中国科技论文统计源期刊 中文核心期刊  
美国《化学文摘》《国际药学文摘》
《乌利希期刊指南》
WHO《西太平洋地区医学索引》来源期刊  
日本科学技术振兴机构数据库(JST)
第七届湖北十大名刊提名奖  
医药导报, 2019, 38(5): 544-550
doi: 10.3870/j.issn.1004-0781.2019.05.002
替加环素临床应用剖析*
Critical Review on Clinical Application of Tigecycline
肖婷婷, 肖永红

摘要:

替加环素是一种广谱抗菌药物,被誉为抗感染治疗的最后选择,于2011年进入我国临床后,被广泛用于多重耐药菌感染的治疗,其已获批的适应证包括成人复杂性腹腔感染、复杂性皮肤和皮肤软组织感染以及社区获得性肺炎。然而,近几年众多临床研究与荟萃分析结果引起了人们对其临床疗效和安全性的关注与质疑,该文就有关内容进行客观阐述,以供临床应用参考。

关键词: 替加环素 ; 耐药菌 ; 临床应用 ; 病死率

Abstract:

Tigecycline,a broad-spectrum antibiotic known as the last resort for the treatment of multiple drug resistance bacterial infections,has been widely used in clinic since its approval in China in 2011. It is currently approved for complicated intra-abdominal infections,complicated skin and skin structure infections,and community-acquired pneumonia.However,numerous clinical studies and meta-analyses have raised concerns about its clinical efficacy and safety.Therefore,in this review,we objectively analyze the researches of tigecycline in recent years,including the mechanism of action and clinical application,which may provide references for clinical application.

Key words: Tigecycline ; Drug-resistant bacteria ; Clinical application ; Mortality

细菌耐药已经成为全球严峻的公共卫生挑战,尤其是泛耐药的肺炎克雷伯菌、鲍曼不动杆菌等,临床对这类细菌感染缺乏有效的抗菌药物[1,2]。2017年2月27日,世界卫生组织(WHO)发布了目前急需新药研发的20种耐药细菌清单,如碳青霉烯类耐药肠杆菌科(Carbapenem-resistant Enterobacteriaceae,CRE)。

替加环素(tigecycline)作为一种新型甘氨酰环素类抗菌药物于2005年6月被美国食品药品管理局(FDA)批准上市,用于治疗成人复杂性腹腔感染和复杂性皮肤及皮肤软组织感染,2008年批准用于治疗社区获得性肺炎[3]。目前全球共有50多个国家批准和上市了替加环素。替加环素于2011年末在中国批准上市,用于3个适应证[4]。但是基于体外抗菌活性结果,替加环素也被广泛用于治疗多重耐药菌(MDR)引起的医院内获得性肺炎(hospital acquired pneumonia,HAP)、呼吸机相关性肺炎(ventilator associated pneumonia,VAP)和血流感染(bloodstream infection,BSI)等[5,6]。这些临床应用大多属于超适应证使用,缺乏严格的临床研究结果支持,无法证明其有效性和安全性。相反,近年来部分文献报道,替加环素的应用会增加这些感染患者的病死率[7,8]。替加环素作为新一代抗菌药物给患者带来希望的同时又有些困惑,笔者将国内外相关文献进行归纳整理,关注替加环素的相关作用机制、药动学/药效学(PK/PD),客观分析其临床疗效及安全性,为临床合理应用替加环素提供参考依据。

1 抗菌作用与耐药现状

作为一种广谱抗菌药物,替加环素对革兰阴性需氧菌如肠杆菌科细菌、大多数革兰阳性需氧菌包括耐甲氧西林葡萄球菌(MRS)、厌氧菌和非典型病原体均具有较好的体外抗菌活性[9]。替加环素主要作用机制类似于其他四环素类药物:通过与核糖体30S亚基结合并阻止氨酰化tRNA分子进入核糖体A位,抑制细菌蛋白质翻译(肽链延长)。初期的研究提示替加环素作为在米诺环素9位分子上添加叔丁基甘氨酞胺基团而衍生的一种新型四环素类抗菌药物,与其他四环素或抗菌药物不易产生交叉耐药,能够克服或限制核糖体保护和外排泵这两种四环素耐药机制[10]

随着替加环素的临床利用逐年增加,世界范围内革兰阴性菌中替加环素耐药逐渐发生并呈增加趋势,常见于鲍曼不动杆菌、肠杆菌科细菌[11]。PFALLER等[12]收集了2016年来自亚太地区(3443株分离株)、欧洲(13 530株分离株)、拉丁美洲(3327株)的33 348份非重复菌株,进行替加环素耐药监测,结果显示CRE对替加环素的耐药率为2%,碳青霉烯类药物不敏感鲍曼不动杆菌对替加环素的耐药率高达25.6%。

细菌对替加环素耐药主要有以下4种机制[13]:①RND型外排系统过表达,肠杆菌科主要是ramR失活和ramA上调的突变引起AcrAB外排泵活性增加,导致替加环素抗性;鲍曼不动杆菌adeS和adeR基因的点突变或者在adeS 基因前插入序列ISAba1,引起AdeABC外排泵过度表达,导致鲍曼不动杆菌对替加环素敏感性下降。②核糖体蛋白结构变化,如核糖体蛋白S10结构的变化可引起鲍曼不动杆菌、粪肠球菌、金黄色葡萄球菌等对替加环素敏感性下降。③细菌细胞膜的改变,如替加环素耐药的大肠埃希菌中发现了脂多糖中核心多糖生物合成途径的相关基因(lpcA、rfaE、rfaD、rfaC和 rfaF)的突变。④各种酶类,如RecA蛋白酶、甲基转移酶等;在替加环素不敏感鲍曼不动杆菌中亦检出tetX1基因编码的依赖黄素的单加氧酶TetX,后者可羟化替加环素,从而产生耐药。

2 PK/PD

替加环素静脉注射后呈线性PK特点[14]。在健康受试者的I期临床试验中,推荐剂量为首剂静脉注射给予100 mg,后每12 h(q12h)50 mg续注。替加环素峰值血清稳态浓度(Cmax)可达(866± 233)μg·L-1,半衰期为37~67 h,血药浓度-时间曲线下面积(AUC) 为300~580 μg·h·L-1。替加环素具有良好的组织穿透力,能广泛分布于身体各组织器官,其稳态分布容积约为7.2~8.6 L·kg-1。研究表明,替加环素在肺、骨骼、肝、脾、肾和皮肤等部位有较好的分布[15]。药物主要通过胆汁/粪便排泄消除[16]。其总剂量的22%以原型经尿液排泄代谢产物,肾功能不全(包括血液透析)患者无需调整剂量;但建议在严重肝功能障碍(Child Pugh C)患者中需要调整剂量(首剂100 mg后维持剂量为25 mg,q12h),并密切关注后续情况。

替加环素与华法林合用时会降低后者的清除率,故需监测凝血酶原时间,并进行抗凝检测。与地高辛联用时两者剂量均不需调整。替加环素不抑制以下6种细胞色素P450(CYP)的亚型:1A2、2C8、2C9、2C19、2D6和3A4,与此相关药物不会影响替加环素的清除。目前未报道替加环素具有明显的药物相互作用,体外研究显示替加环素与其他常用抗菌药物之间没有拮抗作用,并显示出与某些抗生素可能具有协同作用,如替加环素联合阿米卡星对40%~100%肠杆菌属、肺炎克雷伯菌、变形杆菌属具有的协同作用[17]

替加环素属于时间依赖性抗菌药并且有较长的抗菌药物后效应,因此AUC/MIC常作为替加环素疗效评价的PK/PD指标。BHAVNANI等[18]研究结果显示,替加环素fAUC0-24 h/MIC与临床疗效和微生物根除率有关。当fAUC0-24 h/MIC>0.9时,78%的HAP能取得满意的临床疗效;当fAUC0-24 h/MIC>0.35 时,77.8%的患者感染部位的微生物根除率高。但其常规剂量所能达到的最大稳态血药浓度远低于常用的试验药敏折点标准(≤2 mg·L-1),且其为抑菌剂,可导致细菌的不完全清除,从而造成临床疗效不理想。

有关儿童的PK/PD研究较少,一项纳入58例年龄为8~11岁的严重感染患儿接受替加环素多剂量梯度(0.75,1.00,1.25 mg·kg-1,q12h,最大剂量不超过50 mg)的II期多中心的临床研究,提示替加环素剂量为1.2 mg·kg-1,q12h可至适当的AUC/MIC水平,从而达到理想的治愈率[19]

3 临床研究现状与挑战
3.1 替加环素抗耐药菌作用

目前,替加环素主要被用于多重/泛耐药(MDR/XDR)菌株感染的患者群体中,如产碳青霉烯酶的肺炎克雷伯菌、产I型新德里金属β-内酰胺酶菌、碳青霉烯类耐药鲍曼不动杆菌感染等,属于超适应证使用,大多缺乏严格临床研究依据。

3.1.1 CRE感染 CRE存在多种耐药机制,对临床常用抗菌药物广泛耐药,常常仅对多粘菌素和替加环素呈现较高体外敏感性[20]。有多篇临床研究表明替加环素与其他抗菌药物联合治疗产碳青霉烯酶细菌感染有一定疗效,NI等[21]系统性评价了26项临床研究,发现替加环素组在总死亡率方面与对照组差异无统计学意义(P=0.73),亚组分析显示接受替加环素联合治疗的患者30 d死亡率明显低于接受单药治疗(P=0.03)和其他抗生素治疗方案的患者(P=0.01);此外,对重症监护室(ICU)患者病死率而言,大剂量替加环素方案与标准剂量方案显著不同(P=0.006);研究结果表明,替加环素治疗CRE感染的疗效与其他抗生素相似,替加环素联合治疗和高剂量方案可能分别比单一疗法和标准剂量方案更有效。同时也有报道称替加环素单药治疗CRE所致严重感染的疗效差,患者具有较高的死亡风险,可能与其不理想的药动学特征有关[22]。因此,FDA不推荐常规使用该类药物,只有当细菌几乎全耐药时,可基于其体外敏感性,推荐与多黏菌素类、碳青霉烯类或氨基苷类等联合应用[3]

3.1.2 多重/泛耐药鲍曼不动杆菌(MDR/XDRAB)感染 2016中国CHINET监测提示鲍曼不动杆菌碳青霉烯耐药率接近70%,泛耐药鲍曼不动杆菌达到18.7%,后者仅对替加环素或者多粘菌素敏感率较高[20]。王佳等[23]纳入10篇关于替加环素联合头孢哌酮/舒巴坦钠治疗MDR/XDRAB所致的肺炎疗效的随机、对照试验(RCTs),结果显示替加环素联合头孢哌酮/舒巴坦钠组明显高于单用头孢哌酮/舒巴坦钠组;联合用药组细菌清除率也显著高于单用头孢哌酮/舒巴坦钠组。同时需注意替加环素耐药鲍曼不动杆菌也日益增多,其耐药机制主要是AdeABC外排泵系统的过度表达。目前认为替加环素联合治疗方案是治疗替加环素耐药鲍曼不动杆菌的主要选择,LI等[24]进行了1159株菌株的Meta分析得出,替加环素与多粘菌素、阿米卡星等体外联合药敏及时间杀菌曲线提示联合用药对于耐药菌株具有一定的杀菌作用。国内对于多重耐药革兰阴性菌感染治疗的专家共识中认为对于MDR/XDRAB感染可选用替加环素,但建议联合用药[25]

