Introduction/Aim. Herbs have been a vital renewable source of medicine throughout human history as a large proportion of the global population still depends on them for their health benefits. The increasing popularity of herbal supplements has raised an obvious concern about the overall safety and potential interaction with other drugs in situ. The intent was to spur future research on herb-drug interactions as well as the mechanisms of interaction to understand the consequences of such interactions. Methods. The review was conducted by a systematic search of relevant literature using the databases of Google Scholar, Science Direct, Mendeley, Scopus, and PubMed. Publications written in English were used. Many herbal products are reported to exhibit herb-drug interaction with known orthodox medicines. The inhibition-induction mechanism triggers chain reactions which often result in reduced drug bioavailability, toxicities, or undesirable side effects. Some herbal phytoconstituents reportedly bind CYP2C9, CYP2C19, CYP2E1, and CYP3A1 among numerous others temporarily or irreversibly. Conclusion. The study was concluded by reiterating the imperativeness to routinely and regularly inform both physicians and patients of the inherent dangers such as reduced efficacy and increased toxicities associated with herb-drug interactions (HDI). Herb users should be regularly advised on the appropriate use of herbal supplements to avoid the risk of adverse drug interactions during co-administrations or in combination therapies. As both synergistic and antagonistic effects could be observed in HDI, further preclinical and clinical empirical studies are required to underscore the mechanism and extent of HDI.
References
1.
Cheng W, Xia K, Wu S, Li Y. Herb-Drug Interactions and Their Impact on Pharmacokinetics: An Update. Current Drug Metabolism. 2023;24(1):28–69.
2.
Liu D, Zhang L, Duan L, Wu J, Hu M, Liu Z, et al. Potential of herb-drug / herb interactions between substrates and inhibitors of UGTs derived from herbal medicines. Pharmacological Research. 2019;150:104510.
3.
Currie GM. Pharmacology, Part 2: Introduction to Pharmacokinetics. Journal of Nuclear Medicine Technology. 2018;46(3):221–30.
4.
Yan J, He X, Feng S, Zhai Y, Ma Y, Liang S, et al. Up-regulation on cytochromes P450 in rat mediated by total alkaloid extract from Corydalis yanhusuo. BMC Complementary and Alternative Medicine. 2014;14(1).
5.
Ma BL, Ma YM. Pharmacokinetic herb–drug interactions with traditional Chinese medicine: progress, causes of conflicting results and suggestions for future research. Drug Metabolism Reviews. 2016;48(1):1–26.
6.
Grimstein M, Huang SM. A regulatory science viewpoint on botanical–drug interactions. Journal of Food and Drug Analysis. 2018;26(2):S12–25.
Bertram G, Susan B, Anthony J. Basic and ClinicalPharmacology, 11 th ed, Chapter 52, Antiprotozoal Drugs. In: The McGraw-Hill Companies, Inc. 2015. p. 56-71,.
9.
Garza AZ, Park SB, Elimination KRD. 2023;
10.
Sharma V, Madaan R, Bala R, Goyal A, K. Sindhu R. PHARMACODYNAMIC AND PHARMACOKINETIC INTERACTIONS OF HERBS WITH PRESCRIBED DRUGS: A REVIEW. PLANT ARCHIVES. 2021;21(Suppliment-1):185–98.
11.
Xie Y, Wang C. Herb–drug interactions between Panax notoginseng or its biologically active compounds and therapeutic drugs: A comprehensive pharmacodynamic and pharmacokinetic review. Journal of Ethnopharmacology. 2023;307:116156.
12.
Ulbricht C, Chao W, Costa D, Rusie-Seamon E, Weissner W, Woods J. Clinical Evidence of Herb-Drug Interactions: A Systematic Review by the Natural Standard Research Collaboration. Current Drug Metabolism. 2008;9(10):1063–120.
13.
Magdalou J, Fournel-Gigleux S, Testa B, Ouzzine M. BIOTRANSFORMATION REACTIONS. The Practice of Medicinal Chemistry. 2003. p. 517–43.
14.
Gan J, Ma S, Zhang D. Non-cytochrome P450-mediated bioactivation and its toxicological relevance. Drug Metabolism Reviews. 2016;48(4):473–501.
15.
Mazzari ALDA, Prieto JM. Herbal medicines in Brazil: pharmacokinetic profile and potential herb-drug interactions. Frontiers in Pharmacology. 5.
16.
Fasinu PS, Bouic PJ, Rosenkranz B. An Overview of the Evidence and Mechanisms of Herb–Drug Interactions. Frontiers in Pharmacology. 3.
17.
Esteves F, Rueff J, Kranendonk M. The Central Role of Cytochrome P450 in Xenobiotic Metabolism—A Brief Review on a Fascinating Enzyme Family. Journal of Xenobiotics. 11(3):94–114.
18.
Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacology & Therapeutics. 2013;138(1):103–41.
19.
Nelson DR. Cytochrome P450 Nomenclature, 2004. Cytochrome P450 Protocols. p. 1–10.
20.
