Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta: a GWAS

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Standard

Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta : a GWAS. / Andlauer, Till F M; Link, Jenny; Martin, Dorothea; Ryner, Malin; Hermanrud, Christina; Grummel, Verena; Auer, Michael; Hegen, Harald; Aly, Lilian; Gasperi, Christiane; Knier, Benjamin; Müller-Myhsok, Bertram; Jensen, Poul Erik Hyldgaard; Sellebjerg, Finn; Kockum, Ingrid; Olsson, Tomas; Pallardy, Marc; Spindeldreher, Sebastian; Deisenhammer, Florian; Fogdell-Hahn, Anna; Hemmer, Bernhard; ABIRISK Consortium.

I: BMC Medicine, Bind 18, 298, 2020.

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Harvard

Andlauer, TFM, Link, J, Martin, D, Ryner, M, Hermanrud, C, Grummel, V, Auer, M, Hegen, H, Aly, L, Gasperi, C, Knier, B, Müller-Myhsok, B, Jensen, PEH, Sellebjerg, F, Kockum, I, Olsson, T, Pallardy, M, Spindeldreher, S, Deisenhammer, F, Fogdell-Hahn, A, Hemmer, B & ABIRISK Consortium 2020, 'Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta: a GWAS', BMC Medicine, bind 18, 298. https://doi.org/10.1186/s12916-020-01769-6

APA

Andlauer, T. F. M., Link, J., Martin, D., Ryner, M., Hermanrud, C., Grummel, V., Auer, M., Hegen, H., Aly, L., Gasperi, C., Knier, B., Müller-Myhsok, B., Jensen, P. E. H., Sellebjerg, F., Kockum, I., Olsson, T., Pallardy, M., Spindeldreher, S., Deisenhammer, F., ... ABIRISK Consortium (2020). Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta: a GWAS. BMC Medicine, 18, [298]. https://doi.org/10.1186/s12916-020-01769-6

Vancouver

Andlauer TFM, Link J, Martin D, Ryner M, Hermanrud C, Grummel V o.a. Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta: a GWAS. BMC Medicine. 2020;18. 298. https://doi.org/10.1186/s12916-020-01769-6

Author

Andlauer, Till F M ; Link, Jenny ; Martin, Dorothea ; Ryner, Malin ; Hermanrud, Christina ; Grummel, Verena ; Auer, Michael ; Hegen, Harald ; Aly, Lilian ; Gasperi, Christiane ; Knier, Benjamin ; Müller-Myhsok, Bertram ; Jensen, Poul Erik Hyldgaard ; Sellebjerg, Finn ; Kockum, Ingrid ; Olsson, Tomas ; Pallardy, Marc ; Spindeldreher, Sebastian ; Deisenhammer, Florian ; Fogdell-Hahn, Anna ; Hemmer, Bernhard ; ABIRISK Consortium. / Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta : a GWAS. I: BMC Medicine. 2020 ; Bind 18.