3.2 替加环素已经获批的适应证临床研究现状

替加环素国内已获批准的适应证包括:18岁(含)以上由敏感菌株引起的成人复杂性腹腔内感染(complicated intraperitoneal infection,cIAI)、18岁(含)以上由敏感菌株引起的成人复杂性皮肤和皮肤软组织感染(complicated skin and skin soft tissue infections,cSSSI),社区获得性细菌性肺炎(community acquired bacterial pneumonia,CAP)。治疗cIAI及cSSSI推荐疗程5~14 d,治疗CAP推荐7~14 d;实际疗程应根据患者的临床感染的严重程度及部位和细菌学进展情况而定。

3.2.1 cIAI cIAI常由多种细菌尤其是耐药菌引起,临床常给予患者广谱抗生素治疗。国内外已在替加环素治疗成人cIAI进行了数个多中心的随机双盲的临床研究,提示替加环素与亚胺培南/西司他丁治疗效果相似,并且在单病原体和多病原体感染间差异无统计学意义[26,27]。陈昭燕等[28]对5项共计4185例患者的关于替加环素对比IMI/CIS治疗复杂腹腔感染的随机对照试验分析,发现TG并不比IMI/CIS显著提高cIAI临床疗效,但增加了胃肠道不良反应与二次感染的发生率。2017年美国外科感染协会修订了腹腔感染治疗指南指出[29]:在大多数情况下,不建议使用替加环素进行经验性治疗(1-B级);如果是耐药菌感染的成年患者,无敏感药物的情况下可考虑使用含替加环素的联合治疗方案(2-B级)。综合国内外研究成果发现,发生cIAI时,应优先选用敏感药物,如碳青霉烯类等广谱抗生素,若无敏感药物可选时可选择替加环素治疗。

3.2.2 cSSSI cSSSI常需外科手术治疗,目前常规抗生素治疗方案为万古霉素和氨曲南联用。ELLIS-GROSSE 等[30]对cSSSI住院成人患者进行的两项III期双盲研究,结果显示不良事件相似,替加环素组恶心和呕吐反应发生率增加,万古霉素-氨曲南组皮疹出现率高和肝转氨酶水平升高;替加环素单药治疗与万古霉素-氨曲南组合治疗cSSSI患者一样安全有效。但LAUF 等[31]的III期随机双盲试验纳入了糖尿病足患者944例,分为TG组(每日大剂量静脉注射替加环素150 mg)与厄他培南组(每日静脉注射1 g或联合万古霉素),两组治愈率分别为77.5%和82.5% ;合并骨髓炎的糖尿病组患者中,TG组治愈率低于36%,且胃肠道不良反应发生率高,其可能与疗程不足有关。

3.2.3 CAP DARTOIS等[32]对574例成人CAP随机双盲对照的III期临床研究结果显示,替加环素组与左氧氟沙星组的疗效接近。对两个CAP感染临床试验组的患者进行分析发现,替加环素fAUC0-24 h/MIC≥12.8组的HAP患者临床疗效好(退热更快)(P=0.05);同时多变量逻辑回归模型证明替加环素AUC高于阈值6.87 mg·h·L-1和女性是恶心或呕吐等不良反应的危险因素(P=0.004)[33]。2016年中国CAP指南中指出应根据患者年龄、疾病严重程度及药物敏感性等综合评估,选择恰当的抗感染药物;替加环素可作为产超广谱β-内酰胺酶肠杆菌科、高产AmpC酶肠杆菌科、产碳青霉烯酶肠杆菌科以及不动杆菌属的次选抗感染药物[34]

3.3 替加环素超适应证应用研究

由于替加环素具有广谱的微生物覆盖性,体外药敏试验常呈敏感,故临床仍用其来治疗泛耐药菌引起的各种感染,如HAP、VAP和严重复杂性难治性艰难梭菌性肠炎(severe complex and refractory clostridium difficile enteritis,sscCDI)。但有关临床研究结果不完全一致,国内外指南的推荐也存在差异。

3.3.1 HAP/VAP HAP患者中最常见的病原体是鲍曼不动杆菌、大肠埃希菌、铜绿假单胞菌和耐甲氧西林金黄色葡萄球菌,为了提高临床治愈率并降低病死率,在HAP管理指南中强调了恰当的广谱抗生素治疗的重要性。目前关于替加环素治疗HAP的疗效仍具争议。 2010年9月FDA发布了安全公告,指出HAP患者替加环素治疗组病死率上升。PRASAD等[35]对13篇临床研究分析发现,替加环素会增加患者病死率[风险差异(RD)0.7%;95%CI(0.1%,1.2%);P=0.01]和非治愈率[RD 2.9%;95%CI(0.6%,5.2%);P=0.01]。随之2013年9月,FDA要求替加环素说明书添加黑框警告,不推荐常规使用该类药物治疗HAP,只有当细菌几乎全耐药时,可基于替加环素具有较高的体外敏感率,推荐与多黏菌素类、碳青霉烯类或氨基苷类等联合应用[3]。2018年AMBARAS等[36]综述了IDSA/ATS 和JAID/JSC等相关指南,表明对于早发性HAP / VAP,经验性治疗应从窄谱抗生素如青霉素类或头孢菌素类开始,而对于迟发性HAP/VAP,指南建议使用更广谱的经验性抗生素,如碳青霉烯类和氨基苷类。2018年中国成人HAP/VAP指南中将替加环素推荐作为HAP中MDRAB和CRE感染的主要药物之一[37]

3.3.2 sscCDI sscCDI的治疗方案包括口服万古霉素、非达霉素或者联合万古霉素口服与甲硝唑静脉注射。替加环素对于艰难梭菌体外抑菌效果好,并通过粪便排泄,多篇案例文献报道了替加环素单一或联合可用于sscCDI,但效果仍未确定。2014年欧洲临床微生物学和传染病学会和2016年澳大利亚传染病学会CDI指南将替加环素列为严重病例的三线治疗药物[38,39]。BISHOP等[40]对澳大利亚某一医院2013—2016年13例患有sssCDI并接受了替加环素治疗的患者(在两个情况下使用替加环素联合治疗:①腹腔感染或者全身感染需要抗生素治疗的严重CDI患者;②需入住ICU的严重复杂疾病患者)进行评价,30 d 总体的全因死亡率为8%(严重CDI者无病死,严重复杂性CDI患者病死率为25%),77%患者实现了临床治愈;从而认为替加环素在严重CDI中作为联合治疗的是安全有效的,并且可早期给予。目前替加环素单一或者联合用药治疗严重复杂性难治性艰难梭菌性肠炎的有效性以及用药时机仍未确认,故需要大型前瞻性随机对照试验进行评估;另外,替加环素本身导致CDI发生的情况也需要加以考虑。

3.3.3 血流感染 无论是动物实验或是临床个案报道,替加环素治疗BSI的疗效结果不一。 WANG 等[41]进行了替加环素治疗血流感染疗效的系统评价,发现替加环素的全因病死率低于对照组,但差异无统计学意义[OR=0.85,95%CI(0.31,2.33), P=0.745];替加环素单药治疗组的病死率与替加环素联合治疗组(6项研究,250例患者)相比,OR为2.73,95%CI(1.53,4.87),差异无统计学意义;其中5项研究共报告了398例肺炎克雷伯菌血流感染患者,其替加环素治疗组的病死率明显低于对照组。QURESHI等[42]使用替加环素联合其他抗菌药物治疗产碳青霉烯酶肺炎克雷伯菌菌血症,单用与联合用药组28 d病死率分别是57.8%和13.3%(P=0.02)。然而,多数临床研究仍报道了替加环素会增加了感染患者的总体病死率,可能与药物血药浓度低、患者病情严重等因素有关。CHENG等[7]报道了在替加环素MIC>2 mg ·L-1的MDRAB血流感染亚组中粘菌素-替加环素组14 d病死率超过了粘菌素-碳青霉烯组[(OR=6.93,95%CI(1.61,29.78);P=0.009]。XIAO等[43]回顾分析了370例肺炎克雷伯菌血流感染,发现替加环素组30 d病死率明显高于非替加环素组(51.2%和12.2%;P<0.001),同时多因素分析显示感染后替加环素的使用是患者死亡的独立危险因素且OR高达2.300。上述研究结果与2010年FDA就静脉内注射替加环素的安全性发布说明相符,其指出当患者出现严重感染时应考虑选择其他药物来代替替加环素[3]。目前治疗严重耐药菌BSI缺乏有效抗菌药物,本品与粘菌素联合治疗是否可能是治疗BSI的选择值得研究。

3.4 替加环素临床疗效不满意的可能原因

3.4.1 替加环素治疗剂量 随着多重耐药菌感染的出现,越来越多的临床案例提示按照替加环素说明书推荐给药方案无法达到预期的治疗效果。替加环素在血浆浓度较低,常规用药后其峰浓度(0.87 mg·L-1)甚至低于肠杆菌、不动杆菌或厌氧菌的MIC值(2~4 mg·L-1)。为此,大剂量方案可能比小剂量方案更有效。对于传统剂量的替加环素未能治愈感染的患者,常常需增加剂量,这也是近期中国专家共识中推荐的方法[24]。然而,目前支持大剂量替加环素方案对治疗有效的临床证据仍有限。JEFFREY等[44]对34例患者进行了为期3个月的替加环素疗效评估,发现每日一次大剂量替加环素治疗MDR革兰阳性/阴性病原体以及艰难梭菌引起的严重全身感染非常有效。同时,也有部分研究显示大剂量方案也无法提高疗效。DE PASCALE等[45]研究了大剂量(4例替加环素单药和29例联合)和标准剂量组(6例替加环素单药和24例联合)治疗多重耐药菌感染,两组病死率差异无统计学意义[OR=2.25;95%CI(0.52,9.63)]。

3.4.2 单一用药或联合用药 就替加环素疗效是否与单一用药有关方面也有争议。GOMEZ-SIMMONDS等 [46]回顾性地分析了2006—2013年美国纽约市两家医院141例碳青霉烯类耐药肺炎克雷伯菌BSI,发现单一用药与联合用药患者病死率无明显差异(P=0.4)。KENGKLA等[47]对2529例MD/XDRAB感染患者的29项研究进行Meta分析,结果显示虽然各治疗方案之间差异无统计学意义,但是使用粘菌素、舒巴坦和替加环素的三联疗法具有较高的临床治愈率。对此,国内对于多重耐药革兰阴性菌感染治疗的专家共识建议[24],不推荐替加环素单药用于多重耐药菌血流感染的治疗,但可联合用药或提高剂量来治疗严重感染,如成人cIAI、HAP和VAP等。

4 结束语

替加环素是一种广谱抗生素,体外对大部分革兰阳性需氧菌、革兰阴性需氧菌、厌氧菌和不典型病原体有良好的抗菌活性,具有较长的半衰期和广泛的组织分布。已经获批的适应证包括:cIAI、cSSSI和CAP,但基于其本身药学属性和临床研究,大多数国内外指南也不推荐其作为一线治疗药物。对于泛耐药细菌所导致的HAP/VAP、BSI等,已经进行了大量的临床观察,但效果尚不确定,该药物相关的总病死率增加的现象仍然是一个问题,故在重症耐药菌感染,不推荐作为一线治疗药物,只有在缺乏有效药物情况下作为备选治疗方案,且需要进一步研究用药剂量、联合用药等有关问题,确保患者治疗有效安全。

The authors have declared that no competing interests exist.

