Next Article in Journal
First Chemical Investigation of Korean Wild Mushroom, Amanita hemibapha subsp. javanica and the Identification of Anti-Helicobacter pylori Compounds
Next Article in Special Issue
Evening Primrose Oil Improves Chemotherapeutic Effects in Human Pancreatic Ductal Adenocarcinoma Cell Lines—A Preclinical Study
Previous Article in Journal
Beyond Lipid-Lowering: Effects of Statins on Cardiovascular and Cerebrovascular Diseases and Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug-Herb Interactions among Thai Herbs and Anticancer Drugs: A Scoping Review

by
Apisada Jiso
1,†,
Phisit Khemawoot
1,†,
Pinnakarn Techapichetvanich
2,
Sutinee Soopairin
3,
Kittiphong Phoemsap
3,
Panrawee Damrongsakul
3,
Supakit Wongwiwatthananukit
4 and
Pornpun Vivithanaporn
1,*
1
Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn 10540, Thailand
2
Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
3
Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
4
Department of Pharmacy Practice, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI 96720, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to the work.
Pharmaceuticals 2022, 15(2), 146; https://doi.org/10.3390/ph15020146
Submission received: 21 December 2021 / Revised: 17 January 2022 / Accepted: 19 January 2022 / Published: 26 January 2022
(This article belongs to the Special Issue Synergistic Effects of Plant Derivatives with Other Drugs)

Abstract

:
More than half of Thai patients with cancer take herbal preparations while receiving anticancer therapy. There is no systematic or scoping review on interactions between anticancer drugs and Thai herbs, although several research articles have that Thai herbs inhibit cytochrome P450 (CYP) or efflux transporter. Therefore, we gathered and integrated information related to the interactions between anticancer drugs and Thai herbs. Fifty-two anticancer drugs from the 2020 Thailand National List of Essential Medicines and 75 herbs from the 2020 Thai Herbal Pharmacopoeia were selected to determine potential anticancer drug–herb interactions. The pharmacological profiles of the selected anticancer drugs were reviewed and matched with the herbal pharmacological activities to determine possible interactions. A large number of potential anticancer drug–herb interactions were found; the majority involved CYP inhibition. Efflux transporter inhibition and enzyme induction were also found, which could interfere with the pharmacokinetic profiles of anticancer drugs. However, there is limited knowledge on the pharmacodynamic interactions between anticancer drugs and Thai herbs. Therefore, further research is warranted. Information regarding interactions between anticancer drugs and Thai herbs should provide as a useful resource to healthcare professionals in daily practice. It could enable the prediction of possible anticancer drug–herb interactions and could be used to optimize cancer therapy outcomes.

Graphical Abstract

1. Introduction

According to the World Health Organization, cancer was one of the top 10 causes of worldwide death in 2019 [1]. In 2020, there were 190,636 new cases of patients with cancer and 124,866 deaths from cancer reported in Thailand [2]. Cancer is a group of diseases caused by an abnormality in cell proliferation and differentiation, which results in an invasion into organs, leading to metastasis and death [3]. All cancer survivors are at risk of cancer recurrence despite receiving effective treatments, as some cancer cells remain in their bodies [4]. Currently, patients with cancer are treated with many types of chemotherapeutic agents, which predispose them to high incidences of adverse drug reactions and put them at high risk of drug–drug interactions, resulting in sub-therapeutic effects or increased unwanted toxicities that could potentiate the negative outcomes of cancer therapy [5]. Moreover, there are reports on herbal medicines used by patients with cancer as an alternative or supportive treatment. In one study, 433 out of 806 patients with cancer used herbal medicines while receiving chemotherapy [6]. Herbal medicine commonly used in European and Middle Eastern countries is associated with the potential risks of cytochrome P450 (CYP) induction or inhibition, altered pharmacodynamics or the reduction of anticancer resistance in in vitro models [7,8]. Since patients with cancer often take herbs to prevent and relieve the symptoms and adverse effects from anticancer drugs [9], healthcare professionals should be aware and must be vigilant against anticancer drug–herb interaction (DHI) problems arising from the use of herbs as an alternative or supportive treatment [10,11].
Using tropical herbs as an alternative cancer treatment may cause potential DHI and affect the efficacy and safety of anticancer drugs. Thus, information on anticancer drug–herb interactions could minimize or prevent problems and assist healthcare professionals to educate their patients about DHI. There is no systematic or scoping review available in which researchers have discussed interaction between anticancer drugs and commonly used Thai herbs that are relevant to clinical practice and have identified and searched for potential interactions. Therefore, we developed a scoping review of DHIs by selecting anticancer drugs from the 2020 Thailand National List of Essential Medicines (NLEM) [12] and herbs from the 2020 Thai Herbal Pharmacopoeia (THP) [13]. These herbs, such as turmeric (Curcuma longa), garlic (Allium sativum), pepper (Piper nigrum), and green chiretta (Andrographis paniculata), are commonly found in Thailand, China, India and other Southeast Asian countries. This information could be a useful resource to allow healthcare professionals to identify possible anticancer drug–herb interactions and optimize cancer therapy outcomes.

2. Results

The majority of the anticancer drugs in the 2020 NLEM are alkylating agents (23%) and antimetabolites (19%) (Figure 1A). Approximately half of the anticancer drugs are metabolized by phase I biotransformation (Figure 1B). Among phase I metabolism, 80% of anticancer pharmacokinetic profiles involve biotransformation by oxidation, especially via CYP isoforms and, to a lesser degree, by hydrolysis and reduction (Figure 1C). The major enzyme in anticancer metabolism is CYP3A4 (Figure 1D). Several anticancer drugs are excreted via the renal tubules and/or the hepatobiliary system by transmembrane transporters, especially P-glycoprotein. The pharmacokinetic profiles of the selected anticancer drugs are shown in Supporting information (Table S1).
The Thai herbs in the 2020 THP are distributed in 33 families and 13% of them are in the Apiaceae or Umbelliferae family (Figure 2A). Fruits, leaves and rhizomes are common parts that have medicinal properties (Figure 2B). The major bioactive components in these herbs are volatile oils (28%), followed by terpenoids (including triterpenoid saponins, 19%), flavonoids and phenylpropanoids (16%) (Figure 2C). Approximately half of the Thai herbs in the 2020 THP (44%) could alter drug metabolizing enzymatic activities in an in vitro setting, especially inhibition of CYP3A4 and CYP2D6. In addition, some Thai herbs could inhibit efflux transporters, particularly P-glycoprotein (Figure 2D).
Among the 52 anticancer drugs and 75 Thai herbs we selected, there are 565 potential anticancer drug–herb interactions. Approximately 90% of these interactions involve CYP inhibition, while some of the interactions exhibit potent CYP inhibitory activity. Potential anticancer drug–herb interactions might occur via drug metabolizing enzymes and efflux transporter inhibition. When categorized by the level of documentation according to the criteria in Table S2, 15 pairs are classified as good and 550 pairs are classified as fair.
All potential interferences with the activities of drug metabolizing enzymes and transporters by Thai herbs are shown in Table 1.
Andrographis paniculata, Centella asiatica, Curcuma longa, Kaempferia parviflora, and Zingiber montanum are most commonly used in Thai herbal medicine, sometimes referred to as the Thai herbal product champions [106,107]. Our findings have revealed multiple anticancer drugs–herb interactions involving various CYP isoforms and P-glycoprotein transporters. These interactions could have effects on the therapeutic activities and toxicities of anticancer drugs (Table 2).
Interestingly, many Thai herbs in our study exhibit anticancer activities (Table S3). More than of the half (39 out of 75) have been reported to show cytotoxic effects against cancer cell lines or in in vivo models. The most common cell types used in in vitro studies have been liver (16%), breast (15%) and colorectal (12%) (Figure 3A), whereas only 16 herbs (21%) have shown anticancer activity in in vivo studies. The most reported cell types have been cholangiocarcinoma (14%), lung (14%) and colorectal (9%) (Figure 3B).

