留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development

Yurong Lai Xiaoyan Chu Li Di Wei Gao Yingying Guo Xingrong Liu Chuang Lu Jialin Mao Hong Shen Huaping Tang Cindy Q. Xia Lei Zhang Xinxin Ding

Yurong Lai, Xiaoyan Chu, Li Di, Wei Gao, Yingying Guo, Xingrong Liu, Chuang Lu, Jialin Mao, Hong Shen, Huaping Tang, Cindy Q. Xia, Lei Zhang, Xinxin Ding. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development[J]. 机械工程学报. doi: 10.1016/j.apsb.2022.03.009
引用本文: Yurong Lai, Xiaoyan Chu, Li Di, Wei Gao, Yingying Guo, Xingrong Liu, Chuang Lu, Jialin Mao, Hong Shen, Huaping Tang, Cindy Q. Xia, Lei Zhang, Xinxin Ding. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development[J]. 机械工程学报. doi: 10.1016/j.apsb.2022.03.009
Yurong Lai, Xiaoyan Chu, Li Di, Wei Gao, Yingying Guo, Xingrong Liu, Chuang Lu, Jialin Mao, Hong Shen, Huaping Tang, Cindy Q. Xia, Lei Zhang, Xinxin Ding. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development[J]. JOURNAL OF MECHANICAL ENGINEERING. doi: 10.1016/j.apsb.2022.03.009
Citation: Yurong Lai, Xiaoyan Chu, Li Di, Wei Gao, Yingying Guo, Xingrong Liu, Chuang Lu, Jialin Mao, Hong Shen, Huaping Tang, Cindy Q. Xia, Lei Zhang, Xinxin Ding. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development[J]. JOURNAL OF MECHANICAL ENGINEERING. doi: 10.1016/j.apsb.2022.03.009

Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development

doi: 10.1016/j.apsb.2022.03.009
基金项目: 

We would like to thank Dr. Ping Zhao and Dr. Donglu Zhang for providing scientific insights on this review article. Research in Xinxin Ding's laboratory was supported in part by grants from the National Institutes of Health (CA023074, CA092596, ES004940, ES006694, and ES020867, USA).

详细信息
    通讯作者:

    Yurong Lai,E-mail:Yurong.lai@gilead.com

    Xinxin Ding,E-mail:xding@pharmacy.arizona.edu

  • 中图分类号: https://www.sciencedirect.com/science/article/pii/S2211383522001083/pdf?md5=c46dcd9d5479b580a38bd281101c0b7f&pid=1-s2.0-S2211383522001083-main.pdf

Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development

Funds: 

We would like to thank Dr. Ping Zhao and Dr. Donglu Zhang for providing scientific insights on this review article. Research in Xinxin Ding's laboratory was supported in part by grants from the National Institutes of Health (CA023074, CA092596, ES004940, ES006694, and ES020867, USA).

  • 摘要: Drug metabolism and pharmacokinetics (DMPK) is an important branch of pharmaceutical sciences. The nature of ADME (absorption, distribution, metabolism, excretion) and PK (pharmacokinetics) inquiries during drug discovery and development has evolved in recent years from being largely descriptive to seeking a more quantitative and mechanistic understanding of the fate of drug candidates in biological systems. Tremendous progress has been made in the past decade, not only in the characterization of physiochemical properties of drugs that influence their ADME, target organ exposure, and toxicity, but also in the identification of design principles that can minimize drug-drug interaction (DDI) potentials and reduce the attritions. The importance of membrane transporters in drug disposition, efficacy, and safety, as well as the interplay with metabolic processes, has been increasingly recognized. Dramatic increases in investments on new modalities beyond traditional small and large molecule drugs, such as peptides, oligonucleotides, and antibody-drug conjugates, necessitated further innovations in bioanalytical and experimental tools for the characterization of their ADME properties. In this review, we highlight some of the most notable advances in the last decade, and provide future perspectives on potential major breakthroughs and innovations in the translation of DMPK science in various stages of drug discovery and development.

     

