299
Views
0
CrossRef citations to date
0
Altmetric
Review

Innovation in Bioanalytical Strategies and in vitro Drug–drug Interaction Study Approaches in Drug Discovery

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 513-532 | Received 02 Jan 2021, Accepted 18 Feb 2021, Published online: 08 Mar 2021

References

  • Takebe T , ImaiR, OnoS. The current status of drug discovery and development as originated in United States academia: the influence of industrial and academic collaboration on drug discovery and development. Clin. Transl. Sci.11(6), 597–606 (2018).
  • Hennessy S , FlockhartDA. The need for translational research on drug–drug interactions. Clin. Pharmacol. Ther.91(5), 771–773 (2012).
  • Hughes JP , ReesS, KalindjianSB, PhilpottKL. Principles of early drug discovery. Br. J. Pharmacol.162(6), 1239–1249 (2011).
  • Lu C , DiL. In vitro and in vivo methods to assess pharmacokinetic drug– drug interactions in drug discovery and development. Biopharm. Drug Dispos.41(1–2), 3–31 (2020).
  • Hebenstreit D , PichlerR, HeideggerI. Drug–drug interactions in prostate cancer treatment. Clin. Genitourin Cancer18(2), e71–e82 (2020).
  • Gujjarlamudi HB . Polytherapy and drug interactions in elderly. J. Midlife Health7(3), 105–107 (2016).
  • Fahmi OA , RippSL. Evaluation of models for predicting drug–drug interactions due to induction. Expert Opin. Drug Metab. Toxicol.6(11), 1399–1416 (2010).
  • Palleria C , DiPaolo A, GiofrèCet al. Pharmacokinetic drug–drug interaction and their implication in clinical management. J. Res. Med. Sci.18(7), 601 (2013).
  • Hakkola J , HukkanenJ, TurpeinenM, PelkonenO. Inhibition and induction of CYP enzymes in humans: an update. Arch. Toxicol.94, 3671–3722 (2020).
  • Tornio A , FilppulaAM, NiemiM, BackmanJT. Clinical studies on drug–drug interactions involving metabolism and transport: methodology, pitfalls, and interpretation. Clin. Pharmacol. Ther.105(6), 1345–1361 (2019).
  • Yu H , TweedieD. A perspective on the contribution of metabolites to drug–drug interaction potential: the need to consider both circulating levels and inhibition potency. Drug Metab. Dispos.41(3), 536–540 (2013).
  • USFDA . Guidances (Drugs) (2012). http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm
  • European Medicine Agency . Guideline on the Investigation of Drug Interactions. 44, 1–59 (2015).
  • Stampella A , RizzitelliG, DonatiFet al. Human hepatoma cell lines on gas foaming templated alginate scaffolds for in vitro drug–drug interaction and metabolism studies. Toxicol. In Vitro30(1), 331–340 (2015).
  • Shinha K , NiheiW, OnoT, NakazatoR, KimuraH. A pharmacokinetic–pharmacodynamic model based on multi-organ-on-a-chip for drug–drug interaction studies. Biomicrofluidics14(4), 044108 (2020).
  • Shibata Y , TakahashiH, ChibaM, IshiiY. A novel approach to the prediction of drug–drug interactions in humans based on the serum incubation method. Drug Metab. Pharmacokinet.23(5), 328–339 (2008).
  • Horspool AM , WangT, ScaringellaYS, TaubME, ChanTS. Human liver microsomes immobilized on magnetizable beads: a novel approach to study in vitro drug metabolism. Drug Metab. Dispos.48(8), 645–654 (2020).
  • Mattei G , GiustiS, AhluwaliaA. Design criteria for generating physiologically relevant in vitro models in bioreactors. Processes2(3), 548–569 (2014).
  • Rodriguez-Garcia A , Oliva-RamirezJ, Bautista-FloresC, HosseiniS. 3D in vitro human organ mimicry devices for drug discovery, development, and assessment. Adv. Polym. Tech.2, 2020 (2020).
