Skip to main content
Top
Published in: Arthritis Research & Therapy 1/2020

Open Access 01-12-2020 | Adalimumab | Research article

Interleukin-6 receptor blockade or TNFα inhibition for reducing glycaemia in patients with RA and diabetes: post hoc analyses of three randomised, controlled trials

Authors: Mark C. Genovese, Gerd R. Burmester, Owen Hagino, Karthinathan Thangavelu, Melitza Iglesias-Rodriguez, Gregory St John, Miguel A. González-Gay, Thomas Mandrup-Poulsen, Roy Fleischmann

Published in: Arthritis Research & Therapy | Issue 1/2020

Login to get access

Abstract

Background

Diabetes is common in patients with rheumatoid arthritis (RA). Interleukin (IL)-6 is implicated in both the pathogenesis of RA and in glucose homeostasis; this post hoc analysis investigated the effects of IL-6 receptor vs. tumour necrosis factor inhibition on glycosylated haemoglobin (HbA1c) in patients with RA with or without diabetes.

Methods

Data were from two placebo-controlled phase III studies of subcutaneous sarilumab 150/200 mg q2w + methotrexate or conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) and a phase III monotherapy study of sarilumab 200 mg q2w vs. adalimumab 40 mg q2w. Patients with diabetes were identified by medical history or use of antidiabetic medication (patients with HbA1c ≥ 9% were excluded from all three studies). HbA1c was measured at baseline and weeks 12/24. Safety and efficacy were assessed in RA patients with or without diabetes.

Results

Patients with diabetes (n = 184) were older, weighed more and exhibited higher RA disease activity than patients without diabetes (n = 1928). Regardless of diabetes status, in patients on background csDMARDs, least squares (LS) mean difference (95% CI) in change from baseline in HbA1c for sarilumab 150 mg/200 mg vs. placebo at week 24 was − 0.28 (− 0.40, − 0.16; nominal p <  0.0001) and − 0.42 (− 0.54, − 0.31; nominal p <  0.0001), respectively. Without csDMARDs, LS mean difference for sarilumab 200 mg vs. adalimumab 40 mg at week 24 was − 0.13 (− 0.22, − 0.04; nominal p = 0.0043). Greater reduction in HbA1c than placebo or adalimumab was observed at week 24 with sarilumab in patients with diabetes and/or baseline HbA1c ≥ 7%. There was no correlation between baseline/change from baseline in HbA1c and baseline/change from baseline in C-reactive protein, 28-joint Disease Activity Score, or haemoglobin, nor between HbA1c change from baseline and baseline glucocorticoid use. Medical history of diabetes or use of diabetes treatments had limited impact on safety and efficacy of sarilumab and was consistent with overall phase III findings in patients with RA.

Conclusions

In post hoc analyses, sarilumab was associated with a greater reduction in HbA1c than csDMARDs or adalimumab, independent of sarilumab anti-inflammatory effects. Prospective studies are required to further assess these preliminary findings.

