分享

国家银屑病基金会医学董事会:伴炎症性肠病的银屑病患者的治疗

 6226cy 2018-07-14

Scott M. Whitlock, MD, Clinton W. Enos, MD, April W.Armstrong, MD, MPH, Alice Gottlieb, MD, Richard G. Langley, MD, Mark Lebwohl,MD, Joseph F. Merola, MD, MMSc, Caitriona Ryan, MD, Michael P. Siegel, PhD,Jeffrey M. Weinberg, MD, Jashin J. Wu, MD, and Abby S. Van Voorhees, MD

主题负责人:吴文育  上海华山医院

审校:贾雪松  新疆石河子大学医学院第一附属医院

翻译:唐教清  四川大学华西医院

摘要

背景:银屑病与炎症性肠病(inflammatory bowel disease,IBD)有密切关联,众多针对银屑病和银屑病性关节炎的治疗同样可用于IBD。

目的:评估伴IBD的银屑病患者的治疗。

方法:对银屑病、银屑病性关节炎、溃疡性结肠炎和克罗恩病系统用药和生物制剂的临床研究进行系统文献检索,检索时间限定为1947年1月1日至2017年2月14日。优先选择随机对照双盲研究,若没有则选择证据级别最高者。

结果:共检索2282篇文献,最终选定其中的132篇。英夫利昔单抗和阿达木单抗对银屑病、银屑病性关节炎、溃疡性结肠炎和克罗恩病有效。赛妥珠单抗对银屑病性关节炎和克罗恩病有效。依那西普、苏金单抗、布罗达单抗和艾克司单抗对银屑病和银屑病性关节炎有效,但可能加重或诱发IBD。古塞库单抗对银屑病有效。

不足:尚无临床试验专门评估银屑病合并IBD的治疗。

结论:英夫利昔单抗和阿达木单抗对银屑病、银屑病性关节炎、溃疡性结肠炎和克罗恩病有效,其他药物对上述部分(非所有)疾病有效。(J Am Acad Dermatol 2018;78:383-94.)

关键词:克罗恩病;炎症性肠病;银屑病;银屑病性关节炎;溃疡性结肠炎

内容提要

  • 银屑病与炎症性肠病存在密切关联。

  • 英夫利昔单抗和阿达木单抗对银屑病、银屑病性关节炎、溃疡性结肠炎和克罗恩病有效。

  • 炎症性肠病可能与IL-17抑制剂相关,或可考虑使用此类药物治疗炎症性肠病患者。

缩略词表

炎症性肠病:inflammatorybowel disease,IBD

白细胞介素:interleukin,IL

美国食品药品监督管理局:US Food andDrug Administration,FDA

肿瘤坏死因子:tumor necrosisfactor,TNF

引言

银屑病与炎症性肠病(inflammatorybowel disease,IBD)存在密切关联1-3。尽管银屑病确切发生率不明,据估计克罗恩病患者出现银屑病的比例为9.6%(一般人群出现银屑病的比例为2.2%)2,而银屑病患者中出现克罗恩病的比例预估为0.5%(一般人群出现克罗恩病的比例为0.2%)。溃疡性结肠炎情况类似3。重度银屑病患者中,出现克罗恩病的相对风险为2.85,出现溃疡性结肠炎的相对风险为1.964。银屑病和IBD均为上皮组织的多因性慢性炎症性疾病,均涉及细胞免疫活化。IL-23R的基因多态性可转变IL-22/23信号通路,均可见于银屑病和IBD5-7。银屑病和克罗恩病还共有众多其他易感位点8

尽管有众多药物可治疗银屑病和IBD,但某些银屑病治疗药物可能诱发或加重IBD。有大量病例报道显示TNF抑制剂治疗IBD时会诱发银屑病样皮损9,10。尽管IBD常会系统应用糖皮质激素,但指南并不推荐此药用于银屑病,因停药后会致疾病反弹11,12。当为合并银屑病和IBD患者制定治疗计划时,需了解哪些药物对IBD有改善作用,哪些药物又有加重作用。治疗IBD时,与患者的内科医生或消化科医生合作将有助于优化相关疾病的治疗策略。本综述旨在总结各种用于银屑病和银屑病性关节炎的药物,并简要讨论相应药物在IBD的证据级别。

方法

对PubMed数据库进行系统文献检索(图1),时间为1947年1月1日至2017年2月14日,检索词为“infliximab”、“adalimumab”、“golimumab”、“certolizumab”、“etanercept”、“ustekinumab”、“secukinumab”、“ixekixumab”、“acitretin”、“cyclosporine”、“methotrexate”、“apremilast”、“mycophenolate”、“mofetil”、“sulfasalazine”、“hydroxyurea”、“azathioprine”、 “6-thioguanine”、“tacrolimus”、“leflunomide”和“fumaric acid esters”。

文章类型为临床试验(clinical trial)。这些药物分别与“psoriasis”、“psoriatic arthritis”、“inflammatory boweldisease”、“Crohn’s disease”和“ulcerativecolitis”配对检索。其他文章来自初始检索中的参考文献。

后续综述优先选择随机对照双盲试验,其次选择Ⅲ期单药临床试验,若均无上述类型试验则选择Ⅱ期临床试验或证据级别最高的试验类型。另外会剔除针对药物输送技术的临床试验,会纳入每个药物和疾病相关的Meta分析,方法为上述检索词分别与“meta-analysis”进行联合检索。

根据Shekelle等人的指南13,记录每种药物对于每种疾病的最高证据级别。ⅠA级指证据来自随机对照试验的Meta分析;ⅠB级证据指来自随机对照试验;ⅡA级指证据来自非随机的对照试验;ⅡB级提示证据来自其他准实验研究(quasi-experimental study);Ⅲ级指证据来自非实验的观察性研究,例如对比研究、相关性研究和病例对照研究;Ⅳ级指专家会议报道、观点或权威临床经验。有效性推荐强度分为“强”、“中”和“弱”。

结果

共检索2282篇文献,2164篇文献不满足纳入标准,最终选定132篇文献,其中包括15篇Meta分析。

FDA获批的银屑病和/或银屑病性关节炎生物制剂

英夫利昔单抗(Infliximab)。英夫利昔单抗(商品名为Remicade和Inflectra)是抗TNF的单克隆抗体,由FDA获批用于治疗银屑病(2006年)、银屑病性关节炎(2005年)、溃疡性结肠炎(2006年)、克罗恩病(1998年)、儿童克罗恩病(2006年)、儿童溃疡性结肠炎(2011年)及其他疾病(表Ⅰ)14-47。Ⅲ期临床试验证实英夫利昔单抗对银屑病14和银屑病性关节炎15有效。另有Ⅲ期临床试验证实英夫利昔单抗对克罗恩病患者16,48,49和溃疡性结肠炎患者17的维持缓解治疗有效。

总之,英夫利昔单抗对银屑病50和银屑病性关节炎51疗效(有效性推荐强度均为强推荐)证据为ⅠA级。同时对克罗恩病52,53和溃疡性结肠炎54的有效(均为强推荐)证据亦为ⅠA级。