  • [1] Chung TDY, Terry DB, Smith LH. In vitro and in vivo assessment of ADME and PK properties during lead selection and lead optimization-guidelines, benchmarks and rules of thumb. In:Markossian S, Grossman A, Brimacombe K, Arkin M, Auld D, Austin CP, et al, editors. Assay guidance manual. Bethesda (MD):Eli Lilly & Company and the National Center for Advancing Translational Sciences; 2004-; 2015
    [2] Li Y, Meng Q, Yang M, Liu D, Hou X, Tang L, et al. Current trends in drug metabolism and pharmacokinetics. Acta Pharm Sin B 2019; 9:1113-1144
    [3] Wood FL, Houston JB, Hallifax D. Clearance prediction methodology needs fundamental improvement:trends common to rat and human hepatocytes/microsomes and implications for experimental methodology. Drug Metab Dispos 2017; 45:1178-1188
    [4] Edson KZ, Rettie AE. CYP4 enzymes as potential drug targets:focus on enzyme multiplicity, inducers and inhibitors, and therapeutic modulation of 20-hydroxyeicosatetraenoic acid (20-HETE) synthase and fatty acid omega-hydroxylase activities. Curr Top Med Chem 2013; 13:1429-1440
    [5] Saravanakumar A, Sadighi A, Ryu R, Akhlaghi F. Physicochemical properties, biotransformation, and transport pathways of established and newly approved medications:a systematic review of the top 200 most prescribed drugs vs. the FDA-approved drugs between 2005 and 2016. Clin Pharmacokinet 2019; 58:1281-1294
    [6] Xie F, Ding X, Zhang QY. An update on the role of intestinal cytochrome P450 enzymes in drug disposition. Acta Pharm Sin B 2016; 6:374-383
    [7] Li AP, Ho MD, Alam N, Mitchell W, Wong S, Yan Z, et al. Inter-individual and inter-regional variations in enteric drug metabolizing enzyme activities:results with cryopreserved human intestinal mucosal epithelia (CHIM) from the small intestines of 14 donors. Pharmacol Res Perspect 2020; 8:e00645
    [8] Zhang H, Wolford C, Basit A, Li AP, Fan PW, Murray BP, et al. Regional proteomic quantification of clinically relevant non-cytochrome P450 enzymes along the human small intestine. Drug Metab Dispos 2020; 48:528-536
    [9] Di L. The role of drug metabolizing enzymes in clearance. Expert Opin Drug Metab Toxicol 2014; 10:379-393
    [10] Foti RS, Dalvie DK. Cytochrome P450 and non-cytochrome P450 oxidative metabolism:contributions to the pharmacokinetics, safety, and efficacy of xenobiotics. Drug Metab Dispos 2016; 44:1229-1245
    [11] Yang G, Ge S, Singh R, Basu S, Shatzer K, Zen M, et al. Glucuronidation:driving factors and their impact on glucuronide disposition. Drug Metab Rev 2017; 49:105-138
    [12] Bohnert T, Patel A, Templeton I, Chen Y, Lu C, Lai G, et al. Evaluation of a new molecular entity as a victim of metabolic drug-drug interactions-an industry perspective. Drug Metab Dispos 2016; 44:1399-1423
    [13] Seo KA, Kim HJ, Jeong ES, Abdalla N, Choi CS, Kim DH, et al. In vitro assay of six UDP-glucuronosyltransferase isoforms in human liver microsomes, using cocktails of probe substrates and liquid chromatography-tandem mass spectrometry. Drug Metab Dispos 2014; 42:1803-1810
    [14] Miners JO, Rowland A, Novak JJ, Lapham K, Goosen TC. Evidence-based strategies for the characterisation of human drug and chemical glucuronidation in vitro and UDP-glucuronosyltransferase reaction phenotyping. Pharmacol Ther 2021; 218:107689
    [15] Furukawa T, Naritomi Y, Tetsuka K, Nakamori F, Moriguchi H, Yamano K, et al. Species differences in intestinal glucuronidation activities between humans, rats, dogs and monkeys. Xenobiotica 2014; 44:205-216
    [16] Kaivosaari S, Finel M, Koskinen M. N-Glucuronidation of drugs and other xenobiotics by human and animal UDP-glucuronosyltransferases. Xenobiotica 2011; 41:652-669
    [17] Docci L, Klammers F, Ekiciler A, Molitor B, Umehara K, Walter I, et al. In vitro to in vivo extrapolation of metabolic clearance for UGT substrates using short-term suspension and long-term co-cultured human hepatocytes. AAPS J 2020; 22:131
    [18] Mueller JW, Idkowiak J, Gesteira TF, Vallet C, Hardman R, van den Boom J, et al. Human DHEA sulfation requires direct interaction between PAPS synthase 2 and DHEA sulfotransferase SULT2A1. J Biol Chem 2018; 293:9724-9735
    [19] Riches Z, Stanley EL, Bloomer JC, Coughtrie MW. Quantitative evaluation of the expression and activity of five major sulfotransferases (SULTs) in human tissues:the SULT "pie". Drug Metab Dispos 2009; 37:2255-2261
    [20] Li AP, Hartman NR, Lu C, Collins JM, Strong JM. Effects of cytochrome P450 inducers on 17α-ethinyloestradiol (EE2) conjugation by primary human hepatocytes. Br J Clin Pharmacol 1999; 48:733-742
    [21] Di L. Reaction phenotyping to assess victim drug-drug interaction risks. Expert Opin Drug Discov 2017; 12:1105-1115
    [22] Dalvie D, Di L. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol Ther 2019; 201:137-180
    [23] Garattini E, Terao M. The role of aldehyde oxidase in drug metabolism. Expert Opin Drug Metab Toxicol 2012; 8:487-503
    [24] Coelho C, Mahro M, Trincao J, Carvalho AT, Ramos MJ, Terao M, et al. The first mammalian aldehyde oxidase crystal structure:insights into substrate specificity. J Biol Chem 2012; 287:40690-40702
    [25] Garattini E, Fratelli M, Terao M. Mammalian aldehyde oxidases:genetics, evolution and biochemistry. Cell Mol Life Sci 2008; 65:1019-1048
    [26] Zientek M, Jiang Y, Youdim K, Obach RS. In vitro-in vivo correlation for intrinsic clearance for drugs metabolized by human aldehyde oxidase. Drug Metab Dispos 2010; 38:1322-1327
    [27] Akabane T, Gerst N, Masters JN, Tamura K. A quantitative approach to hepatic clearance prediction of metabolism by aldehyde oxidase using custom pooled hepatocytes. Xenobiotica 2012; 42:863-871
    [28] Uehara S, Yoneda N, Higuchi Y, Yamazaki H, Suemizu H. Human aldehyde oxidase 1-mediated carbazeran oxidation in chimeric TK-NOG mice transplanted with human hepatocytes. Drug Metab Dispos 2020; 48:580-586
    [29] Di L. The impact of carboxylesterases in drug metabolism and pharmacokinetics. Curr Drug Metab 2019; 20:91-102
    [30] Hermant P, Bosc D, Piveteau C, Gealageas R, Lam B, Ronco C, et al. Controlling plasma stability of hydroxamic acids:a medchem toolbox. J Med Chem 2017;60:9067-9089
    [31] Ahmed F, Vyas V, Cornfield A, Goodin S, Ravikumar TS, Rubin EH, et al. In vitro activation of irinotecan to SN-38 by human liver and intestine. Anticancer Res 1999; 19:2067-2071
    [32] Shimizu M, Fukami T, Nakajima M, Yokoi T. Screening of specific inhibitors for human carboxylesterases or arylacetamide deacetylase. Drug Metab Dispos 2014; 42:1103-1109
    [33] Umehara K, Zollinger M, Kigondu E, Witschi M, Juif C, Huth F, et al. Esterase phenotyping in human liver in vitro:specificity of carboxylesterase inhibitors. Xenobiotica 2016; 46:862-867
    [34] Trapa PE, Beaumont K, Atkinson K, Eng H, King-Ahmad A, Scott DO, et al. In vitro-in vivo extrapolation of intestinal availability for carboxylesterase substrates using portal vein-cannulated monkey. J Pharm Sci 2017; 106:898-905
    [35] Rautio J, Meanwell NA, Di L, Hageman MJ. The expanding role of prodrugs in contemporary drug design and development. Nat Rev Drug Discov 2018; 17:559-587
    [36] Weber BW, Kimani SW, Varsani A, Cowan DA, Hunter R, Venter GA, et al. The mechanism of the amidases:mutating the glutamate adjacent to the catalytic triad inactivates the enzyme due to substrate mispositioning. J Biol Chem 2013; 288:28514-28523
    [37] Rawal J, Jones R, Payne A, Gardner I. Strategies to prevent N-acetyltransferase-mediated metabolism in a series of piperazine-containing pyrazalopyrimidine compounds. Xenobiotica 2008; 38:1219-1239
    [38] Sun Q, Harper TW, Dierks EA, Zhang L, Chang S, Rodrigues AD, et al. 1-Aminobenzotriazole, a known cytochrome P450 inhibitor, is a substrate and inhibitor of N-acetyltransferase. Drug Metab Dispos 2011; 39:1674-1679
    [39] Cordes H, Thiel C, Aschmann HE, Baier V, Blank LM, Kuepfer L. A physiologically based pharmacokinetic model of isoniazid and its application in individualizing tuberculosis chemotherapy. Antimicrob Agents Chemother 2016; 60:6134-6145
    [40] Kalgutkar AS, Castagnoli N, Jr. Selective inhibitors of monoamine oxidase (MAO-A and MAO-B) as probes of its catalytic site and mechanism. Med Res Rev 1995; 15:325-388
    [41] Masuo Y, Nagamori S, Hasegawa A, Hayashi K, Isozumi N, Nakamichi N, et al. Utilization of liver microsomes to estimate hepatic intrinsic clearance of monoamine oxidase substrate drugs in humans. Pharm Res 2017; 34:1233-1243
    [42] Harada R, Hayakawa Y, Ezura M, Lerdsirisuk P, Du Y, Ishikawa Y, et al. 18F-SMBT-1:a selective and reversible PET tracer for monoamine oxidase-B imaging. J Nucl Med 2021; 62:253-258
    [43] Cashman JR, Zhang J. Human flavin-containing monooxygenases. Annu Rev Pharmacol Toxicol 2006; 46:65-100
    [44] Jones BC, Srivastava A, Colclough N, Wilson J, Reddy VP, Amberntsson S, et al. An investigation into the prediction of in vivo clearance for a range of flavin-containing monooxygenase substrates. Drug Metab Dispos 2017; 45:1060-1067
