全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Biologics in Dermatology

DOI: 10.3390/ph6040557

Keywords: biologics, biological response modifiers, immunopathogenesis, dermatology, skin disease

Full-Text   Cite this paper   Add to My Lib

Abstract:

Skin and subcutaneous diseases affect millions of people worldwide, causing significant morbidity. Biologics are becoming increasingly useful for the treatment of many skin diseases, particularly as alternatives for patients who have failed to tolerate or respond to conventional systemic therapies. Biological therapies provide a targeted approach to treatment through interaction with specific components of the underlying immune and inflammatory disease processes. This review article examines the increasing evidence base for biologics in dermatology, including well-established treatments and novel agents.

References

[1]  Parisi, R.; Griffiths, C.E.M.; Ashcroft, D.M. Systematic review of the incidence and prevalence of psoriasis. Brit. J. Dermatol. 2011, 165, e5, doi:10.1111/j.1365-2133.2011.10217.x.
[2]  Parisi, R.; Symmons, D.P.; Griffiths, C.E.; Ashcroft, D.M. Global Epidemiology of Psoriasis: A Systematic Review of Incidence and Prevalence. J. Invest. Dermatol. 2013, 133, 377–385, doi:10.1038/jid.2012.339.
[3]  Zachariae, H. Prevalence of joint disease in patients with psoriasis: Implications for therapy. Am. J. Clin. Dermatol. 2003, 4, 441–447, doi:10.2165/00128071-200304070-00001.
[4]  Ibrahim, G.; Waxman, R.; Helliwell, P.S. The prevalence of psoriatic arthritis in people with psoriasis. Arthritis. Rheum. 2009, 61, 1373–1378, doi:10.1002/art.24608.
[5]  Lowes, M.A.; Kikuchi, T.; Fuentes-Duculan, J.; Cardinale, I.; Zaba, L.C.; Haider, A.S.; Bowman, E.P.; Krueger, J.G. Psoriasis vulgaris lesions contain discrete populations of Th1 and Th17 T cells. J. Invest. Dermatol. 2008, 128, 1207–1211, doi:10.1038/sj.jid.5701213.
[6]  Di Meglio, P.; Perera, G.K.; Nestle, F.O. The multitasking organ: Recent insights into skin immune function. Immunity 2011, 35, 857–869, doi:10.1016/j.immuni.2011.12.003.
[7]  Woolacott, N.; Bravo, V.Y.; Hawkins, N.; Kainth, A.; Khadjesari, Z.; Misso, K.; Light, K.; Asseburg, C.; Palmer, S.; Claxton, K.; Bruce, I.; Sculpher, M.; Riemsma, R. Etanercept and infliximab for the treatment of psoriatic arthritis: A systematic review and economic evaluation. Health Technol. Assess. 2006, 10, 1–239.
[8]  Mease, P.J.; Antoni, C.E. Psoriatic arthritis treatment: Biological response modifiers. Ann. Rheum Dis. 2005, 64 (Suppl. 2), 78–82.
[9]  Gottlieb, A.B.; Langley, R.G.; Strober, B.E.; Papp, K.A.; Klekotka, P.; Creamer, K.; Thompson, E.H.; Hooper, M.; Kricorian, G. A randomized, double-blind, placebo-controlled study to evaluate the addition of methotrexate to etanercept in patients with moderate to severe plaque psoriasis. Br. J. Dermatol. 2012, 167, 649–657, doi:10.1111/j.1365-2133.2012.11015.x.
[10]  Strober, B.E.; Sobell, J.M.; Duffin, K.C.; Bao, Y.; Guérin, A.; Yang, H.; Goldblum, O.; Okun, M.M.; Mulani, P.M. Sleep quality and other patient-reported outcomes improve after patients with psoriasis with suboptimal response to other systemic therapies are switched to adalimumab: Results from PROGRESS, an open-label Phase IIIB trial. Br. J. Dermatol. 2012, 167, 1374–1381, doi:10.1111/bjd.12000.
[11]  Reich, K.; Ortonne, J.P.; Gottlieb, A.B.; Terpstra, I.J.; Coteur, G.; Tasset, C.; Mease, P. Successful treatment of moderate to severe plaque psoriasis with the PEGylated Fab′ certolizumab pegol: Results of a phase II randomized, placebo-controlled trial with a re-treatment extension. Br. J. Dermatol. 2012, 167, 180–190, doi:10.1111/j.1365-2133.2012.10941.x.
[12]  Nesbitt, A.; Fossati, G.; Bergin, M.; Stephens, P.; Stephens, S.; Foulkes, R.; Brown, D.; Robinson, M.; Bourne, T. Mechanism of action of certolizumab pegol (CDP870): In vitro comparison with other anti-tumor necrosis factor alpha agents. Inflamm. Bowel Dis. 2007, 13, 1323–1332, doi:10.1002/ibd.20225.
[13]  Kanakaraj, P.; Puffer, B.A.; Yao, X.T.; Kankanala, S.; Boyd, E.; Shah, R.R.; Wang, G.; Patel, D.; Krishnamurthy, R.; Kaithamana, S.; et al. Simultaneous targeting of TNF and Ang2 with a novel bispecific antibody enhances efficacy in an in vivo model of arthritis. MAbs 2012, 4, 600–613, doi:10.4161/mabs.21227.
[14]  Jung, K.; Lee, D.; Lim, H.S.; Lee, S.I.; Kim, Y.J.; Lee, G.M.; Kim, S.C.; Koh, G.Y. Double anti-angiogenic and anti-inflammatory protein Valpha targeting VEGF-A and TNF-alpha in retinopathy and psoriasis. J. Biol. Chem. 2011, 286, 14410–14418.
