Indian Journal of Inflammation Research

"Understanding Inflammation, Enhancing Health"

Welcome to the Indian Journal of Inflammation Research (IJIR), where knowledge fuels healing. Our platform provides a vital hub for researchers, clinicians, and policymakers to explore the multifaceted aspects of inflammation and its impact on human health. Delve into our curated selection of articles, studies, and reviews aimed at deepening our understanding of inflammation and developing innovative strategies for prevention and treatment.

Submit Article

Mechanism-based pharmacodynamic interactions of glucocorticoids and disease-modifying antirheumatic drugs: A review

Authors

  • Chandrashekara S Director, ChanRe Rheumatology and Immunology Center, Bangalore, India

Keywords:

Pharmacodynamics, RA, DMARDs, glucocorticoids, NF-κB, T cells

Abstract

Combination of conventional disease-modifying antirheumatic drugs (cDMARDs), which assist in achieving effective disease control and functional outcomes in rheumatoid arthritis (RA), is widely accepted as an effective treatment regimen. Steroid is often used in combination with DMARDs and there are several uncertainties associated with the regimen, including the therapeutic benefits of the combination. Further understanding the pharmacodynamics and mechanism of actions of steroids, and possible drug interactions may assist in improving the treatment outcomes. While the treatment has shown to be effective with better outcome, the use of steroids in the combination regime has been a topic of contradiction. 

 

The pharmacodynamic actions of corticosteroids and cDMARDs is variable and overlapping in some domains. In a highly dynamic and constantly adopting immune system, the expression of receptors and signal pathways depends on various factors including the status of cytokine and antigen load. The pattern of introduction of the drugs is one of the major challenges that hampers the effective use of synergism and other interactions associated with the combination. Understanding the drug mechanism and interaction pathways and their variability is essential for customizing the treatment strategies. This review article addresses the current trends of combined treatment in the management of RA. It is also aimed at providing consolidated information on the pharmacodynamic interactions of glucocorticoids and DMARDs in RA.

 

References

Mikuls TR, Fay BT, Michaud K, et al. Associations of disease activity and treatments with mortality in men with rheumatoid arthritis: results from the VARA registry. Rheumatology (Oxford). 2011;50(1):101–9.

Caplan L, Wolfe F, Russell AS, et al. Corticosteroid use in rheumatoid arthritis: prevalence, predictors, correlates, and outcomes. J Rheumatol. 2007;34(4):696–705.

del Rincón I, Battafarano DF, Restrepo JF, et al. Glucocorticoid dose thresholds associated with all-cause and cardiovascular mortality in rheumatoid arthritis. Arthritis Rheumatol. 2014;66(2):264–72.

Wasko MC, Dasgupta A, Ilse Sears G, et al. Prednisone use and risk of mortality in patients with rheumatoid arthritis: moderation by use of disease-modifying antirheumatic drugs. Arthritis Care Res (Hoboken). 2016;68(5):706–10.

Chandrashekara S. Pharmacokinetic consideration of synthetic DMARDs in rheumatoid arthritis. Expert Opin Drug Metab Toxicol. 2013;9(8):969–81.

Honoré PM, Jacobs R, De Waele E, et al. What do we know about steroids metabolism and “PK/PD approach” in AKI and CKD especially while on RRT--current status in 2014. Blood Purif. 2014;38(2):154–7.

Wessels J a. M, Huizinga TWJ, Guchelaar H-J. Recent insights in the pharmacological actions of methotrexate in the treatment of rheumatoid arthritis. Rheumatology (Oxford). 2008;47(3):249–55.

Chikanza IC. Mechanisms of Corticosteroid Resistance in Rheumatoid Arthritis. Ann N Y Acad Sci. 966(1):39–48.

Majumdar S, Aggarwal BB. Methotrexate Suppresses NF-κB Activation Through Inhibition of IκBα Phosphorylation and Degradation. J Immunol. 2001;167(5):2911–20.

Hedman LA, Röckert L-L, Lundin PM. The effect of steroids on the circulating lymphocyte population — VI. Studies of the thoracic duct T- and B-lymphocyte populations after neonatal thymectomy and prednisolone treatment. An immunofluorescence. In J Immunopharmacol. 1984;6(4):357–63.