Bibtex

@article{19e08609f4fc4575975b7f40c428c912,
title = "Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta: a GWAS",
abstract = "BACKGROUND: Upon treatment with biopharmaceuticals, the immune system may produce anti-drug antibodies (ADA) that inhibit the therapy. Up to 40% of multiple sclerosis patients treated with interferon β (IFNβ) develop ADA, for which a genetic predisposition exists. Here, we present a genome-wide association study on ADA and predict the occurrence of antibodies in multiple sclerosis patients treated with different interferon β preparations.METHODS: We analyzed a large sample of 2757 genotyped and imputed patients from two cohorts (Sweden and Germany), split between a discovery and a replication dataset. Binding ADA (bADA) levels were measured by capture-ELISA, neutralizing ADA (nADA) titers using a bioassay. Genome-wide association analyses were conducted stratified by cohort and treatment preparation, followed by fixed-effects meta-analysis.RESULTS: Binding ADA levels and nADA titers were correlated and showed a significant heritability (47% and 50%, respectively). The risk factors differed strongly by treatment preparation: The top-associated and replicated variants for nADA presence were the HLA-associated variants rs77278603 in IFNβ-1a s.c.- (odds ratio (OR) = 3.55 (95% confidence interval = 2.81-4.48), p = 2.1 × 10-26) and rs28366299 in IFNβ-1b s.c.-treated patients (OR = 3.56 (2.69-4.72), p = 6.6 × 10-19). The rs77278603-correlated HLA haplotype DR15-DQ6 conferred risk specifically for IFNβ-1a s.c. (OR = 2.88 (2.29-3.61), p = 7.4 × 10-20) while DR3-DQ2 was protective (OR = 0.37 (0.27-0.52), p = 3.7 × 10-09). The haplotype DR4-DQ3 was the major risk haplotype for IFNβ-1b s.c. (OR = 7.35 (4.33-12.47), p = 1.5 × 10-13). These haplotypes exhibit large population-specific frequency differences. The best prediction models were achieved for ADA in IFNβ-1a s.c.-treated patients. Here, the prediction in the Swedish cohort showed AUC = 0.91 (0.85-0.95), sensitivity = 0.78, and specificity = 0.90; patients with the top 30% of genetic risk had, compared to patients in the bottom 30%, an OR = 73.9 (11.8-463.6, p = 4.4 × 10-6) of developing nADA. In the German cohort, the AUC of the same model was 0.83 (0.71-0.92), sensitivity = 0.80, specificity = 0.76, with an OR = 13.8 (3.0-63.3, p = 7.5 × 10-4).CONCLUSIONS: We identified several HLA-associated genetic risk factors for ADA against interferon β, which were specific for treatment preparations and population backgrounds. Genetic prediction models could robustly identify patients at risk for developing ADA and might be used for personalized therapy recommendations and stratified ADA screening in clinical practice. These analyses serve as a roadmap for genetic characterizations of ADA against other biopharmaceutical compounds.",
keywords = "Adult, Female, Genome-Wide Association Study/methods, Humans, Interferon-beta/immunology, Male, Middle Aged, Retrospective Studies, Risk Factors",
author = "Andlauer, {Till F M} and Jenny Link and Dorothea Martin and Malin Ryner and Christina Hermanrud and Verena Grummel and Michael Auer and Harald Hegen and Lilian Aly and Christiane Gasperi and Benjamin Knier and Bertram M{\"u}ller-Myhsok and Jensen, {Poul Erik Hyldgaard} and Finn Sellebjerg and Ingrid Kockum and Tomas Olsson and Marc Pallardy and Sebastian Spindeldreher and Florian Deisenhammer and Anna Fogdell-Hahn and Bernhard Hemmer and {ABIRISK Consortium}",
year = "2020",
doi = "10.1186/s12916-020-01769-6",
language = "English",
volume = "18",
journal = "BMC Medicine",
issn = "1741-7015",
publisher = "BioMed Central Ltd.",

}

RIS

TY - JOUR

T1 - Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta

T2 - a GWAS

AU - Andlauer, Till F M

AU - Link, Jenny

AU - Martin, Dorothea

AU - Ryner, Malin

AU - Hermanrud, Christina

AU - Grummel, Verena

AU - Auer, Michael

AU - Hegen, Harald

AU - Aly, Lilian

AU - Gasperi, Christiane

AU - Knier, Benjamin

AU - Müller-Myhsok, Bertram

AU - Jensen, Poul Erik Hyldgaard

AU - Sellebjerg, Finn

AU - Kockum, Ingrid

AU - Olsson, Tomas

AU - Pallardy, Marc

AU - Spindeldreher, Sebastian

AU - Deisenhammer, Florian

AU - Fogdell-Hahn, Anna

AU - Hemmer, Bernhard

AU - ABIRISK Consortium

PY - 2020

Y1 - 2020

N2 - BACKGROUND: Upon treatment with biopharmaceuticals, the immune system may produce anti-drug antibodies (ADA) that inhibit the therapy. Up to 40% of multiple sclerosis patients treated with interferon β (IFNβ) develop ADA, for which a genetic predisposition exists. Here, we present a genome-wide association study on ADA and predict the occurrence of antibodies in multiple sclerosis patients treated with different interferon β preparations.METHODS: We analyzed a large sample of 2757 genotyped and imputed patients from two cohorts (Sweden and Germany), split between a discovery and a replication dataset. Binding ADA (bADA) levels were measured by capture-ELISA, neutralizing ADA (nADA) titers using a bioassay. Genome-wide association analyses were conducted stratified by cohort and treatment preparation, followed by fixed-effects meta-analysis.RESULTS: Binding ADA levels and nADA titers were correlated and showed a significant heritability (47% and 50%, respectively). The risk factors differed strongly by treatment preparation: The top-associated and replicated variants for nADA presence were the HLA-associated variants rs77278603 in IFNβ-1a s.c.- (odds ratio (OR) = 3.55 (95% confidence interval = 2.81-4.48), p = 2.1 × 10-26) and rs28366299 in IFNβ-1b s.c.-treated patients (OR = 3.56 (2.69-4.72), p = 6.6 × 10-19). The rs77278603-correlated HLA haplotype DR15-DQ6 conferred risk specifically for IFNβ-1a s.c. (OR = 2.88 (2.29-3.61), p = 7.4 × 10-20) while DR3-DQ2 was protective (OR = 0.37 (0.27-0.52), p = 3.7 × 10-09). The haplotype DR4-DQ3 was the major risk haplotype for IFNβ-1b s.c. (OR = 7.35 (4.33-12.47), p = 1.5 × 10-13). These haplotypes exhibit large population-specific frequency differences. The best prediction models were achieved for ADA in IFNβ-1a s.c.-treated patients. Here, the prediction in the Swedish cohort showed AUC = 0.91 (0.85-0.95), sensitivity = 0.78, and specificity = 0.90; patients with the top 30% of genetic risk had, compared to patients in the bottom 30%, an OR = 73.9 (11.8-463.6, p = 4.4 × 10-6) of developing nADA. In the German cohort, the AUC of the same model was 0.83 (0.71-0.92), sensitivity = 0.80, specificity = 0.76, with an OR = 13.8 (3.0-63.3, p = 7.5 × 10-4).CONCLUSIONS: We identified several HLA-associated genetic risk factors for ADA against interferon β, which were specific for treatment preparations and population backgrounds. Genetic prediction models could robustly identify patients at risk for developing ADA and might be used for personalized therapy recommendations and stratified ADA screening in clinical practice. These analyses serve as a roadmap for genetic characterizations of ADA against other biopharmaceutical compounds.