[15]  Crawshaw, A.A.; Griffiths, C.E.; Young, H.S. Investigational VEGF antagonists for psoriasis. Expert Opin. Investig. Drugs 2012, 21, 33–43, doi:10.1517/13543784.2012.636351.
[16]  Zheng, Y.; Danilenko, D.M.; Valdez, P.; Kasman, I.; Eastham-Anderson, J.; Wu, J.; Ouyang, W. Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature 2007, 445, 648–651, doi:10.1038/nature05505.
[17]  Griffiths, C.E.; Strober, B.E.; van de Kerkhof, P.; Ho, V.; Fidelus-Gort, R.; Yeilding, N.; Guzzo, C.; Xia, Y.; Zhou, B.; Li, S.; et al. ACCEPT Study group. comparison of ustekinumab and etanercept for moderate-to-severe psoriasis. N Engl. J. Med. 2010, 362, 118–128, doi:10.1056/NEJMoa0810652.
[18]  Reich, K.; Papp, K.A.; Griffiths, C.E.; Szapary, P.O.; Yeilding, N.; Wasfi, Y.; Ott, E.; Hsu, M.C.; Lebwohl, M.; Gordon, K.B. PHOENIX 1, PHOENIX 2, and ACCEPT investigators. An update on the long-term safety experience of ustekinumab: Results from the psoriasis clinical development program with up to four years of follow-up. J. Drugs Dermatol. 2012, 11, 300–312.
[19]  Papp, K.A.; Leonardi, C.; Menter, A.; Ortonne, J.P.; Krueger, J.G.; Kricorian, G.; Aras, G.; Li, J.; Russell, C.B.; Thompson, E.H.; Baumgartner, S. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N Engl. J. Med. 2012, 366, 1181–1189, doi:10.1056/NEJMoa1109017.
[20]  Leonardi, C.; Matheson, R.; Zachariae, C.; Cameron, G.; Li, L.; Edson-Heredia, E.; Braun, D.; Banerjee, S. Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N Engl. J. Med. 2012, 366, 1190–1199, doi:10.1056/NEJMoa1109997.
[21]  Papp, K.A.; Menter, A.; Strober, B.; Langley, R.G.; Buonanno, M.; Wolk, R.; Gupta, P.; Krishnaswami, S.; Tan, H.; Harness, J.A. Efficacy and safety of tofacitinib, an oral Janus kinase inhibitor, in the treatment of psoriasis: A Phase 2b randomized placebo-controlled dose-ranging study. Br. J. Dermatol. 2012, 167, 668–677, doi:10.1111/j.1365-2133.2012.11168.x.
[22]  Skvara, H.; Dawid, M.; Kleyn, E.; Wolff, B.; Meingassner, J.G.; Knight, H.; Dumortier, T.; Kopp, T.; Fallahi, N.; Stary, G.; et al. The PKC inhibitor AEB071 may be a therapeutic option for psoriasis. J. Clin. Invest. 2008, 118, 3151–3159, doi:10.1172/JCI35636.
[23]  Schafer, P.H.; Parton, A.; Gandhi, A.K.; Capone, L.; Adams, M.; Wu, L.; Bartlett, J.B.; Loveland, M.A.; Gilhar, A.; Cheung, Y.F.; et al. Apremilast, a cAMP phosphodiesterase-4 inhibitor, demonstrates anti-inflammatory activity in vitro and in a model of psoriasis. Br. J. Pharmacol. 2010, 159, 842–855, doi:10.1111/j.1476-5381.2009.00559.x.
[24]  Papp, K.; Cather, J.C.; Rosoph, L.; Sofen, H.; Langley, R.G.; Matheson, R.T.; Hu, C.; Day, R.M. Efficacy of apremilast in the treatment of moderate to severe psoriasis: A randomised controlled trial. Lancet 2012, 380, 738–746, doi:10.1016/S0140-6736(12)60642-4.
[25]  Horváth, B.; Huizinga, J.; Pas, H.H.; Mulder, A.B.; Jonkman, M.F. Low-dose rituximab is effective in pemphigus. Br. J. Dermatol. 2012, 166, 405–412, doi:10.1111/j.1365-2133.2011.10663.x.
[26]  Leshem, Y.A.; Hodak, E.; David, M.; Anhalt, G.J.; Mimouni, D. Successful treatment of pemphigus with biweekly 1-g infusions of rituximab: A retrospective study of 47 patients. J. Am. Acad. Dermatol. 2013, 68, 404–411, doi:10.1016/j.jaad.2012.08.010.
[27]  Kasperkiewicz, M.; Shimanovich, I.; Meier, M.; Schumacher, N.; Westermann, L.; Kramer, J.; Zillikens, D.; Schmidt, E. Treatment of severe pemphigus with a combination of immunoadsorption, rituximab, pulsed dexamethasone and azathioprine/mycophenolate mofetil: A pilot study of 23 patients. Br. J. Dermatol. 2012, 166, 154–160, doi:10.1111/j.1365-2133.2011.10585.x.
[28]  Jacobi, A.; Shuler, G.; Hertl, M. Rapid control of therapy-refractory pemphigus vulgaris by treatment with the tumour necrosis factor-alpha inhibitor infliximab. Br. J. Dermatol. 2005, 153, 448–449, doi:10.1111/j.1365-2133.2005.06744.x.