Trial registration

ClinTrials.gov NCT01061736: date of registration February 03, 2010; ClinTrials.gov NCT01709578: date of registration October 18, 2012; ClinTrials.gov NCT02332590: date of registration January 07, 2015.
Appendix
Available only for authorised users
Literature
1.
go back to reference Albrecht K, Luque Ramos A, Hoffmann F, et al. High prevalence of diabetes in patients with rheumatoid arthritis: results from a questionnaire survey linked to claims data. Rheumatology (Oxford). 2018;57(2):329–36.CrossRef Albrecht K, Luque Ramos A, Hoffmann F, et al. High prevalence of diabetes in patients with rheumatoid arthritis: results from a questionnaire survey linked to claims data. Rheumatology (Oxford). 2018;57(2):329–36.CrossRef
2.
go back to reference Maradit-Kremers H, Nicola PJ, Crowson CS, et al. Cardiovascular death in rheumatoid arthritis: a population-based study. Arthritis Rheum. 2005;52(3):722–32.PubMedCrossRef Maradit-Kremers H, Nicola PJ, Crowson CS, et al. Cardiovascular death in rheumatoid arthritis: a population-based study. Arthritis Rheum. 2005;52(3):722–32.PubMedCrossRef
3.
go back to reference Nicolau J, Lequerre T, Bacquet H, et al. Rheumatoid arthritis, insulin resistance, and diabetes. Joint Bone Spine. 2017;84(4):411–6.PubMedCrossRef Nicolau J, Lequerre T, Bacquet H, et al. Rheumatoid arthritis, insulin resistance, and diabetes. Joint Bone Spine. 2017;84(4):411–6.PubMedCrossRef
4.
go back to reference Xu G, Liu B, Sun Y, et al. Prevalence of diagnosed type 1 and type 2 diabetes among US adults in 2016 and 2017: population based study. BMJ. 2018;362:k1497.PubMedPubMedCentralCrossRef Xu G, Liu B, Sun Y, et al. Prevalence of diagnosed type 1 and type 2 diabetes among US adults in 2016 and 2017: population based study. BMJ. 2018;362:k1497.PubMedPubMedCentralCrossRef
5.
go back to reference Giles JT, Danielides S, Szklo M, et al. Insulin resistance in rheumatoid arthritis: disease-related indicators and associations with the presence and progression of subclinical atherosclerosis. Arthritis Rheumatol. 2015;67(3):626–36.PubMedPubMedCentralCrossRef Giles JT, Danielides S, Szklo M, et al. Insulin resistance in rheumatoid arthritis: disease-related indicators and associations with the presence and progression of subclinical atherosclerosis. Arthritis Rheumatol. 2015;67(3):626–36.PubMedPubMedCentralCrossRef
6.
go back to reference Urman A, Taklalsingh N, Sorrento C, et al. Inflammation beyond the joints: rheumatoid arthritis and cardiovascular disease. Scifed J Cardiol. 2018;2(3):1000019.PubMedPubMedCentral Urman A, Taklalsingh N, Sorrento C, et al. Inflammation beyond the joints: rheumatoid arthritis and cardiovascular disease. Scifed J Cardiol. 2018;2(3):1000019.PubMedPubMedCentral
7.
go back to reference Choy E. Understanding the dynamics: pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford). 2012;51(Suppl 5):v3–v11.CrossRef Choy E. Understanding the dynamics: pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford). 2012;51(Suppl 5):v3–v11.CrossRef
8.
go back to reference Park YJ, Yoo SA, Kim GR, et al. Urinary interleukin-6 as a predictor of radiographic progression in rheumatoid arthritis: a 3-year evaluation. Sci Rep. 2016;6:35242.PubMedPubMedCentralCrossRef Park YJ, Yoo SA, Kim GR, et al. Urinary interleukin-6 as a predictor of radiographic progression in rheumatoid arthritis: a 3-year evaluation. Sci Rep. 2016;6:35242.PubMedPubMedCentralCrossRef
10.