参考文献

[1] BARRIERE S L.Clinical,economic and societal impact of antibiotic resistance[J].Expert Opin Pharmacother,2015,16(2):151-153.
Abstract The concern over antibiotic resistance has been voiced since the discovery of modern antibiotics > 75 years ago. The concerns have only increased with time, with efforts to control resistance caused by widespread overuse of antibiotics in human medicine and far more than appreciated use in the feeding of animals for human consumption to promote growth. The problem is worldwide, but certain regions and selected health care institutions report far more resistance, including strains of Gram-negative bacteria that are susceptible only to the once discarded drugs polymyxin B or colistin, and pan-resistant strains are on the rise. One of the central efforts to control resistance, apart from antimicrobial stewardship, is the development of new antimicrobial agents. This has lagged significantly over the past 10 - 15 years, for a variety of reasons; but promising new agents are being developed, unfortunately none thus far addressing all potentially resistant strains. There is the unlikely, but not unreal, possibility that we could return to a pre-antibiotic era, where morbidity and mortality rates have risen dramatically and routine surgical procedures are not performed for fear of post-operative infections. The onus of control of resistance is a moral imperative that falls on the shoulders of all.
DOI:10.1517/14656566.2015.983077      PMID:25483564      URL    
[本文引用:1]
[2] FALAGAS M E,TANSARLI G S,KARAGEORGOPOULOS D E,et al.Deaths Attributable to Carbapenem-Resistant Enterobacteriaceae Infections[J].Emerg Infect Dis,2014,20(7):1170-1175.
Abstract We evaluated the number of deaths attributable to carbapenem-resistant Enterobacteriaceae by using studies from around the world published before April 9, 2012. Attributable death was defined as the difference in all-cause deaths between patients with carbapenem-resistant infections and those with carbapenem-susceptible infections. Online databases were searched, and data were qualitatively synthesized and pooled in a metaanalysis. Nine studies met inclusion criteria: 6 retrospective case-control studies, 2 retrospective cohort studies, and 1 prospective cohort study. Klebsiella pneumoniae was the causative pathogen in 8 studies; bacteremia was the only infection in 5 studies. We calculated that 26%-44% of deaths in 7 studies were attributable to carbapenem resistance, and in 2 studies, which included bacteremia and other infections, -3% and -4% of deaths were attributable to carbapenem resistance. Pooled outcomes showed that the number of deaths was significantly higher in patients with carbapenem-resistant infections and that the number of deaths attributable to carbapenem resistance is considerable.
DOI:10.3201/eid2007.121004      PMID:24959688      URL    
[本文引用:1]
[3] Tigecycline(Marketed as Tygacil) Information.Postmarket drug safety information for patients and providers.FDA/center for drug evaluation and research.2013[EB/OL].Available from:.
URL    
[本文引用:4]
[4] 雍伟哲. 抗超级耐药菌感染新药替加环素上市发布[J].中华医学信息导报,2013,28(9):5.
[本文引用:1]
[5] ARTHUR L E,KIZOR R S,SELIM A G,et al.Antibiotics for ventilator‐associated pneumonia[J].Cochrane Data System Rev,2016,10:CD004267.
Ventilator-associated pneumonia (VAP) is a significant cause of morbidity and mortality, complicating the medical course of approximately 10% of mechanically-ventilated patients, with an estimated attributable mortality of 13%. To treat VAP empirically, the American Thoracic Society currently recommends antibiotic therapy based on the patients' risk of colonisation by an organism with multidrug resistance. The selection of initial antibiotic therapy in VAP is important, as inappropriate initial antimicrobial treatment is associated with higher mortality and longer hospital stay in intensive care unit (ICU) patients.While guidelines exist for the antibiotic treatment of hospital-acquired pneumonia (HAP) from the American Thoracic Society and the British Society for Antimicrobial Chemotherapy, there are many limitations in the quality of available evidence. This systematic review aimed to summarise the results of all randomised controlled trials (RCTs) that compare empirical antibiotic regimens for VAP. The primary objective of this review was to assess the effect of different empirical antimicrobial therapies on the survival and clinical cure of adult patients with ventilator-associated pneumonia (VAP). Secondary objectives included reporting the incidence of adverse events, new superinfections, length of hospital stay, and length of intensive care unit (ICU) stay associated with these therapies. We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, LILACS, CINAHL and Web of Science to December 2015; we searched ClinicalTrials.gov to September 2016. Two review authors independently assessed RCTs comparing empirical antibiotic treatments of VAP in adult patients, where VAP was defined as new-onset pneumonia that developed more than 48 hours after endotracheal intubation. Physicians and researchers were not required to be blinded for inclusion in this review. Two review authors independently extracted study data. We pooled studies and analysed them in two ways. We examined monotherapy, or a single experimental antimicrobial drug, versus combination therapy, or multiple experimental antimicrobial drugs. We also examined carbapenem therapy versus non-carbapenem therapy. We included 12 studies with 3571 participants. All included studies examined the empiric use of one antimicrobial regimen versus another for the treatment of adults with VAP, but the particular drug regimens examined by each study varied. There was potential for bias because some studies did not report outcomes for all participants. All but one study reported sources of funding or author affiliations with pharmaceutical companies.We found no statistical difference in all-cause mortality between monotherapy and combination therapy (N = 4; odds ratio (OR) monotherapy versus combination 0.97, 95% confidence interval (CI) 0.73 to 1.30), clinical cure (N = 2; OR monotherapy versus combination 0.88, 95% CI 0.56 to 1.36), length of stay in ICU (mean difference (MD) 0.65, 95% CI 0.07 to 1.23) or adverse events (N = 2; OR monotherapy versus combination 0.93, 95% CI 0.68 to 1.26). We downgraded the quality of evidence for all-cause mortality, adverse events, and length of ICU stay to moderate for this comparison. We determined clinical cure for this comparison to be of very low-quality evidence.For our second comparison of combination therapy with optional adjunctives only one meta-analysis could be performed due to a lack of trials comparing the same antibiotic regimens. Two studies compared tigecycline versus imipenem-cilastatin for clinical cure in the clinically evaluable population and there was a statistically significant increase in clinical cure for imipenem-cilastatin (N = 2; OR tigecycline versus imipenem-cilastatin 0.44, 95% CI 0.23 to 0.84). Of importance, this effect was due to a single study.We found no statistical difference in all-cause mortality between carbapenem and non-carbapenem therapies (N = 1; OR carbapenem versus non-carbapenem 0.59, 95% CI 0.30 to 1.19) or adverse events (N = 3; OR carbapenem versus non-carbapenem 0.78, 95% CI 0.56 to 1.09), but we found that carbapenems are associated with a statistically significant increase in the clinical cure (N = 3; OR carbapenem versus non-carbapenem 1.53, 95% CI 1.11 to 2.12 for intention-to-treat (ITT) analysis and N = 2; OR carbapenem versus non-carbapenem 2.29, 95% CI 1.19 to 4.43 for clinically evaluable patients analysis). For this comparison we downgraded the quality of evidence for mortality, and clinical cure (ITT and clinically evaluable populations) to moderate. We determined the quality of evidence for adverse events to be low. We did not find a difference between monotherapy and combination therapy for the treatment of people with VAP. Since studies did not identify patients with increased risk for multidrug-resistant bacteria, these data may not be generalisable to all patient groups. However, this is the largest meta-analysis comparing monotherapy to multiple antibiotic therapies for VAP and contributes further evidence to the safety of using effective monotherapy for the empiric treatment of VAP.Due to lack of studies, we could not evaluate the best antibiotic choice for VAP, but carbapenems as a class may result in better clinical cure than other tested antibiotics.
DOI:10.1002/14651858.CD004267.pub4      PMID:27763732      URL    
[本文引用:1]
[6] BAI X R,LIU J M,JIANG D C,et al.Efficacy and safety of tigecycline monotherapy versus combination therapy for the treatment of hospital-acquired pneumonia(HAP):a meta-analysis of cohort studies[J].J Chemother,2018,30(3):1-7.
In 2015, the World Health Organization registered 10.4 million people who developed tuberculosis worldwide and 480,000 new cases of multidrug-resistant tuberculosis were identified. The care of multi and extensively drug-resistant tuberculosis is based on a combination of pyrazinamide and second-line drugs. These regimens are lengthy, partially effective and poorly tolerated. The challenge is to re-evaluate the use of existing molecules and to develop new agents more effective against resistant strains with shorter treatment duration. This literature review gives an overview of the latest research addressing these therapeutic objectives. Some molecules are in late stage clinical development among which pretomanid is showing promising results. Bedaquiline and delamanid have been recently approved by the Food and Drug Administration. The efficacy of drug regimens combining these molecules is under evaluation.
DOI:10.1080/1120009X.2017.1338845      PMID:28651480      URL    
[本文引用:1]
[7] CHENG A,CHUANG Y C,SUN H Y,et al.Excess mortali-ty associated with colistin-tigecycline compared with colistin-carbapenem combination therapy for extensively drug-resistant Acinetobacter baumannii bacteremia:a multicenter prospective observational study[J].Crit Care Med,2015,43(6):1194-1204.
Since few therapeutic options exist for extensively drug resistant Acinetobacter baumannii, an emerging threat in ICUs worldwide, and comparative prospective studies of colistin-based combination therapies are lacking, our objective was to compare the outcomes of patients with extensively drug-resistant A. baumannii bacteremia, treated with colistin-carbapenem and colistin-tigecycline combinations. Prospective, observational, multicenter study. Adults with extensively drug-resistant A. baumannii bacteremia were prospectively followed from 2010 to 2013 at three hospitals in Taiwan. Extensively drug-resistant A. baumannii was defined as A. baumannii (genospecies 2) nonsusceptible to all drug classes except for colistin and tigecycline, and standard combination therapy as use of parenteral colistin-carbapenem or colistin-tigecycline for at least 48 hours after onset of bacteremia. Primary outcome measure was 14-day mortality. Of the 176 episodes of extensively drug-resistant A. baumannii bacteremia evaluated, 55 patients with a median (interquartile range) age of 62 years (44-79 yr) and Sequential Organ Failure Assessment score of 9 (5-13) points received standard combination therapy: colistin-tigecycline in 29 patients and colistin-carbapenem in 26. Crude 14-day and in-hospital mortality rates for patients receiving colistin-tigecycline versus patients receiving colistin-carbapenem were 35% versus 15% (p=0.105) and 69% versus 50% (p=0.152), respectively. Breakthrough extensively drug-resistant A. baumannii bacteremia under steady state concentrations of combination therapy for colistin-tigecycline group was 18% and for colistin-carbapenem group was 0% (p=0.059). Eleven patients (20.0%) developed nephrotoxicity. After adjusting for age, sex, comorbidity, initial disease severity, loading colistin dose, polymicrobial infection, and primary infection site, excess 14-day mortality was associated with the use of colistin-tigecycline in the subgroup with tigecycline minimum inhibitory concentration greater than 2 mg/L compared with the use of colistin-carbapenem (hazard ratio, 6.93; 95% CI, 1.61-29.78; p=0.009). Increased 14-day mortality was associated with colistin-tigecycline therapy given tigecycline minimum inhibitory concentration greater than 2 mg/L compared with colistin-carbapenem therapy for extensively drug-resistant A. baumannii bacteremia.
