#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Biological treatment in dermatology – psoriasis


Authors: K. Ettler
Authors‘ workplace: Klinika nemocí kožních a pohlavních Lékařské fakulty UK a FN Hradec Králové, přednosta doc. MUDr. Karel Ettler, CSc.
Published in: Vnitř Lék 2011; 57(9): 684-692
Category: 65th birthday Mudr. Jany Laciné and and 60th birthday Milana Tržila

Overview

Psoriasis is a chronic, relapsing skin disease affecting 1–2% of the population. Genetically-determined autoimmune inflammation is initiated by an infection (e.g. streptococcal). T-lymphocytes, producing a number of cytokines, mainly of the Th1 profile, including TNFα, play a key role. The role of Th17 and its interleukin IL-23 has recently been confirmed. It contains p40 protein, also a component of IL-12. TNFα and p40 blockade appear to be an effective biological treatment of severe lesional psoriasis. Further research and new drug development is to assure modern individualized treatment of psoriasis as well as other skin diseases.

Key words:
biological treatment of psoriasis – TNFα blockers – IL-12/23 blockers


Sources

1. Gelfand JM, Troxel AB, Lewis JD et al. The risk of mortality in patients with psoriasis: results from a population-based study. Arch Dermatol 2007; 143: 1493–1499.

2. Mak RK, Hundhausen C, Nestle FO. Progress in understanding the immunopathogenesis of psoriasis. Actas Dermosifiliogr 2009; 100 (Suppl 2): 2–13.

3. Capon F, Trembath RC, Barker JN. An update on the genetics of psoriasis. Dermatol Clin 2004; 22: 339–347.

4. Rahman P, Elder JT. Genetic epidemio­logy of psoriasis and psoriatic arthritis. Ann Rheum Dis 2005; 64 (Suppl 2): ii37–ii39, discussion ii40–ii41.

5. Nair RP, Stuart PE, Nistor I et al. Sequence and haplotype analysis supports HLA-C as the psoriasis susceptibility 1 gene. Am J Hum Genet 2006; 78: 827–851.

6. Trembath RC, Clough RL, Rosbotham JL et al. Identification of a major susceptibility locus on chromosome 6p and evidence for further disease loci revealed by a two stage genome-wide search in psoriasis. Hum Mol Genet 1997; 6: 813–820.

7. Gudjonsson JE, Karason A, Antonsdottir A et al. Psoriasis patients who are homozygous for the HLA-Cw*0602 allele have a 2.5-fold increased risk of developing psoriasis compared with Cw6 heterozygotes. Br J Dermatol 2003; 148: 233–235.

8. Jegasothy BV, Ackerman CD, Todo S et al. Tacrolimus (FK 506) – a new therapeutic agent for severe recalcitrant psoriasis. Arch Dermatol 1992; 128: 781–785.

9. Abrams JR, Lebwohl MG, Guzzo CA et al. CTLA4Ig-mediated blockade of T-cell costimulation in patients with psoriasis vulgaris. J Clin Invest 1999; 103: 1243–1252.

10. Conrad C, Boyman O, Tonel G et al. Alpha1beta1 integrin is crucial for accumulation of epidermal T cells and the development of psoriasis. Nat Med 2007; 13: 836–842.

11. Lowes MA, Bowcock AM, Krueger JG. Pathogenesis and therapy of psoriasis. Nature 2007; 445: 866–873.

12. Di Cesare A, Di Meglio P, Nestle FO. The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol 2009; 129: 1339–1350.

13. Zheng Y, Danilenko DM, Valdez P et al. Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature 2007; 445: 648–651.

14. Piskin G, Sylva-Steenland RM, Bos JD et al. In vitro and in situ expression of IL-23 by keratinocytes in healthy skin and psoriasis lesions: enhanced expression in psoriatic skin. J Immunol 2006; 176: 1908–1915.

15. Haider AS, Lowes MA, Suárez-Fariñas M et al. Identification of cellular pathways of „type 1“, Th17 T cells, and TNF- and inducible nitric oxide synthase-producing dendritic cells in autoimmune inflammation through pharmacogenomic study of cyclosporine A in psoriasis. J Immunol 2008: 180: 1913–1920.

16. Sugiyama H, Gyulai R, Toichi E et al. Dysfunctional blood and target tissue CD4+ CD25 high regulatory T cells in psoriasis: mechanism underlying unrestrained pathogenic effector T cell proliferation. J Immunol 2005; 174: 164–173.

17. Nestle FO, Conrad C, Tun-Kyi A et al. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med 2005; 202: 135–143.

18. Fry L. Psoriasis. Br J Dermatol 1988; 119: 445–461.

19. Baker BS, Laman JD, Powles A et al. Peptidoglycan and peptidoglycan-specific Th1 cells in psoriatic skin lesions. J Pathol 2006; 209: 174–181.

20. Johnston A, Gudjonsson JE, Sigmunds­dottir H et al. Peripheral blood T cell responses to keratin peptides that share sequences with streptococcal M proteins are largely restricted to skin-homing CD8(+) T cells. Clin Exp Immunol 2004; 138: 83–93.

21. Lande R, Gregorio J, Facchinetti V et al. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature 2007; 449: 564–569.