  • Chung TDY , TerryDB, SmithLH. In vitro and in vivo assessment of ADME and PK properties during lead selection and lead optimization–guidelines, benchmarks and rules of thumb. Assay Guidance Manual1, 1261–1280 (2015).
  • Andrade EL , BentoAF, CavalliJet al. Non-clinical studies required for new drug development–Part I: early in silico and in vitro studies, new target discovery and validation, proof of principles and robustness of animal studies. Braz. J. Med. Biol. Res.49(11), (2016).
  • Jia L , LiuX. The conduct of drug metabolism studies considered good practice (II): in vitro experiments. Curr. Drug Metab.8(8), 822–829 (2007).
  • Campbell JE , CohallD. Pharmacodynamics–a pharmacognosy perspective. Pharmacognosy. Elsevier, 513–525 (2017).
  • Kremers P . In vitro tests for predicting drug–drug interactions: the need for validated procedures. Pharmacol. Toxicol.91(5), 209–217 (2002).
  • Ogu CC , MaxaJL. Drug interactions due to cytochrome P450. Baylor Univ. Med. Cent. Proc.13(4), 421–423 (2000).
  • Alsanosi SMM , SkiffingtonC, PadmanabhanS. Pharmacokinetic pharmacogenomics. In: Handbook of Pharmacogenomics and Stratified Medicine.Elsevier Inc, 341–364 (2014).
  • Shetty V , ChowtaMN, ChowtaKN, ShenoyA, KamathA, KamathP. Evaluation of potential drug–drug interactions with medications prescribed to geriatric patients in a tertiary care hospital. J. Aging Res.2018, 1–6 (2018).
  • Tripathi KD . General Pharmacological Principles. In: Essentials of Medical Pharmacology (7th Edition). Jaypee, 928–934 (2013).
  • Zhou S , ChanSY, GohBCet al. Mechanism-based inhibition of cytochrome P450 3A4 by therapeutic drugs. Clin. Pharmacokinet.44(3), 279–304 (2005).
  • Cascorbi I . Drug interactions-principles, examples and clinical consequences. Dtsch Arztebl Int.109(33–34), 546–556 (2012).
  • Brodie MJ , MintzerS, PackAM, GidalBE, VechtCJ, SchmidtD. Enzyme induction with antiepileptic drugs: cause for concern?Epilepsia54(1), 11–27 (2013).
  • Maddi S , GoudT, SrivastavaP. Cytochrome P450 enzymes, drug transporters and their role in pharmacokinetic drug–drug interactions of xenobiotics: a comprehensive review. Open J. Chem.3, 001–011 (2017).
  • European Pharmaceutical Review . Drug–Drug Interactions: Tools for Drug Transporter Protein Studies (2009). https://www.europeanpharmaceuticalreview.com/article/782/drug-drug-interactions-tools-for-drug-transporter-protein-studies/
  • Zhang L , StrongJM, QiuW, LeskoLJ, HuangSM. Scientific perspectives on drug transporters and their role in drug interaction. Mol. Pharm.3(1), 62–69 (2006).
  • Joshi A , KieselBF, ChaphekarNet al. In vitro evaluation of the metabolic enzymes and drug interaction potential of triapine. Cancer Chemother. Pharmacol.86(5), 633–640 (2020).
  • Wu Q , JiangH, WangSet al. Effects of avitinib on the pharmacokinetics of osimertinib in vitro and in vivo in rats. Thorac. Cancer11(10), 2775–2781 (2020).
  • Chen N , CuiD, WangQ, WenZ, FinkelmanRD, WeltyD. In vitro drug–drug interactions of budesonide: inhibition and induction of transporters and cytochrome P450 enzymes. Xenobiotica48(6), 637–646 (2018).
