Rheumatoid arthritis: pathological mechanisms and modern pharmacologic therapies

Qiang Guo, Yuxiang Wang, Dan Xu, Johannes Nossent, Nathan J Pavlos, Jiake Xu, Qiang Guo, Yuxiang Wang, Dan Xu, Johannes Nossent, Nathan J Pavlos, Jiake Xu

Abstract

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease that primarily affects the lining of the synovial joints and is associated with progressive disability, premature death, and socioeconomic burdens. A better understanding of how the pathological mechanisms drive the deterioration of RA progress in individuals is urgently required in order to develop therapies that will effectively treat patients at each stage of the disease progress. Here we dissect the etiology and pathology at specific stages: (i) triggering, (ii) maturation, (iii) targeting, and (iv) fulminant stage, concomitant with hyperplastic synovium, cartilage damage, bone erosion, and systemic consequences. Modern pharmacologic therapies (including conventional, biological, and novel potential small molecule disease-modifying anti-rheumatic drugs) remain the mainstay of RA treatment and there has been significant progress toward achieving disease remission without joint deformity. Despite this, a significant proportion of RA patients do not effectively respond to the current therapies and thus new drugs are urgently required. This review discusses recent advances of our understanding of RA pathogenesis, disease modifying drugs, and provides perspectives on next generation therapeutics for RA.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1
RA can be triggered in the potential trigger sites (lung, oral, gut, et al.) by the interaction between the genes and environmental factors, which is characterized by the onset of self-protein citrullination resulting in the production of autoantibodies against citrullinated peptides. Lung exposure to noxious agents, infectious agents (Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, and Epstein-Barr virus), gut microbiome, and dietary factors may induce the self-protein citrullination and maturation of ACPA. Citrullination is catalyzed by the calcium-dependent enzyme PAD, changing a positively charged arginine to a polar but neutral citrulline as the result of a post-translational modification. In RA, PAD can be secreted by the granulocyte and macrophage. ACPA occurs as a result of an abnormal antibody response to a range of citrullinated proteins, including fibrin, vimentin, fibronectin, Epstein-Barr Nuclear Antigen 1, α-enolase, type II collagen, and histones, all of which are distributed throughout the whole body. Many citrullination neoantigens would activate MHC class II-dependent T cells that in turn would help B cells produce more ACPA. The stage is also called loss of tolerance. RA rheumatoid arthritis, PAD peptidyl-arginine-deiminase, ACPA anti-citrullinated  protein antibodies, RF rheumatoid factor.
Fig. 2
Fig. 2
Many cells and their cytokines play critical roles in the development of RA. The synovial compartment is infiltrated by leukocytes and the synovial fluid is inundated with pro-inflammatory mediators that are produced to induce an inflammatory cascade, which is characterized by interactions of fibroblast-like synoviocytes with the cells of the innate immune system, including monocytes, macrophages, mast cells, dendritic cells, and so on, as well as cells of adaptive immune system such as T cells and B cells. Endothelial cells contribute to the extensive angiogenesis. The fulminant stage contains hyperplastic synovium, cartilage damage, bone erosion, and systemic consequence. Bone resorption virtually creates bone erosions, which are usually found at spots where the synovial membrane inserts into the periosteum, which is known as a bare area according to certain anatomical features. The destruction of the subchondral bone can eventually result in the degeneration of the articular cartilage as the result of a decrease in osteoblasts and an increase in osteoclasts and synoviocytes. IL interleukin, TNF tumor necrosis factor, MMP matrix metalloproteinase, TGF transforming growth factor, PDGF platelet-derived growth factor, IFN interferon, GM-CSF granulocyte–macrophage colony-stimulating factor, VEGF vascular endothelial growth factor, FGF fibroblast growth factor.
Fig. 3
Fig. 3
Cells and key receptors/pathways targeted by current therapy strategies. RANKL receptor activator of nuclear factor-ΚB ligand, JAK Janus kinase/signal transducers.

References

    1. Silman AJ, Pearson JE. Epidemiology and genetics of rheumatoid arthritis. Arthritis Res. 2002;4(Suppl. 3):S265–S272.
    1. van der Linden MP, et al. Long-term impact of delay in assessment of patients with early arthritis. Arthritis Rheum. 2010;62:3537–3546.
    1. Moura CS, et al. Early medication use in new-onset rheumatoid arthritis may delay joint replacement: results of a large population-based study. Arthritis Res. Ther. 2015;17:197.
    1. Cho, S. K. et al. Factors associated with time to diagnosis from symptom onset in patients with early rheumatoid arthritis. Korean J. Intern. Med.113, (2017).
