Skip to main content

Advertisement

Log in

Interaction between phytotherapy and oral anticancer agents: prospective study and literature review

  • Review Article
  • Published:
Medical Oncology Aims and scope Submit manuscript

Abstract

Cancer is becoming more prevalent in elderly patient. Due to polypharmacy, older adults with cancer are predisposed to drug-drug interactions. There is also an increasing interest in the use of complementary and alternative medicine (CAM). Thirty to seventy percent of patients with cancer have used CAM. Through pharmaceutical counseling sessions, we can provide advices on herb–drug interactions (HDI). All the patients seen in pharmaceutical counseling sessions were prospectively included. Information was collected during these sessions: prescribed medication (oral anticancer agents (OAA) and other drugs), CAM (phytotherapy especially), and use of over-the-counter (OTC) drugs. If pharmacist considered an interaction or an intervention clinically relevant, the oncologist was notified. Then, a literature review was realized to identify the potential HDI (no interactions, precautions for use, contraindication). Among 201 pharmacist counseling sessions, it resulted in 104 interventions related to 46 HDI, 28 drug-drug interactions and 30 others (wrong dosage, omission…). To determine HDI, we review 73 medicinal plants which are used by our patients with cancer and 31 OAA. A total of 1829 recommendations were formulated about 59 (75%) medical plants and their interaction with an OAA. Herb–drug interactions should not be ignored by healthcare providers in their management of cancer patients in daily practice.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Maher RL, Hanlon JT, Hajjar ER. Clinical consequences of polypharmacy in elderly. Expert Opin Drug Saf. 2014;13:57–65. https://doi.org/10.1517/14740338.2013.827660.

    Article  PubMed  Google Scholar 

  2. Voll ML, Yap KD, Terpstra WE, et al. Potential drug-drug interactions between anti-cancer agents and community pharmacy dispensed drugs. Pharm World Sci. 2010;32:575–80.

    Article  CAS  PubMed  Google Scholar 

  3. Van Leeuwen RWF, Jansman FGA, van den Bemt PMLA, et al. Drug-drug interactions in patients treated for cancer: a prospective study on clinical interventions. Ann Oncol. 2015;26:992–7.

    Article  PubMed  Google Scholar 

  4. van Leeuwen RWF, van Gelder T, Mathijssen RHJ, et al. Drug–drug interactions with tyrosine-kinase inhibitors: a clinical perspective. Lancet Oncol. 2014;15:e315–26.

    Article  PubMed  CAS  Google Scholar 

  5. Van Leeuwen RWF, Brundel DHS, Neef C, et al. Prevalence of potential drug-drug interactions in cancer patients treated with oral anticancer drugs. Br J Cancer. 2013;108:1071–8.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  6. Girre V, Arkoub H, Puts MTE, et al. Potential drug interactions in elderly cancer patients. Crit Rev Oncol/Hematol. 2011;78:220–6.

    Article  CAS  Google Scholar 

  7. Beijnen JH, Schellens JH. Drug interactions in oncology. Lancet Oncol. 2004;5:489–96.

    Article  CAS  PubMed  Google Scholar 

  8. Scripture CD, Figg WD. Drug interactions in cancer therapy. Nat Rev Cancer. 2006;6:546–58.

    Article  CAS  PubMed  Google Scholar 

  9. Hadjibabaie M, Badri S, Ataei S, et al. Potential drug-drug interactions at a referral hematology-oncology ward in Iran: a cross-sectional study. Cancer Chemother Pharmacol. 2013;71:1619–27.

    Article  CAS  PubMed  Google Scholar 

  10. Tanaka E. Clinically important pharmacokinetic drug-drug interactions: role of cytochrome P450 enzymes. J Clin Pharm Ther. 1998;23:403–16.

    Article  CAS  PubMed  Google Scholar 

  11. Segal EM, Flood MR, Mancini RS, et al. Oral chemotherapy food and drug interactions: a comprehensive review of the literature. J Oncol Pract. 2014;10:e255–68.

    Article  PubMed  Google Scholar 

  12. Thomas-Schoemann A, Blanchet B, Bardin C, et al. Drug interactions with solid tumour-targeted therapies. Crit Rev Oncol Hematol. 2014;89:179–96.

    Article  PubMed  Google Scholar 

  13. Riechelmann RP, Del Giglio A. Drug interactions in oncology: how common are they? Ann Oncol. 2009;20:1907–12.

    Article  CAS  PubMed  Google Scholar 

  14. WHO| Traditional Complementary Integrative Medicine. WHO, http://www.who.int/traditional-complementary-integrative-medicine/about/en/. Accessed 14 April 2018.

  15. Guide qualite de la prise en charge medicamenteuse, http://social-sante.gouv.fr/IMG/pdf/Guide_qualite_de_la_prise_en_charge_medicamenteuse.pdf Accessed 27 April 2017.

  16. Vincent L., Giraudier F., Le Rat P., et al. Cytotoxiques oraux : Risques iatrogènes ?—Groupe d’Evaluation et de Recherche sur la Protection en Atmosphère Contrôlée (GERPAC), http://www.gerpac.eu/spip.php?article32. 2010, Accessed 27 April 2017.

  17. Horneber M, Bueschel G, Dennert G, et al. How many cancer patients use complementary and alternative medicine: a systematic review and metaanalysis. Integr Cancer Ther. 2012;11:187–203.

    Article  PubMed  Google Scholar 

  18. Richardson MA, Sanders T, Palmer JL, et al. Complementary/alternative medicine use in a comprehensive cancer center and the implications for oncology. JCO. 2000;18:2505–14.

    Article  CAS  Google Scholar 

  19. Ernst E. How the public is being misled about complementary/alternative medicine. J R Soc Med. 2008;101:528–30.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Sparreboom A, Cox MC, Acharya MR, et al. Herbal remedies in the United States: potential adverse interactions with anticancer agents. J Clin Oncol. 2004;22:2489–503.

    Article  CAS  PubMed  Google Scholar 

  21. Izzo AA. Interactions between herbs and conventional drugs: overview of the clinical data. Med Princ Pract. 2012;21:404–28.

    Article  PubMed  Google Scholar 

  22. Posadzki P, Watson L, Ernst E. Herb–drug interactions: an overview of systematic reviews. Br J Clin Pharmacol. 2013;75:603–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gouws C, Steyn D, Plessis LD, et al. Combination therapy of Western drugs and herbal medicines: recent advances in understanding interactions involving metabolism and efflux. Exp Opin Drug Metab Toxicol. 2012;8:973–84.

    Article  CAS  Google Scholar 

  24. Ben-Arye E, Samuels N, Goldstein LH, et al. Potential risks associated with traditional herbal medicine use in cancer care: a study of Middle Eastern oncology health care professionals. Cancer. 2016;122:598–610.

    Article  PubMed  Google Scholar 

  25. Conde-Estévez D, Albanell J. Oral chemotherapy prescription safety practices in Europe. Int J Clin Pharm. 2014;36:863–4.

    Article  PubMed  Google Scholar 

  26. Lopez-Martin C, Garrido Siles M, Alcaide-Garcia J, et al. Role of clinical pharmacists to prevent drug interactions in cancer outpatients: a single-centre experience. Int J Clin Pharm. 2014;36:1251–9.

    Article  PubMed  Google Scholar 

  27. Bourmaud A, Pacaut C, Melis A, et al. Is oral chemotherapy prescription safe for patients? A cross-sectional survey. Ann Oncol. 2014;25:500–4.

    Article  CAS  PubMed  Google Scholar 

  28. Conde-Estévez D, Salas E, Albanell J. Survey of oral chemotherapy safety and adherence practices of hospitals in Spain. Int J Clin Pharm. 2013;35:1236–44.

    Article  PubMed  CAS  Google Scholar 

  29. Allenet B, Bedouch P, Rose F-X, et al. Validation of an instrument for the documentation of clinical pharmacists’ interventions. Pharm World Sci. 2006;28:181–8.

    Article  PubMed  Google Scholar 

  30. European Medicines Agency, https://www.ema.europa.eu/. Accessed 1 Jan 2019.

  31. Deb S, Chin MY, Adomat H, et al. Abiraterone inhibits 1α,25-dihydroxyvitamin D3 metabolism by CYP3A4 in human liver and intestine in vitro. J Steroid Biochem Mol Biol. 2014;144(Pt A):50–8.

    Article  CAS  PubMed  Google Scholar 

  32. Monbaliu J, Gonzalez M, Bernard A, et al. In vitro and in vivo drug-drug interaction studies to assess the effect of abiraterone acetate, abiraterone, and metabolites of abiraterone on CYP2C8 activity. Drug Metab Dispos. 2016;44:1682–91.

    Article  CAS  PubMed  Google Scholar 

  33. Bernard A, Vaccaro N, Acharya M, et al. Impact on abiraterone pharmacokinetics and safety: open-label drug-drug interaction studies with ketoconazole and rifampicin. Clin Pharmacol Drug Dev. 2015;4:63–73.

    Article  CAS  PubMed  Google Scholar 

  34. Wind S, Giessmann T, Jungnik A, et al. Pharmacokinetic drug interactions of afatinib with rifampicin and ritonavir. Clin Drug Investig. 2014;34:173–82.

    Article  CAS  PubMed  Google Scholar 

  35. Wind S, Schnell D, Ebner T, et al. Clinical pharmacokinetics and pharmacodynamics of afatinib. Clin Pharmacokinet. 2017;56:235–50.

    Article  CAS  PubMed  Google Scholar 

  36. Zhang Y, Wang C, Liu Z, et al. P-gp is involved in the intestinal absorption and biliary excretion of afatinib in vitro and in rats. Pharmacol Rep. 2018;70:243–50.

    Article  CAS  PubMed  Google Scholar 

  37. Koide H, Tsujimoto M, Takeuchi A, et al. Substrate-dependent effects of molecular-targeted anticancer agents on activity of organic anion transporting polypeptide 1B1. Xenobiotica. 2018;48:1059–71.

    Article  CAS  PubMed  Google Scholar 

  38. Stopfer P, Marzin K, Narjes H, et al. Afatinib pharmacokinetics and metabolism after oral administration to healthy male volunteers. Cancer Chemother Pharmacol. 2012;69:1051–61.