  • Mukoyoshi M , NishimuraS, HoshideSet al. In vitro drug–drug interaction studies with febuxostat, a novel non-purine selective inhibitor of xanthine oxidase: plasma protein binding, identification of metabolic enzymes and cytochrome P450 inhibition. Xenobiotica38(5), 496–510 (2008).
  • Wang Y , WangC, WangSet al. Cytochrome P450-based drug–drug interactions of vonoprazan in vitro and in vivo. Front. Pharmacol.11, 1–9 (2020).
  • Seo S-W , ParkJW, HanD-Get al. In vitro and in vivo assessment of metabolic drug interaction potential of dutasteride with ketoconazole. Pharmaceutics11(12), 673 (2019).
  • Shimakura J , TaniN, MizunoY, KomuroS, KanamaruH. In vitro drug–drug interactions with perospirone and concomitantly administered drugs in human liver microsomes. Eur. J. Drug Metab. Pharmacokinet.28(I), 67–72 (2003).
  • Rautio J , HumphreysJE, WebsterLOet al. In vitro p-glycoprotein inhibition assays for assessment of clinical drug interaction potential of new drug candidates: a recommendation for probe substrates. Drug Metab. Dispos.34(5), 786–792 (2006).
  • Kosloski MP , BowDA, KikuchiRet al. Translation of in vitro transport inhibition studies to clinical drug–drug interactions for glecaprevir and pibrentasvir s. J. Pharmacol. Exp. Ther.370(2), 278–287 (2019).
  • Shitara Y , ItohT, SatoH, LiAP, SugiyamaY. Inhibition of transporter-mediated hepatic uptake as a mechanism for drug–drug interaction between cerivastatin and cyclosporin A. J. Pharmacol. Exp. Ther.304(2), 610–616 (2003).
  • Parmentier Y , BossantM-J, BertrandM, WaltherB. In vitro studies of drug metabolism. Compr. Med. Chem. II5, 231–257 (2006).
  • Ackley DC , RockichKT, BakerTR. Metabolic stability assessed by liver microsomes and hepatocytes. Optim. Drug Discov.1, 151–162 (2004).
  • Richardson SJ , BaiA, KulkarniAA, MoghaddamMF. Efficiency in drug discovery: liver S9 fraction assay as a screen for metabolic stability. Drug Metab. Lett.10(2), 83–90 (2016).
  • Pilchova I , KlacanovaK, TatarkovaZ, KaplanP, RacayP. The involvement of Mg 2 + in regulation of cellular and mitochondrial functions. Oxid. Med. Cell Longev.2017, 1–8 (2017).
  • Houston JB , GaletinA. In vitro techniques to study drug–drug interactions of drug metabolism: cytochrome P450. In: Enzyme- and Transporter- Based Drug–Drug Interactions. Springer, 169–215 (2010).
  • Di Marco A , MarcucciI, VerdirameMet al. Development and validation of a high-throughput radiometric CYP3A4/5 inhibition assay using tritiated testosterone. Drug Metab. Dispos.33(3), 349–358 (2005).
  • Fowler S , ZhangH. In vitro evaluation of reversible and irreversible cytochrome P450 inhibition: current status on methodologies and their utility for predicting drug–drug interactions. AAPS J.10(2), 410–424 (2008).
  • Trubetskoy OV , GibsonJR, MarksBD. Highly miniaturized formats for in vitro drug metabolism assays using Vivid® fluorescent substrates and recombinant human cytochrome P450 enzymes. J. Biomol. Screen.10(1), 56–66 (2005).
  • Crespi CL , MillerVP, PenmanBW. Microtiter plate assays for inhibition of human, drug-metabolizing cytochromes P450. Anal. Biochem.248(1), 188–190 (1997).
  • Hossain MJ , SultanMZ, RashidMA, KuddusMR. Does rabeprazole sodium alleviate the anti-diabetic activity of linagliptin? drug–drug interaction analysis by in vitro and in vivo methods. Drug Res.70(11), 519–527 (2020).