    1. Raza K, et al. Delays in assessment of patients with rheumatoid arthritis: variations across Europe. Ann. Rheum. Dis. 2011;70:1822–1825.
    1. Ometto F, et al. Methods used to assess remission and low disease activity in rheumatoid arthritis. Autoimmun. Rev. 2010;9:161–164.
    1. Grennan DM, Gray J, Loudon J, Fear S. Methotrexate and early postoperative complications in patients with rheumatoid arthritis undergoing elective orthopaedic surgery. Ann. Rheum. Dis. 2001;60:214–217.
    1. Nishimura K, et al. Meta-analysis: diagnostic accuracy of anti-cyclic citrullinated peptide antibody and rheumatoid factor for rheumatoid arthritis. Ann. Intern. Med. 2007;146:797–808.
    1. Bizzaro N, et al. Anti-cyclic citrullinated peptide antibody titer predicts time to rheumatoid arthritis onset in patients with undifferentiated arthritis: results from a 2-year prospective study. Arthritis Res. Ther. 2013;15:R16.
    1. Malmstrom V, Catrina AI, Klareskog L. The immunopathogenesis of seropositive rheumatoid arthritis: from triggering to targeting. Nat. Rev. Immunol. 2017;17:60–75.
    1. Padyukov L, et al. A genome-wide association study suggests contrasting associations in ACPA-positive versus ACPA-negative rheumatoid arthritis. Ann. Rheum. Dis. 2011;70:259–265.
    1. Schuerwegh AJ, et al. Evidence for a functional role of IgE anticitrullinated protein antibodies in rheumatoid arthritis. Proc. Natl Acad. Sci. USA. 2010;107:2586–2591.
    1. van Dongen H, et al. Efficacy of methotrexate treatment in patients with probable rheumatoid arthritis: a double-blind, randomized, placebo-controlled trial. Arthritis Rheum. 2007;56:1424–1432.
    1. Sellam J, et al. B cell activation biomarkers as predictive factors for the response to rituximab in rheumatoid arthritis: a six-month, national, multicenter, open-label study. Arthritis Rheum. 2011;63:933–938.
    1. Seegobin SD, et al. ACPA-positive and ACPA-negative rheumatoid arthritis differ in their requirements for combination DMARDs and corticosteroids: secondary analysis of a randomized controlled trial. Arthritis Res. Ther. 2014;16:R13.
    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.
    1. Okada Y, et al. Risk for ACPA-positive rheumatoid arthritis is driven by shared HLA amino acid polymorphisms in Asian and European populations. Hum. Mol. Genet. 2014;23:6916–6926.
    1. Mori M, Yamada R, Kobayashi K, Kawaida R, Yamamoto K. Ethnic differences in allele frequency of autoimmune-disease-associated SNPs. J. Hum. Genet. 2005;50:264–266.
    1. Nabi G, et al. Meta-analysis reveals PTPN22 1858C/T polymorphism confers susceptibility to rheumatoid arthritis in Caucasian but not in Asian population. Autoimmunity. 2016;49:197–210.
    1. Goh LL, et al. NLRP1, PTPN22 and PADI4 gene polymorphisms and rheumatoid arthritis in ACPA-positive Singaporean Chinese. Rheumatol. Int. 2017;37:1295–1302.
    1. McCarthy C, et al. Brief report: genetic variation of the alpha1 -antitrypsin gene is associated with increased autoantibody production in rheumatoid arthritis. Arthritis Rheumatol. 2017;69:1576–1579.
    1. Castaneda-Delgado JE, et al. Type I interferon gene response is increased in early and established rheumatoid arthritis and correlates with autoantibody production. Front. Immunol. 2017;8:285.
    1. Ding B, et al. Different patterns of associations with anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis in the extended major histocompatibility complex region. Arthritis Rheum. 2009;60:30–38.
    1. Schiff MH, et al. Long-term experience with etanercept in the treatment of rheumatoid arthritis in elderly and younger patients: patient-reported outcomes from multiple controlled and open-label extension studies. Drugs Aging. 2006;23:167–178.
    1. Frisell T, et al. Familial risks and heritability of rheumatoid arthritis: role of rheumatoid factor/anti-citrullinated protein antibody status, number and type of affected relatives, sex, and age. Arthritis Rheum. 2013;65:2773–2782.
    1. Kuo CF, et al. Familial aggregation of rheumatoid arthritis and co-aggregation of autoimmune diseases in affected families: a nationwide population-based study. Rheumatology. 2017;56:928–933.
    1. Svendsen AJ, et al. On the origin of rheumatoid arthritis: the impact of environment and genes--a population based twin study. PLoS ONE. 2013;8:e57304.