阿达木单抗(Adalimumab)。阿达木单抗(商品名Humira)是另外一种抗TNF的单克隆抗体,FDA批准用于银屑病(2008年)、银屑病性关节炎(2005年)、溃疡性结肠炎(2012年)、克罗恩病(2007年)、儿童克罗恩病(2014年)、儿童溃疡性结肠炎(2011年)及其他疾病55。Ⅲ期临床试验证实阿达木单抗对银屑病18,56和银屑病性关节炎19,57有效。另有Ⅲ期临床试验证实阿达木单抗对克罗恩病20,58,59和溃疡性结肠炎21,60有效。

总之,阿达木单抗对银屑病50和银屑病性关节炎51疗效(均为强推荐)证据为ⅠA级。同时对克罗恩病53(强推荐)和溃疡性结肠炎54(中推荐)的有效证据亦为ⅠA级。

戈利木单抗(Golimumab)。戈利木单抗(商品名为Simponi)也是抗TNF的单克隆抗体,FDA批准用于银屑病性关节炎(2009)、溃疡性结肠炎(2013年)及其他疾病,但不包括银屑病和克罗恩病61。尽管无发表的临床试验评估此药对银屑病的疗效,但有Ⅲ期临床试验证实此药对银屑病性关节炎和银屑病均有效22。仅有一项非对照的超适应证研究证实戈利木单抗对难治性克罗恩病有一定疗效23。Ⅲ期临床试验证实戈利木单抗对溃疡性结肠炎患者的诱导治疗和维持治疗均有效24,62,63

总之,戈利木单抗对银屑病61(中推荐)疗效证据为ⅡB级,对银屑病性关节炎51(强推荐)疗效证据为ⅠA级。戈利木单抗对克罗恩病23(强推荐)疗效证据为Ⅲ级,对溃疡性结肠炎54(强推荐)的疗效证据为ⅠA级。

聚乙二醇结合赛妥珠单抗(Certolizumabpegol)。聚乙二醇结合赛妥珠单抗(商品名为Cimzia)是一种针对TNF-α的聚乙二醇化单克隆抗体片段。目前FDA批准聚乙二醇结合赛妥珠单抗用于银屑病性关节炎(2013年)、克罗恩病(2008年)及其他疾病的治疗,但不包括银屑病和溃疡性结肠炎64。已有不同Ⅲ期临床试验证实赛妥珠单抗对银屑病性关节炎26和克罗恩病27,65,66有效。另有小型非对照的超适应证研究证实此药对溃疡性结肠炎有一定疗效28

总之,聚乙二醇结合赛妥珠单抗对银屑病61(强推荐)疗效证据为ⅠB级,对银屑病性关节炎67(强推荐)疗效证据为ⅠA级。聚乙二醇结合赛妥珠单抗对克罗恩病53(强推荐)疗效证据为ⅠA级,对溃疡性结肠炎28(弱推荐)的疗效证据亦为Ⅲ级。

依那西普(Etanercept)。依那西普(商品名为Enbrel)是一种抗TNF的重组融合蛋白,FDA批准用于银屑病(2004年)、银屑病性关节炎(2002年)、儿童银屑病(2016年)及其他疾病,但不包括克罗恩病和溃疡性结肠炎68。已有Ⅲ期临床试验证实依那西普对银屑病29,69,70和银屑病性关节炎30,70有效。尚无已发表文献报道依那西普用于溃疡性结肠炎。另有小型对照研究证实此药对克罗恩病无效71

IBD是依那西普的潜在副作用68。尽管Ⅲ期临床试验未报道使用依那西普后IBD新发或加重,但大规模回顾性研究提示依那西普可能诱发或加重部分IBD患者的病情32

总之,依那西普对银屑病50(中推荐)疗效证据为ⅠA级,对银屑病性关节炎51(强推荐)疗效证据为ⅠA级。依那西普对克罗恩病71疗效(无效)证据为ⅠB级,对溃疡性结肠炎疗效无相关研究。考虑到依那西普有加重IBD的风险,IBD患者用药时需谨慎。

优特克单抗(Ustekinumab)。优特克单抗(商品名为Stelara)是一种IL-12/23抑制剂,FDA批准用于银屑病(2009年)、银屑病性关节炎(2013年)和克罗恩病(2016年),但未批准用于溃疡性结肠炎72。已有Ⅲ期临床试验证实优特克单抗对中重度斑块性银屑病33,73和银屑病性关节炎31,34有效。同时优特克单抗对库洛恩兵的诱导缓解和维持治疗有效35,74-76。尚无已发表文献报道优特克单抗用于溃疡性结肠炎。

总之,优特克单抗对银屑病50(强推荐)疗效证据为ⅠA级,对银屑病性关节炎67(中推荐)疗效证据为ⅠA级。优特克单抗对克罗恩病74疗效(强推荐)证据为ⅠB级,对溃疡性结肠炎疗效无相关研究。

苏金单抗(Secukinumab)。苏金单抗(商品名Cosentyx)是一种IL-17A抑制剂,FDA批准用于银屑病(2015年)、银屑病性关节炎(2016年)及其他疾病77。Ⅲ期临床试验证实苏金单抗对银屑病36,78,79和银屑病性关节炎37,79有效。对溃疡性结肠炎疗效无相关研究。Ⅱ期临床试验证实此药对克罗恩病无效,且与安慰剂相比可增加疾病活跃度38

IBD患者应慎重使用苏金单抗77。针对斑块型银屑病的Ⅱ期和Ⅲ期临床试验显示,有9例患者出现IBD新发或加重(发生率为每0.33每100患者年),而依那西普对照组仅有1例(发生率为0.34每100患者年),作者分析认为苏金单抗和IBD无临床联系39。然而,鉴于对照组依那西普本身与IBD相关(正如此前所述),目前很难给出相关与否的确切定论。一项苏金单抗用于强直性脊柱炎的Ⅲ期临床试验显示,苏金单抗治疗组共5例新发或加重的IBD病例(发生率为0.7每100患者年),对照组病例数为080

总之,苏金单抗对银屑病51(强推荐)疗效证据为ⅠA级,对银屑病性关节炎67(强推荐)疗效证据为ⅠA级。苏金单抗对克罗恩病证据为ⅠB级(无效)38,对溃疡性结肠炎疗效无相关研究。考虑到苏金单抗有加重IBD的风险,IBD患者用药时需谨慎。

艾克司单抗(Ixekizumab)。艾克司单抗(商品名Taltz)是一种IL-17A抑制剂,FDA批准用于银屑病(2016年)81。Ⅲ期临床试验证实艾克司单抗对银屑病有效40,41。尽管FDA未批准用于银屑病性关节炎,但近期有Ⅲ期临床试验证实艾克司单抗对银屑病性关节炎82有效。对克罗恩病和溃疡性结肠炎疗效无相关研究。

接受艾克司单抗治疗者应密切监测IBD的情况81。一项未披露的银屑病临床试验40显示,艾克司单抗组出现溃疡性结肠炎的病例数为7(发生率为每0.2每100患者年),克罗恩病病例数为4(发生率为0.1每100患者年),对照组病例数为0。不过,艾克司单抗与IBD的关联还未完全阐明83

总之,艾克司单抗对银屑病和银屑病性关节炎40,41,82(均为强推荐)疗效证据为ⅠB级,对克罗恩病和溃疡性结肠炎疗效无相关研究。考虑到艾克司单抗有加重IBD的风险,IBD患者用药时需谨慎。

