Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Emerging targets of biologic therapies for rheumatoid arthritis

Abstract

Advances in molecular biology and the clinical success of strategies that target tumor necrosis factor (TNF) have led to further research into the pathophysiology of human rheumatoid arthritis. Several novel therapeutic targets have emerged from these efforts, including not only molecules that regulate TNF (e.g. TNF-α converting enzyme), the complex cytokine network (e.g. interleukin [IL]-6, IL-15, IL-17) and several adipokines, but also targets that originate from cellular and subcellular components of the disease. Strategies that aim at cellular targets include antibodies to CD20 or BLyS (also known as TNF ligand family member 13b), which deplete or inhibit B cells, as well as approaches that interfere with membrane-derived microparticles. Components of subcellular pathways, which are predominantly upstream of the central regulator of transcription nuclear factor κB, have also been studied. Of these, strategies that target mitogen-activated protein kinases have a leading role and are on the verge of clinical use; approaches that target specific molecules such as Janus kinases, signal transducer and activator of transcription proteins, and suppressor of cytokine signaling proteins also seem to show promise and might have a clinical application in the future.

Key Points

  • Biologics are a novel class of agents that are designed to target defined pathogenetic mechanisms and molecules with high specificity

  • Interleukin (IL)-6, IL-15, IL-17 and receptor activator of nuclear factor κB ligand form a complex cytokine network with IL-1 and tumor necrosis factor that promotes joint inflammation and joint destruction

  • Adipokines and microparticles are intriguing targets for new biologics

  • The mitogen-activated protein kinases and transcription factors such as nuclear factor κB are essential intracellular mediators of inflammation and, therefore, are potentially powerful targets, but must be selected carefully because of possible adverse effects

  • Components of intracellular signaling pathways upstream of nuclear factor κB seem to be safer, although potentially less effective, than downstream targets

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Potential intercellular targets for biologic therapy of RA
Figure 2: Potential intracellular targets for biologic therapy of RA

Similar content being viewed by others

References

  1. Furst DE et al. (2006) Updated consensus statement on biological agents for the treatment of rheumatic diseases, 2006. Ann Rheum Dis 65 (Suppl 3): iii2–iii15

    PubMed  PubMed Central  Google Scholar 

  2. Pollard L and Choy E (2005) Rheumatoid arthritis: non-tumor necrosis factor targets. Curr Opin Rheumatol 17: 242–246

    Article  CAS  Google Scholar 

  3. Study to assess the effect of tocilizumab plus methotrexate on signs and symptoms in patients with moderate to severe active rheumatoid arthritis [http://www.clinicaltrials.gov/ct/show/NCT00106548?order=1] (accessed 28 March 2007)

  4. Choy EH et al. (2002) Therapeutic benefit of blocking interleukin-6 activity with an anti-interleukin-6 receptor monoclonal antibody in rheumatoid arthritis: a randomized, double-blind, placebo-controlled, dose-escalation trial. Arthritis Rheum 46: 3143–3150

    Article  CAS  Google Scholar 

  5. McInnes IB et al. (2003) New strategies to control inflammatory synovitis: interleukin 15 and beyond. Ann Rheum Dis 62 (Suppl 2): ii51–ii54

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Ferrari-Lacraz S et al. (2004) Targeting IL-15 receptor-bearing cells with an antagonist mutant IL-15–Fc protein prevents disease development and progression in murine collagen-induced arthritis. J Immunol 173: 5818–5826

    Article  CAS  Google Scholar 

  7. Miranda-Carús ME et al. (2004) IL-15 and the initiation of cell contact-dependent synovial fibroblast-T lympocyte cross-talk in rheumatoid arthritis: effect of methotrexate. J Immunol 173: 1463–1476

    Article  Google Scholar 

  8. Ruchatz H et al. (1998) Soluble IL-15 receptor alpha-chain administration prevents murine collagen-induced arthritis: a role for IL-15 in development of antigen-induced immunopathology. J Immunol 160: 5654–5660

    CAS  PubMed  Google Scholar 

  9. Baslund B et al. (2005) Targeting interleukin-15 in patients with rheumatoid arthritis: a proof-of-concept study. Arthritis Rheum 52: 2686–2692