3. Discussion

Drug-herb interactions could result in therapeutic failure and lead to severe adverse events. One of the most well-known natural products that interferes with drug metabolic pathways is grapefruit juice. Naringin from this citrus fruit inhibits major drug metabolizing enzymes, including CYP3A4 [109]. In our database, piperine in pepper (Piper nigrum) also showed strong inhibitory properties against CYP3A4. Therefore, it is possible that the levels of anticancer drugs metabolized mainly by this enzyme would be increased, resulting in more side effects. However, anticancer drugs given as prodrugs (for example, tamoxifen) present decreased efficacy after CYP inhibition due to the reduction in active metabolite [110,111,112,113,114,115,116]. Surprisingly, some of the Thai herbs differentially inhibit several CYP isoforms. For example, Atractylodes lancea markedly inhibits CYP1A2 and moderately inhibits CYP2C19, with weak inhibition of CYP2D6 and CYP3A4. This herb may also interfere with the metabolism of several anticancer drugs [117,118]. The majority of DHIs found in this study are related to CYP inhibition [53]. Therefore, the increased levels of anticancer drugs after concomitant use of some herbs and anticancer drugs should be monitored carefully.
Several Thai herbs that are commonly used as food ingredients show CYP inhibitory properties. Curcuma longa contains curcuminoids as bioactive ingredients, which have been found to be CYP inhibitors (for example, CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4) [19,65,66]. Thus, anticancer drug–spice interactions should also be a concern for patients with cancer due to the ability of these herbal products to inhibit drug metabolizing enzyme. Curcuminoids have recently been proposed as a bioenhancer for several conventional drugs [119]. Hence, elevated anticancer drug bioavailability and toxicity might occur during the coadministration of Curcuma longa and anticancer drugs.
Centella asiatica, a major herbal product of Thailand, has a bioactive component consisting of a triterpenoid glycoside and triterpenic acid. This herbal extract has shown mild-to-moderate inhibitory properties against several CYP isoforms, including CYP2C9 and CYP2C19 [38,61,62,120,121]. Moreover, there are reports of increased blood clotting time after the coadministration of Centella asiatica with warfarin [122]. Thus, practitioners are aware of and are vigilant of potential toxicities in patients taking Centella asiatica with a narrow therapeutic window of drugs metabolized via CYP2C9 or CYP2C19.
Allium sativum, commonly called garlic, is a widely used herb and spice in Thailand that affects anticancer drug levels. A clinical study of patients with breast cancer receiving docetaxel as monotherapy showed that the drug clearance was reduced after garlic administration. Moreover, there were genetic polymorphisms associated with the decline in docetaxel clearance [123]. Although the finding did not reach statistical significance due to a small number of participants and possible compensatory metabolic mechanisms of the drug, these findings suggest that coadministration of garlic and docetaxel affect the anticancer drug pharmacokinetics. Further investigation is required to provide clinical evidence of the undesirable adverse effects due to anticancer drug–herb pharmacokinetic interactions.
Considering pharmacodynamic interactions, several herbs in the 2020 THP show anticancer activity. The majority of the reports have focused on in vitro apoptotic cell death of cancer cell lines via various mechanisms. In addition, some major Thai herbal products (both pure compounds and extracts) show promising in vivo antiproliferative activity. Andrographis paniculata extract and andrographolide inhibit tumor-specific angiogenesis by regulating the production of various pro and antiangiogenic factors such as proinflammatory cytokines, nitric oxide, vascular endothelial growth factor (VEGF), interleukin (IL)-2 and tissue inhibitor of metalloproteinase-1 [124,125]. Co-administration of or pre-treatment with pure compounds from tropical herbs such as curcumin from Curcuma longa, thymoquinone from Nigella sativa, capsaicin from Capsicum annuum, or andrographolide from Andrographis paniculata together with anticancer drugs enhances anticancer activity via a synergistic effect. There are several common anticancer drugs that show synergistic effects when co-administered with herbs, including fluorouracil, topotecan, paclitaxel, docetaxel, and cisplatin. The interaction effect when curcumin is co-administered with anticancer drugs has reviewed by Tan and Norhaizan [126]. Thymoquinone and topotecan separately arrest the S phase of the cell cycle. The combination of thymoquinone and topotecan increases the amount of fragmented DNA and induces apoptosis through p53- and Bax/Bcl2-independent mechanisms [92]. Capsaicin also enhances in vitro and in vivo inhibitory effects and induces autophagy of 5-FU and cisplatin [55,59]. The combination of andrographolide and topotecan, gemcitabine, vincristine, cisplatin, arsenic trioxide, and paclitaxel promotes apoptosis in various cancer cell lines [26,28,29,31,36,43,44,45]. The chemical structures of major compounds from commonly used Thai herbs with potential anticancer–herb interactions are shown in Figure 4.
Pharmacodynamic research in the clinical context is needed to determine the anticancer activities of Thai herbs. An evaluation of benefits and risks should be conducted by considering both pharmacokinetic interactions and pharmacodynamics to optimize cancer therapy.
The management of potential DHI between anticancer drugs and Thai herbs seems to be one of the major problems in patient care in some countries, especially in Thailand. Both phytopharmaceutical products and food ingredients from Thai herbs could affect the outcomes of cancer therapies and increase the side effects. Thus, patient education and consultation from healthcare professionals (i.e., physicians or pharmacists) are necessary before the co-administration of anticancer drugs and Thai herbs. The algorithm ‘ask, check and consult’ could increase the safety of the co-administration of anticancer drugs and Thai herbs [127].
This review on interactions between anticancer drugs and Thai herbs provides healthcare professionals with comprehensive information for patient consultation. This study is limited by the number of anticancer drugs: there are only 52 anticancer drugs on the 2020 NLEM. This might not represent all commercially available anticancer drugs Since these are the drugs covered by Thailand’s universal health insurance, and thus they are used extensively. Another limitation is that we considered only 75 herbs derived from the 2020 THP. We did not include mixtures of preparations of several herbs in this study. Further investigation is needed to complete our database of interactions between anticancer drugs and Thai herbs.

4. Materials and Methods

4.1. Selection of Anticancer Drugs and Herbs

Fifty-two anticancer drugs from the 2020 NLEM and 99 Thai herbs from the 2020 THP were selected. Twenty-four herbal items were excluded due to the fact that they were part of herbal preparations (mixtures of multiple herbs). The selection procedure and lists of anticancer drugs and Thai herbs are shown in Figure 5 and Table 3, respectively.

4.2. Criteria for the Literature Review

We collected pharmacokinetic, pharmacodynamic, toxicological, and drug interaction data of anticancer drugs by using the Micromedex database, which we accessed under the copyright license of Chulalongkorn University (2020). If the drug data were not available in the database, we used PubMed, Science Direct, and Web of Science to find information on metabolic pathways and drug interactions. For the pharmacologic information on Thai herbs, we used the herb database from the Faculty of Pharmacy, Mahidol University, Thailand, and also available online databases (PubMed, Science Direct, and Web of Science). These data provide the pharmacodynamic activities and the possibility of drug–herb interactions. All data were gathered and analyzed from 1 January to 31 December 2020. The keywords for data collection were:
  • (‘Scientific name of herbs’ OR ‘Common name of herbs’ OR ‘major components of herbs’);
  • (‘In vitro’ OR ‘In vivo’ OR case reports OR clinical trials);
  • (cytotoxicity OR antiproliferative activity OR anticancer);
  • (Drug-herbs interaction OR Pharmacokinetic OR Pharmacodynamic);
  • (‘anticancer drug name’)
The classification criteria of the severity level and documentation are reported in Table S2. We matched two sets of collected data (anticancer drugs and Thai herbs) and analyzed them individually for potential of anticancer drug–herb interactions. We then evaluated the information on the severity, documentation, and mechanisms of these interactions.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ph15020146/s1. Table S1: Pharmacokinetic profiles of anticancer drugs; Table S2. Definition and classification of the severity level and documentation; Table S3: Thai herbs with anticancer activities.

Author Contributions

Conceptualization, S.S., K.P., P.D. and P.K.; data collection, P.V. and S.W.; DHI analysis, S.S., K.P., P.D., P.T., A.J. and P.K.; draft preparation, S.S., K.P., P.D., P.T., A.J. and P.K.; review and editing, S.W., P.V. and P.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Mahidol University for the Development of Interaction Database between Thai Herbs and Anti-cancers project (to P.V.) and Postdoctoral Fellowship Award from Mahidol University (to A.J.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data has been presented in main text and supplementary material.