Galon J, Franchimont D, Hiroi N, et al. Gene profiling reveals unknown enhancing and suppressive actions of glucocorticoids on immune cells. FASEB J. 2002;16(1):61–71.

Soyka LF. Treatment of the Nephrotic Syndrome in Childhood: Use of an Alternate-Day Prednisone Regimen. Am J Dis Child. 1967;113(6):693–701.

Portner MM, Thayer KH, Harter JG, et al. Successful initiation of alternate-day prednisone in chronic steroid dependent asthmatic patients. J Allergy Clin Immunol. 1972;49(1):16-26.

Reed W, Lucas ZJ, Cohn R. Alternate-day prednisone therapy after renal transplantation. Lancet. 1970;295(7650):747–9.

McEnery PT, Gonzalez LL, Martin LW, et al. Growth and development of children with renal transplants. Use of alternate-day steroid therapy. J Pediatr 1973;83(5):806–14.

Cocco AE, Mendeloff Al. An evaluation of intermittent corticosteroid therapy in the management of ulcerative colitis. Johns Hopkins Med J. 1967;120(3):162-9.

Hess EV, Goldman JA: Corticosteroids and corticotropin in therapy of rheumatoid arthritis. In: Hollander JL, Mccarty DJ: Arthritis and Allied conditions. 8th ed. Philadelphia, Lea & Febiger, 1972.

Harter JG, Reddy WJ, Thorn GW. Studies on an intermittent corticosteroid dosage regimen. N Engl J Med. 1963;269(12):591-6.

MacGregor RR, Sheagren JN, Lipsett MB, et al. Alternate-day prednisone therapy. Evaluation of delayed hypersensitivity responses, control of disease and steroid side effects. N Engl J Med. 1969;280(26):1427–31.

Cupps TR, Fauci AS. Corticosteroid-mediated immunoregulation in man. Immunol Rev. 1982;65:133–55.

Nara PL, Krakowka S, Poweres TE. Effects of prednisolone on the development of immune responses to canine distemper virus in beagle pups. Am J Vet Res. 1979;40(12):1742-7.

Thorne GW, Forsham PH, Prunty FTG, et al. A test for adrenal corticle insufficiency. JAMA. 1948;137:1005-9.

Cohn LA. The influence of corticosteroids on host defense mechanisms. J Vet Intern Med. 1991;5(2):95–104.

Fauci AS, Dale DC. The effect of in vivo hydrocortisone on subpopulations of human lymphocytes. J Clin Invest. 1974;53(1):240–6.

Butler WT, Rossen RD. Effects of corticosteroids on immunity in man. I. Decreased serum IgG concentration caused by 3 or 5 days of high doses of methylprednisolone. J Clin Invest. 1973;52(10):2629–40.

Haynes BF, Fauci AS. The Differential Effect of In Vivo Hydrocortisone on the Kinetics of Subpopulations of Human Peripheral Blood Thymus-Derived Lymphocytes. J Clin Invest. 1978;61(3):703–7.

Rinehart JJ, Sagone AL, Balcerzak SP, et al. Effects of Corticosteroid Therapy on Human Monocyte Function. N Engl J Med. 1975;292(5):236–41.

Breedveld FC, Dayer JM. Leflunomide: mode of action in the treatment of rheumatoid arthritis. Ann Rheum Dis. 2000;59(11):841–9.

Vuolteenaho K, Kujala P, Moilanen T, et al. Aurothiomalate and hydroxychloroquine inhibit nitric oxide production in chondrocytes and in human osteoarthritic cartilage. Scand J Rheumatol. 2005;34(6):475–9.

Ray A, Prefontaine KE. Physical association and functional antagonism between the p65 subunit of transcription factor NF-kappa B and the glucocorticoid receptor. Proc Natl Acad Sci U S A. 1994;91(2):752–6.

Strauss G, Osen W, Debatin KM. Induction of apoptosis and modulation of activation and effector function in T cells by immunosuppressive drugs. Clin Exp Immunol. 2002;128(2):255–66.

Gerards AH, de Lathouder S, de Groot ER, et al. Inhibition of cytokine production by methotrexate. Studies in healthy volunteers and patients with rheumatoid arthritis. Rheumatology (Oxford). 2003;42(10):1189–96.