    Article  CAS  PubMed  Google Scholar 

  39. Schnell D, Buschke S, Fuchs H, et al. Pharmacokinetics of afatinib in subjects with mild or moderate hepatic impairment. Cancer Chemother Pharmacol. 2014;74:267–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chen Y, Tortorici MA, Garrett M, et al. Clinical pharmacology of axitinib. Clin Pharmacokinet. 2013;52:713–25.

    Article  CAS  PubMed  Google Scholar 

  41. Gu R, Hibbs DE, Ong JA, et al. The multikinase inhibitor axitinib is a potent inhibitor of human CYP1A2. Biochem Pharmacol. 2014;88:245–52.

    Article  CAS  PubMed  Google Scholar 

  42. Reyner EL, Sevidal S, West MA, et al. In vitro characterization of axitinib interactions with human efflux and hepatic uptake transporters: implications for disposition and drug interactions. Drug Metab Dispos. 2013;41:1575–83.

    Article  CAS  PubMed  Google Scholar 

  43. Zientek MA, Goosen TC, Tseng E, et al. In vitro kinetic characterization of axitinib metabolism. Drug Metab Dispos. 2016;44:102–14.

    Article  CAS  PubMed  Google Scholar 

  44. Nguyen L, Holland J, Miles D, et al. Pharmacokinetic (PK) drug interaction studies of cabozantinib: effect of CYP3A inducer rifampin and inhibitor ketoconazole on cabozantinib plasma PK and effect of cabozantinib on CYP2C8 probe substrate rosiglitazone plasma PK. J Clin Pharmacol. 2015;55:1012–23.

    Article  CAS  PubMed  Google Scholar 

  45. Lacy S, Hsu B, Miles D, et al. Metabolism and disposition of cabozantinib in healthy male volunteers and pharmacologic characterization of its major metabolites. Drug Metab Dispos. 2015;43:1190–207.

    Article  CAS  PubMed  Google Scholar 

  46. Lacy SA, Miles DR, Nguyen LT. Clinical pharmacokinetics and pharmacodynamics of cabozantinib. Clin Pharmacokinet. 2017;56:477–91.

    Article  PubMed  Google Scholar 

  47. Miyazaki S, Satoh H, Ikenishi M, et al. Pharmacokinetic model analysis of interaction between phenytoin and capecitabine. Int J Clin Pharmacol Ther. 2016;54:657–65.

    Article  CAS  PubMed  Google Scholar 

  48. Budha NR, Ji T, Musib L, et al. Evaluation of cytochrome P450 3A4-Mediated drug-drug interaction potential for cobimetinib using physiologically based pharmacokinetic modeling and simulation. Clin Pharmacokinet. 2016;55:1435–45.

    Article  CAS  PubMed  Google Scholar 

  49. Choo EF, Ly J, Chan J, et al. Role of P-glycoprotein on the brain penetration and brain pharmacodynamic activity of the MEK inhibitor cobimetinib. Mol Pharm. 2014;11:4199–207.

    Article  CAS  PubMed  Google Scholar 

  50. Choo EF, Woolsey S, DeMent K, et al. Use of transgenic mouse models to understand the oral disposition and drug-drug interaction potential of cobimetinib, a MEK inhibitor. Drug Metab Dispos. 2015;43:864–9.

    Article  CAS  PubMed  Google Scholar 

  51. Arakawa H, Omote S, Tamai I. Inhibitory Effect of Crizotinib on Creatinine Uptake by Renal Secretory Transporter OCT2. J Pharm Sci. 2017;106:2899–903.

    Article  CAS  PubMed  Google Scholar 

  52. Sato T, Mishima E, Mano N, et al. Potential drug interactions mediated by renal organic anion transporter OATP4C1. J Pharmacol Exp Ther. 2017;362:271–7.

    Article  CAS  PubMed  Google Scholar 

  53. Yamazaki S, Johnson TR, Smith BJ. Prediction of drug–drug interactions with crizotinib as the CYP3A substrate using a physiologically based pharmacokinetic model. Drug Metab Dispos. 2015;43:1417–29.

    Article  CAS  PubMed  Google Scholar 

  54. Hamilton G, Rath B, Burghuber O. Pharmacokinetics of crizotinib in NSCLC patients. Expert Opin Drug Metab Toxicol. 2015;11:835–42.

    Article  CAS  PubMed  Google Scholar 

  55. Xu H, O’Gorman M, Tan W, et al. The effects of ketoconazole and rifampin on the single-dose pharmacokinetics of crizotinib in healthy subjects. Eur J Clin Pharmacol. 2015;71:1441–9.

    Article  CAS  PubMed  Google Scholar 

  56. Mao J, Johnson TR, Shen Z, et al. Prediction of crizotinib-midazolam interaction using the Simcyp population-based simulator: comparison of CYP3A time-dependent inhibition between human liver microsomes versus hepatocytes. Drug Metab Dispos. 2013;41:343–52.

    Article  CAS  PubMed  Google Scholar 

  57. Sato T, Ito H, Hirata A, et al. Interactions of crizotinib and gefitinib with organic anion-transporting polypeptides (OATP)1B1, OATP1B3 and OATP2B1: gefitinib shows contradictory interaction with OATP1B3. Xenobiotica. 2018;48:73–8.

    Article  CAS  PubMed  Google Scholar 

  58. El-Serafi I, Afsharian P, Moshfegh A, et al. Cytochrome P450 Oxidoreductase Influences CYP2B6 Activity in Cyclophosphamide Bioactivation. PLoS ONE. 2015;10:e0141979.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  59. Zhang X, Liu J, Ye F, et al. Effects of triptolide on the pharmacokinetics of cyclophosphamide in rats: a possible role of cytochrome P3A4 inhibition. Chin J Integr Med. 2014;20:534–9.

    Article  CAS  PubMed  Google Scholar 

  60. Park D, Yang Y-H, Choi E-K, et al. Licorice extract increases cyclophosphamide teratogenicity by upregulating the expression of cytochrome P-450 2B mRNA. Birth Defects Res B Dev Reprod Toxicol. 2011;92:553–9.

    Article  CAS  PubMed  Google Scholar 

  61. Yang L, Yan C, Zhang F, et al. Effects of ketoconazole on cyclophosphamide metabolism: evaluation of CYP3A4 inhibition effect using the in vitro and in vivo models. Exp Anim. 2018;67:71–82.

    Article  CAS  PubMed  Google Scholar 

  62. Ellens H, Johnson M, Lawrence SK, et al. Prediction of the transporter-mediated drug-drug interaction potential of dabrafenib and its major circulating metabolites. Drug Metab Dispos. 2017;45:646–56.

    Article  CAS  PubMed  Google Scholar 

  63. Puszkiel A, Noé G, Bellesoeur A, et al. Clinical pharmacokinetics and pharmacodynamics of dabrafenib. Clin Pharmacokinet. 2018. https://doi.org/10.1007/s40262-018-0703-0.

    Article  Google Scholar 

  64. Suttle AB, Grossmann KF, Ouellet D, et al. Assessment of the drug interaction potential and single- and repeat-dose pharmacokinetics of the BRAF inhibitor dabrafenib. J Clin Pharmacol. 2015;55:392–400.

    Article  CAS  PubMed  Google Scholar 

  65. Gibbons JA, Ouatas T, Krauwinkel W, et al. Clinical pharmacokinetic studies of enzalutamide. Clin Pharmacokinet. 2015;54:1043–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Backman JT, Filppula AM, Niemi M, et al. Role of cytochrome P450 2C8 in drug metabolism and interactions. Pharmacol Rev. 2016;68:168–241.

    Article  PubMed  Google Scholar 

  67. Weiss J, Kocher J, Mueller C, et al. Impact of enzalutamide and its main metabolite N-desmethyl enzalutamide on pharmacokinetically important drug metabolizing enzymes and drug transporters. Biopharm Drug Dispos. 2017;38:517–25.

    Article  CAS  PubMed  Google Scholar 

  68. Hamilton M, Wolf JL, Drolet DW, et al. The effect of rifampicin, a prototypical CYP3A4 inducer, on erlotinib pharmacokinetics in healthy subjects. Cancer Chemother Pharmacol. 2014;73:613–21.

    Article  CAS  PubMed  Google Scholar 

  69. Calvert H, Twelves C, Ranson M, et al. Effect of erlotinib on CYP3A activity, evaluated in vitro and by dual probes in patients with cancer. Anticancer Drugs. 2014;25:832–40.

    Article  CAS  PubMed  Google Scholar 

  70. Li J, Zhao M, He P, et al. Differential metabolism of gefitinib and erlotinib by human cytochrome P450 enzymes. Clin Cancer Res. 2007;13:3731–7.

    Article  CAS  PubMed  Google Scholar 

  71. Dong P, Fang Z, Zhang Y, et al. Substrate-dependent modulation of the catalytic activity of CYP3A by erlotinib. Acta Pharmacol Sin. 2011;32:399–407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Song J-H, Sun D-X, Chen B, et al. Inhibition of CYP3A4 and CYP2C9 by podophyllotoxin: implication for clinical drug-drug interactions. J Biosci. 2011;36:879–85.

    Article  CAS  PubMed  Google Scholar 

  73. Bossaer JB, Odle BL. Probable etoposide interaction with Echinacea. J Diet Suppl. 2012;9:90–5.

    Article  CAS  PubMed  Google Scholar 

  74. Hsieh Y-W, Huang C-Y, Yang S-Y, et al. Oral intake of curcumin markedly activated CYP 3A4: in vivo and ex vivo studies. Sci Rep. 2014;4:6587.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharmacokinetics of everolimus. Clin Pharmacokinet. 2004;43:83–95.

    Article  CAS  PubMed  Google Scholar 

  76. González F, Valjalo R. Combining cytochrome P-450 3A4 modulators and cyclosporine or everolimus in transplantation is successful. World J Transplant. 2015;5:338–47.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Ravaud A, Urva SR, Grosch K, et al. Relationship between everolimus exposure and safety and efficacy: meta-analysis of clinical trials in oncology. Eur J Cancer. 2014;50:486–95.

    Article  CAS  PubMed  Google Scholar 

  78. Tang SC, Sparidans RW, Cheung KL, et al. P-glycoprotein, CYP3A, and plasma carboxylesterase determine brain and blood disposition of the mTOR Inhibitor everolimus (Afinitor) in mice. Clin Cancer Res. 2014;20:3133–45.