Acknowledgments

All authors are grateful to Kittisak Likhitwitayawuid and Boonchoo Sritularak for their kind guidance regarding the bioactive components of Thai herbs. We sincerely appreciate Boonsong Ongphiphadhanakul and Nutthada Areepium for their kind revision of the manuscript. The Online database and mobile applications to determine DHI among anticancer-Thai herbs were developed by Sithakom Phusanti and the IT team at Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. The Top 10 Causes of Death. Available online: https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death (accessed on 18 May 2021).
  2. World Health Organization. Thailand Fact Sheets. 2021. Available online: http://gco.iarc.fr/today/data/factsheets/populations/764-thailand-fact-sheets.pdf (accessed on 18 May 2021).
  3. World Health Organization. Cancer. 2021. Available online: http://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 18 May 2021).
  4. American Cancer Society. Cancer Treatment & Survivorship Facts & Figures. 2019–2021. Available online: https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/cancer-treatment-and-survivorship-facts-and-figures/cancer-treatment-and-survivorship-facts-and-figures-2019-2021.pdf (accessed on 18 May 2021).
  5. Balis, F.M. Pharmacokinetic drug interactions of commonly used anticancer drugs. Clin. Pharm. 1986, 11, 223–235. [Google Scholar] [CrossRef] [PubMed]
  6. Alsanad, S.M.; Williamson, E.M.; Howard, R.L. Cancer patients at risk of herb/food supplement-drug interactions: A systematic review. Phytother. Res. 2014, 28, 1749–1755. [Google Scholar] [CrossRef]
  7. Ben-Arye, E.; Samuels, N.; Goldstein, L.H.; Mutafoglu, K.; Omran, S.; Schiff, E.; Charalambous, H.; Dweikat, T.; Ghrayeb, I.; Bar-Sela, G.; 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. [Google Scholar] [CrossRef] [PubMed]
  8. Gougis, P.; Hilmi, M.; Geraud, A.; Mir, O.; Funck-Brentano, C. Potential cytochrome P450-mediated pharmacokinetic interactions between herbs, food, and dietary supplements and cancer treatments. Crit. Rev. Oncol. Hematol. 2021, 166, 103342. [Google Scholar] [CrossRef] [PubMed]
  9. Yeung, K.S.; Gubili, J.; Mao, J.J. Herb-drug interactions in cancer care. Oncology 2018, 32, 516–520. [Google Scholar]
  10. Sinuanchaeng, B.; Namvongprom, A.; Pakdevong, N.-O. Educative-supportive care needs, received and satisfaction among patients with early stage cancer. J. Nurs. Sci. Health 2018, 41, 24–33. [Google Scholar]
  11. Damery, S.; Gratus, C.; Grieve, R.; Warmington, S.; Jones, J.; Routledge, P.; Greenfield, S.; Dowswell, G.; Sherriff, J.; Wilson, S. The use of herbal medicines by people with cancer: A cross-sectional survey. Br. J. Cancer 2011, 104, 927–933. [Google Scholar] [CrossRef] [Green Version]
  12. Thailand Development Research Institute. National List of Essential Medicines; Thailand Development Research Institute: Bangkok, Thailand, 2020. [Google Scholar]
  13. Bureau of Drug and Narcotic, Department of Medical Sciences, Ministry of Public Health. Thai Herbal Pharmacopoeia 2020; Bureau of Drug and Narcotic, Department of Medical Sciences, Ministry of Public Health: Bangkok, Thailand, 2020.
  14. Manda, V.K.; Avula, B.; Khan, I.A.; Khan, S.I. Inhibitory effects of Aegle marmelos and its constituents on CYP3A4 and CYP1A2 in human liver microsomes. Planta Med. 2015, 81, PP10. [Google Scholar] [CrossRef]
  15. Abou-Diwan, C.; Ritchie, J. Drug interactions with garlic and ginger supplements. In Efficacy, Toxicity, Interactions with Western Drugs, and Effects on Clinical Laboratory Tests; Wiley: Hoboken, NJ, USA, 2011; pp. 333–350. [Google Scholar] [CrossRef]
  16. Gurley, B.J.; Gardner, S.F.; Hubbard, M.A.; Williams, D.K.; Gentry, W.B.; Cui, Y.; Ang, C.Y. Clinical assessment of effects of botanical supplementation on cytochrome P450 phenotypes in the elderly: St John’s wort, garlic oil, Panax ginseng and Ginkgo biloba. Drugs Aging 2005, 22, 525–539. [Google Scholar] [CrossRef]
  17. Khan, M.; Maryam, A.; Mehmood, T.; Zhang, Y.; Ma, T. Enhancing activity of anticancer drugs in multidrug resistant tumors by modulating P-glycoprotein through dietary nutraceuticals. Asian Pac. J. Cancer Prev. 2015, 16, 6831–6839. [Google Scholar] [CrossRef] [Green Version]
  18. Kim, S.H.; Lee, I.C.; Baek, H.S.; Moon, C.; Kim, S.H.; Yoo, J.C.; Shin, I.S.; Kim, J.C. Induction of cytochrome P450 3A1 expression by diallyl disulfide: Protective effects against cyclophosphamide-induced embryo-fetal developmental toxicity. Food Chem. Toxicol. 2014, 69, 312–319. [Google Scholar] [CrossRef] [PubMed]
  19. Kimura, Y.; Ito, H.; Hatano, T. Effects of mace and nutmeg on human cytochrome P450 3A4 and 2C9 activity. Biol. Pharm. Bull. 2010, 33, 1977–1982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Kimura, Y.; Ito, H.; Ohnishi, R.; Hatano, T. Inhibitory effects of polyphenols on human cytochrome P450 3A4 and 2C9 activity. Food Chem. Toxicol. 2010, 48, 429–435. [Google Scholar] [CrossRef] [PubMed]
  21. Malekzadeh, F.; Rose, C.; Ingvar, C.; Jernstrom, H. Natural remedies and hormone preparations—Potential risk for breast cancer patients. A study surveys the use of agents which possibly counteract with the treatment. Lakartidningen 2005, 102, 3226–3228, 3230–3231. [Google Scholar]
  22. Mooiman, K.D.; Maas-Bakker, R.F.; Hendrikx, J.J.M.A.; Bank, P.C.D.; Rosing, H.; Beijnen, J.H.; Schellens, J.H.M.; Meijerman, I. 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, 865–874. [Google Scholar] [CrossRef]
  23. Williamson, E.M. Interactions between herbal and conventional medicines: The role of cytochrome P450 enzymes and P-glycoprotein. Pharmacologyonline 2006, 2, 200–205. [Google Scholar]
  24. Yang, L.J.; Fan, L.; Liu, Z.Q.; Mao, Y.M.; Guo, D.; Liu, L.H.; Tan, Z.R.; Peng, L.; Han, C.T.; Hu, D.L.; et al. Effects of allicin on CYP2C19 and CYP3A4 activity in healthy volunteers with different CYP2C19 genotypes. Eur. J. Clin. Pharmacol. 2009, 65, 601–608. [Google Scholar] [CrossRef]
  25. Zou, L.; Harkey, M.R.; Henderson, G.L. Effects of herbal components on cDNA-expressed cytochrome P450 enzyme catalytic activity. Life Sci. 2002, 71, 1579–1589. [Google Scholar] [CrossRef]
  26. Bao, G.-Q.; Shen, B.-Y.; Pan, C.-P.; Zhang, Y.-J.; Shi, M.-M.; Peng, C.-H. Andrographolide causes apoptosis via inactivation of STAT3 and Akt and potentiates antitumor activity of gemcitabine in pancreatic cancer. Toxicol. Lett. 2013, 222, 23–35. [Google Scholar] [CrossRef]
  27. Chatuphonprasert, W.; Jarukamjorn, K.; Kondo, S.; Nemoto, N. Synergistic increases of metabolism and oxidation–reduction genes on their expression after combined treatment with a CYP1A inducer and andrographolide. Chem. Biol. Interact. 2009, 182, 233–238. [Google Scholar] [CrossRef]
  28. Chen, S.; Hu, H.; Miao, S.; Zheng, J.; Xie, Z.; Zhao, H. Anti-tumor effect of cisplatin in human oral squamous cell carcinoma was enhanced by andrographolide via upregulation of phospho-p53 in vitro and in vivo. Tumor Biol. 2017, 39, 1010428317705330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Duan, X.; Li, T.; Han, X.; Ren, J.; Chen, P.; Li, H.; Gong, S. The antitumor effect of arsenic trioxide on hepatocellular carcinoma is enhanced by andrographolide. Oncotarget 2017, 8, 90905–90915. [Google Scholar] [CrossRef] [PubMed]
  30. Guo, H.; Zhang, Z.; Su, Z.; Sun, C.; Zhang, X.; Zhao, X.; Lai, X.; Su, Z.; Li, Y.; Zhan, J.Y. Enhanced anti-tumor activity and reduced toxicity by combination andrographolide and bleomycin in ascitic tumor-bearing mice. Eur. J. Pharmacol. 2016, 776, 52–63. [Google Scholar] [CrossRef] [PubMed]
