Use of tigecycline in critically ill patients with serious nosocomial intra-abdominal infections.
替加环素药物利用评估标准的建立及应用
替加环素药物利用评估标准的建立及应用王媛媛;鲁超;赵大海;雷婷【期刊名称】《中国药房》【年(卷),期】2018(029)002【摘要】OBJECTIVE:To provide reference for rational use of tigecycline.METHODS:Based on tigecycline instructions,referring to related specifications and literatures,through pharmacy and clinical expert discussion,DUE criteria for tigecycline was established from medication indications,medication process,medication results and administrative supervision.In retrospective study,DUE criteria was used to evaluate medical records of inpatients in our hospital during Jan.2015-Dec.2016.RESULTS:A total of 71 medical records of inpatients receiving tigecycline were included.The use of tigecycline in our hospital was basically up to DUE criteria.But there still was unreasonable phenomenon,such as microbial inspection rate was 81.7% (aiming at 90%);the rate of medication course meeting the criteria was 76.1% (aiming at 90%);the proportion of patients with consultation records was 81.7%(aiming at 100%);the rate of prescribing authority meeting the criteria was 85.9% (aiming at 100%).CONCLUSIONS:The established DUE criteria for tigecycline shows strong operability and practicability,and provide reference for the work development of doctors and clinical pharmacists.%目的:为临床合理应用替加环素提供参考.方法:以替加环素药品说明书为基础,参考相关规范和文献,并通过与临床专家讨论协商,从用药指征、用药过程、用药结果和行政监管4个方面建立替加环素药物利用评估(DUE)标准;同时,采用回顾性调查方法,对我院2015年1月-2016年12月使用替加环素的住院患者病历应用该DUE标准进行评估.结果:共纳入使用替加环素的住院患者病历71份.经评估我院替加环素使用总体基本符合该DUE标准的要求,但尚存在一些不合理情况,包括微生物送检率为81.7%(目标值为90%),给药疗程符合标准率为76.1%(目标值为90%),病历中有会诊记录的患者比例为81.7%(目标值为100%),处方权限符合标准率为85.9%(目标值为100%).结论:所建立的替加环素DUE标准有较强的可操作性和实用性,可为医师和临床药师开展相关工作提供参考.【总页数】5页(P187-191)【作者】王媛媛;鲁超;赵大海;雷婷【作者单位】安徽医科大学第二附属医院药剂科,合肥230601;安徽医科大学第二附属医院药剂科,合肥230601;安徽医科大学第二附属医院呼吸内科,合肥230601;安徽医科大学第二附属医院药剂科,合肥230601【正文语种】中文【中图分类】R95;R969.3【相关文献】1.某院替加环素药物利用评价标准的建立与应用分析 [J], 王桂凤;李雪芹;刘锐锋;李运景;黎一山;何慧清2.替加环素超说明书药物利用评价标准的建立与应用 [J], 杨雪婷;郑鹏程;曹玮3.泮托拉唑药物利用评价标准的建立与应用 [J], 王鹏;崔健毓;林勇;彭勤;袁浩宇4.万古霉素、美罗培南及替加环素在重症医学科药物利用评价标准的建立与应用分析 [J], 于馨;梁爽;纪俐娜;李飞;冷萍5.万古霉素、美罗培南及替加环素在重症医学科药物利用评价标准的建立与应用分析 [J], 于馨;梁爽;纪俐娜;李飞;冷萍因版权原因,仅展示原文概要,查看原文内容请购买。
肝移植受者围手术期应用替加环素预防感染的效果及低纤维蛋白原血症发生情况
第14卷 第2期2023年3月Vol. 14 No.2Mar. 2023器官移植Organ Transplantation ·论著·肝移植受者围手术期应用替加环素预防感染的效果及低纤维蛋白原血症发生情况徐静 赵圆圆 陈知水 杨博 陈栋 魏来【摘要】 目的 探讨肝移植受者围手术期使用替加环素预防感染的效果及低纤维蛋白原血症发生情况。
方法 回顾性分析40例使用替加环素进行预防感染的肝移植受者的临床资料,分析受者感染事件和供者来源感染事件发生情况;分析替加环素治疗时、结束时及治疗结束后(7±2)d 受者临床指标变化情况;总结低纤维蛋白原血症的发生及治疗情况。
结果 40例肝移植受者中,2例受者发生感染,分别为黑曲霉和巨细胞病毒感染,均不属于替加环素抗菌谱所覆盖的范围,调整抗感染方案后感染情况控制良好。
9例供肝相关培养阳性,但均未发展为供者来源性感染事件。
40例受者均于术后2周左右肝功能恢复良好出院,其中6例于术后2~4 d 出现低纤维蛋白原血症伴凝血功能障碍,而转氨酶、胆红素、感染相关指标术后逐步下降,白蛋白水平稳定,予以补充人纤维蛋白原及凝血酶原复合物,凝血功能好转,但纤维蛋白原水平持续下降。
停用替加环素后,纤维蛋白原水平逐渐恢复至正常,考虑可能为替加环素相关药物不良反应。
结论 肝移植受者围手术期使用包含替加环素在内的预防感染方案可以降低敏感细菌导致的感染发生率,但药物使用期间需密切关注低纤维蛋白原血症的发生。
【关键词】 肝移植;感染;替加环素;低纤维蛋白原血症;药物不良反应;多重耐药菌;供者来源性感染;凝血功能障碍【中图分类号】 R617,R978.1 【文献标志码】A 【文章编号】1674-7445(2023)02-0010-07【Abstract 】 Objective To evaluate the efficacy of perioperative use of tigecycline in preventing infection and the incidence of hypofibrinogenemia in liver transplant recipients. Methods Clinical data of 40 liver transplant recipients given with tigecycline to prevent infection were retrospectively analyzed. The incidence of infection in recipients and donor-derived infection were analyzed. The changes of clinical indexes in recipients during, upon the completion and (7±2) d after tigecycline treatment were analyzed, respectively. The incidence and treatment of hypofibrinogenemia were summarized. Results Among 40 liver transplant recipients, 2 cases were infected by aspergillus niger and cytomegalovirus, out of the antibacterial spectrum of tigecycline. After adjusting the anti-infection regimen, the infection was properly controlled. Liver allografts were positive for relevant culture in 9 cases, whereas none of them progressedEfficacy of perioperative use of tigecycline in preventing infection and incidence of hypofibrinogenemia in liver transplant recipients Xu Jing, Zhao Yuanyuan, Chen Zhishui, Yang Bo, Chen Dong, Wei Lai. Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, Key Laboratory of Organ Transplantation of National Health Commission of China, Key Laboratory of Organ Transplantation of Chinese Academy of Medical Sciences, Wuhan 430030, ChinaCorrespondingauthor:WeiLai,Email:***************DOI: 10.3969/j.issn.1674-7445.2023.02.010基金项目:湖北省重点研发计划项目(2022BCA015)作者单位:430030 武汉,华中科技大学同济医学院附属同济医院器官移植研究所 器官移植教育部重点实验室 国家卫生健康委员会器官移植重点实验室 中国医学科学院器官移植重点实验室作者简介:徐静,硕士研究生,研究方向为器官移植,Email:**********************通信作者:魏来,博士,主任医师,研究方向为肝移植Email :***************·242·第14卷器官移植随着手术技术和免疫抑制药的进步,实体器官移植成为治疗终末期器官衰竭的重要策略,而细菌感染仍然是实体器官移植受者术后死亡的最主要原因[1]。
重症感染
D-二聚体 > 2000 ug/L
持续高水平的血管紧张素Ⅱ
Nat Commun,2014,5:3595.
感染严重程度思维图
重症感染
critically ill patient
关键参数ns in critically ill patients
2016 年 CHINET 耐药监测主要菌种分布
因素
COPD 充血性心力 衰竭
n
166 35
重症
34 10
OR
1.909 2.652
95%CI
(1.194, 3.053) (1.190, 5.911)
P值
0.007 0.017
糖尿病
老年痴呆
107
13
25
5
2.441
4.230
(1.434, 4.154)
(1.242, 14.400)
0.001
0.021
• 舒巴坦及舒巴坦合剂对鲍曼不动杆菌属具良好的抗菌活性,目前 国内多使用头孢哌酮/舒巴坦 • 2010年CHINET细菌耐药显示,目前国内临床使用抗菌药中,头 孢哌酮/舒巴坦的耐药率最低 • 对鲍曼不动杆菌敏感株的抗菌活性强 • 但近年来鲍曼不动杆菌对碳青霉烯类的耐药性上升迅速,全球范 围内(包括中国)的耐药率在50%以上 • 这类药物多与其他抗菌药联合治疗敏感鲍曼不动杆菌感染 • 目前我国鲍曼不动杆菌对阿米卡星的耐药率超过50% • 临床应用的多为多粘菌素E,鲍曼不动杆菌对其耐药率低,但异质 性耐药极易发生 • 近期发现其耐药菌株明显增加、常需与其他抗菌药物联合用
多粘菌素+碳青霉烯类 要点:联合、较大剂量、疗程不小于 2 周、注重临床疗效而非细菌学清除
Chinese XDR Consensus Working Group. Clin Microbiol Infect 2015
青霉素的使用和发现英语作文
青霉素的使用和发现英语作文Penicillin, a powerful antibiotic, has revolutionized the field of medicine since its discovery in the early 20th century. This remarkable drug has saved millions of lives by effectively treating various bacterial infections that were once considered fatal. The journey of penicillin from its accidental discovery to its widespread use is an extraordinary tale that highlights the importance of scientific curiosity and perseverance.The story begins in 1928, when Alexander Fleming, a British bacteriologist, was working on a culture of Staphylococcus aureus, a common bacteria that can cause severe infections. Fleming noticed that a mold growing nearby had produced a substance that inhibited the growth of the bacteria. Intrigued by this observation, he conducted further experiments to isolate and study the mold and its antibacterial properties.The mold was identified as Penicillium notatum, and the substance it produced was named penicillin. Flemingrealized that this substance had the potential to revolutionize the treatment of bacterial infections.However, the road to its clinical use was fraught with challenges. Penicillin was difficult to isolate in pure form, and it was unstable and easily destroyed by heat and acids.Despite these obstacles, Fleming's work caught the attention of other scientists, and a collaboration between the University of Oxford and the pharmaceutical company Boots led to the development of a purer and more stable form of penicillin. In 1942, during the height of World War II, penicillin was first used to treat a patient with a severe infection, and its effectiveness was remarkable. The drug quickly became a staple in hospitals and clinics worldwide, saving countless lives.The discovery and use of penicillin marked asignificant milestone in the history of medicine. It marked the beginning of the antibiotic era, which has transformed the way we treat infectious diseases. Penicillin and other antibiotics have saved millions of lives by eradicating once-fatal infections. However, the widespread use of antibiotics has also led to the emergence of resistantbacteria, a global health challenge that requires urgent attention.The story of penicillin reminds us of the importance of scientific curiosity and perseverance in推动科学进步. Fleming's accidental discovery led to a revolution in medicine, and the collaborative efforts of scientists and pharmaceutical companies resulted in the development of a drug that has saved millions of lives. The challenges faced in the process also highlight the need for continuous research and innovation to address new health challenges. As we look forward, it is crucial that we continue to invest in scientific research and innovation to address the ever-evolving health challenges of our time. The discovery and use of penicillin have taught us that the power of science can transform lives, and it is up to us to harness this power to create a healthier and safer world.**青霉素的发现与使用:医学的革命**自20世纪初被发现以来,青霉素这一强大的抗生素彻底改变了医学领域。
短链氯化石蜡禁用文件
短链氯化石蜡禁用文件英文回答:Short-chain chlorinated paraffins (SCCPs) are a groupof chemicals that have been widely used in variousindustrial applications due to their flame retardant properties. However, their use has been restricted or banned in many countries due to their potential adverse effects on human health and the environment.The main reason for the prohibition of SCCPs is their persistence and bioaccumulative nature. SCCPs have a long half-life in the environment and can accumulate in living organisms, including humans, through various pathways such as inhalation, ingestion, and dermal contact. Once inside the body, SCCPs can interfere with the normal functioningof organs and systems, leading to a range of health issues.For example, studies have shown that SCCPs can havetoxic effects on the liver, kidney, and reproductive system.They have been associated with liver and kidney damage, reduced fertility, and developmental abnormalities in animals. In humans, exposure to SCCPs has been linked to liver and thyroid diseases, as well as adverse effects on the immune system.Furthermore, SCCPs are also known to be persistent organic pollutants (POPs) that can travel long distances through air and water currents. This means that even countries that have banned the use of SCCPs may still be exposed to these chemicals through imported products or environmental contamination.To illustrate this, let's consider the case of a country that has banned the use of SCCPs in consumer products. Despite the ban, the country may still import SCCP-containing products from other countries where their use is not prohibited. These products can then release SCCPs into the environment during their use, leading to local contamination and potential exposure to the population.In addition to the health risks, SCCPs also pose environmental concerns. They are toxic to aquatic organisms and can accumulate in sediments, posing a threat to the aquatic ecosystem. SCCPs have been found in various environmental compartments, including water, soil, and air, indicating their widespread presence and potential forlong-term environmental impact.Given the potential adverse effects of SCCPs on human health and the environment, it is crucial to restrict or ban their use. Many countries have already taken steps to regulate SCCPs and phase out their use in various applications. This includes the development of alternative flame retardants that are less harmful but still effective in preventing fires.In conclusion, the prohibition of short-chain chlorinated paraffins is necessary to protect human health and the environment. The persistence, bioaccumulative nature, and potential toxic effects of SCCPs make them a significant concern. By implementing restrictions and finding safer alternatives, we can minimize the risksassociated with SCCPs and create a safer and healthier environment for all.中文回答:短链氯化石蜡(SCCPs)是一类具有阻燃性能的化学物质,在各种工业应用中广泛使用。
40例使用替加环素的临床用药分析
40例使用替加环素的临床用药分析叶伟红;应小飞;傅军霞;郭晶晶;徐艳艳;田伟强【期刊名称】《医药导报》【年(卷),期】2017(36)1【摘要】Objective To evaluate clinical use of tigecycline in hospital patients. Methods Basic diseases, pathologic examinations, concurrent medication, therapeutic efficacy and side effects of 40 patients in Lishui Central Hospital of Zhejiang Province from January 2012 to December 2014 were analyzed retrospectively. Results The effective rate of patients using tigecycline for anti-infection treatment in hospital was 42. 5%. The rates of rational use, basically rational use and irrational use were 17. 5%, 77. 5% and 5. 0%, respectively. Adverse drug reactions occurred in 6 cases of tigecycline use (15. 0%). Conclusion Clinical use of tigecycline in inpatients was basically reasonable in this hospital. The clinical curative effect of tigecycline was good in a variety of infections caused by sensitive bacteria. However, the incidence of adverse drug reactions was high. Attentions should be paid in clinical application.%目的评价使用替加环素的住院患者临床用药情况.方法采用回顾性研究方法,收集浙江省丽水市中心医院2012年1月-2014年12月使用替加环素住院患者的临床资料(基础疾病、细菌学培养结果、联合用药、疗效、不良反应发生情况等),共40例,并对其应用进行合理性评价.结果该院近3年使用替加环素患者的抗感染治疗有效率为42.5%,合理使用率17.5%,基本合理使用率77.5%,不合理使用率5.0%,出现不良反应6例(15.0%).结论该院住院患者替加环素的使用基本合理;替加环素对各类敏感菌引起的感染临床疗效较好,但不良反应发生率较高,临床应用过程中应引起关注.【总页数】4页(P80-83)【作者】叶伟红;应小飞;傅军霞;郭晶晶;徐艳艳;田伟强【作者单位】浙江省丽水市中心医院药学部,丽水 323000;浙江省丽水市中心医院药学部,丽水 323000;浙江省丽水市中心医院药学部,丽水 323000;浙江省丽水市中心医院药学部,丽水 323000;浙江省丽水市中心医院药学部,丽水 323000;浙江省丽水市中心医院药学部,丽水 323000【正文语种】中文【中图分类】R978;R969.3【相关文献】1.我院2013-2014年720例使用万古霉素患者临床用药分析 [J], 陶娌娜;曲晓宇;张四喜;李雪松2.使用六味地黄汤加减治疗男性骨质疏松症40例临床观察 [J], 杨健3.64例替加环素使用病历临床合理用药评价分析 [J], 吴娜梅;林志航;吴水发;黄晓威;洪珊珊4.临床药师对1例400g超早产儿抗感染治疗的用药分析 [J], 吴小燕;文晓柯;冯彬彬5.使用含替加环素方案挽救性治疗52例脓肿分枝杆菌和龟分枝杆菌感染的临床经验 [J], 刘一典;沙巍因版权原因,仅展示原文概要,查看原文内容请购买。
兴奋剂应该要被禁止吗英语作文
兴奋剂应该要被禁止吗英语作文The use of stimulants is illegal, and the law stipulates that the use of stimulants must be punished. Scientific research has proved that the use of stimulants will cause many direct harm to people's physical and mental health.The harm of doping mainly comes from hormones and stimulants. What is particularly worrying is that many harmful effects only show up after a few years, and even doctors can't tell which athletes are in danger and which won't go wrong for the time being.It is a kind of cheating for athletes to use stimulants. Because the use of illegal drugs and methods will give users an advantage in the competition. This illegal behavior is not in line with the sports ethics of honest and fair competition. Modern sports emphasize the principle of fair competition. Fair competition means "clean competition", proper methods and aboveboard behavior. The use of stimulants is not only against sports laws and regulations, but also against basic sports ethics. Doping makes sports unfair, and athletes are no longer at the same starting point of equality.。
不动杆菌耐药机制及其基因检测进展
to
in—
[9]Bone
and
RC,Balk BA,cara FB,et a1.Definitions for sepsis failure and guidelines for the
use
diseraes[J].Trends
Immunol,2002,23(9):450一
organ
of innoVatiVe
therapies in
sepsis[J].chest,】992,101(3):1644—665. learned[J].Shock,1998,9(6):1—11.