Nebert DW, Dalton TP. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nature Reviews Cancer. 2006;6(12):947–60.
21.
Zhang F, Huang J, He RJ, Wang L, Huo PC, Guan XQ, et al. Herb-drug interaction between Styrax and warfarin: Molecular basis and mechanism. Phytomedicine. 2020;77:153287.
22.
Yim D, Kim MJ, Shin Y, Lee SJ, Shin JG, Kim DH. Inhibition of Cytochrome P450 Activities by Sophora flavescens Extract and Its Prenylated Flavonoids in Human Liver Microsomes. Evidence-Based Complementary and Alternative Medicine. 2019;2019:1–10.
23.
Chen P, Li D, Chen Y, Sun J, Fu K, Guan L, et al. p53‐mediated regulation of bile acid disposition attenuates cholic acid‐induced cholestasis in mice. British Journal of Pharmacology. 2017;174(23):4345–61.
24.
Sahu R, Ahmed T, Sangana R, Punde R, Subudhi BB. Effect of Tinospora cordifolia aqua-alcoholic extract on pharmacokinetic of Glibenclamide in rat: An herb-drug interaction study. Journal of Pharmaceutical and Biomedical Analysis. 2018;151:310–6.
25.
Zhu J, Wang P, Li F, Lu J, Shehu AI, Xie W, et al. CYP1A1 and 1B1-mediated metabolic pathways of dolutegravir, an HIV integrase inhibitor. Biochemical Pharmacology. 2018;158:174–84.
26.
Edeogu CO, Kalu ME, Famurewa AC, Asogwa NT, Onyeji GN, Ikpemo KO. Nephroprotective Effect of Moringa Oleifera Seed Oil on Gentamicin-Induced Nephrotoxicity in Rats: Biochemical Evaluation of Antioxidant, Anti-inflammatory, and Antiapoptotic Pathways. Journal of the American College of Nutrition. 2020;39(4):307–15.
27.
Yang J, He MM, Niu W, Wrighton SA, Li L, Liu Y, et al. Metabolic capabilities of cytochrome P450 enzymes in Chinese liver microsomes compared with those in Caucasian liver microsomes. British Journal of Clinical Pharmacology. 2012;73(2):268–84.
28.
Miyajima A, Sakemi-Hoshikawa K, Usami M, Mitsunaga K, Irie T, Ohno Y, et al. Thyrotoxic rubber antioxidants, 2-mercaptobenzimidazole and its methyl derivatives, cause both inhibition and induction of drug-metabolizing activity in rat liver microsomes after repeated oral administration. Biochemical and Biophysical Research Communications. 2017;492(1):116–20.
29.
Casey Laizure S, Herring V, Hu Z, Witbrodt K, Parker RB. The Role of Human Carboxylesterases in Drug Metabolism: Have We Overlooked Their Importance? Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy. 2013;33(2):210–22.
30.
Li YG, Hou J, Li SY, Lv X, Ning J, Wang P, et al. Fructus Psoraleae contains natural compounds with potent inhibitory effects towards human carboxylesterase 2. Fitoterapia. 2015;101:99–106.
31.
Wang D, Zou L, Jin Q, Hou J, Ge G, Yang L. Human carboxylesterases: a comprehensive review. Acta Pharmaceutica Sinica B. 2018;8(5):699–712.
32.
Sun DX, Ge GB, Dong PP, Cao YF, Fu ZW, Ran RX, et al. Inhibition behavior of fructus psoraleae’s ingredients towards human carboxylesterase 1 (hCES1). Xenobiotica. 2016;46(6):503–10.
33.
Nguyen K, DeSieno MA, Bae B, Johannes TW, Cobb RE, Zhao H, et al. Characterization of the flavin monooxygenase involved in biosynthesis of the antimalarial FR-900098. Organic & Biomolecular Chemistry. 17(6):1506–18.
34.
Di Paolo ML, Cozza G, Milelli A, Zonta F, Sarno S, Minniti E, et al. Benextramine and derivatives as novel human monoamine oxidases inhibitors: an integrated approach. The FEBS Journal. 2019;286(24):4995–5015.
35.
Aydin T, Akincioglu H, Gumustas M, Gulcin I, Kazaz C, Cakir A. human monoamine oxidase (hMAO) A and hMAO B inhibitors from Artemisia dracunculus L. herniarin and skimmin: human mononamine oxidase A and B inhibitors from A. dracunculus L. Zeitschrift für Naturforschung C. 2020;75(11–12):459–66.
36.
Parvez M, Kaisar N, Shin J. Role of Drug Metabolic Enzymes and Transporters in Drug-Drug Interactions Between Antiretroviral and Antituberculosis Drugs. Journal of Advanced Biotechnology and Experimental Therapeutics. 2018;1(1):17.
37.
Lv X, Zhang J, Hou J, Dou T, Ge G, Hu W, et al. Chemical Probes for Human UDP‐Glucuronosyltransferases: A Comprehensive Review. Biotechnology Journal. 2019;14(1).
38.