布罗达单抗(Brodalumab)。布罗达单抗(商品名Siliq)是一种抗IL-17受体A的IgG2单克隆靶向抗体。目前FDA批准用于中重度斑块型银屑病(2017年)84。Ⅲ期临床试验证实布罗达单抗对银屑病有效(其中包括一项与优特克单抗的对比试验)85-87。目前FDA未批准用于银屑病性关节炎,但近期有Ⅱ期临床试验证实布罗达单抗对银屑病性关节炎有效88,89。布罗达单抗对溃疡性结肠炎疗效无相关研究。Ⅱ期临床试验证实布罗达单抗会加重中重度克罗恩病,并导致研究提前终止90

布罗达单抗有自杀意向及行为的黑框警告,由风险评估和减轻战略(Risk Evaluation and Mitigation Strategy)进行严格管束。一项Ⅲ期临床试验报道的4例致命性副作用中有2例为自杀96。此外,患者服用布罗达单抗期间需监测IBD的情况。除了上文提到的Ⅱ期临床试验,另有一项Ⅲ期临床试验报道1例患者在治疗期间出现克罗恩病85

总之,布罗达单抗对银屑病(强推荐)疗效证据为ⅠA级,对银屑病性关节炎疗效证据为ⅠB级。

古塞库单抗(Guselkumab)。古塞库单抗(商品名Tremfya)是一种抗IL-23 p19亚基的IgG1单克隆抗体。FDA批准用于中重度银屑病91。Ⅲ期临床试验证实古塞库单抗有效且耐受性良好92,93。根据检索标准来看,古塞库单抗用于银屑病性关节炎的研究还未发表。检索网址clinicaltrials.gov可见目前有正在进行的银屑病性关节炎Ⅱ期和Ⅲ期临床试验。无涉及古塞库单抗用于克罗恩病和溃疡性结肠炎的研究。

总之,古塞库单抗对银屑病(强推荐)疗效证据为ⅠB级。截止撰稿期间,有关银屑病性关节炎的临床试验还未完成。对克罗恩病和溃疡性结肠炎疗效无相关研究。

FDA批准的银屑病系统用药(非生物制剂)

阿维A阿维A是1997年FDA批准用于银屑病(不包括银屑病性关节炎、克罗恩病及溃疡性结肠炎)的口服维A酸类药物(表Ⅱ)94-105,107-111。随机对照试验证实阿维A对银屑病有效94,112,尽管疗效常弱于其他传统系统用药(归因于剂量限制性毒性)。阿维A被认为对银屑病性关节炎无效,无临床试验对此病进行评估。对IBD无相关研究。

总之,阿维A对银屑病(强推荐)疗效证据为ⅠA级113。此前对银屑病性关节炎、克罗恩病和溃疡性结肠炎疗效无相关研究。

环孢素。环孢素1997年FDA批准用于银屑病(不包括银屑病性关节炎、克罗恩病及溃疡性结肠炎)的免疫抑制剂114。随机对照试验证实环孢素可有效治疗银屑病95,115。同时,另有随机对照试验证实此药对银屑病性关节炎有效96,116,117。有时环孢素会超适应证用于治疗克罗恩病,可有不同程度疗效97,98。小规模随机对照实验提示此药可有效诱导缓解溃疡性结肠炎病情99-101。环孢素神经毒性等有数种严重不良反应,会影响用药安全114

总之,环孢素对银屑病118(强推荐)疗效证据为ⅠA级,对银屑病性关节炎119(强推荐)疗效证据为ⅠA级。环孢素对溃疡性结肠炎120(中推荐)疗效证据为ⅠA级,对克罗恩病98(弱推荐)的疗效证据亦为ⅠA级。

甲氨蝶呤。甲氨蝶呤由FDA批准用于治疗银屑病(1972年),已有随机对照试验证实其疗效95,102。甲氨蝶呤有超适应证用于银屑病性关节炎,随机对照试验证实此药有不同程度的疗效103-105。同时,随机对照试验证实此药对克罗恩病有效107,121,对溃疡性结肠炎疗效有争议108,122。甲氨蝶呤药品信息特别提醒有溃疡性结肠炎时用药需谨慎103

总之,甲氨蝶呤对银屑病106(中推荐)疗效证据为ⅠA级,对银屑病性关节炎119(中推荐)疗效证据为ⅠA级。甲氨蝶呤对克罗恩病123(强推荐)疗效证据为ⅠA级,对溃疡性结肠炎124(弱推荐)的疗效证据亦为ⅠA级。

阿普斯特(Apremilast)。阿普斯特(商品名为Otezla)是口服的磷酸二酯酶-4抑制剂,FDA批准用于银屑病(2014年)和银屑病性关节炎(2014年)的治疗125。Ⅲ期临床试验证实阿普斯特可有效治疗斑块型银屑病109,126和银屑病性关节炎110。无涉及阿普斯特用于克罗恩病和溃疡性结肠炎的研究。最常见不良反应为恶心和腹泻,有IBD的患者服用时需留意此情况。

总之,阿普斯特对银屑病50(中推荐)疗效证据为ⅠA级,对银屑病性关节炎67(强推荐)疗效证据为ⅠA级。截止撰稿期间,有关银屑病性关节炎的临床试验还未完成。对克罗恩病和溃疡性结肠炎疗效无相关研究。

超适应证药物

银屑病超适应证药物及药物对银屑病性关节炎、克罗恩病及溃疡性结肠炎的使用总结于表Ⅲ16,96,127-161

结论

英夫利昔单抗和阿达木单抗对斑块型银屑病、银屑病性关节炎、克罗恩病及溃疡性结肠炎有效。优特克单抗对斑块型银屑病、银屑病性关节炎及克罗恩病有效。赛妥珠单抗对银屑病性关节炎及克罗恩病有效。戈利木单抗对银屑病性关节炎及溃疡性结肠炎有效。布罗达单抗对银屑病有效,对银屑病性关节炎可能有效。古塞库单抗对银屑病有效。尚有众多其他药物有不同程度的疗效。

布罗达单抗会加重克罗恩病症状,依那西普、艾克司单抗和苏金单抗可能加重克罗恩病症状。推断现有银屑病临床试验数据以评估IBD的加重风险存在局限性,部分临床试验并未统计IBD相关数据。此外,本身有IBD病史的患者服药后最容易出现IBD的加重。因此,一般银屑病人群与具有IBD病史的银屑病人群相比,其出现IBD的真实风险可能被低估了。未确定银屑病与IBD的潜在关联,还有必要对长期数据进行严格考察。

参考文献

1.Kim M, Choi KH, Hwang SW, Lee YB, Park HJ, Bae JM. Inflammatory bowel diseaseis associated with an increased risk of inflammatory skin diseases: apopulation-based cross-sectional study. J Am Acad Dermatol. 2017;76:40-48.

2.Lee FI, Bellary SV, Francis C. Increased occurrence of psoriasis in patientswith Crohns disease and their relatives. Am J Gastroenterol.1990;85(8):962-963.

3.Cohen AD, Dreiher J, Birkenfeld S. Psoriasis associated with ulcerative colitisand Crohns disease. J Eur Acad Dermatol Venereol. 2009;23:561.