(收稿臼期:2009一05—12)
口] 邸]
姚榛祥.重症胰腺炎现代治疗的认识口].中华肝胆外科
杂志,1999,5(2):76—78.
[10]Deitch
干扰。
对p内酰胺类抗菌药物的耐药机制不动杆菌对p_内酰
胺类抗菌药物的耐药机制包括:产p-内酰胺酶、青霉素结合蛋 白的改变以及外膜蛋白通透性降低。其中产生p内酰胺酶是 最主要的耐药机制。H ejnar等[3]检测结果表明,有49.4%的 菌株产染色体介导的Ampc酶。Magnet等[4]认为高产染色 体介导的Ampc酶是细菌对P内酰胺类抗菌药物耐药的主要 因素。不动杆菌产TEM一1、TEM一2、0XA-21、0xA一37型内酰 胺酶,对氨苄西林、羧基青霉紊、脲基青霉素耐药。不动杆菌主 要产生诱导性AmpC型,AmpD基因进行性突变或因抗菌药 物选择压力造成的突变均导致AmpC的过度表达,同时外膜
manniiJ[J].Antimicro
1201-1203.
Agents
Chemother,1995,39(5):
PCR扩增
扩增目标DNA使用琼脂糖电泳、探针杂交
Tigecycline_220620-09-7_DataSheet_MedChemExpress
Product Name:Tigecycline CAS No.:220620-09-7Cat. No.:HY-B0117Product Data SheetMWt:585.65Formula:C29H39N5O8Purity :>98%Solubility:DMSO 100 mg/mL; Water 100y Mechanisms:Biological Activity:Tigecycline is a first in class broad spectrum antibiotic with activity against antibiotic resistantPathways:Anti-infection; Target:Antibacterial g ;mg/mLTigecycline is a first-in-class, broad spectrum antibiotic with activity against antibiotic-resistantorganisms.Target: Antibacterial Tigecycline is active against a broad range of gram-negative and gram-positive bacterial species including clinically important multidrug-resistant nosocomial and community-acquired bacterial pathogens. Tigecycline has been shown to inhibit the translation elongation step by binding to the ribosome 30S subunit and preventing aminoacylated tRNAs to accommodate in the ribosomal A site[1]. Tigecycline has also been found to be effective for the treatment of community- as well as hospital-acquired and ventilator-associated pneumonia and bacteremia, sepsis with shock and References:[1]. Seputiene V, et al. Tigecycline - how powerful is it in the fight against antibiotic-resistantbacteria? Medicina (Kaunas). 2010;46(4):240-8.[2]. Bhattacharya M, et al. Tigecycline. J Postgrad Med. 2009 Jan-Mar;55(1):65-8.p q p ,purinary tract infections. Tigecycline appears to be a valuable treatment option for the management ofsuperbugs, especially where conventional therapy has failed [2].Fif...[3]. Moreno BB, et al. Tigecycline therapy for infections due to carbapenemase-producing Klebsiellapneumoniae in critically ill patients. Scand J Infect Dis. 2013 Dec 20.Caution: Not fully tested. For research purposes onlyMedchemexpress LLC18 W i l k i n s o n W a y , P r i n c e t o n , N J 08540,U S AE m a i l : i n f o @m e d c h e m e x p r e s s .c o m W e b : w w w .m e d c h e m e x p r e s s .c om。
高剂量替加环素治疗ICU患者肺部感染的临床疗效
高剂量替加环素治疗ICU患者肺部感染的临床疗效
姚芬V,王曦培1,王依凡I,伍政I,兰金花I,王义蓉I,杨敏]•[ 1.广东省人民医院(广东省医学科学院),广东省心血管 病研究所,广东广州510080;2.华南理工大学生物科学与工程学院,广东广州510006]
摘要:目的 探讨高剂量替加环素治疗ICU患者肺部感染的临床疗效和细菌学疗效。方法 选取2018年3月~2020年10月
YAO Fen12, WANG Xipei',WANG Yifad ,WU Zheng1, LAN Jinhua',WANG Yirong1, YANG Min1 * [ l.Guangtfong
Provincial People's Hospital ( Guangdong Academy of Medical Sciences) , Guangdong Cardiovascular Institute, Guangzhou, Guangdong 510080, China ; 2. School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China]
于广东省人民医院ICU病房接受替加环素治疗的肺部感染患者82例,其中接受标准剂量替加环素治疗(负荷剂量100 mg,维 持剂量50 mg ql2h)的患者26例,接受高剂量替加环素治疗(100mgql2h)的患者56例。结果高剂量组的临床有效率和细
菌清除率62.5%(35/56)和50.8%(33/65),均高于标准剂量组38.5%(10/26)和26.7%(8/30),差异有统计学意义(P<0.05)。 临床有效的独殳预测因素是使用高剂量的替加环素(OR = 3.341,95%C/:1.096~ 10.186,P = 0.034)和替加环素的治疗时长(OR =1.249,95%C/: 1.105-1.411,P=0.000 4)o结论对ICU肺部感染患者,高剂量的替加环素以及恰当的治疗时长具有更好的
注射青霉素的英语作文
注射青霉素的英语作文Title: The Significance of Penicillin Injections。
Penicillin, a revolutionary antibiotic, has undoubtedly transformed the landscape of modern medicine since its discovery by Alexander Fleming in 1928. Among its various forms of administration, the injection of penicillin holds particular significance due to its rapid onset of action and effectiveness in combating bacterial infections. Inthis essay, we delve into the importance of penicillin injections, exploring its mechanism of action, therapeutic benefits, and impact on global healthcare.Penicillin injections serve as a cornerstone in the treatment of numerous bacterial infections, ranging from streptococcal pharyngitis to severe cases of pneumonia. Unlike oral administration, which may be hindered byfactors such as poor absorption or patient non-compliance, injections ensure direct delivery of the antibiotic into the bloodstream, maximizing its bioavailability andtherapeutic efficacy. This is especially crucial incritical situations where prompt intervention is essentialto prevent life-threatening complications.The mechanism of action of penicillin involvesinhibition of bacterial cell wall synthesis, leading to osmotic instability and eventual lysis of the microorganism. By targeting a fundamental component of bacterial structure, penicillin exerts a broad spectrum of activity against various gram-positive and some gram-negative bacteria. This mechanism not only curtails the current infection but also helps in preventing the emergence of antibiotic resistance, a pressing concern in modern healthcare.Moreover, penicillin injections are particularlyfavored in scenarios where oral administration isimpractical or contraindicated, such as in patients with gastrointestinal disturbances, compromised immunity, or inability to swallow. In pediatric populations, where accurate dosing and compliance pose challenges, injections ensure precise delivery of the antibiotic, thereby minimizing the risk of treatment failure or development ofresistance.The therapeutic benefits of penicillin injections extend beyond the treatment of acute infections to the prevention of certain infectious diseases. For instance, individuals prone to recurrent streptococcal infections may receive periodic penicillin injections as prophylaxis to reduce the risk of recurrence and associated complications. Similarly, patients undergoing surgical procedures or those with heart valve abnormalities may receive pre-operative or prophylactic penicillin injections to prevent surgical site infections or bacterial endocarditis, respectively.In addition to its clinical efficacy, the affordability and accessibility of penicillin injections make them indispensable in resource-limited settings and developing countries. Unlike newer generation antibiotics that may be cost-prohibitive or unavailable, penicillin remains a cost-effective option for the management of a wide array of bacterial infections. Its stability at room temperature and ease of administration further enhance its suitability for use in remote areas or during humanitarian crises, whererefrigeration or sophisticated medical facilities may be lacking.Despite its numerous benefits, the use of penicillin injections is not without limitations and potential adverse effects. Allergic reactions, ranging from mild rash tolife-threatening anaphylaxis, pose a significant concern, necessitating careful patient evaluation and monitoring prior to administration. Furthermore, over-reliance on penicillin as a first-line antibiotic has contributed to the emergence of resistant bacterial strains, underscoring the importance of judicious antibiotic stewardship and development of alternative treatment strategies.In conclusion, penicillin injections represent a cornerstone in the management of bacterial infections, offering rapid onset of action, broad-spectrum activity, and cost-effective therapy. Their significance transcends clinical boundaries, encompassing preventive, therapeutic, and humanitarian aspects of healthcare delivery. However, their judicious use, coupled with efforts to combat antibiotic resistance, is imperative to ensure theirsustained efficacy and availability for future generations. As we navigate the complexities of infectious diseases in the 21st century, penicillin injections remain a beacon of hope in our arsenal against microbial adversaries.。
替加环素联合亚胺培南对多重耐药及泛耐药鲍曼不动杆菌体外抗菌作用
替加环素联合亚胺培南对多重耐药及泛耐药鲍曼不动杆菌体外抗菌作用蔡静;许静洁;潘海燕【摘要】目的评价替加环素联合亚胺培南对多重耐药及泛耐药的鲍曼不动杆菌体外抗菌活性,探讨联合用药的可行性,为临床抗菌药物的合理使用提供依据.方法收集南京鼓楼医院2015年1-4月临床分离的多重耐药及泛耐药的鲍曼不动杆菌16株,采用棋盘设计微量内汤稀释法测定替加环素和亚胺培南的单药和联合用药的最低抑菌浓度(MIC),并计算联合药敏指数(FIC),从而判断联合效应.结果替加环素单药平均MIC值(MICG)、MIC50 、MIC90分别是1.73,1,4μg·mL-1,亚胺培南单药MICG、MIC50、MIC90分别是31.00,32,64 μg·mL-1,而在联合用药情况下替加环素MICG、MIC50 、MIC90分别是0.24,0.25,0.5μg· mL-1,亚胺培南MICG、MIC50、MIC90分别是8.16,8.00,16.00 μg·mL-1,联合用药与单用药比较均明显降低,HC≤0.5、0.50<FIC≤1分别占37.50%,62.50%,没有无关和拮抗作用.结论替加环素联合亚胺培南对多重耐药及泛耐药的鲍曼不动杆菌体外抗菌效应主要表现在相加作用和协同作用,提示联合用药对多重耐药及泛耐药鲍曼不动杆菌的治疗可能有效.%Objective To evaluate the in vitro antimicrobial activity of tigecycline in combination with imipenem against multi-drug resistant and pan-drug resistant Acinetobacter baumannii isolates,so as to discuss the feasibility of drug combination and provide the basis for chnical rational use of antimicrobial agents.Methods Sixteen multi-drug resistant and pan-drug resistant Acinetobacter baumannii isolates were collected between January and April in 2015 from all kinds of infected specimens of Nanjing Drun Tower Hospital.The protocol was designed by checkerboardmethod,and the minimum inhibitory concentration (MIC) of antibiotics was determined by microdilution broth method,and the fractional inhibitory concentration (FIC) index was calculated according to MIC results.Results The average value of MIC (MICG),MIC50,MIC90 of tigecycline and imipenem single were 1.73,1,4 μg·mL-1and 31.00,32,64 μg·mL-1.When tigecycline was combined with imipenem,MICG,MIC50,MIC90 of tigecycline and imipenem were 0.24,0.25,0.50 μg·mL-1 and 8.16,8.00,16.00 μg·mL-1,pared with the drug single use groups,MIC was significandy decreased in the drug combination group.In 6 strains(37.50%),synergy effect (FIC≤0.5) was observe d,and in 10 strains(62.50%),additive effect (0.5 <FIC ≤ 1) was found.No negative and independent effects were shown.Conclusion Both additive and synergistic action is observed when tigecyclineis combined with imipenem against multi-drug resistant and pan-drug resistant Acinetobacter baumannii isolates.No negative and independent effects are shown.This combination use against multi-drug resistant and pan-drug resistant Acinetobacter baumannii may be an effective therapy for clinical treatment.【期刊名称】《医药导报》【年(卷),期】2017(036)002【总页数】5页(P149-153)【关键词】替加环素;亚胺培南;鲍曼不动杆菌;联合用药;联合药敏指数【作者】蔡静;许静洁;潘海燕【作者单位】南京大学医学院附属鼓楼医院药学部,南京210008;南京大学医学院附属鼓楼医院药学部,南京210008;南京大学医学院附属鼓楼医院药学部,南京210008【正文语种】中文【中图分类】R978;R37DOI 10.3870/j.issn.1004-0781.2017.02.008鲍曼不动杆菌(Acinetobacter baumannii)属于不动杆菌属,是非发酵的革兰阴性菌,可引起各种严重感染,如呼吸机相关性肺炎、菌血症、脑膜炎、尿路感染、外科伤口感染及软组织感染等,主要发生在重症监护室(ICU)危重患者[1],其次为呼吸内科。
青霉素的发现和应用英语作文
青霉素的发现和应用英语作文The Discovery and Application of PenicillinThe discovery of penicillin in 1928 by Scottish bacteriologist Alexander Fleming marked a significant turning point in the history of modern medicine. This revolutionary antibiotic has had a profound impact on human health, saving countless lives and transforming the way we treat infectious diseases. The story behind the discovery and development of penicillin is a testament to the power of scientific inquiry and the remarkable advancements that can arise from serendipitous discoveries.Alexander Fleming was a brilliant scientist who had a keen interest in the field of bacteriology. He was particularly intrigued by the ways in which various microorganisms could interact with one another. In 1928, while conducting research on Staphylococcus bacteria, Fleming noticed that a petri dish containing the bacteria had been accidentally contaminated by a fungus. To his surprise, he observed that the area around the fungus was devoid of bacterial growth, suggesting that the fungus had produced a substance that inhibited the growth of the Staphylococcus bacteria.Fleming recognized the potential significance of this discovery and began to investigate the nature of the substance produced by the fungus. After extensive experimentation, he identified the active ingredient as a compound he named penicillin. Fleming's groundbreaking work demonstrated that penicillin possessed potent antibacterial properties, capable of killing a wide range of pathogenic bacteria without harming human cells.The discovery of penicillin was a remarkable achievement, but it was only the beginning of a long and arduous journey towards its widespread clinical application. The challenges that followed were numerous and complex. Firstly, Fleming faced the daunting task of purifying and stabilizing the penicillin compound, as it was highly unstable and difficult to produce in large quantities. Despite his efforts, the initial supply of penicillin was limited, and it was not until the early 1940s that researchers at Oxford University, led by Howard Florey and Ernst Chain, were able to develop a more reliable and scalable production process.The outbreak of World War II further complicated the development of penicillin, as the demand for the drug skyrocketed due to the urgent need to treat wounded soldiers. The limited supply of penicillin during this time meant that it had to be rationed and prioritized for the most critical cases. However, the wartime conditions also provided a unique opportunity to test theeffectiveness of penicillin in a clinical setting, and the results were nothing short of remarkable.Penicillin proved to be a game-changer in the treatment of a wide range of bacterial infections, including pneumonia, gonorrhea, and even life-threatening conditions like sepsis. Its ability to target and destroy harmful bacteria without harming human cells made it a revolutionary medical breakthrough. The widespread use of penicillin during the war years not only saved countless lives but also paved the way for the development of other antibiotics and the advancement of modern antimicrobial therapy.The impact of penicillin on human health cannot be overstated. It has been credited with significantly reducing mortality rates from infectious diseases and has played a crucial role in the treatment and prevention of numerous illnesses. The discovery of penicillin also opened the door to a new era of antibiotic development, leading to the creation of a vast array of antimicrobial agents that have transformed the way we approach the treatment of bacterial infections.However, the widespread and often indiscriminate use of antibiotics has also led to the emergence of antibiotic-resistant bacteria, posing a significant threat to global health. This challenge has highlighted the importance of responsible antibiotic stewardship and theongoing need for research and development of new antimicrobial therapies.Despite these challenges, the legacy of Alexander Fleming's discovery of penicillin remains a testament to the power of scientific inquiry and the potential for transformative breakthroughs in the field of medicine. The story of penicillin serves as a shining example of how a single discovery can have far-reaching and profound implications, changing the course of human health and well-being for generations to come.。