Mano ECC, Scott AL, Honorio KM. UDP-glucuronosyltransferases: Structure, Function and Drug Design Studies. Current Medicinal Chemistry. 2018;25(27):3247–55.
39.
Iyanagi T. Molecular Mechanism of Phase I and Phase II Drug‐Metabolizing Enzymes: Implications for Detoxification. International Review of Cytology. 2007. p. 35–112.
40.
Qi C, Fu J, Zhao H, Xing H, Dong D, Wu B. Identification of UGTs and BCRP as potential pharmacokinetic determinants of the natural flavonoid alpinetin. Xenobiotica. 2019;49(3):276–83.
41.
Pettersson Bergstrand M, Richter LHJ, Maurer HH, Wagmann L, Meyer MR. In vitro glucuronidation of designer benzodiazepines by human UDP‐glucuronyltransferases. Drug Testing and Analysis. 2019;11(1):45–50.
42.
Zhou X, Zhao Y, Wang J, Wang X, Chen C, Yin D, et al. Resveratrol represses estrogen-induced mammary carcinogenesis through NRF2-UGT1A8-estrogen metabolic axis activation. Biochemical Pharmacology. 2018;155:252–63.
43.
Wu L, Chen Y, Liu H, Zhan Z, Liang Z, Zhang T, et al. Emodin-induced hepatotoxicity was exacerbated by probenecid through inhibiting UGTs and MRP2. Toxicology and Applied Pharmacology. 2018;359:91–101.
44.
Li Y, Meng Q, Yang M, Liu D, Hou X, Tang L, et al. Current trends in drug metabolism and pharmacokinetics. Acta Pharmaceutica Sinica B. 2019;9(6):1113–44.
45.
Čolić A, Alessandrini M, Pepper MS. Pharmacogenetics of CYP2B6, CYP2A6 and UGT2B7 in HIV treatment in African populations: focus on efavirenz and nevirapine. Drug Metabolism Reviews. 2015;47(2):111–23.
46.
Suh HJ, Yoon SH, Yu KS, Cho JY, Park SI, Lee E, et al. The Genetic Polymorphism UGT1A4*3 Is Associated with Low Posaconazole Plasma Concentrations in Hematological Malignancy Patients Receiving the Oral Suspension. Antimicrobial Agents and Chemotherapy. 2018;62(7).
47.
Kim JH, Dahms HU, Rhee JS, Lee YM, Lee J, Han KN, et al. Expression profiles of seven glutathione S-transferase (GST) genes in cadmium-exposed river pufferfish (Takifugu obscurus). Comparative Biochemistry and Physiology Part C: Toxicology & Pharmacology. 2010;151(1):99–106.
48.
Yang Y, Huycke MM, Herman TS, Wang X. Glutathione S-transferase alpha 4 induction by activator protein 1 in colorectal cancer. Oncogene. 2016;35(44):5795–806.
49.
Jeong SJ, Park JG, Kim S, Kweon HY, Seo S, Na DS, et al. Extract of Rhus verniciflua stokes protects the diet-induced hyperlipidemia in mice. Archives of Pharmacal Research. 2015;38(11):2049–58.
50.
Tolson AH, Wang H. Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CAR. Advanced Drug Delivery Reviews. 2010;62(13):1238–49.
51.
Kalthoff S, Winkler A, Freiberg N, Manns MP, Strassburg CP. Gender matters: Estrogen receptor alpha (ERα) and histone deacetylase (HDAC) 1 and 2 control the gender-specific transcriptional regulation of human uridine diphosphate glucuronosyltransferases genes (UGT1A). Journal of Hepatology. 2013;59(4):797–804.
52.
Zhang Y, Zhang Y, Sun K, Meng Z, Chen L. The SLC transporter in nutrient and metabolic sensing, regulation, and drug development. Journal of Molecular Cell Biology. 2019;11(1):1–13.
Lopes-Rodrigues V, Seca H, Sousa D, Sousa E, Lima RT, Vasconcelos MH. The network of P-glycoprotein and microRNAs interactions. International Journal of Cancer. 2014;135(2):253–63.
55.
Wang X, Zhang H, Chen X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resistance. 2019;
56.
Lai Y. Membrane transporters and the diseases corresponding to functional defects. Transporters in Drug Discovery and Development. 2013. p. 1–146.
57.
Tsai HH, Lin HW, Simon Pickard A, Tsai HY, Mahady GB. Evaluation of documented drug interactions and contraindications associated with herbs and dietary supplements: a systematic literature review. International Journal of Clinical Practice. 2012;66(11):1056–78.
58.
Sun C, Tsai T. Pharmacokinetic and pharmacodynamic herb–drug interactions of common over‐the‐counter pain medications. Biomedical Chromatography. 2023;37(7).
59.
Müller AC, Kanfer I. Potential pharmacokinetic interactions between antiretrovirals and medicinal plants used as complementary and African traditional medicines. Biopharmaceutics & Drug Disposition. 2011;32(8):458–70.
60.