    1. Hensvold AH, et al. Environmental and genetic factors in the development of anticitrullinated protein antibodies (ACPAs) and ACPA-positive rheumatoid arthritis: an epidemiological investigation in twins. Ann. Rheum. Dis. 2015;74:375–380.
    1. van der Woude D, et al. Gene-environment interaction influences the reactivity of autoantibodies to citrullinated antigens in rheumatoid arthritis. Nat. Genet. 2010;42:814–816.
    1. Stolt P, et al. Silica exposure among male current smokers is associated with a high risk of developing ACPA-positive rheumatoid arthritis. Ann. Rheum. Dis. 2010;69:1072–1076.
    1. Mohamed BM, et al. Citrullination of proteins: a common post-translational modification pathway induced by different nanoparticles in vitro and in vivo. Nanomedicine. 2012;7:1181–1195.
    1. Too CL, et al. Occupational exposure to textile dust increases the risk of rheumatoid arthritis: results from a Malaysian population-based case-control study. Ann. Rheum. Dis. 2016;75:997–1002.
    1. Watkin LB, et al. COPA mutations impair ER-Golgi transport and cause hereditary autoimmune-mediated lung disease and arthritis. Nat. Genet. 2015;47:654–660.
    1. Klareskog L, et al. A new model for an etiology of rheumatoid arthritis: smoking may trigger HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by citrullination. Arthritis Rheum. 2006;54:38–46.
    1. Meng W, et al. DNA methylation mediates genotype and smoking interaction in the development of anti-citrullinated peptide antibody-positive rheumatoid arthritis. Arthritis Res. Ther. 2017;19:71.
    1. Konig MF, et al. Aggregatibacter actinomycetemcomitans-induced hypercitrullination links periodontal infection to autoimmunity in rheumatoid arthritis. Sci. Transl. Med. 2016;8:369ra176.
    1. Wegner N, et al. Peptidylarginine deiminase from Porphyromonas gingivalis citrullinates human fibrinogen and alpha-enolase: implications for autoimmunity in rheumatoid arthritis. Arthritis Rheum. 2010;62:2662–2672.
    1. Khandpur R, et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci. Transl. Med. 2013;5:178ra40.
    1. Alspaugh MA, Henle G, Lennette ET, Henle W. Elevated levels of antibodies to Epstein-Barr virus antigens in sera and synovial fluids of patients with rheumatoid arthritis. J. Clin. Invest. 1981;67:1134–1140.
    1. Wu X, et al. Molecular insight into gut microbiota and rheumatoid arthritis. Int. J. Mol. Sci. 2016;17:431.
    1. Chen J, et al. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med. 2016;8:43.
    1. Gan RW, et al. The association between omega-3 fatty acid biomarkers and inflammatory arthritis in an anti-citrullinated protein antibody positive population. Rheumatology. 2017;56:2229–2236.
    1. Hu Y, et al. Long-term dietary quality and risk of developing rheumatoid arthritis in women. Ann. Rheum. Dis. 2017;76:1357–1364.
    1. Orellana C, et al. Oral contraceptives, breastfeeding and the risk of developing rheumatoid arthritis: results from the Swedish EIRA study. Ann. Rheum. Dis. 2017;76:1845–1852.
    1. Alpizar-Rodriguez D, et al. Female hormonal factors and the development of anti-citrullinated protein antibodies in women at risk of rheumatoid arthritis. Rheumatology. 2017;56:1579–1585.
    1. van der Woude D, et al. Epitope spreading of the anti-citrullinated protein antibody response occurs before disease onset and is associated with the disease course of early arthritis. Ann. Rheum. Dis. 2010;69:1554–1561.
    1. Krishnamurthy A, et al. Identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss. Ann. Rheum. Dis. 2016;75:721–729.
    1. Wigerblad G, et al. Autoantibodies to citrullinated proteins induce joint pain independent of inflammation via a chemokine-dependent mechanism. Ann. Rheum. Dis. 2016;75:730–738.
    1. Pianta A, et al. Two rheumatoid arthritis-specific autoantigens correlate microbial immunity with autoimmune responses in joints. J. Clin. Invest. 2017;127:2946–2956.
    1. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011;365:2205–2219.
    1. Burmester GR, Dimitriu-Bona A, Waters SJ, Winchester RJ. Identification of three major synovial lining cell populations by monoclonal antibodies directed to Ia antigens and antigens associated with monocytes/macrophages and fibroblasts. Scand. J. Immunol. 1983;17:69–82.
    1. Lu MC, et al. Anti-citrullinated protein antibodies bind surface-expressed citrullinated Grp78 on monocyte/macrophages and stimulate tumor necrosis factor alpha production. Arthritis Rheum. 2010;62:1213–1223.
