Population pharmacokinetics

群体药代动力学
  • 文章类型: Journal Article
    曲美替尼是一种用于治疗实体瘤的靶向疗法,在现实生活中的研究中报告了显著的变异性。这种可变性增加了次优暴露的风险,这可能导致治疗失败或毒性增加。使用基于模型的仿真,本研究旨在表征和研究曲美替尼目前推荐剂量的药代动力学和适宜性.此外,通过模拟各种次优依从性情景,可以评估患者药物依从性对治疗结果的影响.收集了33名成年患者的人口数据,提供113血浆曲美替尼浓度,最好用具有线性吸收和消除的两室模型来描述。该研究还确定了无脂肪质量的显着积极影响和年龄对清除率的负面影响,解释了66%和21%的初始相关变异性,分别。模拟显示,与72%的女性患者相比,每天2mg的最大剂量在36%的男性患者中达到治疗目标。在65岁以上的患者中,每天1.5mg的剂量达到了相似的比率,男性和女性患者分别占44%和79%,分别,达到治疗目标。较差的依从性导致浓度的显著下降和亚治疗药物水平的高风险。这些结果强调了跨专业合作和患者伙伴关系在患者的旅程中的重要性,以满足患者对曲美替尼的需求并支持药物依从性。
    Trametinib is a targeted therapy used for the treatment of solid tumours, with significant variability reported in real-life studies. This variability increases the risk of suboptimal exposure, which can lead to treatment failure or increased toxicity. Using model-based simulation, this study aims to characterize and investigate the pharmacokinetics and the adequacy of the currently recommended doses of trametinib. Additionally, the simulation of various suboptimal adherence scenarios allowed for an assessment of the impact of patients\' drug adherence on the treatment outcome. The population data collected in 33 adult patients, providing 113 plasmatic trametinib concentrations, were best described by a two-compartment model with linear absorption and elimination. The study also identified a significant positive effect of fat-free mass and a negative effect of age on clearance, explaining 66% and 21% of the initial associated variability, respectively. Simulations showed that a maximum dose of 2 mg daily achieved the therapeutic target in 36% of male patients compared to 72% of female patients. A dose of 1.5 mg per day in patients over 65 years of age achieved similar rates, with 44% and 79% for male and female patients, respectively, reaching the therapeutic target. Poor adherence leads to a significant drop in concentrations and a high risk of subtherapeutic drug levels. These results underline the importance of interprofessional collaboration and patient partnership along the patient\'s journey to address patients\' needs regarding trametinib and support medication adherence.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    MAPK已被报道为犬组织细胞肉瘤的关键致癌途径,可以用曲美替尼进行药理学靶向,MEK1/2的小抑制剂。初步数据在体外和体内模型中显示出有希望的抗肿瘤活性,并代表了将发现从工作台到床边的概念证明。在这个阶段I,使用3+3队列设计的剂量递增研究,曲美替尼在18只癌症犬中进行了评估。根据VCOG-CTCAEv2对不良事件进行分级。收集血液样品和肿瘤活检用于药代动力学和药效学评估。曲美替尼耐受性良好,最大耐受剂量为0.5mg/m2/天,PO.剂量限制性毒性包括全身性高血压,蛋白尿,嗜睡和ALP升高,都是三年级。由于曲美替尼的消除是可饱和的,因此药物暴露随剂量呈线性增加。在500μgQ24h的剂量下(30kg狗中0.5mg/m2/天),大约70%的狗的平均稳态浓度为10ng/mL,大约2周后完成。该阈值与人类的临床疗效相关。在第0天和第7天收集的生物样本中未观察到目标接合。总之,曲美替尼在癌症犬中被认为是安全的,0.5mg/m2/天的剂量是II期研究的推荐剂量。
    MAPK has been reported as a key oncogenic pathway for canine histiocytic sarcoma, which can be pharmacologically targeted with trametinib, a small inhibitor of MEK1/2. Preliminary data showed promising antitumor activity in in vitro and in vivo models and represented a proof of concept to translate the findings from bench to bedside. In this phase I, dose escalating study using a 3 + 3 cohort design, trametinib was evaluated in 18 dogs with cancer. Adverse events were graded according to VCOG-CTCAE v2. Blood samples and tumour biopsies were collected for pharmacokinetic and pharmacodynamic