[29]  Berookhim, B.; Fischer, H.D.; Weinberg, J.M. Treatment of recalcitrant pemphigus vulgaris with the tumor necrosis factor alpha antagonist etanercept. Cutis 2004, 74, 245–247.
[30]  Giamarellos-Bourboulis, E.J.; Antonopoulou, A.; Petropoulou, C.; Mouktaroudi, M.; Spyridaki, E.; Baziaka, F.; Pelekanou, A.; Giamarellou, H.; Stavrianeas, N.G. Altered innate and adaptive immune responses in patients with hidradenitis suppurativa. Br. J. Dermatol. 2007, 156, 51–56, doi:10.1111/j.1365-2133.2006.07556.x.
[31]  Van der Zee, H.H.; de Ruiter, L.; van den Broecke, D.G.; Dik, W.A.; Laman, J.D.; Prens, E.P. Elevated levels of tumour necrosis factor (TNF)-α, interleukin (IL)-1β and IL-10 in hidradenitis suppurativa skin: A rationale for targeting TNF-α and IL-1β. Br. J. Dermatol. 2011, 164, 1292–1298, doi:10.1111/j.1365-2133.2011.10254.x.
[32]  Kimball, A.B.; Kerdel, F.; Adams, D.; Mrowietz, U.; Gelfand, J.M.; Gniadecki, R.; Prens, E.P.; Schlessinger, J.; Zouboulis, C.C.; van der Zee, H.H.; et al. Adalimumab for the treatment of moderate to severe hidradenitis suppurativa: A parallel randomized trial. Ann. Intern. Med. 2012, 157, 846–855, doi:10.7326/0003-4819-157-12-201212180-00004.
[33]  Sotiriou, E.; Goussi, C.; Lallas, A.; Chovarda, E.; Apalla, Z.; Lazaridou, E.; Ioannides, D. A prospective open-label clinical trial of efficacy of the every week administration of adalimumab in the treatment of hidradenitis suppurativa. J. Drugs Dermatol. 2012, 11 (Suppl. 5), S15–S20.
[34]  Van Rappard, D.C.; Limpens, J.; Mekkes, J.R. The off-label treatment of severe hidradenitis suppurativa with TNF-α inhibitors: A systematic review. J. Dermatolog. Treat. 2012, 23, 22397574.
[35]  Van Rappard, D.C.; Leenarts, M.F.; Meijerink-van't Oost, L.; Mekkes, J.R. Comparing treatment outcome of infliximab and adalimumab in patients with severe hidradenitis suppurativa. J. Dermatolog. Treat. 2012, 23, 284–289, doi:10.3109/09546634.2011.571657.
[36]  Schlapbach, C.; H?nni, T.; Yawalkar, N.; Hunger, R.E. Expression of the IL-23/Th17 pathway in lesions of hidradenitis suppurativa. J. Am. Acad. Dermatol. 2011, 65, 790–798, doi:10.1016/j.jaad.2010.07.010.
[37]  Sharon, V.R.; Garcia, M.S.; Bagheri, S.; Goodarzi, H.; Yang, C.; Ono, Y.; Maverakis, E. Management of recalcitrant hidradenitis suppurativa with ustekinumab. Acta. Derm. Venereol. 2012, 92, 320–321, doi:10.2340/00015555-1229.
[38]  Groves, R.W.; Allen, M.H.; Ross, E.L.; Barker, J.N.; MacDonald, D.M. Tumour necrosis factor alpha is pro-inflammatory in normal human skin and modulates cutaneous adhesion molecule expression. Br. J. Dermatol. 1995, 132, 345–352, doi:10.1111/j.1365-2133.1995.tb08666.x.
[39]  Craig, F.F.; Thomas, K.S.; Mitchell, E.J.; Williams, H.C.; Norrie, J.; Mason, J.M.; Ormerod, A.D. UK Dermatology Clinical Trials Network’s STOP GAP trial (a multicentre trial of prednisolone versus ciclosporin for pyoderma gangrenosum): Protocol for a randomised controlled trial. Trials 2012, 13, 51, doi:10.1186/1745-6215-13-51.
[40]  Kaufman, I.; Caspi, D.; Yeshurun, D.; Dotan, I.; Yaron, M.; Elkayam, O. The effect of infliximab on extraintestinal manifestations of Crohn’s disease. Rheumatol. Int. 2005, 25, 406–410, doi:10.1007/s00296-004-0467-8.
[41]  Mooij, J.E.; van Rappard, D.C.; Mekkes, J.R. Six patients with pyoderma gangrenosum successfully treated with infliximab. Int. J. Dermatol. 2012, doi:10.1111/j.1365-4632.2011.05201.x.
[42]  Guenova, E.; Teske, A.; Fehrenbacher, B.; Hoerber, S.; Adamczyk, A.; Schaller, M.; Hoetzenecker, W.; Biedermann, T. Interleukin 23 expression in pyoderma gangrenosum and targeted therapy with ustekinumab. Arch. Dermatol. 2011, 147, 1203–1205, doi:10.1001/archdermatol.2011.168.
[43]  Erdmann, F.; Lortet-Tieulent, J.; Schüz, J.; Zeeb, H.; Greinert, R.; Breitbart, E.W.; Bray, F. International trends in the incidence of malignant melanoma 1953–2008—Are recent generations at higher or lower risk? Int. J. Cancer 2013, 132, 385–400.
[44]  Tarhini, A.A.; Gogas, H.; Kirkwood, J.M. IFN-α in the treatment of melanoma. J. Immunol. 2012, 189, 3789–3793, doi:10.4049/jimmunol.1290060.