    1. Bae S, et al. alpha-Enolase expressed on the surfaces of monocytes and macrophages induces robust synovial inflammation in rheumatoid arthritis. J. Immunol. 2012;189:365–372.
    1. Quero L, Hanser E, Manigold T, Tiaden AN, Kyburz D. TLR2 stimulation impairs anti-inflammatory activity of M2-like macrophages, generating a chimeric M1/M2 phenotype. Arthritis Res. Ther. 2017;19:245.
    1. Fukui S, et al. M1 and M2 monocytes in rheumatoid arthritis: a contribution of imbalance of M1/M2 monocytes to osteoclastogenesis. Front. Immunol. 2017;8:1958.
    1. Hueber AJ, et al. Mast cells express IL-17A in rheumatoid arthritis synovium. J. Immunol. 2010;184:3336–3340.
    1. Suurmond J, et al. Toll-like receptor triggering augments activation of human mast cells by anti-citrullinated protein antibodies. Ann. Rheum. Dis. 2015;74:1915–1923.
    1. Zvaifler NJ, Steinman RM, Kaplan G, Lau LL, Rivelis M. Identification of immunostimulatory dendritic cells in the synovial effusions of patients with rheumatoid arthritis. J. Clin. Invest. 1985;76:789–800.
    1. Yang Z, et al. Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci. Transl. Med. 2016;8:331ra38.
    1. Edwards JC. The nature and origins of synovium: experimental approaches to the study of synoviocyte differentiation. J. Anat. 1994;184(Pt 3):493–501.
    1. Filer A, et al. Differential survival of leukocyte subsets mediated by synovial, bone marrow, and skin fibroblasts: site-specific versus activation-dependent survival of T cells and neutrophils. Arthritis Rheum. 2006;54:2096–2108.
    1. Aupperle KR, et al. Regulation of synoviocyte proliferation, apoptosis, and invasion by the p53 tumor suppressor gene. Am. J. Pathol. 1998;152:1091–1098.
    1. Schett G, et al. Enhanced expression of heat shock protein 70 (hsp70) and heat shock factor 1 (HSF1) activation in rheumatoid arthritis synovial tissue. Differential regulation of hsp70 expression and hsf1 activation in synovial fibroblasts by proinflammatory cytokines, shear stress, and antiinflammatory drugs. J. Clin. Invest. 1998;102:302–311.
    1. Amano T, et al. Synoviolin/Hrd1, an E3 ubiquitin ligase, as a novel pathogenic factor for arthropathy. Genes Dev. 2003;17:2436–2449.
    1. Sergijenko A, Roelofs AJ, Riemen AH, De Bari C. Bone marrow contribution to synovial hyperplasia following joint surface injury. Arthritis Res. Ther. 2016;18:166.
    1. Sabeh F, Fox D, Weiss SJ. Membrane-type I matrix metalloproteinase-dependent regulation of rheumatoid arthritis synoviocyte function. J. Immunol. 2010;184:6396–6406.
    1. Pap T, Korb-Pap A. Cartilage damage in osteoarthritis and rheumatoid arthritis--two unequal siblings. Nat. Rev. Rheumatol. 2015;11:606–615.
    1. Okamoto K, et al. Osteoimmunology: the conceptual framework unifying the immune and skeletal systems. Physiol. Rev. 2017;97:1295–1349.
    1. Pettit AR, Walsh NC, Manning C, Goldring SR, Gravallese EM. RANKL protein is expressed at the pannus-bone interface at sites of articular bone erosion in rheumatoid arthritis. Rheumatology. 2006;45:1068–1076.
    1. Harre U, et al. Induction of osteoclastogenesis and bone loss by human autoantibodies against citrullinated vimentin. J. Clin. Invest. 2012;122:1791–1802.
    1. Borrero CG, Mountz JM, Mountz JD. Emerging MRI methods in rheumatoid arthritis. Nat. Rev. Rheumatol. 2011;7:85–95.
    1. Xu X, et al. Aberrant activation of TGF-beta in subchondral bone at the onset of rheumatoid arthritis joint destruction. J. Bone Miner. Res. 2015;30:2033–2043.
    1. Arts EE, Fransen J, Den Broeder AA, van Riel P, Popa CD. Low disease activity (DAS28</=3.2) reduces the risk of first cardiovascular event in rheumatoid arthritis: a time-dependent Cox regression analysis in a large cohort study. Ann. Rheum. Dis. 2017;76:1693–1699.
    1. Myasoedova E, et al. Total cholesterol and LDL levels decrease before rheumatoid arthritis. Ann. Rheum. Dis. 2010;69:1310–1314.
    1. Chen YJ, Chang YT, Wang CB, Wu CY. The risk of cancer in patients with rheumatoid arthritis: a nationwide cohort study in Taiwan. Arthritis Rheum. 2011;63:352–358.