Herman S, Zurgil N, Langevitz P, et al. The immunosuppressive effect of methotrexate in active rheumatoid arthritis patients vs. its stimulatory effect in nonactive patients, as indicated by cytometric measurements of CD4+ T cell subpopulations. Immunol Invest. 2004;33(3):351–62.

Johnston A, Gudjonsson JE, Sigmundsdottir H, et al. The anti-inflammatory action of methotrexate is not mediated by lymphocyte apoptosis, but by the suppression of activation and adhesion molecules. Clin Immunol. 2005;114(2):154–63.

Wijngaarden S, van Roon JA, van de Winkel JG, et al. Down-regulation of activating Fcgamma receptors on monocytes of patients with rheumatoid arthritis upon methotrexate treatment. Rheumatology (Oxford). 2005;44(6):729–34.

Cherwinski HM, McCarley D, Schatzman R, et al. The immunosuppressant leflunomide inhibits lymphocyte progression through cell cycle by a novel mechanism. J Pharmacol Exp Ther. 1995;272(1):460–8.

Bartlett RR, Anagnostopulos H, Zielinski T, et al. Effects of leflunomide on immune responses and models of inflammation. Springer Semin Immunopathol. 1993;14(4):381–394.

Cherwinski HM, Cohn RG, Cheung P, et al. The immunosuppressant leflunomide inhibits lymphocyte proliferation by inhibiting pyrimidine biosynthesis. J Pharmacol Exp Ther. 1995;275(2):1043–9.

Cherwinski HM, Byars N, Ballaron SJ, et al. Leflunomide interferes with pyrimidine nucleotide biosynthesis. Inflamm Res. 1995;44(8):317–322.

Xu X, Williams JW, Bremer EG, et al. Inhibition of protein tyrosine phosphorylation in T cells by a novel immunosuppressive agent, leflunomide. J Biol Chem. 1995;270(21):12398–403.

Elder RT, Xu X, Williams JW, et al. The immunosuppressive metabolite of leflunomide, A77 1726, affects murine T cells through two biochemical mechanisms. J Immunol. 1997;159(1):22–7.

Siemasko KF, Chong AS, Williams JW, et al. Regulation of B cell function by the immunosuppressive agent leflunomide. Transplantation. 1996;61(4):635–42.

Deage V, Burger D, Dayer J-M. Exposure of T lymphocytes to leflunomide but not to dexamethasone favors the production by monocytic cells of interleukin-1 receptor antagonist and the tissue-inhibitor of metalloproteinases-1 over that of interleukin-1 beta and metalloproteinases. Eur Cytokine Netw. 1998;9(4):663–8.

Goldman FD, Gilman AL, Hollenback C, et al. Hydroxychloroquine inhibits calcium signals in T cells: a new mechanism to explain its immunomodulatory properties. Blood. 2000;95(11):3460–6.

Meng XW, Feller JM, Ziegler JB, et al. Induction of apoptosis in peripheral blood lymphocytes following treatment in vitro with hydroxychloroquine. Arthritis Rheum. 1997;40(5):927–35.

Fox RI. Mechanism of action of hydroxychloroquine as an antirheumatic drug. Semin Arthritis Rheum. 1993;23(2, Supplement 1):82–91.

Kuznik A, Bencina M, Svajger U, et al. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J Immunol. 2011;186(8):4794–804.

Liberman AC, Druker J, Refojo D, et al. Glucocorticoids inhibit GATA-3 phosphorylation and activity in T cells. FASEB J. 2009;23(5):1558–71.

Franchimont D. Overview of the actions of glucocorticoids on the immune response: a good model to characterize new pathways of immunosuppression for new treatment strategies. Ann N Y Acad Sci. 2004;1024(1):124–37.

Hermoso MA, Matsuguchi T, Smoak K, et al. Glucocorticoids and tumor necrosis factor alpha cooperatively regulate Toll-like receptor 2 gene expression. Mol Cell Biol. 2004;24(11):4743–756.

van Ede AE, Laan RF, De Abreu RA, et al. Purine enzymes in patients with rheumatoid arthritis treated with methotrexate. Ann Rheum Dis. 2002;61(12):1060–4.