DOI:10.1097/CCM.0000000000000933      PMID:25793437      URL    
[本文引用:2]
[8] DIXIT D,MADDURI R P,SHARMA R .The role of tigec-ycline in the treatment of infections in light of the new black box warning[J].Exp Rev Anti infecti Ther,2014,12(4):397-400.
Tigecycline is an antibiotic with a broad spectrum of activity. Similar to tetracycline antibiotics, tigecycline exerts bacteriostatic activity. Earlier studies documented the safety and efficacy of tigecycline for complicated intra-abdominal and complicated skin and skin-structure infections, which led to its approval. Recent systematic reviews and meta-analyses have suggested increased risk of death in patients receiving tigecycline compared to other antibiotics. The Food and Drug Administration has warned clinicians about increased risk for death in patients who received tigecycline with certain severe infections and have issued a black box warning. The increased mortality risk with tigecycline is most apparent in patients treated for hospital-acquired pneumonia, particularly ventilator-associated pneumonia. The cause of excess deaths in these trials is uncertain, but it is likely that most deaths in patients with these severe infections were related to progression of the infection. Further experience with tigecycline for serious infections with drug-resistant pathogens is currently warranted.
DOI:10.1586/14787210.2014.894882      PMID:24597542      URL    
[本文引用:1]
[9] GREER N D.Tigecycline(Tygacil):the first in the glycyl-cycline class of antibiotics[J].Proc (Bayl Univ Med Cent),2006,19(2):155-161.
Tigecycline is the first drug in the glycylcycline class of antibiotics. Although it is structurally related to minocycline, alterations to the molecule resulted in its expanded spectrum of activity and decreased susceptibility to the development of resistance when compared with other tetracycline antibiotics. Tigecycline has a broad spectrum of activity, including activity against drug-resistant gram-positive organisms. Randomized trials have shown tigecycline to be efficacious for the treatment of complicated intraabdominal infections and complicated skin and skin structure infections. The dose of tigecycline is 50 mg intravenously every 12 hours after a 100-mg loading dose. Nausea, vomiting, and diarrhea were the most common adverse events reported with tigecycline therapy and may result in discontinuation of therapy. ********** Resistant organisms remain a concern in hospitalized patients and are becoming an increased concern in community-acquired infections. Gram-positive organisms continue to increase in resistance, and very few agents are available to treat these infections. Available options include vancomycin, linezolid, daptomycin, and quinupristin/ dalfopristin. These antimicrobials have improved treatment of resistant gram-positive organisms but may have adverse events that require discontinuation. For this reason, new antimicrobials are needed to treat resistant organisms without sacrificing safety. Tetracyclines are broad-spectrum antibiotics that have been available since the mid-1900s, but use in recent years has been limited by widespread resistance to these agents. Resistance generally occurs by alterations in tetracycline efflux or ribosomal protection. The glycylcyclines were developed to help overcome these resistance mechanisms. Tigecycline, approved by the Food and Drug Administration (FDA) on June 15, 2005, is the first drug in this class of antimicrobials. Alterations to the tetracycline structure allow tigecycline to maintain activity against tetracycline-resistant organisms, including resistant gram-positive organisms such as penicillin-resistant Streptococcus pneumoniae, methicillin-resistant Staphylococcus aureus (MRSA) and Staphylococcus epidermidis (MRSE), and vancomycinresistant Enterococcus (VRE) species. Tigecycline is approved for the treatment of complicated skin and skin structure infections (cSSSI) and complicated intraabdominal infections caused by susceptible organisms (1). Tigecycline is a glycylcycline antimicrobial structurally related to minocycline. Like minocycline, tigecycline binds to the bacterial 30S ribosome, blocking the entry of transfer RNA. This ultimately prevents protein synthesis by halting the incorporation of amino acids into peptide chains and thus limits bacterial growth (1-3). However, the addition of an N,N,-dimethylglycylamido group at the 9 position of the minocycline molecule increases the affinity of tigecycline for the ribosomal target up to 5 times when compared with minocycline or tetracycline (4). This allows for an expanded spectrum of activity and decreased susceptibility to the development of resistance (1, 2, 5). PHARMACOKINETICS AND PHARMACODYNAMICS Tigecycline pharmacokinetics were evaluated after single and multiple doses were given over 30 to 60 minutes and are summarized in Table 1 (1-3, 6). Because it is administered intravenously, tigecycline is 100% bioavailable (1). Tigecycline is highly protein bound and has a large volume of distribution (7-9 L/kg) when compared with the other available tetracyclines. Concentrations of tigecycline in the gallbladder, lung, and colon are higher than serum concentrations (1), while concentrations of tigecycline in the bone and synovial fluid are lower...
DOI:10.1080/08998280.2006.11928154      PMID:16609746      URL    
[本文引用:1]
[10] KELESIDIS T,KARAGEORGOPOULOS D E,KELESIDIS I,et al.Tigecycline for the treatment of multidrug-resistant Enterobacteriaceae:a systematic review of the evidence from microbiological and clinical studies[J].J Antimicrob Chemoth,2008,62(5):895-904.
DOI:10.1093/jac/dkn311      URL    
[本文引用:1]
[11] SUN Y,CAI Y,LIU X,et al.The emergence of clinical resistance to tigecycline[J].Int J Antimicrob Agents,2013,41(2):110-116.
Tigecycline (TIG) exhibits broad-spectrum activity against many Gram-positive and Gram-negative pathogens. However, clinical resistance has emerged recently and has been detected following treatment with TIG. This observation suggests that long-term monotherapy may carry a high risk for TIG resistance. TIG resistance is observed most frequently in Acinetobacter baumannii and Enterobacteriaceae, especially in multidrug-resistant strains. Resistance-nodulation-cell division (RND)-type transporters and other efflux pumps may be factors for decreased sensitivity to TIG. Therefore, TIG should be cautiously used in the clinic, and efflux-mediated resistance should be closely monitored in order to prolong the lifespan of this useful antibiotic. (C) 2012 Elsevier B.V. and the International Society of Chemotherapy. All rights reserved.
DOI:10.1016/j.ijantimicag.2012.09.005      PMID:23127485      URL    
[本文引用:1]
[12] PFALLER M A,HUBAND M D,STREIT J,et al.Survei-llance of tigecycline activity tested against clinical isolates from a global(North America,Europe,Latin America and Asia-Pacific) collection(2016)[J].Int J Antimicrob Agents,2018,51(6):848-853.
Tigecycline and comparators were tested by reference broth microdilution method against 33,348 non-duplicate bacterial isolates collected prospectively in 2016 from medical centers in Asia-Pacific (3,443 isolates), Europe (13,530 isolates), Latin America (3,327 isolates), and United States (13,048 isolates). Among 7,098 Staphylococcus aureus isolates tested, >99.9% were inhibited by ≤0.5 mg/L of tigecycline (MIC 50/90 , 0.06/0.12 mg/L), including >99.9% of methicillin-resistant S. aureus and 100.0% of methicillin-susceptible S. aureus . Tigecycline was slightly more active against E. faecium (MIC 50/90 , 0.03/0.06 mg/L) when compared to E. faecalis (MIC 50/90 , 0.06/0.12 mg/L), and its activity was not adversely affected by vancomycin resistance when tested against these organisms. Tigecycline potency was comparable for Streptococcus pneumoniae (MIC 50/90 , 0.03/0.06 mg/L), viridans group streptococci (MIC 50/90 , 0.03/0.06 mg/L), and β-haemolytic streptococci (MIC 50/90 , 0.06/0.06 mg/L) regardless of species and susceptibility to penicillin. Tigecycline was active against Enterobacteriaceae (MIC 50/90 , 0.25/1 mg/L; 97.8% inhibited at ≤2 mg/L) but was slightly less active against Enterobacteriaceae isolates expressing resistant phenotypes: carbapenem-resistant (CRE; MIC 50/90 , 0.5/2 mg/L; 98.0% susceptible), multidrug-resistant (MDR; MIC 50/90 , 0.5/2 mg/L; 93.1% susceptible), and extensively drug-resistant (XDR; MIC 50/90 , 0.5/4 mg/L; 87.8% susceptible). Tigecycline (MIC 50/90 , 2/4 mg/L) inhibited 74.4% of 888 A. baumannii isolates at ≤2 mg/L and demonstrated good in vitro activity against S. maltophilia (MIC 50/90 , 1/2 mg/L; 90.6% inhibited at ≤2 mg/L) Tigecycline was active against Haemophilus influenzae (MIC 50/90 , 0.12/0.25 mg/L) regardless of β-lactamase status. Tigecycline represents an important treatment option for resistant Gram-negative and Gram-positive infections.
DOI:10.1016/j.ijantimicag.2018.01.006      PMID:29410368      URL    
[本文引用:1]
[13] 王健,沈继 .替加环素耐药机制的研究现状[J].中国感染与化疗杂志,2017,17(2):219-223.
替加环素(TGC)是 FDA 批准的用于治疗复杂性皮肤和软组织感染、复杂性腹腔感染和社区相关细菌性肺炎的抗生素.随着应用越来越广泛,替加环素产生耐药性不可避免.替加环素是一种新型甘氨酰环素类抗菌药物,全称为 9- 叔丁基甘氨酰氨基米诺环素,是继多西环素、米诺环素、美他环素后开发的新一代四环素类衍生抗生素.该药通过可逆地结合于 16S rRNA,阻断 tRNA 进入 A 位点,抑制了翻译过程.替加环素与核糖体的亲和力高于其他四环素类抗生素 20 倍 [1].同时,替加环素克服了四环素的耐药机制,如核糖体保护蛋白(tetM、tetO 等)和外排泵(tetK、tetA 等)[2],使得替加环素对四环素和米诺环素耐药菌均有作用.本文通过介绍替加环素耐药性机制,我们可以更加主动地确定相关基因的转移并通过当前的新型"反突变"药物前瞻性的阻止耐药性的产生.
DOI:10.16718/j.1009-7708.2017.02.021      URL    
[本文引用:1]
[14] KORTH-BRADLEY J M,BAIRD-BELLAIRE S J,PATAT A A,et al.Pharmacokinetics and safety of a single intravenous dose of the antibiotic tigecycline in patients with cirrhosis[J].J Clin Pharmacol,2011,51(1):93-101.
Tigecycline belongs to a new class of tetracyclines, the glycylcyclines, less than 20% of which is metabolized in the liver. Twenty-five patients with cirrhosis with varying degrees of functional hepatic reserve (Child-Pugh A, n = 10; B, n = 10; C, n = 5) and 23 healthy adults, matched by age, sex, weight, and smoking habits, received 100 mg of tigecycline infused intravenously over 60 minutes. Serum and urine samples were collected up to 120 hours after dosing. Pharmacokinetic data were derived using noncompartmental methods. The most common treatment-emergent adverse events in healthy volunteers were nausea (56.5%), vomiting (21.7%), and headache (21.7%) and in the patients with cirrhosis, albuminuria (12%). Mean (00±1 SD) tigecycline clearance values were 29.8 00± 11.3 L/h in healthy subjects and 31.2 00± 13.9 L/h (Child-Pugh A), 22.1 00± 9.3 L/h (Child-Pugh B), and 13.5 00± 2.7 L/h (Child-Pugh C) in the patients. A single intravenous dose of tigecycline 100 mg was safe and well-tolerated in patients with cirrhosis with varying degrees of hepatic functional reserve. No adjustment of tigecycline maintenance dosage is warranted in patients with compensated or moderately decompensated cirrhosis; doses should be reduced by 50%, to 25 mg, every 12 hours in patients with severely decompensated disease.