go back to reference Dayer JM, Choy E. Therapeutic targets in rheumatoid arthritis: the interleukin-6 receptor. Rheumatology (Oxford). 2010;49(1):15–24.CrossRef Dayer JM, Choy E. Therapeutic targets in rheumatoid arthritis: the interleukin-6 receptor. Rheumatology (Oxford). 2010;49(1):15–24.CrossRef
11.
go back to reference Glund S, Krook A. Role of interleukin-6 signalling in glucose and lipid metabolism. Acta Physiol. 2008;192(1):37–48.CrossRef Glund S, Krook A. Role of interleukin-6 signalling in glucose and lipid metabolism. Acta Physiol. 2008;192(1):37–48.CrossRef
12.
go back to reference Akbari M, Hassan-Zadeh V. IL-6 signalling pathways and the development of type 2 diabetes. Inflammopharmacology. 2018;26(3):685–98.PubMedCrossRef Akbari M, Hassan-Zadeh V. IL-6 signalling pathways and the development of type 2 diabetes. Inflammopharmacology. 2018;26(3):685–98.PubMedCrossRef
13.
go back to reference Kristiansen OP, Mandrup-Poulsen T. Interleukin-6 and diabetes: the good, the bad, or the indifferent? Diabetes. 2005;54(Suppl 2):S114–24.PubMedCrossRef Kristiansen OP, Mandrup-Poulsen T. Interleukin-6 and diabetes: the good, the bad, or the indifferent? Diabetes. 2005;54(Suppl 2):S114–24.PubMedCrossRef
14.
go back to reference Pradhan AD, Manson JE, Rifai N, et al. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA. 2001;286(3):327–34.PubMedCrossRef Pradhan AD, Manson JE, Rifai N, et al. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA. 2001;286(3):327–34.PubMedCrossRef
15.
go back to reference Spranger J, Kroke A, Mohlig M, et al. Inflammatory cytokines and the risk to develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes. 2003;52(3):812–7.CrossRefPubMed Spranger J, Kroke A, Mohlig M, et al. Inflammatory cytokines and the risk to develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes. 2003;52(3):812–7.CrossRefPubMed
16.
go back to reference Southern C, Schulster D, Green IC. Inhibition of insulin secretion from rat islets of Langerhans by interleukin-6. An effect distinct from that of interleukin-1. Biochem J. 1990;272(1):243–5.PubMedPubMedCentralCrossRef Southern C, Schulster D, Green IC. Inhibition of insulin secretion from rat islets of Langerhans by interleukin-6. An effect distinct from that of interleukin-1. Biochem J. 1990;272(1):243–5.PubMedPubMedCentralCrossRef
17.
go back to reference Sandler S, Bendtzen K, Eizirik DL, et al. Interleukin-6 affects insulin secretion and glucose metabolism of rat pancreatic islets in vitro. Endocrinology. 1990;126(2):1288–94.PubMedCrossRef Sandler S, Bendtzen K, Eizirik DL, et al. Interleukin-6 affects insulin secretion and glucose metabolism of rat pancreatic islets in vitro. Endocrinology. 1990;126(2):1288–94.PubMedCrossRef
18.
go back to reference Ballak DB, Stienstra R, Tack CJ, et al. IL-1 family members in the pathogenesis and treatment of metabolic disease: focus on adipose tissue inflammation and insulin resistance. Cytokine. 2015;75(2):280–90.PubMedPubMedCentralCrossRef Ballak DB, Stienstra R, Tack CJ, et al. IL-1 family members in the pathogenesis and treatment of metabolic disease: focus on adipose tissue inflammation and insulin resistance. Cytokine. 2015;75(2):280–90.PubMedPubMedCentralCrossRef
19.
go back to reference Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.PubMedCrossRef Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.PubMedCrossRef
20.
go back to reference Larsen CM, Faulenbach M, Vaag A, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356(15):1517–26.PubMedCrossRef Larsen CM, Faulenbach M, Vaag A, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356(15):1517–26.PubMedCrossRef
21.