    [45] Leung L, Yang X, Strelevitz TJ, Montgomery J, Brown MF, Zientek MA, et al. Clearance prediction of targeted covalent inhibitors by in vitro-in vivo extrapolation of hepatic and extrahepatic clearance mechanisms. Drug Metab Dispos 2017; 45:1-7
    [46] Rudewicz PJ, Yue Q, Shin Y. High-throughput strategies for metabolite identification in drug discovery. Boca Raton:CRC Press; 2009
    [47] Di L, Kerns EH. Drug-like properties:concepts, structure design and methods:from ADME to toxicity optimization. 2 edition. Boston:Elsevier/AP; 2016
    [48] Di L, Obach RS. Addressing the challenges of low clearance in drug research. AAPS J 2015; 17:352-357
    [49] Di L, Trapa P, Obach RS, Atkinson K, Bi YA, Wolford AC, et al. A novel relay method for determining low-clearance values. Drug Metab Dispos 2012; 40:1860-1865
    [50] Di L, Atkinson K, Orozco CC, Funk C, Zhang H, McDonald TS, et al. In vitro-in vivo correlation for low-clearance compounds using hepatocyte relay method. Drug Metab Dispos 2013; 41:2018-2023
    [51] Yang X, Atkinson K, Di L. Novel cytochrome P450 reaction phenotyping for low-clearance compounds using the hepatocyte relay method. Drug Metab Dispos 2016; 44:460-465
    [52] Lin C, Shi J, Moore A, Khetani SR. Prediction of drug clearance and drug-drug interactions in microscale cultures of human hepatocytes. Drug Metab Dispos 2016; 44:127-136
    [53] Bonn B, Svanberg P, Janefeldt A, Hultman I, Grime K. Determination of human hepatocyte intrinsic clearance for slowly metabolized compounds:comparison of a primary hepatocyte/stromal cell co-culture with plated primary hepatocytes and HepaRG. Drug Metab Dispos 2016; 44:527-533
    [54] Hultman I, Vedin C, Abrahamsson A, Winiwarter S, Darnell M. Use of HmuREL human coculture system for prediction of intrinsic clearance and metabolite formation for slowly metabolized compounds. Mol Pharm 2016; 13:2796-2807
    [55] Kratochwil NA, Meille C, Fowler S, Klammers F, Ekiciler A, Molitor B, et al. Metabolic profiling of human long-term liver models and hepatic clearance predictions from in vitro data using nonlinear mixed-effects modeling. AAPS J 2017; 19:534-550
    [56] Novik EI, Dwyer J, Morelli JK, Parekh A, Cho C, Pludwinski E, et al. Long-enduring primary hepatocyte-based co-cultures improve prediction of hepatotoxicity. Toxicol Appl Pharmacol 2017; 336:20-30
    [57] Keefer C, Chang G, Carlo A, Novak JJ, Banker M, Carey J, et al. Mechanistic insights on clearance and inhibition discordance between liver microsomes and hepatocytes when clearance in liver microsomes is higher than in hepatocytes. Eur J Pharm Sci 2020; 155:105541
    [58] Di L, Kerns EH, Gao N, Li SQ, Huang Y, Bourassa JL, et al. Experimental design on single-time-point high-throughput microsomal stability assay. J Pharm Sci 2004; 93:1537-1544
    [59] Khetani SR, Bhatia SN. Microscale culture of human liver cells for drug development. Nat Biotechnol 2008;26:120-126
    [60] Chan TS, Yu H, Moore A, Khetani SR, Tweedie D. Meeting the challenge of predicting hepatic clearance of compounds slowly metabolized by cytochrome P450 using a novel hepatocyte model, HepatoPac. Drug Metab Dispos 2013; 41:2024-2032
    [61] Wang WW, Khetani SR, Krzyzewski S, Duignan DB, Obach RS. Assessment of a micropatterned hepatocyte coculture system to generate major human excretory and circulating drug metabolites. Drug Metab Dispos 2010; 38:1900-1905
    [62] Brown HS, Griffin M, Houston JB. Evaluation of cryopreserved human hepatocytes as an alternative in vitro system to microsomes for the prediction of metabolic clearance. Drug Metab Dispos 2007; 35:293-301
    [63] Muller J, Keiser M, Drozdzik M, Oswald S. Expression, regulation and function of intestinal drug transporters:an update. Biol Chem 2017; 398:175-192
    [64] Colas C, Ung PM, Schlessinger A. SLC transporters:structure, function, and drug discovery. Medchemcomm 2016; 7:1069-1081
    [65] Benadiba M, Maor Y. Importance of ABC transporters in drug development. Curr Pharm Des 2016; 22:5817-5829
    [66] International Transporter Consortium, Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, et al. Membrane transporters in drug development. Nat Rev Drug Discov 2010; 9:215-236
    [67] Jani M, Ambrus C, Magnan R, Jakab KT, Beery E, Zolnerciks JK, et al. Structure and function of BCRP, a broad specificity transporter of xenobiotics and endobiotics. Arch Toxicol 2014; 88:1205-1248
    [68] Lund M, Petersen TS, Dalhoff KP. Clinical implications of P-glycoprotein modulation in drug-drug interactions. Drugs 2017; 77:859-883
    [69] Konig J, Muller F, Fromm MF. Transporters and drug-drug interactions:important determinants of drug disposition and effects. Pharmacol Rev 2013; 65:944-966
    [70] Colas C, Schlessinger A, Pajor AM. Mapping functionally important residues in the Na+/dicarboxylate cotransporter, NaDC1. Biochemistry 2017; 56:4432-4441
    [71] Estudante M, Morais JG, Soveral G, Benet LZ. Intestinal drug transporters:an overview. Adv Drug Deliv Rev 2013; 65:1340-1356
    [72] Katsura T, Inui K. Intestinal absorption of drugs mediated by drug transporters:mechanisms and regulation. Drug Metab Pharmacokinet 2003; 18:1-15
    [73] Kruijtzer CM, Beijnen JH, Rosing H, ten Bokkel Huinink WW, Schot M, Jewell RC, et al. Increased oral bioavailability of topotecan in combination with the breast cancer resistance protein and P-glycoprotein inhibitor GF120918. J Clin Oncol 2002; 20:2943-2950
    [74] Greiner B, Eichelbaum M, Fritz P, Kreichgauer HP, von Richter O, Zundler J, et al. The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin. J Clin Invest 1999; 104:147-153
    [75] Zamek-Gliszczynski MJ, Patel M, Yang X, Lutz JD, Chu X, Brouwer KLR, et al. Intestinal P-gp and putative hepatic OATP1B induction:international transporter consortium perspective on drug development implications. Clin Pharmacol Ther 2021; 109:55-64
    [76] Mao Q, Lai Y, Wang J. Drug transporters in xenobiotic disposition and pharmacokinetic prediction. Drug Metab Dispos 2018; 46:561-566
    [77] Varma MV, El-Kattan AF. Transporter-enzyme interplay:deconvoluting effects of hepatic transporters and enzymes on drug disposition using static and dynamic mechanistic models. J Clin Pharmacol 2016; 56 Suppl 7:S99-S109
    [78] Yee SW, Giacomini MM, Shen H, Humphreys WG, Horng H, Brian W, et al. Organic anion transporter polypeptide 1B1 polymorphism modulates the extent of drug-drug interaction and associated biomarker levels in healthy volunteers. Clin Transl Sci 2019; 12:388-399
    [79] Mori D, Kashihara Y, Yoshikado T, Kimura M, Hirota T, Matsuki S, et al. Effect of OATP1B1 genotypes on plasma concentrations of endogenous OATP1B1 substrates and drugs, and their association in healthy volunteers. Drug Metab Pharmacokinet 2019; 34:78-86
    [80] Chu X, Chan GH, Evers R. Identification of endogenous biomarkers to predict the propensity of drug candidates to cause hepatic or renal transporter-mediated drug-drug interactions. J Pharm Sci 2017; 106:2357-2367
    [81] Chu X, Liao M, Shen H, Yoshida K, Zur AA, Arya V, et al. Clinical probes and endogenous biomarkers as substrates for transporter drug-drug interaction evaluation:perspectives from the international transporter consortium. Clin Pharmacol Ther 2018; 104:836-864
    [82] Paulusma CC, van Geer MA, Evers R, Heijn M, Ottenhoff R, Borst P, et al. Canalicular multispecific organic anion transporter/multidrug resistance protein 2 mediates low-affinity transport of reduced glutathione. Biochem J 1999; 338 (Pt 2):393-401
    [83] Keppler D, Arias IM. Hepatic canalicular membrane. Introduction:transport across the hepatocyte canalicular membrane. FASEB J 1997; 11:15-18
    [84] Ito K, Suzuki H, Hirohashi T, Kume K, Shimizu T, Sugiyama Y. Functional analysis of a canalicular multispecific organic anion transporter cloned from rat liver. J Biol Chem 1998; 273:1684-1688
    [85] Konig J, Nies AT, Cui Y, Leier I, Keppler D. Conjugate export pumps of the multidrug resistance protein (MRP) family:localization, substrate specificity, and MRP2-mediated drug resistance. Biochim Biophys Acta 1999; 1461:377-394
    [86] Kusuhara H, Sugiyama Y. Role of transporters in the tissue-selective distribution and elimination of drugs:transporters in the liver, small intestine, brain and kidney. J Control Release 2002; 78:43-54
    [87] Morgan RE, Trauner M, van Staden CJ, Lee PH, Ramachandran B, Eschenberg M, et al. Interference with bile salt export pump function is a susceptibility factor for human liver injury in drug development. Toxicol Sci 2010; 118:485-500
    [88] Hallifax D, Rawden HC, Hakooz N, Houston JB. Prediction of metabolic clearance using cryopreserved human hepatocytes:kinetic characteristics for five benzodiazepines. Drug Metab Dispos 2005; 33:1852-1858
    [89] Davies M, Jones RDO, Grime K, Jansson-Lofmark R, Fretland AJ, Winiwarter S, et al. Improving the accuracy of predicted human pharmacokinetics:lessons learned from the AstraZeneca drug pipeline over two decades. Trends Pharmacol Sci 2020; 41:390-408