DOI:10.1177/0091270010363477      PMID:20308689      URL    
[本文引用:1]
[15] PETERSON L R .A review of tigecycline——the first glyc-ylcycline[J].Int J Antimicrob Agents,2008,32(Suppl 4):S215-222.
The dawn of a troubling post-antibiotic era likely is on the horizon, fuelled by a rise in bacterial resistance to existing antibiotic therapy alongside a waning pipeline of novel antibacterial agents. Tigecycline, a new glycylcycline with an expanded broad spectrum of in vitro activity, was recently approved for the treatment of complicated skin and soft tissue infections (cSSTIs) and complicated intra-abdominal infections (cIAIs). This review will examine how tigecycline evades the common mechanisms of antibiotic resistance, the metabolism and pharmacokinetics of tigecycline, and its spectrum of in vitro activity. The results of randomized clinical trials for the treatment of cSSTIs and cIAIs with tigecycline are also described, as is the patient safety and tolerability observed during these studies. Tigecycline monotherapy has been shown to be as effective as its comparators and, against a backdrop of rising bacterial resistance, the role for tigecycline in monotherapy of infections from Gram-positive, Gram-negative and anaerobic bacteria is a meaningful development.
DOI:10.1016/S0924-8579(09)70005-6      PMID:19134522      URL    
[本文引用:1]
[16] YAMASHITA N,KYLE MATSCHKE M S,GANDHI A,et al.Tigecycline pharmacokinetics,tolerability,safety,and effect on intestinal microflora in healthy Japanese male subjects[J].J Clin Pharmacol,2013,54(5):513-519.
Safety, tolerability, and pharmacokinetics of tigecycline in 76 healthy Japanese subjects were determined in three randomized, double-blind, placebo-controlled studies. Subjects in an ascending single-dose study (n090009=09000940) received 25090009150090009mg intravenously, whereas subjects in two multiple-dose studies received every 12-hour (q12h) dosing with 25090009mg (n090009=09000910) or 25, 50, or 100 then 50090009mg (n090009=09000930). Serial blood samples and urine were collected, drug concentrations determined, and pharmacokinetic parameters calculated. Fecal samples were also collected in the second multiple-dose study. After 10 days of tigecycline 50090009mg q12h, mean09000900±090009standard deviation pharmacokinetics in 8/10 subjects were: maximum concentration 1,11809000900±090009127090009ng/mL, area under the concentration090009time curve 009000912090009hours 3,26109000900±090009937090009ng090009h/mL, clearance 0.2509000900±0900090.05090009L/h/kg, half-life 60.709000900±09000923.4090009hours, and volume of distribution 11.909000900±0900092.3090009L/kg. The most common adverse events were nausea and vomiting. Changes in total bilirubin were also observed. Enterococci in the intestinal microflora were reduced, whereas the number of Enterobacteriaceae and Bacteroides remained relatively constant. Several strains of Bacteroides spp. resistant to tigecycline treatment were found in fecal samples on days 30 and 31. The pharmacokinetic profile of tigecycline was similar to non-Japanese subjects; tolerability and change in intestinal microflora were also similar.
DOI:10.1002/jcph.236      PMID:24243316      URL    
[本文引用:1]
[17] ENTENZA J M,MOREILLON P.Tigecycline in combina-tion with other antimicrobials:a review of in vitro,animal and case report studies[J].Int J Antimicrob Agents,2009,34(8):e1-9.
DOI:10.1016/S0924-8579(09)70540-0      URL    
[本文引用:1]
[18] BHAVNANI S M,RUBINO C M,HAMMEL J P,et al.Pharmacological and patient-specific response determinants in patients with hospital-acquired pneumonia treated with tigecycline[J].Antimicrob Agents Chemother, 2012,56(2):1065-1072.
Abstract Pharmacokinetic and clinical data from tigecycline-treated patients with hospital-acquired pneumonia (HAP) who were enrolled in a phase 3 clinical trial were integrated in order to evaluate pharmacokinetic-pharmacodynamic (PK-PD) relationships for efficacy. Univariable and multivariable analyses were conducted to identify factors associated with clinical and microbiological responses, based on data from 61 evaluable HAP patients who received tigecycline intravenously as a 100-mg loading dose followed by 50 mg every 12 h for a minimum of 7 days and for whom there were adequate clinical, pharmacokinetic, and response data. The final multivariable logistic regression model for clinical response contained albumin and the ratio of the free-drug area under the concentration-time curve from 0 to 24 h (fAUC(0-24)) to the MIC (fAUC(0-24):MIC ratio). The odds of clinical success were 13.0 times higher for every 1-g/dl increase in albumin (P < 0.001) and 8.42 times higher for patients with fAUC(0-24):MIC ratios of ≥0.9 compared to patients with fAUC(0-24):MIC ratios of <0.9 (P = 0.008). Average model-estimated probabilities of clinical success for the albumin/fAUC(0-24):MIC ratio combinations of <2.6/<0.9, <2.6/≥0.9, ≥2.6/<0.9, and ≥2.6/≥0.9 were 0.21, 0.57, 0.64, and 0.93, respectively. For microbiological response, the final model contained albumin and ventilator-associated pneumonia (VAP) status. The odds of microbiological success were 21.0 times higher for every 1-g/dl increase in albumin (P < 0.001) and 8.59 times higher for patients without VAP compared to those with VAP (P = 0.003). Among the remaining variables evaluated, the MIC had the greatest statistical significance, an observation which was not surprising given the differences in MIC distributions between VAP and non-VAP patients (MIC(50)and MIC(90) values of 0.5 and 0.25 mg/liter versus 16 and 1 mg/liter for VAP versus non-VAP patients, respectively; P = 0.006). These findings demonstrated the impact of pharmacological and patient-specific factors on the clinical and microbiological responses.
DOI:10.1128/AAC.01615-10      PMID:22143524      URL    
[本文引用:1]
[19] PURDY J,JOUVE S,YAN J L,et al.Pharmacokinetics and safety profile of tigecycline in children aged 8 to 11 years with selected serious infections:a multicenter,open-label,ascending-dose study[J].Clin Ther,2012,34(2):496-507.
Tigecycline, a broad-spectrum antibiotic used for treating serious bacterial infections in adults, may be suitable for pediatric use once an appropriate dosage is determined. The aim of this study was to assess the pharmacokinetic (PK) properties, safety profile, and descriptive efficacy of tigecycline. In this Phase II, multicenter, open-label clinical trial, children aged 8 to 11 years with community-acquired pneumonia (CAP), complicated intra-abdominal infection (cIAI), or complicated skin and skin structure infections (cSSSI) were administered tigecycline 0.75, 1, or 1.25 mg/kg. A total of 58 patients received ≥1 dose of tigecycline (31 boys; 44 white; mean age, 10 years; mean weight, 35 kg); 47 had data from samples available for PK analysis. The mean (SD) PK values were: Cmax, 1899 (2954) ng/mL; Tmax, 0.56 (0.18) hour; between-dose AUC, 2833 (1557) ng · h/mL; weight-normalized clearance, 0.503 (0.293) L/h/kg; and Vdss, 4.88 (4.84) L/kg. Overall clinical cure rates at test-of-cure were 94% (16/17), 76% (16/21), and 75% (15/20) in the 0.75-, 1-, and 1.25-mg/kg cohorts, respectively. The rates of protocol violations were higher in the 1- and 1.25-mg/kg groups, resulting in higher proportions of indeterminate clinical cure assessments relative to the 0.75-mg/kg cohort (19% and 15% vs 0%). The most frequent adverse event was nausea, which occurred in 50% of patients overall (29/58) and the prevalence of which was significantly higher in the 1.25-mg/kg group versus the 0.75-mg/kg group (65% vs 18%; P = 0.007). Pharmacodynamic simulations using MIC data from an ongoing microbiological surveillance trial predicted that a dosage of 1.2 mg/kg q12h would lead to therapeutic target attainment levels of up to 82% for the target AUC0–24/MIC ratios. A tigecycline dosage of 651.2 mg/kg q12h may represent the most appropriate dosage for subsequent evaluation in Phase III clinical trials in children aged 8 to 11 years with selected serious bacterial infections. ClinicalTrials.gov identifier: NCT00488345.
DOI:10.1016/j.clinthera.2011.12.010      PMID:22249106      URL    
[本文引用:1]
[20] 胡付品,郭燕,朱德妹,.2016年中国CHINET细菌耐药性监测[J].中国感染与化疗杂志,2017,17(5):7-17.
目的了解国内主要地区临床分离菌对常用抗菌药物的敏感性和耐药性。方法对国内主要地区30所教学医院(26所综合性医院、4所儿童医院)临床分离菌采用纸片扩散法或自动化仪器法按统一方案进行抗菌药物敏感性试验。按CLSI 2016版判断结果。结果收集2016年1-12月上述医院临床分离菌共153 059株,其中革兰阳性菌43 462株,占28.4%,革兰阴性菌109 597株,占71.6%。金黄色葡萄球菌和凝固酶阴性葡萄球菌中甲氧西林耐药株的平均检出率分别为38.4%和77.6%。甲氧西林耐药株(MRSA和MRCNS)对绝大多数测试药的耐药率均显著高于甲氧西林敏感株(MSSA和MSCNS)。MRSA中有92.3%菌株对甲氧苄啶-磺胺甲唑敏感;MRCNS中有86.5%菌株对利福平敏感;未发现万古霉素和替考拉宁耐药菌株。肠球菌属中粪肠球菌对多数测试抗菌药物(氯霉素除外)的耐药率均显著低于屎肠球菌,两者中均有少数万古霉素耐药株,经表型或基因型检测结果显示主要为Van A型、Van B型或Van M型耐药。儿童肺炎链球菌非脑膜炎分离株中青霉素敏感和中介(PSSP和PISP)株所占比例较2015年有所上升,青霉素耐药(PRSP)株的检出率有所下降;成人分离株中PISP和PRSP均有所下降。大肠埃希菌、克雷伯菌属(肺炎克雷伯菌和产酸克雷伯菌)和奇异变形杆菌中产ESBL率分别平均为45.2%、25.2%和16.5%,产ESBL株对测试药物的耐药率均比非产ESBL株高。肠杆菌科细菌对碳青霉烯类抗生素仍高度敏感,多数菌属的耐药率低于10%。不动杆菌属(鲍曼不动杆菌占90.6%)对亚胺培南和美罗培南的耐药率分别为68.6%和71.4%。与2015年耐药率数据相比,铜绿假单胞菌中广泛耐药株的检出率有所上升。结论临床分离菌对常用抗菌药物的耐药率仍呈增长趋势,应加强医院感染防控措施和抗菌药物临床应用管理措施,继续做好细菌耐药性监测工作。
URL    
[本文引用:2]
[21] NI W,HAN Y,LIU J,et al.Tigecycline treatment for carbapenem-resistant enterobacteriaceae infections:a systematic review and Meta-analysis[J].Medicine,2016,95(11):e3126.
DOI:10.1097/MD.0000000000003126      URL    
[本文引用:1]
[22] YAHAV D,LADOR A,PAUL M,et al.Efficacy and safety of tigecycline:a systematic review and meta-analysis[J].J Antimicrob Chemother,2011,66(9):1963-1971.
Tigecycline is a novel glycylcycline that exhibits broad-spectrum antibacterial activity. Recently, the US FDA issued a warning concerning increased mortality with tigecycline in randomized controlled trials (RCTs). We conducted a systematic review and meta-analysis of RCTs that compared tigecycline with any other antibiotic regimen for the treatment of any infection. A comprehensive search, without publication status or other restrictions, was conducted. The primary outcome was overall 30 day mortality. The secondary outcome included clinical and microbiological failure, superinfections and adverse events (AEs). The trials' risks of bias and their effects on results were assessed. Two reviewers independently extracted the data. Individual trials' relative risks (RRs) were pooled using a fixed effect meta-analysis. Fifteen trials (7654 patients) were included. Overall mortality was higher with tigecycline compared with other regimens [RR 1.29, 95% confidence interval (CI) 1.02-1.64, without heterogeneity]. The type of infection assessed and the trials' reported risks of bias did not affect this result. Clinical failure was significantly higher with tigecycline (RR 1.16, 95% CI 1.06-1.27) and non-statistically significant higher rates of microbiological failure were demonstrated (RR 1.13, 95% CI 0.99-1.30). Development of septic shock was significantly more frequent with tigecycline (RR 7.01, 95% CI 1.27-38.66). Superinfections were significantly more common with tigecycline and so were AEs, including all AEs and AEs requiring discontinuation. In the light of the increased mortality, probably explained by decreased clinical and microbiological efficacy, clinicians should avoid tigecycline monotherapy in the treatment of severe infections and reserve it as a last-resort drug.
DOI:10.1093/jac/dkr242      PMID:2020202020202032020202020      URL    
[本文引用:1]
[23] 王佳,骆霞,王林海,.替加环素联合头孢哌酮舒巴坦钠治疗多重/泛耐药鲍曼不动杆菌致肺炎的meta分析[J].临床药物治疗杂志,2017,15(1):44-48.