go back to reference Kataria Y, Ellervik C, Mandrup-Poulsen T. Treatment of type 2 diabetes by targeting interleukin-1: a meta-analysis of 2921 patients. Semin Immunopathol. 2019. Kataria Y, Ellervik C, Mandrup-Poulsen T. Treatment of type 2 diabetes by targeting interleukin-1: a meta-analysis of 2921 patients. Semin Immunopathol. 2019.
22.
go back to reference Elena C, Chiara M, Angelica B, et al. Hyperglycemia and diabetes induced by glucocorticoids in nondiabetic and diabetic patients: revision of literature and personal considerations. Curr Pharm Biotechnol. 2019;19(15):1210–1220. Elena C, Chiara M, Angelica B, et al. Hyperglycemia and diabetes induced by glucocorticoids in nondiabetic and diabetic patients: revision of literature and personal considerations. Curr Pharm Biotechnol. 2019;19(15):1210–1220.
23.
go back to reference Movahedi M, Beauchamp ME, Abrahamowicz M, et al. Risk of incident diabetes mellitus associated with the dosage and duration of oral glucocorticoid therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2016;68(5):1089–98.PubMedPubMedCentral Movahedi M, Beauchamp ME, Abrahamowicz M, et al. Risk of incident diabetes mellitus associated with the dosage and duration of oral glucocorticoid therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2016;68(5):1089–98.PubMedPubMedCentral
24.
go back to reference Ozen G, Pedro S, Holmqvist ME, et al. Risk of diabetes mellitus associated with disease-modifying antirheumatic drugs and statins in rheumatoid arthritis. Ann Rheum Dis. 2017;76(5):848–54.PubMedCrossRef Ozen G, Pedro S, Holmqvist ME, et al. Risk of diabetes mellitus associated with disease-modifying antirheumatic drugs and statins in rheumatoid arthritis. Ann Rheum Dis. 2017;76(5):848–54.PubMedCrossRef
25.
go back to reference Solomon DH, Massarotti E, Garg R, et al. Association between disease-modifying antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA. 2011;305(24):2525–31.PubMedCrossRef Solomon DH, Massarotti E, Garg R, et al. Association between disease-modifying antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA. 2011;305(24):2525–31.PubMedCrossRef
26.
go back to reference Rempenault C, Combe B, Barnetche T, et al. Metabolic and cardiovascular benefits of hydroxychloroquine in patients with rheumatoid arthritis: a systematic review and meta-analysis. Ann Rheum Dis. 2018;77(1):98–103.PubMedCrossRef Rempenault C, Combe B, Barnetche T, et al. Metabolic and cardiovascular benefits of hydroxychloroquine in patients with rheumatoid arthritis: a systematic review and meta-analysis. Ann Rheum Dis. 2018;77(1):98–103.PubMedCrossRef
27.
go back to reference Solomon DH, Garg R, Lu B, et al. Effect of hydroxychloroquine on insulin sensitivity and lipid parameters in rheumatoid arthritis patients without diabetes mellitus: a randomized, blinded crossover trial. Arthritis Care Res. 2014;66(8):1246–51.CrossRef Solomon DH, Garg R, Lu B, et al. Effect of hydroxychloroquine on insulin sensitivity and lipid parameters in rheumatoid arthritis patients without diabetes mellitus: a randomized, blinded crossover trial. Arthritis Care Res. 2014;66(8):1246–51.CrossRef
28.
go back to reference Chen X, Wang N, Zhu Y, et al. The antimalarial chloroquine suppresses LPS-induced NLRP3 inflammasome activation and confers protection against murine endotoxic shock. Mediat Inflamm. 2017;2017:6543237. Chen X, Wang N, Zhu Y, et al. The antimalarial chloroquine suppresses LPS-induced NLRP3 inflammasome activation and confers protection against murine endotoxic shock. Mediat Inflamm. 2017;2017:6543237.