22. Henseler T, Christophers E. Disease concomitance in psoriasis. J Am Acad Dermatol 1995; 32: 982–986.

23. Vašků V. Komorbidity u psoriázy, nový pohled na staré onemocnění. Dermatol praxi 2009; 3: 63–66.

24. Gottlieb AB, Chao C, Dann F. Psoriasis comorbidities. J Dermatol Treat 2008; 19: 5–21.

25. Svačina Š. Metabolický syndrom a kožní onemocnění. Čas Lék Čes 2008; 147: 307–310.

26. Mrowietz U, Elder JT, Barker J. The importance of disease associations and concomitant therapy for the long-term management of psoriasis patients. Arch Dermatol Res 2006; 298: 309–319.

27. Benáková N, Štork J. Novinky v léčbě psoriázy biologiky a standardními systémovými léky. Čes Slov Derm 2008: 83: 191–202.

28. Benáková N, Štork J. Léčba psoriázy bio­logiky. Čes Slov Derm 2006: 81: 1–11.

29. Leonardi CL, Papp KA, Gordon KB et al. Efalizumab Study Group. Extended efalizumab therapy improves chronic plaque psoria­sis: Results from a randomize phase III trial. J Am Acad Dermatol 2005: 52: 425–433.

30. Boehncke WH, Brasie RA, Barker J et al. European Dermatology Expert Group. Recommendations for the use of etanercept in psoriasis: a European dermatology expert group consensus. J Eur Acad Dermatol Venereol 2006; 20: 988–998.

31. Papp KA, Tyring S, Lahfa M et al. A global phase III randomized controlled trial of etanercept in psoriasis: safety, efficacy, and effect of dose reduction. Br J Dermatol 2005; 152: 1304–1312.

32. Leonardi CL, Powers JL, Matbeson RT et al. Etanercept as monotherapy in patients with psoriasis. N Engl J Med 2003; 349: 2014–2022.

33. Paller AS, Siegfried EC, Langley RG et al. Etanercept Pediatric Psoriasis Study Group. Etanercept treatment for children and adolescents with plaque psoriasis. N Engl J Med 2008; 358: 241–251.

34. Gordon KB, Langley RG, Leonardi C et al. Clinical response to adalimumab treatment in patients with moderate to severe psoriasis: double-blind, randomized controlled trial and open label extension study. J Am Acad Dermatol 2006; 55: 598–606.

35. Sommer DM, Jenisch S, Suchan M et al. Increased prevalence of the metabolic syndrome in patients with moderate to severe psoriasis. Arch Dermatol Res 2006; 298: 321–328.

36. Dalaker M, Bonesrønning JH. Long--term maintenance treatment of moderate-to-severe plaque psoriasis with infliximab in comabination with methotrexate or azathio­prine in a retrospective cohort. J Eur Acad Dermatol Venereol 2009; 23: 277–282.

37. Benáková N. Ustekinumab v celkové léčbě psoriázy. Farmakoterapie 2009; 5: 249–356.

38. Lebwohl M, Yeilding N, Szapary P et al. Impact of weight on the efficacy and safety of ustekinumab in patients with moderate to severe psoriasis: Rationale for dosing recommendations. J Am Acad Dermatol 2010; 63: 571–579.

39. Leonardi CL, Kimball AB, Papp KA et al. PHOENIX 1 study investigators. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 2008; 371: 1665–1674.

40. Papp KA, Lebwohl M, Krueger GG et al. PHOENIX 2 study investigators. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet 2008; 371: 1675–1684.

41. Griffiths C, Strober B, van de Kerkhof PC et al. A phase 3, multicenter, randomized study comparing ustekinumab and etanercept for the treatment of moderate to severe psoriasis. Presented at: 2008 European Academy of Dermatology & Venereology (EADV), Paris, Poster FP1336. Ref Type: Conference Proceeding.

42. Langowski JL, Zhang X, Wu L et al. IL-23 promotes tumour incidence and growth. Nature 2006; 442: 461–465.

43. Fieschi C, Casanova JL. The role of interleukin-12 in human infectious diseases: only a faint signature. Eur J Immunol 2003; 33: 461–464.

44. Zhu Y, Hu C, Lu M et al. Population pharmacokinetic modeling of ustekinumab, a human monoclonal antibody targetin IL-12/23p40 in patients with moderate to severe plaque psoriasis. J Clin Pharmacol 2009; 49: 162–175.

45. Wu JJ. Interleukin-12, interleukin-23, and psoriasis: ABT-874 in clinical trials. J Am Acad Dermatol 2008; 58: 1083.

46. Strober AE, Crowley JJ, Yamauchi PS et al. Efficacy and safety results from a Phase III, randomized controlled trial comparing the safety and efficacy of bria­kinumab to etanercept and placebo in patients with moderate to severe chronic plaque psoriasis. Presented at Psoriasis 2010, Congress of the Psoriasis International Network, 1–4 July 2010, Paris, France.

47. Kinney MA, Yentzer BA, Feldman SR. IL-12/23 inhibitors and their potential benefits and risks for psoriasis patients. Psoriasis Forum 2010; 16: 16–20.

Labels
Diabetology Endocrinology Internal medicine

Article was published in

Internal Medicine

Issue 9

2011 Issue 9

Most read in this issue
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#