多黏菌素的临床应用
单药治疗、联合治疗
疾病改善治愈率,存活率,联合用药 均是优于单独用药
Antimicrob Agents Chemother. 2014;58(4):2322-8. doi: 10.1128/AAC.02166-13. Epub 2014 Feb 10.
英国Jim O’Neill爵士2016年5月发表《全球抗菌素耐药回顾》报告 及建议,提出一个全面行动计划,呼吁不仅是一个国家、而是 全世界合力预防耐药感染,战胜日益严重的抗菌素耐药威胁。
全球行动方案:五大策略性目标
• To improve awareness and understanding of antimicrobial resistance
耐碳青霉烯类革兰阴性菌感染的治疗药物与联合抗菌策略进展,中华医院感染学杂志2016年第26卷第9期2152-2456 Tumbarelli M.ECCMID 10-13 May 2014 Barcelona Clin Microbiol Rev. 2012 Oct;25(4):682-707. doi: 10.1128/CMR.05035-11.
各种抗生素对产耐碳青霉烯酶的作用
A:≥2种抗菌药联合包括碳青霉烯类 B:≥2种抗菌药联合不包括碳青霉烯类 C:单用氨基糖苷类 D:单用碳青霉烯类 E:单用替加环素 F:单用黏菌素 G:无有效治疗药物
Clin Microbiol Rev. 2012 Oct;25(4):682-707. doi: 10.1128/CMR.05035-11.
我国 G- 菌检出率呈逐年上升趋势
革
常见发生耐碳青霉烯的病原菌:
不动杆菌、铜绿假单胞菌、肠杆菌科细菌、克雷伯菌
耐碳青霉烯的肠杆菌的主要耐药机制为产碳青霉烯酶 即CPE(carbapenamase-producing enterobacteriacae
2012 tigecycline-induced acute pancreatitis about two cases and review of the literature
TigeCyCline-induCed ACuTe pAnCreATiTis: ABouT Two CAses And review of The liTerATure229 Case reportTigecycline-induced acuTe pancreaTiTis:abouT Two cases and review ofThe liTeraTureMarot J-C1, Jonckheere S1 , Munyentwali H2, Belkhir L1, Vandercam B1, Yombi JC11Department of Internal Medecine and Infectious Diseases, Cliniques Universitaires St Luc,2Departement of Nephrology, Cliniques Universitaires St LucCorrespondence and off print requests to: Jean Yombi, E-mail: jean.yombi@uclouvain.beabsTracTTigecycline (formerly GAR-936, Tygacyl®) is the first glycylcycline antibiotic available for clinical use. It has an expanded broad-spectrum antibiotic activity. Phase III studies have identified gastrointestinal side-effects, espe-cially nausea and vomiting. as the most common adverse events. Few cases of acute pancreatitis (AP) have been described in the literature. We report two new cases of mild tigecycline-induced pancreatitis. Tigecycline was given for soft-tissue infection in both cases. Symptoms such as nausea, vomiting and mostly abdominal pain occurred within 5 days after starting Tigecycline. Pancre-atic enzymes elevation occurred five to six days after ini-tiation of treatment, and resolved within a week after drug-discontinuation. Diagnosis of mild pancreatitis was confirmed after performing CT-Scan of the abdomen in both cases. We take this opportunity to review the litera-ture about this potentially serious side-effect induced by tigecycline.Key words:tigecycline, acute pancreatitis, adverse events inTroducTionTigecycline (formerly GAR-936, Tygacyl®) is a 9-t-butylgly-cylamido semi-synthetic derivative of minocycline and is the first glycylcycline antibiotic available for clinical use (1-2). This new antibiotic has been developed in order to overcome the two main mechanisms of tetracyclines resistance (ribosomal and efflux pump). Tigecycline has an expanded broad-spec-trum antibiotic activity. Several large-scale evaluations on many types of bacteria indicate that tigecycline is highly active in vitro against most common Gram-positive and Gram-negative pathogens, anaerobes and atypical patho-gens, including those frequently demonstrating resistance to multiple classes of antimicrobials. It is currently indicated for treatment of complicaterd skin and soft tissue infections (cSSSI), complicated intra-abdominal infections (cIAI) as well as community-aquired pneumonia (CAP). Tigecycline is most likely excreted in bile, its mean t1/2 has been estimated to 40 to 60 h and the drug seems to be well distributed into various tissues. Phase III studies have identified gastrointesti-nal side-effects as the most common adverse events, espe-cially nausea and vomiting (20-45%) (3-5). The occurrence depends on the daily dosage but not on the infusion duration (6-7). These side-effects require anti-vomiting treatments in 30% of cases and discontinuation in only 4% (8). Few cases of acute pancreatitis (AP) have been described in the literature (9-13). We report two new cases of tigecycline-induced mild AP, and take this opportunity to review the literature about this potentially serious side-effect.case 1A 64-years old man was admitted to a surgical ward for treatment of an infection of an arteritic lesion of the second toe of the right foot. He had a previous history of IgE-medi-ated allergy to penicillin. The patient had been diagnosed with rheumatoid arthritis 15 years before admission. Hyper-tension was diagnosed in 2008, an ischaemic colitis in 2008 and an atheromatous arteritis of the lower limbs in 2008 as well. He had a history of dyslipidemia for which he was treated (Total cholesterol level of 188 mg/dl (NL 50-200), Tri-glycerides at 119 mg/dl (NL 0-200 mg/dl)). The patient did not drink alcohol at all and did not smoke. Upon admission the patient treatment was: methylprednisolone 4 mg/day, perindopril 7.5 mg/day, rosuvastatine 10 mg/day, ezetimibe 10 mg/day, AAS 100 mg/day, and allopurinol 100 mg.doi: 10.2143/ACB.67.3.2062663 Acta Clinica Belgica, 2012; 67-3Acta Clinica Belgica, 2012; 67-3revealed an acute pancreatitis, with amylases at 552 UI/L (NL 13-66 UI/L) and lipases at 1660 UI/L (Nl 2-62 UI/L), with a known and stable anicteric cholestasis. The abdominal ultra-sound showed no biliary duct dilatation. The next day, he developed abdominal pain, and was treated with intravenous analgesics on top of fasting and intravenous fluid. A CT-Scan was performed and showed an acute pancreatitis (grade D on Balthazar score, no necrosis visible without contrast injection). It was recommended that tigecycline be discontinued. Treat-ment was shifted to moxifloxacine 400 mg/day for a total of 6 weeks. Shortly after tigecycline discontinuation (i.e. 5 days), the patient’s symptoms resolved and the enzymes returned to baseline (Figure 1). He was discharged with a low-fat diet for 2 weeks. On follow-up, one month after discharge, a control CT-Scan of the abdomen was reported normal. Though tacroli-mus or lisinopril could have been suspected upon diagnosing AP, the patient had been on these medications for several years, and evolution upon discontinuation of tigecycline seemed to confirm the hypothesis of tigecycline-induced AP.discussionThe diagnosis of drug-induced acute pancreatitis is diffi-cult to establish, mainly due to the absence of cause-specific diagnostic tests. Therefore, it is usually based on the following criteria:1. Acute pancreatitis occurring during the administration ofa drug,2. All other common causes are excluded,3. Symptoms of acute pancreatitis disappear after drug with-drawal, and4. Symptoms recur after a re-challenge of the suspecteddrug (14).It seems obvious that it is not always possible to establisha definitive diagnosis of drug-induced AP immediately, and a“”second-look” with the knowledge of the subsequentpatient’s history may be necessary. Due to ethical considera-tions, re-challenge is usually not performed, unless the causeof the first AP episode was not properly recognized. Review-ing the literature, we found five reported cases of tigecycline-induced AP (see Table 1, (9-13)). Among the seven reportedcases (five published and our own two cases) of suspectedtigecycline-induced AP, only one patient meets the re-chal-lenge criteria (unintended) (13), the six others fulfil the 3 firstcriteria only. The correlation can be difficult to establish astreatment might be administered in complex situations, topatients with potentially confounding past medical history oron other potentially AP-inducing medications as was our sec-ond patient (tacrolimus, lisinopril) (15).Acute pancreatitis was not listed in the product labelwhen the drug was originally approved, but in july 2006Wyeth (manufacturer) updated the product label to includeAP as one of the post-marketing adverse events (16). Interest-ingly, it is to be noted that, though tigecylcine was registeredfor treatment of cIAI, treatment for infectious complicationsof acute pancreatitis was excluded from the original studies .These exclusion criteria might have emerged from the similar-ity between tigecycline and other tetracyclines. Erythromycin,as a potent prokinetic agent that stimulates motilin release,may indeed cause acute pancreatitis by causing spasm of theA swab of the foot lesion was positive for staphylococcusaureus susceptible to oxacillin, and resistant to clindamycineand penicillin. The patient was treated for this infectionwith tigecycline at 100 mg IV for the first dose, then 50 mg12-hourly IV, started upon admission to the hospital. A sympa-tholysis was performed 2 days after admission but on day 3,the toe necrosis was extensive and the second toe was ampu-tated, a popliteal-tibial bypass being done at the same time.Six days after admission, the patient developed nausea, epi-gastric pain, and lab studies showed elevation of the lipaseenzyme at 936 U/mL (NL 2-62 UI/L) with triglycerides levels of119 mg/dl (NL 0-200 mg/dl). An abdominal CT-Scan showedan oedematous pancreatitis (grade D on Balthazar score) with-out any biliary duct pathology. Tigecycline was discontinuedon day 6, pancreatitis resolved on day 9 (Figure 1). The patientwas discharged from the hospital on day 10.case 2A 58-years-old kidney (chronic interstitial nephritis) andliver (Hepatitis C-related cirrhosis) transplant recipient diabeticman, with peripheral arterial disease, was first admitted toour ward due to necrosis of a toe that required a phalanxamputation. He had no history of alcohol intake, and nohyperlipemia. At the time of admission, his medicationincluded clopidogrel 75 mg , AAS 80 mg, tacrolimus 2 mg,lisinopril, bisoprolol, allopurinol, omeprazole and actrapidinsulin. A few weeks later, because of an unfavourable changein the scar, he was readmitted to our unit for further surgerywith amputation of the remaining phalanx. According to thesurgeon, the local bone was necrotic and infected, and couldnot be completely removed. We initiated an antibiotic therapywith Piperacillin-Tazobactam after intraoperative bacteriologi-cal samples were taken. Culture of the bone biopsies showed2 types of coagulase-negative Staphylococci – staphyloccusscleiferi methicillin-resistant, and staphylococcus lugdunensismethicillin-sensitive, thus Vancomycin was added two daysafter surgery. Five days later, the residual serum Vancomycin-level was toxic (35.91 mg/l). For an easier management of theantibiotic therapy, we decided to replace the associationPiperacillin-Tazobactam and Vancomycin with Tigecycline.One week after the initiation of Tigecycline, the patient pre-sented with nausea, vomiting and loss of appetite. Lab studiesTime (days)1Lipase Level EvolutionLipase(UI/mL)Figure 1:lipase level evolution – peak of enzyme elevation on day 2,tigecycline was discontinued on day 1 of enzyme elevation inboth cases.Acta Clinica Belgica, 2012; 67-3TigeCyCline-induCed ACuTe pAnCreATiTis: ABouT Two CAses And review of The liTerATure 231T a b l e 1 r e v i e w o f c u r r e n t c a s e s r e p o r t o f t i g e c y c l i n e -i n d u c e d a c u t e p a n c r e a t i t i sc a s e r e p o r t sa g eg e n d e rn a u s e a / v o m i t i n g / a b d o m i n a l p a i no n s e t o f s y m p t o m sd u r a t i o n o f t y ge c y c l i n e p e a k of a m y l a s e /l i p a s e (i u /l )a p s e v e r i t yT i m e T o r e c o v e r y (s y m p t o m s )T i m e T o r e c o v e r y (e n z y m e s l e v e l s )i n d i c a t i o n o f t i g e c y c l i n ei m a g i n gG i l s o n e t a l ,200835m a l e y e s13 d a y s , a b d o m i n a l p a i n15 d a y sN D /1000m i l d2 D a y s43 D a y s O s t e i t i sC T -S c a nL i p s h i t z e t a l64f e m a l ey e s 14 d a y s , e p i g a s t r i c p a i n14 d a y s806/1406m i l d3 D a y s5 D a y sP r o s t h e t i c j o i n t i n f e c t i o nC T -S c a nM a r s h a l l ,200955f e m a l ey e s , s t a r t f r o m d a y 310 d a y s , u n c o n t r o l l e de m e s i s10 d a y s 190/160m i l d2 D a y s7 D a y sS o f t T i s s u e I n f e c t i o nC T -S c a nH u n