4. Egeberg A, Mallbris L, Warren RB, et al. Association between psoriasis and inflammatory boweldisease: a Danish nationwide cohort study. Br J Dermatol. 2016;175:487-492.

5.Cargill M, Schrodi SJ, Chang M, et al. A large-scale genetic association studyconfirms IL12B and leads to the identification of IL23R as psoriasis-riskgenes. Am J Hum Genet. 2007;80:273-290.

6.Nair RP, Duffin KC, Helms C, et al. Genome-wide scan reveals association ofpsoriasis with IL-23 and NF-kappaB pathways. Nat Genet. 2009;41:199-204.

7. Wang K, Zhang H, Kugathasan S, et al. Diverse genome-wide association studies associate theIL12/IL23 pathway with Crohns disease. Am J Hum Genet. 2009;84:399-405.

8.Ellinghaus D, Ellinghaus E, Nair RP, et al. Combined analysis of genome-wideassociation studies for Crohn disease and psoriasis identifies seven sharedsusceptibility loci. Am J Hum Genet. 2012;90(4):636-647.

9. Denadai R, Teixeira FV, Steinwurz F, et al. Induction or exacerbation of psoriatic lesions duringanti-TNF-a therapy for inflammatory bowel disease: a systematic literaturereview based on 222 cases. J Crohns Colitis. 2013;7(7):517-524.

10.Cullen G, Kroshinsky D, Cheifetz AS, et al. Psoriasis associated withanti-tumour necrosis factor therapy in inflammatory bowel disease: a new seriesand a review of 120 cases from the literature. Aliment Pharmacol Ther.2011;34:1318-1327.

11.Brenner M, Molin S, Ruebsam K, et al. Generalized pustular psoriasis induced bysystemic glucocorticosteroids: four cases and recommendations for treatment. BrJ Dermatol. 2009;161:964-966.

12.Coates LC, Helliwell PS. Psoriasis flare with corticosteroid use in psoriaticarthritis. Br J Dermatol. 2016;174:219-221.

13.Shekelle PG,Woolf SH, EcclesM, GrimshawJ. Clinical guidelines: developingguidelines. BMJ. 1999;318(7183):593-596.

14.Reich K, Nestle FO, Papp K, et al. Infliximab induction and maintenance therapyfor moderate-to-severe psoriasis: a phase III, multicentre, double-blind trial.Lancet.2005; 366(9494):1367-1374.

15. Antoni C, Krueger GG, de Vlam K, et al. Infliximab improves signs and symptoms of psoriaticarthritis: results of the IMPACT 2 trial. Ann Rheum Dis. 2005;64:1150-1157.

16.Colombel JF, Sandborn WJ, Reinisch W, et al. Infliximab, azathioprine, orcombination therapy for Crohn’s disease. N Engl J Med. 2010;362:1383-1395.

17. Rutgeerts P, Sandborn WJ, Feagan BG, et al. Infliximab for induction and maintenance therapy forulcerative colitis. N Engl J Med. 2005;353:2462-2476.

18. Gordon KB, Langley RG, Leonardi C, et al. Clinical response to adalimumab treatment in patientswith moderate to severe psoriasis: double-blind, randomized controlled trialand open-label extension study.JAmAcadDermatol. 2006;55:598-606.

19.Mease PJ, Gladman DD, Ritchlin CT, et al. Adalimumab for the treatment ofpatients with moderately to severely active psoriatic arthritis: results of adouble-blind, randomized, placebo-controlled trial. Arthritis Rheum.2005;52:3279-3289.

20. Hanauer SB, Sandborn WJ, Rutgeerts P, et al. Human anti-tumor necrosis factor monoclonal antibody(adalimumab) in Crohn’s disease: the CLASSIC-I trial. Gastroenterology.2006;130(2):323-333.

21.Sandborn WJ, van Assche G, Reinisch W, et al. Adalimumab induces and maintainsclinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2012;142(2):257-265.

22. Kavanaugh A, van der Heijde D, McInnes IB, et al. Golimumab in psoriatic arthritis: one-year clinicalefficacy, radiographic, and safety results from a phase III, randomized,placebo-controlled trial. Arthritis Rheum. 2012;64(8):2504-2517.

23. Ben-Bassat O, Iacono A, Irwin SP, et al. Tu1327a golimumab for treatment of moderate to severeanti-TNF refractory crohn’s disease: open label experience. Gastroenterology.2012;142(5):S-804.

24.Sandborn WJ, Feagan BG, Marano C, et al. Subcutaneous golimumab inducesclinical response and remission in patients with moderate-to-severe ulcerativecolitis. Gastroenterology. 2014;146(1):85-95.

25.Reich K, Ortonne JP, Gottlieb AB. Successful treatment of moderate to severeplaque psoriasis with the PEGylated Fab’ certolizumab pegol: results of a phase IIrandomized, placebo-controlled trial with a re-treatment extension. Br JDermatol. 2012;167(1):180-190.

26.Mease PJ, Fleischmann R, Deodhar AA, et al. Effect of certolizumab pegol onsigns and symptoms in patients with psoriatic arthritis: 24-week results of aphase 3 double-blind randomised placebo-controlled study (RAPID-PsA). Ann Rheum Dis. 2014;73(1):48-55.

27. Sandborn WJ, Feagan BG, Stoinov S, et al. Certolizumab pegol for the treatment of Crohn’s disease. N Engl J Med. 2007;357:228-238.

28.Lee SD, Osterman MT, Parrott SC, Wheat CL. 326 Assessment of the efficacy andsafety of certolizumab pegol for the treatment of moderate to severe ulcerativecolitis. Gastroenterology. 2012;142(5):S-75.

29.Papp KA, Tyring S, Lahfa M. A global phase III randomized controlled trial ofetanercept in psoriasis: safety, efficacy, and effect of dose reduction. Br JDermatol. 2005;152(6):1304-1312.

30.Mease PJ, Kivitz AJ, Burch FX, et al. Etanercept treatment of psoriaticarthritis: safety, efficacy, and effect on disease progression. ArthritisRheum. 2004;50:2264-2272.

31.Ritchlin C, Gottlieb A, McInnes I, et al. Ustekinumab in active psoriaticarthritis including patients previously treated with anti-TNF agents: resultsof a phase 3, multicenter, double-blind, placebo-controlled study. ArthritisRheum. 2014;73(6):990-999.

32.O’TooleA, Lucci M, Korzenik J. Inflammatory bowel disease provoked by etanercept:report of 443 possible cases combined from an IBD referral center and the FDA. J Dig Dis Sci. 2016;61:1772.

33. Papp KA, Langley RG, Lebwohl M, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23monoclonal antibody, in patients with psoriasis: 52-week results from arandomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet.2008;371(9625):1675-1684.

34.Gottlieb A, Menter A, Mendelsohn A, et al. Ustekinumab, a human interleukin12/23 monoclonal antibody, for psoriatic arthritis: randomised, double-blind,placebo-controlled, crossover trial. Lancet. 2009;373(9664):633-640.