Liu N, Chen P, Du X, Sun J, Han S. In vitro inhibitory effect of obtusofolin on the activity of CYP3A4, 2C9, and 2E1. BMC Complementary Medicine and Therapies. 2021;21(1).
61.
Wei Y, Tang C, Sant V, Li S, Poloyac SM, Xie W. A Molecular Aspect in the Regulation of Drug Metabolism: Does PXR-Induced Enzyme Expression Always Lead to Functional Changes in Drug Metabolism? Current Pharmacology Reports. 2016;2(4):187–92.
62.
Rombolà L, Scuteri D, Marilisa S, Watanabe C, Morrone LA, Bagetta G, et al. Pharmacokinetic Interactions between Herbal Medicines and Drugs: Their Mechanisms and Clinical Relevance. Life. 10(7):106.
63.
Liu S, Wang Z, Hou L, Tian X, Zhang X, Cai W. Predicting the effect of tea polyphenols on ticagrelor by incorporating transporter-enzyme interplay mechanism. Chemico-Biological Interactions. 2020;330:109228.
64.
Kumar S, Bouic PJ, Rosenkranz B. In Vitro Assessment of the Interaction Potential of Ocimum basilicum (L.) Extracts on CYP2B6, 3A4, and Rifampicin Metabolism. Frontiers in Pharmacology. 11.
65.
Zhang F, Huang J, Liu W, Wang CR, Liu YF, Tu DZ, et al. Inhibition of drug-metabolizing enzymes by Qingfei Paidu decoction: Implication of herb-drug interactions in COVID-19 pharmacotherapy. Food and Chemical Toxicology. 2021;149:111998.
66.
Mohamed MEF, Tseng T, Frye RF. Inhibitory effects of commonly used herbal extracts on UGT1A1 enzyme activity. Xenobiotica. 2010;40(10):663–9.
67.
Brantley SJ, Argikar AA, Lin YS, Nagar S, Paine MF. Herb–Drug Interactions: Challenges and Opportunities for Improved Predictions. Drug Metabolism and Disposition. 2014;42(3):301–17.
68.
Fimognari C, Berti F, Cantelli-Forti G, Hrelia P. Effect of sulforaphane on micronucleus induction in cultured human lymphocytes by four different mutagens. Environmental and Molecular Mutagenesis. 2005;46(4):260–7.
69.
Prakash C, Zuniga B, Seog Song C, Jiang S, Cropper J, Park S, et al. Nuclear Receptors in Drug Metabolism, Drug Response and Drug Interactions. Nuclear Receptor Research. 2015;2.
70.
Yan R, Yang Y, Chen Y. Pharmacokinetics of Chinese medicines: strategies and perspectives. Chinese Medicine. 2018;13(1).
71.
Markowitz JS. Effect of St John’s Wort on Drug Metabolism by Induction of Cytochrome P450 3A4 Enzyme. JAMA. 2003;290(11):1500.
72.
Hermann R, von Richter O. Clinical Evidence of Herbal Drugs As Perpetrators of Pharmacokinetic Drug Interactions. Planta Medica. 78(13):1458–77.
73.
Gufford B, Barr J, González‐Pérez V, Layton M, White J, Oberlies N, et al. Quantitative prediction and clinical evaluation of an unexplored herb–drug interaction mechanism in healthy volunteers. CPT: Pharmacometrics & Systems Pharmacology. 2015;4(12):701–10.
74.
Brantley S, Gufford B, Dua R, Fediuk D, Graf T, Scarlett Y, et al. Physiologically Based Pharmacokinetic Modeling Framework for Quantitative Prediction of an Herb–Drug Interaction. CPT: Pharmacometrics & Systems Pharmacology. 2014;3(3):1–9.
75.
F.D.A. In vitro drug interaction studiescytochrome P450 enzyme-and transportermediated drug interactions guidance for industry. 2020;
76.
Hakkola J, Hukkanen J, Turpeinen M, Pelkonen O. Inhibition and induction of CYP enzymes in humans: an update. Archives of Toxicology. 2020;94(11):3671–722.
77.
Adler S, Basketter D, Creton S, Pelkonen O, van Benthem J, Zuang V, et al. Alternative (non-animal) methods for cosmetics testing: current status and future prospects—2010. Archives of Toxicology. 2011;85(5):367–485.
78.
Bernasconi C, Pelkonen O, Andersson TB, Strickland J, Wilk-Zasadna I, Asturiol D, et al. Validation of in vitro methods for human cytochrome P450 enzyme induction: Outcome of a multi-laboratory study. Toxicology in Vitro. 2019;60:212–28.
79.
E.M.A. Guideline on the investigation of drug interactions. 2012;
80.
Goey AKL, Mooiman KD, Beijnen JH, Schellens JHM, Meijerman I. Relevance of in vitro and clinical data for predicting CYP3A4-mediated herb–drug interactions in cancer patients. Cancer Treatment Reviews. 2013;39(7):773–83.
81.
Tarek M, Elzanfaly ES, Amer SM, Wagdy HA. Selective analysis of Nadifloxacin in human plasma samples using a molecularly imprinted polymer-based solid-phase extraction proceeded by UPLC-DAD analysis. Microchemical Journal. 2020;158:105162.