g e t a l , 200969f e m a l ey e s s t a r t f r o m d a y 37 d a y s , a b d o m i n a l p a i n8 d a y s926/749m i l d 5 D a y s5 D a y sS o f t t i s s u e i n f e c t i o n / V a s c u l a r g r a f t i n f e c t i o nn o n eP r o t -l a b a r t h e , e t a l 2010 9m a l e y e s 14 d a y s , a b d o m i n a l p a i n 8 w e e k s N l /6033 D a y s 5 D a y s B a c t e r i e m i a / A r t h r i t i s C T -S c a nC a s e 164m a l e y e s 6 d a y s , e p i g a s t r i c p a i n 6 d a y s 750/936m i l d 4D a y s 18 D a y s S o f t t i s s u e i n f e c t i o nC T -S c a n C a s e 258m a l e y e s7 d a y s , + a b d o m i n a l p a i n o n d a y 88 d a y s 552/1660m i l d 5 D a y s 4 D a y sS o f t t i s s u e i n f e c t i o n / O s t e o m y e l i t i sC T -S c a nsphincter of Oddi, leading to abrupt pancreatic-duct hyper-tension and pancreatitis (17).The risk potential of common drugs is difficult to estab-lish. Generally, it is estimated from the absolute numbers of published cases. In the critical reviews of Mallory and Kern (18) and, more recently, McArthur (14), the potential of a drug to induce AP was evaluated as definite, probable, or possible. The seven cases, including our own two cases of mild AP , meet the criteria of probable according to McArthur’s criteria.The mechanism of tigecycline or tetracycline-induced pancreatitis is still unknown. There have been at least three different mechanisms hypothesised, including (i) formation of a toxic metabolite, (ii) hypertriglyceridemia and (iii) a high biliary concentration. Tigecycline is structurally related to minocycline and shares similar pharmacokinetic properties and side effects with tetracyclines, (nausea, vomiting and diarrhea). Steinberg (19) hypothesised that accumulation of an unidentified toxic metabolite may be the cause of tetracy-cline-induced pancreatitis. Tetracycline-induced hypertriglyc-eridemia, and subsequent pancreatitis were proposed by Elmore and Rogge as yet another potential mechanism (20). Tetracycline inhibits protein synthesis by binding to the 30S ribosomal subunit in the messenger ribonucleic acid (mRNA) translation complex. Blockage of protein synthesis could result in an accumulation of defective proteins within hepa-tocytes. This inhibits the release of triglycerides from the liver, which may lead to pancreatitis. Gilson et al. (9) suggested that high biliary concentration of tetracycline might be associated with tetracycline-induced pancreatitis. Concentrations of tigecycline in bile (median 75.2 mg/L, range 15.9-1150 mg/L) were found to be several logs greater than concurrent serum concentration (median 0.112 mg/L, range 0.042-0.25 mg/L) after a single 100 mg dose. The mean and median bile-to-serum 24-h-area under the concentration-time curve (AUC0-24) ratios were 537 and 368, respectively (16).In the five cases of tigecycline-induced pancreatitis pub-lished in the literature, all patients reported nausea and abdominal pain following initiation of tigecycline, and the onset of acute pancreatitis within 6-14 days. This is a more rapid onset than typically seen with tetracycline-induced pancreatitis in patients without liver disease, with erythromy-cin-related latency to AP ranging from 1 to 12 weeks (17, 21). Symptoms such as severe nausea and abdominal pain resolved within 2-9 days after withdrawal of the drug. Amy-lase and lipase levels ranged from two to more than five times the upper limit of normal range. Time to recovery of abnormal pancreatic enzymes varied among individuals and did not correlate with the degree of enzyme elevation. This pattern was similar to that seen with tetracycline-induced pancreatitis. All signs and symptoms of pancreatitis resolved and no long-term complications associated with tigecycline-induced acute pancreatitis were documented.Only five cases of tigecycline-induced AP have been reported. However, based on unpublished data, the esti-mated incidence of tigecycline-induced pancreatitis is around 0.1-1% (22), but it is not consistent throughout four current clinical trials. In a Phase 3 trial on tigecycline for treatment of complicated skin and soft tissue infections, tigecycline was associated with a 3.1% incidence of amylase elevation, com-pared with 1.1% in the control group (vancomycin/aztreo-nam) (3). In the same study, 4.5% of patients had abdominal232TigeCyCline-induCed ACuTe pAnCreATiTis: ABouT Two CAses And review of The liTerATureActa Clinica Belgica, 2012; 67-3 4. Breedt J, Teras J, Gardovskis J, Maritz FJ, Vaasnaand T, Ross DP, et al. Safety andefficacy of tigecycline in treatment of skin and skin-structure infections: results of 2 double – blind phase III comparison study with vancomycin-aztreonam.Antimicrob. Agents Chemother 2005; 49: 4658-4666.5. Ellis-Grosse Ej, Babinschak T, dartois N, Rose G, loh E.The efficacy and safety oftigecycline in the treatment of skin and skin-structure infections: results of2 double-blind phase III comparison studies with vancomycin-aztreonam. Clininf dis 2005; 41(suppl 5): S341-353.6. Muralidharan G, Micalizzi M, Speth J, Raible D, Troy S. Pharmacokinetics of tige-cycline after single and multiple doses in healthy subjects. Antimicrob Agents Chemother 2005; 49: 220-229.7. Chopra I, Roberts M. Tetracycline antibiotics: mode of action, applications,molecular biology, and epidemiology of antibacterial resistance. Microbiol Mol Biol rev 2001; 65: 232-260.8. Zhanel GG, Homenuik k, Nichol K, Noreddin A, Vercaigne L, Embil J, et al.The glycylcyclines, a comparative review with tetracyclines. drugs 2004; 64: 63-68.9. Gilson M, Moachon L, Jeanne L, Dumaine V, Eyrolle L, Morand P, et al. Acutepancreatitis related to tigecycline: case report and review of the literature.scand J infect dis 2008; 40: 681-683.10. Lipshitz J, Kruh J, Cheung P, Cassagnol M. Tigecycline-induced pancreatitis.J Clin gastroenterol 2009; 43: 93.11. Marshall SR. Tigecycline-induced pancreatitis, hosp pharm 2009; 44: 239-241.12. Hung WY, Kogelman L, Volpe G, Iafrati M, Davidson L. Tigecycline-induced pan-creatitis: case report and literature revizw. int Journal of Antimicrobiol Agents 2009; 34: 486-489.13. Prot-Labarthe, Youdaren R, benkerrou M, basmaci R, Lorrot M. Pediatric acutepancreatitis related to tigecycline. The pediatric infectious disease Journal, 2010;29: 890-891.14. McArthur KE. Review article: drug-induced pancreatitis. Aliment pharmacol Ther.1996; 10(1): 23-38.15. Trivedi et al. Drug-induced pancreatitis: an update, J Clin gastroenterol 2005;39(8): 709-716.16. Wyeth Pharmaceutics. Tygacil® (package insert). Philadelphia, PA: WyethPharmaceutics; 2009.17. Badalov N et al. Drug-induced acute pancreatitis: an evidence-based review,Clinical gastroenterology and hepatology 2007; 5: 648-661.18. Mallory A, Kern F Jr. Drug-induced pancreatitis: a critical review. gastroenterol-ogy 1980; 78: 813-820.19. Steinberg WM, Acute drug and toxin induced pancreatitis. hosp pract 1985; 20:95-102.20. Elmore MF, Rogge JD. Tetracycline-induced pancreatitis. gastroenterology 1981;81: 1134-1136.21. Nicolau DP, Mengedoht DE, Kline JJ. Tetracycline-induced pancreatitis. Am Jgastroenterol 1991; 86(11): 1669-1671.22. Unpublished observations. Tygacyl, Second Periodic Safety Update Report-Preliminary assessment report. Rapporteur: Calvo G. Pharmacovigilance asses-sors: Macia Ma, Martin-Serrano G. Clinical assessors: Fernandez-Cortizo MJ.Period cover by this PSUR: 15 juin 2006 to 14 december 2006. Date of the assessment report: 17 April 2007.23. Florescu I, Beuran M, Dimov R, Razbadauskas A, Bochan M, Fichev G, et al. Effi-cacy and safety of tigecycline compared with vancomycin or linezolid for treat-ment of serious infections with methicillin-resistant Staphylococcus aureus or vancomycin-resistant enterococci: a Phase 3, multicenter, double-blind, ran-domized study. J Antimicrob Chemother 2008; 62(S1): i17-i28.24. Vasilev K, Reshedko G, Orasan R, Sanchez M, Teras J, Babinchak T, et al. A Phase3, open-label, non-comparative study of tigecycline in the treatment of patients with selected serious infections due to resistant Gram-negative organisms including Enterobacter species, Acinetobacter baumannii and Klebsiella pneu-moniae. J Antimicrob Chemother 2008; 62(S1): i29-i40.25. Bergallo C, Jasovich A, Teglia O, Oliva ME, Lentnek A, Wouters L, et al. Safety andefficacy of intravenous tigecycline in treatment of community-acquired pneu-monia: results from a double-blind randomized Phase 3 comparison study with levofloxacin. diagn Microbiol infect dis 2009; 63: 52-61.26. Cai Y, et al. Systemic Review and Meta-Analysis of the Effectiveness and Safetyof Tigecycline for the Treatment of Infectious Diseases. AAC 2011; 55(3): 1162-1172.pain in the tigecycline group compared with 2.5% in the con-trol group. On the other hand, neither abdominal pain nor abnormal pancreatic enzymes were reported in the three remaining Phase 3 trials (23-25). In our institution, since tige-cycline was introduced to the pharmacy (i.e. 1245 adminis-tered doses for 53 patients),only two cases of AP are reported.Acute pancreatitis is only one amongst the many side-effects that have triggered a rising concern on tigecycline’s safety. According to Cai et al., though tigecicline has similar effectiveness as other combined antibiotherapies, it is associ-ated with a significant higher risk of adverse events. This meta-analysis on 4,441 patients showed an odd ration to developing AEs of 1.33 overall, and 2.41 on digestive AEs, between the tigecycline group vs. its comparators. Though the mortality was numerically higher in the tigecycline group, data was not sufficient to demonstrate a significant difference in mortality between the groups (26).conclusionTigecycline-induced acute pancreatitis is still considered a rare phenomenon. There are insufficient data to identify significant predicting factors of tigecycline-induced pancre-atitis. Currently, the manufacturer does not recommend rou-tine monitoring of serum amylase and lipase. However, it is important for clinicians to monitor symptoms of abdominal pain during treatment with tigecycline and to have a low threshold to investigate amylase and lipase levels if the clini-cal presentation is in keeping with acute pancreatitis. In absence of other causes of acute pancreatitis, knowledge of this adverse effect of tigecycline is critical to promote prompt and appropriate management of pancreatitis, including drug discontinuation. This potentially severe side-effect should also further emphasise the need for cautious use of tigecy-cline, as it is significantly associated with a higher risk of developing adverse events compared to other similarly effec-tive antibiotic treatments.conflicTs of inTeresT: Nonereferences1. Noskin GA. Tigecycline: a new glycylcycline for treatment of serious infections,Clin infect dis 2005; 41 (5) (2005): S303-S314.2. Pankey GA. Tigecycline. J Antimicrob Chemother 2005; 56: 470-480.3. Sacchidanand S, Penn RL, Embil JM, Campos ME, Curcio D, Cllis-Grosse E et al.Efficacy and safety of tigecycline monotherapy compared with vancomycin plus aztreonam in patients with complicated skin and skin structure infections: results from a phase III randomized double-blind trial. int J infect dis 2005; 9: 251-261.Reproduced with permission of the copyright owner.Further reproduction prohibited without permission.。
替加环素与泰能比较治疗鲍曼