assessment. Trametinib was well tolerated with a maximum tolerated dose of 0.5 mg/m2/day, PO. Dose-limiting toxicities included systemic hypertension, proteinuria, lethargy and elevated ALP, and were all Grade 3. The drug exposures increased more than linearly with dose since the elimination of trametinib was saturable. At a dose of 500 μg Q24h (0.5 mg/m2/day in a 30 kg dog), approximately 70% of dogs had an average steady-state concentration of 10 ng/mL, achieved after approximately 2 weeks. This threshold was associated with clinical efficacy in humans. Target engagement was not observed in biospecimens collected on Days 0 and 7. In conclusion, trametinib was considered safe in dogs with cancer, and the dose of 0.5 mg/m2/day was the recommended dose for phase II studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Teverelix药物产品(DP)是一种肠胃外促性腺激素释放激素(GnRH)拮抗剂,已在激素敏感性晚期前列腺癌(APC)和良性前列腺增生(BPH)的2期试验中成功进行了测试。在之前的APC试验中,teterlixDP作为肌肉内(IM)和皮下(SC)注射,使用负荷剂量和(在单次试验中)维持剂量。我们的目标是为3期临床开发得出最佳给药方案,使用药物计量学建模方法。利用来自9项2期研究(229名患者)的数据来开发群体药代动力学(PK)模型,该模型描述了适应IM和SC给药途径的浓度曲线。具有顺序一阶吸收(缓慢和快速)和滞后时间的2室模型最好地描述了SC和IM给药后的泰维立利的PK曲线。基于生理相关性,拟合具有生产率抑制的间接反应模型来描述睾酮(T)浓度。使用最终群体PK-药效学模型来进行各种候选给药方案的模拟,以选择最佳给药方案来实现临床去势(到第28天T<0.5ng/mL)并维持临床去势26周。模型模拟显示,从第28天开始,360mgSC和180mgIM的负荷剂量与360mgSC6周(Q6W)的维持剂量可以达到≥95%的去势率,直至52周。选择此剂量方案用于3期临床开发,其中包括与GnRH激动剂相比的心血管安全性评估。
    Teverelix drug product (DP) is a parenteral gonadotropin-releasing hormone (GnRH) antagonist that has been successfully tested in phase 2 trials for hormone-sensitive advanced prostate cancer (APC) and benign prostatic hyperplasia (BPH). In previous APC trials, teverelix DP was administered as intramuscular (IM) and subcutaneous (SC) injections, using a loading dose and (in a single trial) a maintenance dose. Our objective was to derive an optimal dosing regimen for phase 3 clinical development, using a pharmacometrics modeling approach. Data from 9 phase 2 studies (229 patients) was utilized to develop a population pharmacokinetic (PK) model that described the concentration profile accommodating both IM and SC routes of administration. A 2-compartment model with sequential first-order absorption (slow and fast) and lag times best described the PK profiles of teverelix following SC and IM administration. An indirect response model with inhibition of production rate was fit to describe testosterone (T) concentrations based on physiological relevance. The final population PK-pharmacodynamic model was used to conduct simulations of various candidate dosing regimens to select the optimal dosing regimen to achieve clinical castration (T < 0.5 ng/mL by day 28) and to sustain clinical castration for 26 weeks. Model simulation showed that a loading dose of 360 mg SC and 180 mg IM with a maintenance dose of 360 mg SC 6-weekly (Q6W) starting at day 28 can achieve a ≥95% castration rate up to 52 weeks. This dose regimen was selected for phase 3 clinical development, which includes cardiovascular safety assessment in comparison to a GnRH agonist.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:手足皮肤反应(HFSR)是多激酶抑制剂(MKI)的常见剂量限制性不良反应,其机制尚未完全了解,预防是不够的。
    目的:在本试验研究中,双盲,我们进行了随机安慰剂对照试验,以评估局部尿素在肾细胞癌患者舒尼替尼诱导的HFSR二级预防中的作用.