29.
go back to reference de Rotte MC, de Jong PH, den Boer E, et al. Effect of methotrexate use and erythrocyte methotrexate polyglutamate on glycosylated hemoglobin in rheumatoid arthritis. Arthritis Rheumatol. 2014;66(8):2026–36.PubMedCrossRef de Rotte MC, de Jong PH, den Boer E, et al. Effect of methotrexate use and erythrocyte methotrexate polyglutamate on glycosylated hemoglobin in rheumatoid arthritis. Arthritis Rheumatol. 2014;66(8):2026–36.PubMedCrossRef
30.
go back to reference Perdan-Pirkmajer K, Pirkmajer S, Thevis M, et al. Methotrexate reduces HbA1c concentration but does not produce chronic accumulation of ZMP in patients with rheumatoid or psoriatic arthritis. Scand J Rheumatol. 2016;45(5):347–55.PubMedCrossRef Perdan-Pirkmajer K, Pirkmajer S, Thevis M, et al. Methotrexate reduces HbA1c concentration but does not produce chronic accumulation of ZMP in patients with rheumatoid or psoriatic arthritis. Scand J Rheumatol. 2016;45(5):347–55.PubMedCrossRef
31.
go back to reference Burmester GR, Lin Y, Patel R, et al. Efficacy and safety of sarilumab monotherapy versus adalimumab monotherapy for the treatment of patients with active rheumatoid arthritis (MONARCH): a randomised, double-blind, parallel-group phase III trial. Ann Rheum Dis. 2017;76(5):840–7.PubMedCrossRef Burmester GR, Lin Y, Patel R, et al. Efficacy and safety of sarilumab monotherapy versus adalimumab monotherapy for the treatment of patients with active rheumatoid arthritis (MONARCH): a randomised, double-blind, parallel-group phase III trial. Ann Rheum Dis. 2017;76(5):840–7.PubMedCrossRef
32.
go back to reference Fleischmann R, van Adelsberg J, Lin Y, et al. Sarilumab and nonbiologic disease-modifying antirheumatic drugs in patients with active rheumatoid arthritis and inadequate response or intolerance to tumor necrosis factor inhibitors. Arthritis Rheumatol. 2017;69(2):277–90.PubMedPubMedCentralCrossRef Fleischmann R, van Adelsberg J, Lin Y, et al. Sarilumab and nonbiologic disease-modifying antirheumatic drugs in patients with active rheumatoid arthritis and inadequate response or intolerance to tumor necrosis factor inhibitors. Arthritis Rheumatol. 2017;69(2):277–90.PubMedPubMedCentralCrossRef
33.
go back to reference Genovese MC, Fleischmann R, Kivitz AJ, et al. Sarilumab plus methotrexate in patients with active rheumatoid arthritis and inadequate response to methotrexate: results of a phase III study. Arthritis Rheumatol. 2015;67(6):1424–37.PubMedCrossRef Genovese MC, Fleischmann R, Kivitz AJ, et al. Sarilumab plus methotrexate in patients with active rheumatoid arthritis and inadequate response to methotrexate: results of a phase III study. Arthritis Rheumatol. 2015;67(6):1424–37.PubMedCrossRef
34.
go back to reference Association AD. Glycemic targets: standards of medical care in diabetes—2018. Diabetes Care. 2018;41(Suppl 1):S55–64.CrossRef Association AD. Glycemic targets: standards of medical care in diabetes—2018. Diabetes Care. 2018;41(Suppl 1):S55–64.CrossRef
35.
go back to reference Simard JF, Mittleman MA. Prevalent rheumatoid arthritis and diabetes among NHANES III participants aged 60 and older. J Rheumatol. 2007;34(3):469–73.PubMed Simard JF, Mittleman MA. Prevalent rheumatoid arthritis and diabetes among NHANES III participants aged 60 and older. J Rheumatol. 2007;34(3):469–73.PubMed
36.
go back to reference Lillegraven S, Greenberg JD, Reed GW, et al. Immunosuppressive treatment and the risk of diabetes in rheumatoid arthritis. PLoS One. 2019;14(1):e0210459.PubMedPubMedCentralCrossRef Lillegraven S, Greenberg JD, Reed GW, et al. Immunosuppressive treatment and the risk of diabetes in rheumatoid arthritis. PLoS One. 2019;14(1):e0210459.PubMedPubMedCentralCrossRef