82.
S. Fasinu P, J. Gurley B, A. Walker L. Clinically Relevant Pharmacokinetic Herb-drug Interactions in Antiretroviral Therapy. Current Drug Metabolism. 2015;17(1):52–64.
83.
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 Pharmaceutica Sinica B. 2021;11(10):2973–82.
84.
Zhang D, Luo G, Ding X, Lu C. Preclinical experimental models of drug metabolism and disposition in drug discovery and development. Acta Pharmaceutica Sinica B. 2012;2(6):549–61.
85.
Wei Y, Yang L, Zhang X, Sui D, Wang C, Wang K, et al. Generation and Characterization of a CYP2C11-Null Rat Model by Using the CRISPR/Cas9 Method. Drug Metabolism and Disposition. 2018;46(5):525–31.
86.
Satoh D, Abe S, Kobayashi K, Nakajima Y, Oshimura M, Kazuki Y. Human and mouse artificial chromosome technologies for studies of pharmacokinetics and toxicokinetics. Drug Metabolism and Pharmacokinetics. 2018;33(1):17–30.
87.
Liu Z, Li L, Wu H, Hu J, Ma J, Zhang QY, et al. Characterization of CYP2B6 in a CYP2B6-Humanized Mouse Model: Inducibility in the Liver by Phenobarbital and Dexamethasone and Role in Nicotine Metabolism In Vivo. Drug Metabolism and Disposition. 2015;43(2):208–16.
88.
Scheer N, Kapelyukh Y, McEwan J, Beuger V, Stanley LA, Rode A, et al. Modeling Human Cytochrome P450 2D6 Metabolism and Drug-Drug Interaction by a Novel Panel of Knockout and Humanized Mouse Lines. Molecular Pharmacology. 2012;81(1):63–72.
89.
Hasegawa M, Kapelyukh Y, Tahara H, Seibler J, Rode A, Krueger S, et al. Quantitative Prediction of Human Pregnane X Receptor and Cytochrome P450 3A4 Mediated Drug-Drug Interaction in a Novel Multiple Humanized Mouse Line. Molecular Pharmacology. 2011;80(3):518–28.
90.
Kumar R, Mota LC, Litoff EJ, Rooney JP, Boswell WT, Courter E, et al. Compensatory changes in CYP expression in three different toxicology mouse models: CAR-null, Cyp3a-null, and Cyp2b9/10/13-null mice. PLOS ONE. 12(3):e0174355.
91.
Gurley BJ, Markowitz JS, Williams DK, Barone GW. Practical Considerations When Designing and Conducting Clinical Pharmacokinetic Herb–Drug Interaction Studies. International Journal of Pharmacokinetics. 2017;2(1):57–69.
92.
Gurley B. Pharmacokinetic Herb-Drug Interactions (Part 1): Origins, Mechanisms, and the Impact of Botanical Dietary Supplements. Planta Medica. 78(13):1478–89.
93.
Zhang D, Luo G, Ding X, Lu C. Preclinical experimental models of drug metabolism and disposition in drug discovery and development. Acta Pharmaceutica Sinica B. 2012;2(6):549–61.
94.
Nduka SO, Okonta MJ, Ajaghaku DL, Amorha KC, Ukwe CV. Inhibition of cytochrome P450 3A enzyme by Millettia aboensis : its effect on the pharmacokinetic properties of efavirenz and nevirapine. Revista Brasileira de Farmacognosia. 2017;27(2):228–35.
95.
Hutter M. In Silico Prediction of Drug Properties. Current Medicinal Chemistry. 2009;16(2):189–202.
96.
Borse SP, Singh DP, Nivsarkar M. Understanding the relevance of herb–drug interaction studies with special focus on interplays: a prerequisite for integrative medicine. Porto Biomedical Journal. 2019;4(2):e15.
97.
Uma KV, Martin JV, Sutheeswaran G, Rajeshkumar R, Ponnusankar S. Molecular docking and in-silico predictive analysis of potential herb-drug interactions between Momordica charantia and Empagliflozin. Journal of Applied Pharmaceutical Science.
98.
Brown AC. An overview of herb and dietary supplement efficacy, safety and government regulations in the United States with suggested improvements. Part 1 of 5 series. Food and Chemical Toxicology. 2017;107:449–71.
99.
Ge B, Zhang Z, Zuo Z. Updates on the Clinical Evidenced Herb‐Warfarin Interactions. Evidence-Based Complementary and Alternative Medicine. 2014;2014(1).
100.
Kahma H, Aurinsalo L, Neuvonen M, Katajamäki J, Paludetto MN, Viinamäki J, et al. An automated cocktail method for in vitro assessment of direct and time-dependent inhibition of nine major cytochrome P450 enzymes – application to establishing CYP2C8 inhibitor selectivity. European Journal of Pharmaceutical Sciences. 2021;162:105810.
101.