目的:系统评价替加环素联合头孢哌酮舒巴坦钠治疗多重/泛耐药鲍曼不动杆菌所致肺炎的疗效。方法:根据相关主题词、自由词从Cochrane Library、Pub Med、Google.scholar、中国知网、维普、万方等数据库检索替加环素联合头孢哌酮舒巴坦钠治疗多重/泛耐药鲍曼不动杆菌所致肺炎的随机对照实验(RCT)的文献。经过Jadad评分标准对所选文献进行筛查并用Excel表对数据进行提取整合。提取的数据用Rev Man 5.3统计软件进行meta分析。结果:最终纳入10篇RCTs,在临床有效率方面,替加环素联合头孢哌酮舒巴坦钠组明显高于单用头孢哌酮舒巴坦钠组[OR=3.86,95%CI(2.81-5.28),P〈0.000 01]。在细菌清除率方面,替加环素联合头孢哌酮舒巴坦钠组也显著高于单用头孢哌酮舒巴坦钠组[OR=2.44,95%CI(1.84-3.24),P〈0.000 01]。不良反应发生二者差异比较无统计学意义[OR=0.87,95%CI(0.58-1.31),P=0.51]。结论:替加环素联合头孢哌酮舒巴坦钠优于单用头孢哌酮舒巴坦钠治疗多重/泛耐药鲍曼不动杆菌所致的肺炎,且安全性较好。
[本文引用:1]
[24] LI J,YANG X,CHEN L,et al.In vitro activity of various Antibiotics in combination with tigecycline against acinetobacter baumannii :a systematic review and Meta-analysis[J].Microb Drug Resist,2017,23(8):982-993.
DOI:10.1089/mdr.2016.0279      URL    
[本文引用:3]
[25] Chinese XDR Consensus Working Group,GUAN X,HE L,et al.Laboratory diagnosis,clinical management and infection control of the infections caused by extensively drug- resistant Gram-negative bacilli:a Chinese consensus statement[J].Clin Microbiol Infect, 2016,22(Suppl 1):S15-25.
Extensively drug-resistant (XDR) Gram-negative bacilli (GNB) are defined as bacterial isolates susceptible to two or fewer antimicrobial categories. XDR-GNB mainly occur inEnterobacteriaceae,Acinetobacter baumannii,Pseudomonas aeruginosa, andStenotrophomonas maltophilia. The prevalence of XDR-GNB is on the rise in China and in other countries, and it poses a major public health threat as a result of the lack of adequate therapeutic options. A group of Chinese clinical experts, microbiologists and pharmacologists came together to discuss and draft a consensus on the laboratory diagnosis, clinical management and infection control of XDR-GNB infections. Lists of antimicrobial categories proposed for antimicrobial susceptibility testing were created according to documents from the Clinical Laboratory Standards Institute (CLSI), the European Committee on Antimicrobial Susceptibility Testing (EUCAST) and the United States Food and Drug Administration (FDA). Multiple risk factors of XDR-GNB infections are analyzed, with long-term exposure to extended-spectrum antimicrobials being the most important one. Combination therapeutic regimens are summarized for treatment of XDR-GNB infections caused by different bacteria based on limited clinical studies and/or laboratory data. Most frequently used antimicrobials used for the combination therapies include aminoglycosides, carbapenems, colistin, fosfomycin and tigecycline. Strict infection control measures including hand hygiene, contact isolation, active screening, environmental surface disinfections, decolonization and restrictive antibiotic stewardship are recommended to curb the XDR-GNB spread.
DOI:10.1016/j.cmi.2015.11.004      PMID:26627340      URL    
[本文引用:1]
[26] ECKMANN C,MONTRAVERS P,BASSETTI M,et al.Efficacy of tigecycline for the treatment of complicated intra-abdominal infections in real-life clinical practice from five European observational studies[J].J Antimicrob Chemother,2013,68(Suppl 2):25-35.
ABSTRACT Objectives: Tigecycline is a broad-spectrum antibiotic approved for the treatment of complicated intra-abdom-inal infections (cIAIs). The efficacy of tigecycline when administered as monotherapy or in combination with other antibacterials in the treatment of cIAIs in routine clinical practice is described. Patients and methods: Individual patient-level data were pooled from five European observational studies (July 2006 to October 2011). Results: A total of 785 cIAI patients who received tigecycline were included (mean age 63.1+14.0 years). Of these, 56.6% were in intensive care units, 65.6% acquired their infection in hospital, 88.1% had at least one comorbidity and 65.7% had secondary peritonitis. The mean Acute Physiology and Chronic Health Evaluation (APACHE) II and Sequential Organ Failure Assessment (SOFA) scores at the beginning of treatment were 16.9+7.6 (n 614) and 7.0+4.2 (n 108), respectively, indicating high disease severity. Escherichia coli (41.8%), Enterococcus faecium (40.1%) and Enterococcus faecalis (21.1%) were the most frequently isolated pathogens; 49.1% of infections were polymicrobial and 17.5% were due to resistant pathogens. Overall, 54.8% (n 430) received tigecycline as monotherapy and 45.2% (n 355) as combination therapy for a mean duration of 10.6 days. Clinical response rates at the end of treatment were 77.4% for all patients (567/733), 80.6% for patients who received tigecycline as monotherapy (329/408), 75.2% for patients with a nosocomial infection (354/471), 75.8% for patients with an APACHE II score .15 (250/330) and 54.2% (32/59) for patients with a SOFA score 7. Conclusions: In these real-life studies, tigecycline, alone and in combination, achieved favourable clinical re-sponse rates in patients with cIAI with a high severity of illness.
DOI:10.1093/jac/dkt142      PMID:23772043      URL    
[本文引用:1]
[27] CHEN Z,WU J,ZHANG Y,et al.Efficacy and safety of tigecycline mono- therapy vs.imipenem/cilastatin in Chinese patients with complicated intra-abdominal infections:a randomized controlled trial[J].BMC Infect Dis,2010,10(1):217.
pAbstract/p pBackground/p pTigecycline, a first-in-class broad-spectrum glycylcycline antibiotic, has broad-spectrum in vitro activity against bacteria commonly encountered in complicated intra-abdominal infections (cIAIs), including aerobic and facultative Gram-positive and Gram-negative bacteria and anaerobic bacteria. In the current trial, tigecycline was evaluated for safety and efficacy vs. imipenem/cilastatin in hospitalized Chinese patients with cIAIs./p pMethods/p pIn this phase 3, multicenter, open-label study, patients were randomly assigned to receive IV tigecycline or imipenem/cilastatin for 2 weeks. The primary efficacy endpoints were clinical response at the test-of-cure visit (12-37 days after therapy) for the microbiologic modified intent-to-treat and microbiologically evaluable populations. Because the study was not powered to demonstrate non-inferiority between tigecycline and imipenem/cilastatin, no formal statistical analysis was performed. Two-sided 95% confidence intervals (CIs) were calculated for the response rates in each treatment group and for differences between treatment groups for descriptive purposes./p pResults/p pOne hundred ninety-nine patients received 1 dose of study drug and comprised the modified intent-to-treat population. In the microbiologically evaluable population, 86.5% (45 of 52) of tigecycline- and 97.9% (47 of 48) of imipenem/cilastatin-treated patients were cured at the test-of-cure assessment (12-37 days after therapy); in the microbiologic modified intent-to-treat population, cure rates were 81.7% (49 of 60) and 90.9% (50 of 55), respectively. The overall incidence of treatment-emergent adverse events was 80.4% for tigecycline vs. 53.9% after imipenem/cilastatin therapy (itP /it 0.001), primarily due to gastrointestinal-related events, especially nausea (21.6% vs. 3.9%; itP /it 0.001) and vomiting (12.4% vs. 2.0%; itP /it= 0.005)./p pConclusions/p pClinical cure rates for tigecycline were consistent with those found in global cIAI studies. The overall safety profile was also consistent with that observed in global studies of tigecycline for treatment of cIAI, as well as that observed in analyses of Chinese patients in those studies; no novel trends were observed./p pTrial Registration/p pClinicalTrials.gov NCT00136201/p
DOI:10.1186/1471-2334-10-217      PMID:2920872      URL    
[本文引用:1]
[28] 陈昭燕,占美,徐珽.替加环素治疗复杂腹腔感染有效性及安全性的Meta分析[J].中国医院药学杂志,2017,37(24):2460-2466.
目的:系统评价替加环素(TGC)与亚胺培南/西司他汀(IMI/CIS)相比治疗复杂腹腔感染的有效性与安全性。方法:计算机检索PubMed、EMbase、Medline、The Cochrane Library(2017年3期)、CBM、CNKI、VIP和WanFang Data,同时检索中国临床试验注册中心(www.chictr.org.cn),检索时间均从建库至2017年3月。纳入关于替加环素对比亚胺培南西司他汀治疗复杂腹腔感染的随机对照试验(RCT)。采用RevMan 5.3统计软件进行Meta分析。结果:共纳入5项RCT,共计4 185例患者。Meta分析结果显示:临床治愈率:(1)ME人群:TGC与IMI/CIS组相比[RR=1.00,95%CI(0.95,1.05),P=0.92]临床治愈率差异不明显,差异无统计学意义;(2)CE人群:TGC组相比IMI/CIS组[RR=0.99,95%CI(0.96,1.02),P=0.50]临床治愈率差异不明显,差异无统计学意义;(3)c-mlTT人群:TGC组相比IMI/CIS组[RR=0.97,95%CI(0.94,1.00),P=0.05]差异有统计学意义,IMI/CIS组稍高于TGC组。不良反应发生率:TGC组相比IMI/CIS组会增加患者二次感染发生率[RR=1.75,95%CI(1.34,2.28),P≤0.000 1]、恶心发生率[RR=1.34,95%CI(1.08,1.65),P=0.008]、呕吐发生率[RR=1.39,95%CI(1.19,1.63),P≤0.000 1],其他不良反应发生率差异无统计学意义。死亡率:TGC组较IMI/CIS组死亡率稍高,但差异无统计学意义[RR=1.45,95%CI(0.94,2.22),P=0.09]。结论:TGC较IMI/CIS并不能显著提高治疗复杂腹腔感染的临床疗效,却增加主要胃肠道不良反应与二次感染的发生率。受纳入研究质量和数量限制,上述结论有待更多高质量的RCT来验证。
[本文引用:1]
[29] MAZUSKI J E,TESSIER J M,MAY A K,et al.The surgical infection society revised guidelines on the management of ntra-abdominal infection[J].Surg Infect,2017,18(1):1-76.
Abstract BACKGROUND: Previous evidence-based guidelines on the management of intra-abdominal infection (IAI) were published by the Surgical Infection Society (SIS) in 1992, 2002, and 2010. At the time the most recent guideline was released, the plan was to update the guideline every five years to ensure the timeliness and appropriateness of the recommendations. METHODS: Based on the previous guidelines, the task force outlined a number of topics related to the treatment of patients with IAI and then developed key questions on these various topics. All questions were approached using general and specific literature searches, focusing on articles and other information published since 2008. These publications and additional materials published before 2008 were reviewed by the task force as a whole or by individual subgroups as to relevance to individual questions. Recommendations were developed by a process of iterative consensus, with all task force members voting to accept or reject each recommendation. Grading was based on the GRADE (Grades of Recommendation Assessment, Development, and Evaluation) system; the quality of the evidence was graded as high, moderate, or weak, and the strength of the recommendation was graded as strong or weak. Review of the document was performed by members of the SIS who were not on the task force. After responses were made to all critiques, the document was approved as an official guideline of the SIS by the Executive Council. RESULTS: This guideline summarizes the current recommendations developed by the task force on the treatment of patients who have IAI. Evidence-based recommendations have been made regarding risk assessment in individual patients; source control; the timing, selection, and duration of antimicrobial therapy; and suggested approaches to patients who fail initial therapy. Additional recommendations related to the treatment of pediatric patients with IAI have been included. SUMMARY: The current recommendations of the SIS regarding the treatment of patients with IAI are provided in this guideline.
DOI:10.1089/sur.2016.261      PMID:28085573      URL    
[本文引用:1]
[30] ELLIS-GROSSE E J,BABINCHAK T,DARTOIS N,et al.Tigecycline 300 cSSSI study group,Tigecycline 305 cSSSI study group.The efficacy and safety of tigecycline in the treatment of skin and skin-structure infections:results of 2 double-blind phase 3 comparison studies with vancomycin-aztreonam[J].Clin Infect Dis,2005,41( Suppl 5):S341-S353.
DOI:10.1086/cid.2005.41.issue-s5      URL    
[本文引用:1]
[31] LAUF L,ZSóFIA O,MITHA I,et al.Phase 3 study comparing tigecycline and ertapenem in patients with diabetic foot infections with and without osteomyelitis[J].Diag Microbio Infect Dis,2014,78(4):469-480.