35.Feagan B, Gasink C, Lang Y, et al. Late-breaking abstracts: OP054-LB4 Amulticenter, double-blind, placebo-controlled Ph3 study of ustekinumab, a humanmonoclonal antibody to IL-12/23P40, in patients with moderately-severely activeCrohn’sdisease who are na€ıve or not refractory to anti-TNFa: UNITI-2. UnitedEuropean Gastroenterol J. 2015;3(6):561-571.

36.Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in plaquepsoriasisdresults of two phase 3 trials. N Engl J Med. 2014;371(4):326-338.

37. McInnes IB, Mease PJ, Kirkham B, et al. Secukinumab, a human anti-interleukin-17A monoclonal antibody,in patients with psoriatic arthritis (FUTURE 2): a randomised, double-blind,placebo-controlled, phase 3 trial. Lancet. 2015; 386(9999):1137-1146.

38.Hueber W, Sands BE, Lewitzky S, et al. Secukinumab, a humananti-IL-17Amonoclonal antibody, formoderate to severe Crohn’s disease: unexpected results of a randomised,double-blind placebo-controlled trial. Gut. 2012;61(12):1693-1700.

39. van de Kerkhoff P, Griffiths C, Reich K, et al. Secukinumab long-term safety experience: a pooledanalysis of 10 phase II and III clinical studies in patients with moderate tosevere plaque psoriasis. J Am Acad Dermatol. 2015;373(26):2534-2548.

40.Gordon KB, Blauvelt A, Papp KA, et al. Phase 3 trials of ixekizumab inmoderate-to-severe plaque psoriasis. N Engl J Med. 2016;375(4):345-356.

41.Griffiths CE, Reich K, Lebwohl M, et al. Comparison of ixekizumab withetanercept or placebo in moderate-to severe psoriasis (UNCOVER-2 andUNCOVER-3): results from two phase 3 randomised trials. Lancet.2015;386(9993):541-551.

42.Travis SP, Higgins PD, Orchard T, et al. Review article: defining remission inulcerative colitis. Aliment Pharmacol Ther. 2011 Jul;34(2):113-124.

43. Peyrin-Biroulet L, Panes J, Sandborn WJ, et al. Defining disease severity in inflammatory bowel diseases:current and future directions. Clin Gastroenterol Hepatol. 2016;14(3):348-354.e17.

44.Celltrion. Inflectra (infliximab-dyyb): full prescribing information 2016.Available at: http://www.accessdata./drugsatfda_ docs/label/2016/125544s000lbl.pdf. Accessed November 12, 2017.

45.Amgen. Amjevita (adalimumab-atto): full prescribing information 2016. Availableat: http://www.accessdata./drug satfda_docs/label/2016/761024lbl.pdf.Accessed November 12, 2017.

46.Novartis Pharma. Erelzi (etanercept-szzs): full prescribing information 2016.Available at: http://pi./united_states/ enbrel/derm/enbrel_pi.pdf.Accessed November 12, 2017.

47.Janssen Biotech. Remicade (infliximab): full prescribing information 2010.Available at: https://www./shared/product/remicade/prescribing-information.pdf. Accessed November 12,2017.

48.Targan SR, Hanauer SB, van Deventer SJH, et al, for the Crohn’s Disease cA2 Study Group. A short-term study ofchimeric monoclonal antibody cA2 to tumor necrosis factor a for Crohn’s disease. N Engl J Med. 1997;337:1029-1035.

49. Hanauer SB, Feagan BG, Lichtenstein GR, et al. Maintenance infliximab for Crohn’s disease: the ACCENT I randomized trial. Lancet.2002;359:1541-1549.

50.Nast A, Jacobs A, Rosumeck S, et al. Efficacy and safety of systemic long-termtreatments for moderate-to-severe psoriasis: a systematic review andmeta-analysis. J Invest Dermatol. 2015 Nov;135(11):2641-2648.

51.Cawson MR, Mitchell SA, Knight C, et al. Systematic review, network meta-analysisand economic evaluation of biological therapy for the management of activepsoriatic arthritis. BMC Musculoskelet Disord. 2014;15:26.

52.Behm BW, Bickston SJ. Tumor necrosis factor-alpha antibody for maintenance ofremission in Crohn’s disease. Cochrane Database Syst Rev. 2008;(1):CD006893.

53. Hazlewood GS, Rezaie A, Borman M, et al. Comparative effectiveness of immunosuppressants andbiologics for inducing and maintaining remission in Crohn’s disease: a network meta-analysis. Gastroenterology.2015;148(2):344-354.e5.

54. Mei WQ, Hu HZ, Liu Y, et al. Infliximab is superior to other biological agents fortreatment of active ulcerative colitis: a meta-analysis. World J Gastroenterol.2015;21(19):6044-6051.

55.AbbVie. Humira (adalimumab): full prescribing information 2016. Available at:http://www./pdf/humira. pdf. Accessed November 12, 2017.

56. Menter A, Tyring SK, Gordon K, et al. Adalimumab therapy for moderate to severe psoriasis:a randomized, controlled phase III trial. J Am Acad Dermatol. 2007;58:106-115.

57.Gladman DD, Mease PJ, Ritchlin CT, et al. Adalimumab for long-term treatment ofpsoriatic arthritis: forty-eight week data from the adalimumab effectiveness inpsoriatic arthritis trial. Arthritis Rheum. 2007;56:476-488.

58.Colombel JF, Sandborn WJ, Rutgeerts P, et al. Adalimumab for maintenance ofclinical response and remission in patients with Crohn’s disease: The CHARM trial. Gastroenterology.2007;132:52-65.

59.Sandborn WJ, Hanauer SB, Rutgeerts P, et al. Adalimumab for maintenancetreatment of Crohn’s disease: results of the CLASSIC II trial. Gut.2007;56:1232-1239.

60.Reinisch W, Sandborn WJ, Hommes DW, et al. Adalimumab for induction of clinicalremission in moderately to severely active ulcerative colitis: results of arandomized controlled trial. Gut. 2011;60(6):780-787.

61.Jannsen Biotech. Simponi (golimumab): full prescribing information 2016.Available at: http://www./shared/product/simponi/prescribing-information.pdf. Accessed November 12, 2017.

62. Sandborn WJ, Feagan BG, Marano C, et al. Subcutaneous golimumab maintains clinical response inpatients with moderate-to-severe ulcerative colitis. Gastroenterology.2014;146(1):96-109.

63.Rutgeerts P, Feagan BG, Marano CW, et al. Randomized clinical trial: aplacebo-controlled study of intravenous golimumab induction therapy forulcerative colitis. Aliment Pharmacol Ther. 2015;42(5):504-514.

64.UCB. Cimzia (certolizumab pegol): full prescribing information 2016. Availableat: http://www./assets/pdf/ Prescribing_Information.pdf. AccessedNovember 12, 2017.

65.Schreiber S, Khaliq-Kareemi M, Lawrance IC, et al. Maintenance therapy withcertolizumab pegol for Crohn’s disease. N Engl J Med. 2007;357:239-250.

66. Sandborn WJ, Lee SD, Randall C, et al. Long-term safety and efficacy of certolizumab pegolin the treatment of Crohn’s disease: 7-year results from the PRECiSE 3 study.Aliment Pharmacol Ther. 2014;40(8):903-916.

67.Ramiro S, Smolen JS, Landewe R, et al. Pharmacological treatment of psoriaticarthritis: a systematic literature review for the 2015 update of the EULARrecommendations for the management of psoriatic arthritis. Ann Rheum Dis.2016;75(3): 490-498.