Gouws C, Steyn D, Du Plessis L, Steenekamp J, Hamman JH. Combination therapy of Western drugs and herbal medicines: recent advances in understanding interactions involving metabolism and efflux. Expert Opinion on Drug Metabolism & Toxicology. 2012;8(8):973–84.
102.
Min JS, Bae SK. Prediction of drug–drug interaction potential using physiologically based pharmacokinetic modeling. Archives of Pharmacal Research. 2017;40(12):1356–79.
103.
Zhang H, Bu F, Li L, Jiao Z, Ma G, Cai W, et al. Prediction of Drug–Drug Interaction between Tacrolimus and Principal Ingredients of Wuzhi Capsule in Chinese Healthy Volunteers Using Physiologically‐Based Pharmacokinetic Modelling. Basic & Clinical Pharmacology & Toxicology. 2018;122(3):331–40.
104.
Palleria C, Paolo A, Giofrè C. Pharmacokinetic drug-drug interaction and their implication in clinical management. J Res Med Sci. 2013;18(7):601–10.
105.
Awortwe C, Makiwane M, Reuter H, Muller C, Louw J, Rosenkranz B. Critical evaluation of causality assessment of herb–drug interactions in patients. British Journal of Clinical Pharmacology. 2018;84(4):679–93.
106.
Qiu Z, Qiao Y, Zhang B, Sun‐Waterhouse D, Zheng Z. Bioactive polysaccharides and oligosaccharides from garlic ( Allium sativum L.): Production, physicochemical and biological properties, and structure–function relationships. Comprehensive Reviews in Food Science and Food Safety. 2022;21(4):3033–95.
107.
Rouf R, Uddin SJ, Sarker DK, Islam MT, Ali ES, Shilpi JA, et al. Antiviral potential of garlic (Allium sativum) and its organosulfur compounds: A systematic update of pre-clinical and clinical data. Trends in Food Science & Technology. 2020;104:219–34.
108.
Bhatwalkar SB, Mondal R, Krishna SBN, Adam JK, Govender P, Anupam R. Antibacterial Properties of Organosulfur Compounds of Garlic (Allium sativum). Frontiers in Microbiology. 12.
109.
Tudu CK, Dutta T, Ghorai M, Biswas P, Samanta D, Oleksak P, et al. Traditional uses, phytochemistry, pharmacology and toxicology of garlic (Allium sativum), a storehouse of diverse phytochemicals: A review of research from the last decade focusing on health and nutritional implications. Frontiers in Nutrition. 9.
110.
Markowitz J. Effects of garlic (Allium sativum L.) supplementation on cytochrome P450 2D6 and 3A4 activity in healthy volunteers. Clinical Pharmacology & Therapeutics. 2003;74(2):170–7.
111.
Izzo AA, Ernst E. Interactions Between Herbal Medicines and Prescribed Drugs. Drugs. 2009;69(13):1777–98.
112.
Piscitelli SC, Burstein AH, Welden N, Gallicano KD, Falloon J. The Effect of Garlic Supplements on the Pharmacokinetics of Saquinavir. Clinical Infectious Diseases. 2002;34(2):234–8.
113.
Chin AC, Baskin LB. Effect of Herbal Supplement–Drug Interactions on Therapeutic Drug Monitoring. Therapeutic Drug Monitoring. 2012. p. 417–45.
114.
Kaye AD, Baluch A, Kaye AM. Mineral, Vitamin, and Herbal Supplements. Anesthesia and Uncommon Diseases. 2012. p. 470–87.
115.
Moltó J, Valle M, Miranda C, Cedeño S, Negredo E, Barbanoj MJ, et al. Herb-Drug Interaction betweenEchinacea purpureaand Darunavir-Ritonavir in HIV-Infected Patients. Antimicrobial Agents and Chemotherapy. 2011;55(1):326–30.
116.
Noor-E-Tabassum, Das R, Lami MS, Chakraborty AJ, Mitra S, Tallei TE, et al. Ginkgo biloba: A Treasure of Functional Phytochemicals with Multimedicinal Applications. Evidence-Based Complementary and Alternative Medicine. 2022;2022:1–30.
117.
Al Mahmud Z, Emran TB, Qais N, Bachar SC, Sarker M, Nasir Uddin MM. Evaluation of analgesic, anti-inflammatory, thrombolytic and hepatoprotective activities of roots ofPremna esculenta(Roxb). Journal of Basic and Clinical Physiology and Pharmacology. 2016;27(1):63–70.
118.
Mohanta TK, Tamboli Y, Zubaidha PK. Phytochemical and medicinal importance ofGinkgo bilobaL. Natural Product Research. 2014;28(10):746–52.
119.
Surana AR, Agrawal SP, Kumbhare MR, Gaikwad SB. Current perspectives in herbal and conventional drug interactions based on clinical manifestations. Future Journal of Pharmaceutical Sciences. 2021;7(1).
120.