37.
go back to reference Frydrych LM, Bian G, O'Lone DE, et al. Obesity and type 2 diabetes mellitus drive immune dysfunction, infection development, and sepsis mortality. J Leukoc Biol. 2018;104(3):525–34.PubMedCrossRef Frydrych LM, Bian G, O'Lone DE, et al. Obesity and type 2 diabetes mellitus drive immune dysfunction, infection development, and sepsis mortality. J Leukoc Biol. 2018;104(3):525–34.PubMedCrossRef
38.
go back to reference International Hypoglycaemia Study Group. Hypoglycaemia, cardiovascular disease, and mortality in diabetes: epidemiology, pathogenesis, and management. Lancet Diabetes Endocrinol. 2019;7(5):358–96. International Hypoglycaemia Study Group. Hypoglycaemia, cardiovascular disease, and mortality in diabetes: epidemiology, pathogenesis, and management. Lancet Diabetes Endocrinol. 2019;7(5):358–96.
39.
go back to reference Nurmohamed MT, Kitas G. Cardiovascular risk in rheumatoid arthritis and diabetes: how does it compare and when does it start? Ann Rheum Dis. 2011;70(6):881–3.PubMedCrossRef Nurmohamed MT, Kitas G. Cardiovascular risk in rheumatoid arthritis and diabetes: how does it compare and when does it start? Ann Rheum Dis. 2011;70(6):881–3.PubMedCrossRef
40.
go back to reference Ogata A, Morishima A, Hirano T, et al. Improvement of HbA1c during treatment with humanised anti-interleukin 6 receptor antibody, tocilizumab. Ann Rheum Dis. 2011;70(6):1164–5.PubMedCrossRef Ogata A, Morishima A, Hirano T, et al. Improvement of HbA1c during treatment with humanised anti-interleukin 6 receptor antibody, tocilizumab. Ann Rheum Dis. 2011;70(6):1164–5.PubMedCrossRef
41.
go back to reference Otsuka Y, Kiyohara C, Kashiwado Y, et al. Effects of tumor necrosis factor inhibitors and tocilizumab on the glycosylated hemoglobin levels in patients with rheumatoid arthritis; an observational study. PLoS One. 2018;13(4):e0196368.PubMedPubMedCentralCrossRef Otsuka Y, Kiyohara C, Kashiwado Y, et al. Effects of tumor necrosis factor inhibitors and tocilizumab on the glycosylated hemoglobin levels in patients with rheumatoid arthritis; an observational study. PLoS One. 2018;13(4):e0196368.PubMedPubMedCentralCrossRef
42.
go back to reference Specker C, Alberding A, Aringer M, et al. Improvement of HbA1c in patients with rheumatoid arthritis and diabetes type 2 during treatment with tocilizumab. 2018. Specker C, Alberding A, Aringer M, et al. Improvement of HbA1c in patients with rheumatoid arthritis and diabetes type 2 during treatment with tocilizumab. 2018.
43.
go back to reference Castaneda S, Remuzgo-Martinez S, Lopez-Mejias R, et al. Rapid beneficial effect of the IL-6 receptor blockade on insulin resistance and insulin sensitivity in non-diabetic patients with rheumatoid arthritis. Clin Exp Rheumatol. 2019;37(3):465473. Castaneda S, Remuzgo-Martinez S, Lopez-Mejias R, et al. Rapid beneficial effect of the IL-6 receptor blockade on insulin resistance and insulin sensitivity in non-diabetic patients with rheumatoid arthritis. Clin Exp Rheumatol. 2019;37(3):465473.
44.
go back to reference Tsigos C, Papanicolaou DA, Kyrou I, et al. Dose-dependent effects of recombinant human interleukin-6 on glucose regulation. J Clin Endocrinol Metab. 1997;82(12):4167–70.PubMedCrossRef Tsigos C, Papanicolaou DA, Kyrou I, et al. Dose-dependent effects of recombinant human interleukin-6 on glucose regulation. J Clin Endocrinol Metab. 1997;82(12):4167–70.PubMedCrossRef
