麻醉领域的个体化用药,药物基因组学(Evan Kharasch)

合集下载

药物基因组学与阿片类镇痛药物需求个体化的研究进展

药物基因组学与阿片类镇痛药物需求个体化的研究进展

药物基因组学与阿片类镇痛药物需求个体化的研究进展药物基因组学是研究药物在个体基因组水平上的表达、变异和相互作用的学科。

随着基因组学研究的不断发展,药物基因组学逐渐成为个体化医学的重要领域之一、阿片类镇痛药物是常用的镇痛药,然而它们的疗效和副作用在不同个体间存在差异。

因此,深入研究药物基因组学与阿片类镇痛药物需求个体化的关系对个体镇痛治疗有重要意义。

研究发现,阿片类镇痛药物的作用主要是通过与μ-阿片受体相互作用来发挥的。

μ-阿片受体是阿片药物的作用靶点,它在不同个体中的表达水平和功能状态可能存在差异,导致个体对阿片类药物的反应不同。

此外,鉴于阿片类药物通过肝脏和肠道被代谢和排泄,药物代谢酶和转运体的多态性也可能对药物的药效和代谢产生影响。

通过药物基因组学研究,可以鉴定与阿片类镇痛药物需求个体化相关的基因多态性。

例如,一些基因多态性会影响阿片类药物在体内的代谢速度,进而影响药物的疗效和副作用。

临床研究表明,CYP2D6基因的多态性可以导致阿片类药物的代谢速度发生改变,从而影响药物的疗效和副作用。

此外,OPRM1基因的变异也会影响个体对阿片类药物的反应,进而影响镇痛效果。

此外,基因多态性还可能与肠道和肝脏中的转运体有关,如P玩家碱泵和乙酰化酶等。

基于药物基因组学研究的结果,可以为个体化阿片类药物治疗提供指导。

根据个体基因型,可以合理选择阿片类药物的种类、剂量和给药间隔,从而提高药物的疗效和减少不良反应。

例如,在一些患者中,由于基因变异导致药物代谢缓慢,应该减少药物剂量或延长给药间隔,以避免药物积聚导致过量和不良反应。

此外,在一些患者中,由于基因变异导致阿片受体的敏感性改变,可以酌情增加药物剂量以获得更好的镇痛效果。

总之,药物基因组学与阿片类镇痛药物需求个体化的研究为个体化阿片类药物治疗提供了重要依据。

通过鉴定与药物代谢和作用相关的基因多态性,可以合理选择药物种类、剂量和给药间隔,优化镇痛效果并减少不良反应。

基因导向的个体化给药在医院临床药学中的应用

基因导向的个体化给药在医院临床药学中的应用

血液
3个工 作日
双胍类 (二 甲双胍 苯乙
1-Oct
预测疗效
基因突变,导致转运功能降低,该类药物清除率变 慢,降糖效应减弱;在突变患者中,建议加大用药 剂量或换药
减少药物不良反应
及安全性的关系
基因水平
个体化用药
11
BaiO
个体化用药
身高/体重






•老年人 •儿童 •新生儿
药物反应个体差异
并发症

12
环境因素
•饮食 •吸烟 •合并用药
脏器功能
•肝功能 •肾功能 • 心功能

BaiO
基因导向的个体化给药
A lifelong, individually tailored health care approach to the detection, prevention and treatment of disease based on knowledge of an individual's precise genetic profile.
用药指导:发现携带突变等位基因的患者给与相对低的 华法林剂量。
月平均基因检测量:20例左右
检测项目举例
3. CYP 2C19检测与氯吡格雷 作用:CYP2C19是氯吡格雷重要的体内代谢酶。 突变型:携带CYP2C19*2,*3基因型患者氯吡格雷抗血 小板聚集的效果出现下降,并呈现基因-剂量效应。 用药指导:发现携带突变等位基因的患者增加氯吡格雷 的剂量,或选用其他不经CYP2C19 代谢的抗血小板药物 如替格瑞洛等。 月平均基因检测量:15例左右
基因导向的个体化给药 在医院临床药学中的应用
主要内容

药物基因组学与个体化给药

药物基因组学与个体化给药

CYP2C18 氟西汀,丙咪嗪, 吡罗昔康,利福平 CYP2C19 氟西汀,丙咪嗪,异烟肼,去甲替林,苯妥英,利福平, 华法林 CYP2D6 CYP2E1 UTG2 氟西汀,地尔硫卓, 丙咪嗪,美托洛尔,去甲替林,茶碱 氟西汀,异烟肼,茶碱,异搏定 布洛芬,萘普生
NAT2
异烟肼
重庆医科大学药学院 秧茂盛
LDG
疾病基因研究室/药物基因组研究中心/生命科学研究院
药物基因组学与个体化给药
重庆医科大学药学院
秧茂盛
LDG
疾病基因研究室/药物基因组研究中心/生命科学研究院
疗效好
药物
无效 不良反应
重庆医科大学药学院
秧茂盛
LDG
疾病基因研究室/药物基因组研究中心/生命科学研究院 年龄
遗传背景
性别
治疗效果
并发症
LDG
疾病基因研究室/药物基因组研究中心/生命科学研究院
疗效好
TPMT+
致死性的骨髓抑制
TPMT-
疗效差
六巯基嘌吟 TPMT:硫嘌呤甲基转移酶
重庆医科大学药学院 秧茂盛
LDG
疾病基因研究室/药物基因组研究中心/生命科学研究院
药物作用的多基因本质
疾病的病源基因 治疗作用 不良反应
宿主易感基因
药物代谢和转运基因
疾病基因研究室/药物基因组研究中心/生命
Alleles C>T C>A A>G C>G C>T C>T G >C G>C G>A C>T G >C 2 4A 4B 10A 10B 10C 17 188 188 188 188 188 1062 1072 1085 1062 1072 1085 1127 1127 1749 1749 1934 1934 1749 1749 1749 2938 4268 4268 4268 4268 4268 4268 2938 4268

药物基因组学与个体化用药

药物基因组学与个体化用药

药物基因组学与个体化用药王晓会12生A 124120035(云南师范大学生命科学学院,云南昆明650500)摘要:药物基因组学是人类开始功能基因组学研究后出现的一门新兴的交叉学科,它阐述了从基因水平研究基因序列的多态性与药物效应多样性之间的关系.药物基因组学应用于临床药学是一个必然的趋势。

将药物基因组学应用于临床药学是合理用药深入发展乃至实现个体化用药的必经之路. 对于深入解释药物治疗的个体差异、减少药物不良反应、提高药物疗效等有重大意义。

药物基因组学作为一门新兴的学科, 致力于研究药物代谢、药物转运和药物靶分子的基因多态性与药物作用, 包括疗效和毒副作用之间的关系。

其在药学研究中, 特别是药物作用机制、药物代谢、提高药物疗效及新药研发等方面发挥重要作用。

本文通过阅读并分析近年国内公开发表的有关药物基因组学的相关文章,根据有关文献, 综合分析、归纳总结了药物基因组学的定义、研究方法、发展和与个体用药的关系,同时阐述了实现个体化用药的基本条件、优点以及个体化用药现阶段的概况、面临的挑战等。

