GastroPlus

Gastroplus
  • 文章类型: Journal Article
    生物等效性研究在仿制药开发中至关重要,其中创新产品提供了治疗等效性。然而,由于与药物相关的多种因素的相互作用,生物等效性研究表现出显著的复杂性,配方,生理学,和药代动力学。诸如基于生理的生物制药建模(PBBM)之类的方法可以通过适当开发和验证的模型来实现虚拟生物等效性(VBE)评估。这些模型现在被广泛用于生物等效性风险评估,内部决策,以及与生物等效性相关的药物和制剂因素的评价。通过结合变异性和开发虚拟种群进行生物等效性评估来描述上述因素对于利用此类模型至关重要。在这份手稿中,我们已经描绘了我们目前对VBE的理解。提供了关于研究设计的详细解释,体内变异性,和生理的影响,药物,和制定VBE种群发展的因素。此外,建议将变异性纳入GastroPlus的策略,重点是受试者内和跨场合的变异性。描绘了两个与立即释放和改良释放制剂有关的工业案例研究,其中VBE用于决策和监管依据。最后,在VBE领域的监管理解,以及未来的前景,是详细的。
    Bioequivalence studies are pivotal in generic drug development wherein therapeutic equivalence is provided with an innovator product. However, bioequivalence studies represent significant complexities due to the interplay of multiple factors related to drug, formulation, physiology, and pharmacokinetics. Approaches such as physiologically based biopharmaceutics modeling (PBBM) can enable virtual bioequivalence (VBE) assessment through appropriately developed and validated models. Such models are now being extensively used for bioequivalence risk assessment, internal decision-making, and the evaluation of drug and formulation factors related to bioequivalence. Depiction of the above-mentioned factors through the incorporation of variability and development of a virtual population for bioequivalence assessment is of paramount importance in utilizing such models. In this manuscript, we have portrayed our current understanding of VBE. A detailed explanation was provided with respect to study designs, in vivo variability, and the impact of physiological, drug, and formulation factors on the development of the population for VBE. Furthermore, strategies are suggested to incorporate variability in GastroPlus with an emphasis on intra-subject and inter-occasion variability. Two industrial case studies pertaining to immediate and modified release formulation were portrayed wherein VBE was utilized for decision-making and regulatory justification. Finally, regulatory understanding in the area of VBE, along with future perspectives, was detailed.
    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
    基于计算机模拟的各种药物的肠道吸收预测已经成为现实。然而,尚未发现基于GastroPlus™的体内药代动力学模拟和虚拟生物等效性评估。本研究旨在模拟具有不同溶解曲线的血浆浓度,并运行群体模拟,以使用GastroPlus软件评估阿托伐他汀的测试和参考产品的生物等效性。阿托伐他汀(20mg片剂)的参考和测试产品的溶出曲线,和参考产品的临床血浆浓度-时间数据用于模拟。结果表明,成功建立了阿托伐他汀片的模拟模型。群体模拟结果表明测试制剂与参考制剂生物等效。研究结果表明,建模是证明阿托伐他汀药代动力学和生物等效性可能性的重要工具。这将有助于了解仿制产品开发过程中的潜在风险。
    The prediction of intestinal absorption of various drugs based on computer simulations has been a reality. However, in vivo pharmacokinetic simulations and virtual bioequivalence evaluation based on GastroPlus™ have not been found. This study aimed to simulate plasma concentrations with different dissolution profiles and run population simulations to evaluate the bioequivalence of test and reference products of atorvastation using GastroPlus software. The dissolution profiles of the reference and test products of atorvastatin (20 mg tablets), and clinical plasma concentration-time data of the reference product were used for the simulations. The results showed that the simulated models were successfully established for atorvastatin tablets. Population simulation results indicated that the test formulation was bioequivalent to the reference formulation. The findings suggest that modelling is an essential tool to demonstrating the possibility of pharmacokinetic and bioequivalence for atorvastatin. It will contribute to understanding the potential risks during the development of generic products.