    [90] Naritomi Y, Terashita S, Kimura S, Suzuki A, Kagayama A, Sugiyama Y. Prediction of human hepatic clearance from in vivo animal experiments and in vitro metabolic studies with liver microsomes from animals and humans. Drug Metab Dispos 2001; 29:1316-1324
    [91] Patilea-Vrana G, Unadkat JD. Transport vs. metabolism:what determines the pharmacokinetics and pharmacodynamics of drugs? Insights from the extended clearance model. Clin Pharmacol Ther 2016; 100:413-418
    [92] El-Kattan AF, Varma MVS. Navigating transporter sciences in pharmacokinetics characterization using the extended clearance classification system. Drug Metab Dispos 2018; 46:729-739
    [93] Watanabe T, Kusuhara H, Maeda K, Shitara Y, Sugiyama Y. Physiologically based pharmacokinetic modeling to predict transporter-mediated clearance and distribution of pravastatin in humans. J Pharmacol Exp Ther 2009; 328:652-662
    [94] Liang X, Lai Y. Overcoming the shortcomings of the extended-clearance concept:a framework for developing a physiologically-based pharmacokinetic (PBPK) model to select drug candidates involving transporter-mediated clearance. Expert Opin Drug Metab Toxicol 2021; 17:869-886
    [95] Lee W, Kim RB. Transporters and renal drug elimination. Annu Rev Pharmacol Toxicol 2004; 44:137-166
    [96] Wright SH, Dantzler WH. Molecular and cellular physiology of renal organic cation and anion transport. Physiol Rev 2004; 84:987-1049
    [97] Liu HC, Goldenberg A, Chen Y, Lun C, Wu W, Bush KT, et al. Molecular properties of drugs interacting with SLC22 transporters OAT1, OAT3, OCT1, and OCT2:a machine-learning approach. J Pharmacol Exp Ther 2016; 359:215-229
    [98] Zamek-Gliszczynski MJ, Giacomini KM, Zhang L. Emerging clinical importance of hepatic organic cation transporter 1 (OCT1) in drug pharmacokinetics, dynamics, pharmacogenetic variability, and drug interactions. Clin Pharmacol Ther 2018; 103:758-760
    [99] Roth M, Obaidat A, Hagenbuch B. OATPs, OATs and OCTs:the organic anion and cation transporters of the SLCO and SLC22A gene superfamilies. Br J Pharmacol 2012; 165:1260-1287
    [100] Nies AT, Koepsell H, Damme K, Schwab M. Organic cation transporters (OCTs, MATEs), in vitro and in vivo evidence for the importance in drug therapy. Handb Exp Pharmacol 2011:105-167
    [101] Chen L, Shu Y, Liang X, Chen EC, Yee SW, Zur AA, et al. OCT1 is a high-capacity thiamine transporter that regulates hepatic steatosis and is a target of metformin. Proc Natl Acad Sci U S A 2014; 111:9983-9988
    [102] Yin J, Wang J. Renal drug transporters and their significance in drug-drug interactions. Acta Pharm Sin B 2016; 6:363-373
    [103] Zhang S, Lovejoy KS, Shima JE, Lagpacan LL, Shu Y, Lapuk A, et al. Organic cation transporters are determinants of oxaliplatin cytotoxicity. Cancer Res 2006; 66:8847-8857
    [104] Yokoo S, Yonezawa A, Masuda S, Fukatsu A, Katsura T, Inui K. Differential contribution of organic cation transporters, OCT2 and MATE1, in platinum agent-induced nephrotoxicity. Biochem Pharmacol 2007; 74:477-487
    [105] Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, et al. Membrane transporters in drug development. Nat Rev Drug Discov 2010; 9:215-236
    [106] Lee WK, Wolff NA, Thevenod F. Organic cation transporters:physiology, toxicology and special focus on ethidium as a novel substrate. Curr Drug Metab 2009; 10:617-631
    [107] Hagos Y, Wolff NA. Assessment of the role of renal organic anion transporters in drug-induced nephrotoxicity. Toxins (Basel) 2010; 2:2055-2082
    [108] Ciarimboli G. Membrane transporters as mediators of cisplatin side-effects. Anticancer Res 2014; 34:547-550
    [109] Cundy KC, Petty BG, Flaherty J, Fisher PE, Polis MA, Wachsman M, et al. Clinical pharmacokinetics of cidofovir in human immunodeficiency virus-infected patients. Antimicrob Agents Chemother 1995; 39:1247-1252
    [110] Yonezawa A, Masuda S, Yokoo S, Katsura T, Inui K. Cisplatin and oxaliplatin, but not carboplatin and nedaplatin, are substrates for human organic cation transporters (SLC22A1-3 and multidrug and toxin extrusion family). J Pharmacol Exp Ther 2006; 319:879-886
    [111] Filipski KK, Loos WJ, Verweij J, Sparreboom A. Interaction of cisplatin with the human organic cation transporter 2. Clin Cancer Res 2008; 14:3875-3880
    [112] Li Q, Guo D, Dong Z, Zhang W, Zhang L, Huang SM, et al. Ondansetron can enhance cisplatin-induced nephrotoxicity via inhibition of multiple toxin and extrusion proteins (MATEs). Toxicol Appl Pharmacol 2013; 273:100-109
    [113] Shen H, Yang Z, Zhao W, Zhang Y, Rodrigues AD. Assessment of vandetanib as an inhibitor of various human renal transporters:inhibition of multidrug and toxin extrusion as a possible mechanism leading to decreased cisplatin and creatinine clearance. Drug Metab Dispos 2013; 41:2095-2103
    [114] Steinway SN, Saleh J, Koo BK, Delacour D, Kim DH. Human microphysiological models of intestinal tissue and gut microbiome. Front Bioeng Biotechnol 2020; 8:725
    [115] Calitz C, Hamman JH, Fey SJ, Wrzesinski K, Gouws C. Recent advances in three-dimensional cell culturing to assess liver function and dysfunction:from a drug biotransformation and toxicity perspective. Toxicol Mech Methods 2018; 28:369-385
    [116] Fang Y, Eglen RM. Three-dimensional cell cultures in drug discovery and development. SLAS Discov 2017; 22:456-472
    [117] Kulthong K, Hooiveld G, Duivenvoorde L, Miro Estruch I, Marin V, van der Zande M, et al. Transcriptome comparisons of in vitro intestinal epithelia grown under static and microfluidic gut-on-chip conditions with in vivo human epithelia. Sci Rep 2021; 11:3234
    [118] Santbergen MJC, van der Zande M, Gerssen A, Bouwmeester H, Nielen MWF. Dynamic in vitro intestinal barrier model coupled to chip-based liquid chromatography mass spectrometry for oral bioavailability studies. Anal Bioanal Chem 2020; 412:1111-1122
    [119] Guo Y, Li Z, Su W, Wang L, Zhu Y, Qin J. A biomimetic human gut-on-a-chip for modeling drug metabolism in intestine. Artif Organs 2018; 42:1196-1205
    [120] Kasendra M, Tovaglieri A, Sontheimer-Phelps A, Jalili-Firoozinezhad S, Bein A, Chalkiadaki A, et al. Development of a primary human small intestine-on-a-chip using biopsy-derived organoids. Sci Rep 2018; 8:2871
    [121] Kasendra M, Luc R, Yin J, Manatakis DV, Kulkarni G, Lucchesi C, et al. Duodenum intestine-chip for preclinical drug assessment in a human relevant model. Elife 2020; 9:1-23
    [122] Niu C, Smith B, Lai Y. Transporter gene regulation in sandwich cultured human hepatocytes through the activation of constitutive androstane receptor (CAR) or aryl hydrocarbon receptor (AhR). Front Pharmacol 2020; 11:620197
    [123] Bi YA, Kimoto E, Sevidal S, Jones HM, Barton HA, Kempshall S, et al. In vitro evaluation of hepatic transporter-mediated clinical drug-drug interactions:hepatocyte model optimization and retrospective investigation. Drug Metab Dispos 2012; 40:1085-1092
    [124] Wang H, Brown PC, Chow ECY, Ewart L, Ferguson SS, Fitzpatrick S, et al. 3D cell culture models:drug pharmacokinetics, safety assessment, and regulatory consideration. Clin Transl Sci 2021; 14:1659-1680
    [125] Baert Y, Ruetschle I, Cools W, Oehme A, Lorenz A, Marx U, et al. A multi-organ-chip co-culture of liver and testis equivalents:a first step toward a systemic male reprotoxicity model. Hum Reprod 2020; 35:1029-1044
    [126] Li J, Zhu HJ. Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based proteomics of drug-metabolizing enzymes and transporters. Molecules 2020; 25:2718
    [127] Couto N, Al-Majdoub ZM, Achour B, Wright PC, Rostami-Hodjegan A, Barber J. Quantification of proteins involved in drug metabolism and disposition in the human liver using label-free global proteomics. Mol Pharm 2019; 16:632-647
    [128] Khatri R, Fallon JK, Rementer RJB, Kulick NT, Lee CR, Smith PC. Targeted quantitative proteomic analysis of drug metabolizing enzymes and transporters by nano LC-MS/MS in the sandwich cultured human hepatocyte model. J Pharmacol Toxicol Methods 2019; 98:106590
    [129] Nakamura K, Hirayama-Kurogi M, Ito S, Kuno T, Yoneyama T, Obuchi W, et al. Large-scale multiplex absolute protein quantification of drug-metabolizing enzymes and transporters in human intestine, liver, and kidney microsomes by SWATH-MS:comparison with MRM/SRM and HR-MRM/PRM. Proteomics 2016; 16:2106-2117
    [130] Ladumor MK, Thakur A, Sharma S, Rachapally A, Mishra S, Bobe P, et al. A repository of protein abundance data of drug metabolizing enzymes and transporters for applications in physiologically based pharmacokinetic (PBPK) modelling and simulation. Sci Rep 2019; 9:9709
    [131] Bhatt DK, Prasad B. Critical issues and optimized practices in quantification of protein abundance level to determine interindividual variability in DMET proteins by LC-MS/MS proteomics. Clin Pharmacol Ther 2018; 103:619-630
    [132] Couto N, Al-Majdoub ZM, Gibson S, Davies PJ, Achour B, Harwood MD, et al. Quantitative proteomics of clinically relevant drug-metabolizing enzymes and drug transporters and their intercorrelations in the human small intestine. Drug Metab Dispos 2020; 48:245-268