Barczyk K, Ehrchen J, Tenbrock K, et al. Glucocorticoids promote survival of anti-inflammatory macrophages via stimulation of adenosine receptor A3. Blood. 2010;116(3):446–55.

de Lathouder S, Gerards AH, Dijkmans BA et al. Two inhibitors of DNA synthesis lead to inhibition of cytokine production via a different mechanism. Nucleosides Nucleotides Nucleic Acids. 2004;23(8-9):1089–100.

Kraan M, Smeets T, van Loon MJ, et al. Differential effects of leflunomide and methotrexate on cytokine production in rheumatoid arthritis. Ann Rheum Dis. 2004;63(9):1056–61.

de Lathouder S, Gerards AH, de Groot ER, et al. Mycophenolic acid and methotrexate inhibit lymphocyte cytokine production via different mechanisms. Eur Cytokine Netw. 2002;13(3):317–23.

Miranda-Carus ME, Balsa A, Benito-Miguel M, et al. IL-15 and the initiation of cell contact-dependent synovial fibroblast-T lymphocyte cross-talk in rheumatoid arthritis: effect of methotrexate. J Immunol. 2004;173(2):1463–76.

Cao WW, Kao PN, Aoki Y, et al. A novel mechanism of action of the immunomodulatory drug, leflunomide: augmentation of the immunosuppressive cytokine, TGF-beta 1, and suppression of the immunostimulatory cytokine, IL-2. Transplant Proc. 1996;28(6):3079–80.

van den Borne BE, Dijkmans BA, de Rooij HH, et al. Chloroquine and hydroxychloroquine equally affect tumor necrosis factor-alpha, interleukin 6, and interferon-gamma production by peripheral blood mononuclear cells. J Rheumatol. 1997;24(1):55–60.

Jeong JY, Choi JW, Jeon KI, et al. Chloroquine decreases cell-surface expression of tumour necrosis factor receptors in human histiocytic U-937 cells. Immunology. 2002;105(1):83–91.

da Silva JC, Mariz HA, da Rocha Júnior LF, et al. Hydroxychloroquine decreases Th17-related cytokines in systemic lupus erythematosus and rheumatoid arthritis patients. Clinics (Sao Paulo). 2013;68(6):766–71.

Guo L, Lichten LA, Ryu MS, et al. STAT5-glucocorticoid receptor interaction and MTF-1 regulate the expression of ZnT2 (Slc30a2) in pancreatic acinar cells. Proc Natl Acad Sci U S A. 2010;107(7):2818–23.

Foster M, Samman S. Zinc and regulation of inflammatory cytokines: implications for cardiometabolic disease. Nutrients. 2012;4(7):676–94.

Liu MJ, Bao S, Galvez-Peralta M, et al. ZIP8 regulates host defense through zinc-mediated inhibition of NF-κB. Cell Rep. 2013;3(2):386–400.

Chen R, Burke TF, Cumberland JE, et al. Glucocorticoids inhibit calcium- and calcineurin-dependent activation of the human IL-4 promoter. J Immunol. 2000;164(2):825–832.

Tian H, Cronstein BN. Understanding the mechanisms of action of methotrexate: implications for the treatment of rheumatoid arthritis. Bull NYU Hosp Jt Dis 2007;65(3):168–73.

Davis JP, Cain GA, Pitts WJ, Magolda RL, Copeland RA. The immunosuppressive metabolite of leflunomide is a potent inhibitor of human dihydroorotate dehydrogenase. Biochemistry. 1996;35(4):1270–3.

Fox RI. Mechanism of action of leflunomide in rheumatoid arthritis. J Rheumatol Suppl. 1998;53:20–6.

Xu X, Blinder L, Shen J, Gong H, Finnegan A, Williams JW, et al. In vivo mechanism by which leflunomide controls lymphoproliferative and autoimmune disease in MRL/MpJ-lpr/lpr mice. J Immunol. 1997;159(1):167–74.

Furst DE, Lindsley H, Baethge B, Botstein GR, Caldwell J, Dietz F, et al. Dose-loading with hydroxychloroquine improves the rate of response in early, active rheumatoid arthritis: a randomized, double-blind six-week trial with eighteen-week extension. Arthritis Rheum. 1999;42(2):357–65.

Leis H, Page A, Ramirez A, et al. Glucocorticoid receptor counteracts tumorigenic activity of Akt in skin through interference with the phosphatidylinositol 3-kinase signaling pathway. Mol Endocrinol. 2004;18(2):303–11.