  31. Hodroj, M.H.; Jardaly, A.; Abi Raad, S.; Zouein, A.; Rizk, S. Andrographolide potentiates the antitumor effect of topotecan in acute myeloid leukemia cells through an intrinsic apoptotic pathway. Cancer Manag. Res. 2018, 10, 1079–1088. [Google Scholar] [CrossRef] [Green Version]
  32. Ismail, S.; Hanapi, N.A.; Ab Halim, M.R.; Uchaipichat, V.; Mackenzie, P.I. Effects of Andrographis paniculata and Orthosiphon stamineus extracts on the glucuronidation of 4-methylumbelliferone in human UGT isoforms. Molecules 2010, 15, 3578–3592. [Google Scholar] [CrossRef] [Green Version]
  33. Jaruchotikamol, A.; Jarukamjorn, K.; Sirisangtrakul, W.; Sakuma, T.; Kawasaki, Y.; Nemoto, N. Strong synergistic induction of CYP1A1 expression by andrographolide plus typical CYP1A inducers in mouse hepatocytes. Toxicol. Appl. Pharmacol. 2007, 224, 156–162. [Google Scholar] [CrossRef]
  34. Jarukamjorn, K.; Don-in, K.; Makejaruskul, C.; Laha, T.; Daodee, S.; Pearaksa, P.; Sripanidkulchai, B.O. Impact of Andrographis paniculata crude extract on mouse hepatic cytochrome P450 enzymes. J. Ethnopharmacol. 2006, 105, 464–467. [Google Scholar] [CrossRef]
  35. Kang, X.; Zheng, Z.; Liu, Z.; Wang, H.; Zhao, Y.; Zhang, W.; Shi, M.; He, Y.; Cao, Y.; Xu, Q.; et al. Liposomal codelivery of doxorubicin and andrographolide inhibits breast cancer growth and metastasis. Mol. Pharm. 2018, 15, 1618–1626. [Google Scholar] [CrossRef]
  36. Lin, H.-H.; Shi, M.-D.; Tseng, H.-C.; Chen, J.-H. Andrographolide sensitizes the cytotoxicity of human colorectal carcinoma cells toward cisplatin via enhancing apoptosis pathways in vitro and in vivo. Toxicol. Sci. 2014, 139, 108–120. [Google Scholar] [CrossRef] [Green Version]
  37. Mao, W.; He, P.; Wang, W.; Wu, X.; Wei, C. Andrographolide sensitizes Hep-2 human laryngeal cancer cells to carboplatin-induced apoptosis by increasing reactive oxygen species levels. Anticancer Drugs 2019, 30, 731–739. [Google Scholar] [CrossRef]
  38. Pan, Y.; Abd-Rashid, B.A.; Ismail, Z.; Ismail, R.; Mak, J.W.; Pook, P.C.; Er, H.M.; Ong, C.E. In vitro modulatory effects of Andrographis paniculata, Centella asiatica and Orthosiphon stamineus on cytochrome P450 2C19 (CYP2C19). J. Ethnopharmacol. 2011, 133, 881–887. [Google Scholar] [CrossRef] [PubMed]
  39. Pekthong, D.; Blanchard, N.; Abadie, C.; Bonet, A.; Heyd, B.; Mantion, G.; Berthelot, A.; Richert, L.; Martin, H. Effects of Andrographis paniculata extract and andrographolide on hepatic cytochrome P450 mRNA expression and monooxygenase activities after in vivo administration to rats and in vitro in rat and human hepatocyte cultures. Chem. Biol. Interact. 2009, 179, 247–255. [Google Scholar] [CrossRef] [PubMed]
  40. Pekthong, D.; Martin, H.; Abadie, C.; Bonet, A.; Heyd, B.; Mantion, G.; Richert, L. Differential inhibition of rat and human hepatic cytochrome P450 by Andrographis paniculata extract and andrographolide. J. Ethnopharmacol. 2008, 115, 432–440. [Google Scholar] [CrossRef] [PubMed]
  41. Su, M.; Qin, B.; Liu, F.; Chen, Y.; Zhang, R. Andrographolide enhanced 5-fluorouracil-induced antitumor effect in colorectal cancer via inhibition of c-MET pathway. Drug Des. Dev. Ther. 2017, 11, 3333–3341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Usia, T.; Iwata, H.; Hiratsuka, A.; Watabe, T.; Kadota, S.; Tezuka, Y. CYP3A4 and CYP2D6 inhibitory activities of Indonesian medicinal plants. Phytomedicine 2006, 13, 67–73. [Google Scholar] [CrossRef] [PubMed]
  43. Yuan, H.; Sun, B.; Gao, F.; Lan, M. Synergistic anticancer effects of andrographolide and paclitaxel against A549 NSCLC cells. Pharm. Biol. 2016, 54, 2629–2635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Yunos, N.M.; Mutalip, S.S.M.; Jauri, M.H.; Yu, J.Q.; Huq, F. Anti-proliferative and pro-apoptotic effects from sequenced combinations of andrographolide and cisplatin on ovarian cancer cell lines. Anticancer Res. 2013, 33, 4365. [Google Scholar]
  45. Zhang, M.; Xue, E.; Shao, W. Andrographolide promotes vincristine-induced SK-NEP-1 tumor cell death via PI3K-AKT-p53 signaling pathway. Drug Des. Dev. Ther. 2016, 10, 3143–3152. [Google Scholar] [CrossRef] [Green Version]
  46. Chen, X.P.; Li, W.; Xiao, X.F.; Zhang, L.L.; Liu, C.X. Phytochemical and pharmacological studies on Radix Angelica sinensis. Chin. J. Nat. Med. 2013, 11, 577–587. [Google Scholar] [CrossRef]
  47. Li, Y.H.; Zhang, Y.Q.; Li, L.; Wang, Q.; Wang, N.S. Effect of Danggui and Honghua on cytochrome P450 1A2, 2C11, 2E1 and 3A1 mRNA expression in liver of rats. Am. J. Chin. Med. 2008, 36, 1071–1081. [Google Scholar] [CrossRef]
  48. Yu, C.; Chai, X.; Yu, L.; Chen, S.; Zeng, S. Identification of novel pregnane X receptor activators from traditional Chinese medicines. J. Ethnopharmacol. 2011, 136, 137–143. [Google Scholar] [CrossRef]
  49. Ashour, M.L.; Youssef, F.S.; Gad, H.A.; Wink, M. Inhibition of cytochrome P450 (CYP3A4) activity by extracts from 57 plants used in Traditional Chinese Medicine (TCM). Pharmacogn. Mag. 2017, 13, 300–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Melillo de Magalhães, P.; Dupont, I.; Hendrickx, A.; Joly, A.; Raas, T.; Dessy, S.; Sergent, T.; Schneider, Y.-J. Anti-inflammatory effect and modulation of cytochrome P450 activities by Artemisia annua tea infusions in human intestinal Caco-2 cells. Food Chem. 2012, 134, 864–871. [Google Scholar] [CrossRef] [PubMed]
  51. Wei, S.; Ji, H.; Yang, B.; Ma, L.; Bei, Z.; Li, X.; Dang, H.; Yang, X.; Liu, C.; Wu, X.; et al. Impact of chrysosplenetin on the pharmacokinetics and anti-malarial efficacy of artemisinin against Plasmodium berghei as well as in vitro CYP450 enzymatic activities in rat liver microsome. Malar. J. 2015, 14, 432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Pao, L.H.; Hu, O.Y.; Fan, H.Y.; Lin, C.C.; Liu, L.C.; Huang, P.W. Herb-drug interaction of 50 Chinese herbal medicines on CYP3A4 activity in vitro and in vivo. Am. J. Chin. Med. 2012, 40, 57–73. [Google Scholar] [CrossRef] [PubMed]
  53. Sumsakul, W.; Mahavorasirikul, W.; Na-Bangchang, K. Inhibitory activities of Thai medicinal plants with promising activities against malaria and cholangiocarcinoma on human cytochrome P450. Phytother. Res. 2015, 29, 1926–1933. [Google Scholar] [CrossRef]
  54. Chen, J.-C.; Ko, J.-C.; Yen, T.-C.; Chen, T.-Y.; Lin, Y.-C.; Ma, P.-F.; Lin, Y.-W. Capsaicin enhances erlotinib-induced cytotoxicity via AKT inactivation and excision repair cross-complementary 1 (ERCC1) down-regulation in human lung cancer cells. Toxicol. Res. 2019, 8, 459–470. [Google Scholar] [CrossRef]
  55. Hong, Z.-F.; Zhao, W.-X.; Yin, Z.-Y.; Xie, C.-R.; Xu, Y.-P.; Chi, X.-Q.; Zhang, S.; Wang, X.-M. Capsaicin enhances the drug sensitivity of cholangiocarcinoma through the inhibition of chemotherapeutic-induced autophagy. PLoS ONE 2015, 10, e0121538. [Google Scholar] [CrossRef]
  56. Lan, Y.; Sun, Y.; Yang, T.; Ma, X.; Cao, M.; Liu, L.; Yu, S.; Cao, A.; Liu, Y. Co-delivery of paclitaxel by a capsaicin prodrug micelle facilitating for combination therapy on breast cancer. Mol. Pharm. 2019, 16, 3430–3440. [Google Scholar] [CrossRef]
  57. Nabekura, T.; Kamiyama, S.; Kitagawa, S. Effects of dietary chemopreventive phytochemicals on P-glycoprotein function. Biochem. Biophys. Res. Commun. 2005, 327, 866–870. [Google Scholar] [CrossRef]