ARTICLEEfficacy of tigecycline vs.imipenem in the treatmentof experimental Acinetobacter baumannii murine pneumoniaC.Pichardo &M.E.Pachón-Ibañez &F.Docobo-Perez &R.López-Rojas &M.E.Jiménez-Mejías &A.Garcia-Curiel &J.PachonReceived:31July 2009/Accepted:1February 2010/Published online:25February 2010#Springer-Verlag 2010Abstract The in vivo activity of tigecycline was evaluated in an experimental pneumonia model (C57BL/6mice)by Acinetobacter baumannii .Two clinical strains were used:minimum inhibitory concentrations (MICs)of imipenem and tigecycline 1and 2µg/mL (imipenem-susceptible,IPM-S),and 8and 2µg/mL (imipenem-intermediate,IPM-I),respectively.For imipenem (30mg/Kg),∆T/CMI (h)were 1.04and 0.51for IPM-S and IPM-I,respectively.For tigecycline (5mg/Kg),the area under the concentration –time curve (AUC)/MIC 0–24h (serum and lung)were 9.24and 4.37(for the two strains),respectively.In the efficacy experiments with the IPM-S,imipenem (log CFU/g 3.59±0.78,p =0.006)and tigecycline (2.82±1.2,p =0.054)decreased the bacterial counts in lungs with respect to its controls;with the IPM-I,both imipenem (1.21±0.52,p =0.002)and tigecycline (3.21±0.28,p =0.035)decreased the bacterial counts with respect to the controls.In the survival experiments,with the IPM-S,the mortality was the same in the control (67%)and in the tigecycline (77%)groups,and imipenem reduced it (21%,p =0.025);with the IPM-I,the mortality was the same in the control (87%)and in thetigecycline (85%)groups,and imipenem (0%)reduced it (p <0.001).In summary,the present study shows that tigecycline is less efficacious than imipenem in the treatment of experimental A.baumannii pneumonia caused by IPM-S and IPM-I strains.IntroductionAcinetobacter baumannii is a common nosocomial patho-gen,especially in intensive care units,causing a great number of clinical conditions,being pneumonia or bacter-emia the more frequent infections [1,2].In the last several years,multi-resistant strains are frequent [1,3]and,with the recent reports of outbreaks of colistin-resistant A.baumannii isolates[4],new antimicrobial agents have been searched.Recent studies have shown that tigecycline exhibits potent activity against organisms isolated from hospitalized patients,including multidrug-resistant nonfermentative gram-negative bacilli,among others [5,6].Specifically,tigecycline was in vitro-active against A.baumannii strains,including those resistant to imipenem,being bacteriostatic in the time –kill studies [7].In the treatment of multi-resistant A.baumannii ventilator-associated pneumonia (VAP),three patients were cured with tigecycline in monotherapy and 15out of 19patients (78.9%)were cured when treated with tigecycline plus imipenem and/or colistin [8].Data from more studies are needed before tigecycline can be recommended for the treatment of Acinetobacter infections.The purpose of this study is to compare the efficacy of tigecycline and imipenem in a murine pneumonia model caused by A.baumannii strains susceptible and intermedi-ate to imipenem.C.Pichardo (*):M.E.Pachón-Ibañez :F.Docobo-Perez :R.López-Rojas :M.E.Jiménez-Mejías :A.Garcia-Curiel :J.PachonInstituto de Biomedicina de Sevilla,Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla,Avda.Manuel Siurot s/n,41013Seville,Spaine-mail:cristina.pichardo.exts@juntadeandalucia.esC.Pichardo :M.E.Pachón-Ibañez :F.Docobo-Perez :R.López-Rojas :M.E.Jiménez-Mejías :A.Garcia-Curiel :J.PachonSpanish Network for Research in Infectious Diseases(REIPI —ISCIII —RD06/0008),Instituto de Salud Carlos III,Madrid,SpainEur J Clin Microbiol Infect Dis (2010)29:527–531DOI 10.1007/s10096-010-0890-6Materials and methodsWe used two A.baumannii strains collected from blood cultures,corresponding to the more prevalent clones isolated in our hospital:imipenem-susceptible strain (IPM-S,953,minimum inhibitory concentration[MIC] imipenem=1mg/L,tigecycline=2mg/L),and imipenem-intermediate strain(IPM-I,1514,MIC imipenem=8mg/L, tigecycline=2mg/L).Imipenem(laboratory standard powder)and imipenem plus cilastatin(IPM)were obtained from Merck Sharp& Dohme(Madrid,Spain)for the in vitro and the in vivo experiments,respectively,and tigecycline(TGC)from Wyeth-Ayerst(Pearl River,NY,USA).The animals used were immunocompetent C57BL/6 female mice weighing14–16g(University of Seville’s facility)and had a sanitary status of murine pathogen-free (MPF).Drug pharmacokineticsThe plasma levels of imipenem were determined after the administration of a single intramuscular(im)dose of 30mg/Kg.The plasma and lung(homogenate)levels of tigecycline were determined after the administration of a single subcutaneal(sc)dose of5mg/Kg.Blood and lungs were extracted from three anesthetized mice per time-point. The total plasmatic drug concentrations and the total drug concentrations in the lung were measured for triplicate by the bioassay method,using Micrococcus luteus ATCC9341 for imipenem and Bacillus cereus ATCC9634for tigecy-cline as the indicator strains.The intraday and interday variation of the assays were2.6%±2.4%and3.2%±1.9% for imipenem,and 3.5%±2.6%and 4.5%±1.1%for tigecycline;the linearity(r2)of the assay was0.91±0.02 and0.92±0.32,respectively,and the lower limits of detection were0.01and0.1mg/L.The maximum plasma concentration(C max,mg/L)and terminal half-life(t1/2,h) were calculated.The time during which the plasma concentration remained above the MIC(ΔT/MIC,h)was estimated by extrapolation from the regression line of plasma elimination.Experimental pneumonia model in miceA modification of the Esposito and Pennington model performed by our group[9]was used to produce pneumo-nia.The animals were inoculated with50μl of the A. baumannii bacterial suspension,with a final inoculum size of approximately108CFU/mL.The treatment was commenced at4h after the inoculation,grouping the mice in each of the following treatment groups over a period of 72h:(a)controls,no treatment;(b)tigecycline,10mg/Kg/d/sc,b.i.d.;and(c)imipenem,120mg/Kg/d/im,t.i.d.(a quarter of the total dose at8:00and14:00h,and the half of the total dose at20:00h).Two types of experiments were performed:survival and bacterial clearance from lung studies.Survival experimentsBecause in this model the mortality in the control group is near100%at72h,the influence on the mortality of the different treatments was evaluated in this period,in groups of15mice.Efficacy experimentsTo evaluate the efficacy of antimicrobials in the clearance of bacteria from lungs,efficacy experiments were per-formed.Groups of five mice were sacrificed at4h after the infection,before the beginning of treatments(control groups),and groups of five mice were sacrificed every 24h,4h after the last dose in the case of IPM and12h after the last dose in the case of tigecycline(treatment groups).At these time-points,the surviving mice were sacrificed and thoracotomy was carried out;the lungs were removed,weighed,and processed for quantitative cultures. The results were expressed as the log10CFU/g of tissue.In order to confirm that imipenem and tigecycline were not toxic to the animals,groups of ten non-infected mice were each given the antibiotics for72h.The use of the experimental pneumonia model was approved by the Ethics Committee of the Hospital Universitario Virgen del Rocío, Sevilla,Spain.Statistical analysisThe numbers of surviving animals were evaluated with Fisher’s exact test.The CFU/g of lung tissue was analyzed using the Kruskal–Wallis test.The SPSS v13.0statistical package(SPSS Inc.,Chicago,IL,USA)was used.A p-value<0.05was considered to be significant.Results and discussionThe pharmacokinetic/pharmacodynamic parameters of each antimicrobial drug(C max,t1/2,AUC,∆T/MIC,and AUC0–24h/ MIC)are shown in Table1.In the survival experiments(Table2),with both strains IPM-I and IPM-S,only imipenem reduces the mortality with respect to the control groups(0and21%in the imipenem groups and87and67%in the controls, respectively,p≤0.025);imipenem was better than tigecy-cline in both groups(p≤0.007).In the bacterial clearance from the lungs experiments (Fig.1),with the IPM-I strain,for the imipenem-treated group,there was a decrease of the bacterial counts from controls(p=0.002).In the tigecycline-treated group,the treatment cleared the lungs compared to the control group (p=0.035).With the IPM-S strain,in the imipenem-treated group,there was a decrease of the bacterial counts(p= 0.006)and in the tigecycline-treated group,the bacterial lung concentration decreased to2.82log CFU/g(p=0.054).These results show that,in general,imipenem is more active than tigecycline in this experimental pneumonia model caused by A.baumannii with the doses examined. Tigecycline decreased the bacterial count in the lung for the strain intermediate to imipenem with respect to the controls. However,in the in vivo experiments performed with the strains susceptible and intermediate to imipenem,imipenem was better than tigecycline regarding the mortality and clearance of bacteria from the lungs.Different studies in experimental A.baumannii pneumo-nia models had shown that imipenem was the most active antimicrobial,including infections caused by susceptible and intermediate strains[9,10].Different works showed the high in vitro activity of tigecycline from multi-resistant pathogens,including A.baumannii[5,6],which is of particular importance because of the high frequency of multi-resistant strains[1,3],with a high number of strains only susceptible to colistin[1,11,12]or the appearance of pan-resistant A.baumannii isolates[13,14].In a previous in vitro study on the activity of imipenem and tigecycline against49isolates of A.baumannii,the MIC90values were 128and2mg/L,respectively;however,in the time–kill studies,tigecycline was bacteriostatic[7].The limited efficacy of tigecycline in the experimental pneumonia model may be explained by the in vitro bacteriostatic instead of bactericidal activity against A.baumannii.Tigecycline has high penetration in tissues[15,16];thus, we found a serum and lung C max of1.92mg/L and8.29μg/ g,respectively.These figures are higher than those found in humans after doses of50and100mg,which show serum C max values of0.38and0.91mg/L[15,17],respectively, and the mean and median concentration values of tigecy-cline in the lungs after100mg were0.50and0.31μg/g (range:0.11–1.89μg/g)[18].There are no definitive data regarding the pharmacodynamic variables predicting the in vivo efficacy of tigecycline.Several studies had addressed this issue,both in clinical trials and in experimental models of infection[15,16].Some authors suggest that,although tetracyclines do not exhibit concentration-dependent kill-ing,if the antimicrobial agent has a moderate to prolonged postantibiotic effect(PAE),the time of exposure is less important and the AUC/MIC ratio is the best pharmacoki-netic/pharmacodynamic parameter correlating with the therapeutic efficacy of these drugs[19].Data obtained in animal models[17,20]suggest that tigecycline activityTable1Pharmacokinetic/pharmacodynamic parametersC max t1/2(h)AUC(mg.h/L)AUC/MIC0–24h∆T/MIC(h)Strain1514IPM-I Strain953IPM-S Strain1514IPM-IStrain953IPM-STigecycline(serum)5mg/Kg 1.92mg/L 2.339.249.249.24 1.70 1.70 Tigecycline(lung)5mg/Kg8.29μg/g0.23 4.37 4.37 4.370.970.97 Imipenem(serum)30mg/Kg16.9mg/L0.158.25 4.12330.51 1.04C max:maximal drug concentration;t1/2:half-life;AUC:area under the concentration–time curve;AUC/MIC0–24h:area under the concentration–time curve at24h to the MIC;∆T/MIC:the time that a drug concentration remains above the MICTable2Survival study using three strains with different susceptibilities to imipenemStrain1514(IPM-I)Strain953(IPM-S)n n died%n n died% Controls151387151067 Imipenem,120mg/Kg/d/im1500a14321b Tigecycline,10mg/Kg/d/sc131185131077 IPM:imipenem,I:intermediate,S:susceptiblea p<0.001compared to the controls and tigecycline.b p=0.025and p=0.007compared to the controls and tigecycline,respectivelydepends on the time above the MIC and on the AUC/MIC.On the other hand,the relationship between the AUC,MIC,and the microbiologic results in clinical trials in complicat-ed skin and soft tissue infections were revised recently [21,22],showing that an AUC/MIC ratio between 6.96and 12.3was necessary for a good clinical and microbiological response,with the predominant pathogens being Staphylo-coccus aureus and Streptococcus spp.However,although in our experiments the serum AUC/MIC of tigecycline was 9.24,we did not obtain the same favorable results in the pneumonia by A.baumannii .The clinical information on the relevance of tigecycline as an antimicrobial agent against A.baumannii is scarce.A clinical case of septic shock due to a multidrug-resistant A.baumannii strain in a patient with intra-abdominal abscess after acute pancreatitis,with failure during treatment with colistin plus meropenem,cured after the addition of tigecycline [23].In the treatment of multi-resistant A.baumannii V AP in a recent study [8],three patients were cured with tigecycline in monotherapy and 15out of 19patients (78.9%)cured when treated with tigecycline plus imipenem and/or colistin.In other study,only two out of five patients with A.baumannii V AP were cured with tigecycline in monotherapy or in combination [24].Peleg et al.[25]show the first clinical description of bloodstream infection caused by tigecycline-non-susceptible A.baumannii .In their work,two patients developed breakthrough A.baumannii bacteraemia while receiving tigecycline for other indications.The MICs oftigecycline for these strains were 4and 16mg/L,respectively,and the resistance appears to be at least partly attributable to an efflux pump mechanism,since the exposure to an efflux pump inhibitor diminished the MIC of tigecycline 4to 1mg/L and from 16to 4mg/L,respectively.The authors conclude that,given the facility of A.baumannii to acquire resistance to other antimicrobials,exposure to sub-therapeutic levels of tigecycline for even short periods of time may promote the rapid emergence of tigecycline resistance.In this sense,a recent multicenter Spanish study obtained an MIC 90of tigecycline for A.baumannii of 8mg/L [26],which is higher than that found previously by us [7].The results of the present experimental study and those from the clinical experience pointed to the necessity of evaluating tigecycline combined with other antimicrobials when treating severe A.baumannii infections.Thus,in a model of A.baumannii experimental murine pneumonia caused by imipenem-susceptible strains [9],the treatment with other tetracyclines,such as doxycycline plus amikacin,was as efficacious as imipenem in reducing the mortality and the clearance of bacteria from the lungs,this combina-tion being synergic in vitro.However,in an in vitro study using time –kill curves [27],the possible synergy of tigecycline plus amikacin,meropenem,imipenem,ciprofloxacin,levofloxacin,ampicillin-sulbactam,and rifampin has been evaluated against A.baumannii strains intermediate or resistant to carbapenems (MIC 90of tigecycline of 2mg/L),showing indifference for tigecycline in combination with these antimicrobials.Also,concentration escalation studies dem-onstrate that tigecycline may need to approach serum concentrations higher than those currently achieved to treat multidrug-resistant A.baumannii .In the same way,using checkerboard testing,tigecycline synergy was observed in only 1of 5strains when combined with cefepime or amikacin,and no synergy was shown with the combination of tigecycline with amoxicillin/cavulanate,piperacillin/tazobactam,ceftriaxone,ceftazidime,imipenem,merope-nem,aztreonam,trimethoprim/sulfamethoxazole,or cipro-floxacin [28].In summary,the present study,taking into account the limitations of the small size of the groups in the efficacy experiments,shows that tigecycline in monotherapy is less efficacious than imipenem in the treatment of experimental A.baumannii pneumonia caused by susceptible or inter-mediate to imipenem strains.However,due to its in vitro activity against multi-resistant A.baumannii ,the high mortality of severe infections caused by this bacterium,such as V AP and bacteremia,and the paucity of therapeutic alternatives for these severe infections,new in vivo studies are needed to evaluate its efficacy in combination with otherantimicrobials.Fig.1Bacterial clearance from lung tissue using two strains with different susceptibilities to imipenem (treatment during a period of 72h).Values are expressed as mean ±standard deviation (SD)(log CFU/g).