[45]  Kirkwood, J.M.; Manola, J.; Ibrahim, J.; Sondak, V.; Ernstoff, M.S.; Rao, U. A pooled analysis of eastern cooperative oncology group and intergroup trials of adjuvant high-dose interferon for melanoma. Clin. Cancer Res. 2004, 10, 1670–1677, doi:10.1158/1078-0432.CCR-1103-3.
[46]  Eggermont, A.M.; Suciu, S.; Testori, A.; Santinami, M.; Kruit, W.H.; Marsden, J.; Punt, C.J.; Salès, F.; Dummer, R.; Robert, C.; et al. Long-term results of the randomized phase III trial EORTC 18991 of adjuvant therapy with pegylated interferon alfa-2b versus observation in resected stage III melanoma. J. Clin. Oncol. 2012, 30, 3810–3818, doi:10.1200/JCO.2011.41.3799.
[47]  Atkins, M.B.; Lotze, M.T.; Dutcher, J.P.; Fisher, R.I.; Weiss, G.; Margolin, K.; Abrams, J.; Sznol, M.; Parkinson, D.; Hawkins, M.; et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: Analysis of 270 patients treated between 1985 and 1993. J. Clin. Oncol. 1999, 17, 2105–2116.
[48]  Dudley, M.E.; Yang, J.C.; Sherry, R.; Hughes, M.S.; Royal, R.; Kammula, U.; Robbins, P.F.; Huang, J.; Citrin, D.E.; Leitman, S.F.; et al. Adoptive cell therapy for patients with metastatic melanoma: Evaluation of intensive myeloablative chemoradiation preparative regimens. J. Clin. Oncol. 2008, 26, 5233–5239, doi:10.1200/JCO.2008.16.5449.
[49]  Ellebaek, E.; Iversen, T.Z.; Junker, N.; Donia, M.; Engell-Noerregaard, L.; Met, O.; H?lmich, L.R.; Andersen, R.S.; Hadrup, S.R.; Andersen, M.H.; et al. Adoptive cell therapy with autologous tumor infiltrating lymphocytes and low-dose Interleukin-2 in metastatic melanoma patients. J. Transl. Med. 2012, 10, 169, doi:10.1186/1479-5876-10-169.
[50]  Weber, J. Overcoming immunologic tolerance to melanoma: Targeting CTLA-4 with ipilimumab (MDX-010). Oncologist 2008, 13 (Suppl. 4), 16–25, doi:10.1634/theoncologist.13-S4-16.
[51]  Robert, C.; Thomas, L.; Bondarenko, I.; O’Day, S.; Weber, J.; Garbe, C.; Lebbe, C.; Baurain, J.F.; Testori, A.; Grob, J.J.; et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl. J. Med. 2011, 364, 2517–2526, doi:10.1056/NEJMoa1104621.
[52]  Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl. J. Med. 2010, 363, 711–723, doi:10.1056/NEJMoa1003466.
[53]  Ribas, A.; Chesney, J.A.; Gordon, M.S.; Abernethy, A.P.; Logan, T.F.; Lawson, D.H.; Chmielowksi, B.; Glaspy, J.A.; Lewis, K.; Huang, B.; et al. Safety profile and pharmacokinetic analyses of the anti-CTLA4 antibody tremelimumab administered as a one hour infusion. J. Transl. Med. 2012, 10, 236, doi:10.1186/1479-5876-10-236.
[54]  Kirkwood, J.M.; Lorigan, P.; Hersey, P.; Hauschild, A.; Robert, C.; McDermott, D.; Marshall, M.A.; Gomez-Navarro, J.; Liang, J.Q.; Bulanhagui, C.A. Phase II trial of tremelimumab (CP-675,206) in patients with advanced refractory or relapsed melanoma. Clin. Cancer Res. 2010, 16, 1042–1048, doi:10.1158/1078-0432.CCR-09-2033.
[55]  Camacho, L.H.; Antonia, S.; Sosman, J.; Kirkwood, J.M.; Gajewski, T.F.; Redman, B.; Pavlov, D.; Bulanhagui, C.; Bozon, V.A.; Gomez-Navarro, J.; et al. Phase I/II trial of tremelimumab in patients with metastatic melanoma. J. Clin. Oncol. 2009, 27, 1075–1081, doi:10.1200/JCO.2008.19.2435.
[56]  Ribas, A.; Hauschild, A.; Kefford, R.; Punt, C.J.; Haanen, J.B.; Marmol, M.; Garbe, C.; Gomez-Navarro, J; Pavlov, D.; Marshall, M. Phase III, open-label, randomized, comparative study of tremelimumab (CP-675,206) and chemotherapy (temozolomide [TMZ] or dacarbazine [DTIC]) in patients with advanced melanoma [oral]. J. Clin. Oncol. 2008, 20 (Suppl. 15), BA9011.
[57]  Sinha, R.; Edmonds, K.; Newton-Bishop, J.A.; Gore, M.E.; Larkin, J.; Fearfield, L. Cutaneous adverse events associated with vemurafenib in patients with metastatic melanoma: Practical advice on diagnosis, prevention and management of the main treatment-related skin toxicities. Br. J. Dermatol. 2012, 167, 987–994, doi:10.1111/bjd.12010.
[58]  Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl. J. Med. 2011, 364, 2507–2516.
[59]  Young, K.; Minchom, A.; Larkin, J. BRIM-1, -2 and -3 trials: Improved survival with vemurafenib in metastatic melanoma patients with a BRAF(V600E) mutation. Future Oncol. 2012, 8, 499–507, doi:10.2217/fon.12.43.