  • Yong YF , TanSC, LiewMWO, YaacobNS. Liquid chromatography-tandem mass spectrometry (LC–MS/MS) method development for screening of potential tamoxifen-drug/herb interaction via in vitro cytochrome P450 inhibition assay. J. Chromatogr. B1148, 122148 (2020).
  • Bjornsson TD , CallaghanJT, EinolfHJet al. The conduct of in vitro and in vivo drug–drug interaction studies: a PhRMA perspective. J. Clin. Pharmacol.43(5), 443–469 (2003).
  • Yu C . Metabolism and in vitro drug–drug interaction assessment of viloxazine. Xenobiotica50(11), 1285–1300 (2020).
  • Peng Y , WuH, ZhangXet al. A comprehensive assay for nine major cytochrome P450 enzymes activities with 16 probe reactions on human liver microsomes by a single LC/MS/MS run to support reliable in vitro inhibitory drug–drug interaction evaluation. Xenobiotica45(11), 961–977 (2015).
  • Scherf-Clavel O , KinzigM, StoffelMS, FuhrU, SörgelF. Quantification of adefovir and pitavastatin in human plasma and urine by LC–MS/MS: a useful tool for drug–drug interaction studies. J. Chromatogr. B.1125, 121718 (2019).
  • Liu J , ZhongX, JiangYet al. Systematic identification metabolites of hemerocallis citrina Borani by high-performance liquid chromatography/quadrupole-time-of-flight mass spectrometry combined with a screening method. J. Pharm. Biomed. Anal.186, 113314 (2020).
  • Elgawish MS , SoltanMK, SebaiyMM. Molecular modeling, spectrofluorimetric, and tandem mass spectrometric analysis reveal a competitive binding of amlodipine and rosuvastatin to plasma albumin: insight into drug–drug interaction. Microchem. J.149, 104014 (2019).
  • Balhara A , LadumorM, SinghDKet al. In vitro evaluation of reactive nature of E- and Z-guggulsterones and their metabolites in human liver microsomes using UHPLC-Orbitrap mass spectrometer. J. Pharm. Biomed. Anal.186, 113275 (2020).
  • Di L . Reaction phenotyping to assess victim drug–drug interaction risks. Expert Opin. Drug Discov.12(11), 1105–1115 (2017).
  • Gabriel L , TodM, GoutelleS. Quantitative prediction of drug interactions caused by CYP1A2 inhibitors and inducers. Clin. Pharmacokinet.55(8), 977–990 (2016).
  • Turpeinen M , RaunioH, PelkonenO. The functional role of CYP2B6 in human drug metabolism: substrates and inhibitors in vitro, in vivo and in silico. Curr. Drug Metab.7(7), 705–714 (2006).
  • Daily EB , AquilanteCL. Cytochrome P450 2C8 pharmacogenetics: a review of clinical studies. Pharmacogenomics10(9), 1489–1510 (2009).
  • Booven DV , MarshS, McLoedH. Cytochrome P450 2C9-CYP2C9. NIH public access. Bone20(4), 277–281 (2010).
  • Joanna KT , RebeccaNC, MichelleDWet al. Cytochrome P450 3A4 and CYP3A5-catalyzed bioactivation of lapatinib. Drug Metab Dispos.44(10), 1584–1597 (2014).
  • Rendic S . Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab. Rev.34(1–2), 83–448 (2002).
  • Dalvie D , DiL. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol. Ther.201, 137–180 (2019).
  • Shen Y , ShiZ, YanB. Carboxylesterases: pharmacological inhibition regulated expression and transcriptional involvement of nuclear receptors and other transcription factors. Nucl. Recept. Res.6, 1–15 (2019).