Haske T, Nakao M, Moudgil VK. Phosphorylation of immunopurified rat liver glucocorticoid receptor by the catalytic subunit of cAMP-dependent protein kinase. Mol Cell Biochem. 1994;132(2):163–71.

Doucas V, Shi Y, Miyamoto S, et al. Cytoplasmic catalytic subunit of protein kinase A mediates cross-repression by NF-κB and the glucocorticoid receptor. Proc Natl Acad Sci U S A. 2000;97(22):11893–8.

Petta I, Dejager L, Ballegeer M. The Interactome of the Glucocorticoid Receptor and Its Influence on the Actions of Glucocorticoids in Combatting Inflammatory and Infectious Diseases. Microbiol Mol Biol Rev. 2016;80(2):495–522.

Herdegen T, Leah JD. Inducible and constitutive transcription factors in the mammalian nervous system: control of gene expression by Jun, Fos and Krox, and CREB/ATF proteins. Brain Res Brain Res Rev. 1998;28(3):370–490.

Bruna A, Nicolas M, Munoz A, et al. Glucocorticoid receptor-JNK interaction mediates inhibition of the JNK pathway by glucocorticoids. EMBO J. 2003;22(22):6035–44.

Beck IM, Berghe WV, Vermeulen L, et al. Altered subcellular distribution of MSK1 induced by glucocorticoids contributes to NF-κB inhibition. EMBO J. 2008;27(12):1682–93.

Vandevyver S, Dejager L, Tuckermann J, et al. New insights into the anti-inflammatory mechanisms of glucocorticoids: an emerging role for glucocorticoid-receptor-mediated transactivation. Endocrinology. 2013;154(3):993–1007.

Barnes PJ. Corticosteroid effects on cell signalling. Eur Respir J. 2006;27(2):413–26.

Li X, Wong J, Tsai SY, et al. Progesterone and glucocorticoid receptors recruit distinct coactivator complexes and promote distinct patterns of local chromatin modification. Mol Cell Biol. 2003;23(11):3763–73.

Vandevyver S, Dejager L, Van Bogaert T, et al. Glucocorticoid receptor dimerization induces MKP1 to protect against TNF-induced inflammation. J Clin Invest. 2012;122(6):2130–40.

Gutierrez-Mecinas M, Trollope AF, Collins A, et al. Long-lasting behavioral responses to stress involve a direct interaction of glucocorticoid receptors with ERK1/2-MSK1-Elk-1 signaling. Proc Natl Acad Sci U S A. 2011;108(33):13806–11.

Bougarne N, Paumelle R, Caron S, et al. PPARalpha blocks glucocorticoid receptor alpha-mediated transactivation but cooperates with the activated glucocorticoid receptor alpha for transrepression on NF-kappaB. Proc Natl Acad Sci U S A. 2009;106(18):7397–402.

Sheppard KA, Phelps KM, Williams AJ, et al. Nuclear integration of glucocorticoid receptor and nuclear factor-kappaB signaling by CREB-binding protein and steroid receptor coactivator-1. J Biol Chem. 1998;273(45):29291–4.

Gupte R, Muse GW, Chinenov Y, et al. Glucocorticoid receptor represses proinflammatory genes at distinct steps of the transcription cycle. Proc Natl Acad Sci U S A. 2013;110(36):14616–21.

Lee CK, Lee EY, Chung SM, et al. Effects of disease-modifying antirheumatic drugs and antiinflammatory cytokines on human osteoclastogenesis through interaction with receptor activator of nuclear factor kappaB, osteoprotegerin, and receptor activator of nuclear factor kappaB ligand. Arthritis Rheum. 2004;50(12):3831–43.

Lowenberg M, Verhaar AP, van den Brink GR, et al. Glucocorticoid signaling: a nongenomic mechanism for T-cell immunosuppression. Trends Mol Med. 2007;13(4):158–63.

Reily MM, Pantoja C, Hu X, et al. The GRIP1:IRF3 interaction as a target for glucocorticoid receptor-mediated immunosuppression. EMBO J. 2006;25(1):108–117.

Ranganathan P, McLeod HL. Methotrexate pharmacogenetics: the first step toward individualized therapy in rheumatoid arthritis. Arthritis Rheum 2006;54(5):1366–77.