    Article  CAS  PubMed  Google Scholar 

  79. Omote S, Matsuoka N, Arakawa H, et al. Effect of tyrosine kinase inhibitors on renal handling of creatinine by MATE1. Sci Rep. 2018;8:9237.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  80. Han S-Y, Zhao H-Y, Zhou N, et al. Marsdenia tenacissima extract inhibits gefitinib metabolism in vitro by interfering with human hepatic CYP3A4 and CYP2D6 enzymes. J Ethnopharmacol. 2014;151:210–7.

    Article  CAS  PubMed  Google Scholar 

  81. Wang D-D, Liu Y, Li N, et al. Induction of CYP1A1 increases gefitinib-induced oxidative stress and apoptosis in A549 cells. Toxicol In Vitro. 2017;44:36–43.

    Article  CAS  PubMed  Google Scholar 

  82. Fang P, Zheng X, He J, et al. Functional characterization of wild-type and 24 CYP2D6 allelic variants on gefitinib metabolism in vitro. Drug Des Devel Ther. 2017;11:1283–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zhao C, Han S-Y, Li P-P. Pharmacokinetics of gefitinib: roles of drug metabolizing enzymes and transporters. Curr Drug Deliv. 2017;14:282–8.

    Article  CAS  PubMed  Google Scholar 

  84. Harivenkatesh N, Kumar L, Bakhshi S, et al. Influence of MDR1 and CYP3A5 genetic polymorphisms on trough levels and therapeutic response of imatinib in newly diagnosed patients with chronic myeloid leukemia. Pharmacol Res. 2017;120:138–45.

    Article  CAS  PubMed  Google Scholar 

  85. Verboom MC, Visser L, Kouwen S, et al. Influence of CYP2C8 polymorphisms on imatinib steady-state trough level in chronic myeloid leukemia and gastrointestinal stromal tumor patients. Pharmacogenet Genomics. 2017;27:223–6.

    Article  CAS  PubMed  Google Scholar 

  86. Osorio S, Escudero-Vilaplana V, Gómez-Centurión I, et al. Inadequate response to imatinib treatment in chronic myeloid leukemia due to a drug interaction with phenytoin. J Oncol Pharm Pract. 2017;25:694–8.

    Article  PubMed  Google Scholar 

  87. Murray M, Gillani TB, Ghassabian S, et al. Differential effects of hepatic cirrhosis on the intrinsic clearances of sorafenib and imatinib by CYPs in human liver. Eur J Pharm Sci. 2018;114:55–63.

    Article  CAS  PubMed  Google Scholar 

  88. Skoglund K, Richter J, Olsson-Strömberg U, et al. In vivo cytochrome P450 3A isoenzyme activity and pharmacokinetics of imatinib in relation to therapeutic outcome in patients with chronic myeloid leukemia. Ther Drug Monit. 2016;38:230–8.

    Article  CAS  PubMed  Google Scholar 

  89. Filppula AM, Neuvonen M, Laitila J, et al. Autoinhibition of CYP3A4 leads to important role of CYP2C8 in imatinib metabolism: variability in CYP2C8 activity may alter plasma concentrations and response. Drug Metab Dispos. 2013;41:50–9.

    Article  CAS  PubMed  Google Scholar 

  90. Filppula AM, Tornio A, Niemi M, et al. Gemfibrozil impairs imatinib absorption and inhibits the CYP2C8-mediated formation of its main metabolite. Clin Pharmacol Ther. 2013;94:383–93.

    Article  CAS  PubMed  Google Scholar 

  91. Koch KM, Smith DA, Botbyl J, et al. Effect of lapatinib on oral digoxin absorption in patients. Clin Pharmacol Drug Dev. 2015;4:449–53.

    Article  CAS  PubMed  Google Scholar 

  92. Koch KM, Dees EC, Coker SA, et al. The effects of lapatinib on CYP3A metabolism of midazolam in patients with advanced cancer. Cancer Chemother Pharmacol. 2017;80:1141–6.

    Article  CAS  PubMed  Google Scholar 

  93. Hardy KD, Wahlin MD, Papageorgiou I, et al. Studies on the role of metabolic activation in tyrosine kinase inhibitor-dependent hepatotoxicity: induction of CYP3A4 enhances the cytotoxicity of lapatinib in HepaRG cells. Drug Metab Dispos. 2014;42:162–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Chan ECY, New LS, Chua TB, et al. Interaction of lapatinib with cytochrome P450 3A5. Drug Metab Dispos. 2012;40:1414–22.

    Article  CAS  PubMed  Google Scholar 

  95. Towles JK, Clark RN, Wahlin MD, et al. Cytochrome P450 3A4 and CYP3A5-Catalyzed Bioactivation of Lapatinib. Drug Metab Dispos. 2016;44:1584–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Gupta A, Jarzab B, Capdevila J, et al. Population pharmacokinetic analysis of lenvatinib in healthy subjects and patients with cancer. Br J Clin Pharmacol. 2016;81:1124–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Shumaker RC, Aluri J, Fan J, et al. Effect of rifampicin on the pharmacokinetics of lenvatinib in healthy adults. Clin Drug Investig. 2014;34:651–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Munroe M, Kolesar J. Olaparib for the treatment of BRCA-mutated advanced ovarian cancer. Am J Health Syst Pharm. 2016;73:1037–41.

    Article  CAS  PubMed  Google Scholar 

  99. Dirix L, Swaisland H, Verheul HMW, et al. Effect of itraconazole and rifampin on the pharmacokinetics of olaparib in patients with advanced solid tumors: results of two phase I open-label studies. Clin Ther. 2016;38:2286–99.

    Article  CAS  PubMed  Google Scholar 

  100. McCormick A, Swaisland H, Reddy VP, et al. In vitro evaluation of the inhibition and induction potential of olaparib, a potent poly(ADP-ribose) polymerase inhibitor, on cytochrome P450. Xenobiotica. 2018;48:555–64.

    Article  CAS  PubMed  Google Scholar 

  101. Pilla Reddy V, Walker M, Sharma P, et al. Development, verification, and prediction of osimertinib drug-drug interactions using pbpk modeling approach to inform drug label. CPT Pharmacometrics Syst Pharmacol. 2018;7:321–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Dickinson PA, Cantarini MV, Collier J, et al. Metabolic Disposition of osimertinib in rats, dogs, and humans: insights into a drug designed to bind covalently to a cysteine residue of epidermal growth factor receptor. Drug Metab Dispos. 2016;44:1201–12.

    Article  CAS  PubMed  Google Scholar 

  103. Chen Z, Chen Y, Xu M, et al. Osimertinib (AZD9291) enhanced the efficacy of chemotherapeutic agents in ABCB1- and ABCG2-overexpressing cells in vitro, in vivo, and ex vivo. Mol Cancer Ther. 2016;15:1845–58.

    Article  CAS  PubMed  Google Scholar 

  104. Hsiao S-H, Lu Y-J, Li Y-Q, et al. Osimertinib (AZD9291) attenuates the function of multidrug resistance-linked atp-binding cassette transporter ABCB1 in vitro. Mol Pharm. 2016;13:2117–25.

    Article  CAS  PubMed  Google Scholar 

  105. Zhang X-Y, Zhang Y-K, Wang Y-J, et al. Osimertinib (AZD9291), a mutant-selective egfr inhibitor, reverses ABCB1-mediated drug resistance in cancer cells. Molecules. 2016;21:1236. https://doi.org/10.3390/molecules21091236.

    Article  CAS  PubMed Central  Google Scholar 

  106. Yu Y, Loi C-M, Hoffman J, et al. Physiologically based pharmacokinetic modeling of palbociclib. J Clin Pharmacol. 2017;57:173–84.

    Article  CAS  PubMed  Google Scholar 

  107. Pabla N, Gibson AA, Buege M, et al. Mitigation of acute kidney injury by cell-cycle inhibitors that suppress both CDK4/6 and OCT2 functions. Proc Natl Acad Sci USA. 2015;112:5231–6.

    Article  CAS  PubMed  Google Scholar 

  108. De Gooijer MC, Zhang P, Thota N, et al. P-glycoprotein and breast cancer resistance protein restrict the brain penetration of the CDK4/6 inhibitor palbociclib. Invest New Drugs. 2015;33:1012–9.

    Article  PubMed  CAS  Google Scholar 

  109. Wang Y-K, Yang X-N, Liang W-Q, et al. A metabolomic perspective of pazopanib-induced acute hepatotoxicity in mice. Xenobiotica. 2018. https://doi.org/10.1080/00498254.2018.1489167.

    Article  PubMed  Google Scholar 

  110. Liu X-J, Lu H, Sun J-X, et al. Metabolic behavior prediction of pazopanib by cytochrome P450 (CYP) 3A4 by molecular docking. Eur J Drug Metab Pharmacokinet. 2016;41:465–8.

    Article  CAS  PubMed  Google Scholar 

  111. Filppula AM, Neuvonen PJ, Backman JT. In vitro assessment of time-dependent inhibitory effects on CYP2C8 and CYP3A activity by fourteen protein kinase inhibitors. Drug Metab Dispos. 2014;42:1202–9.

    Article  PubMed  CAS  Google Scholar 

  112. Ellawatty WEA, Masuo Y, Fujita K-I, et al. Organic cation transporter 1 is responsible for hepatocellular uptake of the tyrosine kinase inhibitor pazopanib. Drug Metab Dispos. 2018;46:33–40.

    Article  CAS  PubMed  Google Scholar 

  113. Sauzay C, White-Koning M, Hennebelle I, et al. Inhibition of OCT2, MATE1 and MATE2-K as a possible mechanism of drug interaction between pazopanib and cisplatin. Pharmacol Res. 2016;110:89–95.

    Article  CAS  PubMed  Google Scholar 

  114. Khurana V, Minocha M, Pal D, et al. Inhibition of OATP-1B1 and OATP-1B3 by tyrosine kinase inhibitors. Drug Metabol Drug Interact. 2014;29:249–59.