45.
go back to reference Lang Lehrskov L, Lyngbaek MP, Soederlund L, et al. Interleukin-6 delays gastric emptying in humans with direct effects on glycemic control. Cell Metab. 2018;27(6):1201–11 e1203.PubMedCrossRef Lang Lehrskov L, Lyngbaek MP, Soederlund L, et al. Interleukin-6 delays gastric emptying in humans with direct effects on glycemic control. Cell Metab. 2018;27(6):1201–11 e1203.PubMedCrossRef
46.
go back to reference Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, et al. Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol. 2006;24(1):83–6.PubMed Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, et al. Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol. 2006;24(1):83–6.PubMed
47.
go back to reference Stagakis I, Bertsias G, Karvounaris S, et al. Anti-tumor necrosis factor therapy improves insulin resistance, beta cell function and insulin signaling in active rheumatoid arthritis patients with high insulin resistance. Arthritis Res Ther. 2012;14(3):R141.PubMedPubMedCentralCrossRef Stagakis I, Bertsias G, Karvounaris S, et al. Anti-tumor necrosis factor therapy improves insulin resistance, beta cell function and insulin signaling in active rheumatoid arthritis patients with high insulin resistance. Arthritis Res Ther. 2012;14(3):R141.PubMedPubMedCentralCrossRef
48.
go back to reference Tam LS, Tomlinson B, Chu TT, et al. Impact of TNF inhibition on insulin resistance and lipids levels in patients with rheumatoid arthritis. Clin Rheumatol. 2007;26(9):1495–8.PubMedCrossRef Tam LS, Tomlinson B, Chu TT, et al. Impact of TNF inhibition on insulin resistance and lipids levels in patients with rheumatoid arthritis. Clin Rheumatol. 2007;26(9):1495–8.PubMedCrossRef
49.
go back to reference Bissell LA, Hensor EM, Kozera L, et al. Improvement in insulin resistance is greater when infliximab is added to methotrexate during intensive treatment of early rheumatoid arthritis-results from the IDEA study. Rheumatology (Oxford). 2016;55(12):2181–90.CrossRef Bissell LA, Hensor EM, Kozera L, et al. Improvement in insulin resistance is greater when infliximab is added to methotrexate during intensive treatment of early rheumatoid arthritis-results from the IDEA study. Rheumatology (Oxford). 2016;55(12):2181–90.CrossRef
50.
go back to reference Ferraz-Amaro I, Arce-Franco M, Muniz J, et al. Systemic blockade of TNF-alpha does not improve insulin resistance in humans. Horm Metab Res. 2011;43(11):801–8.PubMedCrossRef Ferraz-Amaro I, Arce-Franco M, Muniz J, et al. Systemic blockade of TNF-alpha does not improve insulin resistance in humans. Horm Metab Res. 2011;43(11):801–8.PubMedCrossRef
51.
go back to reference Rosenvinge A, Krogh-Madsen R, Baslund B, et al. Insulin resistance in patients with rheumatoid arthritis: effect of anti-TNF alpha therapy. Scand J Rheumatol. 2007;36(2):91–6.PubMedCrossRef Rosenvinge A, Krogh-Madsen R, Baslund B, et al. Insulin resistance in patients with rheumatoid arthritis: effect of anti-TNF alpha therapy. Scand J Rheumatol. 2007;36(2):91–6.PubMedCrossRef
52.
go back to reference Stavropoulos-Kalinoglou A, Metsios GS, Panoulas VF, et al. Anti-tumour necrosis factor alpha therapy improves insulin sensitivity in normal-weight but not in obese patients with rheumatoid arthritis. Arthritis Res Ther. 2012;14(4):R160.PubMedPubMedCentralCrossRef Stavropoulos-Kalinoglou A, Metsios GS, Panoulas VF, et al. Anti-tumour necrosis factor alpha therapy improves insulin sensitivity in normal-weight but not in obese patients with rheumatoid arthritis. Arthritis Res Ther. 2012;14(4):R160.PubMedPubMedCentralCrossRef