    [133] Ohtsuki S, Uchida Y, Kubo Y, Terasaki T. Quantitative targeted absolute proteomics-based ADME research as a new path to drug discovery and development:methodology, advantages, strategy, and prospects. J Pharm Sci 2011; 100:3547-3559
    [134] Prasad B, Unadkat JD. Optimized approaches for quantification of drug transporters in tissues and cells by MRM proteomics. AAPS J 2014; 16:634-648
    [135] Xu M, Saxena N, Vrana M, Zhang H, Kumar V, Billington S, et al. Targeted LC-MS/MS proteomics-based strategy to characterize in vitro models used in drug metabolism and transport studies. Anal Chem 2018; 90:11873-11882
    [136] Uchida Y, Zhang Z, Tachikawa M, Terasaki T. Quantitative targeted absolute proteomics of rat blood-cerebrospinal fluid barrier transporters:comparison with a human specimen. J Neurochem 2015; 134:1104-1115
    [137] Prasad B, Evers R, Gupta A, Hop CE, Salphati L, Shukla S, et al. Interindividual variability in hepatic organic anion-transporting polypeptides and P-glycoprotein (ABCB1) protein expression:quantification by liquid chromatography tandem mass spectroscopy and influence of genotype, age, and sex. Drug Metab Dispos 2014; 42:78-88
    [138] Al-Majdoub ZM, Al Feteisi H, Achour B, Warwood S, Neuhoff S, Rostami-Hodjegan A, et al. Proteomic quantification of human blood-brain barrier SLC and ABC transporters in healthy individuals and dementia patients. Mol Pharm 2019; 16:1220-1233
    [139] Billington S, Salphati L, Hop C, Chu X, Evers R, Burdette D, et al. Interindividual and regional variability in drug transporter abundance at the human blood-brain barrier measured by quantitative targeted proteomics. Clin Pharmacol Ther 2019; 106:228-237
    [140] Al-Majdoub ZM, Achour B, Couto N, Howard M, Elmorsi Y, Scotcher D, et al. Mass spectrometry-based abundance atlas of ABC transporters in human liver, gut, kidney, brain and skin. FEBS Lett 2020; 594:4134-4150
    [141] Uchida Y, Ohtsuki S, Katsukura Y, Ikeda C, Suzuki T, Kamiie J, et al. Quantitative targeted absolute proteomics of human blood-brain barrier transporters and receptors. J Neurochem 2011; 117:333-345
    [142] Prasad B, Johnson K, Billington S, Lee C, Chung GW, Brown CD, et al. Abundance of drug transporters in the human kidney cortex as quantified by quantitative targeted proteomics. Drug Metab Dispos 2016; 44:1920-1924
    [143] Wang L, Prasad B, Salphati L, Chu X, Gupta A, Hop CE, et al. Interspecies variability in expression of hepatobiliary transporters across human, dog, monkey, and rat as determined by quantitative proteomics. Drug Metab Dispos 2015; 43:367-374
    [144] Li CY, Hosey-Cojocari C, Basit A, Unadkat JD, Leeder JS, Prasad B. Optimized renal transporter quantification by using aquaporin 1 and aquaporin 2 as anatomical markers:application in characterizing the ontogeny of renal transporters and its correlation with hepatic transporters in paired human samples. AAPS J 2019; 21:88
    [145] Bhatt DK, Mehrotra A, Gaedigk A, Chapa R, Basit A, Zhang H, et al. Age- and genotype-dependent variability in the protein abundance and activity of six major uridine diphosphate-glucuronosyltransferases in human liver. Clin Pharmacol Ther 2019; 105:131-141
    [146] Billington S, Ray AS, Salphati L, Xiao G, Chu X, Humphreys WG, et al. Transporter expression in noncancerous and cancerous liver tissue from donors with hepatocellular carcinoma and chronic hepatitis C infection quantified by LC-MS/MS proteomics. Drug Metab Dispos 2018; 46:189-196
    [147] Prasad B, Gaedigk A, Vrana M, Gaedigk R, Leeder JS, Salphati L, et al. Ontogeny of hepatic drug transporters as quantified by LC-MS/MS proteomics. Clin Pharmacol Ther 2016; 100:362-370
    [148] El-Khateeb E, Vasilogianni AM, Alrubia S, Al-Majdoub ZM, Couto N, Howard M, et al. Quantitative mass spectrometry-based proteomics in the era of model-informed drug development:applications in translational pharmacology and recommendations for best practice. Pharmacol Ther 2019; 203:107397
    [149] Al Feteisi H, Achour B, Rostami-Hodjegan A, Barber J. Translational value of liquid chromatography coupled with tandem mass spectrometry-based quantitative proteomics for in vitro-in vivo extrapolation of drug metabolism and transport and considerations in selecting appropriate techniques. Expert Opin Drug Metab Toxicol 2015; 11:1357-1369
    [150] Kumar AR, Prasad B, Bhatt DK, Mathialagan S, Varma MVS, Unadkat JD. In vivo-to-in vitro extrapolation of transporter-mediated renal clearance:relative expression factor versus relative activity factor approach. Drug Metab Dispos 2020; 49:470-478
    [151] Kumar V, Yin M, Ishida K, Salphati L, Hop C, Rowbottom C, et al. Prediction of transporter-mediated rosuvastatin hepatic uptake clearance and drug interaction in humans using proteomics-informed REF approach. Drug Metab Dispos 2021; 49:159-168
    [152] Sachar M, Kumar V, Gormsen LC, Munk OL, Unadkat JD. Successful prediction of positron emission tomography-imaged metformin hepatic uptake clearance in humans using the quantitative proteomics-informed relative expression factor approach. Drug Metab Dispos 2020; 48:1210-1216
    [153] Wegler C, Prieto Garcia L, Klinting S, Robertsen I, Wisniewski JR, Hjelmesaeth J, et al. Proteomics-informed prediction of rosuvastatin plasma profiles in patients with a wide range of body weight. Clin Pharmacol Ther 2021; 109:762-771
    [154] Ishida K, Ullah M, Toth B, Juhasz V, Unadkat JD. Successful prediction of in vivo hepatobiliary clearances and hepatic concentrations of rosuvastatin using sandwich-cultured rat hepatocytes, transporter-expressing cell lines, and quantitative proteomics. Drug Metab Dispos 2018; 46:66-74
    [155] Vildhede A, Karlgren M, Svedberg EK, Wisniewski JR, Lai Y, Noren A, et al. Hepatic uptake of atorvastatin:influence of variability in transporter expression on uptake clearance and drug-drug interactions. Drug Metab Dispos 2014; 42:1210-1218
    [156] Wegler C, Gaugaz FZ, Andersson TB, Wisniewski JR, Busch D, Groer C, et al. Variability in mass spectrometry-based quantification of clinically relevant drug transporters and drug metabolizing enzymes. Mol Pharm 2017; 14:3142-3151
    [157] Prasad B, Achour B, Artursson P, Hop C, Lai Y, Smith PC, et al. Toward a consensus on applying quantitative liquid chromatography-tandem mass spectrometry proteomics in translational pharmacology research:a white paper. Clin Pharmacol Ther 2019; 106:525-543
    [158] Sampson KE, Brinker A, Pratt J, Venkatraman N, Xiao Y, Blasberg J, et al. Zinc finger nuclease-mediated gene knockout results in loss of transport activity for P-glycoprotein, BCRP, and MRP2 in Caco-2 cells. Drug Metab Dispos 2015; 43:199-207
    [159] Gartzke D, Delzer J, Laplanche L, Uchida Y, Hoshi Y, Tachikawa M, et al. Genomic knockout of endogenous canine P-glycoprotein in wild-type, human P-glycoprotein and human BCRP transfected MDCKII cell lines by zinc finger nucleases. Pharm Res 2015; 32:2060-2071
    [160] Cheng Y, Chen S, Freeden C, Chen W, Zhang Y, Abraham P, et al. Bile salt homeostasis in normal and Bsep gene knockout rats with single and repeated doses of troglitazone. J Pharmacol Exp Ther 2017; 362:385-394
    [161] Lu J, Liu J, Guo Y, Zhang Y, Xu Y, Wang X. CRISPR-Cas9:a method for establishing rat models of drug metabolism and pharmacokinetics. Acta Pharm Sin B 2021; 11:2973-2982
    [162] Karlgren M, Simoff I, Keiser M, Oswald S, Artursson P. CRISPR-Cas9:a new addition to the drug metabolism and disposition tool box. Drug Metab Dispos 2018; 46:1776-1786
    [163] Yoshimi K, Kunihiro Y, Kaneko T, Nagahora H, Voigt B, Mashimo T. ssODN-mediated knock-in with CRISPR-Cas for large genomic regions in zygotes. Nat Commun 2016; 7:10431
    [164] Ma X, Shang X, Qin X, Lu J, Liu M, Wang X. Characterization of organic anion transporting polypeptide 1b2 knockout rats generated by CRISPR/Cas9:a novel model for drug transport and hyperbilirubinemia disease. Acta Pharm Sin B 2020; 10:850-860
    [165] Chen EC, Broccatelli F, Plise E, Chen B, Liu L, Cheong J, et al. Evaluating the utility of canine Mdr1 knockout Madin-Darby canine kidney I cells in permeability screening and efflux substrate determination. Mol Pharm 2018; 15:5103-5113
    [166] Vaidyanathan J, Yoshida K, Arya V, Zhang L. Comparing various in vitro prediction criteria to assess the potential of a new molecular entity to inhibit organic anion transporting polypeptide 1B1. J Clin Pharmacol 2016; 56 Suppl 7:S59-72
    [167] Mori D, Ishida H, Mizuno T, Kusumoto S, Kondo Y, Izumi S, et al. Alteration in the plasma concentrations of endogenous organic anion-transporting polypeptide 1B biomarkers in patients with non-small cell lung cancer treated with paclitaxel. Drug Metab Dispos 2020; 48:387-394
    [168] Tatosian DA, Yee KL, Zhang Z, Mostoller K, Paul E, Sutradhar S, et al. A microdose cocktail to evaluate drug interactions in patients with renal impairment. Clin Pharmacol Ther 2021; 109:403-415
    [169] Mori D, Kimoto E, Rago B, Kondo Y, King-Ahmad A, Ramanathan R, et al. Dose-dependent inhibition of OATP1B by rifampicin in healthy volunteers:comprehensive evaluation of candidate biomarkers and OATP1B probe drugs. Clin Pharmacol Ther 2020; 107:1004-1013