[60]  Hauschild, A.; Grob, J.J.; Demidov, L.V.; Jouary, T.; Gutzmer, R.; Millward, M.; Rutkowski, P.; Blank, C.U.; Miller, W.H., Jr.; Kaempgen, E.; et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012, 380, 358–365.
[61]  Anforth, R.M.; Blumetti, T.C.; Kefford, R.F.; Sharma, R.; Scolyer, R.A.; Kossard, S.; Long, G.V.; Fernandez-Pe?as, P. Cutaneous manifestations of dabrafenib (GSK2118436): A selective inhibitor of mutant BRAF in patients with metastatic melanoma. Br. J. Dermatol. 2012, 167, 1153–1160, doi:10.1111/j.1365-2133.2012.11155.x.
[62]  Klein, O.; Clements, A.; Menzies, A.M.; O’Toole, S.; Kefford, R.F.; Long, G.V. BRAF inhibitor activity in V600R metastatic melanoma. Eur. J. Cancer 2013, 49, 1073–1079, doi:10.1016/j.ejca.2012.11.004.
[63]  Hatzivassiliou, G.; Song, K.; Yen, I.; Brandhuber, B.J.; Anderson, D.J.; Alvarado, R.; Ludlam, M.J.; Stokoe, D.; Gloor, S.L.; Vigers, G.; et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature 2010, 464, 431–435.
[64]  Halaban, R.; Zhang, W.; Bacchiocchi, A.; Cheng, E.; Parisi, F.; Ariyan, S.; Krauthammer, M.; McCusker, J.P.; Kluger, Y.; Sznol, M. PLX4032, a selective BRAF(V600E) kinase inhibitor, activates the ERK pathway and enhances cell migration and proliferation of BRAF melanoma cells. Pigment. Cell. Melanoma Res. 2010, 23, 190–200, doi:10.1111/j.1755-148X.2010.00685.x.
[65]  Flaherty, K.T.; Robert, C.; Hersey, P.; Nathan, P.; Garbe, C.; Milhem, M.; Demidov, L.V.; Hassel, J.C.; Rutkowski, P.; Mohr, P.; Metric study group. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl. J. Med. 2012, 367, 107–114, doi:10.1056/NEJMoa1203421.
[66]  Flaherty, K.T.; Infante, J.R.; Daud, A.; Gonzalez, R.; Kefford, R.F.; Sosman, J.; Hamid, O.; Schuchter, L.; Cebon, J.; Ibrahim, N.; et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl. J. Med. 2012, 367, 1694–1703, doi:10.1056/NEJMoa1210093.
[67]  Mangana, J.; Levesque, M.P.; Karpova, M.B.; Dummer, R. Sorafenib in melanoma. Expert Opin. Investig. Drugs 2012, 21, 557–568, doi:10.1517/13543784.2012.665872.
[68]  Iyer, R.; Fetterly, G.; Lugade, A.; Thanavala, Y. Sorafenib: A clinical and pharmacologic review. Expert Opin. Pharmacother. 2010, 11, 1943–1955, doi:10.1517/14656566.2010.496453.
[69]  Hauschild, A.; Agarwala, S.S.; Trefzer, U.; Hogg, D.; Robert, C.; Hersey, P.; Eggermont, A.; Grabbe, S.; Gonzalez, R.; Gille, J.; et al. Results of a phase III, randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel as second-line treatment in patients with unresectable stage III or stage IV melanoma. J. Clin. Oncol. 2009, 27, 2823–2830, doi:10.1200/JCO.2007.15.7636.
[70]  Egberts, F.; Kahler, K.C.; Livingstone, E.; Hauschild, A. Metastatic melanoma: Scientific rationale for sorafenib treatment and clinical results. Onkologie 2008, 31, 398–403, doi:10.1159/000137714.
[71]  McDermott, D.F.; Sosman, J.A.; Gonzalez, R.; Hodi, F.S.; Linette, G.P.; Richards, J.; Jakub, J.W.; Beeram, M.; Tarantolo, S.; Agarwala, S.; et al. Double-blind randomized phase II study of the combination of sorafenib and dacarbazine in patients with advanced melanoma: A report from the 11715 Study Group. J. Clin. Oncol. 2008, 26, 2178–2185, doi:10.1200/JCO.2007.14.8288.
[72]  Amaravadi, R.K.; Schuchter, L.M.; McDermott, D.F.; Kramer, A.; Giles, L.; Gramlich, K.; Carberry, M.; Troxel, A.B.; Letrero, R.; Nathanson, K.L.; et al. Phase II Trial of Temozolomide and Sorafenib in Advanced Melanoma Patients with or without Brain Metastases. Clin. Cancer Res. 2009, 15, 7711–7718, doi:10.1158/1078-0432.CCR-09-2074.
[73]  Ferguson, J.; Arozarena, I.; Ehrhardt, M.; Wellbrock, C. Combination of MEK and SRC inhibition suppresses melanoma cell growth and invasion. Oncogene 2013, 32, 86–96, doi:10.1038/onc.2012.25.
[74]  Gangadhar, T.C.; Clark, J.I.; Karrison, T.; Gajewski, T.F. Phase II study of the Src kinase inhibitor saracatinib (AZD0530) in metastatic melanoma. Invest. New Drugs 2012, 30, 23151808.
[75]  Agata, Y.; Kawasaki, A.; Nishimura, H.; Ishida, Y.; Tsubata, T.; Yagita, H.; Honjo, T. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int. Immunol. 1996, 8, 765–772, doi:10.1093/intimm/8.5.765.