68.Amgen. Enbrel (etanercept): full prescribing information 2013. Available at:http://pi./united_states/enbrel/derm/ enbrel_pi.pdf. Accessed November12, 2017.

69.Tyring S, Gottlieb A, Papp K, et al. Etanercept and clinical outcomes, fatigue,and depression in psoriasis: double-blind placebo-controlled randomized phaseIII trial. Lancet. 2006; 367:29-35.

70.Mease PJ, Goffe BS, Metz J, et al. Etanercept in the treatment of psoriaticarthritis and psoriasis: a randomized trial. Lancet. 2000;356:385-390.

71.Sandborn WJ, Hanauer SB, Katz S, et al. Etanercept for active Crohn’s disease: a randomized, double-blind,placebocontrolled trial. Gastroenterology. 2001;121(5):1088-1094.

72.Janssen Biotech. Stelara (ustekinumab): full prescribing information 2016.Available at: https://www./ pdf/prescribinginformation.pdf.Accessed November 12, 2017.

73. Krueger GG, Langley RG, Leonardi C, et al. A human interleukin-12/23 monoclonal antibody for thetreatment of psoriasis. N Engl J Med. 2007;356(6):580-592.

74. Feagan B, Sandborn W, Gasink C, et al. Ustekinumab as induction and maintenance therapy forCrohn’sdisease. N EnglJ Med. 2016;375:1946-1960.

75. Sandborn W, Gasink C, Gao L, et al. Ustekinumab induction and maintenance therapy inrefractory Crohn’s disease. N Engl J Med. 2012;367(16):1519-1528.

76. Sands B, Han C, Gasink C, et al. Tu2006 ustekinumab improves general health status anddisease-specific health related quality of life of patients with moderate tosevere crohn’s disease: results from the UNITI and IMUNITI phase 3clinical trial. Gastroenterology. 2016;150(4):S1004.

77.Novartis Pharma. Cosentyx (secukinumab): full prescribing information 2016.Available at: https://www.pharma.us./sites/www.pharma.us./files/cosentyx.pdf. Accessed November 12, 2017.

78. Ohtsuki M, Morita A, Abe M, et al. Secukinumab efficacy and safety in Japanese patientswith moderate-to-severe plaque psoriasis: subanalysis from ERASURE, arandomized, placebocontrolled, phase 3 study. J Dermatol.2014;41(12):1039-1046.

79. Gottlieb AB, Langley RG, Philipp S, et al. Secukinumab Improves physical function in subjectswith plaque psoriasis and psoriatic arthritis: results from two randomized,phase 3 trials. J Drugs Dermatol. 2015;14(8):821-833.

80. Baeten D, Sieper J, Braun J, et al. Secukinumab, an interleukin-17A inhibitor, inankylosing spondylitis. N Engl J Med. 2015;373:2534-2548.

81.Eli Lilly. Taltz (ixekizumab): full prescribing information 2016. Available at:http://pi./us/taltz-uspi.pdf. Accessed November 12, 2017.

82. Mease P, van der Heijde D, Ritchlin CT, et al. Ixekizumab, an interleukin-17A specific monoclonalantibody, for the treatment of biologic-naive patients with active psoriaticarthritis: results from the 24-week randomised, double-blind, placebo-controlledand active (adalimumab)-controlled period of the phase III trial SPIRIT-P1. AnnRheum Dis. 2017; 76:79-87.

83.Langley R, Romiti R, Leonardi C, et al. Evaluation of inflammatory boweldisease in patients with plaque psoriasis studied in phase 3 trials ofixekizumab (UNCOVER trials): adjudicated data from the induction period. J AmAcad Dermatol. 2016;74(5):AB24.

84.Valeant. Siliq (brodalumab): full prescribing information 2017. Available at:http://www./Portals/25/Pdf/PI/ Siliq-pi.pdf. Accessed November 12,2017.

85. Lebwohl M, Strober B, Menter A, et al. Phase 3 studies comparing brodalumab with ustekinumabin psoriasis. N Engl J Med. 2015;373(14):1318-1328.

86. Papp KA, Reich K, Paul C, et al. A prospective phase III, randomized, double-blind,placebo-controlled study of brodalumab in patients with moderate-to-severeplaque psoriasis. Br J Dermatol. 2016;175(2):273-286.

87.Attia A, Abushouk A, Ahmed H, et al. Safety and efficacy ofbrodalumabformoderate-to-severeplaquepsoriasis:Asystematic review andmeta-analysis. Clin Drug Investig. 2017;37(5):439-451.

88.Mease PJ, Genovese MC, Greenwald MW, et al. Brodalumab, an anti-IL17RAmonoclonal antibody, in psoriatic arthritis. N Engl J Med.2014;370(24):2295-2306.

89.Nakagawa H, Niiro H, Ootaki K. Brodalumab, a human anti-interleukin-17-receptorantibody in the treatment of Japanese patients with moderate-to-severe plaquepsoriasis: Efficacy and safety results from a phase II randomized controlledstudy. J Dermatol Sci. 2016;81(1):44-52.

90.Targan SR, Feagan B, Vermeire S, et al. A randomized, double-blind,placebo-controlled phase 2 study of brodalumab in patients withmoderate-to-severe Crohn’s disease. Am J Gastroenterol.2016;111(11):1599-1607.

91.Janssen Biotech. Tremfya (guslekumab): full prescribing information. 2017.Available at: https://www./ pdf/PrescribingInformation.pdf.Accessed November 12, 2017.

92. Blauvelt A, Papp KA, Griffiths CE, et al. Efficacy and safety of guselkumab, ananti-interleukin-23 monoclonal antibody, compared with adalimumab for thecontinuous treatment of patients withmoderate tosevere psoriasis: Resultsfromthe phase III, double-blinded, placebo- and active comparator-controlledVOYAGE 1 trial. J AmAcad Dermatol. 2017;76(3):405-417.

93.Reich K, Armstrong A, Foley P, et al. Efficacy and safety of guselkumab, ananti-interleukin-23 monoclonal antibody, compared with adalimumab for thetreatment of patients with moderate to severe psoriasis with randomizedwithdrawal and retreatment: Results from the phase III, double-blind, placebo-and active comparator-controlled VOYAGE 2 trial. J Am Acad Dermatol.2017;76(3):418-431.

94.Dogra S, Jain A, Kanwar AJ. Efficacy and safety of acitretin in three fixeddoses of 25, 35 and 50 mg in adult patients with severe plaque type psoriasis:a randomized, double blind, parallel group, dose ranging study. J Eur AcadDermatol Venereol. 2013;27(3):e305-e311.

95.Heydendael VM, Spuls PI, Opmeer BC, et al. Methotrexate versus cyclosporine inmoderate-to-severe chronic plaque psoriasis. N Engl J Med. 2003;349:658-665.

96. Salvarani C, Macchioni P, Olivieri I, et al. A comparison of cyclosporine, sulfasalazine, andsymptomatic therapy in the treatment of psoriatic arthritis. J Rheumatol. 2001;28:2274-2282.