    [170] Cheung KWK, Yoshida K, Cheeti S, Chen B, Morley R, Chan IT, et al. GDC-0810 pharmacokinetics and transporter-mediated drug interaction evaluation with an endogenous biomarker in the first-in-human, dose escalation study. Drug Metab Dispos 2019; 47:966-973
    [171] Yoshida K, Guo C, Sane R. Quantitative prediction of OATP-mediated drug-drug interactions with model-based analysis of endogenous biomarker kinetics. CPT Pharmacometrics Syst Pharmacol 2018; 7:517-524
    [172] Muller F, Sharma A, Konig J, Fromm MF. Biomarkers for in vivo assessment of transporter function. Pharmacol Rev 2018; 70:246-277
    [173] Rodrigues AD, Taskar KS, Kusuhara H, Sugiyama Y. Endogenous probes for drug transporters:balancing vision with reality. Clin Pharmacol Ther 2018; 103:434-448
    [174] Mochizuki T, Mizuno T, Maeda K, Kusuhara H. Current progress in identifying endogenous biomarker candidates for drug transporter phenotyping and their potential application to drug development. Drug Metab Pharmacokinet 2021; 37:100358
    [175] Lai Y, Mandlekar S, Shen H, Holenarsipur VK, Langish R, Rajanna P, et al. Coproporphyrins in plasma and urine can be appropriate clinical biomarkers to recapitulate drug-drug interactions mediated by organic anion transporting polypeptide inhibition. J Pharmacol Exp Ther 2016; 358:397-404
    [176] Shen H, Christopher L, Lai Y, Gong J, Kandoussi H, Garonzik S, et al. Further studies to support the use of coproporphyrin I and III as Novel clinical biomarkers for evaluating the potential for organic anion transporting polypeptide 1B1 and OATP1B3 inhibition. Drug Metab Dispos 2018; 46:1075-1082
    [177] Barnett S, Ogungbenro K, Menochet K, Shen H, Lai Y, Humphreys WG, et al. Gaining mechanistic insight into coproporphyrin I as endogenous biomarker for OATP1B-mediated drug-drug interactions using population pharmacokinetic modeling and simulation. Clin Pharmacol Ther 2018; 104:564-574
    [178] Neuvonen M, Hirvensalo P, Tornio A, Rago B, West M, Lazzaro S, et al. Identification of glycochenodeoxycholate 3-O-glucuronide and glycodeoxycholate 3-O-glucuronide as highly sensitive and specific OATP1B1 biomarkers. Clin Pharmacol Ther 2021; 109:646-657
    [179] Suzuki Y, Sasamoto Y, Koyama T, Yoshijima C, Nakatochi M, Kubo M, et al. Substantially increased plasma coproporphyrin-I concentrations associated with OATP1B1~*15 allele in japanese general population. Clin Transl Sci 2021; 14:382-388
    [180] Chu X, Bleasby K, Chan GH, Nunes I, Evers R. The complexities of interpreting reversible elevated serum creatinine levels in drug development:does a correlation with inhibition of renal transporters exist?. Drug Metab Dispos 2016; 44:1498-1509
    [181] Tsuruya Y, Kato K, Sano Y, Imamura Y, Maeda K, Kumagai Y, et al. Investigation of endogenous compounds applicable to drug-drug interaction studies involving the renal organic anion transporters, OAT1 and OAT3, in humans. Drug Metab Dispos 2016; 44:1925-1933
    [182] Shen H, Nelson DM, Oliveira RV, Zhang Y, McNaney CA, Gu X, et al. Discovery and validation of pyridoxic acid and homovanillic acid as novel endogenous plasma biomarkers of organic anion transporter (OAT) 1 and OAT3 in cynomolgus monkeys. Drug Metab Dispos 2018; 46:178-188
    [183] Shen H, Holenarsipur VK, Mariappan TT, Drexler DM, Cantone JL, Rajanna P, et al. Evidence for the validity of pyridoxic acid (PDA) as a plasma-based endogenous probe for OAT1 and OAT3 function in healthy subjects. J Pharmacol Exp Ther 2019; 368:136-145
    [184] Willemin ME, Van Der Made TK, Pijpers I, Dillen L, Kunze A, Jonkers S, et al. Clinical investigation on endogenous biomarkers to predict strong OAT-mediated drug-drug interactions. Clin Pharmacokinet 2021; 60:1187-1199
    [185] Ahmad A, Ogungbenro K, Kunze A, Jacobs F, Snoeys J, Rostami-Hodjegan A, et al. Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters. CPT Pharmacometrics Syst Pharmacol 2021; 10:467-477
    [186] Diczfalusy U, Miura J, Roh HK, Mirghani RA, Sayi J, Larsson H, et al. 4β-Hydroxycholesterol is a new endogenous CYP3A marker:relationship to CYP3A5 genotype, quinine 3-hydroxylation and sex in Koreans, Swedes and Tanzanians. Pharmacogenet Genomics 2008; 18:201-208
    [187] Bodin K, Andersson U, Rystedt E, Ellis E, Norlin M, Pikuleva I, et al. Metabolism of 4β-hydroxycholesterol in humans. J Biol Chem 2002; 277:31534-31540
    [188] Kasichayanula S, Boulton DW, Luo WL, Rodrigues AD, Yang Z, Goodenough A, et al. Validation of 4β-hydroxycholesterol and evaluation of other endogenous biomarkers for the assessment of CYP3A activity in healthy subjects. Br J Clin Pharmacol 2014; 78:1122-1134
    [189] Suzuki Y, Itoh H, Fujioka T, Sato F, Kawasaki K, Sato Y, et al. Association of plasma concentration of 4β-hydroxycholesterol with CYP3A5 polymorphism and plasma concentration of indoxyl sulfate in stable kidney transplant recipients. Drug Metab Dispos 2014; 42:105-110
    [190] Suzuki Y, Itoh H, Sato F, Kawasaki K, Sato Y, Fujioka T, et al. Significant increase in plasma 4β-hydroxycholesterol concentration in patients after kidney transplantation. J Lipid Res 2013; 54:2568-2572
    [191] Bjorkhem-Bergman L, Backstrom T, Nylen H, Ronquist-Nii Y, Bredberg E, Andersson TB, et al. Comparison of endogenous 4β-hydroxycholesterol with midazolam as markers for CYP3A4 induction by rifampicin. Drug Metab Dispos 2013; 41:1488-1493
    [192] Shin KH, Choi MH, Lim KS, Yu KS, Jang IJ, Cho JY. Evaluation of endogenous metabolic markers of hepatic CYP3A activity using metabolic profiling and midazolam clearance. Clin Pharmacol Ther 2013; 94:601-609
    [193] Mao J, Martin I, McLeod J, Nolan G, van Horn R, Vourvahis M, et al. Perspective:4β-hydroxycholesterol as an emerging endogenous biomarker of hepatic CYP3A. Drug Metab Rev 2017; 49:18-34
    [194] Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism:regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther 2013; 138:103-141
    [195] Tornio A, Backman JT. Cytochrome P450 in pharmacogenetics:an update. Adv Pharmacol 2018; 83:3-32
    [196] Stingl JC, Bartels H, Viviani R, Lehmann ML, Brockmoller J. Relevance of UDP-glucuronosyltransferase polymorphisms for drug dosing:a quantitative systematic review. Pharmacol Ther 2014; 141:92-116
    [197] Merali Z, Ross S, Pare G. The pharmacogenetics of carboxylesterases:CES1 and CES2 genetic variants and their clinical effect. Drug Metabol Drug Interact 2014; 29:143-151
    [198] Niemi M, Pasanen MK, Neuvonen PJ. Organic anion transporting polypeptide 1B1:a genetically polymorphic transporter of major importance for hepatic drug uptake. Pharmacol Rev 2011; 63:157-181
    [199] Hira D, Terada T. BCRP/ABCG2 and high-alert medications:biochemical, pharmacokinetic, pharmacogenetic, and clinical implications. Biochem Pharmacol 2018; 147:201-210
    [200] FDA. Clinical pharmacogenomics:premarket evaluation in early-phase clinical studies and recommendations for labeling. Published[January 2013]; Accessed[February, 2022]. Available from:https://www.fda.gov/files/drugs/published/Clinical-Pharmacogenomics-Premarket-Evaluation-in-Early-Phase-Clinical-Studies-and-Recommendations-for-Labeling.pdf
    [201] EMA/CHMP. Guideline on the use of pharmacogenetic methodologies in the pharmacokinetic evaluation of medicinal products. Published[January 19, 2011]. Updated[August 1, 2012]. Accessed[February, 2022]. Available from:https://www.ema.europa.eu/documents/scientific-guideline/guideline-use-pharmacogenetic-methodologies-pharmacokinetic-evaluation-medicinal-products_en.pdf
    [202] Brian W, Tremaine LM, Arefayene M, de Kanter R, Evers R, Guo Y, et al. Assessment of drug metabolism enzyme and transporter pharmacogenetics in drug discovery and early development:perspectives of the I-PWG. Pharmacogenomics 2016; 17:615-631
    [203] Tremaine L, Brian W, DelMonte T, Francke S, Groenen P, Johnson K, et al. The role of ADME pharmacogenomics in early clinical trials:perspective of the Industry Pharmacogenomics Working Group (I-PWG). Pharmacogenomics 2015; 16:2055-2067
    [204] Giacomini KM, Balimane PV, Cho SK, Eadon M, Edeki T, Hillgren KM, et al. International transporter consortium commentary on clinically important transporter polymorphisms. Clin Pharmacol Ther 2013; 94:23-26
    [205] Guo Y, Lucksiri A, Dickinson GL, Vuppalanchi RK, Hilligoss JK, Hall SD. Quantitative prediction of CYP3A4- and CYP3A5-mediated drug interactions. Clin Pharmacol Ther 2020; 107:246-256
    [206] Tsamandouras N, Dickinson G, Guo Y, Hall S, Rostami-Hodjegan A, Galetin A, et al. Identification of the effect of multiple polymorphisms on the pharmacokinetics of simvastatin and simvastatin acid using a population-modeling approach. Clin Pharmacol Ther 2014; 96:90-100
    [207] Fowler S, Kletzl H, Finel M, Manevski N, Schmid P, Tuerck D, et al. A UGT2B10 splicing polymorphism common in african populations may greatly increase drug exposure. J Pharmacol Exp Ther 2015; 352:358-367
    [208] Morrissey KM, Stocker SL, Wittwer MB, Xu L, Giacomini KM. Renal transporters in drug development. Annu Rev Pharmacol Toxicol 2013; 53:503-529