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Wang Y-J, Zhang Y-K, Zhang G-N, et al. Regorafenib overcomes chemotherapeutic multidrug resistance mediated by ABCB1 transporter in colorectal cancer: in vitro and in vivo study. Cancer Lett. 2017;396:145–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Ohya H, Shibayama Y, Ogura J, et al. Regorafenib is transported by the organic anion transporter 1B1 and the multidrug resistance protein 2. Biol Pharm Bull. 2015;38:582–6.

    Article  CAS  PubMed  Google Scholar 

  117. Kort A, Durmus S, Sparidans RW, et al. Brain and testis accumulation of regorafenib is restricted by breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (P-GP/ABCB1). Pharm Res. 2015;32:2205–16.

    Article  CAS  PubMed  Google Scholar 

  118. Wang Y-K, Xiao X-R, Xu K-P, et al. Metabolic profiling of the anti-tumor drug regorafenib in mice. J Pharm Biomed Anal. 2018;159:524–35.

    Article  CAS  PubMed  Google Scholar 

  119. Paech F, Mingard C, Grünig D, et al. Mechanisms of mitochondrial toxicity of the kinase inhibitors ponatinib, regorafenib and sorafenib in human hepatic HepG2 cells. Toxicology. 2018;395:34–44.

    Article  CAS  PubMed  Google Scholar 

  120. Di Gion P, Kanefendt F, Lindauer A, et al. Clinical pharmacokinetics of tyrosine kinase inhibitors: focus on pyrimidines, pyridines and pyrroles. Clin Pharmacokinet. 2011;50:551–603.

    Article  PubMed  Google Scholar 

  121. Zimmerman EI, Hu S, Roberts JL, et al. Contribution of OATP1B1 and OATP1B3 to the disposition of sorafenib and sorafenib-glucuronide. Clin Cancer Res. 2013;19:1458–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Ting C-T, Cheng Y-Y, Tsai T-H. Herb–drug interaction between the traditional hepatoprotective formulation and sorafenib on hepatotoxicity, histopathology and pharmacokinetics in rats. Molecules. 2017;22:1034. https://doi.org/10.3390/molecules22071034.

    Article  CAS  PubMed Central  Google Scholar 

  123. Amaya GM, Durandis R, Bourgeois DS, et al. Cytochromes P450 1A2 and 3A4 catalyze the metabolic activation of sunitinib. Chem Res Toxicol. 2018;31:570–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Teo YL, Wee HL, Chue XP, et al. Effect of the CYP3A5 and ABCB1 genotype on exposure, clinical response and manifestation of toxicities from sunitinib in Asian patients. Pharmacogenomics J. 2016;16:47–53.

    Article  CAS  PubMed  Google Scholar 

  125. Patel ND, Chakrabory K, Messmer G, et al. Severe sunitinib-induced myelosuppression in a patient with a CYP 3A4 polymorphism. J Oncol Pharm Pract. 2018;24:623–6.

    Article  PubMed  Google Scholar 

  126. Reustle A, Fisel P, Renner O, et al. Characterization of the breast cancer resistance protein (BCRP/ABCG2) in clear cell renal cell carcinoma. Int J Cancer. 2018;143:3181–93. https://doi.org/10.1002/ijc.31741.

    Article  CAS  PubMed  Google Scholar 

  127. Melchardt T, Magnes T, Weiss L, et al. Liver toxicity during temozolomide chemotherapy caused by Chinese herbs. BMC Complement Altern Med. 2014;14:115.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  128. De Gooijer MC, Zhang P, Weijer R, et al. The impact of P-glycoprotein and breast cancer resistance protein on the brain pharmacokinetics and pharmacodynamics of a panel of MEK inhibitors. Int J Cancer. 2018;142:381–91.

    Article  PubMed  CAS  Google Scholar 

  129. Katayama K, Fujiwara C, Noguchi K, et al. RSK1 protects P-glycoprotein/ABCB1 against ubiquitin-proteasomal degradation by downregulating the ubiquitin-conjugating enzyme E2 R1. Sci Rep. 2016;6:36134.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Qiu J-G, Zhang Y-J, Li Y, et al. Trametinib modulates cancer multidrug resistance by targeting ABCB1 transporter. Oncotarget. 2015;6:15494–509.

    PubMed  PubMed Central  Google Scholar 

  131. Johansson S, Read J, Oliver S, et al. Pharmacokinetic evaluations of the co-administrations of vandetanib and metformin, digoxin, midazolam, omeprazole or ranitidine. Clin Pharmacokinet. 2014;53:837–47.

    Article  CAS  PubMed  Google Scholar 

  132. Jovelet C, Deroussent A, Broutin S, et al. Influence of the multidrug transporter P-glycoprotein on the intracellular pharmacokinetics of vandetanib. Eur J Drug Metab Pharmacokinet. 2013;38:149–57.

    Article  CAS  PubMed  Google Scholar 

  133. Harmsen S, Meijerman I, Maas-Bakker RF, et al. PXR-mediated P-glycoprotein induction by small molecule tyrosine kinase inhibitors. Eur J Pharm Sci. 2013;48:644–9.

    Article  CAS  PubMed  Google Scholar 

  134. Jovelet C, Bénard J, Forestier F, et al. Inhibition of P-glycoprotein functionality by vandetanib may reverse cancer cell resistance to doxorubicin. Eur J Pharm Sci. 2012;46:484–91.

    Article  CAS  PubMed  Google Scholar 

  135. Martin P, Oliver S, Robertson J, et al. Pharmacokinetic drug interactions with vandetanib during coadministration with rifampicin or itraconazole. Drugs R D. 2011;11:37–51.

    Article  PubMed  Google Scholar 

  136. MacLeod AK, McLaughlin LA, Henderson CJ, et al. Activation status of the pregnane X receptor influences vemurafenib availability in humanized mouse models. Cancer Res. 2015;75:4573–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Zhang W, Heinzmann D, Grippo JF. Clinical Pharmacokinetics of Vemurafenib. Clin Pharmacokinet. 2017;56:1033–43.

    Article  CAS  PubMed  Google Scholar 

  138. Durmus S, Sparidans RW, Wagenaar E, et al. Oral availability and brain penetration of the B-RAFV600E inhibitor vemurafenib can be enhanced by the P-GLYCOprotein (ABCB1) and breast cancer resistance protein (ABCG2) inhibitor elacridar. Mol Pharm. 2012;9:3236–45.

    Article  CAS  PubMed  Google Scholar 

  139. Mittapalli RK, Vaidhyanathan S, Sane R, et al. Impact of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) on the brain distribution of a novel BRAF inhibitor: vemurafenib (PLX4032). J Pharmacol Exp Ther. 2012;342:33–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Michaelis M, Rothweiler F, Wurglics M, et al. Substrate-specific effects of pirinixic acid derivatives on ABCB1-mediated drug transport. Oncotarget. 2016;7:11664–76.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Beulz-Riché D, Grudé P, Puozzo C, et al. Characterization of human cytochrome P450 isoenzymes involved in the metabolism of vinorelbine. Fundam Clin Pharmacol. 2005;19:545–53.

    Article  PubMed  CAS  Google Scholar 

  142. Topletz AR, Dennison JB, Barbuch RJ, et al. The relative contributions of CYP3A4 and CYP3A5 to the metabolism of vinorelbine. Drug Metab Dispos. 2013;41:1651–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Lagas JS, Damen CWN, van Waterschoot RAB, et al. P-glycoprotein, multidrug-resistance associated protein 2, Cyp3a, and carboxylesterase affect the oral availability and metabolism of vinorelbine. Mol Pharmacol. 2012;82:636–44.

    Article  CAS  PubMed  Google Scholar 

  144. Press RR, Buckle T, Beijnen JH, et al. The effect of P-glycoprotein and cytochrome P450 3a on the oral bioavailability of vinorelbine in mice. Cancer Chemother Pharmacol. 2006;57:819–25.

    Article  CAS  PubMed  Google Scholar 

  145. Sen S, Sharma H, Singh N. Curcumin enhances Vinorelbine mediated apoptosis in NSCLC cells by the mitochondrial pathway. Biochem Biophys Res Commun. 2005;331:1245–52.

    Article  CAS  PubMed  Google Scholar 

  146. Zhou XJ, Rahmani R. Preclinical and clinical pharmacology of vinca alkaloids. Drugs. 1992;44(Suppl 4):1–16 discussion 66–69.

    Article  CAS  PubMed  Google Scholar 

  147. Kajita J, Kuwabara T, Kobayashi H, et al. CYP3A4 is mainly responsibile for the metabolism of a new vinca alkaloid, vinorelbine, in human liver microsomes. Drug Metab Dispos. 2000;28:1121–7.

    CAS  PubMed  Google Scholar 

  148. Abou-Alfa GK, Lewis LD, LoRusso P, et al. Pharmacokinetics and safety of vismodegib in patients with advanced solid malignancies and hepatic impairment. Cancer Chemother Pharmacol. 2017;80:29–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Malhi V, Colburn D, Williams SJ, et al. A clinical drug-drug interaction study to evaluate the effect of a proton-pump inhibitor, a combined P-glycoprotein/cytochrome 450 enzyme (CYP)3A4 inhibitor, and a CYP2C9 inhibitor on the pharmacokinetics of vismodegib. Cancer Chemother Pharmacol. 2016;78:41–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Williamson E, Driver S, Baxter K. Stockley’s book herbal medicines interactions. 2009. https://www.stonybrookmedicine.edu/sites/default/files/herbal_medicines_interactions-1.pdf. Accessed 23 Sept 2018.

  151. Chen S-H, Lin K-Y, Chang C-C, et al. Aloe-emodin-induced apoptosis in human gastric carcinoma cells. Food Chem Toxicol. 2007;45:2296–303.

    Article  CAS  PubMed  Google Scholar 

  152. Djuv A, Nilsen OG. Aloe vera juice: iC50 and dual mechanistic inhibition of CYP3A4 and CYP2D6. Phytother Res. 2012;26:445–51.

    CAS  PubMed  Google Scholar 

  153. Vahabi S, Eatemadi A. Phyto-anesthetics: a mini-review on herb–anesthesia drug interactions. Biomed Pharmacother. 2016;84:1885–90.

    Article  CAS  PubMed  Google Scholar 

  154. Zhang Y, Huang L, Bi H, et al. Study of the upregulation of the activity of cytochrome P450 3A isoforms by Astragalus injection and Astragalus granules in rats and in cells. Eur J Drug Metab Pharmacokinet. 2013;38:105–13.