  • Polasek TM , ElliotDJ, SomogyiAA, GillamEMJ, LewisBC, MinersJO. An evaluation of potential mechanism-based inactivation of human drug metabolizing cytochromes P450 by monoamine oxidase inhibitors, including isoniazid. Br. J. Clin. Pharmacol.61(5), 570–584 (2006).
  • Ryu SD , YiHG, ChaYNet al. Flavin-containing monooxygenase activity can be inhibited by nitric oxide-mediated S-nitrosylation. Life Sci.75(21), 2559–2572 (2004).
  • Celius T , PansoyA, MatthewsJet al. Flavin-containing monooxygenase-3: induction by 3-methylcholanthrene and complex regulation by xenobiotic chemicals in hepatoma cells and mouse liver. Toxicol. Appl. Pharmacol.247(1), 60–69 (2010).
  • Mohos V , Fliszár-NyúlE, PoórM. Inhibition of xanthine oxidase-catalyzed xanthine and 6-mercaptopurine oxidation by flavonoid aglycones and some of their conjugates. Int. J. Mol. Sci.21(9), 3256 (2020).
  • Ercan S , KencebayC, BasaranlarG, DerinN, AslanM. Induction of xanthine oxidase activity, endoplasmic reticulum stress and caspase activation by sodium metabisulfite in rat liver and their attenuation by Ghrelin. Food Chem. Toxicol.76, 27–32 (2015).
  • Koppaka V , ThompsonDC, ChenYet al. Aldehyde dehydrogenase inhibitors: a comprehensive review of the pharmacology, mechanism of action, substrate specificity, and clinical application. Pharmacol. Rev.64(3), 520–539 (2012).
  • Lv X , XiaY, FinelM, WuJ, GeG, YangL. Recent progress and challenges in screening and characterization of UGT1A1 inhibitors. Acta Pharm. Sin. B9(2), 258–278 (2019).
  • Marques SC , IkediobiON. The clinical application of UGT1A1 pharmacogenetic testing: gene-environment interactions. Hum. Genom.4(4), 238 (2010).
  • You BH , GongEC, ChoiYH. Inhibitory effect of sauchinone on UDP-glucuronosyltransferase (UGT) 2B7 activity. Molecules23(2), 366 (2018).
  • Wang Z , WongT, HashizumeTet al. Human UGT1A4 and UGT1A3 conjugate 25-hydroxyvitamin D3: metabolite structure, kinetics, inducibility, and interindividual variability. Endocrinology155(6), 2052–2063 (2014).
  • Jiang L , LiangSC, WangCet al. Identifying and applying a highly selective probe to simultaneously determine the O-glucuronidation activity of human UGT1A3 and UGT1A4. Sci. Rep.5(1), 1–7 (2015).
  • Krishnaswamy S , HaoQ, Von MoltkeLL, GreenblattDJ, CourtMH. Evaluation of 5-hydroxytryptophol and other endogenous serotonin (5-hydroxytryptamine) analogs as substrates for UDP-glucuronosyltransferase 1A6. Drug Metab. Dispos.32(8), 862–869 (2004).
  • Hu DG , MackenziePI, LuL, MeechR, McKinnonRA. Induction of human UDP-Glucuronosyltransferase 2B7 gene expression by cytotoxic anticancer drugs in liver cancer HepG2 Cells. Drug Metab. Dispos.43(5), 660–668 (2015).
  • Barre L , Fournel-GigleuxS, FinelM, NetterP, MagdalouJ, OuzzineM. Substrate specificity of the human UDP-glucuronosyltransferase UGT2B4 and UGT2B7: identification of a critical aromatic amino acid residue at position 33. FEBS J.274(5), 1256–1264 (2007).
  • Sten T , FinelM, AskB, RaneA, EkströmL. Non-steroidal anti-inflammatory drugs interact with testosterone glucuronidation. Steroids74(12), 971–977 (2009).
  • De Leon J . Glucuronidation enzymes, genes and psychiatry. Int. J. Neuropsychopharmacol.6(1), 57–72 (2003).