关键词:药物基因组学;个体化用药1 药物基因组学1.1 药物基因组学的概念药物基因组学是基因功能学与分子药理学的有机结合,是研究基因序列变异及其药物不同反应的科学,以药物效应及安全性为目标,运用已知的基因理论研究各种基因突变与药效及安全性的关系,药物基因组学强调个体化。

通过它可为患者或者特定人群寻找合适的药物及恰当的剂量,改善病人的治疗效果[1]。

药物基因组学的核心是药物反应(药酶)的遗传多态性,宗旨是实现用药个体化,以求得到最佳疗效和最少不良反应。

由此可见,药物基因组学研究方法有别于一般的基因组学,它并不是通过研究新的基因来寻找疾病的发病机理,是通过已知基因组学理论来探讨基因因素对药物效应的影响,以明确药物作用靶点,从而准确预测患者对临床治疗反应[2].1.2 药物基因组学研究药物遗传学研究发现人体对药物的反应性与基因多态性存在极大关联,参与编码药物代谢酶、转运体、受体等基因的多态性能明显影响药物不良反应发生的概率,并改变药物疗效,导致药物“低代谢”或“超速代谢”表型发生,并在群体中构成一定比例。

药物基因组学对临床个体化用药的指导作用

药物基因组学对临床个体化用药的指导作用
步阶段, 有待 于更深入 的研究 。随着药物基因组学研 究方法及新技术 的不 断完 善, 以及个 体化用药基因芯片的研发 , 不久 的将来 就可 以实 现治疗学上按基 因选药的个体化用药医疗模式。
【 关键词 】 药物基因组学 ; 个体化用药 遗传 多态性 ; 疗效 ; 毒副作用
早在 2 0世纪 5 0年代我们就知道遗传 因素对药物反 应
药 的 意义 。 21 氨 基糖 苷 类 药 物 与 耳 聋 .
的能力下 降 , 可导致血 药浓度过高, 易诱发严重 的不 良反应 如支气管 哮喘、 心血管疾病 , 甚至死 亡 , 此基因型病人 , 对 临
床用药应减少 药量 。I 型者属于强代谢 者 中较 弱的一部 M
分, 因基 因突变导致酶 活性 略微降低 , 此类病人用药也应适 当减少剂量 。E M是正常人 群的代谢表型 , 临床上使用常 故
阻断药和钙通道阻滞药 ,大 多数情况下医生制定治疗方案 主要根据病人的年龄 、 体重 、 高血压程度 、 有无并发症等 , 凭
代谢异常 , 一般情况下患者无症状 , 但在吃蚕豆或使用抗疟
药伯氨喹啉类及其他具有强氧化作用 的药物后就会 出现 急 性溶血反应 ; 再有就是异烟肼的乙酰化作用 , 因个体 乙酰化 速度不同 ,导致不 同个体使用 同等剂量异烟肼时出现疗效 差异 , 甚或 发生 毒副反应的现象。2 0世纪 9 0年代 , 药物基 因组学的出现使我们对不 同个体用药后的药物反应差异有 了更深入了解 ,对很多 以前难以解释的药物反应现象有 了
规治疗剂量有效。U M则是 由于 出现 C P D E Y 2 6的多基 因拷
氨基糖苷类抗生素 自 14 9 5年问世以来 , 因其杀菌作 用 强 、 菌谱较宽且价格低廉而在临床上广为应用 , 其致 耳 抗 但 聋 的毒 性 反 应 也 一 直 困 扰着 全 世 界 的 医生 。我 国 有 听力 残 疾 20 0 0万 人 ,其 中 6 %一 0 0 8 %为 氨基糖 苷类 药物 中毒所 致。 氨基糖 苷类抗生 素致聋 可分为两类 , 一类 因接受 了毒 性剂量而致聋 ; 另一类则与遗传因素相关 。 国内外学者均证 实 :线粒体基因第 1 5 5 5位点 A G的均值性点突变和氨基 —

药物治疗个体化与药物基因组学

药物治疗个体化与药物基因组学

药物治疗个体化与药物基因组学随着科技的不断进步,药物治疗也进入了个体化的时代。

传统上,临床医生会根据患者的病情、身体特征和临床经验来选择适合的药物治疗方案。

然而,由于每个人的基因组有所差异,对于同一种药物的反应也会存在差异。

因此,药物基因组学的出现为个体化药物治疗提供了新的方向。

药物基因组学是研究药物在个体层面上的作用机制以及基因与药物反应之间的关联的学科。

它通过对个体的遗传信息进行分析,来预测他们对特定药物的反应和药物的代谢情况。

这种个体化的药物治疗方法可以帮助医生更准确地选择药物、调整药物剂量,以提高治疗效果和减少不良反应的发生。

药物基因组学的实施包括两个主要方面:药物代谢相关基因的检测和个体化的药物治疗方案的制定。

在药物代谢相关基因的检测方面,通过对患者的基因组进行测序或者对特定的代谢酶基因进行检测,可以了解到患者对某些药物的代谢情况。

例如,CYP2D6基因的变异会导致对不同药物的代谢能力存在差异,从而影响到药物的疗效和毒副作用的发生。

在个体化的药物治疗方案制定方面,根据患者的基因信息和药物代谢能力,可以制定出更加符合患者个体情况的用药方案。

个体化的药物治疗在很多疾病的治疗中都具有重要意义。

例如,在抗癌药物治疗中,药物基因组学可以帮助医生选择哪些患者可以获益于某种特定的抗癌药物,从而减少对于无效药物治疗的浪费。

此外,对于具有突变基因的患者来说,可能会存在对某些药物过敏或者毒副作用的风险。

通过个体化药物治疗的方案制定,可以减少对无效或者有害药物的使用,提高治疗的安全性和有效性。

药物基因组学不仅可以在药物治疗方面发挥重要作用,还可以为新药物的研发提供指导。

通过对药物分子和靶标基因的相互作用进行研究,可以更准确地判断某种药物是否会对特定人群产生疗效。

这种个体化的研发模式可以避免无效药物的开发,提高药物研发的效率和成功率。

尽管药物基因组学在个体化药物治疗中具有很大的潜力,但还存在一些挑战需要克服。

胡欣--药物基因组学与个体化用药

胡欣--药物基因组学与个体化用药

•因基因多态性致严重毒性而撤市的药物
•由于严重毒性,近年来被FDA召回的药物达40余种! •制药企业损失:400亿美元!
撤市药物
适用症
毒性
原因
阿洛司琼 阿司咪唑 西立伐他汀 西沙必利 右芬氟拉明 罗非考昔 特非那定 地来洛尔 舍吲哚 特罗地林
PPT文档演模板
肠道综合症 变态反应 高脂血症 胃十二指肠返流 肥胖 疼痛 变态反应 高血压 精神分裂症 尿失禁
缺血性结肠炎
QT 延长
横纹肌溶解
QT 延长
肺动脉高压 心脏猝死
基因多态性
QT, 扭转型室速
肝毒性
QT, 扭转型室速
扭转型室性心胡动欣过--药速物基因组学与个体化用药
•目的与意• 义药物基因组学在个体化医学中的作用
预测药物的疗效 预测药物的剂量 规避药物的ADR 寻找药物的新靶点 促使“成药性差”的新药及时终止研发
胡欣--药物基因组学与个体化用药
•根据非小细胞肺癌患者EGFR基因型应用吉非替尼
售价:550元/片。每天口服药物费用550 元,每月费用16,500元。
基因检测EGFR无突变患者可节省1-6个月 的药费:16,500元至99,000元。
•基于药物基因组学的个体化用药降低非 小细胞肺癌患者治疗费用!
2003年人类基因组序列完整 版绘制完成 人类基因组计划 (HGP) 启动 了个体化医学/基因组医学
•基于药物基因组学的个体化 药物治疗成为个体化医学中 的先行领域
PPT文档演模板
胡欣--药物基因组学与个体化用药
药物基因组学的概念
药物基因组学 ( Pharmacogenomics, PGx) : −研究DNA如何影响药物反应
•无效

药物基因组学在个体化治疗中的应用

药物基因组学在个体化治疗中的应用

药物基因组学在个体化治疗中的应用随着科技的进步和科学的发展,个体化医疗成为了医疗领域的一个重要研究方向。

药物基因组学,作为个体化治疗的重要组成部分,通过研究药物与个体基因之间的相互作用,可以帮助医生更好地选择和调整药物疗法,提高治疗效果,减少不良反应。

本文将探讨药物基因组学在个体化治疗中的应用。

一、药物基因组学的基本原理药物基因组学是研究个体基因对药物作用的一门学科。

它通过研究个体基因的多样性,寻找与药物治疗反应相关的遗传变异,从而预测个体对特定药物的反应。

基因多态性是个体对药物反应差异的一个重要原因,通过检测个体基因的多态性,可以确定个体对药物的敏感性和耐受性,从而实现个体化治疗。

二、药物基因组学在药物选择中的应用个体对药物的反应受多个基因的影响,其中包括药物转运基因、药物受体基因和药物代谢基因等。

在选择药物治疗方案时,可以根据患者的基因型信息来预测药物的疗效和不良反应风险,从而选择最适合的治疗方案。

例如,某些基因型的患者对特定药物的反应会更好,而另一些基因型的患者则对同一药物更为敏感,这些信息可以帮助医生更好地选择药物并确定剂量。

三、药物基因组学在药物调整中的应用在个体化治疗中,药物调整也是非常重要的一步。

根据患者的基因型信息,医生可以合理调整药物剂量,避免不良反应的发生。

例如,某些基因型的患者对药物代谢较慢,需要减少药物剂量,而另一些基因型的患者对药物代谢较快,需要增加药物剂量才能达到疗效。

通过了解患者的基因信息,可以进行个体化的药物调整,提高治疗的效果。

四、药物基因组学在不良反应预测中的应用药物基因组学还可以用于预测患者不良反应的风险。

某些患者的基因型可能会导致对某些药物的不良反应更为敏感,通过检测患者的基因信息,可以预测患者对某些药物的不良反应风险,并采取相应的预防措施。

例如,在使用某些化疗药物时,某些基因型的患者可能会出现严重的骨髓抑制,通过预测风险可以避免不必要的副作用。

五、药物基因组学的应用前景个体化治疗已经在临床实践中取得了一些积极的成果,但目前仍存在一些挑战。

药物基因组学在个体化药物治疗中的应用与前景

药物基因组学在个体化药物治疗中的应用与前景

药物基因组学在个体化药物治疗中的应用与前景引言药物治疗是治疗疾病的重要手段之一,然而由于人体基因组的个体差异性,同一种药物在不同患者中的疗效和副作用可能存在差异。

药物基因组学通过研究药物与基因之间的相互作用,可以更好地实现个体化药物治疗,提高治疗效果,降低不良反应的发生。

本文将深入探讨药物基因组学在个体化药物治疗中的应用与前景。

一、药物基因组学的基本概念药物基因组学是研究药物与基因之间的相互作用的学科,其核心是研究个体基因组对药物代谢、药效和毒性的影响。

通过了解患者的基因型,可以预测患者对药物的代谢速度、药效和不良反应的风险,从而为个体化用药提供依据。

二、药物基因组学在个体化药物治疗中的应用1.个体化用药指导根据患者基因型的不同,可以对药物的剂量进行个性化调整,以达到更好的治疗效果。

例如,对于对乙肝病毒感染的患者,部分患者对拉米夫定代谢能力较差,容易出现不良反应,通过检测患者的基因型,可以确定合适的用药剂量,降低不良反应的风险。

2.不良反应预测一些药物在代谢过程中会形成毒性代谢产物,导致不良反应的发生。

通过研究患者基因型,可以预测患者对这些药物的代谢能力,从而提前预警可能出现的不良反应。

例如,对于使用华法林抗凝治疗的患者,部分患者对华法林的代谢速度较快,需要更高的剂量才能达到治疗效果,而过高的剂量会增加出血的风险,通过检测患者的基因型,可以避免这种风险的发生。

3.新药研发药物基因组学可以通过研究药物与基因之间的相互作用,加速新药的研发和上市。

通过了解患者的基因型,可以在早期筛选出对某种药物具有较好疗效的患者群体,提高临床试验的成功率,缩短新药上市的时间。

三、药物基因组学在个体化药物治疗中的前景1.应用范围扩大随着技术的不断发展,药物基因组学可以应用于越来越多的药物和疾病。

目前已经有一些药物基因组学指南被纳入临床指南中,成为患者用药的重要参考依据,随着研究的深入,药物基因组学将在越来越多的领域发挥作用。

个体化用药与基因技术--什么是个体化用药(下)

个体化用药与基因技术--什么是个体化用药(下)

血栓形成性疾病,包括心肌梗死、缺血性脑中风、静脉
血栓以及心瓣膜臵换术、房室纤颤后。然而,Warfarin 的有效剂量范围很窄,特定剂量下人群的反应差异至少 相差十倍,从而导致剂量不够达不到防血栓的效果,剂 量过大又易引起出血的危险
个体化用药的案例
华法林(warfarin)的化学结构为3-(a-苯基丙酮)-4-羟基香 豆素分子式为C19H15NaO4
对药物不同反应的科学,所以它是研究高效、特效药物
的重要途径,通过它为患者或者特定人群寻找合适的药 物,药物基因组学强调个体化,因人制宜
药物基因组学
药物相关基因的多态性以及患者的基因的变异是导致个体 药物反应差异的原因。药物基因组学从基因水平给出了基因
多态性与药物效应的相关性。这种关系能辅助临床人员在使
正 常
•多基因病 •体细胞遗传病 坏
药物基因组学 药物基因组学综合药理学和遗传学、研究基因结构多态 性与不同药物反应之间关系的交叉学科,即个体基因遗
传因素如何影响机体对药物反应,解释由于个体之间差
异所表现出药物的不同治疗效果,趋向于用药个性化。 用药个性化将产生最大的效果和安全性
药物基因组学 药物基因组学也可以说是基因功能学与分子药理学的有 机结合。正因为药物基因组学是研究基因序列变异及其
患者不同的药物基因组学信息是导致华法林剂量差异的重
要因素 基因突变使得华法林的治疗产生了两种显著的表型:增加 了低凝血酶原血的概率和产生华法林的耐药性。在血多情 况下,基因型是决定华法林达到最佳抗凝血效果的关键变
量。由遗传因素导致低凝血酶原血的患者最常见的原因是
由CYP2C9代谢活性降低和药物靶标VKORC低表达引起的
预测药物异常反应的个体性差异,正确掌握用药的个体化原

药物基因组学

药物基因组学
药物基因组学
张巍
概念及研究背景 研究内容与目的 研究方法和技术 案例
药物基因组学概述
药物基因组学的诞生
早在20世纪50年代,人们就发现不同的遗传背景会导致 药物反应的差异,如一些遗传性葡萄糖-6-磷酸脱氢酶缺 陷患者在接受抗疟药伯氨喹治疗后,引发严重的溶血。 1959年Vogel提出的“遗传药理学”就是药物基因组学的 一种雏形,它从单基因的角度研究遗传因素对药物代谢和 药物反应的影响,特别是遗传因素引起的异常药物反应。
• 研究目的 • 合理用药, 个性化治疗 • 新药的发现和开发 • 药物经济学意义
为什么药物疗效不一样?
药物受体基因多态性
大多数药物作用于药物靶蛋白,使其产生药理学效 应,如受体、酶或涉及信号转导、细胞周期调控或其他细 胞功能的蛋白质。许多编码药物靶蛋白的基因具有多态性。 药物靶蛋白的基因多态性对于药物作用可有明显的影响。 已发现25种以上药物靶蛋白的遗传变异能影响药物效应。
药物从进人体内到发挥作用直至被清除,是一个较为 复杂的过程。在这个过程中的任何一个环节出现问题都可 能引起药物效应的各种异常。药物作用的差异可以是药物 动力学或药效学差异。
个性化治疗
Michael Kauffman预言,未来疾病的划分将基于生物 作用机制和分子标记。利用药物基因组学的技术和方法 ,实现个性化、可预测、可预防的治疗策略,由此可发 展为临床药物基因组学(clinical pharmacog-enomics) 。
对于每一个药物来说,大约有10%~40%对人无效,对百 分之几或更多的人有副作用。如果制药公司利用药物基因组 学理论可以事先预见结果或筛选试验人群的话,其成功率就 会高得多。
药物基因组学应用案例
• 癫痫是一种临床常见的由多病因引起的慢性反复性发作的 神经系统常见疾病,具有发病率高、病程长、治愈率低等特 点,主要通过长期合理服用抗癫痫药物(AEDs)来控制疾病发 作。

药物基因组学在精神疾病治疗中的应用与挑战

药物基因组学在精神疾病治疗中的应用与挑战

药物基因组学在精神疾病治疗中的应用与挑战一、绪论精神疾病是一类影响认知、情感、行为等方面的疾病,给患者和家庭带来沉重的负担。

药物治疗是精神疾病的主要治疗手段之一,但由于患者对药物的反应各异,传统的经验治疗不一定适用于每位患者。

药物基因组学的发展为精神疾病的个体化治疗提供了新的思路和方法。

本文将探讨药物基因组学在精神疾病治疗中的应用与挑战。

二、药物基因组学的基本原理药物基因组学是研究药物与个体基因组之间相互作用的学科,旨在通过分析个体基因组的差异来预测患者对特定药物的反应。

在精神疾病治疗中,药物基因组学可以帮助医生选择最适合患者的药物和剂量,提高治疗效果,减少不良反应。

药物基因组学的基本原理包括:基因多态性、药物代谢途径、药物靶点等。

三、药物基因组学在精神疾病治疗中的应用1. 个体化用药药物基因组学可以帮助医生根据患者的遗传特征选择最有效的药物和最合适的剂量。

例如,对于抗抑郁药物来说,一些患者可能由于基因的差异而对某些药物敏感,而另一些患者则可能需要更高的剂量才能达到治疗效果。

通过个体化用药,可以提高治疗效果,减少试错,缩短治疗时间。

2. 预测药物不良反应一些药物在部分患者身上可能会引起不良反应,甚至危及生命。

药物基因组学可以帮助医生预测患者对特定药物的不良反应风险,从而及时调整治疗方案,避免发生不良事件。

3. 新药研发药物基因组学还可以为新药的研发提供重要参考。

通过分析药物与靶点基因之间的相互作用,可以更好地理解药物的药效、毒性等特性,加速新药的研发过程。

四、药物基因组学在精神疾病治疗中的挑战1. 数据获取与解读药物基因组学需要大量的基因组数据来支持个体化治疗的决策,然而基因测序等技术的高昂成本限制了数据的获取。

同时,基因数据的解读也需要专业知识和工具支持,对临床医生的要求较高。

2. 伦理道德问题药物基因组学涉及患者的个人基因信息和隐私,如何保护患者的隐私权成为一个重要课题。

同时,基因检测的结果可能会带来心理负担和社会歧视,对患者造成不良影响。

药物基因组学 案例

药物基因组学 案例

药物基因组学案例
【原创版】
目录
一、药物基因组学的概念与背景
二、药物基因组学的研究方法与技术
三、药物基因组学的应用案例
四、药物基因组学的前景与挑战
正文
一、药物基因组学的概念与背景
药物基因组学是一门研究药物在人体内过程差异的基因特性以及基因变异所致患者对药物的不同反应的学科。

它旨在通过研究遗传基因及基因变异对药物效应的影响,从而开发出新的药物和合理的用药方法。

药物基因组学是基因功能学与分子药理学的有机结合,对于提高药物疗效和减少不良反应具有重要意义。

二、药物基因组学的研究方法与技术
药物基因组学的研究方法包括基因芯片技术、基因组测序、转录组测序、蛋白质组测序等。

这些技术可以帮助研究者深入了解基因变异与药物反应之间的关系,为药物研发和个体化治疗提供重要依据。

三、药物基因组学的应用案例
药物基因组学在很多领域都有广泛应用,以下是一些具体案例:
1.肿瘤个体化治疗:通过药物基因组学研究,可以发现肿瘤患者对药物的敏感性和耐药性,从而实现个体化治疗,提高疗效。

2.心血管疾病治疗:药物基因组学可以帮助研究者了解患者对心血管药物的反应,降低药物治疗的心血管事件发生率。

3.药物基因组学在精神病治疗中的应用:通过研究患者基因变异与药物反应的关系,可以为精神病患者提供更合适的药物治疗方案。

四、药物基因组学的前景与挑战
药物基因组学在提高药物疗效、减少不良反应等方面具有巨大潜力。

然而,它也面临许多挑战,如数据分析方法、样本量、标准化等问题。

此外,随着研究的深入,药物基因组学还需解决伦理、隐私保护等诸多问题。

麻醉药物基因组学研究进展

麻醉药物基因组学研究进展

麻醉药物基因组学研究进展本文从网络收集而来,上传到平台为了帮到更多的人,如果您需要使用本文档,请点击下载按钮下载本文档(有偿下载),另外祝您生活愉快,工作顺利,万事如意!本文对药物基因组学的基本概念和常用麻醉药的药物基因组学研究进展进行综述。

药物基因组学是伴随人类基因组学研究的迅猛发展而开辟的药物遗传学研究的新领域,主要阐明药物代谢、药物转运和药物靶分子的基因多态性及药物作用包括疗效和毒副作用之间关系的学科。

基因多态性是药物基因组学的研究基础。

药物效应基因所编码的酶、受体、离子通道作为药物作用的靶,是药物基因组学研究的关键所在。

基因多态性可通过药物代谢动力学和药物效应动力学改变来影响麻醉药物的作用。

基因多态性对药代动力学的影响主要是通过相应编码的药物代谢酶及药物转运蛋白等的改变而影响药物的吸收、分布、转运、代谢和生物转化等方面。

与麻醉药物代谢有关的酶有很多,其中对细胞色素-P450家族与丁酰胆碱酯酶的研究较多。

基因多态性对药效动力学的影响主要是受体蛋白编码基因的多态性使个体对药物敏感性发生差异。

苯二氮卓类药与基因多态性:咪唑安定由CYP3A 代谢,不同个体对咪唑安定的清除率可有五倍的差异。

地西泮是由CYP2C19和CYP2D6代谢,基因的差异在临床上可表现为用药后镇静时间的延长。

吸入麻醉药与基因多态性:RYR1基因变异与MH 密切相关,现在已知至少有23种不同的RYR1基因多态性与MH有关。

氟烷性肝炎可能源于机体对在CYP2E1作用下产生的氟烷代谢产物的一种免疫反应。

神经肌肉阻滞药与基因多态性:丁酰胆碱酯酶是水解琥珀酰胆碱和美维库铵的酶,已发现该酶超过40种的基因多态性,其中最常见的是被称为非典型的(A)变异体,与用药后长时间窒息有关。

镇痛药物与基因多态性:μ-阿片受体是阿片类药的主要作用部位,常见的基因多态性是A118G和G2172T。

可待因和曲马多通过CYP2D6代谢。

此外,美沙酮的代谢还受CYP3A4的作用。

  1. 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. 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. 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)。

Pharmacogenetics in AnesthesiaEvan D. Kharasch, M.D., Ph.D. St. Louis, Missouri 302 Page 1Pharmacogenetics (or pharmacogenomics) aims to understand the inherited basis for variability in drug response. The promise of pharmacogenetics has been a change from “one drug and dose fits all” to individualized predictive medicine, or “the right drug at the right dose in the right patient”. Anesthesiology as a specialty played a key role in developing pharmacogenetics. Prolonged apnea after succinylcholine, thiopental-induced acute porphyria, and malignant hyperthermia were clinical problems of the 1960’s whose investigation helped craft the new science of pharmacogenetics. Today we perhaps take for granted the knowledge that they are genetically-based problems, due to variants in pseudocholinesterase, heme synthesis and the ryanodine receptor, respectively. This review will address basic principles of pharmacogenetics and their application to drugs used in anesthetic practice.The term pharmacogenetics was originally defined (1959) as “the role of genetics in drug response”. Since the science of pharmacokinetics (drug absorption, distribution, metabolism, excretion) evolved earlier than pharmacodynamics, early pharmacogenetic studies addressed mainly pharmaco-kinetics. Application (fusion) of the genomic revolution and associated technologies to pharmaco-genetics spawned pharmacogenomics. Pharmacogenetics has been used by some in a more narrow sense, to refer only to genetic factors which influence drug kinetics and dynamics (drug receptor actions), while pharmacogenomics has been used more broadly to refer to the application of genomic technologies (whole-genome or individual gene changes) to drug discovery, pharmacokinetics and pharmacodynamics, pharmacologic response, and therapeutic outcome. Nonetheless, many consider this distinction unimportant and use the two terms interchangeably, as will this review.BASIC CONCEPTSA polymorphism is a discontinuous variation in a population (a bimodal or trimodal distribution). It is different than simple continuous variability (i.e. a unimodal population distribution, even if quite wide). A genetic polymorphism is the presence of multiple discrete states (i.e. for a particular trait) within a population, which has an inherited difference. The complete human genome consists of approximately 3 billion base pairs, which encode approximately 30,000 genes. A single nucleotide polymorphism (SNP) is a variation in the DNA sequence which occurs at a specific base. Polymorphisms are relatively common, occurring by definition in ≥1% of the population, while mutations are less common, occurring in <1%. Only 3% of DNA consists of sequences which code for protein (exons). Other portions of the DNA include promoter regions (near the transcription initiation site), enhancer regions (which bind regulatory transcription factors), and introns (DNA sequences which do not code for protein). After exons and introns are transcribed, the intronic mRNA is excised and the exonic mRNA is spliced together to form the final mature mRNA, which then undergoes translation into protein. SNPs are frequent, occurring in approximately 1:100-1:1000 bases. SNPs and mutations may occur in the coding or noncoding regions of the DNA. Since most occur in the latter, they are usually synonymous (or silent, having no effect on proteins), although intronic changes and promoter variants can change protein expression. Non-synonymous SNPs result in a change in an amino acid. A conservative change results in a similar amino acid that does not alter protein function, while a non-conservative change yields an amino acid which alters protein structure or function. These latter SNPs may be clinically significant. SNPs are not the onlyevents which can cause RNA and protein changes; others are deletions, insertions, duplications, andsplice variants, however these are not inherited.Multiple SNPs can occur in the DNA which encodesa particular protein. A haplotype is a set of closely linked alleles or DNA polymorphisms which are inherited together. While SNPs are important, haplotypes are more clinically relevant.Polymorphisms can be classified at the DNA locus (which depicts the normal “wild-type” and the altered base pair; for example the mu opioid receptor gene polymorphism at base pair 118 which codes for changing an adenine nucleotide to aguanine is abbreviated as A118G, or 118 A>G); atpolymorphism changes the amino acid at position 40Asn40Asp); or at the allele level of whole gene or protein (for example, the second variant of the wild-type P450 (CYP) enzyme CYP2C9*1, is CYP2C9*3). Individuals may be homozygous or heterozygous for a particular allele (i.e. CYP2C9*1/*1 or CYP2C9*1/*3). Typically, function is altered more in homozygotes than in heterozygotes. APPLIED CONCEPTSPharmacogenetic variability may influence drug disposition, drug transport, receptor structure and function, cell signaling, and the myriad of downstream responses which ultimately produce a therapeutic effect or adverse reaction. The figure schematically depicts the role of genes in determining drug pharmacokinetics and pharmaco-dynamics. It is important to remember that causes of interindividual variability include genetics, as well as age, disease, environment (diet, smoking, ethanol), and interactions with drugs and herbals.1. PharmacokineticsThis section will focus on the polymorphic proteins responsible for human drug disposition, and the role of genetic variability in metabolism and response. Although transport proteins do not catalyze metabolism per se, they influence drug absorption and bioavailability, act in concert with enzymes of biotransformation, and influence effect site concentrations, and will be discussed in this section.1A. MetabolismMetabolism converts lipophilic (fat soluble) drugs to more polar (water soluble) molecules more amenable to excretion. Phase I reactions introduce or uncover a functional group that increases drug polarity and prepares it for a Phase II reaction. Phase II reactions link the drug or metabolite with a highly polar molecule which renders the conjugate water soluble and thus excreted. Although considered mainly as a route of drug inactivation, phase I and II reactions can convert an inactive prodrug to an active metabolite (i.e. oxidation of inactive codeine to the more active metabolite morphine); an active drug to an active metabolite (morphine conversion to morphine-6-glucuronide); an active drug to an inactive metabolite (morphine to normorphine); or an active drug to a toxic metabolite (meperidine to normeperidine, which can cause seizures).Phase I EnzymesCytochrome P450 (CYP): CYPs are a superfamily of drug metabolizing enzymes. The individual CYPs are classified according to their sequence evolution. CYPs that share >40% sequence homology are grouped in a family (designated by an Arabic number, i.e. CYP2), those with >55% homology are grouped in a subfamily (designated by a letter, i.e. CYP2A), and individual CYPs are identified by a third number (i.e. CYP2A6). Allelic variants are designated by an asterisk and number following the protein identifier (i.e. CYP2A6*1). The majority of drugs are metabolized in humans by CYPs 1, 2 and 3 (mainly CYPs 2C, 2D6 and 3A). Some, but not all P450s exhibit pharmacogenetic variability.CYP2B6: CYP2B6 is expressed mainly in liver; also in intestine, brain and kidney. Initially considered to be expressed in only some individuals (~20%), in very low levels, and thus not important, CYP2B6 is now known to be expressed in greater amounts, in all/most individuals, and metabolizes many drugs. It is the main enzyme responsible for methadone N-demethylation, propofol hydroxylation, and (R)- and (S)-ketamine N-demethylation. Many drugs thought previously to be metabolized mainly by CYP3A, may instead (or also) be metabolized by CYP2B6. CYP2B6 is one of the most polymorphic CYPs, with several mutations occurring with high frequency, particularly in African-Americans, one of which results in low CYP2B6 activity.CYP2C: The human CYP2C gene family consists of CYPs 2C8, 2C9, 2C18 and 2C19, which together account for ~one-fourth of total hepatic CYP and metabolizing 15-20% of all therapeutic drugs. The pharmacogenetically most important CYP2C enzymes are 2C9 and 2C19. Others (CYP2C8, CYP2C18) are expressed in low levels or have narrow substrate specificities, and are not polymorphically expressed.CYP2C9 is the major 2C isoform in human liver. The best known CYP2C9 substrates are (S)-warfarin, phenytoin, tolbutamide, diclofenac, and several NSAIDs. Several variant 2C9 proteins with substantially decreased activity (as low as one-fifth) have been identified. The frequency of these mutant alleles occurs more commonly in Caucasians (10-20%) than in Asians and African-Americans (<5%). Homozygotes (CYP2C9*3/*3) have very low enzyme activity (1/10 of normal) while heterozygotes (*1/*3, *2/*3 ) have moderately reduced activity (1/3-2/3 of normal) compared with wild-types (*1/*1), with correspondingly reduced clearances. Warfarin is an excellent example of pharmacogenetics and its application in modern therapeutics. Patients with CYP2C9 variants havehigher warfarin plasma concentrations, and are more susceptible to over-anticoagulation, serious or life-threatening bleeding complications, and longer hospitalizations. CYP2C9 genotyping, and correspondingly adjusted doses for CYP2C9 variants, results in more rapid attainment of proper anticoagulation, and fewer complications.CYP2C19 is a minor P450, but has been well-studied since it was one of the early CYPs for which a genetic polymorphism was identified. The prototype 2C19 reaction is S-mephenytoin hydroxylation; genetic variability was initially termed the mephenytoin polymorphism. Other substrates include barbiturates (R-mephobarbital, hexobarbital) and diazepam. Individuals are classified as extensive (EM) or poor (PM) metabolizers of S-mephenytoin, with a PM frequency of 2-5% in Caucasians, 2% in African-Americans, 18-23% in Japanese, and 15-17% in Chinese. PM is caused by an autosomal recessive mutation, and PMs do not express CYP2C19. The CYP2C19 polymorphism is important in the disposition and clinical effect of diazepam, particularly PM homozygotes. Diazepam clearance in PM is half that in EM, with dose requirements correspondingly less, and heterozygotes are intermediate. This is thought to account for lower diazepam dose requirements in Chinese.CYP2D6 metabolizes ~25% of all drugs, even though it accounts for only 2-5% of total human liver P450. It is also of historical significance, being the first identified CYP genetic polymorphism, termed the “debrisoquine/sparteine polymorphism”. The list of CYP2D6 substrates is notable for a group of opioids which undergo CYP2D6-mediated O-demethylation (codeine, dextromethorphan, dihydrocodeine, hydrocodone, oxycodone, tramadol), beta-blockers (alprenolol, bufurolol, labetolol, metoprolol, propranolol, timolol) as well as various antiarrhythmics, antipsychotics, tricyclic anti-depressants, serotonin reuptake inhibitors. and antiemetic 5-HT3 antagonists. The debrisoquine/sparteine phenotype is an autosomal recessive monogenic trait. Patients are extensive (EM, wildtype), poor (PM), or ultrarapid (UM) metabolizers. The overall PM frequency is 7-8% in Caucasians, 2% in African-Americans, and <1% in Asians. Over 20 mutant CYP2D6 alleles have been identified. Most code for inactive proteins, and homozygotes are PM. Some alleles, however, which are particularly frequent in Asians, code for a protein with reduced activity. UM are caused by gene duplication, with up to 13 copies. UMs (20% Ethiopians, 7% Spanish, 1% Scandinavians) generally require substantially higher drug doses. There is even a spectrum of CYP2D6 activity within the EMs, owing to the diversity of 2D6 alleles. Commercial assays are now available which allow for accurate CYP2D6 genotyping.The role of pharmacogenetics in interindividual variability in drug disposition and response is well illustrated by codeine, a prodrug requiring CYP2D6-mediated metabolic activation (O-demethylation) to the mu agonist morphine, and even greater bioactivation to the more potent agonist morphine-6-glucuronide. CYP2D6 deficient individuals (PMs) are poor metabolizers of codeine, have markedly diminished or absent morphine formation, and minimal if any analgesia. Conversely, individuals with CYP2D6 gene amplification and/or duplication (UM) have greater morphine formation from codeine than extensive metabolizers and poor metabolizers. Ethnic variations attributable to codeine biotransformation have also been observed. For example, Chinese produced less morphine from codeine, were less sensitive to the opioid effects of codeine, and might therefore experience reduced analgesia.CYP3A: The CYP3A family metabolizes >50% of all therapeutic drugs. In adults it is comprised of CYP3A4 (which exhibits wide variability in activity but no genetic polymorphism) and the polymorphically expressed CYP3A5. CYP3A4 is the most quantitatively abundant CYP in human liver, accounting for ~30% (but as much as 60%) of total CYP. It is also the predominant CYP in human intestine. It exhibits the broadest substrate specificity of all CYPs, including opioids, benzodiazepines, local anesthetics, calcium channel antagonists, and immunosuppressants.CYP3A5 is similar to CYP3A4, and metabolizes many but not all CYP3A4 substrates, often with diminished activity. Hepatic CYP3A5 is polymorphically expressed, and was once thought present in only 25% of human livers and at lower levels than CYP3A4. However, more recent studies suggest that hepatic CYP3A5 expression is more frequent, occurs at a higher level, and is more contributory to the metabolism of certain CYP3A substrates than previously appreciated. CYP3A5 may account for >50% of CYP3A in some livers. Individuals expressing CYP3A5 along with 3A4 may have greater metabolism of CYP3A drugs. CYP3A5 is also polymorphically expressed in intestine, although levels are less than CYP3A4. The clinical role of polymorphic CYP3A5 expression is under investigation.Non-P450 EnzymesCarboxylesterases have a wide tissue distribution, found in greatest amounts in the liver, and also in the gastrointestinal tract, brain, and possibly blood. They have a nonselective substrate specificity, and hydrolyze estersand amides. Two broad-substrate human liver microsomal carboxylesterases have been isolated (hCE-1 and -2). hCE-1 catalyzes the hydrolysis of cocaine to benzoylecgonine, the hydrolysis of meperidine to meperidinic acid (the major route of metabolism), and the hydrolysis of heroin (3,6-diacetylmorphine) to 6-monoacetylmorphine, the active metabolite. hCE-02 also hydrolyzes heroin to both 6-monoacetylmorphine and then to morphine. It also catalyzes the bioactivation of the anticancer drug irinotecan into its active metabolite. There are numerous polymorphisms in the hCE-02 gene, however none result in altered enzyme activity.One major human cholinesterase is plasma cholinesterase (also known as pseudocholinesterase, serum cholinesterase, butyrylcholine esterase, and nonspecific cholinesterase), found in plasma as well as liver and other tissues. Plasma cholinesterase was one of the early enzymes for which pharmacogenetic variation was elucidated, based on observed heritable variability in the response to succinylcholine. A single autosomal locus is responsible for all plasma cholinesterase variants. Plasma cholinesterase activity is evaluating by its inhibition by dibucaine. The dibucaine number is the percent inhibition; normal is 71-85, intermediate ~60, and atypical ~20. The activity of atypical cholinesterase is decreased by 70%, homozygotes are very sensitive to succinylcholine, and the frequency of this variant is about 2%. Another method for evaluating activity is inhibition by fluoride, which typically parallels dibucaine inhibition. A small number of patients (0.3%), however, express a fluoride-resistant enzyme whose activity is decreased by 60%, and who are moderately sensitive to succinylcholine. Patients with the silent variant (0.03%) have no enzyme activity. Succinylcholine hydrolysis is rapid (90% within one minute) and robust (70% reduction in enzyme activity only moderately prolongs neuromuscular blockade). Mivacurium is also hydrolyzed by plasma cholinesterase, at a rate 70-90% of that of succinylcholine. Patients with cholinesterase variants respond similarly to mivacurium and succinylcholine. Recovery of neuromuscular function can be prolonged up to 60-fold in atypical cholinesterase homozygotes. Plasma cholinesterase also metabolizes the three ester local anesthetics cocaine, procaine and chloroprocaine. Heroin undergoes extensive deacetylation in blood to 6-monoacetylmorphine, and then to morphine. The former reaction is catalyzed rapidly by plasma cholinesterase, and more slowly by erythrocyte AchE (see below), although the latter reaction accounts for most hydrolysis in vivo and only erythrocyte AchE can convert 6-monoacetylmorphine to morphine.A second major human cholinesterase is acetylcholinesterase (AChE). The major function of AChE is to hydrolyze acetylcholine at the neuromuscular junction thereby terminating synaptic transmission. However there are two other AChE forms, in erythrocytes and the brain. Like succinylcholine, esmolol and remifentanil, also undergo extensive metabolic inactivation in blood. However their metabolism is catalyzed exclusively by erythrocyte AchE, and plasma cholinesterase has no catalytic activity. Hence neither remifentanil nor esmolol elimination are altered by pseudocholinesterase deficiency.Phase II EnzymesGlucuronosyltransferase: The glucuronosyltransferases (UGT) catalyze addition of glucuronic acid to lipophilic compounds to form water soluble metabolites which are rapidly excreted. Glucuronides may be more or less pharmacologically active than their parent drugs. Human UGTs are broadly classified into the UGT1A (phenol/bilirubin) and UGT2 (steroid/bile) families. Humans express UGT1As which glucuronidate endogenous compounds and many drugs (phenols, amines, anthraquinones, flavones), and UGT2s which glucuronidate endogenous compounds and a few drugs. There are several polymorphisms in the UGT1A family, most notably in UGT1A1, which results in much lower enzyme activity and mild hyperbilirubinemia (Gilbert's syndrome). Glucuronidation is an important pathway for certain drugs used in anesthesia. Propofol glucuronidation by human UGT1A9 is the major route of systemic elimination. UGT2B7 glucuronidates many opioids such as morphine, codeine, naloxone, nalorphine, buprenorphine, oxymorphone, and hydromorphone. Morphine 6-glucuronidation is important since this metabolite is more potent than its parent drug, is thought to play a significant role in morphine analgesia, and the finding of UGT2B7 in human brain suggests that in situ formation of this metabolite may play a role in analgesia. The glucuronide of 1-hydroxymidazolam is pharmacologically active, is renally excreted, and is thought to underlie the prolonged effects of midazolam in patients with renal insufficiency when used for ICU sedation. The role of UGT polymorphisms in anesthetic metabolism and response are unknown.Glutathione-S-transferase: Glutathione-S-transferases (GST) catalyze the reaction of the tripeptide glutathione with a diverse array of drugs and toxins, and are primarily a defensive system for detoxification. There isconsiderable polymorphism in GST expression, however this enzyme is of little relevance to anesthesia (except in the metabolism of the sevoflurane degradation product ‘compound A’).N-acetyl-transferase: N-Acetylation by N-acetyl-transferases (NAT1 & NAT2) is a common Phase II reaction, and can result in drug inactivation (isonizaid), an active therapeutic metabolite (N-acetylprocainamide) or an active toxic metabolite (carcinogens). NAT is of great historical significance, since it was the first drug metabolizing enzyme for which a genetic polymorphism was discovered (fast- and slow isoniazid N-acetylation). Slow acetylators show a greater therapeutic response than fast acetylators to several drugs (i.e. isoniazid, hydralazine, dapsone), and the latter group may require a greater dose. In contrast, slow acetylators may be more susceptible to the side effects of drugs mediated by acetylated metabolites, such as isoniazid hepatotoxicity and lupus-like syndrome after hydralazine or procainamide. There are NAT1 polymorphisms, but NAT2 polymorphisms cause the slow-acetylator phenotype (50-60% in Caucasians, 40% in African-Americans, 20% in Chinese, 8% in Japanese.1B Transport ProteinsP-glycoprotein (P-gp) is a plasma membrane efflux pump that actively transports a structurally diverse array of compounds out of the interior of several cell types. It is also known as the multidrug resistance protein (MDR1) because it was first associated with cancer cell resistance. P-gp is expressed on the apical (luminal) surface of epithelial cells, including the brush border of intestinal cells, hepatocyte biliary canalicular cell membranes, and the luminal surface of renal proximal tubular cells. In the gut, it actively pumps drugs from the intracellular milieu against a concentration gradient back into the intestinal lumen, actively countering intestinal drug absorption, and thereby limiting oral drug absorption. P-gp plays a major role in the intestinal absorption, first-pass elimination, and bioavailability of many drugs. P-gp is also expressed on the luminal surface of brain capillary endothelial cells, where it is an intrinsic part of the blood brain barrier and limits CNS access of drugs. Thus P-gp can be a major determinant of drug pharmacodynamics. There is considerable interindividual variability in human P-gp expression, and numerous polymorphisms and haplotypes in humans have been identified.Of particular potential significance in anesthesia is that opioids may be substrates for P-gp, both in the intestine and the brain. Morphine, the best-characterized opioid, is extruded by P-gp in blood-brain barrier brain capillary endothelial cells in rodents, actively limiting morphine access to the CNS. Alfentanil is also thought to be a brain P-gp substrate in rodents. Although P-gp is a major determinant of analgesic opioid CNS access and analgesia in animals, its role in humans is presently unknown. The role of P-gp polymorphisms in anesthetic transport and response is also unknown.A second subfamily of transporters are the multidrug resistance-associated proteins, also known as the multispecific organic anion transporter. The first protein to be discovered in this family was MRP1, whose overexpression is responsible for the majority of non-P-gp mediated multidrug resistance. Because MRPs were only recently discovered, little is known about their significance in anesthetic transport and clinical effects.2. PharmacodynamicsPharmacodynamic polymorphisms are generally attributable to variants in cell surface receptors or intracellular signaling pathways Compared with genetic influences on drug disposition, less information is currently available about genetic influences on pharmacodynamics. There are however several notable examples. Malignant hyperthermia is due to polymorphism in the skeletal muscle calcium release channel (ryanodine receptor) gene.Of considerable interest in anesthesiology is the occurrence and potentialA clinical significance in opioid receptor polymorphisms. The best studied SNP is the A118G mu opioid receptor polymorphism. Nevertheless, it is a confusing picture. Some but not all studies show decreased A118G mu receptor affinity for ß-endorphin and morphine. Some have shown a 2- to 3 fold decrease in the potency of morphine and morphine-6-glucuronide, while others have shown no difference. Some have shown a 1.5 to 2-fold increase in postop morphine PCA requirements in A118G homozygotes (but not heterozygotes), while othes suggest no major difference. If there is a major difference among A118G carriers, it is small.Several nonsynonymous polymorphisms in adrenergic receptors have been identified, although their functional significance awaits full determination. Some ß-adrenergic receptor polymorphisms have been implicated in the response to asthma therapy, and speculated to be involved in the response to drugs used for hypertension and congestive heart failure.SummaryPharmacogenetics is a rapidly evolving science. Genetic polymorphisms have been identified which alter drug metabolism, bioavailability, clearance, receptor binding, clinical effect, and toxicity. The ultimate clinical utility and cost-effectiveness of pharmacogenetic testing remains to be determined.ReferencesA. General references on pharmacogenetics1.Dervieux T: Pharmacogenetic testing: proofs of principle and pharmacoeconomic implications. Mutat Res2005;573:180-942.Lesko LJ, Woodcock J: Translation of pharmacogenomics and pharmacogenetics: a regulatory perspective. NatRev Drug Discov 2004;3:763-93.Ma JD: Genetic polymorphisms of cytochrome P450 enzymes and the effect on interindividual,pharmacokinetic variability in extensive metabolizers. J Clin Pharmacology 2004;44:447-564.Wilkinson GR: Drug metabolism and variability among patients in drug response. N Engl J Med 2005; 352:2211-215.Need AC: Priorities and standards in pharmacogenetic research. Nat Genet 2005; 37: 671-816.Williams JA: So many studies, too few subjects: establishing functional relevance of genetic polymorphisms onpharmacokinetics. J Clin Pharmacol 2006; 46: 258-647.Leschziner GD: ABCB1 genotype and PGP expression, function and therapeutic drug response: a criticalreview and recommendations for future research. Pharmacogenomics J 2007; 7: 154-79B. Pharmacogenetics and anesthesia8.Belfer I: Candidate gene studies of human pain mechanisms: methods for optimizing choice of polymorphismsand sample size. Anesthesiology 2004; 100:1562-72.9.Bukaveckas BL: Pharmacogenetics as related to the practice of cardiothoracic and vascular anesthesia. JCardiothorac Vasc Anesth 2004;18:353-65.10.Freeman BD: Challenges of implementing pharmacogenetics in the critical care environment. Nat Rev DrugDiscov 2004; 3: 88-9311.Iohom G: Principles of pharmacogenetics--implications for the anaesthetist. Br J Anaesth 2004; 93:440-5012.Palmer SN: Pharmacogenetics of anesthetic and analgesic agents. Anesthesiology 2005;102:663-7113.Zaugg M: Genetic modulation of adrenergic activity in the heart and vasculature: implications for perioperativemedicine. Anesthesiology 2005; 102:429-4614.Ziegeler S: Influence of genotype on perioperative risk and outcome. Anesthesiology 2003;99:212-915.Samer CF: Individualizing analgesic prescription Part I: pharmacogenetics of opioid analgesics. PersonalizedMedicine 2006; 3: 239-6916.Lötsch J: Current evidence for a genetic modulation of the response to analgesics. Pain 2006; 121: 1-517.Kharasch ED: Influence of CYP3A5 genotype on the pharmacokinetics and pharmacodynamics of alfentaniland midazolam. Clin Pharmacol Ther. 2007 in pressC. WebsitesGlossary of Genetic Terms /10002096AMA overview /ama/pub/category/2306.htmlNCBI pharmacogenomic primer /about/primer/pharm.htmlHuman Genome Project /sci/techresources/Human_Genome/home.shtml Haplotype map (HapMap) project /NIH pharmacogenetics research network http://www.nigms。

相关文档
最新文档