    Article  CAS  PubMed  Google Scholar 

  155. Or PMY, Lam FFY, Kwan YW, et al. Effects of Radix Astragali and Radix Rehmanniae, the components of an anti-diabetic foot ulcer herbal formula, on metabolism of model CYP1A2, CYP2C9, CYP2D6, CYP2E1 and CYP3A4 probe substrates in pooled human liver microsomes and specific CYP isoforms. Phytomedicine. 2012;19:535–44.

    Article  CAS  PubMed  Google Scholar 

  156. Lau C, Mooiman KD, Maas-Bakker RF, et al. Effect of Chinese herbs on CYP3A4 activity and expression in vitro. J Ethnopharmacol. 2013;149:543–9.

    Article  CAS  PubMed  Google Scholar 

  157. He S-M, Yang A-K, Li X-T, et al. Effects of herbal products on the metabolism and transport of anticancer agents. Expert Opin Drug Metab Toxicol. 2010;6:1195–213.

    Article  PubMed  Google Scholar 

  158. Tian QE, De Li H, Yan M, et al. Effects of Astragalus polysaccharides on P-glycoprotein efflux pump function and protein expression in H22 hepatoma cells in vitro. BMC Complement Altern Med. 2012;12:94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Pao LH, Hu OYP, Fan HY, et al. Herb–drug interaction of 50 Chinese herbal medicines on CYP3A4 activity in vitro and in vivo. Am J Chin Med. 2012;40:57–73.

    Article  PubMed  Google Scholar 

  160. Savranoglu S, Tumer TB. Inhibitory effects of spirulina platensis on carcinogen-activating cytochrome P450 isozymes and potential for drug interactions. Int J Toxicol. 2013;32:376–84.

    Article  PubMed  CAS  Google Scholar 

  161. Lu Y, Zhong H, Tang Q, et al. Construction and verification of CYP3A5 gene polymorphisms using a Saccharomyces cerevisiae expression system to predict drug metabolism. Mol Med Rep. 2017;15:1593–600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Rahman H, Kim M, Leung G, et al. Drug–herb interactions in the elderly patient with IBD: a growing concern. Curr Treat Options Gastroenterol. 2017;15:618–36.

    Article  PubMed  Google Scholar 

  163. Sprouse AA, van Breemen RB. Pharmacokinetic interactions between drugs and botanical dietary supplements. Drug Metab Dispos. 2016;44:162–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Colombo D, Lunardon L, Bellia G. Cyclosporine and herbal supplement interactions. J Toxicol. 2014;2014:145325.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Arellano AL, Papaseit E, Romaguera A, et al. Neuropsychiatric and general interactions of natural and synthetic cannabinoids with drugs of abuse and medicines. CNS Neurol Disord Drug Targets. 2017;16:554–66. https://doi.org/10.2174/1871527316666170413104516.

    Article  CAS  PubMed  Google Scholar 

  166. Ashino T, Hakukawa K, Itoh Y, et al. Inhibitory effect of synthetic cannabinoids on CYP1A activity in mouse liver microsomes. J Toxicol Sci. 2014;39:815–20.

    Article  CAS  PubMed  Google Scholar 

  167. Kim JH, Kwon SS, Kong TY, et al. AM-2201 inhibits multiple cytochrome P450 and uridine 5’-diphospho-glucuronosyltransferase enzyme activities in human liver microsomes. Molecules. 2017;22:443. https://doi.org/10.3390/molecules22030443.

    Article  CAS  PubMed Central  Google Scholar 

  168. Kong TY, Kim J-H, Kwon S-S, et al. Inhibition of cytochrome P450 and uridine 5′-diphospho-glucuronosyltransferases by MAM-2201 in human liver microsomes. Arch Pharm Res. 2017;40:727–35.

    Article  CAS  PubMed  Google Scholar 

  169. Grabowsky JA. Drug interactions and the pharmacist: focus on everolimus. Ann Pharmacother. 2013;47:1055–63.

    Article  PubMed  CAS  Google Scholar 

  170. Dadkhah A, Allameh A, Khalafi H, et al. Inhibitory effects of dietary caraway essential oils on 1,2-dimethylhydrazine-induced colon carcinogenesis is mediated by liver xenobiotic metabolizing enzymes. Nutr Cancer. 2011;63:46–54.

    CAS  PubMed  Google Scholar 

  171. Naderi-Kalali B, Allameh A, Rasaee MJ, et al. Suppressive effects of caraway (Carum carvi) extracts on 2, 3, 7, 8-tetrachloro-dibenzo-p-dioxin-dependent gene expression of cytochrome P450 1A1 in the rat H4IIE cells. Toxicol In Vitro. 2005;19:373–7.

    Article  CAS  PubMed  Google Scholar 

  172. Tarirai C, Viljoen AM, Hamman JH. Herb–drug pharmacokinetic interactions reviewed. Expert Opin Drug Metab Toxicol. 2010;6:1515–38.

    Article  CAS  PubMed  Google Scholar 

  173. Fujita K-I, Hidaka M, Takamura N, et al. Inhibitory effects of citrus fruits on cytochrome P450 3A (CYP3A) activity in humans. Biol Pharm Bull. 2003;26:1371–3.

    Article  CAS  PubMed  Google Scholar 

  174. Satoh H, Yamashita F, Tsujimoto M, et al. Citrus juices inhibit the function of human organic anion-transporting polypeptide OATP-B. Drug Metab Dispos. 2005;33:518–23.

    Article  CAS  PubMed  Google Scholar 

  175. Adeyemi DO, Komolafe OA, Adewole OS, et al. Anti hyperglycemic activities of Annona muricata (Linn). Afr J Tradit Complement Altern Med. 2008;6:62–9.

    PubMed  PubMed Central  Google Scholar 

  176. Fu L, He L, Liang Y, et al. Experimental chemotherapy against xenografts derived from multidrug resistant KBv200 cells and parental drug-sensitive KB cells in nude mice by annonaceous acetogenin 89-2. Yao Xue Xue Bao. 2003;38:565–70.

    CAS  PubMed  Google Scholar 

  177. Holanda CMDCX, Barbosa DA, Demeda VF, et al. Influence of Annona muricata (soursop) on biodistribution of radiopharmaceuticals in rats. Acta Cir Bras. 2014;29:145–50.

    Article  PubMed  Google Scholar 

  178. Scott IM, Leduc RI, Burt AJ, et al. The inhibition of human cytochrome P450 by ethanol extracts of North American botanicals. Pharma Biol. 2006;44:315–27.

    Article  CAS  Google Scholar 

  179. Teksin ZS, Lee IJ, Nemieboka NN, et al. Evaluation of the transport, in vitro metabolism and pharmacokinetics of Salvinorin A, a potent hallucinogen. Eur J Pharm Biopharm. 2009;72:471–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Kim E, Sy-Cordero A, Graf TN, et al. Isolation and identification of intestinal CYP3A inhibitors from cranberry (Vaccinium macrocarpon) using human intestinal microsomes. Planta Med. 2011;77:265–70.

    Article  CAS  PubMed  Google Scholar 

  181. Dave AA, Samuel J. Suspected interaction of cranberry juice extracts and tacrolimus serum levels: a case report. Cureus. 2016;8:e610.

    PubMed  PubMed Central  Google Scholar 

  182. Srinivas NR. Cranberry juice ingestion and clinical drug-drug interaction potentials; review of case studies and perspectives. J Pharm Pharm Sci. 2013;16:289–303.

    Article  CAS  PubMed  Google Scholar 

  183. Mohamed MEF, Frye RF. Effects of herbal supplements on drug glucuronidation. Review of clinical, animal, and in vitro studies. Planta Med. 2011;77:311–21.

    Article  CAS  PubMed  Google Scholar 

  184. Foster B, Vandenhoek S, Hana J, et al. In vitro inhibition of human cytochrome P450-mediated metabolism of marker substrates by natural products. Phytomedicine. 2003;10:334–42.

    Article  CAS  PubMed  Google Scholar 

  185. Huet M. Medicinal plants in cancer patients: current practices and evaluation data. Bull Cancer. 2013;100:485–95.

    PubMed  Google Scholar 

  186. Pourroy B, Letellier C, Helvig A, et al. Development of a rapid risk evaluation tool for herbs/drugs interactions in cancer patients: a multicentric experience in south of France. Eur J Cancer Care (Engl). 2017;1:1–10. https://doi.org/10.1111/ecc.12752.

    Article  Google Scholar 

  187. Shamsi S, Chen Y, Lim LY. Characterization and biological properties of NanoCUR formulation and its effect on major human cytochrome P450 enzymes. Int J Pharm. 2015;495:194–203.

    Article  CAS  PubMed  Google Scholar 

  188. Cheng Y-Y, Hsieh C-H, Tsai T-H. Concurrent administration of anticancer chemotherapy drug and herbal medicine on the perspective of pharmacokinetics. J Food Drug Anal. 2018;26:S88–95.

    Article  CAS  PubMed  Google Scholar 

  189. Maliakal PP, Wanwimolruk S. Effect of herbal teas on hepatic drug metabolizing enzymes in rats. J Pharm Pharmacol. 2001;53:1323–9.

    Article  CAS  PubMed  Google Scholar 

  190. Dufay S, Worsley A, Monteillier A, et al. Herbal tea extracts inhibit Cytochrome P450 3A4 in vitro. J Pharm Pharmacol. 2014;66:1478–90.

    Article  CAS  PubMed  Google Scholar 

  191. Calitz C, Steenekamp JH, Steyn JD, et al. Impact of traditional African medicine on drug metabolism and transport. Expert Opin Drug Metab Toxicol. 2014;10:991–1003.

    Article  PubMed  Google Scholar 

  192. Unger M, Frank A. Simultaneous determination of the inhibitory potency of herbal extracts on the activity of six major cytochrome P450 enzymes using liquid chromatography/mass spectrometry and automated online extraction. Rapid Commun Mass Spectrom. 2004;18:2273–81.

    Article  CAS  PubMed  Google Scholar 

  193. Romiti N, Tramonti G, Corti A, et al. Effects of Devil’s Claw (Harpagophytum procumbens) on the multidrug transporter ABCB1/P-glycoprotein. Phytomedicine. 2009;16:1095–100.