    方法:在55名筛查患者中,14人随机接受局部尿素或安慰剂4周。HFSR与舒尼替尼及其代谢物(正去乙基舒尼替尼)的药物水平的关联,VEGFR2基因的遗传多态性,还评估了生活质量(QOL)和生化指标.
    结果:结果表明,基于尿素的乳膏并不优于安慰剂(P=.075)。两组的QOL均无变化。检查了位于4号染色体VEGFR2基因中的两个核苷酸rs1870377和rs2305948的单核苷酸多态性。与野生型相比,rs1870377的SNP(变体T>A)与新HFSR的出现有关,尽管相关性无统计学意义(OR0.714).与安慰剂组相比,尿素组中舒尼替尼和N-去乙基舒尼替尼的平均血浆水平之间没有统计学上的显著差异。舒尼替尼的最佳群体药代动力学模型是具有一阶吸收和线性消除的单室模型。根据Ka的群体药代动力学模型计算的群体参数的中位数(IQR),V和Cl为0.22(0.21-0.24)h-1,4.4(4.09-4.47)L,0.049(0.042-0.12)L/hr,分别。
    结论:研究表明,在接受舒尼替尼4:2方案的肾癌患者中,尿素乳膏在减少新的HFSR出现方面并不优于安慰剂。
    BACKGROUND: Hand foot skin reaction (HFSR) is a common dose-limiting adverse effect of multi kinase inhibitors (MKI) whose mechanism is not fully understood, and the prophylaxis is inadequate.
    OBJECTIVE: In this pilot study, a double-blind, randomized placebo-controlled trial was conducted to evaluate the effect of topical urea in secondary prevention of sunitinib-induced HFSR in renal cell cancer patients.
    METHODS: Out of 55 screened patients, 14 were randomized to receive topical urea or placebo for four weeks. The association of HFSR with drug levels of sunitinib and its metabolite (n-desethyl sunitinib), genetic polymorphism of VEGFR2 gene, quality of life (QOL) and biochemical markers was also assessed.
    RESULTS: The results showed that urea-based cream was not superior to placebo (P = .075). There was no change in the QOL in both the groups. Single nucleotide polymorphism was checked for two nucleotides rs1870377 and rs2305948 located in VEGFR2 gene on chromosome 4. SNP (variant T > A) at rs1870377 was associated with appearance of new HFSR as compared to the wild type, although the association was not statistically significant (OR 0.714). There was no statistically significant difference between mean plasma levels of sunitinib and N-desethyl sunitinib in urea arm as compared to placebo arm as compared to placebo. The best fit population pharmacokinetic model for sunitinib was one compartment model with first order absorption and linear elimination. The median (IQR) of population parameters calculated from the population pharmacokinetics model for Ka, V and Cl was 0.22 (0.21-0.24) h-1, 4.4 (4.09-4.47) L, 0.049 (0.042-0.12) L/hr, respectively.
    CONCLUSIONS: The study suggested that the urea-based cream was not superior to placebo in decreasing the appearance of new HFSR in renal cancer patients receiving 4:2 regimen of sunitinib.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:本研究的目的是探讨癫痫患儿的药物动力学,确定导致受试者之间药代动力学变化的因素,评估PER暴露与临床结果之间的联系,并建立了一种基于证据的方法来定制该特定人群的个体化抗癫痫治疗。
    方法:在这项前瞻性研究中,从194名年龄小于18岁的患者中获得了PER血浆浓度和代谢酶的遗传信息。使用非线性混合效应模型表征口服给药后儿科患者中PER的处置动力学。通过评估PER治疗的有效性和安全性以及分析药物暴露与临床反应之间的关系来确定PER血浆浓度的有效范围。然后进行蒙特卡罗模拟以评估和优化当前的给药方案。
    结果:PER的药代动力学曲线由一级吸收和消除的一室模型充分描述。体重,总胆红素水平,发现并伴有奥卡西平对PER药代动力学有重大影响。表观清除率和分布体积的模型估计值分别为.016±.009L/h/kg和1.47±.78L/kg,分别。根据响应者的血浆浓度数据预测的有效范围为215-862μg/L。通过模拟分析,提出了根据基本协变量分层的给药方案。
    结论:在这项研究中,我们通过对真实世界数据的分析,捕获了小儿癫痫患者中PER的药代动力学/药效学特征,并采用了药物计量学方法,以支持该特定人群中PER的个体化给药策略.