    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
    该研究基于使用SeDeM专家系统开发盐酸曲马多控释渗透片及其基于体内药代动力学评估的计算机生理学药代动力学(PBPK)模型。在开发SeDEM驱动的全因子渗透药物递送中应用了基于设计质量(QbD)的方法。使用24全因子设计,以NaCl为渗透剂,制作盐酸曲马多渗透片的试验配方,MethocelK4M作为速率控制聚合物,和avicelpH101作为稀释剂。通过应用SeDeM专家工具测定制剂(F1-F16)的预制剂特征。使用GastroPlus™的PBPK“ACAT”模型对制剂进行优化,然后进行体内预测药代动力学评估。FTIR结果显示各成分之间没有相互作用。所有试验配方共混物的良好可压缩性指数(ICG)值≥5,表明直接压缩是最合适的方法。基于在2、10和16小时的药物释放以及零级动力学释放(r2=0.992和0.994)来优化制剂F3和F4。SEM图像证实在药物完全释放后在渗透片剂的表面上形成微孔。F3和F4也是稳定的(保质期29.41和23.46个月)。PBPK计算机模型的药代动力学的体内模拟显示,相对于盐酸曲马多50mgIR制剂,F3和F4具有出色的相对生物利用度。在设计每日一次渗透控释盐酸曲马多片剂时,最好利用SeDeM专家工具来评估选定的制剂赋形剂及其混合物的压缩特性,以用于直接压缩方法。SimuloGastroPlus™PBPK模型提供了对优化制剂的全面药代动力学评估,作为盐酸曲马多的体内研究的替代品。
    The study is based on using SeDeM expert system in developing controlled-release tramadol HCl osmotic tablets and its in-silico physiologically based pharmacokinetic (PBPK) modeling for in-vivo pharmacokinetic evaluation. A Quality by Design (QbD) based approach in developing SeDEM-driven full factorial osmotic drug delivery was applied. A 24 Full-factorial design was used to make the trial formulations of tramadol HCl osmotic tablets using NaCl as osmogen, Methocel K4M as rate controlling polymer, and avicel pH 101 as diluent. The preformulation characteristics of formulations (F1-F16) were determined by applying SeDeM Expert Tool. The formulation was optimized followed by in-vivo predictive pharmacokinetic assessment using PBPK \"ACAT\" model of GastroPlus™. The FTIR results showed no interaction among the ingredients. The index of good compressibility (ICG) values of all trial formulation blends were ≥5, suggesting direct compression is the best-suited method. Formulation F3 and F4 were optimized based on drug release at 2, 10, and 16 h with a zero-order kinetic release (r 2 = 0.992 and 0.994). The SEM images confirmed micropores formation on the surface of the osmotic tablet after complete drug release. F3 and F4 were also stable (shelf life 29.41 and 23.46 months). The in vivo simulation of the pharmacokinetics of the PBPK in-silico model revealed excellent relative bioavailability of F3 and F4 with reference to tramadol HCl 50 mg IR formulations. The SeDeM expert tool was best utilized to evaluate the compression characteristics of selected formulation excipients and their blends for direct compression method in designing once-daily osmotically controlled-release tramadol HCl tablets. The in-silico GastroPlus™ PBPK modeling provided a thorough pharmacokinetic assessment of the optimized formulation as an alternative to tramadol HCl in vivo studies.