*p <0.05,#p =0.054Acknowledgments This work was partly supported by a grant fromthe Wyeth-Ayerst Research(Pearl River,NY,USA).The authorsdeclare no conflict of interest with respect to this article. References1.Cisneros JM,Reyes MJ,Pachón J,Becerril B,Caballero FJ,García-Garmendía JL,Ortiz C,Cobacho AR(1996)Bacteremiadue to Acinetobacter baumannii:epidemiology,clinical findings,and prognostic features.Clin Infect Dis22:1026–10322.Garnacho-Montero J,Ortiz-Leyba C,Jiménez-Jiménez FJ,Barrero-Almodóvar AE,García-Garmendia JL,Bernabeu-WittellM,Gallego-Lara SL,Madrazo-Osuna J(2003)Treatment ofmultidrug-resistant Acinetobacter baumannii ventilator-associatedpneumonia(V AP)with intravenous colistin:a comparison withimipenem-susceptible V AP.Clin Infect Dis36:1111–11183.Fernández-Cuenca F,Pascual A,Ribera A,Vila J,Bou G,Cisneros JM,Rodríguez-Baño J,Pachón J,Martínez-Martínez L;Grupo de Estudio de Infección Hospitalaria(2004)Diversidadclonal y sensibilidad a los antimicrobianos de Acinetobacter baumannii aislados en hospitales españoles.Estudio multicéntrico nacional:proyecto GEIH-Ab2000.Enferm Infecc Microbiol Clin22(5):267–2714.Vila J,Pachón J(2008)Therapeutic options for Acinetobacterbaumannii infections.Expert Opin Pharmacother9(4):587–599 5.Bouchillon SK,Hoban DJ,Johnson BM,Johnson JL,Hsiung A,Dowzicky MJ;Tigecycline Evaluation and Surveillance Trial(TEST)Group(2005)In vitro activity of tigecycline against3989gram-negative and gram-positive clinical isolates from the UnitedStates Tigecycline Evaluation and Surveillance Trial(TESTProgram;2004).Diagn Microbiol Infect Dis52(3):173–1796.Fritsche TR,Sader HS,Stilwell MG,Dowzicky MJ,Jones RN(2005)Potency and spectrum of tigecycline tested against aninternational collection of bacterial pathogens associated with skinand soft tissue infections(2000–2004).Diagn Microbiol InfectDis52(3):195–2017.Pachón-Ibáñez ME,Jiménez-Mejías ME,Pichardo C,Llanos AC,Pachón J(2004)Activity of tigecycline(GAR-936)against Acinetobacter baumannii strains,including those resistant to imipenem.Antimicrob Agents Chemother48:4479–44818.Schafer JJ,Goff DA,Stevenson KB,Mangino JE(2007)Earlyexperience with tigecycline for ventilator-associated pneumoniaand bacteremia caused by multidrug-resistant Acinetobacter baumannii.Pharmacotherapy27(7):980–9879.Rodríguez-Hernández MJ,Pachón J,Pichardo C,Cuberos L,Ibáñez-Martínez J,García-Curiel A,Caballero FJ,Moreno I,Jiménez-Mejías ME(2000)Imipenem,doxycycline and ami-kacin in monotherapy and in combination in Acinetobacter baumannii experimental pneumonia.J Antimicrob Chemother 45:493–50110.Wolff M,Joly-Guillou ML,Farinotti R,Carbon C(1999)In vivoefficacies of combinations ofβ-lactams,β-lactamase inhibitors,and rifampin against Acinetobacter baumannii in a mousepneumonia model.Antimicrob Agents Chemother43:1406–141111.Bergogne-Bérézin E,Towner KJ(1996)Acinetobacter spp.asnosocomial pathogens:microbiological,clinical,and epidemio-logical features.Clin Microbiol Rev9:148–16512.Bou G,CerveróG,Domínguez MA,Quereda C,Martínez-BeltránJ(2000)Characterization of a nosocomial outbreak caused by amultiresistant Acinetobacter baumannii strain with a carbapenem-hydrolyzing enzyme:high-level carbapenem resistance in A.baumannii is not due solely to the presence ofβ-lactamases.J Clin Microbiol38:3299–330513.Henwood CJ,Gatward T,Warner M,James D,Stockdale MW,Spence RP,Towner KJ,Livermore DM,Woodford N(2002) Antibiotic resistance among clinical isolates of Acinetobacter in the UK,and in vitro evaluation of tigecycline(GAR-936).J Antimicrob Chemother49:479–48714.Urban C,Mariano N,Rahal JJ,Tay E,Ponio C,Koprivnjak T,Weiss J(2000)Polymyxin B-resistant Acinetobacter baumannii clinical isolate susceptible to recombinant BPI21and cecropin P1.Antimicrob Agents Chemother45:994–99515.Meagher AK,Ambrose PG,Grasela TH,Ellis-Grosse EJ(2005)Pharmacokinetic/pharmacodynamic profile for tigecycline—a new glycylcycline antimicrobial agent.Diagn Microbiol Infect Dis52(3):165–17116.Muralidharan G,Micalizzi M,Speth J,Raible D,Troy S(2005)Pharmacokinetics of tigecycline after single and multiple doses in healthy subjects.Antimicrob Agents Chemother49(1):220–229 17.Rubinstein E,Vaughan D(2005)Tigecycline:a novel glycylcy-cline.Drugs65(10):1317–133618.Rodvold KA,Gotfried MH,Cwik M,Korth-Bradley JM,DukartG,Ellis-Grosse EJ(2006)Serum,tissue and body fluid concen-trations of tigecycline after a single100mg dose.J Antimicrob Chemother58(6):1221–122919.Craig WA(1998)Pharmacokinetic/pharmacodynamic parameters:rationale for antibacterial dosing of mice and men.Clin Infect Dis 26:1–1220.van Ogtrop ML,Andes D,Stamstad TJ,Conklin B,Weiss WJ,Craig WA,Vesga O(2000)In vivo pharmacodynamic activities of two glycylcyclines(GAR-936and WAY152,288)against various gram-positive and gram-negative bacteria.Antimicrob Agents Chemother44(4):943–94921.Agwuh KN,MacGowan A(2006)Pharmacokinetics and phar-macodynamics of the tetracyclines including glycylcyclines.J Antimicrob Chemother58(2):256–26522.Doan TL,Fung HB,Mehta D,Riska PF(2006)Tigecycline:aglycylcycline antimicrobial agent.Clin Ther28(8):1079–1106,Review 23.Taccone FS,Rodriguez-Villalobos H,De Backer D,De Moor V,Deviere J,Vincent JL,Jacobs F(2006)Successful treatment of septic shock due to pan-resistant Acinetobacter baumannii using combined antimicrobial therapy including tigecycline.Eur J Clin Microbiol Infect Dis25:257–26024.Anthony KB,Fishman NO,Linkin DR,Gasink LB,Edelstein PH,Lautenbach E(2008)Clinical and microbiological outcomes of serious infections with multidrug-resistant gram-negative organ-isms treated with tigecycline.Clin Infect Dis46:567–57025.Peleg AY,Potoski BA,Rea R,Adams J,Sethi J,Capitano B,Husain S,Kwak EJ,Bhat SV,Paterson DL(2007)Acinetobacter baumannii bloodstream infection while receiving tigecycline:a cautionary report.J Antimicrob Chemother59(1):128–13126.Betriu C,Rodriguez-Avial I,Gómez M,Culebras E,López F,Alvarez J,Picazo JJ;Spanish Tigecycline Group(2006)Antimi-crobial activity of tigecycline against clinical isolates from Spanish medical centers.Second multicenter study.Diagn Micro-biol Infect Dis56(4):437–44427.Scheetz MH,Qi C,Warren JR,Postelnick MJ,Zembower T,Obias A,Noskin GA(2007)In vitro activities of various antimicrobials alone and in combination with tigecycline against carbapenem-intermediate or-resistant Acinetobacter baumannii.Antimicrob Agents Chemother51(5):1621–162628.V ouillamoz J,Moreillon P,Giddey M,Entenza JM(2008)In vitroactivities of tigecycline combined with other antimicrobials against multiresistant gram-positive and gram-negative pathogens.J Antimicrob Chemother61:371–374。
青霉素的使用和发现英语作文
青霉素的使用和发现英语作文{z}Title: The Use and Discovery of PenicillinPenicillin, one of the most significant discoveries in the history of medicine, has saved countless lives since its creation.It is a type of antibiotic that is widely used to treat various bacterial infections.This essay will discuss the discovery and the uses of penicillin.The discovery of penicillin can be attributed to Scottish scientist Alexander Fleming in 1928.While studying influenza, Fleming noticed that a mold called Penicillium notatum had grown on one of his cultures.He found that the mold prevented the growth of staphylococcus bacteria, which was causing the infection.Fleming realized that the mold was producing a substance that killed the bacteria, and he named this substance penicillin.Since its discovery, penicillin has been extensively studied and developed, leading to the creation of various types of penicillin antibiotics.These antibiotics are classified based on their chemical structure and spectrum of activity.The most common types of penicillin include penicillin G, penicillin V, and amoxicillin.Penicillin is used to treat a wide range of bacterial infections, including skin infections, urinary tract infections, respiratory tract infections, and gastrointestinal infections.It is also used to treat syphilis and gonorrhea, which are sexually transmitted infections.Theeffectiveness of penicillin depends on the type of bacteria causing the infection and its susceptibility to the antibiotic.When using penicillin, it is essential to follow the doctor"s instructions and complete the full course of medication, even if symptoms improve before the end of the course.This is because incomplete treatment can lead to the development of antibiotic resistance, making the infection harder to treat in the future.Additionally, some individuals may experience side effects when taking penicillin, such as nausea, vomiting, and skin rash.In rare cases, penicillin can cause a severe allergic reaction, known as anaphylaxis, which can be life-threatening.In conclusion, penicillin is a vital antibiotic that has revolutionized the treatment of bacterial infections.Its discovery by Alexander Fleming in 1928 has saved countless lives, and its use has significantly reduced the mortality rate from these infections.However, with the development of antibiotic resistance, it is crucial to use penicillin responsibly and complete the full course of medication as prescribed by a doctor.。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
5668Rev Esp Quimioter 2013;26(1):56-63Uso de tigeciclina en pacientes críticos con infección intraabdominal nosocomial graveRESUMENLa infección intraabdominal (IIA) es una patología habitual en la unidad de cuidados intensivos quirúrgica (UCIQ) y se asocia a una considerable mortalidad. Tigeciclina es el primer antibiótico de la familia de las glicilciclinas que presenta un amplio espectro de actividad frente a las bacterias habituales responsables de la IIA. Este estudio observacional retrospectivo tiene como objetivo describir la experiencia con tigeciclina en los pacientes con IIA nosocomial grave ingresados en la UCIQ. Los datos fueron recogidos en 23 pacientes consecutivos admitidos en la UCIQ con IIA nosocomial grave que habían recibido tratamiento antibiótico empírico con tigeciclina. En todos los casos, la IIA fue diagnosticada mediante cirugía urgente. En el 56,5% de los pacientes se encontró sepsis grave y el 43,5% presentaron shock séptico. La enfermedad concomitante más fre-cuente fue la enfermedad oncológica (60%). El SAPS III (Simplified Acute Physiology Score ) a las 24 h del diagnóstico de la IIA fue 57,5 ± 14,7 y un 87% de los pacientes presentaron un McCabe > 1 (2 o 3). El patógeno más frecuente fue Escherichia coli (43,5%), seguido de Bacteroides spp y Streptococcus spp (30,4%, respectivamente). Todos los pacientes excepto uno de ellos, recibieron tigeciclina en combinación con otros antimicrobianos (95,7%), con más frecuen-cia fluconazol (52,2%), seguido de piperacilina-tazobactam (43,5%). El tratamiento antibiótico empírico fue considerado adecuado en el 95% de los pacientes. La duración media del tratamiento antibiótico fue 8,5 ± 4,5 días. Se consiguió una respuesta favorable en el 78% de los pacientes. No se observó fracaso del tratamiento antibiótico en ningún paciente. Tampoco en ningún caso hubo que suspender el tratamiento con tigeciclina debido a la presencia de reacciones adversas. La mortalidad en la UCIQ fue del 13%, y ninguna muerte fue atribuible a tigeciclina. E stos hallazgos sugieren que la uti-lización de tigeciclina en combinación con otros antimicrobianos es un tratamiento eficaz y bien tolerado en los pacientes con IIA noso-comial grave ingresados en la UCIQ.Palabras clave: infección intraabdominal, pacientes críticos, UCIQ, tratamiento empírico, tigeciclina.ABSTRACTIntra-abdominal infection (IAI) is a frequent complica-tion found in surgical intensive care unit (SICU) and continues to be associated with considerable mortality. Tigecycline, the first-in-class glycylcycline has demonstrated a broad spec-trum of activity against a wide range of bacteria commonly found in IAI. This observational retrospective study aimed to describe the experience with tigecycline for serious nosocomial IAI in the SICU. Data were collected from 23 consecutive pa-tients admitted to SICU with serious nococomial IAI who had received empirical treatment with tigecycline. In all cases, IAI was diagnosed via emergency surgery. Severe sepsis