was excluded due to missing data; 3/15 had quantifiable CZP levels (maximum 0.048 μg/mL).

Conclusions: There was no to minimal placental transfer of CZP from mothers to infants, suggesting lack of in utero foetal exposure during the third trimester. These results support continuation of CZP treatment during pregnancy, when considered necessary.

Trial registration number: NCT02019602; Results.

Keywords: anti-tnf; psoriatic arthritis; rheumatoid arthritis; spondyloarthritis; treatment.

Conflict of interest statement

Competing interests: XM: grant/research support: Biogen, Pfizer, UCB Pharma; consultant for BMS, GSK, LFB, Pfizer, UCB Pharma. FF: grant/research support: UCB Pharma; speaker’s fees: Mepha, Roche, UCB Pharma. BA: grant/research support: Janssen-Cilag, UCB Pharma; speaker’s fees: AbbVie, American Reagent, Janssen-Cilag, UCB Pharma. AF: grant/research support: UCB Pharma. AM: grant/research support: MSD, AbbVie, Pfizer and UCB Pharma; consultant for: MSD, AbbVie, Pfizer, UCB Pharma. R-MF: grant/research support and consultant for: UCB Pharma. AvT: grant/research support: Pfizer, AbbVie, UCB Pharma, Janssen-Cilag, Celgene, Novartis; speaker’s fees: MSD, Janssen-Cilag, Pfizer; consultant for: AbbVie, Novartis, Janssen-Cilag, Pfizer. LS: employee of UCB Pharma. JS: employee of UCB Pharma. MT: employee of UCB Pharma. EH: employee of UCB Pharma. MW: employee of UCB Pharma. EC: grant/research support: UCB Pharma.

© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.

Figures

Figure 1
Figure 1
CRIB study design. aLast certolizumab pegol (CZP) dose given within 35 days prior to delivery.
Figure 2
Figure 2
Plasma CZP concentrations in mothers and infants (n=14 mother–infant pairsa). aTwo of 16 infants were excluded from the final per-protocol set: one due to missing data at birth and one due to implausible PK data (ie, data not consistent with a paediatric CZP PK model, based on the expected range of clearance, volume of distribution and subsequent elimination half-life; see online supplementary appendix); bInfant samples were collected within 24 hours post-delivery, while mother samples could be collected within 24 hours before or after delivery; c±7 days (two samples missing); d±7 days. BLQ, below the LLOQ (<0.032 μg/mL); CZP, certolizumab pegol; LLOQ, lower limit of quantification.