    Article  CAS  Google Scholar 

  10. McKenzie BS et al. (2006) Understanding the IL-23–IL-17 immune pathway. Trends Immunol 27: 17–23

    Article  CAS  Google Scholar 

  11. Kim HR et al. (2007) Up-regulation of IL-23p19 expression in rheumatoid arthritis synovial fibroblasts by IL-17 through PI3-kinase-, NF-κB- and p38 MAPK-dependent signaling pathways. Rheumatology 46: 57–64

    Article  CAS  Google Scholar 

  12. Hwang SY et al. (2004) IL-17 induces production of IL-6 and IL-8 in rheumatoid arthritis synovial fibroblasts via NF-κB- and PI3-kinase/Akt-dependent pathways. Arthritis Res Ther 6: R120–R128

    Article  CAS  Google Scholar 

  13. Lubberts E (2003) The role of IL-17 and family members in the pathogenesis of arthritis. Curr Opin Investig Drugs 4: 572–577

    CAS  PubMed  Google Scholar 

  14. Lubberts E et al. (2005) The role of T-cell interleukin-17 in conducting destructive arthritis: lessons from animal models. Arthritis Res Ther 7: 29–37

    Article  CAS  Google Scholar 

  15. Lubberts E et al. (2004) Treatment with a neutralizing anti-murine interleukin-17 antibody after the onset of collagen-induced arthritis reduces joint inflammation, cartilage destruction, and bone erosion. Arthritis Rheum 50: 650–659

    Article  CAS  Google Scholar 

  16. Schett G et al. (2005) Mechanisms of disease: the link between RANKL and arthritic bone disease. Nat Clin Pract Rheumatol 1: 47–54

    Article  CAS  Google Scholar 

  17. Neumann E et al. (2005) The RANK/RANKL/osteoprotegerin system in rheumatoid arthritis: new insights from animal models. Arthritis Rheum 52: 2960–2967

    Article  CAS  Google Scholar 

  18. Geusens PP et al. (2006) The ratio of circulating osteoprotegerin to RANKL in early rheumatoid arthritis predicts later joint destruction. Arthritis Rheum 54: 1772–1777

    Article  CAS  Google Scholar 

  19. Bekker PJ et al. (2001) The effect of a single dose of osteoprotegerin in postmenopausal women. J Bone Miner Res 16: 348–360

    Article  CAS  Google Scholar 

  20. Bekker PJ et al. (2004) A single-dose placebo controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res 19: 1059–1066

    Article  CAS  Google Scholar 

  21. Edwards JC et al. (2006) B cell depletion therapy in rheumatic disease. Best Pract Res Clin Rheumatol 20: 915–928

    Article  CAS  Google Scholar 

  22. Cheema GS et al. (2001) Elevated serum B lymphocyte stimulator levels in patients with systemic immune-based rheumatic diseases. Arthritis Rheum 44: 1313–1319

    Article  CAS  Google Scholar 

  23. Seyler TM et al. (2005) BLyS and APRIL in rheumatoid arthritis. J Clin Invest 115: 1083–1092

    Article  Google Scholar 

  24. Ohata J et al. (2005) Fibroblast-like synoviocytes of mesenchymal origin express functional B cell-activating factor of the TNF family in response to proinflammatory cytokines. J Immunol 174: 864–870

    Article  CAS  Google Scholar 

  25. Wang H et al. (2001) TACI-ligand interactions are required for T cell activation and collagen-induced arthritis in mice. Nat Immunol 2: 632–637

    Article  CAS  Google Scholar 

  26. Gross JA et al. (2001) TACI-Ig neutralizes molecules critical for B cell development and autoimmune disease: impaired B cell maturation in mice lacking BLyS. Immunity 15: 289–302

    Article  CAS  Google Scholar 

  27. Ding C and Jones G (2006) Belimumab Human Genome Sciences/Cambridge Antibody Technology/GlaxoSmithKline. Curr Opin Investig Drugs 7: 464–472

    CAS  PubMed  Google Scholar 

  28. Cohen SB et al. (2006) Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks. Arthritis Rheum 54: 2793–2806

    Article  CAS  Google Scholar 

  29. Roschke V et al. (2002) BLyS and APRIL form biologically active heterotrimers that are expressed in patients with systemic immune-based rheumatic diseases. J Immunol 169: 4314–4321