53.
go back to reference Bach E, Nielsen RR, Vendelbo MH, et al. Direct effects of TNF-α on local fuel metabolism and cytokine levels in the placebo-controlled, bilaterally infused human leg: increased insulin sensitivity, increased net protein breakdown, and increased IL-6 release. Diabetes. 2013;62(12):4023–9.PubMedPubMedCentralCrossRef Bach E, Nielsen RR, Vendelbo MH, et al. Direct effects of TNF-α on local fuel metabolism and cytokine levels in the placebo-controlled, bilaterally infused human leg: increased insulin sensitivity, increased net protein breakdown, and increased IL-6 release. Diabetes. 2013;62(12):4023–9.PubMedPubMedCentralCrossRef
54.
go back to reference Krogh-Madsen R, Plomgaard P, Moller K, et al. Influence of TNF-alpha and IL-6 infusions on insulin sensitivity and expression of IL-18 in humans. Am J Physiol Endocrinol Metab. 2006;291(1):E108–14.PubMedCrossRef Krogh-Madsen R, Plomgaard P, Moller K, et al. Influence of TNF-alpha and IL-6 infusions on insulin sensitivity and expression of IL-18 in humans. Am J Physiol Endocrinol Metab. 2006;291(1):E108–14.PubMedCrossRef
55.
go back to reference Boulton JG, Bourne JT. Unstable diabetes in a patient receiving anti-TNF-alpha for rheumatoid arthritis. Rheumatology (Oxford). 2007;46:178–9.CrossRef Boulton JG, Bourne JT. Unstable diabetes in a patient receiving anti-TNF-alpha for rheumatoid arthritis. Rheumatology (Oxford). 2007;46:178–9.CrossRef
56.
go back to reference Bonilla E, Lee YY, Phillips PE, et al. Hypoglycaemia after initiation of treatment with etanercept in a patient with type 2 diabetes mellitus. Ann Rheum Dis. 2007;66:1688.PubMedPubMedCentralCrossRef Bonilla E, Lee YY, Phillips PE, et al. Hypoglycaemia after initiation of treatment with etanercept in a patient with type 2 diabetes mellitus. Ann Rheum Dis. 2007;66:1688.PubMedPubMedCentralCrossRef
57.
go back to reference Wambier CG, Foss-Freitas MC, Paschoal RS, et al. Severe hypoglycemia after initiation of anti-tumor necrosis factor therapy with etanercept in a patient with generalized pustular psoriasis and type 2 diabetes mellitus. J Am Acad Dermatol. 2009;60:883–5.PubMedCrossRef Wambier CG, Foss-Freitas MC, Paschoal RS, et al. Severe hypoglycemia after initiation of anti-tumor necrosis factor therapy with etanercept in a patient with generalized pustular psoriasis and type 2 diabetes mellitus. J Am Acad Dermatol. 2009;60:883–5.PubMedCrossRef
58.
go back to reference Cheung D, Bryer-Ash M. Persistent hypoglycemia in a patient with diabetes taking etanercept for the treatment of psoriasis. J Am Acad Dermatol. 2009;60(6):1032–6.PubMedCrossRef Cheung D, Bryer-Ash M. Persistent hypoglycemia in a patient with diabetes taking etanercept for the treatment of psoriasis. J Am Acad Dermatol. 2009;60(6):1032–6.PubMedCrossRef
Metadata
Title
Interleukin-6 receptor blockade or TNFα inhibition for reducing glycaemia in patients with RA and diabetes: post hoc analyses of three randomised, controlled trials
Authors
Mark C. Genovese
Gerd R. Burmester
Owen Hagino
Karthinathan Thangavelu
Melitza Iglesias-Rodriguez
Gregory St John
Miguel A. González-Gay
Thomas Mandrup-Poulsen
Roy Fleischmann
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Arthritis Research & Therapy / Issue 1/2020
Electronic ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-020-02229-5

Other articles of this Issue 1/2020

Arthritis Research & Therapy 1/2020 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.