  58. Sánchez, B.G.; Bort, A.; Mateos-Gómez, P.A.; Rodríguez-Henche, N.; Díaz-Laviada, I. Combination of the natural product capsaicin and docetaxel synergistically kills human prostate cancer cells through the metabolic regulator AMP-activated kinase. Cancer Cell Int. 2019, 19, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Wang, Y.; Deng, X.; Yu, C.; Zhao, G.; Zhou, J.; Zhang, G.; Li, M.; Jiang, D.; Quan, Z.; Zhang, Y. Synergistic inhibitory effects of capsaicin combined with cisplatin on human osteosarcoma in culture and in xenografts. J. Exp. Clin. Cancer Res. 2018, 37, 251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Wang, J.; Huang, M.; Hu, H.; Yu, L.; Zeng, S. Pregnane X receptor-mediated transcriptional activation of UDP-glucuronosyltransferase 1A1 by natural constituents from foods and herbs. Food Chem. 2014, 164, 74–80. [Google Scholar] [CrossRef] [PubMed]
  61. Kulthong, K.; Tantisira, M.; Niwattisaiwong, N.; Apipalakul, K.; Chevapat, S.; Lawanprasert, S. Effects of the standard extract of Centella Asiatica (ECa233) on rat hepatic cytochrome P450. Thai J. Pharm. Sci. 2009, 33, 91–100. [Google Scholar]
  62. Pan, Y.; Abd-Rashid, B.A.; Ismail, Z.; Ismail, R.; Mak, J.W.; Pook, P.C.; Er, H.M.; Ong, C.E. In vitro modulatory effects on three major human cytochrome P450 enzymes by multiple active constituents and extracts of Centella asiatica. J. Ethnopharmacol. 2010, 130, 275–283. [Google Scholar] [CrossRef]
  63. Savai, J.; Varghese, A.; Pandita, N.; Chintamaneni, M. In vitro assessment of CYP1A2 and 2C9 inhibition potential of Withania somnifera and Centella asiatica in human liver microsomes. Drug Metab. Pers. Ther. 2015, 30, 137–141. [Google Scholar] [CrossRef]
  64. Piyapolrungroi, N.; Sotanaphun, U.; Phattanawasin, P. Effect of leech lime juice on cytochrome P450 3A4 and P-glycoprotein activities. In Proceedings of the 3rd Asian Pacific Regional Meeting, Bangkok, Thailand, 10–12 May 2009; p. 122. [Google Scholar]
  65. Appiah-Opong, R.; Commandeur, J.N.; van Vugt-Lussenburg, B.; Vermeulen, N.P. Inhibition of human recombinant cytochrome P450s by curcumin and curcumin decomposition products. Toxicology 2007, 235, 83–91. [Google Scholar] [CrossRef]
  66. Foster, B.C.; Vandenhoek, S.; Hana, J.; Krantis, A.; Akhtar, M.H.; Bryan, M.; Budzinski, J.W.; Ramputh, A.; Arnason, J.T. In vitro inhibition of human cytochrome P450-mediated metabolism of marker substrates by natural products. Phytomedicine 2003, 10, 334–342. [Google Scholar] [CrossRef]
  67. Pouyfung, P.; Sarapusit, S.; Rongnoparut, P. Effects of Vernonia cinerea compounds on drug-metabolizing cytochrome P450s in human liver microsomes. Phytother. Res. 2017, 31, 1916–1925. [Google Scholar] [CrossRef]
  68. Han, Y.M.; Kim, I.S.; Rehman, S.U.; Choe, K.; Yoo, H.H. In vitro evaluation of the effects of Eurycoma longifolia extract on CYP-mediated drug metabolism. Evid. Based Complement. Altern. Med. 2015, 2015, 631329. [Google Scholar] [CrossRef] [Green Version]
  69. Muthiah, Y.D.; Ong, C.E.; Sulaiman, S.A.; Ismail, R. Inhibition of human cytochrome P450 2C8-catalyzed amodiaquine N-desethylation: Effect of five traditionally and commonly used herbs. Pharmacogn. Res. 2016, 8, 292–297. [Google Scholar] [CrossRef] [Green Version]
  70. Langhammer, A.J.; Nilsen, O.G. In vitro inhibition of human CYP1A2, CYP2D6, and CYP3A4 by six herbs commonly used in pregnancy. Phytother. Res. 2014, 28, 603–610. [Google Scholar] [CrossRef] [PubMed]
  71. Langhammer, A.J.; Nilsen, O.G. Fennel and raspberry leaf as possible inhibitors of acetaminophen oxidation. Phytother. Res. 2014, 28, 1573–1576. [Google Scholar] [CrossRef] [PubMed]
  72. He, M.; Jiang, J.; Qiu, F.; Liu, S.; Peng, P.; Gao, C.; Miao, P. Inhibitory effects of gypenosides on seven human cytochrome P450 enzymes in vitro. Food Chem. Toxicol. 2013, 57, 262–265. [Google Scholar] [CrossRef] [PubMed]
  73. Johnson, S.S.; Oyelola, F.T.; Ari, T.; Juho, H. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Dumrongsakunchai, W.; Attakornvattana, V.; Somanabandhu, A.; Vannaprasaht, S.; Tassaneeyakul, W. Inhibitory effect and mechanism-based inhibition of Thai herbal plants on CYP3A4 and CYP2D6 activities. Thai J. Pharmacol. 2007, 29, 35–39. [Google Scholar]
  75. Suwannakul, S.; Kumpangngam, N.; Panprom, S.; Watcharathanakij, S.; Sethabouppha, B. Survey study of herbal medicine used in out-patients of Prasrimahabhodi psychiatric hospital, Thailand. In Proceedings of the The 7th Indochina Conference on Pharmaceutical Sciences: Advancing Pharmacy for ASEAN Community, Bangkok, Thailand, 14–16 December 2011; pp. 255–257. [Google Scholar]
  76. Noysang, C.; Mahringer, A.; Zeino, M.; Saeed, M.; Luanratana, O.; Fricker, G.; Bauer, R.; Efferth, T. Cytotoxicity and inhibition of P-glycoprotein by selected medicinal plants from Thailand. J. Ethnopharmacol. 2014, 155, 633–641. [Google Scholar] [CrossRef]
  77. Appiah-Opong, R.; Commandeur, J.N.; Axson, C.; Vermeulen, N.P. Interactions between cytochromes P450, glutathione S-transferases and Ghanaian medicinal plants. Food Chem. Toxicol. 2008, 46, 3598–3603. [Google Scholar] [CrossRef]
  78. Konishi, T.; Satsu, H.; Hatsugai, Y.; Aizawa, K.; Inakuma, T.; Nagata, S.; Sakuda, S.H.; Nagasawa, H.; Shimizu, M. Inhibitory effect of a bitter melon extract on the P-glycoprotein activity in intestinal Caco-2 cells. Br. J. Pharmacol. 2004, 143, 379–387. [Google Scholar] [CrossRef] [Green Version]
  79. Monera, T.G.; Wolfe, A.R.; Maponga, C.C.; Benet, L.Z.; Guglielmo, J. Moringa oleifera leaf extracts inhibit 6β-hydroxylation of testosterone by CYP3A4. J. Infect. Dev. Ctries. 2008, 2, 379–383. [Google Scholar] [CrossRef]
  80. Taesotikul, T.; Navinpipatana, V.; Tassaneeyakul, W. Selective inhibition of human cytochrome P450 1A2 by Moringa oleifera. Thai J. Pharmacol. 2010, 32, 256–258. [Google Scholar]
  81. Huang, L.; Bi, H.C.; Liu, Y.H.; Wang, Y.T.; Xue, X.P.; Huang, M. CAR-mediated up-regulation of CYP3A4 expression in LS174T cells by Chinese herbal compounds. Drug Metab. Pharm. 2011, 26, 331–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Limtrakul, P. Research and Development of Herbal Tea for Drug Resistance in Cervical Cancer; Department of Biochemistry Chiang Mai University: Bangkok, Thailand, 2003. [Google Scholar] [CrossRef]
  83. Liu, Y.H.; Mo, S.L.; Bi, H.C.; Hu, B.F.; Li, C.G.; Wang, Y.T.; Huang, L.; Huang, M.; Duan, W.; Liu, J.P.; et al. Regulation of human pregnane X receptor and its target gene cytochrome P450 3A4 by Chinese herbal compounds and a molecular docking study. Xenobiotica 2011, 41, 259–280. [Google Scholar] [CrossRef] [PubMed]
  84. Ye, L.H.; He, X.X.; Kong, L.T.; Liao, Y.H.; Pan, R.L.; Xiao, B.X.; Liu, X.M.; Chang, Q. Identification and characterization of potent CYP2D6 inhibitors in lotus leaves. J. Ethnopharmacol. 2014, 153, 190–196. [Google Scholar] [CrossRef] [PubMed]
  85. Zhao, Y.; Hellum, B.H.; Liang, A.; Nilsen, O.G. The in vitro inhibition of human CYP1A2, CYP2D6 and CYP3A4 by tetrahydropalmatine, neferine and berberine. Phytother. Res. 2012, 26, 277–283. [Google Scholar] [CrossRef]
  86. Zhao, Y.; Hellum, B.H.; Liang, A.; Nilsen, O.G. Inhibitory mechanisms of human CYPs by three alkaloids isolated from traditional Chinese herbs. Phytother. Res. 2015, 29, 825–834. [Google Scholar] [CrossRef]
  87. Ahmad, A.; Husain, A.; Mujeeb, M.; Khan, S.A.; Najmi, A.K.; Siddique, N.A.; Damanhouri, Z.A.; Anwar, F. A review on therapeutic potential of Nigella sativa: A miracle herb. Asian Pac. J. Trop. Biomed. 2013, 3, 337–352. [Google Scholar] [CrossRef] [Green Version]
  88. Albassam, A.A.; Ahad, A.; Alsultan, A.; Al-Jenoobi, F.I. Inhibition of cytochrome P450 enzymes by thymoquinone in human liver microsomes. Saudi Pharm. J. 2018, 26, 673–677. [Google Scholar] [CrossRef]
  89. Dogar, M.Z.U.H.; Adi, H.; Akhtar, M.S.; Sheikh, M.A. Preliminary assessment of efficacy of Nigella sativa seeds in acute lymphoblastic leukemia in local children. Pharmacol. Online 2009, 2, 769–777. [Google Scholar]