  • Lévesque É , BeaulieuM, GuillemetteC, HumDW, BélangerA. Effect of interleukins on UGT2B15 and UGT2B17 steroid uridine diphosphate-glucuronosyltransferase expression and activity in the LNCaP cell line. Endocrinology139(5), 2375–2381 (1998).
  • Zhou T , ChenY, HuangC, ChenG. Caffeine induction of sulfotransferases in rat liver and intestine. J. Appl. Toxicol.32(10), 804–809 (2012).
  • Wang L-Q , JamesM. Inhibition of sulfotransferases by xenobiotics. Curr. Drug Metab.7(1), 83–104 (2006).
  • Allocati N , MasulliM, DiIlio C, FedericiL. Glutathione transferases: substrates, inihibitors and pro-drugs in cancer and neurodegenerative diseases. Oncogenesis7(1), 1–15 (2018).
  • Vogel HG , HockFJ, MaasJ, MayerD. Safety and pharmacokinetic assays. In: Drug Discovery and Evaluation (2nd Edition). Springer Science & Business Media, Heidelberg, Germany (2006).
  • Li Y , ZhouD, FergusonSS, DorffP, SimpsonTR, GrimmSW. In vitro assessment of metabolic drug–drug interaction potential of AZD2624, neurokinin-3 receptor antagonist, through cytochrome P450 enzyme identification, inhibition, and induction studies. Xenobiotica40(11), 721–729 (2010).
  • US FDA . In Vitro Drug Interaction Studies–Cytochrome P450 Enzyme- and Drug Interactions Guidance for Industry In Vitro Drug Interaction Studies (2020). https://www.fda.gov/media/134582/download
  • Hofstaedter F , EbnerR. The use of 3-D cultures for high-throughput screening. J. Biomol. Screen.9(4), 273–285 (2004).
  • Bhatia SN , IngberDE. Microfluidics organ-on-chips: a perspective. Nat. Biotechnol.32, 760–772 (2014).
  • Esch EW , BahinskiA, HuhD. Organs-on-chips at the frontiers of drug discovery. Nat. Rev. Drug Discov.14(4), 248–260 (2015).
  • Deng J , ZhangX, ChenZet al. A cell lines derived microfluidic liver model for investigation of hepatotoxicity induced by drug–drug interaction. Biomicrofluidics13, 024101 (2019).
  • Tuschl G , HrachJ, WalterY, HewittPG, MuellerSO. Serum-free collagen sandwich cultures of adult rat hepatocytes maintain liver-like properties long term: a valuable model for in vitro toxicity and drug–drug interaction studies. Chem. Biol. Interact.181(1), 124–137 (2009).
  • Yamamoto N , WuJ, ZhangYet al. An optimal culture condition maintains human hepatocyte phenotype after long-term culture. J. Hepatol.35(3), 169–177 (2006).
  • Das A , SligarSG. Modulation of the cytochrome P450 reductase redox potential by the phospholipid bilayer. Biochemistry48, 12104–12112 (2009).
  • Rowland A , GaganisP, ElliotDJ, MackenziePI, KnightsKM, MinersJO. Binding of inhibitory fatty acids is responsible for the enhancement of UDP-Glucuronosyltransferase 2B7 activity by albumin: implications for in vitro-in vivo extrapolation. J. Pharmacol. Exp. Ther.321(1), 137–147 (2007).
  • Pharmaceuticals and Medical Devices Agency (Japan) . Guideline on Drug Interaction for Drug Development and Appropriate Provision of Information (2019). https://www.xenotech.com/access-adme-research-resources/resources/2019-japanese-pmda-guideline-english-translation/
  • Yoshida K , ZhaoP, ZhangLet al. In vitro–in vivo extrapolation of metabolism- and transporter-mediated drug–drug interactions—overview of basic prediction methods. J. Pharm. Sci.106(9), 2209–2213 (2017).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.