Kremer JM. Toward a better understanding of methotrexate. Arthritis Rheum. 2004;50(5):1370–82.

Herman S, Zurgil N, Deutsch M. Low dose methotrexate induces apoptosis with reactive oxygen species involvement in T lymphocytic cell lines to a greater extent than in monocytic lines. Inflamm Res. 2005;54(7):273–80.

Winter-Vann AM, Kamen BA, Bergo MO, et al. Targeting Ras signaling through inhibition of carboxyl methylation: an unexpected property of methotrexate. Proc Natl Acad Sci USA. 2003;100(11):6529–34.

Giacomelli R, Cipriani P, Cerinic MM, et al. Combination therapy with cyclosporine and methotrexate in patients with early rheumatoid arthritis soon inhibits TNFalpha production without decreasing TNF alpha mRNA levels. An in vivo and in vitro study. Clin Exp Rheumatol. 2002;20(3):365–72.

Sawah S Al, Zhang X, Zhu B, et al. Effect of corticosteroid use by dose on the risk of developing organ damage over time in systemic lupus erythematosus-the Hopkins Lupus Cohort. Lupus Sci Med. 2015;2(1):e000066.

Greene S, Watanabe K, Braatz-Trulson J, et al. Inhibition of dihydroorotate dehydrogenase by the immunosuppressive agent leflunomide. Biochem Pharmacol. 1995;50(6):861–7.

Herrmann M, Frangou CG, Kirschbaum B. Cell cycle control of the de novo pyrimidine synthesis inhibitor leflunomide through the p53 and p21WAF-1 pathways. Arthritis Rheum. 1997;40(9):S177-S177.

Xu X, Williams JW, Gong H, et al. Two activities of the immunosuppresive metabolite of leflunomide, A77 1726: inhibition of pyrimidine nucleotide synthesis and protein tyrosine phosphorylation. Biochem Pharmacol. 1996;52(4):527–34.

Poole B, Ohkuma S. Effect of weak bases on the intralysosomal pH in mouse peritoneal macrophages. J Cell Biol. 1981;90(3):665-9.

Sperber K, Quraishi H, Kalb TH, et al. Selective regulation of cytokine secretion by hydroxychloroquine: Inhibition of interleukin 1 alpha (IL-1-alpha) and IL-6 in human monocytes and T cells. J Rheumatol. 1993;20(5):803-8.

Picot S, Peyton F, Vuillez JP. Chloroquine inhibits tumor necrosis factor production by human macrophages in vitro. J Infect Dis. 1991;164(4):830.

Müller-Calleja N, Manukyan D, Canisius A, et al. Hydroxychloroquine inhibits proinflammatory signalling pathways by targeting endosomal NADPH oxidase. Ann Rheum Dis. 2017;76(5):891–7.

Hasko G, Cronstein BN. Adenosine: an endogenous regulator of innate immunity. Trends Immunol. 2004;25(1):33–9.

Montesinos MC, Takedachi M, Thompson LF, et al. The antiinflammatory mechanism of methotrexate depends on extracellular conversion of adenine nucleotides to adenosine by ecto-5’-nucleotidase: findings in a study of ecto-5’-nucleotidase gene-deficient mice. Arthritis Rheum. 2007;56(5):1440–5.

Ralph JA, McEvoy AN, Kane D, et al. Modulation of orphan nuclear receptor NURR1 expression by methotrexate in human inflammatory joint disease involves adenosine A2A receptor-mediated responses. J Immunol. 2005;175(1):555–65.

Kremer JM. Methotrexate and leflunomide: Biochemical basis for combination therapy in the treatment of rheumatoid arthritis. Semin Arthritis Rheum. 1999;29(1):14–26.

Weinblatt ME, Kremer JM, Coblyn JS, et al. Pharmacokinetics, safety, and efficacy of combination treatment with methotrexate and leflunomide in patients with active rheumatoid arthritis. Arthritis Rheum. 1999;42(7):1322–8.

Chou TC. Theoretical Basis, Experimental Design, and Computerized Simulation of Synergism and Antagonism in Drug Combination Studies. Pharmacol Rev. 2006;58 (3):621-81.

Downloads

Published

2018-09-27

Issue

Section

Articles