References

    1. Kavanaugh A, Cush JJ, Ahmed MS, et al. . Proceedings from the American College of Rheumatology Reproductive Health Summit: the management of fertility, pregnancy, and lactation in women with autoimmune and systemic inflammatory diseases. Arthritis Care Res 2015;67:313–25. 10.1002/acr.22516
    1. Chakravarty E, Clowse ME, Pushparajah DS, et al. . Family planning and pregnancy issues for women with systemic inflammatory diseases: patient and physician perspectives. BMJ Open 2014;4:e004081 10.1136/bmjopen-2013-004081
    1. Bröms G, Granath F, Linder M, et al. . Birth outcomes in women with inflammatory bowel disease: effects of disease activity and drug exposure. Inflamm Bowel Dis 2014;20:1091–8. 10.1097/MIB.0000000000000060
    1. de Man YA, Hazes JM, van der Heide H, et al. . Association of higher rheumatoid arthritis disease activity during pregnancy with lower birth weight: results of a national prospective study. Arthritis Rheum 2009;60:3196–206. 10.1002/art.24914
    1. Jakobsson GL, Stephansson O, Askling J, et al. . Pregnancy outcomes in patients with ankylosing spondylitis: a nationwide register study. Ann Rheum Dis 2016;75:1838–42. 10.1136/annrheumdis-2015-207992
    1. Mahadevan U, Sandborn WJ, Li DK, et al. . Pregnancy outcomes in women with inflammatory bowel disease: a large community-based study from Northern California. Gastroenterology 2007;133:1106–12. 10.1053/j.gastro.2007.07.019
    1. Nørgaard M, Larsson H, Pedersen L, et al. . Rheumatoid arthritis and birth outcomes: a Danish and Swedish nationwide prevalence study. J Intern Med 2010;268:329–37. 10.1111/j.1365-2796.2010.02239.x
    1. Mouyis MA, Thornton CC, Williams D, et al. . Pregnancy outcomes in patients with psoriatic arthritis. J Rheumatol 2017;44:128–9. 10.3899/jrheum.160929
    1. Jethwa H, Lam S, Giles I. O26 Does inflammatory arthritis really improve during pregnancy? A systematic review and meta-analysis. Rheumatology 2014;53(suppl 1):i40 10.1093/rheumatology/keu090.002
    1. Polachek A, Li S, Polachek IS, et al. . Psoriatic arthritis disease activity during pregnancy and the first-year postpartum. Semin Arthritis Rheum 2017;46:740–5. 10.1016/j.semarthrit.2017.01.002
    1. de Man YA, Dolhain RJ, van de Geijn FE, et al. . Disease activity of rheumatoid arthritis during pregnancy: results from a nationwide prospective study. Arthritis Rheum 2008;59:1241–8. 10.1002/art.24003
    1. Pedersen N, Bortoli A, Duricova D, et al. . The course of inflammatory bowel disease during pregnancy and postpartum: a prospective European ECCO-EpiCom Study of 209 pregnant women. Aliment Pharmacol Ther 2013;38:501–12. 10.1111/apt.12412
    1. Tracey D, Klareskog L, Sasso EH, et al. . Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol Ther 2008;117:244–79. 10.1016/j.pharmthera.2007.10.001
    1. Gisbert JP, Chaparro M. Safety of anti-TNF agents during pregnancy and breastfeeding in women with inflammatory bowel disease. Am J Gastroenterol 2013;108:1426–38. 10.1038/ajg.2013.171
    1. Hazes JM, Coulie PG, Geenen V, et al. . Rheumatoid arthritis and pregnancy: evolution of disease activity and pathophysiological considerations for drug use. Rheumatology 2011;50:1955–68. 10.1093/rheumatology/ker302
    1. Hyrich KL, Verstappen SM. Biologic therapies and pregnancy: the story so far. Rheumatology 2014;53:1377–85. 10.1093/rheumatology/ket409
    1. Baker T, Kevorkian L, Nesbitt A. FRI0162 Investigation into the binding affinity of certolizumab pegol to FcRn and functional consequences for FcRn-mediated transcytosis: comparison to infliximab, adalimumab and etanercept. Ann Rheum Dis 2013;72(Suppl 3):A426.1–A426. 10.1136/annrheumdis-2013-eular.1289
    1. Brown D, Nesbitt A, Stephens S, et al. . Lack of placental transfer and accumulation in milk of an anti‐TNF PEGylated Fab' fragment in rats: P‐0030. Inflammatory bowel diseases 2007;13:656.
    1. Förger F, Zbinden A, Villiger PM. Certolizumab treatment during late pregnancy in patients with rheumatic diseases: low drug levels in cord blood but possible risk for maternal infections. A case series of 13 patients. Joint Bone Spine 2016;83:341–3. 10.1016/j.jbspin.2015.07.004
    1. Mahadevan U, Wolf DC, Dubinsky M, et al. . Placental transfer of anti-tumor necrosis factor agents in pregnant patients with inflammatory bowel disease. Clin Gastroenterol Hepatol 2013;11:286–92. 10.1016/j.cgh.2012.11.011