    Article  CAS  PubMed  Google Scholar 

  194. Hostanska K, Melzer J, Rostock M, et al. Alteration of anti-inflammatory activity of Harpagophytum procumbens (devil’s claw) extract after external metabolic activation with S9 mix. J Pharm Pharmacol. 2014;66:1606–14.

    Article  CAS  PubMed  Google Scholar 

  195. Okada N, Murakami A, Urushizaki S, et al. Extracts of immature orange (Aurantii fructus immaturus) and Citrus Unshiu Peel (Citri unshiu pericarpium) Induce P-Glycoprotein and Cytochrome P450 3A4 Expression via Upregulation of Pregnane X Receptor. Front Pharmacol. 2017;8:84.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  196. Pandit S, Mukherjee PK, Ponnusankar S, et al. Metabolism mediated interaction of α-asarone and Acorus calamus with CYP3A4 and CYP2D6. Fitoterapia. 2011;82:369–74.

    Article  CAS  PubMed  Google Scholar 

  197. Hellum BH, Nilsen OG. In vitro inhibition of CYP3A4 metabolism and P-glycoprotein-mediated transport by trade herbal products. Basic Clin Pharmacol Toxicol. 2008;102:466–75.

    Article  CAS  PubMed  Google Scholar 

  198. Izzo AA, Ernst E. Interactions between herbal medicines and prescribed drugs: an updated systematic review. Drugs. 2009;69:1777–98.

    Article  CAS  PubMed  Google Scholar 

  199. Budzinski JW, Foster BC, Vandenhoek S, et al. An in vitro evaluation of human cytochrome P450 3A4 inhibition by selected commercial herbal extracts and tinctures. Phytomedicine. 2000;7:273–82.

    Article  CAS  PubMed  Google Scholar 

  200. Wanwimolruk S, Prachayasittikul V. Cytochrome P450 enzyme mediated herbal drug interactions (Part 1). EXCLI J. 2014;13:347–91.

    PubMed  PubMed Central  Google Scholar 

  201. Langhammer AJ, Nilsen OG. In vitro inhibition of human CYP1A2, CYP2D6, and CYP3A4 by six herbs commonly used in pregnancy. Phytother Res. 2014;28:603–10.

    Article  PubMed  Google Scholar 

  202. Gorski JC, Huang S-M, Pinto A, et al. The effect of echinacea (Echinacea purpurea root) on cytochrome P450 activity in vivo. Clin Pharmacol Ther. 2004;75:89–100.

    Article  PubMed  Google Scholar 

  203. Liu R, Tam TW, Mao J, et al. The effect of natural health products and traditional medicines on the activity of human hepatic microsomal-mediated metabolism of oseltamivir. J Pharm Pharm Sci. 2010;13:43–55.

    Article  PubMed  Google Scholar 

  204. Awortwe C, Bouic PJ, Masimirembwa CM, et al. Inhibition of major drug metabolizing CYPs by common herbal medicines used by HIV/AIDS patients in Africa– implications for herb–drug interactions. Drug Metab Lett. 2014;7:83–95.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  205. Hellum BH, Hu Z, Nilsen OG. Trade herbal products and induction of CYP2C19 and CYP2E1 in cultured human hepatocytes. Basic Clin Pharmacol Toxicol. 2009;105:58–63.

    Article  CAS  PubMed  Google Scholar 

  206. Gurley BJ, Swain A, Williams DK, et al. Gauging the clinical significance of P-glycoprotein-mediated herb–drug interactions: comparative effects of St. John’s wort, Echinacea, clarithromycin, and rifampin on digoxin pharmacokinetics. Mol Nutr Food Res 2008;52:772–779.

    Article  CAS  Google Scholar 

  207. Na DH, Ji HY, Park EJ, et al. Evaluation of metabolism-mediated herb–drug interactions. Arch Pharm Res. 2011;34:1829–42.

    Article  CAS  PubMed  Google Scholar 

  208. Haefeli WE, Carls A. Drug interactions with phytotherapeutics in oncology. Expert Opin Drug Metab Toxicol. 2014;10:359–77.

    Article  CAS  PubMed  Google Scholar 

  209. Nguyen S, Huang H, Foster BC, et al. Antimicrobial and P450 inhibitory properties of common functional foods. J Pharm Pharm Sci. 2014;17:254–65.

    Article  PubMed  Google Scholar 

  210. Subehan null, Zaidi SFH, Kadota S, et al. Inhibition on human liver cytochrome P450 3A4 by constituents of fennel (Foeniculum vulgare): identification and characterization of a mechanism-based inactivator. J Agric Food Chem 2007; 55: 10162–10167.

    Article  CAS  PubMed  Google Scholar 

  211. Ganzera M, Schneider P, Stuppner H. Inhibitory effects of the essential oil of chamomile (Matricaria recutita L.) and its major constituents on human cytochrome P450 enzymes. Life Sci 2006; 78: 856–861.

    Article  CAS  PubMed  Google Scholar 

  212. Nowack R, Nowak B. Herbal teas interfere with cyclosporin levels in renal transplant patients. Nephrol Dial Transplant. 2005;20:2554–6.

    Article  PubMed  Google Scholar 

  213. Qiu J-X, Zhou Z-W, He Z-X, et al. Estimation of the binding modes with important human cytochrome P450 enzymes, drug interaction potential, pharmacokinetics, and hepatotoxicity of ginger components using molecular docking, computational, and pharmacokinetic modeling studies. Drug Des Devel Ther. 2015;9:841–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Kim HJ, Kim IS, Rehman SU, et al. Effects of 6-paradol, an unsaturated ketone from gingers, on cytochrome P450-mediated drug metabolism. Bioorg Med Chem Lett. Epub ahead of print 20 February 2017. https://doi.org/10.1016/j.bmcl.2017.02.047.

    Article  CAS  Google Scholar 

  215. Shalansky S, Lynd L, Richardson K, et al. Risk of warfarin-related bleeding events and supratherapeutic international normalized ratios associated with complementary and alternative medicine: a longitudinal analysis. Pharmacotherapy. 2007;27:1237–47.

    Article  CAS  PubMed  Google Scholar 

  216. Yap KY-L, See CS, Chan A. Clinically-relevant chemotherapy interactions with complementary and alternative medicines in patients with cancer. Recent Pat Food Nutr Agric 2010; 2: 12–55.

    Article  CAS  Google Scholar 

  217. Cho H-J, Yoon I-S. Pharmacokinetic Interactions of Herbs with Cytochrome P450 and P-Glycoprotein. Evid Based Complement Alternat Med; 2015. Epub ahead of print 2015. https://doi.org/10.1155/2015/736431.

    CAS  Google Scholar 

  218. Etheridge AS, Black SR, Patel PR, et al. An in vitro evaluation of cytochrome P450 inhibition and P-glycoprotein interaction with goldenseal, Ginkgo biloba, grape seed, milk thistle, and ginseng extracts and their constituents. Planta Med. 2007;73:731–41.

    Article  CAS  PubMed  Google Scholar 

  219. Mooiman KD, Maas-Bakker RF, Hendrikx JJMA, et al. The effect of complementary and alternative medicines on CYP3A4-mediated metabolism of three different substrates: 7-benzyloxy-4-trifluoromethyl-coumarin, midazolam and docetaxel. J Pharm Pharmacol. 2014;66:865–74.

    Article  CAS  PubMed  Google Scholar 

  220. Zou L, Harkey MR, Henderson GL. Effects of herbal components on cDNA-expressed cytochrome P450 enzyme catalytic activity. Life Sci. 2002;71:1579–89.

    Article  CAS  PubMed  Google Scholar 

  221. Shi S, Klotz U. Drug interactions with herbal medicines. Clin Pharmacokinet. 2012;51:77–104.

    Article  CAS  PubMed  Google Scholar 

  222. Deng Y, Bi H, Zhao L, et al. Induction of cytochrome P450 3A by the Ginkgo biloba extract and bilobalides in human and rat primary hepatocytes. Drug Metab Lett. 2008;2:60–6.

    Article  CAS  PubMed  Google Scholar 

  223. Scott GN, Elmer GW. Update on natural product–drug interactions. Am J Health Syst Pharm. 2002;59:339–47.

    Article  PubMed  Google Scholar 

  224. Unger M. Pharmacokinetic drug interactions involving Ginkgo biloba. Drug Metab Rev. 2013;45:353–85.

    Article  CAS  PubMed  Google Scholar 

  225. Naccarato M, Yoong D, Gough K. A potential drug-herbal interaction between Ginkgo biloba and efavirenz. J Int Assoc Physicians AIDS Care (Chic). 2012;11:98–100.

    Article  Google Scholar 

  226. Wang R, Zhang H, Sun S, et al. Effect of Ginkgo Leaf Tablets on the Pharmacokinetics of Amlodipine in Rats. Eur J Drug Metab Pharmacokinet. 2016;41:825–33.

    Article  CAS  PubMed  Google Scholar 

  227. Hao M, Ba Q, Yin J, et al. Deglycosylated ginsenosides are more potent inducers of CYP1A1, CYP1A2 and CYP3A4 expression in HepG2 cells than glycosylated ginsenosides. Drug Metab Pharmacokinet. 2011;26:201–5.

    Article  CAS  PubMed  Google Scholar 

  228. Hao M, Zhao Y, Chen P, et al. Structure-activity relationship and substrate-dependent phenomena in effects of ginsenosides on activities of drug-metabolizing P450 enzymes. PLoS ONE. 2008;3:e2697.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  229. Hwang S-W, Han H-S, Lim KY, et al. Drug interaction between complementary herbal medicines and gefitinib. J Thorac Oncol. 2008;3:942–3.

    Article  PubMed  Google Scholar 

  230. Pal D, Mitra AK. MDR- and CYP3A4-mediated drug-herbal interactions. Life Sci. 2006;78:2131–45.

    Article  CAS  PubMed  Google Scholar 

  231. Goey AKL, Mooiman KD, Beijnen JH, et al. Relevance of in vitro and clinical data for predicting CYP3A4-mediated herb–drug interactions in cancer patients. Cancer Treat Rev. 2013;39:773–83.

    Article  CAS  PubMed  Google Scholar 

  232. Bilgi N, Bell K, Ananthakrishnan AN, et al. Imatinib and Panax ginseng: a potential interaction resulting in liver toxicity. Ann Pharmacother. 2010;44:926–8.