was found in 56.5% and 43.5% developed septic shock. Oncological dis-ease was the most common comorbidity (60%). The mean Sim-plified Acute Physiology Score (SAPS) III within 24 hours from IAI diagnosis was 57.5±14.7, and 87% showed a McCabe score >1 (2 or 3). Escherichia coli was the most common pathogen (43.5%), followed by Bacteroides spp. and Streptococcus spp. (30.4%, respectively). All but one patient received tigecycline in combination (95.7%), particularly with fluconazole (52.2%), followed by piperacillin-tazobactam (43.5%). E mpirical an-tibiotic therapy was considered adequate in 95%. The mean duration of treatment was 8.5±4.5 days. A favorable response was achieved in 78%. Failure of the antibiotic therapy was not observed in any patient. None of the patients discontinued ti-gecycline due to adverse reactions. SICU mortality was 13%, with no deaths attributable to tigecycline. These findings sug-gest that tigecycline combination therapy is an effective and well tolerated empirical treatment of serious nosocomial IAI in the SICU.Keywords: intra-abdominal infections, critical patients, SICU, empirical treatment, tygeciclineUse of tigecycline in critically ill patients with serious nosocomial intra-abdominal infectionsServicio de Anestesia y Reanimación, Hospital Universitario La Paz, Madrid, SpainEmilio MasedaSantiago Ernesto Denis Ana Riquelme Fernando GilsanzCorrespondence:Dr. Emilio MasedaServicio de Anestesiología y ReanimaciónHospital Universitario La Paz, Pº de la Castellana, 261 28046. MadridTel: +34 629018689 - Fax: +34 914561105E-mail: emilio.maseda@OriginalUse of tigecycline in critically ill patients with serious nosocomial intra-abdominal infectionsE. Maseda, et al.6957Rev Esp Quimioter 2013;26(1):56-63The present study describes the experience with tigecy-cline in patients with serious nosocomial IAIs in the surgical ICU (SICU) of a University hospital and their clinical outcome in routine clinical practice.PATIENTS AND METHODSStudy population and design. All consecutive patients admitted for serious nosocomial IAI to the SICU of a University hospital were retrospectively analyzed. The study population included patients of both sexes and 18 years or older who had received at least one dose of tigecycline for treatment of IAI.This was a non-interventional retrospective registry lim-ited to collecting the information recorded in the data base of patients with IAI, which reflects the experience with tigecy-cline in the SICU in routine clinical practice. The data collected for each patient included: demographic data (age, gender), an-thropometric data (weight, height, BMI), co-morbidity, data on IAI, microbiological data, treatment related data (tigecycline therapy, previous and concomitant antibiotic treatment, ad-equacy of antibiotic treatment in the first hours of infection), and data on patient outcome (clinical efficacy, death).Assessment of severity of underlying disease at 24 hours from diagnosis of infection was performed using the follow-ing severity scores: Simplified Acute Physiology Score III (SAPS III)21, Sequential Organ Failure Assessment (SOFA)22, McCabe score 23 and serum lactate levels.Identification of microorganisms was performed according to the standard microbiological procedures of our institution.Definitionsa) Treatment adequacy.Empirical antibiotic treatment in the first 6 hours in the SICU was considered adequate or inadequate in terms of cov-erage of the antimicrobial agents used against the microor-ganisms isolated, and based on the clinical improvement of the patient.b) Clinical outcome of patients.The clinical outcome of patients was analyzed by assess-ing whether they showed a favorable clinical response, suf-fered a treatment failure, experienced a new suture failure, or if they died.Clinical response was determined based on the investiga-tor’s judgment as a favorable clinical response or treatment failure according to usual clinical practice criteria. A favorable clinical response was considered when the clinical outcome of the patient was assessed as cure or improvement based on resolution or improvement of signs and symptoms; so that the patient did not require new antibiotic treatment, or there were no serious adverse effects requiring discontinuation of tigecy-cline-based therapy. Treatment failure was defined as persis-tence or worsening of clinical signs and symptoms of IAI or ap-pearance of new signs or symptoms associated with infection, requirement of additional antibiotic treatment, or replacement of tigecycline for an alternative antibiotic treatment.INTRODUCTIONIntra-abdominal infection (IAI) is one to the most fre-quent complications treated in the surgical area. At present, a high proportion of patients with IAI require admission to an intensive care unit (ICU) and mortality continues to be above 20%1. Increased bacterial resistance 2, inadequate empirical treatment 3, and poor control of the infectious focus 4, may be, among others, the factors responsible for failure of its man-agement.In recent years we have witnessed a significant increase in the incidence of multiresistant microorganisms both in the community and hospital setting 5, particularly in ICUs 6,7. This scenario has created an urgent need to develop new antibi-otics with a broad antimicrobial spectrum that overcome the usual mechanisms of resistance. Moreover, given the polymi-crobial aerobic and anaerobic etiology of IAIs, optimizing the spectrum of antibiotic therapy is crucial to the approach to their treatment.Tigecycline is the first agent in a new class of antibiotics belonging to the glycylcycline group, structurally similar to the tetracyclines whose structure confers a broader antimicrobial spectrum and a decreased susceptibility to the development of resistance than tetracycline antibiotics. Thus, this antimicro-bial agent is not affected by the main mechanisms of bacte-rial resistance of the tetracyclines, based on the activity of ef-flux pumps that reduce that the intracellular concentration of the antibiotic and ribosomal protection 8. The broad spectrum of action of tigecycline covers gram-positive, gram-negative, anaerobic and atypical pathogens, including microorganisms resistant to multiple antimicrobial agents 9,10. In this regard, ti-gecycline has been reported to have microbiological efficacy against a large number of multiresistant gram-positive patho-gens, including methicillin-resistant Staphylococcus aureus (MRSA), penicillin-resistant Streptococcus pneumoniae (PRSP) and vancomycin-resistant Enterococcus spp.11. In addition, it has been reported to possess a broad spectrum of action against multiresistant gram-negative bacteria such as extend-ed-spectrum beta-lactamase (SBL)-producing Escherichia coli and Klebsiella pneumoniae or carbapenamase-producing Acinetobacter spp.12,13. However, resistance to tigecycline has been shown in Pseudomonas aeruginosa and decreased sensi-tivity in Proteus spp.14.The broad spectrum of action against multidrug resistant bacteria, its wide tissue distribution 15, and the lack of adjust-ments for hepatic or renal impairment, make tigecycline an especially useful antibiotic agent for early treatment of IAIs in the ICU setting 13,16,17. However, available experience on the use of tigecycline in critical patients is scarce and the studies con-ducted have evaluated its use in different hospital settings and in different infections and microorganisms against which it is applied 16,18-20. There are limited observational studies available describing the use of tigecycline in the ICU, and furthermore they consist of heterogeneous reports in terms of the indica-tion for treatment with tigecycline 16,20.Use of tigecycline in critically ill patients with serious nosocomial intra-abdominal infectionsE. Maseda, et al.5870Rev Esp Quimioter 2013;26(1):56-63Statistical analysis of the data was performed using the Statistical Package for the Social Sciences (SPSS) version 17.0 statistical software (SPSS, Chicago, IL, USA).RESULTSClinical and demographic characteristics of patients. A total of 23 patients with serious nosocomial IAI who hadStatistical analysis. To describe the clinical outcome of pa-tients admitted for severe IAI and treated with tigecycline, a de-scriptive analysis was carried out on all qualitative and quantitative variables. Qualitative variables were analyzed using absolute and relative frequencies and quantitative variables using the main mea-sures of central tendency and dispersion (mean, median, standard deviation 5, minimum, maximum, first quartile and third quartile). The results were expressed as mean ± SD and median (range).Table 1 D emographic and clinical characteristics of patients and IAI-related data at SICU admissionASA: American Society Anesthesiology; BMI: Body Mass Index; COPD: Chronic Obstructive Pulmonary Disease; CVV-HDF: Continous Veno-Venous Hemodiafiltration; IAI: Intra-abdominal Infection; LAR: Low Anterior Resection; SAPS III: Simplified Acute Physiology Score; SB: Small Bowel; SICU: Surgical Intensive Care Unit; III; SOFA: Sequential Organ Failure Assessment. aPatients may suffer more than one comorbid condition (multiple response variable). Comorbidities present in at least 5% of patients are presented. bMost frequent surgical procedures used in at least 10% of patients are presented. Data have been calculated over 20 patients. cData have been calculated over 20 patients. dTwo (8.7%) patients showed SAPS III score between 1% and 10%, 10 (43.5%) patients between 10% and 20% and the 3 patients remaining (13.0%) between 30% and 40%.Demographics and clinical characteristics No. patients (%)Demographic and anthropometric data Gender: men, n (%)16 (69.6%)Age (yr, mean ± SD)63.6±16,6BMI (Kg/m2, mean ± SD)24.9±5,1Comorbidity a Cancer 14 (60.9)Neutropenia 4 (17.4)Diabetes mellitus 4 (17.4)Corticotherapy 3 (13.0)Cardiovascular disease3 (13.0)Health care setting within last year 3 (13.0) Chronic kidney disease 2 (8.7)COPD 2 (8.7)IAI related data No. patients (%)IAI non preceded by surgery 4 (17.4)Nosocomial infection 2 (8.7)Community-acquired infection 2 (8.7)Postoperatory IAI19 (82.6)Type of surgical intervention Programmed 11 (55.0)Emergency9 (45.0)Type of surgical procedure b Hemicolectomy 3 (15.0)Esophagectomy2 (10.0)IAI related data No. patients (%)Subtotal gastrectomy 2 (10.0)LAR2 (10.0)SB resection + Anastomosis 2 (10.0)Sigmoidectomy2 (10.0)ASA classification for patients who underwent surgery c Healthy2 (10.0)Mild-moderate systemic disease10 (50.0)Severe systemic disease not associated to disability 8 (40.0)Diagnosis of IAI Septic shock 13 (56.5)Emergency surgery 23 (100.0)Location Colon11 (47.8)Stomach-duodenum 4 (17.4)Small bowel2 (8.7)Complicated cholecystitis-cholangitis 1 (4.3)Pancreas1 (4.3)Severity score within 24h from diagnosis of IAI (mean ± SD) SAPS III scored57.5 ± 14,7Predicted mortality according to SAPS III (%)30SOFA score 7.0 ± 2,2Serum lactate 1.8 ± 1.3McCabe score >1; n (%)20 (87.0)Measures taken for management of infectious process Mechanic ventilation ≥24h 8 (34.8)CVV-HDF6 (27.3)Use of tigecycline in critically ill patients with serious nosocomial intra-abdominal infectionsE. Maseda, et al.7159Rev Esp Quimioter 2013;26(1):56-63Microbiology. The most frequent microorganisms in IAIs,identified with a frequency higher than 15%, were mainly gram-negative pathogens. E. coli was the microorganism most frequently identified in cultures, present in 10 (43.5%) patients. Anaerobes and gram-positive cocci ranked second in frequency among microbiological cultures of the patients. Bacteroides spp. and Streptococcus spp. were detected in the microbiological cultures of 7 (30.4%) patients, respectively. Enterococcus faecalis was identified in 5 (21.7%) patients and Bacteroides fragilis in 4 (17.4%). The remaining microorgan-isms were identified with a frequency less than 15%. Only 2 patients had P. aeruginosa in their infectious diagnosis as part of their polymicrobial flora (tables 2 and 4).Prior antibiotic therapy and tigecycline treatment a)Prior antibiotic therapy.Only 2 (8.7%) patients had received more than two cy-cles of antibiotic therapy within the year previous to diagno-sis of IAI, whereas 17 (73.9%) patients had received antibiotic therapy within the 2 previous months, and 3 (13.0%) had been treated with fluconazole (table 3).b) Tigecycline therapy and concomitant antibiotic treatment.Tigecycline was used as monotherapy in only one patient, whereas in the remaining 22 (95.7%) patients tigecycline was administered in combination with other antibiotic agents. To-gether with tigecycline, the most frequently selected treat-ment for management of IAI was the antifungal agent flucon-azole, received by 12 (52.2%) patients, followed by the antibi-otic combination piperacillin-tazobactam, given to 10 (43.5%) patients. Caspofungin and anidulafungin were administered in 7 (30.4%) patients, respectively, and 4 (17.4%) received ce-fepime (tables 3 and 4).The mean duration of antibiotic therapy with tigecycline was 8.5 ± 4.5 days. With regard to the drugs used in combina-tion with tigecycline, mean duration was 6.6 ± 4.1 and 5.9 ± 2.4 days for fluconazole and piperacillin-tazobactam, respec-tively (table 3).Among the 21 patients in whom adequacy of treatment was