A phase 3, randomized, double-blind trial was conducted in subjects with diabetic foot infections without osteomyelitis (primary study) or with osteomyelitis (substudy) to determine the efficacy and safety of parenteral (intravenous [iv]) tigecycline (150 mg once-daily) versus 1 g once-daily iv ertapenem ± vancomycin. Among 944 subjects in the primary study who received ≥1 dose of study drug, >85% had type 2 diabetes; ~90% had Perfusion, Extent, Depth/tissue loss, Infection, and Sensation infection grade 2 or 3; and ~20% reported prior antibiotic failure. For the clinically evaluable population at test-of-cure, 77.5% of tigecycline- and 82.5% of ertapenem ± vancomycin–treated subjects were cured. Corresponding rates for the clinical modified intent-to-treat population were 71.4% and 77.9%, respectively. Clinical cure rates in the substudy were low (<36%) for a subset of tigecycline-treated subjects with osteomyelitis. Nausea and vomiting occurred significantly more often after tigecycline treatment (P = 0.003 and P < 0.001, respectively), resulting in significantly higher discontinuation rates in the primary study (nausea P = 0.007, vomiting P < 0.001). In the primary study, tigecycline did not meet criteria for noninferiority compared with ertapenem ± vancomycin in the treatment of subjects with diabetic foot infections.
DOI:10.1016/j.diagmicrobio.2013.12.007      PMID:24439136      URL    
[本文引用:1]
[32] DARTOIS N,COOPER C A,CASTAING N,et al.Tigecycline versus levofloxacin in hospitalized patients with community-acquired pneumonia:an analysis of risk factors[J].Open Res Med J,2013,7(1):13-20.
This study was conducted to evaluate the efficacy of tigecycline (TGC) versus levofloxacin (LEV) in hospitalized patients with community-acquired pneumonia (CAP) using pooled data and to perform exploratory analyses of risk factors associated with poor outcome. Pooled analyses of 2 phase 3 studies in patients randomized to intravenous (IV) TGC (100 mg, then 50 mg q12h) or IV LEV (500 mg q24h or q12h). Clinical responses at test of cure visit for the clinically evaluable (CE) and clinical modified intention to treat populations were assessed for patients with risk factors including aged 65 years, prior antibiotic failure, bacteremia, multilobar disease, chronic obstructive pulmonary disease, alcohol abuse, altered mental status, hypoxemia, renal insufficiency, diabetes mellitus, white blood cell count >30 x 109/L or <4 x 109/L, CURB-65 score 2, Fine score category of III to V and at least 2 clinical instability criteria on physical examination. In the CE population of 574 patients, overall cure rates were similar: TGC (253/282, 89.7%); LEV (252/292, 86.3%). For all but one risk factor, cure rates for TGC were similar to or higher than those for LEV. For individual risk factors, the greatest difference between treatment groups was observed in patients with diabetes mellitus (difference of 22.9 for TGC versus LEV; 95% confidence interval, 4.8 - 39.9). TGC achieved cure rates similar to those of LEV in hospitalized patients with CAP. For patients with risk factors, TGC provided generally favorable clinical outcomes.
DOI:10.2174/1874306401307010013      PMID:3601338      URL    
[本文引用:1]
[33] FORREST A,COOPER A,AMBROSE P G,et al.Pharma-cokinetics-pharmacodynamics of tigecycline in patients with community-acquired pneumonia[J].Antimicrob Agents Chemother,2012,56(1):130-136.
Exposure-response analyses for efficacy and safety were performed for tigecycline-treated patients suffering from community-acquired pneumonia. Data were collected from two randomized, controlled clinical trials in which patients were administered a 100-mg loading dose followed by 50 mg of tigecycline every 12 h. A categorical endpoint, success or failure, 7 to 23 days after the end of therapy (test of cure) and a continuous endpoint, time to fever resolution, were evaluated for exposure-response analyses for efficacy. Nausea/vomiting, diarrhea, headache, and changes in blood urea nitrogen concentration (BUN) and total bilirubin were evaluated for exposure-response analyses for safety. For efficacy, ratios of the free-drug area under the concentration-time curve at 24 h to the MIC of the pathogen (fAUC(0-24):MIC) of 12.8 were associated with a faster time to fever resolution; patients with lower drug exposures had a slower time to fever resolution (P = 0.05). For safety, a multivariable logistic regression model demonstrated that a tigecycline AUC above a threshold of 6.87 mg hr/liter (P = 0.004) and female sex were predictive of the occurrence of nausea and/or vomiting (P = 0.004). Although statistically significant, the linear relationship between tigecycline exposure and maximum change from baseline in total bilirubin is unlikely to be clinically significant.
DOI:10.1128/AAC.00277-10      PMID:3256083      URL    
[本文引用:1]
[34] 瞿介明,曹彬.中国成人社区获得性肺炎诊断和治疗指南(2016年版)修订要点[J].中华结核和呼吸杂志,2016,39(4):241-242.
@@
[本文引用:1]
[35] PRASAD P,SUN J,DANNER R L,et al.Excess deaths associated with tigecycline after approval based on noninferiority trials[J].Clin Infect Dis,2012,54(12):1699-1709.
Background. On the basis of noninferiority trials, tigecycline received Food and Drug Administration (FDA) approval in 2005. In 2010, the FDA warned in a safety communication that tigecycline was associated with an increased risk of death.
DOI:10.1093/cid/cis270      PMID:22467668      URL    
[本文引用:1]
[36] AMBARAS R K,AZIZ Z.The methodological quality of guidelines for hospital-acquired pneumonia and ventilator-associated pneumonia:A systematic review[J].J Clin Pharm Ther,2018,43(4):450-549.
DOI:10.1111/jcpt.12696      PMID:29722052      URL    
[本文引用:1]
[37] 中华医学会呼吸病学分会感染学组.中国成人医院获得性肺炎与呼吸机相关性肺炎诊断和治疗指南(2018年版)[J].中华结核和呼吸杂志,2018,41(4):255-280.
[本文引用:1]
[38] DEBAST S B,BAUER M P,KUIJPER E J.European society of clinical microbiology and infectious diseases:update of the treatment guidance document for clostridium difficile infection[J].Clin Microbiol Infect,2014,20(1):1-26.
In 2009 the first European Society of Clinical Microbiology and Infection (ESCMID) treatment guidance document for Clostridium difficile infection (CDI) was published. The guideline has been applied widely in clinical practice. In this document an update and review on the comparative effectiveness of the currently available treatment modalities of CDI is given, thereby providing evidence-based recommendations on this issue. A computerized literature search was carried out to investigate randomized and non-randomized trials investigating the effect of an intervention on the clinical outcome of CDI. The Grades of Recommendation Assessment, Development and Evaluation (GRADE) system was used to grade the strength of our recommendations and the quality of the evidence. The ESCMID and an international team of experts from 11 European countries supported the process. To improve clinical guidance in the treatment of CDI, recommendations are specified for various patient groups, e.g. initial non-severe disease, severe CDI, first recurrence or risk for recurrent disease, multiple recurrences and treatment of CDI when oral administration is not possible. Treatment options that are reviewed include: antibiotics, toxin-binding resins and polymers, immunotherapy, probiotics, and faecal or bacterial intestinal transplantation. Except for very mild CDI that is clearly induced by antibiotic usage antibiotic treatment is advised. The main antibiotics that are recommended are metronidazole, vancomycin and fidaxomicin. Faecal transplantation is strongly recommended for multiple recurrent CDI. In case of perforation of the colon and/or systemic inflammation and deteriorating clinical condition despite antibiotic therapy, total abdominal colectomy or diverting loop ileostomy combined with colonic lavage is recommended.
DOI:10.1111/1469-0691.12418      PMID:24118601      URL    
[本文引用:1]
[39] NELSON R L,SUDA K J,EVANS C T.Antibiotic treatment for Clostridium difficile-associated diarrhoea in adults[J].Cochrane Database Syst Rev,2017,3:CD004610.
Clostridium difficile (C. difficile) is recognized as a frequent cause of antibiotic-associated diarrhea and colitis. The aim of this review is to establish the efficacy of antibiotic therapy for C. difficile-associated diarrhea (CDAD), to identify the most effective antibiotic treatment for CDAD in adults and to determine the need for stopping the causative antibiotic during therapy. MEDLINE (1966 to 2003), EMBASE (1980 to 2003), Cochrane Central Database of Controlled Trials and the Cochrane IBD Review Group Specialized Trials Register were searched using the following search terms: "pseudomembranous colitis and randomized trial"; "Clostridium difficile and randomized trial"; "antibiotic associated diarrhea and randomized trial". Only randomized, controlled trials assessing antibiotic treatment for CDAD were included in the review. Probiotic trials are excluded. The following outcomes were sought: initial resolution of diarrhea; initial conversion of stool to C. difficile cytotoxin and/or stool culture negative; recurrence of diarrhea; recurrence of fecal C. difficile cytotoxin and/or positive stool culture; patient response to cessation of prior antibiotic therapy; sepsis; emergent surgery: fecal diversion or colectomy; and death. Data were analyzed using the MetaView statistical package in Review Manager. For dichotomous outcomes, relative risks (RR) and 95% confidence intervals (CI) were derived from each study. When appropriate, the results of included studies were combined for each outcome. For dichotomous outcomes, pooled RR and 95% CI were calculated using a fixed effect model, except where significant heterogeneity was detected, at which time the random effects model was used. Data heterogeneity was calculated using MetaView. Of eleven studies identified, two were subsequently excluded because patients were stool positive for C. difficile, but did not have diarrhea or because the study was not a randomized controlled trial. All of the remaining nine studies involved patients with diarrhea who recently received antibiotics for an infection other than C. difficile. The definition of diarrhea ranged from at least two loose stools per day with an associated symptom such as rectal temperature > 38(o)C, to at least six loose stools in 36 hours. In terms of symptomatic cure, metronidazole, bacitracin and fusidic acid were not shown to be less effective than vancomycin. Teicoplanin may be slightly more effective than vancomycin with a relative risk of 1.21 [95% CI 1.00 to 1.46] and a p-value of 0.06. In terms of initial symptomatic resolution, vancomycin is more effective than placebo with a relative risk of 6.75 [95% CI 1.16 to 48.43] and a p-value of 0.03. This result should be interpreted with caution given the small number of patients in this comparison (12 in the vancomycin group and nine in the placebo group) and the poor methodological quality of the trial. Metronidazole, bacitracin, teicoplanin, fusidic acid and rifaximine are as effective as vancomycin for initial symptomatic resolution. The other secondary outcomes measured in this review: surgery, sepsis and death occurred infrequently in all of the studies. Current evidence leads to uncertainty whether mild CDAD needs to be treated. Patients with mild CDAD may resolve their symptoms as quickly without treatment. The only placebo-controlled study shows vancomycin's superior efficacy. However, this result should be treated with caution due to the small number of patients enrolled and the poor methodological quality of the trial. The Johnson study of asymptomatic carriers also shows that placebo is better than vancomycin or metronidazole for eliminating C. difficile in stool during follow-up. If one does decide to treat, then two goals of therapy need to be kept in mind: improvement of the patient's clinical condition and prevention of spread of C. difficile infection to other patients. Given these two considerations, one should choose the antibiotic that brings both symptomatic cure and bacteriologic cure. In this regard, teicoplanin appears to be the best choice because the available evidence suggests that it is better than vancomycin for bacteriologic cure and has borderline superior effectiveness in terms of symptomatic cure. Teicoplanin is not readily available in the United States, which must be taken into account when making treatment decisions in that country.
