Genome-wide pharmacogenetics of anti-drug antibody response to bococizumab highlights key residues in HLA DRB1 and DQB1

Daniel I Chasman, Craig L Hyde, Franco Giulianini, Rebecca D Danning, Ellen Q Wang, Timothy Hickling, Paul M Ridker, A Katrina Loomis, Daniel I Chasman, Craig L Hyde, Franco Giulianini, Rebecca D Danning, Ellen Q Wang, Timothy Hickling, Paul M Ridker, A Katrina Loomis

Abstract

In this largest to-date genetic analysis of anti-drug antibody (ADA) response to a therapeutic monoclonal antibody (MAb), genome-wide association was performed for five measures of ADA status among 8844 individuals randomized to bococizumab, which targets PCSK9 for LDL-C lowering and cardiovascular protection. Index associations prioritized specific amino acid substitutions at the DRB1 and DQB1 MHC class II genes rather than canonical haplotypes. Two clusters of missense variants at DRB1 were associated with general ADA measures (residues 9, 11, 13; and 96, 112, 120, 180) and a third cluster of missense variants in DQB1 was associated with ADA measures including neutralizing antibody (NAb) titers (residues 66, 67, 71, 74, 75). The structural disposition of the missense substitutions implicates peptide antigen binding and CD4 effector function, mechanisms that are potentially generalizable to other therapeutic mAbs.Clinicaltrials.gov: NCT01968954, NCT01968967, NCT01968980, NCT01975376, NCT01975389, NCT02100514.

Conflict of interest statement

C. Hyde, E. Wang, and A. K. Loomis are employees of Pfizer, Inc. T. Hickling was an employee of Pfizer, Inc. during the design and implementation of the study. P. Ridker has received research grant support from Novartis, Kowa, Amarin, Pfizer, and the NHLBI; and has served as a consultant to Corvidia, Novartis, Flame, Agepha, Inflazome, AstraZeneca, Jannsen, Civi Biopharm, SOCAR, Novo Nordisk, Uptton, and Omeicos, and Boehringer-Ingelheim. D. Chasman has received funding support from Pfizer Inc. related to the genetics of bococizumab treatment. R. Danning and F. Giulianini have no disclosures.

© 2022. The Author(s).

Figures

Figure 1
Figure 1
Genome-wide associations with anti-drug antibody response to bococizumab in SPIRE. (A) Circular Manhattan plot of genome-wide associations using genome-wide genotype data imputed to the HRC reference panel. Each ring corresponds to an anti-body response phenotype ordered from the central ring toward the perimeter as ADA positive, ADA maximum titer, ADA maximum titer top 10%, NAb positive, NAb maximum titer (see “Methods”). Thin green dashed lines indicate genome-wide significance threshold (p = 5 × 10–8) for each genome-wide scan. Red dots indicate SNPs reaching genome-wide significance. See Supplementary Figure S1 for corresponding quantile–quantile (QQ) plots. (B) Manhattan plot restricted to the MHC region (chr6:29-33 Mb) derived from MHC-specific imputation. Antibody response phenotypes as indicated are in the same order, top to bottom, as phenotypes in (A) center to perimeter. Thin green dashed line and red dots as in (A).
Figure 2
Figure 2
Effects of index SNPs across the several measures of immunogenicity. Diameters of circles are proportional to the magnitude of variant effects (i.e. beta coefficients). Circles with red outlines indicate associations reaching genome-wide significance. See also Table S5.
Figure 3
Figure 3
Mapping of index non-synonymous variants and their proxies to structural models of DR1 and DQ1. (A) ADA positive status on DR1. Index non-synonymous variant at DRB1 residue 120 and its proxies at residues 96, 112, 166, and 180 shown as red spheres are mapped onto the crystal structure of DR1 (PDB ID: 3S4S). DRA1 and DRB1 peptides are represented in light and dark gray ribbons, respectively. A 13 residue peptide in the peptide-binding grove is indicated by a green coil. The index variant and its proxies map to the β2 beta-sandwich domain of DRB1. (B) ADA positive status on DR1 with CD4. Same representation of the DRB1 index variant 96 and its proxies as in (A), but now including CD4, making contact with the β2 beta-sandwich domain of DRB1. Also shown are residues in DRB1 inferred from mutagenesis to interfere with CD4-mediated T-cell response from Brogdon et al. (residues, 46, 54, 55, 56, orange spheres) and Konig et al. (residues, 137–143, cyan spheres). (C) ADA maximum titer top 10% and NAb positive status on DQ1. Index non-synonymous variant at DQB1 residue 71 and its proxies at residues 66, 67, 74, and 75 are mapped onto the crystal structure of DQ1 (PDB ID: 6DIG), and represented as spheres alternating in darker and lighter red to aid visualization. DRA1 and DRB1 peptides are represented in light and dark gray ribbons, respectively. A 13 residue peptide in the peptide-binding grove is indicated by a green coil. (D) ADA maximum titer on DR1. Index non-synonymous variant at residue 13 and its proxies at residues 9 and 11 are mapped onto the crystal structure of DR1 (PDB ID: 6CQN), and represented as red spheres. DRA1 and DRB1 peptides are represented in light and dark gray ribbons, respectively. A 13 residue peptide in the peptide-binding grove is indicated by a green coil.