    1. Deane KD. Can rheumatoid arthritis be prevented? Best Pract. Res. Clin. Rheumatol. 2013;27:467–485.
    1. Finckh A, Liang MH, van Herckenrode CM, de Pablo P. Long-term impact of early treatment on radiographic progression in rheumatoid arthritis: a meta-analysis. Arthritis Rheum. 2006;55:864–872.
    1. Fuchs HA, Kaye JJ, Callahan LF, Nance EP, Pincus T. Evidence of significant radiographic damage in rheumatoid arthritis within the first 2 years of disease. J. Rheumatol. 1989;16:585–591.
    1. Cohen S, Emery P. The American College of Rheumatology/European League Against Rheumatism criteria for the classification of rheumatoid arthritis: a game changer. Arthritis Rheum. 2010;62:2592–2594.
    1. Sanmarti R, et al. Update of the Consensus Statement of the Spanish Society of Rheumatology on the use of biological therapies in rheumatoid arthritis. Reumatol. Clin. 2015;11(5):279–94.
    1. Lopez-Olivo MA, et al. Risk of malignancies in patients with rheumatoid arthritis treated with biologic therapy: a meta-analysis. JAMA. 2012;308:898–908.
    1. Rezaei H, et al. Evaluation of hand bone loss by digital X-ray radiogrammetry as a complement to clinical and radiographic assessment in early rheumatoid arthritis: results from the SWEFOT trial. BMC Musculoskelet. Disord. 2013;14:79.
    1. Brown PM, Pratt AG, Isaacs JD. Mechanism of action of methotrexate in rheumatoid arthritis, and the search for biomarkers. Nat. Rev. Rheumatol. 2016;12:731–742.
    1. Owen SA, et al. Genetic polymorphisms in key methotrexate pathway genes are associated with response to treatment in rheumatoid arthritis patients. Pharmacogenomics J. 2013;13:227–234.
    1. Schiff MH, Sadowski P. Oral to subcutaneous methotrexate dose-conversion strategy in the treatment of rheumatoid arthritis. Rheumatol. Int. 2017;37(2):213–218.
    1. Al Maashari R, Hamodat MM. Methotrexate-induced panniculitis in a patient with rheumatoid arthritis. Acta Dermatovenerol. Alp. Pannonica Adriat. 2016;25:79–81.
    1. Herrmann ML, Schleyerbach R, Kirschbaum BJ. Leflunomide: an immunomodulatory drug for the treatment of rheumatoid arthritis and other autoimmune diseases. Immunopharmacology. 2000;47:273–289.
    1. Smolen JS, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann. Rheum. Dis. 2014;73:492–509.
    1. Strand V, et al. Treatment of active rheumatoid arthritis with leflunomide compared with placebo and methotrexate. Leflunomide Rheumatoid Arthritis Investigators Group. Arch. Intern. Med. 1999;159:2542–2550.
    1. Kalden JR, et al. The efficacy and safety of leflunomide in patients with active rheumatoid arthritis: a five-year followup study. Arthritis Rheum. 2003;48:1513–1520.
    1. Schiff MH, Strand V, Oed C, Loew-Friedrich I. Leflunomide: efficacy and safety in clinical trials for the treatment of rheumatoid arthritis. Drugs Today (Barc.). 2000;36:383–394.
    1. Janssen NM, Genta MS. The effects of immunosuppressive and anti-inflammatory medications on fertility, pregnancy, and lactation. Arch. Intern. Med. 2000;160:610–619.
    1. Schuna AA. Update on treatment of rheumatoid arthritis. J. Am. Pharm. Assoc. 1998;38:728–735.
    1. Plosker GL, Croom KF. Sulfasalazine: a review of its use in the management of rheumatoid arthritis. Drugs. 2005;65:1825–1849.
    1. Smolen JS, et al. Efficacy and safety of leflunomide compared with placebo and sulphasalazine in active rheumatoid arthritis: a double-blind, randomised, multicentre trial. European Leflunomide Study Group. Lancet. 1999;353:259–266.
    1. Lee CK, 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;50:3831–3843.
    1. Rodenburg RJ, Ganga A, van Lent PL, van de Putte LB, van Venrooij WJ. The antiinflammatory drug sulfasalazine inhibits tumor necrosis factor alpha expression in macrophages by inducing apoptosis. Arthritis Rheum. 2000;43:1941–1950.
    1. Hirohata S, Ohshima N, Yanagida T, Aramaki K. Regulation of human B cell function by sulfasalazine and its metabolites. Int. Immunopharmacol. 2002;2:631–640.