    OBJECTIVE: The purposes of this study were to explore the pharmacokinetics of perampanel (PER) in children with epilepsy, identify factors that contribute to pharmacokinetic variations among subjects, evaluate the connection between PER exposure and clinical outcome, and establish an evidence-based approach for tailoring individualized antiepileptic treatment in this specific population.
    METHODS: In this prospective study, PER plasma concentrations and genetic information on metabolic enzymes were obtained from 194 patients younger than 18 years. The disposition kinetics of PER in pediatric patients following oral dosing were characterized using nonlinear mixed effect models. The effective range for the plasma concentration of PER was determined by assessing the efficacy and safety of PER treatment and analyzing the relationship between drug exposure and clinical response. Monte Carlo simulations were then performed to evaluate and optimize the current dosing regimens.
    RESULTS: The pharmacokinetic profile of PER was adequately described by a one-compartment model with first-order absorption and elimination. Body weight, total bilirubin level, and concomitant oxcarbazepine were found to have significant influences on PER pharmacokinetics. Model estimates of apparent clearance and volume of distribution were .016 ± .009 L/h/kg and 1.47 ± .78 L/kg, respectively. The effective range predicted from plasma concentration data in responders was 215-862 μg/L. Dosing scenarios stratified according to essential covariates were proposed through simulation analysis.
    CONCLUSIONS: In this study, we captured the pharmacokinetic/pharmacodynamic characteristics of PER in pediatric epilepsy patients through analysis of real-world data and adopted a pharmacometric approach to support an individualized dosing strategy for PER in this specific population.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:西替利嗪在临床实践中经常以增加的剂量给药,并由一些指南推荐。尽管如此,中国儿科人群的药代动力学(PK)概况和真实世界安全性数据仍然不足.本研究的目的是在中国儿科患者中开发西替利嗪的群体药代动力学(PPK)模型,并研究其标签外使用的基本原理。方法:进行前瞻性队列研究,招募被诊断患有过敏性疾病并处方西替利嗪的儿童。结果是安全性和药代动力学(PK)参数。使用预先建立的分析方法测量西替利嗪浓度。随后,开发了一个PK模型,其次是模型评估和仿真。开发的PK模型用于研究不同年龄组的药物暴露差异,并模拟潜在的过量情况。结果:63名儿童入组,其中24人接受的西替利嗪剂量超过推荐剂量。PPK模型,根据已发表的文献,作为我们分析的基础,进行调整以估计某些参数。最终的模型评估和验证表明,预测性能准确,参数估计稳健。在1-12岁之间对标签剂量进行的模拟表明,稳态下的中位数最大浓度(Cmax,ss)的7岁儿童可能是最高的。该模型还用于预测标签外剂量情景和过量患者以支持临床决策。两组均无药物不良反应。结论:本研究为优化中国儿科患者西替利嗪的使用提供了循证和基于模型的探索。西替利嗪PPK模型显示出准确的预测性能,可用于模拟真实世界临床场景中的个体患者暴露。
    Aims: Cetirizine is frequently administered at an increased dosage in clinical practice and recommended by several guidelines. Nonetheless, the pharmacokinetic (PK) profile and real-world safety data remain insufficient in the Chinese pediatric population. The objective of the current study is to develop a population pharmacokinetic (PPK) model for cetirizine in Chinese pediatric patients and to investigate the rationale behind its off-label usage. Methods: A prospective cohort study was conducted, enrolling children who had been diagnosed with allergic diseases and prescribed cetirizine. The outcomes were safety and pharmacokinetic (PK) parameters. Cetirizine concentrations were measured using a pre-established analytical method. Subsequently, a PK model was developed, followed by model evaluation and simulation. The developed PK model was employed to investigate the drug exposure differences across various age groups and to simulate scenarios of potential overdose. Results: Sixty-three children were enrolled, and 24 of them received a cetirizine dose exceeding the recommended dosage. A PPK model, based on published literature, served as the basis of our analysis, with adjustment made to estimate certain parameters. The final model evaluation and validation indicated accurate predictive performance and robust parameter estimation. Simulations conducted for the label-dose among age 1-12 indicated median maximum concentration at steady state (Cmax,ss) of 7 year old children could be the highest. The model was also used to predict the off-label dose scenarios and overdose patient to support the clinical decision. There were no adverse drug reactions in either group. Conclusion: This study provides evidence-based and model-based exploration for optimizing cetirizine usage in Chinese pediatric patients. The cetirizine PPK model showed accurate predictive performance and could be utilized to simulate individual patient exposure in real-world clinical scenarios.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:外用布雷帕替尼,酪氨酸激酶(TYK)2/Janus激酶(JAK)1抑制剂,正在发展为牛皮癣(PsO)和特应性皮炎(AD)。先前临床试验数据的定量分析用于为未来的临床试验设计提供信息。
    方法:使用线性混合效应回归(LMER),对患有AD和PsO的患者进行了两项2b期研究,以表征局部布雷帕替尼的量和由此产生的全身波谷浓度(CTrough)。该模型用于预测成人和儿童的较高治疗体表面积(BSA)的布雷帕替尼全身性CTrough。利用口服布雷帕替尼的非临床和临床试验的信息来设定安全阈值。该组合方法用于告知未来的剂量强度选择和治疗的BSA限制。
    结果:分析了256例患者的数据。患者类型,剂量强度,和频率对剂量-暴露关系有显著影响。预测PsO患者的全身浓度比相同剂量的AD患者低45%。当局部应用于相同百分比的BSA时,预计布雷帕替尼的全身暴露在成人和儿童之间具有可比性.在AD和PsO患者中,每天一次和每天两次(2mg/cm2)乳膏应用于少于50%BSA后的全身稳态暴露量为3%,分别,与非临床安全性发现和临床血液学标志物保持至少三倍的界限。
    结论:应用活性药物的量与布波替尼全身性CTrough之间的关系,由LMER描述,可能会在布雷帕替尼主题计划中提供剂量优化的开发策略。
    BACKGROUND: Topical brepocitinib, a tyrosine kinase (TYK)2/Janus kinase (JAK)1 inhibitor, is in development for psoriasis (PsO) and atopic dermatitis (AD). Quantitative analyses of prior clinical trial data were used to inform future clinical trial designs.
    METHODS: Two phase 2b studies in patients with AD and PsO were used to characterize the amount of topical brepocitinib and the resultant systemic trough concentration (CTrough) using a linear mixed-effects regression (LMER). This model was used to predict brepocitinib systemic CTrough for higher treated body surface areas (BSAs) in adults and children. Information from non-clinical and clinical trials with oral brepocitinib was leveraged to set safety thresholds. This combined approach was used to inform future dose-strength selection and treated BSA limits.
    RESULTS: Data from 256 patients were analyzed. Patient type, dose strength, and frequency had significant impacts on the dose-exposure relationship. Systemic concentration in patients with PsO was predicted to be 45% lower than in patients with AD from the same dose. When topically applied to the same percentage BSA, brepocitinib systemic exposures are expected to be comparable between adults and children. The systemic steady-state exposure after 3% once daily and twice daily (2 mg/cm2) cream applied to less than 50% BSA in patients with AD and PsO, respectively, maintains at least a threefold margin to non-clinical safety findings and clinical hematologic markers.
    CONCLUSIONS: The relationship between the amount of active drug applied and brepocitinib systemic CTrough, described by LMER, may inform the development strategy for dose optimization in the brepocitinib topical program.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:利福平治疗药物易感结核病(TB)的最佳给药策略仍存在很大争议。在3期临床试验研究31/ACTG5349(NCT02410772)中,对照方案组的所有参与者均接受600mg利福平每日固定剂量.这里,我们评估了利福平暴露与疗效和安全性结局之间的关系.
    方法:我们使用群体非线性混合效应模型分析了利福平浓度时间曲线。我们比较了平分和重分给药的模拟利福平暴露。我们评估了利福平暴露对6个月时稳定培养物转化的影响,使用Cox比例风险模型在9、12和18个月时与结核病相关的不利结果,和使用逻辑回归的所有试验定义的安全性结果。
    结果:我们的模型衍生的利福平暴露范围为4.57mg·h/L至140.0mg·h/L,中位数为41.8mg·h/L。药代动力学模拟表明,统一剂量的利福平提供了类似于体重带状剂量的暴露覆盖率。暴露效果分析(N=680)显示,利福平暴露低于中位数的参与者与暴露高于中位数的参与者相比,经历了类似的稳定培养物转化和结核病相关不利结果的危险。暴露-安全性分析(N=722)显示,增加利福平暴露与增加3级或更高的不良事件无关。或严重不良事件。
    结论:对于6个月的标准治疗方案,每日600mg利福平的平分给药可能是现有的体重带给药策略的合理替代方案。未来的研究应该评估利福平的最佳给药策略,剂量高于目前的建议。
    BACKGROUND: The optimal dosing strategy for rifampicin in treating drug-susceptible tuberculosis (TB) is still highly debated. In the phase 3 clinical trial Study 31/ACTG 5349 (NCT02410772), all participants in the control regimen arm received 600 mg rifampicin daily as a flat dose. Here, we evaluated relationships between rifampicin exposure and efficacy and safety outcomes.
    METHODS: We analyzed rifampicin concentration time profiles using population nonlinear mixed-effects models. We compared simulated rifampicin exposure from flat- and weight-banded dosing. We evaluated the effect of rifampicin exposure on stable culture conversion at 6 months; TB-related unfavorable outcomes at 9, 12, and 18 months using Cox proportional hazard models; and all trial-defined safety outcomes using logistic regression.
    RESULTS: Our model-derived rifampicin exposure ranged from 4.57 mg · h/L to 140.0 mg · h/L with a median of 41.8 mg · h/L. Pharmacokinetic simulations demonstrated that flat-dosed rifampicin provided exposure coverage similar to the weight-banded dose. Exposure-efficacy analysis (n = 680) showed that participants with rifampicin exposure below the median experienced similar hazards of stable culture conversion and TB-related unfavorable outcomes compared with those with exposure above the median. Exposure-safety analysis (n = 722) showed that increased rifampicin exposure was not associated with increased grade 3 or higher adverse events or serious adverse events.