97. Brynskov J, Freund L, Rasmussen SN, et al. A Placebo-Controlled, Double-Blind, Randomized Trialof Cyclosporine Therapy in Active Chronic Crohn’s Disease. N Engl J Med. 1989;321:845-850.

98.McDonald J, Feagan B, Jewell D, et al. Cyclosporine for induction of remissionin Crohn’s disease. Cochrane Database Syst Rev.2005;(2):CD000297.

99.Lichtiger S, Present DH, Kornbluth A, et al. Cyclosporine in severe ulcerativecolitis refractory to steroid therapy. N Engl J Med. 1994;330:1841-1845.

100. Arts J, D’Haens G, Zeegers M, et al. Long-term outcome of treatment with intravenouscyclosporin in patients with severe ulcerative colitis. Inflamm Bowel Dis. 2004;10:73-78.

101. D’Haens G, Lemmens L, Geboes K, etal. Intravenous cyclosporine vs.intravenous corticosteroids as single therapy for severe attacks of ulcerativecolitis. Gastroenterology. 2001; 120:1323-1329.

102.Saurat JH, Stingl G, Dubertret L, et al. Efficacy and safety results from therandomized controlled comparative study of adalimumab vs methotrexate vsplacebo in patients with psoriasis (CHAMPION). Br J Dermatol. 2008;158:558-566.

103.Coates LC, Helliwell PS. Methotrexate Efficacy in the Tight Control inPsoriatic Arthritis (TICOPA) study. J Rheumatol. 2016;43(2):356-361.

104.Willkens RF,Williams HJ,Ward JR, et al.Randomized,double-blind, placebocontrolled trial of low-dose pulse methotrexate in psoriatic arthritis.Arthritis Rheum. 1984;27:376-381.

105.Kingsley GH, Kowalczyk A, Taylor H, et al. A randomized placebo-controlledtrial of methotrexate in psoriatic arthritis. Rheumatology (Oxford, England).2012;51(8):1368-1377. 106. West J, Ogston S, Foerster J. Safety and efficacy ofmethotrexate in psoriasis: a meta-analysis of published trials. PLoS One.2016;11(5):e0153740.

107.Feagan BG, Rochon J, Fedorak RN, et al. Methotrexate for the treatment of Crohn’s disease. N Engl JMed. 1995;332:292-297.

108.Carbonell F, Colombel JF, Filippi J, et al. Methotrexate is not superior toplacebo for inducing steroid-free remission, but induces steroid-free clinicalremission in a larger proportion of patients with ulcerative colitis.Gastroenterology. 2016;150(2): 380-388.e4.

109.Papp K, Reich K, Leonardi CL, et al. Apremilast, an oral phosphodiesterase 4(PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: resultsof a phase III, randomized, controlled trial (Efficacy and Safety TrialEvaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am AcadDermatol. 2015;73(1):37-49.

110.Paul C, Cather J, Gooderham M, et al. Efficacy and Safety of Apremilast, anOral Phosphodiesterase 4 Inhibitor, in Patients with Moderate-to-Severe PlaquePsoriasis over 52 Weeks: a phase III, randomized Controlled Trial (ESTEEM 2).Br J Dermatol. 2015;173(6):1387-1399.

111.Connetics. Soriatane (acitretin): full prescribing information 2004. Availableat: http://www.accessdata./drugsatfda_docs/ label/2004/19821s011lbl.pdf.Accessed November 12, 2017.

112.Olsen EA, Weed WW, Meyer CJ, Cobo LM. A double-blind, placebo-controlled trialof acitretin for the treatment of psoriasis. J Am Acad Dermatol.1989;21:681-686.

113. Haushalter K, Murad EJ, Dabade TS, et al. Efficacy of low-dose acitretin in the treatment ofpsoriasis. J Dermatolog Treat. 2012 Dec;23(6):400-403.

114.Novartis Pharma. Neoral (cyclosporine):full prescribing information 2015.Available at: https://www.pharma.vartis. com/product-list. Accessed November 12, 2017.

115. Ellis CN, Fradin MS, Messana JM, et al. Cyclosporine for plaque-type psoriasis: results of amultidose, double-blind trial. N Engl J Med. 1991;324(5):277-284.

116. Mahrle G, Schulze HJ, Brautigam M, et al. Anti-inflammatory efficacy of low-dose Cyclosporin Ain psoriatic arthritis: a prospectivemulticenter study. Br J Dermatol. 1996;135:752-757.

117. Fraser AD, van Kuijk AW, Westhovens R, et al. A randomized, double blind, placebocontrolled,multicenter trial of combination therapywithmethotrexatepluscyclosporin in patientswith active psoriatic arthritis. Ann Rheum Dis. 2005;64:859-864.

118. Faerber L, Braeutigam M, Weidinger G, et al. Cyclosporine in severe psoriasis: results of ameta-analysis in 579 patients. Am J Clin Dermatol. 2001;2:41-47.

119. Ash Z, Gaujoux-Viala C, Gossec L, et al. A systematic literature review of drug therapies forthe treatment of psoriatic arthritis: current evidence and meta analysisinforming the EULAR recommendations for the management of psoriatic arthritis. Ann Rheum Dis.2012;71(3):319-326.

120. Feuerstein JD, Akbari M, Tapper EB, et al. Systematic review and meta-analysis of third-linesalvage therapy with infliximab or cyclosporine in severe ulcerative colitis. Ann Gastroenterol.2016;29(3):341-347.

121. Feagan BG, Fedorak RN, Irvine EJ, et al. A comparison of methotrexate with placebo for themaintenance of remission in Crohn’s disease. N Engl J Med. 2000;342(22): 1627-1632.

122. Oren R, Arber N, Odes S, et al. Methotrexate in chronic active ulcerative colitis: adouble-blind, randomized, Israeli multicenter trial. Gastroenterology.1996;110:1416-1421.

123.Patel V, Wang Y, MacDonald JK, et al. Methotrexate for maintenance of remissionin Crohn’s disease. Cochrane Database Syst Rev.2014;(8):CD006884.

124.Chande N, Wang Y, MacDonald JK, et al. Methotrexate for induction of remissionin ulcerative colitis. Cochrane Database Syst Rev. 2014;(8):CD006618.

125.Celgene. Otezla (apremilast): full prescribing information 2015. Available at: https://www./content/uploads/otezla-pi.pdf. Accessed November 12, 2017.

126. Kavanaugh A, Mease PJ, Gomez-Reino JJ, et al. Treatment of psoriatic arthritis in a phase 3randomised, placebo-controlled trial with apremilast, an oral phosphodiesterase4 inhibitor. Ann Rheum Dis. 2014;73(6):1020-1026.

127.Akhyani M, Chams-Davatchi C, Hemami MR, et al. Efficacy and safety ofmycophenolate mofetil vs. methotrexate for the treatment of chronic plaquepsoriasis. J Eur Acad Dermatol Venereol. 2010;24(12):1447-1451.

128. Grundmann-Kollmann M, Mooser G, Schraeder P, et al. Treatment of chronic plaque-stage psoriasis andpsoriatic arthritis with mycophenolate mofetil. J Am Acad Dermatol. 2000;42(5Pt 1):835-837.

129.Tan T, Lawrance IC. Use of mycophenolate mofetil in inflammatory bowel disease.World J Gastroenterol. 2009; 15(13):1594-1599.