  90. Effenberger-Neidnicht, K.; Schobert, R. Combinatorial effects of thymoquinone on the anti-cancer activity of doxorubicin. Cancer Chemother. Pharmacol. 2011, 67, 867–874. [Google Scholar] [CrossRef] [Green Version]
  91. Elbarbry, F.; Ung, A.; Abdelkawy, K. Studying the inhibitory effect of quercetin and thymoquinone on human cytochrome P450 enzyme activities. Pharm. Mag. 2017, 13, 895–899. [Google Scholar] [CrossRef]
  92. Khalife, R.; Hodroj, M.H.; Fakhoury, R.; Rizk, S. Thymoquinone from Nigella sativa seeds promotes the antitumor activity of noncytotoxic doses of topotecan in human colorectal cancer cells in vitro. Planta Med. 2016, 82, 312–321. [Google Scholar] [CrossRef] [PubMed]
  93. Khan, A.; Aldebasy, Y.H.; Alsuhaibani, S.A.; Khan, M.A. Thymoquinone augments cyclophosphamide-mediated inhibition of cell proliferation in breast cancer cells. Asian Pac. J. Cancer Prev. 2019, 20, 1153–1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lei, X.; Lv, X.; Liu, M.; Yang, Z.; Ji, M.; Guo, X.; Dong, W. Thymoquinone inhibits growth and augments 5-fluorouracil-induced apoptosis in gastric cancer cells both in vitro and in vivo. Biochem. Biophys. Res. Commun. 2012, 417, 864–868. [Google Scholar] [CrossRef] [PubMed]
  95. Mu, G.-G.; Zhang, L.-l.; Li, H.-Y.; Liao, Y.; Yu, H.-G. Thymoquinone pretreatment overcomes the insensitivity and potentiates the antitumor effect of gemcitabine through abrogation of Notch1, PI3K/Akt/mTOR regulated signaling pathways in pancreatic cancer. Dig. Dis. Sci. 2015, 60, 1067–1080. [Google Scholar] [CrossRef]
  96. Pan, Y.; Abd-Rashid, B.A.; Ismail, Z.; Ismail, R.; Mak, J.W.; Pook, P.C.; Er, H.M.; Ong, C.E. In vitro effects of active constituents and extracts of Orthosiphon stamineus on the activities of three major human cDNA-expressed cytochrome P450 enzymes. Chem. Biol. Interact. 2011, 190, 1–8. [Google Scholar] [CrossRef] [PubMed]
  97. Sornsuvit, C.; Phosuya, C.; Jaroonwanichkul, D.; Piriyachananusorn, N. The use of herbal and dietary supplements and potential interactions with drugs in patients with chronic diseases. Thai Pharm. Health Sci. J. 2012, 7, 149–154. [Google Scholar]
  98. Anannarukan, N.; Niwattisaiwong, N.; Warisnoicharoen, W.; Winitthana, T.; Pramyothin, P.; Chaichantipayuth, C.; Lawanprasert, S. Inhibition of human cytochrome P450 in vitro by Phyllanthus amarus and Phyllanthus emblica aqueous extracts. Thai J. Pharm. Sci. 2012, 36, 135–143. [Google Scholar]
  99. Junyaprasert, V.B.; Soonthornchareonnon, N.; Thongpraditchote, S.; Murakami, T.; Takano, M. Inhibitory effect of Thai plant extracts on P-glycoprotein mediated efflux. Phytother. Res. 2006, 20, 79–81. [Google Scholar] [CrossRef]
  100. Pinmai, K.; Chunlaratthanabhorn, S.; Ngamkitidechakul, C.; Soonthornchareon, N.; Hahnvajanawong, C. Synergistic growth inhibitory effects of Phyllanthus emblica and Terminalia bellerica extracts with conventional cytotoxic agents: Doxorubicin and cisplatin against human hepatocellular carcinoma and lung cancer cells. World J. Gastroenterol. 2008, 14, 1491–1497. [Google Scholar] [CrossRef]
  101. Harwansh, R.K.; Mukherjee, K.; Bhadra, S.; Kar, A.; Bahadur, S.; Mitra, A.; Mukherjee, P.K. Cytochrome P450 inhibitory potential and RP-HPLC standardization of trikatu--a Rasayana from Indian Ayurveda. J. Ethnopharmacol. 2014, 153, 674–681. [Google Scholar] [CrossRef] [PubMed]
  102. Jin, M.J.; Han, H.K. Effect of piperine, a major component of black pepper, on the intestinal absorption of fexofenadine and its implication on food-drug interaction. J. Food Sci. 2010, 75, H93–H96. [Google Scholar] [CrossRef] [PubMed]
  103. Larson, E.C.; Hathaway, L.B.; Lamb, J.G.; Pond, C.D.; Rai, P.P.; Matainaho, T.K.; Piskaut, P.; Barrows, L.R.; Franklin, M.R. Interactions of Papua New Guinea medicinal plant extracts with antiretroviral therapy. J. Ethnopharmacol. 2014, 155, 1433–1440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wu, G.; Dong, Z.; Dong, J.; Wei, L.; Shi, R.; Kang, S.; Zhang, D. Effects of mongolian medicine Terminalia chebula Retz. on 6 CYP450 enzymes in rats. Int. J. Clin. Exp. Pathol. 2020, 13, 3128–3138. [Google Scholar]
  105. Dumrongsakunchai, W.; Attakornvattana, V.; Somanabandhu, A.; Vannaprasaht, S.; Tassaneeyakul, W. Inhibitory effect of Thai herbal plants on CYP3A activity. Thai J. Pharmacol. 2006, 28, 88. [Google Scholar]
  106. Sompopcharoen, M.; Sresumatchai, V. Systematic Review: Marketing Communication of Thai Herbal Products to Enhance Potential in Becoming Global Products. In Proceedings of the The 1st International Conference on Innovative Communication and Sustainable Development in ASEAN, Bangkok, Thailand, 9–10 July 2015; pp. 243–253. [Google Scholar]
  107. Thai Herbal Product Champions. Available online: https://pharmacy.mahidol.ac.th/th/knowledge/article/404/ProductChampion%E0%B8%82%E0%B8%AD%E0%B8%87%E0%B8%AA%E0%B8%A1%E0%B8%B8%E0%B8%99%E0%B9%84%E0%B8%9E%E0%B8%A3%E0%B9%84%E0%B8%97%E0%B8%A2/ (accessed on 16 December 2020).
  108. Ye, L.-H.; Kong, L.-T.; Yan, M.-Z.; Cao, F.-R.; Wang, L.-S.; Liao, Y.-H.; Pan, R.-L.; Chang, Q. Lotus leaf alkaloid fraction can strongly inhibit CYP2D6 isoenzyme activity. J. Ethnopharmacol. 2016, 194, 913–917. [Google Scholar] [CrossRef]
  109. Bailey, D.G.; Dresser, G.; Arnold, J.M.O. Grapefruit-medication interactions: Forbidden fruit or avoidable consequences? CMAJ 2013, 185, 309–316. [Google Scholar] [CrossRef] [Green Version]
  110. AstraZeneca. Product Information: Nolvadex, Tamoxifen Citrate; AstraZeneca: North Ryde, Australia, 2003. [Google Scholar]
  111. BC Cancer Drug Manual. Tamoxifen. Available online: http://www.bccancer.bc.ca/drug-database-site/Drug%20Index/Tamoxifen_monograph.pdf (accessed on 18 May 2021).
  112. Mayne Pharma Group Limited. Product Information: SOLTAMOX Oral Solution, Tamoxifen Citrate Oral Solution; Mayne Pharma Group Limited: Salisbury South, Australia, 2018. [Google Scholar]
  113. Heel, R.C.; Brogden, R.N.; Speight, T.M.; Avery, G.S. Tamoxifen: A review of its pharmacological properties and therapeutic use in the treatment of breast cancer. Drugs 1978, 16, 1–24. [Google Scholar] [CrossRef]
  114. Klein, D.J.; Thorn, C.F.; Desta, Z.; Flockhart, D.A.; Altman, R.B.; Klein, T.E. PharmGKB summary: Tamoxifen pathway, pharmacokinetics. Pharmacogenet. Genom. 2013, 23, 643–647. [Google Scholar] [CrossRef] [Green Version]
  115. Sanchez-Spitman, A.; Dezentje, V.; Swen, J.; Moes, D.; Bohringer, S.; Batman, E.; van Druten, E.; Smorenburg, C.; van Bochove, A.; Zeillemaker, A.; et al. Tamoxifen pharmacogenetics and metabolism: Results from the prospective CYPTAM study. J. Clin. Oncol. 2019, 37, 636–646. [Google Scholar] [CrossRef]
  116. Junsaeng, D.; Anukunwithaya, T.; Songvut, P.; Sritularak, B.; Likhitwitayawuid, K.; Khemawoot, P. Comparative pharmacokinetics of oxyresveratrol alone and in combination with piperine as a bioenhancer in rats. BMC Complement. Altern. Med. 2019, 19, 235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Balis, F.M.; Holcenberg, J.S.; Bleyer, W.A. Clinical pharmacokinetics of commonly used anticancer drugs. Clin. Pharm. 1983, 8, 202–232. [Google Scholar] [CrossRef] [PubMed]
  118. Marsh, S.; McLeod, H.; Dolan, E.; Shukla, S.J.; Rabik, C.A.; Gong, L.; Hernandez-Boussard, T.; Lou, X.J.; Klein, T.E.; Altman, R.B. Platinum pathway. Pharm. Genom. 2009, 19, 563–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Kesarwani, K.; Gupta, R. Bioavailability enhancers of herbal origin: An overview. Asian Pac. J. Trop. Biomed. 2013, 3, 253–266. [Google Scholar] [CrossRef] [Green Version]