References

    1. Bachelet D, et al. Occurrence of anti-drug antibodies against interferon-beta and natalizumab in multiple sclerosis: A collaborative cohort analysis. PLoS One. 2016;11:e0162752. doi: 10.1371/journal.pone.0162752.
    1. Calabresi PA, et al. The incidence and significance of anti-natalizumab antibodies: Results from AFFIRM and SENTINEL. Neurology. 2007;69:1391–1403. doi: 10.1212/01.wnl.0000277457.17420.b5.
    1. Cassotta A, et al. A single T cell epitope drives the neutralizing anti-drug antibody response to natalizumab in multiple sclerosis patients. Nat. Med. 2019;25:1402–1407. doi: 10.1038/s41591-019-0568-2.
    1. Liang S, Zhang C. Prediction of immunogenicity for humanized and full human therapeutic antibodies. PLoS One. 2020;15:e0238150. doi: 10.1371/journal.pone.0238150.
    1. Meunier S, et al. Impact of human sequences in variable domains of therapeutic antibodies on the location of CD4 T-cell epitopes. Cell Mol. Immunol. 2020;17:656–658. doi: 10.1038/s41423-019-0304-3.
    1. Oliver-Martos B, et al. Early development of anti-natalizumab antibodies in MS patients. J. Neurol. 2013;260:2343–2347. doi: 10.1007/s00415-013-6991-2.
    1. Burmester GR, et al. Association of HLA-DRB1 alleles with clinical responses to the anti-interleukin-17A monoclonal antibody secukinumab in active rheumatoid arthritis. Rheumatology (Oxford) 2016;55:49–55. doi: 10.1093/rheumatology/kev258.
    1. Liu M, et al. Identification of HLA-DRB1 association to adalimumab immunogenicity. PLoS One. 2018;13:e0195325. doi: 10.1371/journal.pone.0195325.
    1. Billiet T, et al. Immunogenicity to infliximab is associated with HLA-DRB1. Gut. 2015;64:1344–1345. doi: 10.1136/gutjnl-2015-309698.
    1. Sazonovs A, 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;158:189–199. doi: 10.1053/j.gastro.2019.09.041.
    1. Bournazos S, Gupta A, Ravetch JV. The role of IgG Fc receptors in antibody-dependent enhancement. Nat. Rev. Immunol. 2020;20:633–643. doi: 10.1038/s41577-020-00410-0.
    1. Kastbom A, et al. Influence of FCGR3A genotype on the therapeutic response to rituximab in rheumatoid arthritis: An observational cohort study. BMJ Open. 2012 doi: 10.1136/bmjopen-2012-001524.
    1. Romero-Cara P, et al. A FCGR3A polymorphism predicts anti-drug antibodies in chronic inflammatory bowel disease patients treated with anti-TNF. Int. J. Med. Sci. 2018;15:10–15. doi: 10.7150/ijms.22812.
    1. Ternant D, et al. Assessment of the influence of inflammation and FCGR3A genotype on infliximab pharmacokinetics and time to relapse in patients with Crohn's disease. Clin. Pharmacokinet. 2015;54:551–562. doi: 10.1007/s40262-014-0225-3.
    1. Ballantyne CM, et al. Results of bococizumab, a monoclonal antibody against proprotein convertase subtilisin/kexin type 9, from a randomized, placebo-controlled, dose-ranging study in statin-treated subjects with hypercholesterolemia. Am. J. Cardiol. 2015;115:1212–1221. doi: 10.1016/j.amjcard.2015.02.006.
    1. Ridker PM, et al. Lipid-reduction variability and antidrug-antibody formation with bococizumab. N. Engl. J. Med. 2017;376:1517–1526. doi: 10.1056/NEJMoa1614062.
    1. Ridker PM, et al. Cardiovascular efficacy and safety of bococizumab in high-risk patients. N. Engl. J. Med. 2017;376:1527–1539. doi: 10.1056/NEJMoa1701488.
    1. Chang CC, et al. Second-generation PLINK: Rising to the challenge of larger and richer datasets. Gigascience. 2015;4:7. doi: 10.1186/s13742-015-0047-8.
    1. The 1000 Genomes Project Consortium et al. A global reference for human genetic variation. Nature. 2015;526:68–74. doi: 10.1038/nature15393.
    1. Das S, et al. Next-generation genotype imputation service and methods. Nat. Genet. 2016;48:1284–1287. doi: 10.1038/ng.3656.