    1. Porter C, Armstrong-Fisher S, Kopotsha T, et al. . Certolizumab pegol does not bind the neonatal Fc receptor (FcRn): consequences for FcRn-mediated in vitro transcytosis and ex vivo human placental transfer. J Reprod Immunol 2016;116:7–12. 10.1016/j.jri.2016.04.284
    1. Smeraglia J, Silva JP, Jones K. Improving the sensitivity and specificity of a bioanalytical assay for the measurement of certolizumab pegol. Bioanalysis 2017;9:1217–26. 10.4155/bio-2017-0124
    1. Lacroix BD, Parker GL. S1029 Dosing with certolizumab pegol (CZP) 200 mg every 2 weeks (Q2W) provides higher plasma trough concentrations than 400 mg every 4 weeks (Q4W). Gastroenterology 2010;138:S-163–4. 10.1016/S0016-5085(10)60749-4
    1. Wade JR, Parker G, Kosutic G, et al. . Population pharmacokinetic analysis of certolizumab pegol in patients with Crohn’s disease. J Clin Pharmacol 2015;55:866–74. 10.1002/jcph.491
    1. Götestam Skorpen C, Hoeltzenbein M, Tincani A, et al. . The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis 2016;75:795–810. 10.1136/annrheumdis-2015-208840
    1. Mahadevan U, Cucchiara S, Hyams JS, et al. . The London Position Statement of the World Congress of Gastroenterology on Biological Therapy for IBD with the European Crohn’s and Colitis Organisation: pregnancy and pediatrics. Am J Gastroenterol 2011;106:214–23. 10.1038/ajg.2010.464
    1. Flint J, Panchal S, Hurrell A, et al. . BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding-Part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids. Rheumatology 2016;55:1693–7. 10.1093/rheumatology/kev404
    1. Kliegman R, Stanton B, Saint Geme J, et al. . Nelson Textbook of Pediatrics. 20th ed: Elsevier Health Sciences, 2015.
    1. Fomsgaard A, Svenson M, Bendtzen K. Auto-antibodies to tumour necrosis factor alpha in healthy humans and patients with inflammatory diseases and gram-negative bacterial infections. Scand J Immunol 1989;30:219–23. 10.1111/j.1365-3083.1989.tb01204.x
    1. Arsenescu R, Arsenescu V, de Villiers WJ. TNF-α and the development of the neonatal immune system: implications for inhibitor use in pregnancy. Am J Gastroenterol 2011;106:559–62. 10.1038/ajg.2011.5
    1. Pasparakis M, Alexopoulou L, Episkopou V, et al. . Immune and inflammatory responses in TNF alpha-deficient mice: a critical requirement for TNF alpha in the formation of primary B cell follicles, follicular dendritic cell networks and germinal centers, and in the maturation of the humoral immune response. J Exp Med 1996;184:1397–411. 10.1084/jem.184.4.1397
    1. Wen L, Shinton SA, Hardy RR, et al. . Association of B-1 B cells with follicular dendritic cells in spleen. J Immunol 2005;174:6918–26. 10.4049/jimmunol.174.11.6918
    1. Holsapple MP, West LJ, Landreth KS. Species comparison of anatomical and functional immune system development. Birth Defects Res B Dev Reprod Toxicol 2003;68:321–34. 10.1002/bdrb.10035
    1. Martin PL, Oneda S, Treacy G. Effects of an anti-TNF-alpha monoclonal antibody, administered throughout pregnancy and lactation, on the development of the macaque immune system. Am J Reprod Immunol 2007;58:138–49. 10.1111/j.1600-0897.2007.00499.x
    1. Julsgaard M, Christensen LA, Gibson PR, et al. . Concentrations of adalimumab and infliximab in mothers and newborns, and effects on infection. Gastroenterology 2016;151:110–9. 10.1053/j.gastro.2016.04.002
    1. Mahadevan U, Martin C, Kane SV, et al. . 437 Do infant serum levels of biologic agents at birth correlate with risk of adverse outcomes? Results from the PIANO registry. Gastroenterology 2016;150:S91–S92. 10.1016/S0016-5085(16)30422-X
    1. Komaki F, Komaki Y, Micic D, et al. . Outcome of pregnancy and neonatal complications with anti-tumor necrosis factor-α use in females with immune mediated diseases; a systematic review and meta-analysis. J Autoimmun 2017;76:38–52. 10.1016/j.jaut.2016.11.004
    1. Mozaffari S, Abdolghaffari AH, Nikfar S, et al. . Pregnancy outcomes in women with inflammatory bowel disease following exposure to thiopurines and antitumor necrosis factor drugs: a systematic review with meta-analysis. Hum Exp Toxicol 2015;34:445–59. 10.1177/0960327114550882
    1. Clowse MEB, Scheuerle AE, Chambers CD, et al. . Characteristics and outcomes of prospectively reported pregnancies exposed to certolizumab pegol from a safety database. Arthritis Rheumatol 2017;69(Suppl 10). [abstract 1309].

Source: PubMed

3
S'abonner