    [209] Li Z, Litchfield J, Tess DA, Carlo AA, Eng H, Keefer C, et al. A physiologically based in silico tool to assess the risk of drug-related crystalluria. J Med Chem 2020; 63:6489-6498
    [210] Di L, Whitney-Pickett C, Umland JP, Zhang H, Zhang X, Gebhard DF, et al. Development of a new permeability assay using low-efflux MDCKII cells. J Pharm Sci 2011; 100:4974-4985
    [211] Zamek-Gliszczynski MJ, Taub ME, Chothe PP, Chu X, Giacomini KM, Kim RB, et al. Transporters in drug development:2018 ITC recommendations for transporters of emerging clinical importance. Clin Pharmacol Ther 2018; 104:890-899
    [212] Mathialagan S, Piotrowski MA, Tess DA, Feng B, Litchfield J, Varma MV. Quantitative prediction of human renal clearance and drug-drug interactions of organic anion transporter substrates using in vitro transport data:a relative activity factor approach. Drug Metab Dispos 2017; 45:409-417
    [213] Paine SW, Menochet K, Denton R, McGinnity DF, Riley RJ. Prediction of human renal clearance from preclinical species for a diverse set of drugs that exhibit both active secretion and net reabsorption. Drug Metab Dispos 2011; 39:1008-1013
    [214] Chen J, Yang H, Zhu L, Wu Z, Li W, Tang Y, et al. In silico prediction of human renal clearance of compounds using quantitative structure-pharmacokinetic relationship models. Chem Res Toxicol 2020; 33:640-650
    [215] Doddareddy MR, Cho YS, Koh HY, Kim DH, Pae AN. In silico renal clearance model using classical Volsurf approach. J Chem Inf Model 2006; 46:1312-1320
    [216] Kotal P, Van der Veere CN, Sinaasappel M, Elferink RO, Vitek L, Brodanova M, et al. Intestinal excretion of unconjugated bilirubin in man and rats with inherited unconjugated hyperbilirubinemia. Pediatr Res 1997; 42:195-200
    [217] van der Velde AE, Vrins CL, van den Oever K, Kunne C, Oude Elferink RP, Kuipers F, et al. Direct intestinal cholesterol secretion contributes significantly to total fecal neutral sterol excretion in mice. Gastroenterology 2007; 133:967-975
    [218] Mayer U, Wagenaar E, Beijnen JH, Smit JW, Meijer DK, van Asperen J, et al. Substantial excretion of digoxin via the intestinal mucosa and prevention of long-term digoxin accumulation in the brain by the mdr 1a P-glycoprotein. Br J Pharmacol 1996; 119:1038-1044
    [219] Sparreboom A, van Asperen J, Mayer U, Schinkel AH, Smit JW, Meijer DK, et al. Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sci U S A 1997; 94:2031-2035
    [220] Gramatte T, Oertel R. Intestinal secretion of intravenous talinolol is inhibited by luminal R-verapamil. Clin Pharmacol Ther 1999; 66:239-245
    [221] Zhang D, He K, Herbst JJ, Kolb J, Shou W, Wang L, et al. Characterization of efflux transporters involved in distribution and disposition of apixaban. Drug Metab Dispos 2013; 41:827-835
    [222] Zhang D, Wei C, Hop C, Wright MR, Hu M, Lai Y, et al. Intestinal excretion, intestinal recirculation, and renal tubule reabsorption are underappreciated mechanisms that drive the distribution and pharmacokinetic behavior of small molecule drugs. J Med Chem 2021; 64:7045-7059
    [223] Zhang D, Frost CE, He K, Rodrigues AD, Wang X, Wang L, et al. Investigating the enteroenteric recirculation of apixaban, a factor Xa inhibitor:administration of activated charcoal to bile duct-cannulated rats and dogs receiving an intravenous dose and use of drug transporter knockout rats. Drug Metab Dispos 2013; 41:906-915
    [224] Raghavan N, Frost CE, Yu Z, He K, Zhang H, Humphreys WG, et al. Apixaban metabolism and pharmacokinetics after oral administration to humans. Drug Metab Dispos 2009; 37:74-81
    [225] Stass H, Kubitza D, Moller JG, Delesen H. Influence of activated charcoal on the pharmacokinetics of moxifloxacin following intravenous and oral administration of a 400 mg single dose to healthy males. Br J Clin Pharmacol 2005; 59:536-541
    [226] Gramatte T, Oertel R, Terhaag B, Kirch W. Direct demonstration of small intestinal secretion and site-dependent absorption of the beta-blocker talinolol in humans. Clin Pharmacol Ther 1996; 59:541-549
    [227] Ramon J, Ben-Haim M, Shabtai M, Rubinstein E. Transepithelial intestinal excretion of ciprofloxacin in humans. Clin Infect Dis 2001; 32:822-823
    [228] Garner CE, Solon E, Lai CM, Lin J, Luo G, Jones K, et al. Role of P-glycoprotein and the intestine in the excretion of DPC 333[(2R)-2-{(3R)-3-amino-3-[4-(2-methylquinolin-4-ylmethoxy)phenyl]-2-oxopyrrolidin-1-yl}-N-hydroxy-4-methylpentanamide] in rodents. Drug Metab Dispos 2008; 36:1102-1110
    [229] Gnoth MJ, Buetehorn U, Muenster U, Schwarz T, Sandmann S. In vitro and in vivo P-glycoprotein transport characteristics of rivaroxaban. J Pharmacol Exp Ther 2011; 338:372-380
    [230] Wang X, Mondal S, Wang J, Tirucherai G, Zhang D, Boyd RA, et al. Effect of activated charcoal on apixaban pharmacokinetics in healthy subjects. Am J Cardiovasc Drugs 2014; 14:147-154
    [231] Wong PC, Pinto DJ, Zhang D. Preclinical discovery of apixaban, a direct and orally bioavailable factor Xa inhibitor. J Thromb Thrombolysis 2011; 31:478-492
    [232] Agrawal S, Jiang Z, Zhao Q, Shaw D, Cai Q, Roskey A, et al. Mixed-backbone oligonucleotides as second generation antisense oligonucleotides:in vitro and in vivo studies. Proc Natl Acad Sci U S A 1997; 94:2620-2625
    [233] Yu AM, Choi YH, Tu MJ. RNA drugs and RNA targets for small molecules:principles, progress, and challenges. Pharmacol Rev 2020; 72:862-898
    [234] Bennett CF. Therapeutic antisense oligonucleotides are coming of age. Annu Rev Med 2019; 70:307-321
    [235] Roberts TC, Langer R, Wood MJA. Advances in oligonucleotide drug delivery. Nat Rev Drug Discov 2020; 19:673-694
    [236] Shemesh CS, Yu RZ, Warren MS, Liu M, Jahic M, Nichols B, et al. Assessment of the drug interaction potential of unconjugated and GalNAc3-conjugated 2'-MOE-ASOs. Mol Ther Nucleic Acids 2017; 9:34-47
    [237] Society TA. Therapeutic monoclonal antibodies approved or in review in the EU or US. Accessed[May 1, 2021]. Available from:https://www.antibodysociety.org/antibody-therapeutics-product-data.
    [238] Lu RM, Hwang YC, Liu IJ, Lee CC, Tsai HZ, Li HJ, et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci 2020; 27:1
    [239] Wang W, Wang EQ, Balthasar JP. Monoclonal antibody pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther 2008; 84:548-558
    [240] Tibbitts J, Canter D, Graff R, Smith A, Khawli LA. Key factors influencing ADME properties of therapeutic proteins:a need for ADME characterization in drug discovery and development. MAbs 2016; 8:229-245
    [241] Datta-Mannan A. Mechanisms influencing the pharmacokinetics and disposition of monoclonal antibodies and peptides. Drug Metab Dispos 2019; 47:1100-1110
    [242] Mould DR, Green B. Pharmacokinetics and pharmacodynamics of monoclonal antibodies:concepts and lessons for drug development. BioDrugs 2010; 24:23-39
    [243] Shah DK, Betts AM. Towards a platform PBPK model to characterize the plasma and tissue disposition of monoclonal antibodies in preclinical species and human. J Pharmacokinet Pharmacodyn 2012; 39:67-86
    [244] Sand KM, Bern M, Nilsen J, Noordzij HT, Sandlie I, Andersen JT. Unraveling the interaction between FcRn and albumin:opportunities for design of albumin-based therapeutics. Front Immunol 2014; 5:682
    [245] Deng R, Iyer S, Theil FP, Mortensen DL, Fielder PJ, Prabhu S. Projecting human pharmacokinetics of therapeutic antibodies from nonclinical data:what have we learned?. MAbs 2011; 3:61-66
    [246] Dua P, Hawkins E, van der Graaf PH. A tutorial on target-mediated drug disposition (TMDD) models. CPT Pharmacometrics Syst Pharmacol 2015; 4:324-337
    [247] Morgan ET, Goralski KB, Piquette-Miller M, Renton KW, Robertson GR, Chaluvadi MR, et al. Regulation of drug-metabolizing enzymes and transporters in infection, inflammation, and cancer. Drug Metab Dispos 2008; 36:205-216
    [248] FDA. Drug-drug interaction assessment for therapeutic proteins Published[August 2020]; Accessed[February, 2022]. Available from:https://www.fda.gov/media/140909/download.
    [249] Tumey LN. An overview of the current ADC discovery landscape. Methods Mol Biol 2020; 2078:1-22
    [250] Liu-Kreyche P, Shen H, Marino AM, Iyer RA, Humphreys WG, Lai Y. Lysosomal P-gp-MDR1 confers drug resistance of brentuximab vedotin and its cytotoxic payload monomethyl auristatin E in tumor cells. Front Pharmacol 2019; 10:749
    [251] Przepiorka D, Deisseroth A, Kane R, Kaminskas E, Farrell AT, Pazdur R. Gemtuzumab ozogamicin. J Clin Oncol 2013; 31:1699-1700
    [252] Matsuda Y, Mendelsohn BA. An overview of process development for antibody-drug conjugates produced by chemical conjugation technology. Expert Opin Biol Ther 2021; 21:963-975
    [253] Li P, Gong Y, Kim J, Liu X, Gilbert J, Kerns HM, et al. Hybridization liquid chromatography-tandem mass spectrometry:an alternative bioanalytical method for antisense oligonucleotide quantitation in plasma and tissue samples. Anal Chem 2020; 92:10548-10559