    Article  CAS  Google Scholar 

  30. Wagner G and Laufer S (2006) Small molecular anti-cytokine agents. Med Res Rev 26: 1–62

    Article  CAS  Google Scholar 

  31. Conway JG et al. (2001) Inhibition of tumor necrosis factor-α (TNF-α) production and arthritis in the rat by GW3333, a dual inhibitor of TNF-α-converting enzyme and matrix metalloproteinases. J Pharmacol Exp Ther 298: 900–908

    CAS  PubMed  Google Scholar 

  32. Schäffler A et al. (2006) Role of adipose tissue as an inflammatory organ in human diseases. Endocr Rev 27: 449–467

    Article  Google Scholar 

  33. Neumeier M et al. (2006) Different effects of adiponectin isoforms in human monocytic cells. J Leukoc Biol 79: 803–808

    Article  CAS  Google Scholar 

  34. Weigert J et al. (2005) The adiponectin paralog CORS-26 has anti-inflammatory properties and is produced by human monocytic cells. FEBS Lett 579: 5565–5570

    Article  CAS  Google Scholar 

  35. Otero M et al. (2006) Towards a pro-inflammatory and immunomodulatory emerging role of leptin. Rheumatology 45: 944–950

    Article  CAS  Google Scholar 

  36. Otero M et al. (2006) Changes in plasma levels of fat-derived hormones adiponectin, leptin, resistin and visfatin in patients with rheumatoid arthritis. Ann Rheum Dis 65: 1198–1201

    Article  CAS  Google Scholar 

  37. Schäffler A et al. (2003) Adipocytokines in synovial fluid. JAMA 290: 1709–1710

    Article  Google Scholar 

  38. Ehling A et al. (2006) The potential of adiponectin in driving arthritis. J Immunol 176: 4468–4478

    Article  CAS  Google Scholar 

  39. Busso N et al. (2002) Leptin signaling deficiency impairs humoral and cellular immune responses and attenuates experimental arthritis. J Immunol 168: 875–882

    Article  CAS  Google Scholar 

  40. Bokarewa M et al. (2005) Resistin, an adipokine with potent proinflammatory properties. J Immunol 174: 5789–5795

    Article  CAS  Google Scholar 

  41. Distler JH et al. (2005) Microparticles as regulators of inflammation: novel players of cellular crosstalk in the rheumatic diseases. Arthritis Rheum 52: 3337–3348

    Article  CAS  Google Scholar 

  42. Knijff-Dutmer EA et al. (2002) Elevated levels of platelet microparticles are associated with disease activity in rheumatoid arthritis. Arthritis Rheum 46: 1498–1503

    Article  CAS  Google Scholar 

  43. Berckmans RJ et al. (2002) Cell-derived microparticles in synovial fluid from inflamed arthritic joints support coagulation exclusively via a factor VII-dependent mechanism. Arthritis Rheum 46: 2857–2866

    Article  CAS  Google Scholar 

  44. Berckmans RJ et al. (2005) Synovial microparticles from arthritic patients modulate chemokine and cytokine release by synoviocytes. Arthritis Res Ther 7: R536–R544

    Article  CAS  Google Scholar 

  45. Kim SH et al. (2005) Exosomes derived from IL-10-treated dendritic cells can suppress inflammation and collagen-induced arthritis. J Immunol 174: 6440–6448

    Article  CAS  Google Scholar 

  46. Zhang HG et al. (2006) A membrane form of TNF-alpha presented by exosomes delays T cell activation-induced cell death. J Immunol 176: 7385–7393

    Article  CAS  Google Scholar 

  47. McIntyre KW et al. (2003) A highly selective inhibitor of I kappa B kinase, BMS-345541, blocks both joint inflammation and destruction in collagen-induced arthritis in mice. Arthritis Rheum 48: 2652–2659

    Article  CAS  Google Scholar 

  48. Sweeney SE and Firestein GS (2004) Signal transduction in rheumatoid arthritis. Curr Opin Rheumatol 16: 231–237

    Article  CAS  Google Scholar 

  49. Firestein GS (2006) Inhibiting inflammation in rheumatoid arthritis. N Engl J Med 354: 80–82

    Article  CAS  Google Scholar 

  50. Görtz B et al. (2005) Tumour necrosis factor activates the mitogen-activated protein kinases p38α and ERK in the synovial membrane in vivo. Arthritis Res Ther 7: R1140–R1147