DOI:10.1002/14651858.CD004610.pub4      PMID:15674956      URL    
[本文引用:1]
[40] BISHOP E J,TIRUVOIPATI R,METCALFE J,et al.The outcome of patients with severe and severe-complicated,clostridium difficile,infection treated with tigecycline combination therapy:a retrospective observational study[J].Inter Med J,2018,48(6):651-660.
DOI:10.1111/imj.2018.48.issue-6      URL    
[本文引用:1]
[41] WANG J,PAN Y,SHEN J,et al.The efficacy and safety of tigecycline for the treatment of bloodstream infections:a systematic review and meta-analysis[J].Ann Clin Microbiol Antimicrob,2017,16(1):24.
Abstract Patients with bloodstream infections (BSI) are associated with high mortality rates. Due to tigecycline has shown excellent in vitro activity against most pathogens, tigecycline is selected as one of the candidate drugs for the treatment of multidrug-resistant organisms infections. The purpose of this study was to evaluate the effectiveness and safety of the use of tigecycline for the treatment of patients with BSI. The PubMed and Embase databases were systematically searched, to identify published studies, and we searched clinical trial registries to identify completed unpublished studies, the results of which were obtained through the manufacturer. The primary outcome was mortality, and the secondary outcomes were the rate of clinical cure and microbiological success. 24 controlled studies were included in this systematic review. All-cause mortality was lower with tigecycline than with control antibiotic agents, but the difference was not significant (OR 0.85, [95% confidence interval (CI) 0.31-2.33; P = 0.745]). Clinical cure was significantly higher with tigecycline groups (OR 1.76, [95% CI 1.26-2.45; P = 0.001]). Eradication efficiency did not differ between tigecycline and control regimens, but the sample size for these comparisons was small. Subgroup analyses showed good clinical cure result in bacteremia patients with CAP. Tigecycline monotherapy was associated with a OR of 2.73 (95% CI 1.53-4.87) for mortality compared with tigecycline combination therapy (6 studies; 250 patients), without heterogeneity. Five studies reporting on 398 patients with Klebsiella pneumoniae carbapenemase-producing K. pneumoniae BSI showed significantly lower mortality in the tigecycline arm than in the control arm. The combined treatment with tigecycline may be considered the optimal option for severely ill patients with BSI.
DOI:10.1186/s12941-017-0199-8      PMID:28381268      URL    
[本文引用:1]
[42] QURESHI Z A,PATERSON D L,POTOSKI B A,et al.Treatment outcome of bacteremia due to KPC-producing Klebsiella pneumoniae:superiority of combination antimicrobial regimens[J].Antimicrob Agents Chemother,2012,56(4):2108-2113.
DOI:10.1128/AAC.06268-11      URL    
[本文引用:1]
[43] XIAO T,YU W,NIU T,et al.A retrospective,comparative analysis of risk factors and outcomes in carbapenem-susceptible and carbapenem-nonsusceptible Klebsiella pneumoniaebloodstream infections:tigecycline significantly increases the mortality[J].Infect Drug Resist,2018, 11:595-606.
DOI:10.2147/IDR      URL    
[本文引用:1]
[44] JEFFREY B,SHUNTAO C,NATALIE K,et al.Once daily high dose tigecycline is optimal:tigecycline PK/PD parameters predict clinical effectiveness[J].J Clin Med,2018,7(3):49.
Objective: The clinical effectiveness of tigecycline depends on appropriate use, and PK/PD (pharmacokinetic/pharmacodynamic) parameters related to dose and dosing interval. Methods: In our 600-bed university-affiliated teaching hospital, we conducted a tigecycline efficacy review over a three-month period in 34 evaluable patients. Parameters assessed included clinical response, cure or treatment failure, once daily as q12h dosing, maintenance dosing, high dose vs. standard loading regimens, adverse effects, and the effect of infectious disease consultation on outcomes. Results: We found once daily high dose tigecycline (HDT) was highly effective in treating serious systemic infections due to MDR Gram-positive/negative pathogens as well asC. difficilecolitis. Adverse effects were infrequent and limited to mild nausea/vomiting. Once daily HDT was highly effective, and the few treatment failures were related to suboptimal/split dosing regimens. Conclusion: Once daily HDT was highly effective when used to treat susceptible pathogens and when optimally dosed, i.e., 200–400 mg (IV) loading dose ×1, followed by a once daily maintenance dose of 100–200 mg (IV) q24h.
DOI:10.3390/jcm7030049      PMID:29522431      URL    
[本文引用:1]
[45] DE PASCALE G,MONTINI L,PENNISI M A,et al.High dose tigecycline in critically ill patients with severe infections due to multidrug-resistant bacteria[J].Crit Care,2014,18(3):R90.
Introduction The high incidence of multidrug-resistant (MDR) bacteria among patients admitted to ICUs has determined an increase of tigecycline (TGC) use for the treatment of severe infections. Many concerns have been raised about the efficacy of this molecule and increased dosages have been proposed. Our purpose is to investigate TGC safety and efficacy at higher than standard doses. Methods We conducted a retrospective study of prospectively collected data in the ICU of a teaching hospital in Rome. Data from all patients treated with TGC for a microbiologically confirmed infection were analyzed. The safety profile and efficacy of high dosing regimen use were investigated. Results Over the study period, 54 patients (pts) received TGC at a standard dose (SD group: 50 mg every 12 hours) and 46 at a high dose (HD group: 100 mg every 12 hours). Carbapenem-resistant Acinetobacter.baumannii (blaOXA-58 and blaOXA-23 genes) and Klebsiella pneumoniae (blaKPC-3 gene) were the main isolated pathogens (n???=???79). There were no patients requiring TGC discontinuation or dose reduction because of adverse events. In the ventilation-associated pneumonia population (VAP) subgroup (63 patients: 30 received SD and 33 HD), the only independent predictor of clinical cure was the use of high tigecycline dose (odds ratio (OR) 6.25; 95% confidence interval (CI) 1.59 to 24.57; P???=???0.009) whilst initial inadequate antimicrobial treatment (IIAT) (OR 0.18; 95% CI 0.05 to 0.68; P???=???0.01) and higher Sequential Organ Failure Assessment (SOFA) score (OR 0.66; 95% CI 0.51 to 0.87; P???=???0.003) were independently associated with clinical failure. Conclusions TGC was well tolerated at a higher than standard dose in a cohort of critically ill patients with severe infections. In the VAP subgroup the high-dose regimen was associated with better outcomes than conventional administration due to Gram-negative MDR bacteria.
DOI:10.1186/cc13858      PMID:24887101      URL    
[本文引用:1]
[46] GOMEZ-SIMMONDS A,NELSON B,EIRAS D P,et al.Combination regimens for treatment of carbapenem-resistant Klebsiella pneumoniae bloodstream infections[J].Antimicrob Agents Chemother,2016,60(6):3601-3607.
Previous studies reported decreased mortality in patients with carbapenemase-producing Klebsiella pneumoniae bloodstream infections (BSIs) treated with combination therapy but included carbapenem-susceptible and -intermediate isolates, as per revised CLSI breakpoints. Here, we assessed outcomes in patients with BSIs caused by phenotypically carbapenem-resistant K. pneumoniae (CRKP) according to the number of in vitro active agents received and whether an extended-spectrum beta-lactam (BL) antibiotic, including meropenem, or an extended-spectrum cephalosporin was administered. We retrospectively reviewed CRKP BSIs at two New York City hospitals from 2006 to 2013, where all isolates had meropenem or imipenem MICs of ≥4 μg/ml. Univariate and multivariable models were created to identify factors associated with mortality. Of 141 CRKP BSI episodes, 23% were treated with a single active agent (SAA), 26% were treated with an SAA plus BL, 28% were treated with multiple active agents (MAA), and 23% were treated with MAA plus BL. Ninety percent of isolates had meropenem MICs of ≥16 μg/ml. Thirty-day mortality was 33% overall and did not significantly differ across the four treatment groups in a multivariable model (P = 0.4); mortality was significantly associated with a Pitt bacteremia score of ≥4 (odds ratio [OR], 7.7; 95% confidence interval [CI], 3.2 to 18.1; P = 0.1), and immunosuppression was protective (OR, 0.4; 95% CI, 0.2 to 1.0; P = 0.04). Individual treatment characteristics were also not significantly associated with outcome, including use of SAAs versus MAA (26% versus 38%, P = 0.1) or BL versus no BL (26% versus 39%, P = 0.1). In summary, in patients with CRKP BSIs caused by isolates with high carbapenem MICs, the role of combination therapy remains unclear, highlighting the need for prospective studies to identify optimal treatment regimens.
DOI:10.1128/AAC.03007-15      PMID:27044555      URL    
[本文引用:1]
[47] KENGKLA K,KONGPAKWATTANA K,SAOKAEW S,et al.Comparative efficacy and safety of treatment options for MDR and XDR Acinetobacter baumannii infections:a systematic review and network meta-analysis[J].J Antimicrob Chemother,2017,73(1):22-32.
Abstract Objectives: To comprehensively compare and rank the efficacy and safety of available treatment options for patients with MDR and XDR Acinetobacter baumannii (AB) infection. Methods: We searched PubMed, Embase and the Cochrane register of trials systematically for studies that examined treatment options for patients with MDR- and XDR-AB infections until April 2016. Network meta-analysis (NMA) was performed to estimate the risk ratio (RR) and 95% CI from both direct and indirect evidence. Primary outcomes were clinical cure and microbiological cure. Secondary outcomes were all-cause mortality and nephrotoxic and non-nephrotoxic adverse events. Results: A total of 29 studies with 2529 patients (median age 60 years; 65% male; median APACHE II score 19.0) were included. Although there were no statistically significant differences between treatment options, triple therapy with colistin, sulbactam and tigecycline had the highest clinical cure rate. Colistin in combination with sulbactam was associated with a significantly higher microbiological cure rate compared with colistin in combination with tigecycline (RR 1.23; 95% CI 1.03-1.47) and colistin monotherapy (RR 1.21; 95% CI 1.06-1.38). No significant differences in all-cause mortality were noted between treatment options. Tigecycline-based therapy also appeared less effective for achieving a microbiological cure and is not appropriate for treating bloodstream MDR- and XDR-AB infections. Conclusions: Combination therapy of colistin with sulbactam demonstrates superiority in terms of microbiological cure with a safety profile similar to that of colistin monotherapy. Thus, our findings support the use of this combination as a treatment for MDR- and XDR-AB infections.
DOI:10.1093/jac/dkx368      PMID:29069421      URL    
[本文引用:1]
资源
PDF下载数    
RichHTML 浏览数    
摘要点击数    

分享
导出

相关文章:
关键词(key words)
替加环素
耐药菌
临床应用
病死率

Tigecycline
Drug-resistant bacteria
Clinical application
Mortality

作者
肖婷婷
肖永红

XIAO Tingting
XIAO Yonghong