  120. Hengjumrut, P.; Anukunwithaya, T.; Tantisira, M.H.; Tantisira, B.; Khemawoot, P. Comparative pharmacokinetics between madecassoside and asiaticoside presented in a standardised extract of Centella asiatica, ECa 233 and their respective pure compound given separately in rats. Xenobiotica 2018, 48, 18–27. [Google Scholar] [CrossRef] [PubMed]
  121. Songvut, P.; Chariyavilaskul, P.; Tantisira, M.H.; Khemawoot, P. Safety and pharmacokinetics of standardized extract of Centella asiatica (ECa 233) capsules in healthy Thai volunteers: A phase 1 clinical study. Planta Med. 2019, 85, 483–490. [Google Scholar] [CrossRef]
  122. Temeesak, N.; Kheokasem, N.; Phatcharawongsagorn, N.; Nontakulwiwat, P.; Boonmuang, P.; Santimaleeworagun, W.; Nulsopapon, P. The effects of herbs or dietary supplements on international normalized ratio in warfarin users: A retrospective study at Phramongkutklao hospital. Thai Pharm. Health Sci. J. 2015, 10, 139–146. [Google Scholar]
  123. Cox, M.C.; Low, J.; Lee, J.; Walshe, J.; Denduluri, N.; Berman, A.; Permenter, M.G.; Petros, W.P.; Price, D.K.; Figg, W.D.; et al. Influence of garlic (Allium sativum) on the pharmacokinetics of docetaxel. Clin. Cancer Res. 2006, 12, 4636–4640. [Google Scholar] [CrossRef] [Green Version]
  124. Lai, Y.H.; Yu, S.L.; Chen, H.Y.; Wang, C.C.; Chen, H.W.; Chen, J.J. The HLJ1-targeting drug screening identified Chinese herb andrographolide that can suppress tumour growth and invasion in non-small-cell lung cancer. Carcinogenesis 2013, 34, 1069–1080. [Google Scholar] [CrossRef] [Green Version]
  125. Sheeja, K.; Guruvayoorappan, C.; Kuttan, G. Antiangiogenic activity of Andrographis paniculata extract and andrographolide. Int. Immunopharmacol. 2007, 7, 211–221. [Google Scholar] [CrossRef]
  126. Tan, B.L.; Norhaizan, M.E. Curcumin combination chemotherapy: The implication and efficacy in cancer. Molecules 2019, 24, 2527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Samuels, N.; Ben-Arye, E. Exploring herbal medicine use during palliative cancer care: The integrative physician as a facilitator of pharmacist–patient–oncologist communication. Pharmaceuticals 2020, 13, 455. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Characteristics of anticancer drugs: (A) mechanism of action; (B) metabolic pathways; (C) phase I biotransformation; and (D) cytochrome P450 (CYP) isoforms responsible for metabolism.
Figure 1. Characteristics of anticancer drugs: (A) mechanism of action; (B) metabolic pathways; (C) phase I biotransformation; and (D) cytochrome P450 (CYP) isoforms responsible for metabolism.
Pharmaceuticals 15 00146 g001
Figure 2. Characteristics of Thai herbs: (A) plant families; (B) plant parts used; (C) bioactive components; and (D) potential interactions. Most herbs could inhibit cytochrome P450 (CYP) isoforms and P-glycoprotein; 10% of herbs could inhibit one or more CYP isoform, while inducing other CYP isoforms.
Figure 2. Characteristics of Thai herbs: (A) plant families; (B) plant parts used; (C) bioactive components; and (D) potential interactions. Most herbs could inhibit cytochrome P450 (CYP) isoforms and P-glycoprotein; 10% of herbs could inhibit one or more CYP isoform, while inducing other CYP isoforms.
Pharmaceuticals 15 00146 g002
Figure 3. In vitro (A) and in vivo (B) experiments of cancer cells used in Thai herbs studies.
Figure 3. In vitro (A) and in vivo (B) experiments of cancer cells used in Thai herbs studies.
Pharmaceuticals 15 00146 g003
Figure 4. Major compounds found in commonly used Thai herbs.
Figure 4. Major compounds found in commonly used Thai herbs.
Pharmaceuticals 15 00146 g004
Figure 5. Selection process of anticancer drugs and Thai herbs for the development of DHI information.
Figure 5. Selection process of anticancer drugs and Thai herbs for the development of DHI information.
Pharmaceuticals 15 00146 g005
Table 1. Potential interactions of drug metabolizing enzyme and transporter activities by Thai herbs.
Table 1. Potential interactions of drug metabolizing enzyme and transporter activities by Thai herbs.
Thai HerbsPotential InteractionsReferences
Acorus calamus
-
N/A
Aegle marmelos
-
CYP3A4 and CYP1A2 inhibition
[14]
Albizia procera
-
N/A
Allium ascalonicum
-
N/A
Allium sativum
-
CYP1A, CYP2B, CYP2C, CYP2E1, CYP3A induction
-
CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4, CYP3A5, P-glycoprotein inhibition
-
Reduce cyclophosphamide-induced developmental toxicity
-
Interact with tamoxifen
[15,16,17,18,19,20,21,22,23,24,25]
Andrographis paniculata
-
Potent CYP2A4 and CYP2B9 induction (Andrographolide)
-
CYP1A2, CYP2B1, CYP2C, CYP2C9, CYP2C19, CYP2C11, CYP2D6, CYP3A, CYP3A1, CYP3A4, UGT1A1, UGT1A3, UGT1A6, UGT1A7, UGT1A8, UGT1A10, UGT2B7, and P-glycoprotein inhibition
-
Strong synergistic induction of CYP1A1 and CYP1B1 expression (Combination of Andrographolide and CYP1A1 inducers)
-
Synergistic effects on anticancer activity of 5-FU, arsenic trioxide, bleomycin, carboplatin, cisplatin, doxorubicin, gemcitabine, paclitaxel, topotecan, and vincristine
[26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45]
Anethum graveolens
-
CYP3A4 inhibition
[19]
Angelica dahurica
-
N/A
Angelica sinensis
-
CYP2D6, CYP3A4, CYP1A2 induction and CYP2E1, CYP3A inhibition
[46,47,48]
Arcangelisia flava
-
N/A
Areca catechu
-
CYP3A4 inhibition
[49]
Artemisia annua
-
CYP1A1, CYP3A4, moderate CYP1A2, CYP2C19, CYP3A inhibition, and weak CYP2E1inhibition
[49,50,51]
Atractylodes lancea
-
Potent CYP1A2 inhibition, moderate CYP2E1 and CYP2C19 inhibition, low CYP2D6 and CYP3A4 inhibition
[52,53]
Aucklandia lappa
-
N/A
Caesalpinia bonduc
-
N/A
Capsicum annuum
-
CYP3A4 and CYP2C9 inhibition
-
Potent P-glycoprotein inhibition
-
Increase daunorubicin and vinblastine accumulation in cancer cells and increases anticancer activity of the drugs in KB-C2 cells
-
Synergistic effects on anticancer activity of 5-FU, cisplatin, docetaxel, erlotinib, and paclitaxel
[19,54,55,56,57,58,59]
Carum carvi
-
CYP2C9 and CYP3A4 inhibition
-
UGT1A1 induction
[19,60]
Cassia fistula
-
N/A
Centella asiatica
-
CYP1A2, CYP2B1, CYP2B2, CYP2C19, CYP2C9, CYP2D6, CYP2E1, CYP3A inhibition
[38,61,62,63]
Cissus quadrangularis
-
N/A
Citrus hystrix
-
CYP3A4 and P-glycoprotein inhibition
[64]
Clerodendrum indicum
-
N/A
Clinacanthus nutans
-
N/A
Cuminum cyminum
-
CYP2C9 and CYP3A4 inhibition
[19]
Curcuma longa
-
CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A, CYP3A4 and P-glycoprotein inhibition
[19,65,66]
Curcuma spp.
-
N/A
Cyanthillium cinereum (Vernonia cinerea)
-
CYP1A2, CYP2A6, and CYP2D6 inhibition
[67]
Dracaena cochinchinensis
-
N/A
Eurycoma longifolia
-
CYP2C8 inhibition, weak CYP1A2, CYP2A6, and CYP2C19 inhibition
[68,69]
Ficus racemosa
-
N/A
Foeniculum vulgare
-
CYP2C9, CYP3A4, CYP1A2, CYP2D6 and CYP2E1 inhibition
[19,42,70,71]
Gynostemma pentaphyllum
-
CYP2D6 (major), CYP2C8, CYP3A4, and CYP2C9 inhibition
[72]
Harrisonia perforata
-
N/A
Hibiscus sabdariffa
-
weak CYP1A2, CYP2C8, CYP2D6, CYP2B6, CYP2E1, CYP2C19, CYP3A4, CYP2C9, and CYP2A6 inhibition
[73]
Hyptis suaveolens
-
N/A
Kaempferia parviflora
-
CYP2D6, CYP1A2, and CYP3A4 inhibition
[74,75]
Lepidium sativum
-
N/A
Ligusticum sinense
-
N/A
Mesua ferrea
-
P-glycoprotein inhibition
[76]
Mimusops elengi
-
N/A
Momordica charantia
-
CYP2C9 and P-glycoprotein inhibition
[17,77,78]
Moringa oleifera
-
CYP1A2 inhibition
[79,80]
Morus alba
-
CYP3A4, CYP2D6, P-glycoprotein inhibition, and CYP3A4 induction
[52,74,81,82,83]
Murdannia loriformis
-
N/A
Nardostachys jatamansi