    1. Volin MV, Campbell PL, Connors MA, Woodruff DC, Koch AE. The effect of sulfasalazine on rheumatoid arthritic synovial tissue chemokine production. Exp. Mol. Pathol. 2002;73:84–92.
    1. Ostensen M. Treatment with immunosuppressive and disease modifying drugs during pregnancy and lactation. Am. J. Reprod. Immunol. 1992;28:148–152.
    1. Sames E, Paterson H, Li C. Hydroxychloroquine-induced agranulocytosis in a patient with long-term rheumatoid arthritis. Eur. J. Rheumatol. 2016;3:91–92.
    1. Katz SJ, Russell AS. Re-evaluation of antimalarials in treating rheumatic diseases: re-appreciation and insights into new mechanisms of action. Curr. Opin. Rheumatol. 2011;23:278–281.
    1. van der Heijde DM, van Riel PL, Nuver-Zwart IH, van de Putte LB. Alternative methods for analysis of radiographic damage in a randomized, double blind, parallel group clinical trial comparing hydroxychloroquine and sulfasalazine. J. Rheumatol. 2000;27:535–538.
    1. Kim JW, et al. Risk of retinal toxicity in longterm users of hydroxychloroquine. J. Rheumatol. 2017;44:1674–1679.
    1. Van Doornum S, McColl G, Wicks IP. Tumour necrosis factor antagonists improve disease activity but not arterial stiffness in rheumatoid arthritis. Rheumatology. 2005;44:1428–1432.
    1. Parameswaran N, Patial S. Tumor necrosis factor-alpha signaling in macrophages. Crit. Rev. Eukaryot. Gene. Expr. 2010;20:87–103.
    1. Dulai R, et al. The effect of tumor necrosis factor-alpha antagonists on arterial stiffness in rheumatoid arthritis: a literature review. Semin. Arthritis Rheum. 2012;42:1–8.
    1. Olofsson T, et al. Predictors of work disability after start of anti-TNF therapy in a national cohort of Swedish patients with rheumatoid arthritis: does early anti-TNF therapy bring patients back to work? Ann. Rheum. Dis. 2017;76(7):1245–1252.
    1. Low AS, et al. Relationship between exposure to tumour necrosis factor inhibitor therapy and incidence and severity of myocardial infarction in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2017;76:654–660.
    1. Schneeweiss S, et al. Anti-tumor necrosis factor alpha therapy and the risk of serious bacterial infections in elderly patients with rheumatoid arthritis. Arthritis Rheum. 2007;56:1754–1764.
    1. Lemery SJ, Ricci MS, Keegan P, McKee AE, Pazdur R. FDA’s approach to regulating biosimilars. Clin. Cancer Res. 2016;23(8):1882–1885.
    1. Schaible TF. Long term safety of infliximab. Can. J. Gastroenterol. 2000;14(Suppl. C):29C–32C.
    1. Braun, J. & Kay, J. The safety of emerging biosimilar drugs for the treatment of rheumatoid arthritis. Expert. Opin. Drug. Saf.16, 289–302 (2017).
    1. Baeten D, et al. Immunomodulatory effects of anti-tumor necrosis factor alpha therapy on synovium in spondylarthropathy: histologic findings in eight patients from an open-label pilot study. Arthritis Rheum. 2001;44:186–195.
    1. Weinblatt ME, et al. Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA trial. Arthritis Rheum. 2003;48:35–45.
    1. van Schouwenburg PA, et al. Adalimumab elicits a restricted anti-idiotypic antibody response in autoimmune patients resulting in functional neutralisation. Ann. Rheum. Dis. 2013;72:104–109.
    1. Cohen S, et al. Efficacy and safety of the biosimilar ABP 501 compared with adalimumab in patients with moderate to severe rheumatoid arthritis: a randomised, double-blind, phase III equivalence study. Ann. Rheum. Dis. 2017;76:1679–1687.
    1. Moreland LW, et al. Etanercept therapy in rheumatoid arthritis. A randomized, controlled trial. Ann. Intern. Med. 1999;130:478–486.
    1. Genovese MC, et al. Longterm safety, efficacy, and radiographic outcome with etanercept treatment in patients with early rheumatoid arthritis. J. Rheumatol. 2005;32:1232–1242.
    1. Singh, J. A., Noorbaloochi, S. & Singh, G. Golimumab for rheumatoid arthritis. The Cochrane database of systematic reviews (2010). CD008341.
    1. Kay J, et al. Golimumab 3-year safety update: an analysis of pooled data from the long-term extensions of randomised, double-blind, placebo-controlled trials conducted in patients with rheumatoid arthritis, psoriatic arthritis or ankylosing spondylitis. Ann. Rheum. Dis. 2015;74:538–546.
