中华皮肤科杂志 ›› 2023, Vol. 56 ›› Issue (3): 273-278.doi: 10.35541/cjd.20200715
何谐 栗玉珍
收稿日期:
2020-07-14
修回日期:
2021-02-02
发布日期:
2023-03-06
通讯作者:
栗玉珍
E-mail:liyuzhenchina@126.com
基金资助:
He Xie, Li Yuzhen
Received:
2020-07-14
Revised:
2021-02-02
Published:
2023-03-06
Contact:
Li Yuzhen
E-mail:liyuzhenchina@126.com
Supported by:
摘要: 【摘要】 银屑病的发生与Janus激酶(JAK)-信号转导及转录激活因子(STAT)通路密切相关,白细胞介素(IL)-23、IL-22、干扰素γ等多种细胞因子均可通过该通路传递信号,从而促进银屑病中角质形成细胞增殖及异常分化、炎症细胞浸润等病理过程,提示靶向该通路是治疗银屑病的新策略。近年来,小分子JAK抑制剂治疗银屑病显示出较好的疗效和安全性,而靶向STAT的药物正在开发阶段,为银屑病的治疗提供了更多选择。该文综述靶向JAK-STAT信号通路治疗银屑病的研究进展。
何谐 栗玉珍. 靶向JAK-STAT信号通路治疗银屑病的研究进展[J]. 中华皮肤科杂志, 2023,56(3):273-278. doi:10.35541/cjd.20200715
He Xie, Li Yuzhen. Targeting the JAK-STAT signaling pathway in the treatment of psoriasis[J]. Chinese Journal of Dermatology, 2023, 56(3): 273-278.doi:10.35541/cjd.20200715
[1] | Banerjee S, Biehl A, Gadina M, et al. JAK⁃STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects[J]. Drugs, 2017,77(5):521⁃546. doi: 10.1007/s40265⁃017⁃0701⁃9. |
[2] | Ciechanowicz P, Rakowska A, Sikora M, et al. JAK⁃inhibitors in dermatology: current evidence and future applications[J]. J Dermatolog Treat, 2019,30(7):648⁃658. doi: 10.1080/09546634. 2018.1546043. |
[3] | Chen M, Dai SM. A novel treatment for psoriatic arthritis: Janus kinase inhibitors[J]. Chin Med J (Engl), 2020,133(8):959⁃967.doi: 10.1097/CM9.0000000000000711. |
[4] | Schwartz DM, Kanno Y, Villarino A, et al. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases[J]. Nat Rev Drug Discov, 2017,16(12):843⁃862. doi: 10.1038/nrd.2017.201. |
[5] | Chalmin F, Humblin E, Ghiringhelli F, et al. Transcriptional programs underlying Cd4 T cell differentiation and functions[J]. Int Rev Cell Mol Biol, 2018,341:1⁃61. doi: 10.1016/bs.ircmb. 2018.07.002. |
[6] | Long D, Chen Y, Wu H, et al. Clinical significance and immunobiology of IL⁃21 in autoimmunity[J]. J Autoimmun, 2019,99:1⁃14. doi: 10.1016/j.jaut.2019.01.013. |
[7] | Yang L, Li B, Dang E, et al. Impaired function of regulatory T cells in patients with psoriasis is mediated by phosphorylation of STAT3[J]. J Dermatol Sci, 2016,81(2):85⁃92. doi: 10.1016/j.jdermsci.2015.11.007. |
[8] | Chen L, Shen Z. Tissue⁃resident memory T cells and their biological characteristics in the recurrence of inflammatory skin disorders[J]. Cell Mol Immunol, 2020,17(1):64⁃75. doi: 10. 1038/s41423⁃019⁃0291⁃4. |
[9] | Deng Y, Wang Z, Chang C, et al. Th9 cells and IL⁃9 in autoimmune disorders: pathogenesis and therapeutic potentials[J]. Hum Immunol, 2017,78(2):120⁃128. doi: 10.1016/j.humimm. 2016.12.010. |
[10] | Morelli M, Scarponi C, Mercurio L, et al. Selective immunomodulation of inflammatory pathways in keratinocytes by the Janus kinase (JAK) inhibitor tofacitinib: implications for the employment of JAK⁃targeting drugs in psoriasis[J]. J Immunol Res, 2018,2018:7897263. doi: 10.1155/2018/7897263. |
[11] | Johansen C, Rittig AH, Mose M, et al. STAT2 is involved in the pathogenesis of psoriasis by promoting CXCL11 and CCL5 production by keratinocytes[J/OL]. PLoS One, 2017,12(5):e0176994. doi: 10.1371/journal.pone.0176994. |
[12] | Liu L, Wu Y, Cao K, et al. Shikonin inhibits IFN⁃γ⁃induced K17 over⁃expression of HaCaT cells by interfering with STAT3 signaling[J]. Int J Clin Exp Pathol, 2015,8(8):9202⁃9207. |
[13] | Papp KA, Menter MA, Raman M, et al. A randomized phase 2b trial of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor, in patients with moderate⁃to⁃severe psoriasis[J]. Br J Dermatol, 2016,174(6):1266⁃1276. doi: 10.1111/bjd.14403. |
[14] | Punwani N, Scherle P, Flores R, et al. Preliminary clinical activity of a topical JAK1/2 inhibitor in the treatment of psoriasis[J]. J Am Acad Dermatol, 2012,67(4):658⁃664. doi: 10.1016/j.jaad.2011.12.018. |
[15] | Krueger J, Clark JD, Suárez⁃Fariñas M, et al. Tofacitinib attenuates pathologic immune pathways in patients with psoriasis: a randomized phase 2 study[J]. J Allergy Clin Immunol, 2016,137(4):1079⁃1090. doi: 10.1016/j.jaci.2015.12.1318. |
[16] | Abdulrahim H, Sharlala H, Adebajo AO. An evaluation of tofacitinib for the treatment of psoriatic arthritis[J]. Expert Opin Pharmacother, 2019,20(16):1953⁃1960. doi: 10.1080/14656566. 2019.1657404. |
[17] | Burmester GR, Curtis JR, Yun H, et al. An integrated analysis of the safety of tofacitinib in psoriatic arthritis across phase III and long⁃term extension studies with comparison to real⁃world observational data[J]. Drug Saf, 2020,43(4):379⁃392. doi: 10.1007/s40264⁃020⁃00904⁃9. |
[18] | Strober BE, Gottlieb AB, van de Kerkhof P, et al. Benefit⁃risk profile of tofacitinib in patients with moderate⁃to⁃severe chronic plaque psoriasis: pooled analysis across six clinical trials[J]. Br J Dermatol, 2019,180(1):67⁃75. doi: 10.1111/bjd.17149. |
[19] | Zhang J, Tsai TF, Lee MG, et al. The efficacy and safety of tofacitinib in Asian patients with moderate to severe chronic plaque psoriasis: aphase 3, randomized, double⁃blind, placebo⁃controlled study[J]. J Dermatol Sci, 2017,88(1):36⁃45. doi: 10. 1016/j.jdermsci.2017.05.004. |
[20] | Papp KA, Bissonnette R, Gooderham M, et al. Treatment of plaque psoriasis with an ointment formulation of the Janus kinase inhibitor, tofacitinib: a phase 2b randomized clinical trial[J]. BMC Dermatol, 2016,16(1):15. doi: 10.1186/s12895⁃016⁃0051⁃4. |
[21] | Kathuria H, Nguyen D, Handral HK, et al. Proposome for transdermal delivery of tofacitinib[J]. Int J Pharm, 2020,585:119558. doi: 10.1016/j.ijpharm.2020.119558. |
[22] | Mössner R, Hoff P, Mohr J, et al. Successful therapy of palmoplantar pustulosis with tofacitinib⁃report on three cases[J]. Dermatol Ther, 2020,33(4):e13753. doi: 10.1111/dth.13753. |
[23] | Wang YA, Rosenbach M. Successful treatment of refractory tumor necrosis factor inhibitor⁃induced palmoplantar pustulosis with tofacitinib: report of case[J]. JAAD Case Rep, 2020,6(2):115⁃118. doi: 10.1016/j.jdcr.2019.12.006. |
[24] | Shibata T, Muto J, Hirano Y, et al. Palmoplantar pustulosis⁃like eruption following tofacitinib therapy for juvenile idiopathic arthritis[J]. JAAD Case Rep, 2019,5(6):518⁃521. doi: 10.1016/j.jdcr.2019.03.024. |
[25] | Koumaki D, Koumaki V, Lagoudaki E, et al. Palmoplantar pustulosis⁃like eruption induced by baricitinib for treatment of rheumatoid arthritis[J]. Eur J Case Rep Intern Med, 2020,7(1):001383. doi: 10.12890/2019_001383. |
[26] | Di Domizio J, Castagna J, Algros MP, et al. Baricitinib⁃induced paradoxical psoriasis[J]. J Eur Acad Dermatol Venereol, 2020,34(8):e391⁃e393. doi: 10.1111/jdv.16293. |
[27] | Forman SB, Pariser DM, Poulin Y, et al. TYK2/JAK1 inhibitor PF⁃06700841 in patients with plaque psoriasis: phase IIa, randomized, double⁃blind, placebo⁃controlled trial[J]. J Invest Dermatol, 2020,140(12):2359⁃2370. doi: 10.1016/j.jid.2020.03. 962. |
[28] | Works MG, Yin F, Yin CC, et al. Inhibition of TYK2 and JAK1 ameliorates imiquimod⁃induced psoriasis⁃like dermatitis by inhibiting IL⁃22 and the IL⁃23/IL⁃17 axis[J]. J Immunol, 2014,193(7):3278⁃3287. doi: 10.4049/jimmunol.1400205. |
[29] | Papp K, Pariser D, Catlin M, et al. A phase 2a randomized, double⁃blind, placebo⁃controlled, sequential dose⁃escalation study to evaluate the efficacy and safety of ASP015K, a novel Janus kinase inhibitor, in patients with moderate⁃to⁃severe psoriasis[J]. Br J Dermatol, 2015,173(3):767⁃776. doi: 10.1111/bjd.13745. |
[30] | Jones P, Storer RI, Sabnis YA, et al. Design and synthesis of a pan⁃Janus kinase inhibitor clinical candidate (PF⁃06263276) suitable for inhaled and topical delivery for the treatment of inflammatory diseases of the lungs and skin[J]. J Med Chem, 2017,60(2):767⁃786. doi: 10.1021/acs.jmedchem.6b01634. |
[31] | Tanimoto A, Shinozaki Y, Yamamoto Y, et al. A novel JAK inhibitor JTE⁃052 reduces skin inflammation and ameliorates chronic dermatitis in rodent models: comparison with conventional therapeutic agents[J]. Exp Dermatol, 2018,27(1):22⁃29. doi: 10.1111/exd.13370. |
[32] | Mease P, Coates LC, Helliwell PS, et al. Efficacy and safety of filgotinib, a selective Janus kinase 1 inhibitor, in patients with active psoriatic arthritis (EQUATOR): results from a randomised, placebo⁃controlled, phase 2 trial[J]. Lancet, 2018,392(10162):2367⁃2377. doi: 10.1016/S0140⁃6736(18)32483⁃8. |
[33] | Orbai AM, Ogdie A, Gossec L, et al. Effect of filgotinib on health⁃related quality of life in active psoriatic arthritis: a randomized phase 2 trial (EQUATOR)[J]. Rheumatology (Oxford), 2020,59(7):1495⁃1504. doi: 10.1093/rheumatology/kez408. |
[34] | Bissonnette R, Luchi M, Fidelus⁃Gort R, et al. A randomized, double⁃blind, placebo⁃controlled, dose⁃escalation study of the safety and efficacy of INCB039110, an oral janus kinase 1 inhibitor, in patients with stable, chronic plaque psoriasis[J]. J Dermatolog Treat, 2016,27(4):332⁃338. doi: 10.3109/09546634. 2015.1115819. |
[35] | Schmieder GJ, Draelos ZD, Pariser DM, et al. Efficacy and safety of the Janus kinase 1 inhibitor PF⁃04965842 in patients with moderate⁃to⁃severe psoriasis: phase II, randomized, double⁃blind, placebo⁃controlled study[J]. Br J Dermatol, 2018,179(1):54⁃62. doi: 10.1111/bjd.16004. |
[36] | Ludbrook VJ, Hicks KJ, Hanrott KE, et al. Investigation of selective JAK1 inhibitor GSK2586184 for the treatment of psoriasis in a randomized placebo⁃controlled phase IIa study[J]. Br J Dermatol, 2016,174(5):985⁃995. doi: 10.1111/bjd.14399. |
[37] | Burke JR, Cheng L, Gillooly KM, et al. Autoimmune pathways in mice and humans are blocked by pharmacological stabilization of the TYK2 pseudokinase domain[J]. Sci Transl Med, 2019,11(502):eaaw1736. doi: 10.1126/scitranslmed.aaw1736. |
[38] | Papp K, Gordon K, Thaçi D, et al. Phase 2 trial of selective tyrosine kinase 2 inhibition in psoriasis[J]. N Engl J Med, 2018,379(14):1313⁃1321. doi: 10.1056/NEJMoa1806382. |
[39] | Singh R, Pradhan V, Roberts ES, et al. Safety and pharmacokinetics of the oral TYK2 inhibitor PF⁃06826647: a phase I, randomized, double⁃blind, placebo⁃controlled, dose⁃escalation study[J/OL]. Clin Transl Sci, 2020[2021⁃02⁃02].https://ascpt.onlinelibrary.wiley.com/doi/10.1111/cts.12929. doi: 10.1111/cts.12929. |
[40] | Miyoshi K, Takaishi M, Nakajima K, et al. Stat3 as a therapeutic target for the treatment of psoriasis: a clinical feasibility study with STA⁃21, a Stat3 inhibitor[J]. J Invest Dermatol, 2011,131(1):108⁃117. doi: 10.1038/jid.2010.255. |
[41] | Park JS, Kwok SK, Lim MA, et al. STA⁃21, a promising STAT⁃3 inhibitor that reciprocally regulates Th17 and Treg cells, inhibits osteoclastogenesis in mice and humans and alleviates autoimmune inflammation in an experimental model of rheumatoid arthritis[J]. Arthritis Rheumatol, 2014,66(4):918⁃929. doi: 10.1002/art.38305. |
[42] | Kim JY, Ahn J, Kim J, et al. Nanoparticle⁃assisted transcutaneous delivery of a signal transducer and activator of transcription 3⁃inhibiting peptide ameliorates psoriasis⁃like skin inflammation[J]. ACS Nano, 2018,12(7):6904⁃6916. doi: 10. 1021/acsnano.8b02330. |
[43] | Schöneberger H, Weiss A, Brill B, et al. The integration of a Stat3 specific peptide aptamer into the thioredoxin scaffold protein strongly enhances its inhibitory potency[J]. Horm Mol Biol Clin Investig, 2011,5(1):1⁃9. doi: 10.1515/HMBCI.2011. 013. |
[44] | Mack L, Brill B, Delis N, et al. Stat3 is activated in skin lesions by the local application of imiquimod, a ligand of TLR7, and inhibited by the recombinant peptide aptamer rS3⁃PA[J]. Horm Mol Biol Clin Investig, 2012,10(2):265⁃272.doi: 10.1515/hmbci⁃2012⁃0007. |
[45] | Miyoshi K, Takaishi M, Digiovanni J, et al. Attenuation of psoriasis⁃like skin lesion in a mouse model by topical treatment with indirubin and its derivative E804[J]. J Dermatol Sci, 2012,65(1):70⁃72. doi: 10.1016/j.jdermsci.2011.10.001. |
[46] | Tang L, He S, Wang X, et al. Cryptotanshinone reduces psoriatic epidermal hyperplasia via inhibiting the activation of STAT3[J]. Exp Dermatol, 2018,27(3):268⁃275. doi: 10.1111/exd.13511. |
[47] | Skyvalidas DΝ, Mavropoulos A, Tsiogkas S, et al. Curcumin mediates attenuation of pro⁃inflammatory interferon γ and interleukin 17 cytokine responses in psoriatic disease, strengthening its role as a dietary immunosuppressant[J]. Nutr Res, 2020,75:95⁃108. doi: 10.1016/j.nutres.2020.01.005. |
[48] | Marepally S, Boakye CH, Patel AR, et al. Topical administration of dual siRNAs using fusogenic lipid nanoparticles for treating psoriatic⁃like plaques[J]. Nanomedicine (Lond), 2014,9(14):2157⁃2174. doi: 10.2217/nnm.13.202. |
[49] | Ran LW, Wang H, Lan D, et al. Effect of RNA interference targeting STAT3 gene combined with ultrasonic irradiation and SonoVue microbubbles on proliferation and apoptosis in keratinocytes of psoriatic lesions[J]. Chin Med J (Engl), 2018,131(17):2097⁃2104. doi: 10.4103/0366⁃6999.239297. |
[50] | Ran LW, Wang H, Lan D, et al. Effects of RNA interference combined with ultrasonic irradiation and SonoVue microbubbles on expression of STAT3 gene in keratinocytes of psoriatic lesions[J]. J Huazhong Univ Sci Technolog Med Sci, 2017,37(2):279⁃285. doi: 10.1007/s11596⁃017⁃1728⁃6. |
[51] | Hu XP, Xie Q, Chen CF, et al. Let⁃7a inhibits T⁃cell proliferation and IFN⁃γ secretion by down⁃regulating STAT3 expression in patients with psoriasis[J]. Cell Physiol Biochem, 2017,42(1):115⁃125. doi: 10.1159/000477120. |
[52] | Chowdhari S, Saini N. hsa⁃miR⁃4516 mediated downregulation of STAT3/CDK6/UBE2N plays a role in PUVA induced apoptosis in keratinocytes[J]. J Cell Physiol, 2014,229(11):1630⁃1638. doi: 10.1002/jcp.24608. |
[53] | Wang Y, Yu X, Wang L, et al. miR⁃320b is down⁃regulated in psoriasis and modulates keratinocyte proliferation by targeting AKT3[J]. Inflammation, 2018,41(6):2160⁃2170. doi: 10.1007/s10753⁃018⁃0859⁃7. |
[54] | Yan JJ, Qiao M, Li RH, et al. Downregulation of miR⁃145⁃5p contributes to hyperproliferation of keratinocytes and skin inflammation in psoriasis[J]. Br J Dermatol, 2019,180(2):365⁃372. doi: 10.1111/bjd.17256. |
[1] | 中华医学会皮肤性病学分会免疫学组. [开放获取] 中国慢性诱导性荨麻疹诊治专家共识(2023)[J]. 中华皮肤科杂志, 2023, 0(4): 20220819-e20220819. |
[2] | 中华医学会皮肤性病学分会 中国医师协会皮肤科医师分会. [开放获取] 慢性自发性荨麻疹达标治疗专家共识(2023)[J]. 中华皮肤科杂志, 2023, 0(4): 20230004-e20230004. |
[3] | 阮诗钒 张鹏 林婷婷 罗仁炜 鲍思怡 薛晨瑶 童泽群 张亮亮 龚婷 纪超. 寻常型银屑病与血脂异常的相关性研究[J]. 中华皮肤科杂志, 2023, 0(3): 20220322-e20220322. |
[4] | 中华医学会皮肤性病学分会银屑病专业委员会. 银屑病生物制剂达标治疗专家共识[J]. 中华皮肤科杂志, 2023, 56(3): 191-203. |
[5] | 周健 俞晨 王刚. 司库奇尤单抗治疗13例难治性局限性脓疱型银屑病的临床疗效与安全性观察[J]. 中华皮肤科杂志, 2023, 56(3): 247-251. |
[6] | 胡琨 杨晶 王巧林 陈军臣 张谧 朱武 张斌 窦冠珅 Wendong Chen, 匡叶红. [开放获取] 依奇珠单抗治疗银屑病短期疗效的单中心病例回顾性研究[J]. 中华皮肤科杂志, 2023, 56(3): 210-215. |
[7] | 万立 胡彬 罗红玉 方美珍 韩丽娟 陈强 周小勇 陈柳青 陈金波. 天疱疮或大疱性类天疱疮合并单纯疱疹病毒感染8例临床特征及治疗分析[J]. 中华皮肤科杂志, 2023, 56(3): 229-233. |
[8] | 罗帅寒天 龙海 陆前进. 2022年系统性红斑狼疮研究新进展[J]. 中华皮肤科杂志, 2023, 56(3): 266-269. |
[9] | 陈奇权 杨显杰 顾恒 徐金华 郝飞 姚煦 宋志强. [开放获取] 基于德尔菲法构建《中国慢性诱导性荨麻疹专家诊治共识(2023)》[J]. 中华皮肤科杂志, 2023, 0(3): 20220814-e20220814. |
[10] | 苑辰 闫言 王宝玺. 反常性痤疮的生物治疗进展[J]. 中华皮肤科杂志, 2023, 56(3): 270-273. |
[11] | 张红燕 李丽 王明霞 董银卯 孟宏 易帆. 益生菌在痤疮治疗中的应用进展[J]. 中华皮肤科杂志, 2023, 56(3): 283-285. |
[12] | 洪芷榆 谢红付 李吉 黄莹雪. 发光二极管红黄光治疗急性发作期面部炎症性皮肤病的支付意愿分析[J]. 中华皮肤科杂志, 2023, 0(3): 20220128-e20220128. |
[13] | 任芬芬 王鹏 张景展 康晓静. 基于网络药理学预测中药在抗卡波西肉瘤血管生成治疗中的潜在有效成分及分子作用机制[J]. 中华皮肤科杂志, 2023, 0(3): 20220140-e20220140. |
[14] | 王召阳 向欣 陈云刘 苗朝阳 赵欣荣 张振华 刘元香 马琳 徐子刚. 生物制剂与甲氨蝶呤治疗儿童重度斑块状银屑病疗效及安全性比较[J]. 中华皮肤科杂志, 2023, 56(2): 112-117. |
[15] | 贾元源 毛秋雨 杨婧怡 闵玮. 度普利尤单抗治疗中重度老年特应性皮炎临床疗效观察[J]. 中华皮肤科杂志, 2023, 56(2): 125-129. |
|