130.Neurath M, Wanitschke R, Peters M, et al. Randomised trial of mycophenolatemofetil versus azathioprine for treatment of chronic active Crohn’s disease. Gut. 1999;44(5): 625-628.

131.GuptaAK, Ellis CN, SiegelMT, et al. Sulfasalazine improvespsoriasis: adouble-blind analysis. Arch Dermatol. 1990;126:487-493.

132.Clegg DO, Reda DJ, Mejias E, et al. Comparison of sulfasalazine and placebo inthe treatment of psoriatic arthritis: a Department of Veterans Affairscooperative study. Arthritis Rheum. 1996;39(12):2013-2020.

133.Misiewicz JJ, Lennard, Jones JE, et al. Controlled trial of sulphasalazine inmaintenance therapy for ulcerative colitis. Lancet. 1965;1:185-188.

134.Dissanayake AS, Truelove SC. A controlled therapeutic trial of long-termmaintenance treatment of ulcerative colitis with sulphasalazine (Salazopyrin).Gut. 1973;14(12):923-926.

135.Layton AM, Sheehan-Dare RA, Goodfield MJ, Cotterill JA. Hydroxyurea in themanagement of therapy resistant psoriasis. Br J Dermatol. 1989;121(5):647-653.

136.Kumar B, Saraswat A, Kaur I. Rediscovering hydroxyurea: its role inrecalcitrant psoriasis. Int J Dermatol. 2001;40(8):530-534.

137. Ranjan N, Sharma NL, Shanker V, et al. Methotrexate versus hydroxycarbamide (hydroxyurea) asa weekly dose to treat moderate-to-severe chronic plaque psoriasis: acomparative study. J Dermatolog Treat. 2007;18(5):295-300.

138.Du Vivier A, Munro DD, Verbov J. Treatment of psoriasis with azathioprine. BrMed J. 1974;1(5897):49-51.

139.Greaves MW, Dawber R. Azathioprine in psoriasis. Br Med J.1970;2(5703):237-238.

140.Lee JC, Gladman DD, Schentag CT, Cook RJ. The long-term use of azathioprine inpatients with psoriatic arthritis. J Clin Rheumatol. 2001;7:160-165.

141.Candy S, Wright J, Gerber M, et al. A controlled double blind study ofazathioprine in the management of Crohn’s disease. Gut. 1995;37:674-678.

142.Willoughby JM, Beckett J, Kumar PJ, Dawson AM. Controlled trial of azathioprinein Crohn’s disease. Lancet. 1971;2:944-947.

143.Rosenberg JL, Levin B, Wall AJ, Kirsner JB. A controlled trial of azathioprinein Crohn’s disease. Am J Dig Dis. 1975;20:721-726.

144.Kirk AP, Lennard-Jones JE. Controlled trial of azathioprine in chroniculcerativecolitis. BrMedJ (Clin Res Ed).1982;284:1291-1292.

145.Sood A, Midha V, Sood N, Kaushal V. Role of azathioprine in severe ulcerativecolitis: one-year, placebo-controlled, randomized trial. Indian JGastroenterol. 2000;19(1):14-16.

146.Mantzaris GJ, Sfakianakis M, Archavlis E, et al. A prospective randomizedobserver-blind 2-year trial of azathioprine monotherapy vs. azathioprine andolsalazine for the maintenance of remission of steroid-dependent ulcerativecolitis. Am J Gastroenterol. 2004;99:1122-1128.

147.Zackheim HS, Maibach HI. Treatment of psoriasis with 6-thioguanine. Australas JDermatol. 1988;29(3):163-167.

148.Zackheim HS, Glogau RG, Fisher DA, Maibach HI. 6-Thioguanine treatment ofpsoriasis: experience in 81 patients. J Am Acad Dermatol. 1994;30(3):452-458.

149.Mason C, Krueger GG. Thioguanine for refractory psoriasis: a 4-year experience.J Am Acad Dermatol. 2001;44(1):67-72.

150.Herrlinger KR, Kreisel W, Schwab M, et al. 6-thioguanineeefficacy and safety inchronic active Crohn’s disease. Aliment Pharmacol Ther.2003;17(4):503-508.

151.Ansari A, Elliott T, Fong F, et al. Further experience with the use of6-thioguanine in patients with Crohn’s disease. Inflamm Bowel Dis. 2008;14:1399-1405.

152. Teml A, Schwab M, Harrer M, et al. A prospective, open-label trial of 6-thioguanine inpatients with ulcerative or indeterminate colitis. Scand J Gastroenterol.2005;40(10):1205-1213.

153. Bos JD, Witkamp L, Zonnevald IM, et al. Systemic tacrolimus (FK 506) is effective for thetreatment of psoriasis in a double-blind, placebo-controlled study: theEuropean FK 506 multicenter psoriasis study group. Arch Dermatol.1996;132:419-423.

154.Lythgoe M, Abraham S. Tacrolimus: an effective treatment in refractorypsoriatic arthritis following biologic failure. Clin Exp Rheumatol. 2016;34(1suppl 95):S12-S13.

155.Fellermann K, Ludwig D, StahlM, et al. Steroid-unresponsive acute attacks ofinflammatory bowel disease: immunomodulation by tacrolimus (FK506). Am JGastroenterol. 1998;93:1860-1866.

156.Ogata H, Matsui T, Nakamura M, et al. A randomised dose finding study of oraltacrolimus (FK506) therapy in refractory ulcerative colitis. Gut.2006;55:1255-1262.

157.Kaltwasser JP, Nash P, Gladman D, et al. Efficacy and safety of leflunomide inthe treatment of psoriatic arthritis and psoriasis: a multinational,double-blind, randomized, placebo-controlled clinical trial. Arthritis Rheum.2004;50(6):1939-1950.

158.Holtmann MH, Gerts AL, Weinman A, et al. Treatment of Crohn’s disease with leflunomide as second-lineimmunosuppression: a phase 1 open-label trial on efficacy, tolerability andsafety. Dig DisSci. 2008;53(4):1025-1032.

159. Altmeyer PJ, Matthes U, Pawlak F, et al. Antipsoriatic effect of fumaric acid derivatives:results of a multicenter double-blind study in 100 patients. J Am AcadDermatol. 1994;30:977-981.

160.Fallah Arani S, Neumann H, Hop WC, Thio HB. Fumarates vs. methotrexate inmoderate to severe chronic plaque psoriasis: a multicentre prospectiverandomized controlled clinical trial. Br J Dermatol. 2011;164(4):855-861.

161. Peeters AJ, Dijkmans BA, van der Schroeff JG. Fumaric acid therapy for psoriatic arthritis: arandomized, double-blind, placebo-controlled study. Br J Rheumatol. 1992;31:502-504.

http://dx./10.1016/j.jaad.2017.06.043

JAm Acad Dermatol 2018;78:383-94.© 2017 by the American Academy of Dermatology,Inc


    本站是提供个人知识管理的网络存储空间,所有内容均由用户发布,不代表本站观点。请注意甄别内容中的联系方式、诱导购买等信息,谨防诈骗。如发现有害或侵权内容,请点击一键举报。
    转藏 分享 献花(0

    0条评论

    发表

    请遵守用户 评论公约

    类似文章 更多