[76]  Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704, doi:10.1146/annurev.immunol.26.021607.090331.
[77]  Okazaki, T.; Honjo, T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006, 27, 195–201, doi:10.1016/j.it.2006.02.001.
[78]  Brahmer, J.R.; Drake, C.G.; Wollner, I.; Powderly, J.D.; Picus, J.; Sharfman, W.H.; Stankevich, E.; Pons, A.; Salay, T.M.; McMiller, T.L.; et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: Safety, clinical activity, pharmacodynamics, and immunologic correlates. J. Clin. Oncol. 2010, 28, 3167–3175, doi:10.1200/JCO.2009.26.7609.
[79]  Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl. J. Med. 2012, 366, 2455–2465, doi:10.1056/NEJMoa1200694.
[80]  Bordignon, M.; Rottoli, P.; Agostini, C.; Alaibac, M. Adaptive immune responses in primary cutaneous sarcoidosis. Clin. Dev. Immunol. 2011, 2011, 235142.
[81]  Facco, M.; Cabrelle, A.; Teramo, A.; Olivieri, V.; Gnoato, M.; Teolato, S.; Ave, E.; Gattazzo, C.; Fadini, G.P.; Calabrese, F.; Semenzato, G.; Agostini, C. Sarcoidosis is a Th1/Th17 multisystem disorder. Thorax 2011, 66, 144–150, doi:10.1136/thx.2010.140319.
[82]  Marques, L.J.; Zheng, L.; Poulakis, N.; Guzman, J.; Costabel, U. Pentoxifylline inhibits TNF-alpha production from human alveolar macrophages. Am. J. Respir. Crit. Care Med. 1999, 159, 508–511, doi:10.1164/ajrccm.159.2.9804085.
[83]  Tuchinda, P.; Bremmer, M.; Gaspari, A.A. A case series of refractory cutaneous sarcoidosis successfully treated with infliximab. Dermatol. Ther. (Heidelb) 2012, 2, 11, doi:10.1007/s13555-012-0011-9.
[84]  Wanat, K.A.; Rosenbach, M. Case series demonstrating improvement in chronic cutaneous sarcoidosis following treatment with TNF inhibitors. Arch. Dermatol. 2012, 148, 1097–1100, doi:10.1001/archdermatol.2012.1322.
[85]  Sené, T.; Juillard, C.; Rybojad, M.; Cordoliani, F.; Lebbé, C.; Morel, P.; Tazi, A.; Guibal, F. Infliximab as a steroid-sparing agent in refractory cutaneous sarcoidosis: Single-center retrospective study of 9 patients. J. Am. Acad. Dermatol. 2012, 66, 328–332, doi:10.1016/j.jaad.2011.05.040.
[86]  Tuchinda, C.; Wong, H.K. Etanercept for chronic progressive cutaneous sarcoidosis. J. Drugs Dermatol. 2006, 5, 538–540.
[87]  Khanna, D.; Liebling, M.R.; Louie, J.S. Etanercept ameliorates sarcoidosis arthritis and skin disease. J. Rheumatol. 2003, 30, 1864–1867.
[88]  Gregoriou, S.; Chiolou, Z.; Stefanaki, C.; Zakopoulou, N.; Rigopoulos, D.; Kontochristopoulos, G. Pityriasis rubra pilaris presenting with an abnormal autoimmune profile: Two case reports. J. Med. Case Rep. 2009, 3, 123, doi:10.1186/1752-1947-3-123.
[89]  Magro, C.M.; Crowson, A.N. The clinical and histomorphological features of pityriasis rubra pilaris. A comparative analysis with psoriasis. J. Cutan. Pathol. 1997, 24, 416–424, doi:10.1111/j.1600-0560.1997.tb00816.x.
[90]  Zhang, Y.H.; Zhou, Y.; Ball, N.; Su, M.W.; Xu, J.H.; Zheng, Z.Z. Type I pityriasis rubra pilaris: Upregulation of tumor necrosis factor alpha and response to adalimumab therapy. J. Cutan. Med. Surg. 2010, 14, 185–188.
[91]  Müller, H.; Gattringer, C.; Zelger, B.; H?pfl, R.; Eisendle, K. Infliximab monotherapy as first-line treatment for adult-onset pityriasis rubra pilaris: Case report and review of the literature on biologic therapy. J. Am. Acad. Dermatol. 2008, 59 (Suppl. 5), S65–S70.
[92]  Garcovich, S.; Di Giampetruzzi, A.R.; Antonelli, G.; Garcovich, A.; Didona, B. Treatment of refractory adult-onset pityriasis rubra pilaris with TNF-alpha antagonists: A case series. J. Eur. Acad. Dermatol. Venereol. 2010, 24, 881–884.
[93]  Ruiz Villaverde, R.; Sánchez Cano, D. Successful treatment of type 1 pityriasis rubra pilaris with ustekinumab therapy. Eur. J. Dermatol. 2010, 20, 630–631.
[94]  Wohlrab, J.; Kreft, B. Treatment of pityriasis rubra pilaris with ustekinumab. Br. J. Dermatol. 2010, 163, 655–656, doi:10.1111/j.1365-2133.2010.09855.x.
[95]  Shellow, W.V.; Edwards, J.E.; Koo, J.Y. Profile of alopecia areata: A questionnaire analysis of patient and family. Int. J. Dermatol. 1992, 31, 186–189, doi:10.1111/j.1365-4362.1992.tb03932.x.