    1. Ruiz Garcia, V. et al. Certolizumab pegol (CDP870) for rheumatoid arthritis in adults. The Cochrane database of systematic reviews (2014). CD007649.
    1. Steed PM, et al. Inactivation of TNF signaling by rationally designed dominant-negative TNF variants. Science. 2003;301:1895–1898.
    1. Melet J, et al. Rituximab-induced T cell depletion in patients with rheumatoid arthritis: association with clinical response. Arthritis Rheum. 2013;65:2783–2790.
    1. Cohen SB, et al. Continued inhibition of structural damage over 2 years in patients with rheumatoid arthritis treated with rituximab in combination with methotrexate. Ann. Rheum. Dis. 2010;69:1158–1161.
    1. Cancro MP. Signalling crosstalk in B cells: managing worth and need. Nat. Rev. Immunol. 2009;9:657–661.
    1. Moreland L, Bate G, Kirkpatrick P. Abatacept. Nat. Rev. Drug Discov. 2006;5:185–186.
    1. McInnes IB, Schett G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet. 2017;389:2328–2337.
    1. Ruiz-Limon P, et al. Tocilizumab improves the proatherothrombotic profile of rheumatoid arthritis patients modulating endothelial dysfunction, NETosis, and inflammation. Transl. Res. 2017;183:87–103.
    1. Navarro-Millan I, Singh JA, Curtis JR. Systematic review of tocilizumab for rheumatoid arthritis: a new biologic agent targeting the interleukin-6 receptor. Clin. Ther. 2012;34:788–802.
    1. Burmester GR, et al. Low immunogenicity of tocilizumab in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2017;76(6):1078–1085.
    1. Moots RJ, et al. Effect of tocilizumab on neutrophils in adult patients with rheumatoid arthritis: pooled analysis of data from phase 3 and 4 clinical trials. Rheumatology. 2017;56(4):541–549.
    1. Aletaha D, et al. Efficacy and safety of sirukumab in patients with active rheumatoid arthritis refractory to anti-TNF therapy (SIRROUND-T): a randomised, double-blind, placebo-controlled, parallel-group, multinational, phase 3 study. Lancet. 2017;389:1206–1217.
    1. Weinblatt ME, et al. The efficacy and safety of subcutaneous clazakizumab in patients with moderate-to-severe rheumatoid arthritis and an inadequate response to methotrexate: results from a multinational, phase IIb, randomized, double-blind, placebo/active-controlled, dose-ranging study. Arthritis Rheumatol. 2015;67:2591–2600.
    1. Kim GW, et al. IL-6 inhibitors for treatment of rheumatoid arthritis: past, present, and future. Arch. Pharm. Res. 2015;38:575–584.
    1. Boissier MC. Cell and cytokine imbalances in rheumatoid synovitis. Joint Bone Spine. 2011;78:230–234.
    1. Ikonomidis I, et al. Lowering interleukin-1 activity with anakinra improves myocardial deformation in rheumatoid arthritis. Heart. 2009;95:1502–1507.
    1. Schatz A, et al. Resolution of pericardial constriction with Anakinra in a patient with effusive-constrictive pericarditis secondary to rheumatoid arthritis. Int. J. Cardiol. 2016;223:215–216.
    1. Blanco FJ, et al. Secukinumab in active rheumatoid arthritis: a phase III randomized, double-blind, active comparator- and placebo-controlled Study. Arthritis Rheumatol. 2017;69:1144–1153.
    1. Smolen JS, et al. A randomised phase II study evaluating the efficacy and safety of subcutaneously administered ustekinumab and guselkumab in patients with active rheumatoid arthritis despite treatment with methotrexate. Ann. Rheum. Dis. 2017;76:831–839.
    1. Chiu YG, Ritchlin CT. Denosumab: targeting the RANKL pathway to treat rheumatoid arthritis. Expert Opin. Biol. Ther. 2017;17:119–128.
    1. Yue J, et al. Repair of bone erosion in rheumatoid arthritis by denosumab: a high-resolution peripheral quantitative computed tomography study. Arthritis Care Res. 2016;69(8):1156–1163.
    1. Burmester GR, et al. A randomised phase IIb study of mavrilimumab, a novel GM-CSF receptor alpha monoclonal antibody, in the treatment of rheumatoid arthritis. Ann. Rheum. Dis. 2017;76:1020–1030.
    1. Venkatesha SH, Dudics S, Acharya B, Moudgil KD. Cytokine-modulating strategies and newer cytokine targets for arthritis therapy. Int. J. Mol. Sci. 2014;16:887–906.
    1. Strand V, et al. Tofacitinib versus methotrexate in rheumatoid arthritis: patient-reported outcomes from the randomised phase III ORAL Start trial. RMD Open. 2016;2:e000308.
    1. Semerano L, Decker P, Clavel G, Boissier MC. Developments with investigational Janus kinase inhibitors for rheumatoid arthritis. Expert Opin. Investig. Drugs. 2016;25:1355–1359.
    1. Naldini L. Gene therapy returns to centre stage. Nature. 2015;526:351–360.
    1. Kim MJ, et al. Notch1 targeting siRNA delivery nanoparticles for rheumatoid arthritis therapy. J. Control. Release. 2015;216:140–148.
    1. Lee SJ, et al. TNF-alpha gene silencing using polymerized siRNA/thiolated glycol chitosan nanoparticles for rheumatoid arthritis. Mol. Ther. 2014;22:397–408.
    1. Lundstrom SL, et al. IgG Fc galactosylation predicts response to methotrexate in early rheumatoid arthritis. Arthritis Res. Ther. 2017;19:182.
    1. Whang JA, Chang BY. Bruton’s tyrosine kinase inhibitors for the treatment of rheumatoid arthritis. Drug Discov. Today. 2014;19:1200–1204.
    1. Alvaro-Gracia JM, et al. Intravenous administration of expanded allogeneic adipose-derived mesenchymal stem cells in refractory rheumatoid arthritis (Cx611): results of a multicentre, dose escalation, randomised, single-blind, placebo-controlled phase Ib/IIa clinical trial. Ann. Rheum. Dis. 2017;76:196–202.
    1. Kasarello K, Cudnoch-Jedrzejewska A, Czlonkowski A, Mirowska-Guzel D. Mechanism of action of three newly registered drugs for multiple sclerosis treatment. Pharmacol. Rep. 2017;69:702–708.
    1. Linares V, Alonso V, Domingo JL. Oxidative stress as a mechanism underlying sulfasalazine-induced toxicity. Expert Opin. Drug. Saf. 2011;10:253–263.
    1. Rainsford KD, Parke AL, Clifford-Rashotte M, Kean WF. Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology. 2015;23:231–269.
    1. Kim EY, Moudgil KD. Immunomodulation of autoimmune arthritis by pro-inflammatory cytokines. Cytokine. 2017;98:87–96.
    1. Mota P, Reddy V, Isenberg D. Improving B-cell depletion in systemic lupus erythematosus and rheumatoid arthritis. Expert Rev. Clin. Immunol. 2017;13:667–676.
    1. Mellado M, et al. T cell migration in rheumatoid arthritis. Front. Immunol. 2015;6:384.
    1. Raimondo MG, Biggioggero M, Crotti C, Becciolini A, Favalli EG. Profile of sarilumab and its potential in the treatment of rheumatoid arthritis. Drug Des. Dev. Ther. 2017;11:1593–1603.
    1. Cavalli G, Dinarello CA. Treating rheumatological diseases and co-morbidities with interleukin-1 blocking therapies. Rheumatology. 2015;54:2134–2144.
    1. Kim EK, et al. IL-17-mediated mitochondrial dysfunction impairs apoptosis in rheumatoid arthritis synovial fibroblasts through activation of autophagy. Cell Death Dis. 2017;8:e2565.
    1. Fassio A, et al. New strategies for the prevention and treatment of systemic and local bone loss; from pathophysiology to clinical application. Curr. Pharm. Des. 2017;23(41):6241–6250.
    1. Yamaoka K. Janus kinase inhibitors for rheumatoid arthritis. Curr. Opin. Chem. Biol. 2016;32:29–33.
    1. Winthrop KL, et al. The safety and immunogenicity of live zoster vaccination in patients with rheumatoid arthritis before starting tofacitinib: a randomized phase II trial. Arthritis Rheumatol. 2017;69:1969–1977.
    1. Elshabrawy HA, Essani AE, Szekanecz Z, Fox DA, Shahrara S. TLRs, future potential therapeutic targets for RA. Autoimmun. Rev. 2017;16:103–113.
    1. Bartok B, Hammaker D, Firestein GS. Phosphoinositide 3-kinase delta regulates migration and invasion of synoviocytes in rheumatoid arthritis. J. Immunol. 2014;192:2063–2070.
    1. Fechtner S, Fox DA, Ahmed S. Transforming growth factor beta activated kinase 1: a potential therapeutic target for rheumatic diseases. Rheumatology. 2016;56(7):1060–1068.
    1. Cheung TT, McInnes IB. Future therapeutic targets in rheumatoid arthritis? Semin. Immunopathol. 2017;39:487–500.
    1. Benham H, et al. Citrullinated peptide dendritic cell immunotherapy in HLA risk genotype-positive rheumatoid arthritis patients. Sci. Transl. Med. 2015;7:290ra87.

Source: PubMed

3
Abonnieren