    CONCLUSIONS: Flat-dosing of rifampicin at 600 mg daily may be a reasonable alternative to the incumbent weight-banded dosing strategy for the standard-of-care 6-month regimen. Future research should assess the optimal dosing strategy for rifampicin, at doses higher than the current recommendation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:由于危重病人的异质性,替加环素的药代动力学尚不清楚,最佳给药策略是有争议的。方法:进行单中心前瞻性临床研究,包括接受替加环素治疗的危重患者。对血液样本进行了密集采样(每个样本八个),并测定血浆药物浓度。建立了群体药代动力学(PPK)模型,并通过拟合优度图进行了评估,引导分析和视觉预测检查。进行蒙特卡罗模拟以优化给药方案。结果:总体而言,包括来自98例患者的751个观察结果。最终的PPK模型是包含肌酐清除率(CL)协变量的两室模型,中心和外周分布体积上的体重(V1和V2),γ-谷氨酰转移酶和总胆红素对室间清除(Q),和白蛋白在V2上。CL的典型值,Q,V1和V2为3.09L/h,39.7L/h,32.1升和113升,分别。50mg/12h的给药方案适用于复杂的腹腔内感染,但是社区获得性肺炎需要100毫克/12小时,由敏感性较低的细菌引起的皮肤和皮肤结构感染。结论:成功建立并验证了替加环素PPK模型。替加环素的个体化给药可能对危重患者有益。
    Background: Due to the heterogeneity of critically ill patients, the pharmacokinetics of tigecycline are unclear, and the optimal dosing strategy is controversial. Methods: A single-center prospective clinical study that included critically ill patients who received tigecycline was performed. Blood samples were intensively sampled (eight samples each), and plasma drug concentrations were determined. A population pharmacokinetic (PPK) model was developed and evaluated by goodness-of-fit plots, bootstrap analysis and visual predictive checks. Monte Carlo simulation was conducted to optimize the dosage regimen. Results: Overall, 751 observations from 98 patients were included. The final PPK model was a two-compartment model incorporating covariates of creatinine clearance on clearance (CL), body weight on both central and peripheral volumes of distribution (V1 and V2), γ-glutamyl transferase and total bilirubin on intercompartment clearance (Q), and albumin on V2. The typical values of CL, Q, V1 and V2 were 3.09 L/h, 39.7 L/h, 32.1 L and 113 L, respectively. A dosage regimen of 50 mg/12 h was suitable for complicated intra-abdominal infections, but 100 mg/12 h was needed for community-acquired pneumonia, skin and skin structure infections and infections caused by less-susceptive bacteria. Conclusion: The Tigecycline PPK model was successfully developed and validated. Individualized dosing of tigecycline could be beneficial for critically ill patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    霉酚酸酯(MMF)通常用于异基因造血干细胞移植(HSCT)后的急性移植物抗宿主病(aGVHD)。然而,有限群体药代动力学(PPK)数据可用于儿科HSCT患者.本研究旨在开发PPK模型并推荐小儿HSCT患者的最佳口服MMF剂量。这项前瞻性研究涉及三级学术机构的小儿HSCT患者。患者每天两次口服MMF15-20mg/kg用于aGVHD预防和治疗。使用非线性混合效应建模方法进行PPK分析。考虑不同体表面积(BSA)(0.5m2、1.0m2、1.5m2)和给药(400mg/m2、600mg/m2、900mg/m2,每天两次)进行模拟。基于模拟,建议口服MMF的最佳剂量。在PPK模型开发中包括总共20名患者和80个样本。一级吸收的单室模型充分描述了霉酚酸(MPA)的药代动力学。BSA是Vd/F上具有统计学意义的协变量。模拟建议口服MMF的最佳剂量为每天两次900mg/m2,分别。建立了可靠的PPK模型,具有良好的预测性能。这种模型告知儿科HSCT患者的最佳MMF剂量可以在现实世界的临床实践中提供有价值的给药指导。
    Mycophenolate mofetil (MMF) is commonly used for acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation (HSCT). However, limited population pharmacokinetic (PPK) data are available for pediatric HSCT patients. This study aimed to develop a PPK model and recommend optimal oral MMF dosage in pediatric HSCT patients. This prospective study involved pediatric HSCT patients at a tertiary academic institution. Patients received oral MMF 15-20 mg/kg twice daily for aGVHD prophylaxis and treatment. The PPK analysis was conducted using a nonlinear mixed-effects modeling method. Simulation was performed considering different body surface areas (BSAs) (0.5 m2, 1.0 m2, 1.5 m2) and dosing (400 mg/m2, 600 mg/m2, 900 mg/m2 twice daily). Based on the simulation, an optimal dosage of oral MMF was suggested. A total of 20 patients and 80 samples were included in the PPK model development. A one-compartment model with first-order absorption adequately described the pharmacokinetics of mycophenolic acid (MPA). BSA was a statistically significant covariate on Vd/F. Simulation suggested the optimal dosage of oral MMF as 900 mg/m2 twice daily, respectively. A reliable PPK model was developed with good predictive performance. This model-informed optimal MMF dosage in pediatric HSCT patients can provide valuable dosing guidance in real-world clinical practice.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号