    Article  PubMed  Google Scholar 

  233. Engdal S, Nilsen OG. In vitro inhibition of CYP3A4 by herbal remedies frequently used by cancer patients. Phytother Res. 2009;23:906–12.

    Article  PubMed  Google Scholar 

  234. Fritz H, Seely D, Kennedy DA, et al. Green tea and lung cancer: a systematic review. Integr Cancer Ther. 2013;12:7–24.

    Article  CAS  PubMed  Google Scholar 

  235. Chung J, Choi D, Choi J. Effects of oral epigallocatechin gallate on the oral pharmacokinetics of verapamil in rats. Biopharm Drug Dispos. 2009;30:90–3.

    Article  CAS  PubMed  Google Scholar 

  236. Johnson SS, Oyelola FT, Ari T, et al. In vitro inhibitory activities of the extract of Hibiscus sabdariffa L. (family Malvaceae) on selected cytochrome P450 isoforms. Afr J Tradit Complement Altern Med 2013; 10: 533–540.

  237. Jacquin-Porretaz C, Nardin C, Blanc D, et al. Cutaneous Toxicity Induced by Hibiscus Tea in a Patient Treated with Erlotinib. J Thorac Oncol. 2017;12:e47–8.

    Article  PubMed  Google Scholar 

  238. Guo J, Nikolic D, Chadwick LR, et al. Identification of human hepatic cytochrome P450 enzymes involved in the metabolism of 8-prenylnaringenin and isoxanthohumol from hops (Humulus lupulus L.). Drug Metab Dispos 2006; 34: 1152–1159.

    Article  CAS  PubMed  Google Scholar 

  239. Huang Y, Zheng S, Zhu H, et al. Effects of aescin on cytochrome P450 enzymes in rats. J Ethnopharmacol. 2014;151:583–90.

    Article  CAS  PubMed  Google Scholar 

  240. Pandit S, Ponnusankar S, Bandyopadhyay A, et al. Exploring the possible metabolism mediated interaction of Glycyrrhiza glabra extract with CYP3A4 and CYP2D6. Phytother Res. 2011;25:1429–34.

    Article  CAS  PubMed  Google Scholar 

  241. Nabekura T, Yamaki T, Ueno K, et al. Inhibition of P-glycoprotein and multidrug resistance protein 1 by dietary phytochemicals. Cancer Chemother Pharmacol. 2008;62:867–73.

    Article  CAS  PubMed  Google Scholar 

  242. Kawaguchi-Suzuki M, Frye RF, Zhu H-J, et al. The effects of milk thistle (Silybum marianum) on human cytochrome P450 activity. Drug Metab Dispos. 2014;42:1611–6.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  243. Doehmer J, Eisenbraun J. Assessment of extracts from mistletoe (Viscum album) for herb–drug interaction by inhibition and induction of cytochrome P450 activities. Phytother Res. 2012;26:11–7.

    Article  CAS  PubMed  Google Scholar 

  244. Agus HH, Tekin P, Bayav M, et al. Drug interaction potential of the seed extract of Urtica urens L. (dwarf nettle). Phytother Res 2009; 23: 1763–1770.

    Article  PubMed  Google Scholar 

  245. Yu EL, Sivagnanam M, Ellis L, et al. Acute hepatotoxicity after ingestion of Morinda citrifolia (Noni Berry) juice in a 14-year-old boy. J Pediatr Gastroenterol Nutr. 2011;52:222–4.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Carr M, Klotz J, Bergeron M. Coumadin resistance and the vitamin supplement ‘Noni’. Am J Hematol. 2004;77:103.

    Article  PubMed  Google Scholar 

  247. Kang Y-C, Chen M-H, Lai S-L. Potentially Unsafe Herb–drug Interactions Between a Commercial Product of Noni Juice and Phenytoin- A Case Report. Acta Neurol Taiwan. 2015;24:43–6.

    PubMed  Google Scholar 

  248. Dresser GK, Wacher V, Wong S, et al. Evaluation of peppermint oil and ascorbyl palmitate as inhibitors of cytochrome P4503A4 activity in vitro and in vivo. Clin Pharmacol Ther. 2002;72:247–55.

    Article  CAS  PubMed  Google Scholar 

  249. Hidaka M, Nagata M, Kawano Y, et al. Inhibitory effects of fruit juices on cytochrome P450 2C9 activity in vitro. Biosci Biotechnol Biochem. 2008;72:406–11.

    Article  CAS  PubMed  Google Scholar 

  250. Li Z, Dong X, Wang D, et al. Effect of oligosaccharide esters and polygalaxanthone Ill from Polygala tenuifolia willd towards cytochrome P450. Zhongguo Zhong Yao Za Zhi. 2014;39:4459–63.

    PubMed  Google Scholar 

  251. Ryu CS, Oh SJ, Oh JM, et al. Inhibition of Cytochrome P450 by Propolis in Human Liver Microsomes. Toxicol Res. 2016;32:207–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Hanlon PR, Webber DM, Barnes DM. Aqueous extract from Spanish black radish (Raphanus sativus L. Var. niger) induces detoxification enzymes in the HepG2 human hepatoma cell line. J Agric Food Chem 2007; 55: 6439–6446.

    Article  CAS  PubMed  Google Scholar 

  253. Prasad GVR, Wong T, Meliton G, et al. Rhabdomyolysis due to red yeast rice (Monascus purpureus) in a renal transplant recipient. Transplantation. 2002;74:1200–1.

    Article  PubMed  Google Scholar 

  254. Chen C-H, Uang Y-S, Wang S-T, et al. Interaction between Red Yeast Rice and CYP450 Enzymes/P-Glycoprotein and Its Implication for the Clinical Pharmacokinetics of Lovastatin. Evid Based Complement Alternat Med. 2012;2012:127043.

    PubMed  PubMed Central  Google Scholar 

  255. Fung WT, Subramaniam G, Lee J, et al. Assessment of extracts from red yeast rice for herb–drug interaction by in vitro and in vivo assays. Sci Rep. 2012;2:298.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  256. Tang J-C, Zhang J-N, Wu Y-T, et al. Effect of the water extract and ethanol extract from traditional Chinese medicines Angelica sinensis (Oliv.) Diels, Ligusticum chuanxiong Hort. and Rheum palmatum L. on rat liver cytochrome P450 activity. Phytother Res 2006; 20: 1046–1051.

    Article  CAS  PubMed  Google Scholar 

  257. Gao J, Shi Z, Zhu S, et al. Influences of processed rhubarbs on the activities of four CYP isozymes and the metabolism of saxagliptin in rats based on probe cocktail and pharmacokinetics approaches. J Ethnopharmacol. 2013;145:566–72.

    Article  PubMed  Google Scholar 

  258. Hellum BH, Nilsen OG. The in vitro inhibitory potential of trade herbal products on human CYP2D6-mediated metabolism and the influence of ethanol. Basic Clin Pharmacol Toxicol. 2007;101:350–8.

    Article  CAS  PubMed  Google Scholar 

  259. Schrøder-Aasen T, Molden G, Nilsen OG. In vitro inhibition of CYP3A4 by the multiherbal commercial product Sambucus Force and its main constituents Echinacea purpurea and Sambucus nigra. Phytother Res. 2012;26:1606–13.

    Article  PubMed  CAS  Google Scholar 

  260. Chen F, Li L, Tian D-D. Salvia miltiorrhiza Roots against Cardiovascular Disease: consideration of Herb–Drug Interactions. Biomed Res Int. 2017;2017:9868694.

    PubMed  PubMed Central  Google Scholar 

  261. Quaye O, Cramer P, Ofosuhene M, et al. Acute and Subchronic Toxicity Studies of Aqueous Extract of Desmodium adscendens (Sw) DC. J Evid Based Complementary Altern Med. 2017;22:753–9.

    Article  PubMed  PubMed Central  Google Scholar 

  262. Herb–Drug Interactions: Starflower oil | Starflower | Medicinal Plants, http://medicinalplants.us/herb-drug-interactions-starflower-oil (accessed 16 June 2018).

  263. Xie R, Tan LH, Polasek EC, et al. CYP3A and P-glycoprotein activity induction with St. John’s Wort in healthy volunteers from 6 ethnic populations. J Clin Pharmacol 2005; 45: 352–356.

    Article  CAS  PubMed  Google Scholar 

  264. Markowitz JS, Donovan JL, DeVane CL, et al. Effect of St John’s wort on drug metabolism by induction of cytochrome P450 3A4 enzyme. JAMA. 2003;290:1500–4.

    Article  CAS  PubMed  Google Scholar 

  265. Xu H, Williams KM, Liauw WS, et al. Effects of St John’s wort and CYP2C9 genotype on the pharmacokinetics and pharmacodynamics of gliclazide. Br J Pharmacol. 2008;153:1579–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Barnes J, Anderson LA, Phillipson JD. St John’s wort (Hypericum perforatum L.): a review of its chemistry, pharmacology and clinical properties. J Pharm Pharmacol 2001; 53: 583–600.

    Article  CAS  PubMed  Google Scholar 

  267. Mouly S, Lloret-Linares C, Sellier P-O, et al. Is the clinical relevance of drug-food and drug-herb interactions limited to grapefruit juice and Saint-John’s Wort? Pharmacol Res. 2017;118:82–92.

    Article  CAS  PubMed  Google Scholar 

  268. Schwarz UI, Hanso H, Oertel R, et al. Induction of intestinal P-glycoprotein by St John’s wort reduces the oral bioavailability of talinolol. Clin Pharmacol Ther. 2007;81:669–78.

    Article  CAS  PubMed  Google Scholar 

  269. Hennessy M, Kelleher D, Spiers JP, et al. St John’s Wort increases expression of P-glycoprotein: implications for drug interactions. Br J Clin Pharmacol. 2002;53:75–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Imai H, Kotegawa T, Tsutsumi K, et al. The recovery time-course of CYP3A after induction by St John’s wort administration. Br J Clin Pharmacol. 2008;65:701–7.

    Article  PubMed  PubMed Central  Google Scholar 

  271. Yang A-K, He S-M, Liu L, et al. Herbal interactions with anticancer drugs: mechanistic and clinical considerations. Curr Med Chem. 2010;17:1635–78.