    Article  Google Scholar 

  51. Mbalaviele G et al. (2006) Inhibition of p38 mitogen-activated protein kinase prevents inflammatory bone destruction. J Pharmacol Exp Ther 317: 1044–1053

    Article  CAS  Google Scholar 

  52. Medicherla S et al. (2006) A selective p38 alpha mitogen-activated protein kinase inhibitor reverses cartilage and bone destruction in mice with collagen-induced arthritis. J Pharmacol Exp Ther 318: 132–141

    Article  CAS  Google Scholar 

  53. A study of P38 inhibitor (4) in patients with active rheumatoid arthritis (RA) on stable methotrexate therapy [www.clinicaltrials.gov/ct/show/NCT00316771?order=1] (accessed 28 March 2007)

  54. Han Z et al. (2001) c-Jun N-terminal kinase is required for metalloproteinase expression and joint destruction in inflammatory arthritis. J Clin Invest 108: 73–81

    Article  CAS  Google Scholar 

  55. Chabaud-Riou M and Firestein GS (2004) Expression and activation of mitogen-activated protein kinase kinase-3 and -6 in rheumatoid arthritis. Am J Pathol 164: 177–184

    Article  CAS  Google Scholar 

  56. O'Shea JJ et al. (2005) New strategies for immunosuppression: interfering with cytokines by targeting the JAK/STAT pathway. Curr Opin Rheumatol 17: 305–311

    Article  CAS  Google Scholar 

  57. Egan PJ et al. (2003) Suppressor of cytokine signaling-1 regulates acute inflammatory arthritis and T cell activation. J Clin Invest 111: 915–924

    Article  CAS  Google Scholar 

  58. Wong PK et al. (2006) SOCS-3 negatively regulates innate and adaptive immune mechanisms in acute IL-1-dependent inflammatory arthritis. J Clin Invest 116: 1571–1581

    Article  CAS  Google Scholar 

  59. Shouda T et al. (2001) Induction of the cytokine signal regulator SOCS3/CIS3 as a therapeutic strategy for treating inflammatory arthritis. J Clin Invest 108: 1781–1788

    Article  CAS  Google Scholar 

  60. Kremer JM et al. (2006) A randomized, double-blind, placebo-controlled trial of 3 dose levels of CP-690,550 versus placebo in the treatment of active rheumatoid arthritis. In Program and abstracts of the American College of Rheumatology 70th Annual Meeting; November 11–15, 2006: Washington, DC.

    Google Scholar 

  61. Weimar W et al. (2006) Phase 1 evaluation of the safety of a JAK3 inhibitor, CP-690,550 in stable kidney transplant recipients [abstract]. Transplantation 82 (Suppl 2): 86

    Google Scholar 

  62. Comparison of six CP-690,550 doses vs placebo, each combined with methotrexate, for the treatment of rheumatoid arthritis [www.clinicaltrials.gov/ct/show/NCT00413660?order=2] (accessed 28 March 2007)

  63. Ospelt C et al. (2005) Gene analysis for exploring the effects of drugs in rheumatoid arthritis. Arthritis Rheum 52: 2248–2256

    Article  CAS  Google Scholar 

  64. Bongartz T et al. (2006) Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA 295: 2275–2285

    Article  CAS  Google Scholar 

  65. Setoguchi S et al. (2006) Tumor necrosis factor alpha antagonist use and cancer in patients with rheumatoid arthritis. Arthritis Rheum 54: 2757–2764

    Article  CAS  Google Scholar 

Download references

Acknowledgements

IH Tarner was funded in part by the Deutsche Forschungsgemeinschaft. U Müller-Ladner was funded by the Deutsche Forschungsgemeinschaft. S Gay was funded by the Swiss National Science Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Steffen Gay.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Tarner, I., Müller-Ladner, U. & Gay, S. Emerging targets of biologic therapies for rheumatoid arthritis. Nat Rev Rheumatol 3, 336–345 (2007). https://doi.org/10.1038/ncprheum0506

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncprheum0506

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing