阿片类药物被广泛用于中度至重度疼痛的治疗,然而其镇痛效果及不良反应存在广泛的个体差异。药物遗传学研究表明,基因多态性与上述个体差异有密切关系,研究较多的为CYP2D6、 μ阿片受体(OPRM1)和儿茶酚-
Opioids are widely used in the treatment of moderate to severe pain.However,there are wide inter-individual variabilities in analgesic efficacy and adverse reactions.Pharmacogenetics study shows that gene polymorphism was closely related to the above interindividual variabilities,and more research was focused on CYP2D6 (cytochrome P450 2D6),μ opioid receptor (OPRM1),and catechol-
开放科学(资源服务)标识码(OSID)
阿片类药物是常用的镇痛药,广泛用于治疗中至重度疼痛,其镇痛效果、有效剂量以及不良反应存在极大的个体差异[1]。造成个体差异的原因可能与疼痛感知差异、社会文化、环境、性别、年龄和遗传变异有关,其中,遗传变异占主导地位[2]。这些变异可能归因于与阿片类药物代谢动力学和药效学有关基因的单核苷酸多态性(single nucleotide polymorphism,SNP)[3]。多个基因SNP如 μ阿片受体(μ opioid receptor,OPRM1)(A118G)、儿茶酚-
CYP2D6是CYP450家族中的最重要的氧化代谢酶之一。可待因、曲马多等常用阿片类镇痛药都是作为前体药物通过CYP2D6脱甲基化成活性代谢物才具有药理作用,因此,CYP2D6酶活性对这些阿片类药物发挥临床疗效具有关键性作用。而CYP2D6基因多态性对酶的活性又有着至关重要的影响。迄今为止,已发现的CYP2D6基因变异超过130种[7],不同等位基因的组合用于确定不同的双倍型。将CYP2D6每个等位基因活性值范围设为0~1,0为无功能(例如CYP2D6*3~*6),0.25或0.5为功能下降(例如CYP2D6*9、*10、*17、*29和*41),1为正常功能(例如CYP2D6*1、*2和*35),若等位基因包含功能基因的多个拷贝,则该值乘以存在的拷贝数,因此,CYP2D6活性评分是每个等位基因的活性值总和,通常范围为0~3[8]。《指南》根据CYP2D6酶的活性评分将其分为4种表型:慢代谢型(poor metabolizer,PM)、中间代谢型(intermediate metabolizer,IM)、正常代谢型(normal metabolizer,NM)、超快代谢型(ultrarapid metabolizer,UM)。CYP2D6活性评分及患者的双倍型与表型转化关系见
1.1.1 CYP2D6基因多态性对可待因的影响 可待因是一种在临床中广泛使用的前药,具有镇痛、镇咳和止泻的作用。可待因通过CYP2D6去甲基化形成吗啡。与可待因比较,吗啡对 μ阿片受体的亲和力高200倍,镇痛效力高50倍[9]。CYP2D6代谢表型与可待因形成吗啡具有相关性[10]。对接受可待因的健康志愿者开展药动学研究,结果表明正常代谢型和中间代谢型的志愿者均可产生镇痛作用,而慢代谢型志愿者使用可待因后产生的镇痛作用与使用安慰药比较无显著差异[10]。与正常代谢型和中间代谢型比较,可待因慢代谢型患者的血清中吗啡的血药浓度-时间曲线下面积(AUC)、峰浓度(peak concentration,
1.1.2 CYP2D6基因多态性对曲马多的影响 曲马多是一种人工合成的阿片类药物,对术后疼痛有显著的镇痛作用。与可待因类似,曲马多也是经CYP2D6代谢的一种前药,CYP2D6将其转化为其活性代谢物(+)-
1.1.3 CYP2D6基因多态性对氢可酮的影响 氢可酮是一种半合成的阿片类药物,类似于吗啡和可待因。在CYP2D6正常代谢型中,约5%的氢可酮被CYP2D6
1.1.4 CYP2D6基因多态性对羟考酮的影响 羟考酮是一种半合成的阿片类药物,通常用于急性和慢性术后疼痛。在CYP2D6正常代谢型中,约11%的羟考酮被CYP2D6
1.1.5 CYP2D6基因多态性对美沙酮的影响 美沙酮是一种合成阿片类镇痛药,目前被用于对阿片类药物依赖的维持治疗[24]。《指南》指出,美沙酮由CYP2D6代谢为无活性代谢物的程度较小,因此,CYP2D6基因多态性对美沙酮的镇痛效果、有效剂量及不良反应影响较小。
2.1.1 可待因与曲马多 CYP2D6基因变异对可待因和曲马多至关重要,因为它们影响药物的镇痛效果和不良反应。《指南》中对可待因和曲马多的建议是,对于CYP2D6正常代谢型的患者,使用说明书推荐的可待因或曲马多的年龄特异性或体质量特异性的起始剂量。对于中间代谢型也建议按照说明书推荐的起始剂量,并密切监测此类患者是否出现不良反应,如有必要,应提供替代镇痛药。对于CYP2D6慢代谢型,因可能出现镇痛效果不佳的情况,应避免使用可待因和曲马多,而选择其他替代镇痛药。由于文献中没有足够的证据推荐慢代谢型使用更大剂量的可待因或曲马多,特别是考虑到慢代谢型和正常代谢型之间一些不良事件没有差异[10],因此,《指南》中未推荐加大药物的剂量。对于CYP2D6超快代谢型,《指南》建议不应使用可待因或曲马多,以避免出现重度中毒风险。根据治疗疼痛的类型、严重程度和长期性,非阿片类镇痛药和不受CYP2D6表型影响的其他阿片类药物是CYP2D6慢代谢型和超快代谢型患者的替代治疗。基于CYP2D6表型的可待因和曲马多治疗建议总结见
2.1.2 氢可酮 《指南》对氢可酮的建议是,对于CYP2D6中间代谢型和慢代谢型,推荐使用氢可酮说明书推荐的年龄特异性或体质量特异性给药。由于目前尚不清楚增加氢可酮的剂量是否会影响中间代谢型或慢代谢型的镇痛效果,因此,《指南》提出,若CYP2D6中间代谢型和慢代谢型患者对氢可酮无反应时,不能增加剂量,而应考虑使用替代镇痛药即非阿片类药物或不受CYP2D6表型影响的阿片类药物。对于CYP2D6超快代谢型,由于目前缺乏足够的证据,《指南》中未提供指导临床实践的建议。基于CYP2D6表型的氢可酮治疗建议总结见
2.1.3 羟考酮与美沙酮 对于羟考酮和美沙酮,目前证据不足,不同CYP2D6基因型与羟考酮和美沙酮个体差异之间的关系尚不明确,因此,《指南》没有提供根据CYP2D6基因型为羟考酮或美沙酮指导临床实践的建议。
由于目前没有足够的证据,因此,《指南》中尚无基于OPRM1或COMT基因型的阿片类药物给药的治疗建议。
临床研究发现,儿童在使用治疗剂量的可待因后,CYP2D6超快速代谢型患儿可出现严重呼吸抑制和死亡[32]。对于扁桃体切除术后使用处方可待因的儿童来说,与无任何正常功能等位基因的儿童比较,如果至少有一个正常功能CYP2D6等位基因,那么该患儿的药物不良反应发生率将大大升高[33]。美国食品药品管理局(FDA)于2013年评估了可待因的安全性特征,并发布了关于可待因和含可待因产品用于儿童扁桃体和(或)腺样体切除术后疼痛管理的风险的黑框警告[4]。目前,全球已有包括FDA和欧洲药品管理局在内的多个监管机构反对12岁以下儿童以及扁桃体和(或)腺样体切除术后的18岁以下青少年使用可待因和曲马多[10]。出于同样的原因,FDA最近建议12岁以下儿童和18岁以下青少年在耳、鼻、喉手术后不要使用曲马多,并警告肥胖或患有肺部疾病的儿童谨慎使用曲马多[34]。2018年《阿片类药物在慢性非癌性疼痛中的规范化应用》建议阿片类药物应仅在特殊情况下以及在儿童和青少年的疼痛治疗专门机构中考虑使用[35]。
对于哺乳期妇女,FDA已在说明书中增加警告,即服用可待因或曲马多时不建议哺乳[36,37]。已有证据表明母亲摄入可待因会导致婴儿过度嗜睡、哺乳困难或严重呼吸问题等[4]。可待因及其代谢产物(包括吗啡)可分泌至人乳汁中,浓度通常较低且呈剂量依赖性,但CYP2D6超快代谢型的哺乳期女性在标准剂量的可待因治疗期间吗啡可达到较高的血药浓度,这可能导致乳汁中吗啡水平升高,从而使母乳喂养的婴儿暴露于高浓度的吗啡中[38]。研究报道1例接受可待因的超快代谢型母亲的母乳喂养新生儿发生致死性阿片类药物中毒事件[39]。另一方面,美国妇产科学会为产后疼痛管理提供的临床建议指出哺乳期妇女未经治疗或治疗不充分的疼痛也会对产后母亲及其母乳喂养的婴儿产生不良后果[40]。因此,根据基因型使用适当的镇痛药物为哺乳期妇女进行疼痛治疗至关重要。
通过解读CPIC发布的《指南》,明确根据检测患者的基因型来指导阿片类药物个体化给药方案的制定,可以最大程度地发挥阿片类药物的镇痛效应,减少不良反应,实现阿片类药物在临床上的精准治疗。然而,迄今为止,阿片类药物遗传学尚未被临床广泛应用,只有美国少数机构真正采用CYP2D6基因分型来指导可待因或曲马多的使用。此外,临床上更常用的其他阿片类药物的基因多态性相关指南尚属空白。因此,利用药物遗传学指导阿片类药物治疗仍然是一个有前途的领域,也是探索个体化镇痛方式、提高镇痛满意度、减少镇痛不良反应的必要途径。基因多态性与阿片类药物镇痛的关系尚有待进一步研究。
[1] |
|
[2] |
|
[3] |
|
[4] |
|
[5] |
|
[6] |
|
[7] |
|
[8] |
|
[9] |
|
[10] |
|
[11] |
Codeine is widely used as an analgesic and antitussive drug. The analgesic effect of codeine is mediated by its metabolite morphine, which is formed by the polymorphically expressed enzyme CYP2D6; therefore poor metabolizers have no analgesia after administration of codeine. Like other opiates, codeine causes a delay of gastric emptying and spastic constipation. It is not yet known whether the effect on gastrointestinal motility is mediated by codeine or its metabolite morphine.
To test the hypothesis that the metabolite morphine is responsible for the effects of codeine on gastrointestinal motility, a randomized, double-blind, two-way crossover study was performed. The orocecal transit time was studied in five extensive and five poor metabolizers of sparteine with the sulfasalazine-sulfapyridine method, assuming that no effects are observed in poor metabolizers because negligible amounts of morphine are formed.
No differences of orocecal transit times were observed between extensive metabolizers and poor metabolizers after oral placebo administration. However, after oral codeine administration orocecal transit time was significantly prolonged in extensive metabolizer but not poor metabolizer subjects. All pharmacokinetic parameters of codeine showed no differences between extensive metabolizers and poor metabolizers. The pharmacokinetic parameters (mean +/- SD) of the metabolite morphine were significantly different between extensive metabolizer and poor metabolizer subjects (peak serum concentration, 13.9 +/- 10.5 versus 0.68 +/- 0.15 pmol/ml; area under the serum concentration-time curve, 27.8 +/- 16.0 versus 1.9 +/- 0.7 hr.pmol/ml; total amount of morphine excreted in urine, 0.160 +/- 0.036 versus 0.015 +/- 0.007 mumol).
Because the orocecal transit time prolongation after codeine administration was observed only in extensive metabolizers, the effect of codeine on gastrointestinal motility, like the analgesia, is mediated by its metabolite morphine.
DOI:10.1016/S0009-9236(97)90196-X
PMID:9129563
[本文引用:1]
|
[12] |
Codeine is an analgesic drug acting on mu-opiate receptors predominantly via its metabolite morphine, which is formed almost exclusively by the genetically polymorphic enzyme cytochrome P450 2D6 (CYP2D6). Whereas it is known that individuals lacking CYP2D6 activity (poor metabolizers, PM) suffer from poor analgesia from codeine, ultra-fast metabolizers (UM) due to the CYP2D6 gene duplication may experience exaggerated and even potentially dangerous opioidergic effects and no systematical study has been performed so far on this question. A single dose of 30 mg codeine was administered to 12 UM of CYP2D6 substrates carrying a CYP2D6 gene duplication, 11 extensive metabolizers (EM) and three PM. Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism methods and a single-base primer extension method for characterization of the gene-duplication alleles. Pharmacokinetics was measured over 24 h after drug intake and codeine and its metabolites in plasma and urine were analyzed by liquid chromatography with tandem mass spectrometry. Significant differences between the EM and UM groups were detected in areas under the plasma concentration versus time curves (AUCs) of morphine with a median (range) AUC of 11 (5-17) microg h l(-1) in EMs and 16 (10-24) microg h l(-1) in UM (P=0.02). In urine collected over 12 h, the metabolic ratios of the codeine+codeine-6-glucuronide divided by the sum of morphine+its glucuronides metabolites were 11 (6-17) in EMs and 9 (6-16) in UM (P=0.05). Ten of the 11 CYP2D6 UMs felt sedation (91%) compared to six (50%) of the 12 EMs (P=0.03). CYP2D6 genotypes predicting ultrarapid metabolism resulted in about 50% higher plasma concentrations of morphine and its glucuronides compared with the EM. No severe adverse effects were seen in the UMs in our study most likely because we used for safety reasons a low dose of only 30 mg. It might be good if physicians would know about the CYP2D6 duplication genotype of their patients before administering codeine.
DOI:10.1038/sj.tpj.6500406
PMID:16819548
[本文引用:1]
|
[13] |
|
[14] |
|
[15] |
|
[16] |
Tramadol inhibits norepinephrine reuptake, stimulates serotonin release, and acts with mu-opioid receptors by way of its metabolite (+)-M1. Formation of M1 seems to depend on the genetic polymorphic CYP2D6. The analgesic effect of 2 mg/kg tramadol was evaluated in 15 extensive and 12 poor metabolizers of sparteine in two parallel, randomized, double-blind, placebo-controlled crossover studies that used experimental pain models. In extensive metabolizers, tramadol increased pressure pain detection (p = 0.03) and tolerance (p = 0.06) thresholds, as well as thresholds for eliciting nociceptive reflexes, after single (p = 0.0002) and repeated (p = 0.06) stimulation of the sural nerve. Peak pain and pain area in the cold pressor test were reduced (p = 0.0006 and 0.0009). In poor metabolizers, only thresholds to pressure pain tolerance (p = 0.02) and nociceptive reflexes after single stimulation (p = 0.04) were increased and the reflex threshold was less increased in poor metabolizers than in extensive metabolizers (p = 0.02). The serum concentration of (+)-M1 2 to 10 hours after tramadol ranged from 10 to 100 ng/L in extensive metabolizers, whereas in poor metabolizers serum concentrations of (+)-M1 were below or around the detection limit of 3 ng/ml. It is concluded that formation of (+)-M1 by way of CYP2D6 is important for the effect of tramadol on experimental pain.
PMID:8988065
[本文引用:1]
|
[17] |
|
[18] |
Tramadol is a synthetic, centrally acting analgesic for the treatment of moderate to severe pain. The marketed tramadol is a racemic mixture containing 50% (+)tramadol and 50% (-)tramadol and is mainly metabolized to O-desmethyltramadol (M1) by the cytochrome P450 CYP2D6. Tramadol is generally considered to be devoid of any serious adverse effects of traditional opioid receptor agonists, such as respiratory depression and drug dependence.
A 22-year-old Caucasian female patient was admitted to our ICU in refractory cardiac arrest requiring extracorporeal membrane oxygenation. This aggressive support allowed resolution of multi-organ dysfunction syndrome. Repeated blood analyses using liquid chromatography-tandem mass spectrometry confirmed high concentrations of both tramadol and its main metabolite O-desmethyltramadol. Genotyping of CYP2D6 revealed the patient to be heterozygous for a duplicated wild-type allele, predictive of a CYP2D6 ultrarapid metabolizer (UM) phenotype, confirmed by calculation of the tramadol/M1 (MR1) metabolic ratio at all time points.
We here report a case of near-fatal isolated tramadol cardiotoxicity. Because of the inhibition of norepinephrine reuptake, excessive blood epinephrine levels in this CYP2D6R UM patient following excessive tramadol ingestion could explain the observed strong myocardial stunning. This patient admitted intermittent tramadol consumption to gain a "high" sensation. In patients with excessive morphinomimetic effects, levels of tramadol and its main metabolite M1could be measured, ideally combined with CYP2D6 genotyping, to identify individuals at risk of tramadol-related cardiotoxicity. Tramadol treatment could be optimized in these at-risk individuals, consequently improving patient outcome and safety.
DOI:10.1007/s00228-011-1080-x
PMID:21691803
[本文引用:1]
|
[19] |
|
[20] |
The contribution of cytochrome P450 2D6 (CYP2D6) to the formation of hydrocodone's active metabolite, hydromorphone, was examined in vitro and in vivo. Human liver microsomes prepared from an individual homozygous for the D6-B mutation of the CYP2D6 gene catalyzed this reaction at a negligible rate. Urinary metabolic ratios of hydrocodone/hydromorphone were highly correlated with O-demethylation ratios for dextromethorphan, an established marker drug of CYP2D6 activity (rs = 0.85; n = 18). The kinetics of hydrocodone after a single oral dose and its partial metabolic clearance to hydromorphone were investigated in five extensive metabolizers of dextromethorphan, six poor metabolizers, and four extensive metabolizers after pretreatment with quinidine, a selective inhibitor of CYP2D6 activity. The mean values for partial metabolic clearance by O-demethylation in the three groups were 28.1 +/- 10.3, 3.4 +/- 2.4, and 5.0 +/- 3.6 ml/hr/kg, respectively. No statistically significant phenotypic differences in physiologic measures were observed. However, over the first hour after dosing, the extensive metabolizers reported more "good opiate effects" and fewer "bad opiate effects" than poor metabolizers and extensive metabolizers in whom CYP2D6 was inhibited by quinidine. These data establish the importance of CYP2D6 in the formation of hydromorphone from hydrocodone and suggest that the activity of this enzyme may limit the abuse liability of hydrocodone.
DOI:10.1038/clpt.1993.177
PMID:7693389
[本文引用:1]
|
[21] |
|
[22] |
|
[23] |
Opioids are recommended by the World Health Organization for moderate to severe cancer pain. Oxycodone is one of the most commonly used opioids and is metabolized in the liver by CYP3A4 and CYP2D6 enzymes. The aim of this cross-sectional study was to assess the relationship between oxycodone pharmacokinetics, pharmacodynamics and the CYP2D6 genotypes "poor metaboliser" (PM), "extensive metaboliser" (EM) and "ultra-rapid metaboliser" (URM) in a cohort of patients with cancer pain.
The patients were genotyped for the most common CYP2D6 variants and serum concentrations of oxycodone and metabolites were determined. Pain was assessed using the Brief Pain Inventory (BPI). The EORTC QLQ-C30 was used to assess the symptoms of tiredness and nausea. Cognitive function was assessed by the Mini Mental State (MMS) examination. Associations were examined by analyses of variance (ANOVA) and covariance (ANCOVA), or ordinal logistic regressions with and without covariates.
The sample consisted of 27 PM, 413 EM (including heterozygotes) and 10 URM. PM had lower oxymorphone and noroxymorphone serum concentrations and oxymorphone to oxycodone ratios than EM and URM. No differences between PM, EM and URM in pain intensity, nausea, tiredness or cognitive function was found.
CYP2D6 genotypes caused expected differences in pharmacokinetics, but they had no pharmacodynamic consequence. CYP2D6 genotypes did not influence pain control, the adverse symptoms nausea and sedation or the risk for cognitive failure in this study of patients treated with oxycodone for cancer pain.
DOI:10.1007/s00228-011-1093-5
PMID:21735164
[本文引用:1]
|
[24] |
|
[25] |
|
[26] |
|
[27] |
|
[28] |
|
[29] |
|
[30] |
|
[31] |
|
[32] |
|
[33] |
|
[34] |
Acute nociceptive pain management in children is a major public health concern. Effective and safe pain treatment is essential, but safety data cannot be simply extrapolated from adults to children due to pharmacokinetic and pharmacodynamic specificities. In addition, the frequent absence of child-specific data, the difficulty to assess drug tolerability, and the infants' inability to communicate properly and voluntarily report adverse drug reactions make children more vulnerable to safety issues. Awareness of the possible toxicity of analgesics is important but should not lead to suboptimal dosing and underuse of analgesia. A better assessment and individualization of treatment should allow effective prescribing of analgesics in more secure conditions. This article aims to review the safety of acetaminophen, nonsteroidal anti-inflammatory drugs, and opioids in children and the precautions that should be taken.
© 2019 The Authors Clinical Pharmacology & Therapeutics © 2019 American Society for Clinical Pharmacology and Therapeutics.
DOI:10.1002/cpt.1358
PMID:30648741
[本文引用:1]
|
[35] |
|
[36] |
|
[37] |
|
[38] |
|
[39] |
|
[40] |
|