known, empirical antibiotic therapy administered in the first 6 hours from diagnosis of infection was adequate in 20 (95.2%) patients. Treatment was considered inadequate in only 1 patient (table 3).Clinical outcome. Five (21.7%) patients died prior to dis-charge from the hospital. In no case was death attributable to treatment of IAI with tigecycline. Three (13.0%) patients died during their stay in the SICU, whereas the remaining 20 (87.0%) patients continued in the hospital ward after dis-charge from the SICU. The median total duration of hospital-ization was 43 days (range 37-84 days) (table 4).E ighteen (78.3%) patients achieved a favorable response with tigecycline therapy. Failure of the antibiotic therapy for IAI was not observed in any patient. None of the patients needed to discontinue tigecycline treatment due to adverse reactions. Three (13.0%) patients suffered a new suture failure (table 4).received empirical treatment with tigecycline were retrospec-tively analyzed.The main clinical and demographical characteristics of the patients at admission and during their stay in the SICU are shown in table 1. Sixteen (69.6%) patients were men and the mean age was 63.6 ± 16.6 years. Cancer was the most frequent underlying co-morbidity recorded in the medical history, af-fecting 14 (60.9%) patients. Other underlying diseases among patients with IAI were neutropenia and diabetes mellitus in 4 (17.4%) patients respectively.Thirteen (56.5%) patients presented severe sepsis and 10 (43.5%) developed septic shock. In 19 (82.6%) patients, IAI was postoperative in origin. Among patients who developed an IAI after a surgical operation, the most frequent procedure was hemicolectomy, carried out in 4 (20.0%) patients. In all cases, infection was diagnosed via emergency surgery.The mean SAPS III score in the first 24 hours since IAI di-agnosis was 57.5±14.7. Twenty one (87.0%) patients showed a McCabe score of more than 1 (2 or 3).Eight (34.8%) patients required mechanical ventilation for at least 48 hours during their stay in the SICU, while 6 (27.3%) required continuous veno-venous hemodiafiltration (CVV-HDF).Table 2 M icrobiology of IAIMost frequent pathogens identified in microbiologic cultures in more than 5% of patients are presented in the table.aPseudomonas aeruginosa is found as a part of the polymicrobial flora of infection focus.Microorganisms No. patients (%)Gram negatives Escherichia coli 10 (43.5)Bacteroides spp.7 (30.4)Bacteroides fragilis 4 (17.4)Klebsiella spp.3 (13.0)Pseudomonas aeruginosa a 3 (13.0)Prevotella spp. 3 (13.0)Gram positives Streptococcus spp.7 (30.4)Enterococcus faecalis 5 (21.7)Enterococcus faecium 3 (13.0)Clostridium spp. 2 (8.7)Staphylococcus aureus 2 (8.7)Candida albicans 3 (13.0)Candida tropicalis2 (8.7)6072Rev Esp Quimioter 2013;26(1):56-63Use of tigecycline in critically ill patients with serious nosocomial intra-abdominal infectionsE. Maseda, et ed 16,18-20. To our knowledge, this is the first homogeneous se-ries of critically ill patients describing experience with tigecy-cline in the SICU setting in which the specific indication of this antibiotic for the treatment of complicated IAI was assessed. The study population comprised patients with severe sep-sis and septic shock (57%), who underwent emergency ab-dominal surgery. Most patients had some relevant comorbid-ity, and 60% were oncological patients. The severely ill patient population analyzed in the present study is underrepresented or even excluded from pivotal clinical trials of tigecycline, since the number of patients with severe underlying disease, such as immunosuppressed patient, those with an acute Physi-ology and Chronic Health Evaluation II (APACHE II) score> 15 (4%), or with multiple intraabdominal abscesses (10%), was very limited in these trials 24.The pivotal trials on which approval of the use of tige-cycline in IAIs was based are part of a pooled analysis of the randomized trials that were the basis for the recent United States Food and Drug Administration (FDA) alert describing the increased mortality risk associated with the use of tigecycline compared to other drugs in the treatment of a variety of se-rious infections, including complicated IAIs 25. In this scenario, there are discrepancies between the evidence of the clinical trials, which generally exclude severely ill patients treated with tigecycline, and the situation the clinician must face in daily management of critically ill patients as a result of the high rate of multiresistant pathogens and the few available treatment options evaluated for this type of patients, especially in sep-sis 26. In this regard, clinical studies are needed to evaluate the efficacy of tigecycline in critically ill patients.Morbidity and mortality for severe IAI is high, particularly in patients with more severe infections and greater comorbid-ity 1. The mortality rate (21.7%) for the population of critically ill patients was lower than the predicted mortality according to SAPS III severity score shown within the 24h from diagnosis of IAI based on their baseline comorbidity (30%). Even though they were critically ill patients with relevant associated comor-bidities, the mortality rate was relatively low (21.7%). Further-more, the mortality seen in the current series was even lower than that reported in a German study in patients with severe sepsis and septic shock in the SICU setting (30%)20.The favorable clinical response achieved with tigecycline therapy in our study was consistent regardless of the etiology of the infections. Among the most common pathogens treat-ed were E. coli and B. fragilis , two enterobacteria strains that typically colonize the gastrointestinal tract. After the Entero-bacteriaceae family, the next most common infection was that caused by enterococci, with a greater presence of E. faecalis and to a lesser extent E. faecium . Accordingly, piperacillin-tazobactam in combination with tigecycline was the predomi-nant regimen used in order to provide adequate enterococcal coverage due to the worse prognosis of patients infected with Enterococcus spp.27.An incidence of Candida spp. infection of around 6% has been reported In the ICU setting, with a higher mortality rateDISCUSSIONThe findings of the present investigation demonstrated that tigecycline combination therapy is an effective and well tolerated empirical treatment for critical patients with serious nosocomial IAIs. Although these were critically ill patients with severe sepsis and septic shock, 78% achieved a favorable re-sponse with tigecycline therapy. None of the patients showed failure of the antibiotic therapy for IAI. The mortality rate in this series was 21.7%, and there were no deaths attributable to tigecycline therapy.Available experience on the use of tigecycline in critically ill patients is scant and the studies conducted to date dif-fer with regard to its use (empirical or rescue therapy), types of infection treated and microorganisms against which it isTable 3 Tigecycline treatment and previousand concomitant therapyaThree patients received antifungal treatment with fluconazole within the 2 previous months before the start of treatment with tigecycline.bMultiple response variable. cDuration of antibiotic treatments administered to more than 40% of pa-tients are presented.dAdequacy of empiric treatment administered within the first 6h of IAI. Adequacy of treatment was unknown in 2 patients.Antibiotic treatmentNo. patients (%)Antbiotic treatment prior to tigecycline >2 cycles of antibiotics within the year prior to tigecycline2 (8.7)Antibiotic therapy within the 2 previous months a 17 (73.9)Tigecycline treatment Combined therapy22 (95.7)Concomitant treatment with tigecycline b Fluconazole 12 (52.2)Piperacillin-tazobactam 10 (43.5)Caspofungin 7 (30.4)Anidulafungin 7 (30.4)Cefepime 4 (17.4)Voriconazole 2 (17.4)Amikacin1 (8.7)Treatment duration c (days, mean ± SD)Tigecycline 8.5±4,5Fluconazol6.6±4,1Piperaciline-Tazobactam5.9±2,4Adequate treatment within the first 6 h d Adequate 20 (95.2)Inadequate1 (4.8)7361Rev Esp Quimioter 2013;26(1):56-63T a b l e 4 M a i n c h a r a c t e r i s t i c s o f p a t i e n t s , a n t i b i o t i c t r e a t m e n t a n d c l i n i c a l o u t c o m eN o .A g e (y r )C o m o r b i d i t y aS A P S I I I bM i c r o b i o l o g yT i g e c y c l i n e t r e a t m e n t d u r a t i o n (d a y s )C o m b i n a t i o n t r e a t m e n t s C l i n i c a l o u t c o m e169C a n c e r , D M52B a c t e r o i d e s s p p . E . f a e c i u m , o t h e r 9p i p e r a c i l l i n e -t a z o b a c t a m a n i d u l a f u n g i n , f l u c o n a z o l e F a v o r a b l e267N R38P . a e r u g i n o s a , B . f r a g i l i s , P r e v o t e l l a , E . f a e c a l i s , S t r e p t o c o c c u s s p p .4c a s p o f u n g i n , f l u c o n a z o l e F a v o r a b l e327N R49N A c6p i p e r a c i l l i n e -t a z o b a c t a m , f l u c o n a z o l eF a v o r a b l e475C a n c e r71E . c o l i A M C R , E -c o l i T Z P R , E . c o l i C I P R10c a s p o f u n g i n F a v o r a b l e557C a n c e r46E .c o l i , B .f r a g i l i s , C l o s t r i d i u m , C . t r o p i c a l i s8f l u c o n a z o l e F a v o r a b l e682C a n c e r , C O P D , n e u t r o p e n i a , D M , C V D60E . c o l i B L E E , E . c o l i A M C R , E . c o l i T Z P R , E . c o l i C I P R ,B a c t e r o i d e s s p p .,C l o s t r i d i u m , P r e v o t e l l a , E . f a e c a l i s , o t h e r23c a s p o f u n g i n F a v o r a b l e746C a n c e r , n e u t r o p e n i a50B a c t e r o i d e s s p p .7c e f e p i m e , c a s p o f u n g i nF a v o r a b l e866N R25E . c o l i , B .f r a g i l i s , B a c t e r o i d e s s p p ., C l o s t r i d i u m , V e i o n e l l a , S t r e p t o c o c c u s s p p .8p i p e r a c i l l i n e -t a z o b a c t a m , v o r i c o n a z o l eF a v o r a b l e ; n e w s u t u r e f a i l u r e982C a n c e r48E . c o l i B L E E8n o n eF a v o r a b l e1084C a n c e r70E . f a e c a l i s7p i p e r a c i l l i n e -t a z o b a c t a m , c a s p o f u n g i n , f l u c o n a z o l eÉx i t u s1170C a n c e r , n e u t r o p e n i a , c o r t i c o t h e r a p y83E n t e r o b a c t e r B L E E , C . t r o p i c a l i s8p i p e r a c i l l i n e -t a z o b a c t a m , a n i d u l a f u n g i nÉx i t u s1240N R70N A c9p i p e r a c i l l i n e -t a z o b a c t a m , v o r i c o n a z o l eF a v o r a b l e1373C a n c e r50E . f a e c a l i s , o t h e r8p i p e r a c i l l i n e -t a z o b a c t a m , c a s p o f u n g i n ,F a v o r a b l e1481C a n c e r , C K D72S t r e p t o c o c c u s s p p .8f l u c o n a z o l eN e w s u t u r e f a i l u r e ; d e a t h d1564C a n c e r50E . c o l i , S t r e p t o c o c c u s s p p .9f l u c o n a z o l eF a v o r a b l e ; n e w s u t u r e f a i l u r e1683C V D58K l e i b s e l l a s p p .8a n i d u l a f u n g i nD e a t h1751C a n c e r , n e u t r o p e n i a , c o r t i c o t h e r a p y80A c i n e t o b a c t e r s p p ., P . a e r u g i n o s a , E . f a e c i u m , C a n d i d a s p p .11c e f e p i m e , c a s p o f u n g i n a , f l u c o n a z o l eD e a t h1864N R58E . c o l i , K l e i b s e l l a s p p ., P . a e r u g i n o s a , B a c t e r o i d e s s p p ., E . f a e c a l i s , C . a l b i c a n s19p i p e r a c i l l i n e -t a z o b a c t a m , a n i d u l a f u n g i n , f l u c o n a z o l eF a v o r a b l e1926C o r t i c o t h e r a p y50E . c o l i , P r o t e u s s p p ., S t r e p t o c o c c u s s p p ., S . a u r e u s6a n i d u l a f u n g i n , f l u c o n a z o l e , a m i k a c i n eF a v o r a b l e2071C a n c e r74E . f a e c i u m8c e f e p i m e , a n id u l a f u n g i nF a v o r a b l e2159C O P D , D M , C V D , s u r g e r y w i t h i n l a s t y r76E . c o l i , P r e v o t e l l a , S . a u r e u s4c e f e p i m e , a n id u l a f u n g i nF a v o r a b l e2258D M , C K D46K l e i b s e l l a s p p ., S t r e p t o c o c c u s s p p .6p i p e r a c i l l i n e -t a z o b a c t a m , f l u c o n a z o l eF a v o r a b l e2368C a n c e r46E . c o l i , B . f r a g i l i s , B a c t e r o i d e s s p p ., P e p t o s t r e p t o c o c c u s s p p ., S t r e p t o c o c c u s s p p .2p i p e r a c i l l i n e -t a z o b a c t a mF a v o r a b l eA M C R : a m o x i c l a v -r e s i s t a n t (a m o x i c i l l i n + c l a v u l a n i c a c i d ), C I P R : c i p r o f l o x a c i n -r e s i s t a n t ; C K D : C h r o n i c K i d n e y D i s e a s e , C O P D : C h r o n i c O b s t r u c t i v e P u l m o n a r y D i s e a s e , C V D : C a r d i o v a s c u l a r D i s e a s e , D M : D i a b e t e s M e l l i t u s , N A : n o n a v a i l a b l e , N R : n o n e r e p o r t e d . S A P S I I I : S i m p l i f i e d A c u t e P h y s i o l o g y S c o r e I I I ; T Z P R : p i p e r a c i l l i n e -t a z o b a c t a m -r e s i s t a n t .a C o m o r b i d c o n d i t i o n s r e c o r d e d i n t h e m e d i c a l h i s t o r y a r e d e s c r i b e d b S A P S I I I s c o r e w i t h i n t h e 24h o f i n f e c t i o n c A s a m p l e f o r m i c r o b i o l o g i c c u l t u r e w a s n o t t a k e n f r o m t h i s p a t i e n t .d F o r t h i s p a t i e n t , d e a t h w a s a t t r i b u t a b l e t o a s e c o n d s u t u r i n g p r o c e d u r e .。