    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
    埃洛石纳米管(HNTs)由于其天然可用性,是一种多功能且高度研究的粘土矿物,低成本,机械强度强,生物相容性,和绑定属性。本工作探讨了其在延缓和控制药物从复合聚合物基质材料释放中的作用。
    为此,使用“溶液流延法”配制包含盐酸普萘洛尔和不同浓度的HNT的纳米复合膜。使用浓度为1%w/v的薄荷醇作为渗透促进剂,并测定了其对释放和渗透的影响。确定了纳米复合材料的质量特性,使用Franz扩散系统进行了体外释放和渗透研究。使用各种数学模型和渗透参数分析数据。优化的配方也进行了皮肤刺激试验,FTIR,DSC,和SEM研究。使用GastroPlusTCAT®模型预测纳米复合材料中普萘洛尔HCl的系统吸收和处置。
    药物释放速率的控制与较高的HNTs浓度有关。F8在8小时内释放了50%的普萘洛尔(药物,HNTs比率,1:2).优化配方(F6)与药物:HNTs(2:1),在4小时内表现出80%的药物释放,最大通量为145.812µg/cm2hr。发现优化的配方对皮肤无刺激性,保质期为35.46个月(28-30℃)。计算机模拟模型预测了Cmax,Tmax,AUCt,和AUCinf为32.113ng/mL,16.58h,942.34ng/mL×h,1102.9ng/mL×h,分别。
    研究证明HNT可有效地用作基质型透皮制剂中的速率控制剂。
    UNASSIGNED: Halloysite nanotubes (HNTs) are a versatile and highly investigated clay mineral due to their natural availability, low cost, strong mechanical strength, biocompatibility, and binding properties. The present work explores its role for retarding and controlling the drug release from the composite polymer matrix material.
    UNASSIGNED: For this purpose, nanocomposite films comprising propranolol HCl and different concentrations of HNTs were formulated using the \"solution casting method\". The menthol in a concentration of 1% w/v was used as a permeation enhancer, and its effect on release and permeation was also determined. Quality characteristics of the nanocomposite were determined, and in vitro release and permeation studies were performed using the Franz diffusion system. The data was analyzed using various mathematical models and permeation parameters. Optimized formulation was also subjected to skin irritation test, FTIR, DSC, and SEM study. Systemic absorption and disposition of propranolol HCl from the nanocomposites were predicted using the GastroPlus TCAT® model.
    UNASSIGNED: The control in drug release rate was associated with the higher concentration of HNTs. F8 released 50% of propranolol within 8 hours (drug, HNTs ratio, 1:2). The optimized formulation (F6) with drug: HNTs (2:1), exhibited drug release 80% in 4 hours, with maximum flux of 145.812 µg/cm2hr. The optimized formulation was found to be a non-irritant for skin with a shelf life of 35.46 months (28-30 ℃). The in silico model predicted Cmax, Tmax, AUCt , and AUCinf as 32.113 ng/mL, 16.58 h, 942.34 ng/mL×h, and 1102.9 ng/mL×h, respectively.
    UNASSIGNED: The study demonstrated that HNTs could be effectively used as rate controlling agent in matrix type transdermal formulations.
    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
    The present work aimed to differentiate between in vitro dissolution profiles of ibuprofen as input for GastroPlus™ and to see the impact on systemic exposure. In vitro dissolution profiles of ibuprofen obtained under low- and high-buffered dissolution media were used as input using the z-factor approach. In a second step, a customized surface pH calculator was applied to predict the surface pH of ibuprofen under these low- and high-buffered dissolution conditions. These surface pH values were adopted in GastroPlus™ and simulations were performed to predict the systemic outcome. Simulated data were compared with systemic data of ibuprofen obtained under fasted state conditions in healthy subjects. The slower dissolution rate observed when working under low-buffered conditions nicely matched with the slower dissolution rate as observed during the clinical aspiration study and was in line with the systemic exposure of the drug. Finally, a population simulation was performed to explore the impact of z-factor towards bioequivalence (BE) criteria (so-called safe space). Concerning future perspectives, the customized calculator should be developed in such a way to make it possible to predict the dissolution rate (being informed by the particle size distribution) which, in its turn, can be used as a surrogate to predict the USP2 dissolution curve. Subsequently, validation can be done by using this profile as input for PBPK platforms.