[96]  Br?cker, E.B.; Echternacht-Happle, K.; Hamm, H.; Happle, R. Abnormal expression of class I and class II major histocompatibility antigens in alopecia areata: Modulation by topical immunotherapy. J. Invest. Dermatol. 1987, 88, 564–568.
[97]  Mcdonagh, A.J.; Snowden, J.A.; Stierle, C.; Elliott, K.; Messenger, A.G. HLA and ICAM-1 expression in alopecia areata in vivo and in vitro: The role of cytokines. Br. J. Dermatol. 1993, 129, 250–256.
[98]  McElwee, K.J.; Hoffmann, R.; Freyschmidt-Paul, P.; Wenzel, E.; Kissling, S.; Sundberg, J.P.; Z?ller, M. Resistance to alopecia areata in C3H/HeJ mice is associated with increased expression of regulatory cytokines and a failure to recruit CD4+ and CD8+ cells. J. Invest. Dermatol. 2002, 119, 1426–1433, doi:10.1046/j.1523-1747.2002.19620.x.
[99]  Le Bidre, E.; Chaby, G.; Martin, L.; Perrussel, M.; Sassolas, B.; Sigal, M.L.; Kaassis, C.; Lespessailles, E.; Nseir, A.; Estève, E. Alopecia areata during anti-TNF alpha therapy: Nine cases. Ann. Dermatol. Venereol. 2011, 138, 285–293, doi:10.1016/j.annder.2011.01.047.
[100]  Heffernan, M.P.; Hurley, M.Y.; Martin, K.S.; Smith, D.I.; Anadkat, M.J. Alefacept for alopecia areata. Arch. Dermatol. 2005, 141, 1513–1516, doi:10.1001/archderm.141.12.1513.
[101]  Strober, B.E.; Menon, K.; McMichael, A.; Hordinsky, M.; Krueger, G.; Panko, J.; Siu, K.; Lustgarten, J.L.; Ross, E.K.; Shapiro, J. Alefacept for severe alopecia areata: A randomized, double-blind, placebo-controlled study. Arch. Dermatol. 2009, 145, 1262–1266, doi:10.1001/archdermatol.2009.264.
[102]  Kaelin, U.; Hassan, A.S.; Braathen, L.R.; Yawalkar, N. Treatment of alopecia areata partim universalis with efalizumab. J. Am. Acad. Dermatol. 2006, 55, 529–532, doi:10.1016/j.jaad.2006.05.062.
[103]  Kolde, G.; Meffert, H.; Rowe, E. Successful treatment of alopecia areata with efalizumab. J. Eur. Acad. Dermatol. Venereol. 2008, 22, 1519–1520, doi:10.1111/j.1468-3083.2008.02665.x.
[104]  Price, V.H.; Hordinsky, M.K.; Olsen, E.A.; Roberts, J.L.; Siegfried, E.C.; Rafal, E.S.; Korman, N.J.; Altrabulsi, B.; Leung, H.M.; Garovoy, M.R.; et al. Subcutaneous efalizumab is not effective in the treatment of alopecia areata. J. Am. Acad. Dermatol. 2008, 58, 395–402, doi:10.1016/j.jaad.2007.10.645.
[105]  Confino-Cohen, R.; Chodick, G.; Shalev, V.; Leshno, M.; Kimhi, O.; Goldberg, A. Chronic urticaria and autoimmunity: Associations found in a large population study. J. Allergy Clin. Immunol. 2012, 129, 1307–1313.
[106]  Kaplan, A.P. Treatment of chronic spontaneous urticaria. Allergy Asthma Immunol. Res. 2012, 4, 326–331, doi:10.4168/aair.2012.4.6.326.
[107]  Kaplan, A.P.; Joseph, K.; Maykut, R.J.; Geba, G.P.; Zeldin, R.K. Treatment of chronic autoimmune urticaria with omalizumab. J. Allergy Clin. Immunol. 2008, 122, 569–573, doi:10.1016/j.jaci.2008.07.006.
[108]  Saini, S.; Rosen, K.E.; Hsieh, H.J.; Wong, D.A.; Conner, E.; Kaplan, A.; Spector, S.; Maurer, M. A randomized, placebo-controlled, dose-ranging study of single-dose omalizumab in patients with H1-antihistamine-refractory chronic idiopathic urticaria. J. Allergy Clin. Immunol. 2011, 128, 567–573, doi:10.1016/j.jaci.2011.06.010.
[109]  Ferrer, M.; Gamboa, P.; Sanz, M.L.; Goikoetxea, M.J.; Cabrera-Freitag, P.; Javaloyes, G.; Berroa, F.; Kaplan, A.P. Omalizumab is effective in nonautoimmune urticaria. J. Allergy Clin. Immunol. 2011, 127, 1300–1302, doi:10.1016/j.jaci.2010.12.1085.
[110]  Ivyanskiy, I.; Sand, C.; Thomsen, S.F. Omalizumab for chronic urticaria: A case series and overview of the literature. Case Rep. Dermatol. 2012, 4, 19–26, doi:10.1159/000336205.
[111]  NICE clinical guideline 57. Atopic eczema in children: Management of atopic eczema in children from birth up to the age of 12 years. Issued December 2007. Available online: http://www.nice.org.uk/CG057/ (accessed on 10 April 2013).
[112]  Koga, C.; Kabashima, K.; Shiraishi, N.; Kobayashi, M.; Tokura, Y. Possible pathogenic role of Th17 cells for atopic dermatitis. J. Invest. Dermatol. 2008, 128, 2625–2630, doi:10.1038/jid.2008.111.