Yoshioka M, Ohnishi N, Sone N, Egami S, Takara K, Yokoyama T, et al. Studies on Interactions between Functional Foods or Dietary Supplements and Medicines. III. Effects of Ginkgo biloba Leaf Extract on the Pharmacokinetics of Nifedipine in Rats. Biological and Pharmaceutical Bulletin. 2004;27(12):2042–5.
121.
Yin OQ, Tomlinson B, Waye MM, Chow AH, Chow MS. Pharmacogenetics and herb???drug interactions. Pharmacogenetics. 2004;14(12):841–50.
122.
Kahraman C, Ceren Arituluk Z, Irem Tatli Cankaya I. The Clinical Importance of Herb-Drug Interactions and Toxicological Risks of Plants and Herbal Products. Medical Toxicology.
123.
Kupiec T, Raj V. Fatal Seizures Due to Potential Herb-Drug Interactions with Ginkgo Biloba. Journal of Analytical Toxicology. 2005;29(7):755–8.
124.
Shi S, Klotz U. Drug Interactions with Herbal Medicines. Clinical Pharmacokinetics. 2012;51(2):77–104.
125.
Bian T, Corral P, Wang Y, Botello J, Kingston R, Daniels T, et al. Kava as a Clinical Nutrient: Promises and Challenges. Nutrients. 12(10):3044.
126.
Olsen LR, Grillo MP, Skonberg C. Constituents in Kava Extracts Potentially Involved in Hepatotoxicity: A Review. Chemical Research in Toxicology. 2011;24(7):992–1002.
127.
Pluskal T, Torrens-Spence MP, Fallon TR, De Abreu A, Shi CH, Weng JK. The biosynthetic origin of psychoactive kavalactones in kava. Nature Plants. 5(8):867–78.
128.
GURLEY B, GARDNER S, HUBBARD M, WILLIAMS D, GENTRY W, KHAN I, et al. In vivo effects of goldenseal, kava kava, black cohosh, and valerian on human cytochrome P450 1A2, 2D6, 2E1, and 3A4/5 phenotypes. Clinical Pharmacology & Therapeutics. 2005;77(5):415–26.
129.
Gurley BJ, Swain A, Hubbard MA, Williams DK, Barone G, Hartsfield F, et al. Clinical assessment of CYP2D6‐mediated herb–drug interactions in humans: Effects of milk thistle, black cohosh, goldenseal, kava kava, St. John’s wort, andEchinacea. Molecular Nutrition & Food Research. 2008;52(7):755–63.
130.
Karimi G, Vahabzadeh M, Lari P. Silymarin", a promising pharmacological agent for treatment of diseases. Iran J Basic Med Sci. 2011;14(4):308–17.
131.
Bijak M. Silybin, a Major Bioactive Component of Milk Thistle (Silybum marianum L. Gaernt.)—Chemistry, Bioavailability, and Metabolism. Molecules. 22(11):1942.
132.
Rajnarayana K, Reddy M, Vidyasagar J, Krishna D. Study on the Influence of Silymarin Pretreatment on Metabolism and Disposition of Metronidazole. Arzneimittelforschung. 54(02):109–13.
133.
Xie Y, Zhang D, Zhang J, Yuan J. Metabolism, Transport and Drug–Drug Interactions of Silymarin. Molecules. 24(20):3693.
134.
Han Y, Guo D, Chen Y, Tan ZR, Zhou HH. Effect of continuous silymarin administration on oral talinolol pharmacokinetics in healthy volunteers. Xenobiotica. 2009;39(9):694–9.
135.
Mills E, Wilson K, Clarke M, Foster B, Walker S, Rachlis B, et al. Milk thistle and indinavir: a randomized controlled pharmacokinetics study and meta-analysis. European Journal of Clinical Pharmacology. 2005;61(1):1–7.
136.
Gurley B, Hubbard MA, Williams DK, Thaden J, Tong Y, Gentry WB, et al. Assessing the Clinical Significance of Botanical Supplementation on Human Cytochrome P450 3A Activity: Comparison of a Milk Thistle and Black Cohosh Product to Rifampin and Clarithromycin. The Journal of Clinical Pharmacology. 2006;46(2):201–13.
137.
Kawaguchi-Suzuki M, Frye RF, Zhu HJ, Brinda BJ, Chavin KD, Bernstein HJ, et al. The Effects of Milk Thistle (Silybum marianum) on Human Cytochrome P450 Activity. Drug Metabolism and Disposition. 2014;42(10):1611–6.
138.
Kawaguchi-Suzuki M, Frye RF, Zhu HJ, Brinda BJ, Chavin KD, Bernstein HJ, et al. The Effects of Milk Thistle (Silybum marianum) on Human Cytochrome P450 Activity. Drug Metabolism and Disposition. 2014;42(10):1611–6.
139.
Ratan ZA, Haidere MF, Hong YH, Park SH, Lee JO, Lee J, et al. Pharmacological potential of ginseng and its major component ginsenosides. Journal of Ginseng Research. 2021;45(2):199–210.
140.
Li KK, Li SS, Xu F, Gong XJ. Six new dammarane-type triterpene saponins from Panax ginseng flower buds and their cytotoxicity. Journal of Ginseng Research. 2020;44(2):215–21.