    [254] Hock MB, Thudium KE, Carrasco-Triguero M, Schwabe NF. Immunogenicity of antibody drug conjugates:bioanalytical methods and monitoring strategy for a novel therapeutic modality. AAPS J 2015; 17:35-43
    [255] Carrasco-Triguero M, Dere RC, Milojic-Blair M, Saad OM, Nazzal D, Hong K, et al. Immunogenicity of antibody-drug conjugates:observations across 8 molecules in 11 clinical trials. Bioanalysis 2019; 11:1555-1568
    [256] Teorell T. Kinetics of distribution of substances administered to the body. I. The extravascular modes of administration. Arch. Int. Pharmacodyn. Ther. 1937; 57:205-225
    [257] Rowland M, Peck C, Tucker G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol 2011; 51:45-73
    [258] Heimbach T, Chen Y, Chen J, Dixit V, Parrott N, Peters SA, et al. Physiologically-based pharmacokinetic modeling in renal and hepatic impairment populations:a pharmaceutical industry perspective. Clin Pharmacol Ther 2021; 110:297-310
    [259] Jones HM, Chen Y, Gibson C, Heimbach T, Parrott N, Peters SA, et al. Physiologically based pharmacokinetic modeling in drug discovery and development:a pharmaceutical industry perspective. Clin Pharmacol Ther 2015; 97:247-262
    [260] Sinha V, Zhao P, Huang SM, Zineh I. Physiologically based pharmacokinetic modeling:from regulatory science to regulatory policy. Clin Pharmacol Ther 2014; 95:478-480
    [261] Wagner C, Zhao P, Pan Y, Hsu V, Grillo J, Huang SM, et al. Application of physiologically based pharmacokinetic (PBPK) modeling to support dose selection:report of an FDA public workshop on PBPK. CPT Pharmacometrics Syst Pharmacol 2015; 4:226-230
    [262] Riedmaier AE, DeMent K, Huckle J, Bransford P, Stillhart C, Lloyd R, et al. Use of physiologically based pharmacokinetic (PBPK) modeling for predicting drug-food interactions:an industry perspective. AAPS J 2020; 22:123
    [263] Taskar KS, Pilla Reddy V, Burt H, Posada MM, Varma M, Zheng M, et al. Physiologically-based pharmacokinetic models for evaluating membrane transporter mediated drug-drug interactions:current capabilities, case studies, future opportunities, and recommendations. Clin Pharmacol Ther 2020; 107:1082-1115
    [264] Zhang X, Yang Y, Grimstein M, Fan J, Grillo JA, Huang SM, et al. Application of PBPK modeling and simulation for regulatory decision making and its impact on US prescribing information:an update on the 2018-2019 submissions to the US FDA's Office of Clinical Pharmacology. J Clin Pharmacol 2020; 60 Suppl 1:S160-S178
    [265] Mueller KT, Waldron E, Grupp SA, Levine JE, Laetsch TW, Pulsipher MA, et al. Clinical pharmacology of tisagenlecleucel in B-cell acute lymphoblastic leukemia. Clin Cancer Res 2018; 24:6175-6184
    [266] Stein AM, Grupp SA, Levine JE, Laetsch TW, Pulsipher MA, Boyer MW, et al. Tisagenlecleucel model-based cellular kinetic analysis of chimeric antigen receptor-T cells. CPT Pharmacometrics Syst Pharmacol 2019; 8:285-295
    [267] Liu C, Ayyar VS, Zheng X, Chen W, Zheng S, Mody H, et al. Model-based cellular kinetic analysis of chimeric antigen receptor-T cells in humans. Clin Pharmacol Ther 2021; 109:716-727
    [268] Singh AP, Zheng X, Lin-Schmidt X, Chen W, Carpenter TJ, Zong A, et al. Development of a quantitative relationship between CAR-affinity, antigen abundance, tumor cell depletion and CAR-T cell expansion using a multiscale systems PK-PD model. MAbs 2020; 12:1688616
    [269] Yu RZ, Grundy JS, Geary RS. Clinical pharmacokinetics of second generation antisense oligonucleotides. Expert Opin Drug Metab Toxicol 2013; 9:169-182
    [270] Yu RZ, Kim TW, Hong A, Watanabe TA, Gaus HJ, Geary RS. Cross-species pharmacokinetic comparison from mouse to man of a second-generation antisense oligonucleotide, ISIS 301012, targeting human apolipoprotein B-100. Drug Metab Dispos 2007; 35:460-468
    [271] Geary RS. Antisense oligonucleotide pharmacokinetics and metabolism. Expert Opin Drug Metab Toxicol 2009; 5:381-391
    [272] Huang SM, Strong JM, Zhang L, Reynolds KS, Nallani S, Temple R, et al. New era in drug interaction evaluation:US Food and Drug Administration update on CYP enzymes, transporters, and the guidance process. J Clin Pharmacol 2008; 48:662-670
    [273] Zhang L, Reynolds KS, Zhao P, Huang SM. Drug interactions evaluation:an integrated part of risk assessment of therapeutics. Toxicol Appl Pharmacol 2010; 243:134-145
    [274] Rekic D, Reynolds KS, Zhao P, Zhang L, Yoshida K, Sachar M, et al. Clinical drug-drug interaction evaluations to inform drug use and enable drug access. J Pharm Sci 2017; 106:2214-2218
    [275] Yoshida K, Zhao P, Zhang L, Abernethy DR, Rekic D, Reynolds KS, et al. In vitro-in vivo extrapolation of metabolism- and transporter-mediated drug-drug interactions-overview of basic prediction methods. J Pharm Sci 2017; 106:2209-2213
    [276] Huang SM, Zhang L, Giacomini KM. The International Transporter Consortium:a collaborative group of scientists from academia, industry, and the FDA. Clin Pharmacol Ther 2010; 87:32-36
    [277] Giacomini KM, Huang SM. Transporters in drug development and clinical pharmacology. Clin Pharmacol Ther 2013; 94:3-9
    [278] Giacomini KM, Galetin A, Huang SM. The International Transporter Consortium:summarizing advances in the role of transporters in drug development. Clin Pharmacol Ther 2018; 104:766-771
    [279] FDA. In vitro drug interaction studies-cytochrome P450 enzyme- and transporter-mediated drug interactions. Published[January 2020]. Accessed[February 2022]. Available form:https://www.fda.gov/media/134582/download.
    [280] FDA. Clinical drug interaction studies-cytochrome P450 enzyme- and transporter-mediated drug interactions. Published[January 2020]. Accessed[February 2022]. Available form:https://www.fda.gov/media/134581/download.
    [281] FDA. Drug interaction & labeling. Published[January 2020]. Accessed[February 2022]. Available from:https://www.fda.gov/drugs/development-resources/drug-interactions-labeling.
    [282] Zhao P, Zhang L, Grillo JA, Liu Q, Bullock JM, Moon YJ, et al. Applications of physiologically based pharmacokinetic (PBPK) modeling and simulation during regulatory review. Clin Pharmacol Ther 2011; 89:259-267
    [283] Zhao P, Rowland M, Huang SM. Best practice in the use of physiologically based pharmacokinetic modeling and simulation to address clinical pharmacology regulatory questions. Clin Pharmacol Ther 2012; 92:17-20
    [284] Wagner C, Pan Y, Hsu V, Sinha V, Zhao P. Predicting the effect of CYP3A inducers on the pharmacokinetics of substrate drugs using physiologically based pharmacokinetic (PBPK) modeling:an analysis of PBPK submissions to the US FDA. Clin Pharmacokinet 2016; 55:475-483
    [285] Wagner C, Pan Y, Hsu V, Grillo JA, Zhang L, Reynolds KS, et al. Predicting the effect of cytochrome P450 inhibitors on substrate drugs:analysis of physiologically based pharmacokinetic modeling submissions to the US Food and Drug Administration. Clin Pharmacokinet 2015; 54:117-127
    [286] Zhang L, Wu F, Lee SC, Zhao H, Zhang L. pH-dependent drug-drug interactions for weak base drugs:potential implications for new drug development. Clin Pharmacol Ther 2014; 96:266-277
    [287] Dong Z, Li J, Wu F, Zhao P, Lee SC, Zhang L, et al. Application of physiologically-based pharmacokinetic modeling to predict gastric pH-dependent drug-drug interactions for weak base drugs. CPT Pharmacometrics Syst Pharmacol 2020; 9:456-465
    [288] Mitra A, Parrott N, Miller N, Lloyd R, Tistaert C, Heimbach T, et al. Prediction of pH-dependent drug-drug interactions for basic drugs using physiologically based biopharmaceutics modeling:industry case studies. J Pharm Sci 2020; 109:1380-1394
    [289] Evers R, Dallas S, Dickmann LJ, Fahmi OA, Kenny JR, Kraynov E, et al. Critical review of preclinical approaches to investigate cytochrome P450-mediated therapeutic protein drug-drug interactions and recommendations for best practices:a white paper. Drug Metab Dispos 2013; 41:1598-1609
    [290] Kenny JR, Liu MM, Chow AT, Earp JC, Evers R, Slatter JG, et al. Therapeutic protein drug-drug interactions:navigating the knowledge gaps-highlights from the 2012 AAPS NBC Roundtable and IQ Consortium/FDA workshop. AAPS J 2013; 15:933-940
    [291] Huang SM, Zhao H, Lee JI, Reynolds K, Zhang L, Temple R, et al. Therapeutic protein-drug interactions and implications for drug development. Clin Pharmacol Ther 2010; 87:497-503
    [292] Lee JI, Zhang L, Men AY, Kenna LA, Huang SM. CYP-mediated therapeutic protein-drug interactions:clinical findings, proposed mechanisms and regulatory implications. Clin Pharmacokinet 2010; 49:295-310
    [293] Jing X, Ji P, Schrieber SJ, Fletcher EP, Sahajwalla C. Update on therapeutic protein-drug interaction:information in labeling. Clin Pharmacokinet 2020; 59:25-36
    [294] Bissig KD, Han W, Barzi M, Kovalchuk N, Ding L, Fan X, et al. P450-humanized and human liver chimeric mouse models for studying xenobiotic metabolism and toxicity. Drug Metab Dispos 2018; 46:1734-1744
  • 加载中
计量
  • 文章访问数:  58
  • HTML全文浏览量:  41
  • PDF下载量:  0
  • 被引次数: 0
出版历程
  • 收稿日期:  2021-09-22
  • 修回日期:  2021-11-07
  • 录用日期:  2021-11-10
  • 网络出版日期:  2023-03-17

目录

    /

    返回文章
    返回