    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
    Uridine 5\'-diphospho-glucuronosyltransferases (UGTs) are expressed in the small intestines, but prediction of first-pass extraction from the related metabolism is not well studied. This work assesses physiologically based pharmacokinetic (PBPK) modeling as a tool for predicting intestinal metabolism due to UGTs in the human gastrointestinal tract. Available data for intestinal UGT expression levels and in vitro approaches that can be used to predict intestinal metabolism of UGT substrates are reviewed. Human PBPK models for UGT substrates with varying extents of UGT-mediated intestinal metabolism (lorazepam, oxazepam, naloxone, zidovudine, cabotegravir, raltegravir, and dolutegravir) have demonstrated utility for predicting the extent of intestinal metabolism. Drug-drug interactions (DDIs) of UGT1A1 substrates dolutegravir and raltegravir with UGT1A1 inhibitor atazanavir have been simulated, and the role of intestinal metabolism in these clinical DDIs examined. Utility of an in silico tool for predicting substrate specificity for UGTs is discussed. Improved in vitro tools to study metabolism for UGT compounds, such as coculture models for low clearance compounds and better understanding of optimal conditions for in vitro studies, may provide an opportunity for improved in vitro-in vivo extrapolation (IVIVE) and prospective predictions. PBPK modeling shows promise as a useful tool for predicting intestinal metabolism for UGT substrates.
    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
    无定形固体分散体(ASD)剂型可以提高水溶性差的药物的口服生物利用度,实现新化学实体的商业化,并提高现有药物的疗效和患者依从性。然而,稳健的发展,高性能ASD剂型可能具有挑战性,通常需要多次配方迭代,很长的时间线,和高成本。在之前的研究中,阿卡拉布替尼/醋酸羟丙基甲基纤维素琥珀酸酯(HPMCAS)-H级ASD片剂被证明在比格犬中克服了市售Calquence的pH效应。这项研究描述了用于开发这些ASD片剂的流线型体外和计算机模拟方法。HPMCAS-H和-M级聚合物在初步筛选研究中提供了最长的acalabrutinib超饱和维持,和HPMCAS-H级ASD在胃至肠转移溶出试验中在升高的胃pH下提供了最高的体外曲线下面积(AUC)。在HPMCAS-HASD片剂和Calquence胶囊的计算机模拟中,使用自下而上的方法(AUC0-inf的绝对平均倍数误差<2,Calquence法莫替丁≈3除外)提供了良好的体内研究预测准确性。这种简化的方法结合了对关键药物的理解,聚合物,和胃肠道特性与体外和硅工具,以克服阿卡拉布替尼pH效应,而无需重新配制或多项研究,显示出减少开发ASD药物产品的时间和成本的希望。
    Amorphous solid dispersion (ASD) dosage forms can improve the oral bioavailability of poorly water-soluble drugs, enabling the commercialization of new chemical entities and improving the efficacy and patient compliance of existing drugs. However, the development of robust, high-performing ASD dosage forms can be challenging, often requiring multiple formulation iterations, long timelines, and high cost. In a previous study, acalabrutinib/hydroxypropyl methylcellulose acetate succinate (HPMCAS)-H grade ASD tablets were shown to overcome the pH effect of commercially marketed Calquence in beagle dogs. This study describes the streamlined in vitro and in silico approach used to develop those ASD tablets. HPMCAS-H and -M grade polymers provided the longest acalabrutinib supersaturation sustainment in an initial screening study, and HPMCAS-H grade ASDs provided the highest in vitro area under the curve (AUC) in gastric to intestinal transfer dissolution tests at elevated gastric pH. In silico simulations of the HPMCAS-H ASD tablet and Calquence capsule provided good in vivo study prediction accuracy using a bottom-up approach (absolute average fold error of AUC0-inf < 2 except for Calquence + famotidine ≈ 3). This streamlined approach combined an understanding of key drug, polymer, and gastrointestinal properties with in vitro and in silico tools to overcome the acalabrutinib pH effect without the need for reformulation or multiple studies, showing promise for reducing time and costs to develop ASD drug products.