    Article  CAS  PubMed  Google Scholar 

  272. Smith P, Bullock JM, Booker BM, et al. The influence of St. John’s wort on the pharmacokinetics and protein binding of imatinib mesylate. Pharmacotherapy 2004; 24: 1508–1514.

    Article  CAS  PubMed  Google Scholar 

  273. Borrelli F, Izzo AA. Herb–drug interactions with St John’s wort (Hypericum perforatum): an update on clinical observations. AAPS J. 2009;11:710–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Brahmi Z, Niwa H, Yamasato M, et al. Effective cytochrome P450 (CYP) inhibitor isolated from thyme (Thymus saturoides) purchased from a Japanese market. Biosci Biotechnol Biochem. 2011;75:2237–9.

    Article  CAS  PubMed  Google Scholar 

  275. Aristatile B, Al-Assaf AH, Pugalendi KV. Carvacrol ameliorates the PPAR-A and cytochrome P450 expression on D-galactosamine induced hepatotoxicity rats. Afr J Tradit Complement Altern Med. 2014;11:118–23.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  276. De-Oliveira AC, Ribeiro-Pinto LF, Otto SS, et al. Induction of liver monooxygenases by beta-myrcene. Toxicology. 1997;124:135–40.

    Article  CAS  PubMed  Google Scholar 

  277. Chang-Liao WL, Chien CF, Lin LC, et al. Isolation of gentiopicroside from Gentianae Radix and its pharmacokinetics on liver ischemia/reperfusion rats. J Ethnopharmacol. 2012;141:668–73.

    Article  CAS  PubMed  Google Scholar 

  278. García JJ, Fernández N, Diez MJ, et al. Influence of two dietary fibers in the oral bioavailability and other pharmacokinetic parameters of ethinyloestradiol. Contraception. 2000;62:253–7.

    Article  PubMed  Google Scholar 

  279. Bromley J, Hughes BGM, Leong DCS, et al. Life-threatening interaction between complementary medicines: cyanide toxicity following ingestion of amygdalin and vitamin C. Ann Pharmacother. 2005;39:1566–9.

    Article  PubMed  Google Scholar 

  280. Mbeunkui F, Grace MH, Lategan C, et al. In vitro antiplasmodial activity of indole alkaloids from the stem bark of Geissospermum vellosii. J Ethnopharmacol. 2012;139:471–7.

    Article  CAS  PubMed  Google Scholar 

  281. Samojlik I, Petković S, Stilinović N, et al. Pharmacokinetic Herb–Drug Interaction between Essential Oil of Aniseed (Pimpinella anisum L., Apiaceae) and Acetaminophen and Caffeine: A Potential Risk for Clinical Practice. Phytother Res 2016; 30: 253–259.

    Article  PubMed  CAS  Google Scholar 

  282. Samojlik I, Mijatović V, Petković S, et al. The influence of essential oil of aniseed (Pimpinella anisum, L.) on drug effects on the central nervous system. Fitoterapia 2012; 83: 1466–1473.

    Article  CAS  PubMed  Google Scholar 

  283. Juanbeltz Zurbano R, Pérez-Fernández MD, Tirapu Nicolás B, et al. Complementary medicine use in cancer patients receiving intravenous antineoplastic treatment. Farm Hosp. 2017;41:589–600.

    PubMed  Google Scholar 

  284. McLay JS, Stewart D, George J, et al. Complementary and alternative medicines use by Scottish women with breast cancer. What, why and the potential for drug interactions? Eur J Clin Pharmacol 2012; 68: 811–819.

    Article  PubMed  CAS  Google Scholar 

  285. Kremser T, Evans A, Moore A, et al. Use of complementary therapies by Australian women with breast cancer. Breast. 2008;17:387–94.

    Article  CAS  PubMed  Google Scholar 

  286. Morris KT, Johnson N, Homer L, et al. A comparison of complementary therapy use between breast cancer patients and patients with other primary tumor sites. Am J Surg. 2000;179:407–11.

    Article  CAS  PubMed  Google Scholar 

  287. Molassiotis A, Scott JA, Kearney N, et al. Complementary and alternative medicine use in breast cancer patients in Europe. Support Care Cancer. 2006;14:260–7.

    Article  PubMed  Google Scholar 

  288. Boon H, Stewart M, Kennard MA, et al. Use of complementary/alternative medicine by breast cancer survivors in Ontario: prevalence and perceptions. J Clin Oncol. 2000;18:2515–21.

    Article  CAS  PubMed  Google Scholar 

  289. Amini A, Masoumi-Moghaddam S, Ehteda A, et al. Bromelain and N-acetylcysteine inhibit proliferation and survival of gastrointestinal cancer cells in vitro: significance of combination therapy. J Exp Clin Cancer Res. 2014;33:92.

    PubMed  PubMed Central  Google Scholar 

  290. Amini A, Masoumi-Moghaddam S, Ehteda A, et al. Potentiation of chemotherapeutics by bromelain and N-acetylcysteine: sequential and combination therapy of gastrointestinal cancer cells. Am J Cancer Res. 2016;6:350–69.

    CAS  PubMed  PubMed Central  Google Scholar 

  291. Hsu C-H, Cheng A-L. Clinical studies with curcumin. Adv Exp Med Biol. 2007;595:471–80.

    Article  PubMed  Google Scholar 

  292. Vallianou NG, Evangelopoulos A, Schizas N, et al. Potential anticancer properties and mechanisms of action of curcumin. Anticancer Res. 2015;35:645–51.

    CAS  PubMed  Google Scholar 

  293. Wilson KS. Regression of follicular lymphoma with Devil’s Claw: coincidence or causation? Curr Oncol. 2009;16:67–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  294. Chang H-K, Shin M-S, Yang H-Y, et al. Amygdalin induces apoptosis through regulation of Bax and Bcl-2 expressions in human DU145 and LNCaP prostate cancer cells. Biol Pharm Bull. 2006;29:1597–602.

    Article  CAS  PubMed  Google Scholar 

  295. Khacha-Ananda S, Tragoolpua K, Chantawannakul P, et al. Propolis extracts from the northern region of Thailand suppress cancer cell growth through induction of apoptosis pathways. Invest New Drugs. 2016;34:707–22.

    Article  PubMed  Google Scholar 

  296. Elnakady YA, Rushdi AI, Franke R, et al. Characteristics, chemical compositions and biological activities of propolis from Al-Bahah. Saudi Arabia. Sci Rep. 2017;7:41453.

    Article  CAS  PubMed  Google Scholar 

  297. Ren K, Zhang W, Wu G, et al. Synergistic anti-cancer effects of galangin and berberine through apoptosis induction and proliferation inhibition in oesophageal carcinoma cells. Biomed Pharmacother. 2016;84:1748–59.

    Article  CAS  PubMed  Google Scholar 

  298. Rouhollahi E, Zorofchian Moghadamtousi S, Paydar M, et al. Inhibitory effect of Curcuma purpurascens BI: Rhizome on HT-29 colon cancer cells through mitochondrial-dependent apoptosis pathway. BMC Complement Altern Med 2015; 15: 15.

  299. Cassileth BR, Lucarelli CD. Herb–drug Interactions in Oncology. PMPH-USA, 2003.

  300. Zeller T, Muenstedt K, Stoll C, et al. Potential interactions of complementary and alternative medicine with cancer therapy in outpatients with gynecological cancer in a comprehensive cancer center. J Cancer Res Clin Oncol. 2013;139:357–65.

    Article  CAS  PubMed  Google Scholar 

  301. Cheng C-W, Fan W, Ko S-G, et al. Evidence-based management of herb–drug interaction in cancer chemotherapy. Explore (NY). 2010;6:324–9.

    Article  Google Scholar 

  302. Brantley SJ, Graf TN, Oberlies NH, et al. A systematic approach to evaluate herb–drug interaction mechanisms: investigation of milk thistle extracts and eight isolated constituents as CYP3A inhibitors. Drug Metab Dispos. 2013;41:1662–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  303. Butterweck V, Derendorf H, Gaus W, et al. Pharmacokinetic herb–drug interactions: are preventive screenings necessary and appropriate? Planta Med. 2004;70:784–91.

    Article  CAS  PubMed  Google Scholar 

  304. Raucy JL. Regulation of CYP3A4 expression in human hepatocytes by pharmaceuticals and natural products. Drug Metab Dispos. 2003;31:533–9.

    Article  CAS  PubMed  Google Scholar 

  305. Harkey MR, Henderson GL, Gershwin ME, et al. Variability in commercial ginseng products: an analysis of 25 preparations. Am J Clin Nutr. 2001;73:1101–6.

    Article  CAS  PubMed  Google Scholar 

  306. Tsai H-H, Lin H-W, Simon Pickard A, et al. Evaluation of documented drug interactions and contraindications associated with herbs and dietary supplements: a systematic literature review. Int J Clin Pract. 2012;66:1056–78.

    Article  CAS  PubMed  Google Scholar 

  307. Tadic D, Spasojevic IB, Tomasevic ZI, et al. Oral administration of antineoplastic agents: the challenges for healthcare professionals. J BUON. 2015;20:690–8.

    PubMed  Google Scholar 

  308. Yap KYL, Kuo EY, Lee JJJ, et al. An onco-informatics database for anticancer drug interactions with complementary and alternative medicines used in cancer treatment and supportive care: an overview of the OncoRx project. Support Care Cancer. 2010;18:883–91.

    Article  PubMed  Google Scholar 

  309. Memorial Sloan Kettering Cancer Center, https://www.mskcc.org/. Accessed 27 March 2018.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Virginie Nerich.

Ethics declarations

Conflict of interest

All authors declared that they have no competing interests.

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the national research committee and with the 1964 Helsinki Declaration and its later amendments. In France, this search is considered like a non-interventional study according to European legislation and only a declaration to the CNIL (French data protection authority) is required: authorization granted number 1907874. All patients were individually informed that their data should be used to scientific research.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (XLSX 25 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Clairet, AL., Boiteux-Jurain, M., Curtit, E. et al. Interaction between phytotherapy and oral anticancer agents: prospective study and literature review. Med Oncol 36, 45 (2019). https://doi.org/10.1007/s12032-019-1267-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s12032-019-1267-z

Keywords

Navigation