AB - BACKGROUND: Upon treatment with biopharmaceuticals, the immune system may produce anti-drug antibodies (ADA) that inhibit the therapy. Up to 40% of multiple sclerosis patients treated with interferon β (IFNβ) develop ADA, for which a genetic predisposition exists. Here, we present a genome-wide association study on ADA and predict the occurrence of antibodies in multiple sclerosis patients treated with different interferon β preparations.METHODS: We analyzed a large sample of 2757 genotyped and imputed patients from two cohorts (Sweden and Germany), split between a discovery and a replication dataset. Binding ADA (bADA) levels were measured by capture-ELISA, neutralizing ADA (nADA) titers using a bioassay. Genome-wide association analyses were conducted stratified by cohort and treatment preparation, followed by fixed-effects meta-analysis.RESULTS: Binding ADA levels and nADA titers were correlated and showed a significant heritability (47% and 50%, respectively). The risk factors differed strongly by treatment preparation: The top-associated and replicated variants for nADA presence were the HLA-associated variants rs77278603 in IFNβ-1a s.c.- (odds ratio (OR) = 3.55 (95% confidence interval = 2.81-4.48), p = 2.1 × 10-26) and rs28366299 in IFNβ-1b s.c.-treated patients (OR = 3.56 (2.69-4.72), p = 6.6 × 10-19). The rs77278603-correlated HLA haplotype DR15-DQ6 conferred risk specifically for IFNβ-1a s.c. (OR = 2.88 (2.29-3.61), p = 7.4 × 10-20) while DR3-DQ2 was protective (OR = 0.37 (0.27-0.52), p = 3.7 × 10-09). The haplotype DR4-DQ3 was the major risk haplotype for IFNβ-1b s.c. (OR = 7.35 (4.33-12.47), p = 1.5 × 10-13). These haplotypes exhibit large population-specific frequency differences. The best prediction models were achieved for ADA in IFNβ-1a s.c.-treated patients. Here, the prediction in the Swedish cohort showed AUC = 0.91 (0.85-0.95), sensitivity = 0.78, and specificity = 0.90; patients with the top 30% of genetic risk had, compared to patients in the bottom 30%, an OR = 73.9 (11.8-463.6, p = 4.4 × 10-6) of developing nADA. In the German cohort, the AUC of the same model was 0.83 (0.71-0.92), sensitivity = 0.80, specificity = 0.76, with an OR = 13.8 (3.0-63.3, p = 7.5 × 10-4).CONCLUSIONS: We identified several HLA-associated genetic risk factors for ADA against interferon β, which were specific for treatment preparations and population backgrounds. Genetic prediction models could robustly identify patients at risk for developing ADA and might be used for personalized therapy recommendations and stratified ADA screening in clinical practice. These analyses serve as a roadmap for genetic characterizations of ADA against other biopharmaceutical compounds.

KW - Adult

KW - Female

KW - Genome-Wide Association Study/methods

KW - Humans

KW - Interferon-beta/immunology

KW - Male

KW - Middle Aged

KW - Retrospective Studies

KW - Risk Factors

U2 - 10.1186/s12916-020-01769-6

DO - 10.1186/s12916-020-01769-6

M3 - Journal article

C2 - 33143745

VL - 18

JO - BMC Medicine

JF - BMC Medicine

SN - 1741-7015

M1 - 298

ER -

ID: 261587494