    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
    The application of in silico modeling to predict the in vivo outcome of an oral drug product is gaining a lot of interest. Fully relying on these models as a surrogate tool requires continuous optimization and validation. To do so, intraluminal and systemic data are desirable to judge the predicted outcomes. The aim of this study was to predict the systemic concentrations of ibuprofen after oral administration of an 800 mg immediate-release (IR) tablet to healthy subjects in fasted-state conditions. A mechanistic oral absorption model coupled with a two-compartmental pharmacokinetic (PK) model was built in Phoenix WinNonlinWinNonlin® software and in the GastroPlus™ simulator. It should be noted that all simulations were performed in an ideal framework as we were in possession of a plethora of in vivo data (e.g., motility, pH, luminal and systemic concentrations) in order to evaluate and optimize these models. All this work refers to the fact that important, yet crucial, gastrointestinal (GI) variables should be integrated into biopredictive dissolution testing (low buffer capacity media, considering phosphate versus bicarbonate buffer, hydrodynamics) to account for a valuable input for physiologically-based pharmacokinetic (PBPK) platform programs. While simulations can be performed and mechanistic insights can be gained from such simulations from current software, we need to move from correlations to predictions (IVIVC → IVIVP) and, moreover, we need to further determine the dynamics of the GI variables controlling the dosage form transit, disintegration, dissolution, absorption and metabolism along the human GI tract. Establishing the link between biopredictive in vitro dissolution testing and mechanistic oral absorption modeling (i.e., physiologically-based biopharmaceutics modeling (PBBM)) creates an opportunity to potentially request biowaivers in the near future for orally administered drug products, regardless of its classification according to the Biopharmaceutics Classification System (BCS).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Recently published studies have proposed that amorphous drug nanoparticles in gastrointestinal fluids may be beneficial for the absorption of poorly soluble compounds. Nanosized drug particles are known to provide rapid dissolution rates and, in some instances, a slight increase in solubility. However, in recent studies, the differences observed in vivo could not be explained solely by these attributes. Given the high dose and very low aqueous solubility of the study compounds, rapid equilibration to the drug-saturated solubility in gastrointestinal fluid would occur independent of the presence of nanoparticles. Alternatively, it has been proposed that drug nanoparticles (ca. ≤ 200 to 300 nm) may provide a \"shuttle\" for drug across the unstirred water layer (UWL) adjacent to the intestinal epithelium, particularly for low solubility/lipophilic compounds where absorption may be largely UWL-limited. This transport mechanism would result in a higher unbound drug concentration at the surface of the epithelium for absorption. This study evaluates this mechanism using a simple modification of the effective permeability to account for the effect of drug nanoparticles diffusing across the UWL. The modification can be made using inputs for solubility and nanoparticle size. The permeability modification was evaluated using three published case studies for amorphous formulations of itraconazole, anacetrapib, and enzalutamide, where the formation of amorphous drug nanoparticles upon dissolution resulted in improved drug absorption. Absorption modeling was performed using GastroPlus to assess the impact of the nanomodified permeability method on the accuracy of model prediction compared to in vivo data. Simulation results were compared to those for baseline simulations using an unmodified effective permeability. The results show good agreement using the nanomodified permeability, which described the data better than the standard baseline predictions. The nanomodified permeability method can be a suitable, fit-for-purpose in silico approach for evaluating or predicting oral absorption of poorly soluble, UWL-limited drugs from formulations that produce a significant number of amorphous drug nanoparticles.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Topiroxostat is a selective xanthine oxidoreductase (XOR) inhibitor for the management of hyperuricemia in patients with or without gout. In this work, we aim to employ the physiologically based pharmacokinetic (PBPK) model with the drug-target residence time model to predict and characterize both the pharmacokinetics (PK) and pharmacodynamics (PD) of topiroxostat in humans. The plasma concentration-time profile of topiroxostat was simulated based on drug properties and human physiology parameters. The predictive power of this PBPK model was then demonstrated by comparison of stimulated to observed pharmacokinetic parameters. The utility of the model was further demonstrated through predicting the oral absorption and disposition characteristics of topiroxostat in humans. Finally, by combining the PBPK model and the drug-target residence time model, we successfully predicted the target occupancy and built the relationship between PK and PD using in vitro, in vivo and in silico information. The results showed that topiroxostat exhibited significant in vivo pharmacological activity even after the complete clearance of this drug from the liver (target site), which may be due to the long residence time of the binary topiroxostat-XOR complex. This work may be helpful to guide future investigations of topiroxostat and also provides a novel strategy for PK/PD studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号