[113]  Hanifin, J.M.; Schneider, L.C.; Leung, D.Y.; Ellis, C.N.; Jaffe, H.S.; Izu, A.E.; Bucalo, L.R.; Hirabayashi, S.E.; Tofte, S.J.; Cantu-Gonzales, G.; et al. Recombinant interferon gamma therapy for atopic dermatitis. J. Am. Acad. Dermatol. 1993, 28, 189–197, doi:10.1016/0190-9622(93)70026-P.
[114]  Schneider, L.C.; Baz, Z.; Zarcone, C.; Zurakowski, D. Long-term therapy with recombinant interferon-gamma (rIFN-gamma) for atopic dermatitis. Ann. Allergy Asthma Immunol. 1998, 80, 263–268, doi:10.1016/S1081-1206(10)62968-7.
[115]  Jang, I.G.; Yang, J.K.; Lee, H.J.; Yi, J.Y.; Kim, H.O.; Kim, C.W.; Kim, T.Y. Clinical improvement and immunohistochemical findings in severe atopic dermatitis treated with interferon gamma. J. Am. Acad. Dermatol. 2000, 42, 1033–1040, doi:10.1067/mjd.2000.104793.
[116]  Nielsen, B.W.; Reimert, C.M.; Hammer, R.; Schi?tz, P.O.; Thestrup-Pedersen, K. Interferon therapy for atopic dermatitis reduces basophil histamine release, but does not reduce serum IgE or eosinophilic proteins. Allergy 1994, 49, 120–128, doi:10.1111/j.1398-9995.1994.tb00811.x.
[117]  Jullien, D.; Nicolas, J.F.; Frappaz, A.; Thivolet, J. Alpha interferon treatment in atopic dermatitis. Acta. Derm. Venereol. 1993, 73, 130–132.
[118]  Noh, G.W.; Lee, K.Y. Blood eosinophils and serum IgE as predictors for prognosis of interferon-gamma therapy in atopic dermatitis. Allergy 1998, 53, 1202–1207, doi:10.1111/j.1398-9995.1998.tb03842.x.
[119]  Jolles, S.; Hughes, J.; Rustin, M. The treatment of atopic dermatitis with adjunctive high-dose intravenous immunoglobulin: A report of three patients and review of the literature. Br. J. Dermatol. 2000, 142, 551–554, doi:10.1046/j.1365-2133.2000.03377.x.
[120]  Jolles, S.; Sewell, C.; Webster, D.; Ryan, A.; Heelan, B.; Waite, A.; Rustin, M. Adjunctive high-dose intravenous immunoglobulin treatment for resistant atopic dermatitis: Efficacy and effects on intracellular cytokine levels and CD4 counts. Acta. Derm. Venereol. 2003, 83, 433–437, doi:10.1080/00015550310020549.
[121]  Paul, C.; Lahfa, M.; Bachelez, H.; Chevret, S.; Dubertret, L. A randomized controlled evaluator-blinded trial of intravenous immunoglobulin in adults with severe atopic dermatitis. Br. J. Dermatol. 2002, 147, 518–522.
[122]  Bemanian, M.H.; Movahedi, M.; Farhoudi, A.; Gharagozlou, M.; Seraj, M.H.; Pourpak, Z.; Nabavi, M.; Aghamohammadi, A.; Shirkhoda, Z. High doses intravenous immunoglobulin versus oral cyclosporine in the treatment of severe atopic dermatitis. Iran. J. Allergy Asthma Immunol. 2005, 4, 139–143.
[123]  Moul, D.K.; Routhouska, S.B.; Robinson, M.R.; Korman, N.J. Alefacept for moderate to severe atopic dermatitis: A pilot study in adults. J. Am. Acad. Dermatol. 2008, 58, 984–989, doi:10.1016/j.jaad.2008.02.007.
[124]  Takiguchi, R.; Tofte, S.; Simpson, B.; Harper, E.; Blauvelt, A.; Hanifin, J.; Simpson, E. Efalizumab for severe atopic dermatitis: A pilot study in adults. J. Am. Acad. Dermatol. 2007, 56, 222–227.
[125]  Simon, D.; H?sli, S.; Kostylina, G.; Yawalkar, N.; Simon, H.U. Anti-CD20 (rituximab) treatment improves atopic eczema. J. Allergy Clin. Immunol. 2008, 121, 122–128, doi:10.1016/j.jaci.2007.11.016.
[126]  Sheinkopf, L.E.; Rafi, A.W.; Do, L.T.; Katz, R.M.; Klaustermeyer, W.B. Efficacy of omalizumab in the treatment of atopic dermatitis: A pilot study. Allergy Asthma Proc. 2008, 29, 530–537, doi:10.2500/aap.2008.29.3160.
[127]  Phipps, S.; Flood-Page, P.; Menzies-Gow, A.; Ong, Y.E.; Kay, A.B. Intravenous anti-IL-5 monoclonal antibody reduces eosinophils and tenascin deposition in allergen-challenged human atopic skin. J. Invest. Dermatol. 2004, 122, 1406–1412, doi:10.1111/j.0022-202X.2004.22619.x.
[128]  Rullan, P.; Murase, J. Two cases of chronic atopic dermatitis treated with soluble tumor necrosis factor receptor therapy. J. Drugs Dermatol. 2009, 8, 873–876.
[129]  Jacobi, A.; Antoni, C.; Manger, B.; Schuler, G.; Hertl, M. Infliximab in the treatment of moderate to severe atopic dermatitis. J. Am. Acad. Dermatol. 2005, 52, 522–526, doi:10.1016/j.jaad.2004.11.022.

Full-Text

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133