    1. Jia X, et al. Imputing amino acid polymorphisms in human leukocyte antigens. PLoS One. 2013;8:e64683. doi: 10.1371/journal.pone.0064683.
    1. Price AL, et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 2006;38:904–909. doi: 10.1038/ng1847.
    1. Aulchenko YS, Struchalin MV, van Duijn CM. ProbABEL package for genome-wide association analysis of imputed data. BMC Bioinform. 2010;11:134. doi: 10.1186/1471-2105-11-134.
    1. Schrödinger, L. & DeLano, W. PyMOL, Available at: (2020).
    1. Galperin M, et al. CD4(+) T cell-mediated HLA class II cross-restriction in HIV controllers. Sci. Immunol. 2018 doi: 10.1126/sciimmunol.aat0687.
    1. Jiang W, et al. In vivo clonal expansion and phenotypes of hypocretin-specific CD4(+) T cells in narcolepsy patients and controls. Nat. Commun. 2019;10:5247. doi: 10.1038/s41467-019-13234-x.
    1. Wang XX, et al. Affinity maturation of human CD4 by yeast surface display and crystal structure of a CD4-HLA-DR1 complex. Proc. Natl. Acad. Sci. USA. 2011;108:15960–15965. doi: 10.1073/pnas.1109438108.
    1. Berman HM, et al. The Protein Data Bank. Nucleic Acids Res. 2000;28:235–242. doi: 10.1093/nar/28.1.235.
    1. A language and environment for statistical computing (R Foundation for Statistical Computing, Vienna, Austria, 2019).
    1. Circle Manhattan Plot v. 3.6.2 (CRAN, 2020).
    1. Mahaweni NM, et al. A comprehensive overview of FCGR3A gene variability by full-length gene sequencing including the identification of V158F polymorphism. Sci. Rep. 2018;8:15983. doi: 10.1038/s41598-018-34258-1.
    1. Erbe AK, Wang W, Gallenberger M, Hank JA, Sondel PM. Genotyping single nucleotide polymorphisms and copy number variability of the FCGRs expressed on NK Cells. Methods Mol. Biol. 2016;1441:43–56. doi: 10.1007/978-1-4939-3684-7_4.
    1. Konig R, Huang LY, Germain RN. MHC class II interaction with CD4 mediated by a region analogous to the MHC class I binding site for CD8. Nature. 1992;356:796–798. doi: 10.1038/356796a0.
    1. Brogdon J, Eckels DD, Davies C, White S, Doyle C. A site for CD4 binding in the beta 1 domain of the MHC class II protein HLA-DR1. J. Immunol. 1998;161:5472–5480.
    1. Evseeva I, Nicodemus KK, Bonilla C, Tonks S, Bodmer WF. Linkage disequilibrium and age of HLA region SNPs in relation to classic HLA gene alleles within Europe. Eur. J. Hum. Genet. 2010;18:924–932. doi: 10.1038/ejhg.2010.32.
    1. Zhang L, et al. TEPITOPEpan: Extending TEPITOPE for peptide binding prediction covering over 700 HLA-DR molecules. PLoS One. 2012;7:e30483. doi: 10.1371/journal.pone.0030483.
    1. Degoot AM, Chirove F, Ndifon W. Trans-allelic model for prediction of peptide:MHC-II interactions. Front. Immunol. 2018;9:1410. doi: 10.3389/fimmu.2018.01410.
    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;2:256–265. doi: 10.4161/mabs.2.3.11641.
    1. Lu RM, et al. Development of therapeutic antibodies for the treatment of diseases. J. Biomed. Sci. 2020;27:1. doi: 10.1186/s12929-019-0592-z.
    1. Hamze M, et al. Characterization of CD4 T cell epitopes of infliximab and rituximab identified from healthy donors. Front. Immunol. 2017;8:500. doi: 10.3389/fimmu.2017.00500.
    1. Hu X, et al. Additive and interaction effects at three amino acid positions in HLA-DQ and HLA-DR molecules drive type 1 diabetes risk. Nat. Genet. 2015;47:898–905. doi: 10.1038/ng.3353.
    1. Molineros JE, et al. Amino acid signatures of HLA Class-I and II molecules are strongly associated with SLE susceptibility and autoantibody production in Eastern Asians. PLoS Genet. 2019;15:e1008092. doi: 10.1371/journal.pgen.1008092.
    1. Raychaudhuri S, et al. Five amino acids in three HLA proteins explain most of the association between MHC and seropositive rheumatoid arthritis. Nat. Genet. 2012;44:291–296. doi: 10.1038/ng.1076.

Source: PubMed

3
Abonnieren