141.
Yang L, Wang Y, Xu H, Huang G, Zhang Z, Ma Z, et al. Panax ginseng Inhibits Metabolism of Diester Alkaloids by Downregulating CYP3A4 Enzyme Activity via the Pregnane X Receptor. Evidence-Based Complementary and Alternative Medicine. 2019;2019:1–13.
142.
Zhang R, Jie J, Zhou Y, Cao Z, Li W. Long-Term Effects of Panax Ginseng on Disposition of Fexofenadine in Ratsin vivo. The American Journal of Chinese Medicine. 2009;37(04):657–67.
143.
Choi MK, Song IS. Interactions of ginseng with therapeutic drugs. Archives of Pharmacal Research. 2019;42(10):862–78.
144.
Markham MJ, Dog TL. Dietary Supplements and Hemostasis. Consultative Hemostasis and Thrombosis. 2013. p. 595–600.
145.
El Hamdaoui Y, Zheng F, Fritz N, Ye L, Tran MA, Schwickert K, et al. Analysis of hyperforin (St. John’s wort) action at TRPC6 channel leads to the development of a new class of antidepressant drugs. Molecular Psychiatry. 2022;27(12):5070–85.
146.
Vacek J, Klejdus B, Kubán V. Hypericin a hyperforin: biologicky aktivní komponenty trezalky teckované (Hypericum perforatum). Jejich izolace, analýza a studium fyziologických úcinků [Hypericin and hyperforin: bioactive components of St. John’s Wort (Hypericum perforatum. Their isolation, analysis and study of physiological effect] Ceska Slov Farm. 2007;56(2):62–6.
147.
Alahmad A, Alghoraibi I, Zein R, Kraft S, Dräger G, Walter JG, et al. Identification of Major Constituents of Hypericum perforatum L. Extracts in Syria by Development of a Rapid, Simple, and Reproducible HPLC-ESI-Q-TOF MS Analysis and Their Antioxidant Activities. ACS Omega. 2022;7(16):13475–93.
148.
Murphy PA. ST. JOHN’S WORT AND ORAL CONTRACEPTIVES: REASONS FOR CONCERN? Journal of Midwifery & Women’s Health. 2002;47(6):447–50.
149.
Hall S. The interaction between St John’s wort and an oral contraceptive. Clinical Pharmacology & Therapeutics. 2003;74(6):525–35.
150.
Wanwimolruk S, Prachayasittikul V. Cytochrome P450 enzyme mediated herbal drug interactions (Part 1. EXCLI J. 2014;13:347–91.
151.
Igbinoba SI, Onyeji CO, Akanmu MA, Soyinka JO, Pullela SSVV, Cook JM, et al. Effect of dehuskedGarcinia kolaseeds on the overall pharmacokinetics of quinine in healthy Nigerian volunteers. The Journal of Clinical Pharmacology. 2015;55(3):348–54.
152.
Akinleye M, Amaeze O, Opeodu O, Okubanjo O. Effect of Ciklavit® - a Nigerian Poly-herbal Formulation on the Dissolution Profile of Proguanil Tablets: Potential for Herb-drug Interaction. British Journal of Pharmaceutical Research. 12(6):1–9.
153.
Odunke NS, Eleje O, Christiana AC, Peter IC, Uchenna E. Herb drug interaction: effect of Manix® on pharmacokinetic parameters of pefloxacin in rat model. Asian Pacific Journal of Tropical Biomedicine. 2014;4:S413–6.
154.
Olawoye O, Adeagbo B, Bolaji O. Effects of Moringa oleifera Leaf Powder Suspension on the Pharmacokinetics of Amodiaquine in Rats. Journal of Complementary and Alternative Medical Research. 3(4):1–8.
155.
Olawoye OS, Adeagbo BA, Bolaji OO. Moringa oleifera leaf powder alters the pharmacokinetics of amodiaquine in healthy human volunteers. Journal of Clinical Pharmacy and Therapeutics. 2018;43(5):626–32.
156.
Adepiti AO, Adeagbo BA, Adehin A, Bolaji OO, Elujoba AA. Influence of MAMA decoction, an Herbal Antimalarial, on the Pharmacokinetics of Amodiaquine in Mice. European Journal of Drug Metabolism and Pharmacokinetics. 2020;45(1):81–8.
157.
Kolade YT, Babalola CP, Olaniyi AA, Scriba GKE. Effect of kolanut on the pharmacokinetics of the antimalarial drug halofantrine. European Journal of Clinical Pharmacology. 2008;64(1):77–81.
158.
Nwafor SV, Akah PA, Okoli CO. Interaction between chloroquine sulphate and aqueous extract of Azadirachta indica A. Juss (Meliaceae) in rabbits. Acta Pharm. 2003;53(4):305–11.
The statements, opinions and data contained in the journal are solely those of the individual authors and contributors and not of the publisher and the editor(s). We stay neutral with regard to jurisdictional claims in published maps and institutional affiliations.