-
N/A
Nelumbo nucifera
-
CYP1A2, CYP2C19, CYP2C9, CYP2D6, CYP2E1 and CYP3A4 inhibition
[84,85,86]
Neopicrorhiza scrophulariiflora
-
N/A
Nigella sativa
-
CYP1A2, CYP2C9, and CYP3A4, and CYP2C19inhibition (Thymoquinone)
-
Synergistic effects on anticancer activity of 5-FU, cyclophosphamide, doxorubicin, gemcitabine, and topotecan
[87,88,89,90,91,92,93,94,95]
Ocimum sanctum
-
N/A
Orthosiphon aristatus (Orthosiphon stamineus)
-
CYP2C19, CYP2C9, CYP2D6, CYP3A4, UGT1A7, UGT1A1, UGT1A6 and UGT1A8 inhibition
-
P-glycoprotein inhibition results in decreasing resistance of KB-V-1 cells to vinblastine
[32,38,74,82,96,97]
Phyllanthus emblica
-
Weak CYP1A2, CYP2C9, CYP2D6, CYP2E1, CYP3A4 inhibition, P-glycoprotein inhibition, and synergistic growth inhibitory effect with cisplatin and doxorubicin
[98,99,100]
Pimpinella anisum
-
CYP2C9 and CYP3A4 inhibition
[19]
Piper betle
-
N/A
Piper nigrum
-
CYP2C9 and CYP3A4 inhibition
-
P-glycoprotein, MRP1 and BCRP1 transporter inhibition
[17,19,42,101,102]
Piper retrofractum
-
N/A
Piper sarmentosum
-
N/A
Piper wallichii
-
N/A
Plantago ovata
-
N/A
Pterocarpus santalinus
-
N/A
Santalum album
-
CYP3A4 and CYP2D6 inhibition
[42]
Senna alata(Casssia alata)
-
CYP1A2, CYP2C19, CYP2D6, CYP3A4 inhibition
[74,77,103]
Senna garrettiana(Cassia garrettiana)
-
N/A
Senna tora(Cassia tora)
-
N/A
Solanum trilobatum
-
P-glycoprotein inhibition
[99]
Solori scandens(Derris scandens)
-
N/A
Tarlmounia elliptica
-
N/A
Terminalia bellirica
-
Synergistic effects on growth inhibitory effects of cisplatin in A549 cells and doxorubicin in HepG2 cells
[100]
Terminalia chebula
-
CYP2E1 and CYP2C19 inhibition
[104]
Thunbergia laurifolia
-
CYP1A4, CYP2D6 and CYP3A4 inhibition
[74,82,97,105]
Tiliacora triandra
-
N/A
Tinospora crispa
-
CYP3A4 and CYP2D6 inhibition
[42]
Trachyspermum ammi
-
CYP2C9 and CYP3A4 inhibition
[19]
Zingiber montanum (Zingiber cassumunar)
-
CYP2D6 and CYP3A4 inhibition
[42]
Zingiber officinale
-
N/A
Zingiber zerumbet(Zingiber aromaticum)
-
CYP2D6 and CYP3A4 inhibition
[42]
N/A, Not available.
Table 2. Pharmacokinetics-based anticancer-herb interactions with Thai herbs.
Table 2. Pharmacokinetics-based anticancer-herb interactions with Thai herbs.
Thai HerbsEffects of Thai Herbal
Products
Potential Drug
Interaction
Possible Effects on
Anticancer Drugs
References
Aegle marmelosCYP1A2 inhibition
In vitro: Methanolic extract of
Aegle marmelos inhibits CYP1A2 with IC50 = 0.8 μg/mL.
Dasatinib
Imatinib
Increase concentrations[14]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Methanolic extract of
Aegle marmelos inhibits CYP3A4 in pooled human liver microsomes with IC50 = 5 μg/mL.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[14]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Allium sativumCYP1A2 inhibition
In vitro: Allicin inhibits CYP1A2 with IC50 = 44.22 µM.
Dasatinib
Imatinib
Increase concentrations[25]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Allicin, apigenin and myricetin inhibit CYP3A4 with IC50 = 43.73, 0.4, and 44.5 μM, respectively.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[20,25]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
CYP2C9 inhibition
In vitro: Allicin, apigenin, and myricetin inhibit CYP2C9 with IC50 = 5.41, 6.4, and 32.1 μM, respectively.
Dasatinib
Imatinib
Increase concentrations[20,25]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
CYP2C19 inhibition
In vitro: Allicin inhibits CYP1A2 with IC50 = 3.52 µM.
ImatinibIncrease concentrations[25]
TamoxifenDecrease levels of active
metabolites
CYP2D6 inhibition
In vitro: Allicin inhibits CYP1A2 with IC50 = 47.10 µM.
Doxorubicin
Imatinib
Increase concentrations[25]
TamoxifenDecrease levels of active
metabolites
Andrographis
paniculata
CYP1A2 inhibition
In vitro: Extract of Andrographis
paniculata inhibits CYP1A2 with IC50 = 5.1 μg/mL.
Dasatinib
Imatinib
Increase concentrations[39,40]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
CYP2C19 inhibition
In vitro: Ethanolic extract of Andrographis paniculata inhibits CYP2C19 with IC50 = 91.7 μg/mL.
ImatinibIncrease concentrations[38]
TamoxifenDecrease levels of active
metabolites
UGT1A1 inhibition
In vitro: Ethanolic extract of Andrographis paniculata inhibits UGT1A1 with IC50 = 5.00 µg/mL.
Etoposide
Dasatinib
Increase concentrations[32]
UGT2B7 inhibition
In vitro: Spray-dried 50% methanolic powder of Andrographis paniculata inhibits UGT2B7 with IC50 = 2.82 µg/mL.
TamoxifenDecrease levels of active
metabolites
[32]
Anethum graveolensCYP3A4 inhibition
In vitro: 100 µg/mL of Anethum graveolens extract inhibit CYP3A4 with percent inhibition more than 50%.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Angelica sinensisCYP3A4 induction
In vivo: Ethanolic crude extract,
ligustilide, linoleic acid, ferulic acid, and beta-sitosterol from Angelica sinensis induces CYP3A4 activity in HepG2 cells with maximum induction at 118 ± 2.26%
relative rifampin.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Decrease concentration[48]
Cyclophosphamide
Ifosfamide
Tamoxifen
Increase levels of active
metabolites
Areca catechuCYP3A4 inhibition
In vitro: 100 μg/mL of Areca catechu aqueous extracts inhibits CYP3A4 with percent inhibition 85%
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[49]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Carum carviCYP2C9 inhibition
In vitro: 100 μg/mL of Carum carvi extract inhibits CYP2C9 with percent inhibition more than 50%.
Dasatinib
Imatinib
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
CYP3A4 inhibition
In vitro: 100 μg/mL of Carum carvi extract inhibits CYP3A4 with percent inhibition more than 50%.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Centella asiaticaCYP2C19 inhibition
In vitro: Dichloromethane extract of
Centella asiatica inhibits CYP2C19 with IC50 = 30.2 μg/mL.
ImatinibIncrease concentrations[38]
TamoxifenDecrease levels of active
metabolites
CYP2C9 inhibition
In vitro: Ethanolic extract of Centella asiatica inhibits CYP2C9 with IC50 = 48.41 ± 4.64 μg/mL.
Dasatinib
Imatinib
Increase concentrations[63]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
CYP1A2 inhibition
In vitro: Ethanolic extract of Centella asiatica inhibits CYP1A2 with IC50 = 42.23 ± 3.65 μg/mL.
Dasatinib
Imatinib
Increase concentrations[63]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
Cuminum cyminumCYP2C9 inhibition
In vitro: 100 µg/mL of Cuminum cyminum extract inhibits CYP2C9 with percent inhibition more than 50%.
Dasatinib
Imatinib
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Decrease levels of active metabolites
CYP3A4 inhibition
In vitro: 100 µg/mL of Cuminum cyminum extract inhibits CYP3A4 with percent inhibition more than 75%.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Curcuma longaCYP1A2 inhibition
In vitro: Curcumin inhibits CYP1A2 with IC50 = 40 µM.
Dasatinib
Imatinib
Increase concentrations[65]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
CYP2C9 inhibition
In vitro: Curcumin inhibits CYP2C9 with IC50 = 14.8 µg/mL.
Aqueous extract of Curcuma longa inhibits CYP2C9 with IC50 = 82.3 ± 6.05 µg/mL.
Dasatinib
Imatinib
Increase concentrations[19,66]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Extract of Curcuma longa inhibits CYP3A4 with IC50 = 17 µg/mL.
Curcumin inhibits CYP3A4 with IC50 = 16.3 µM.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[19,65]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Cyanthillium
cinereum
(Vernonia cinerea)
CYP2A6 inhibition
In vitro: Flavonoid chrysoeriol inhibits CYP2A6 with Ki = 1.93 ± 0.05 µM,
hirsutinolides inhibits CYP2A6 with IC50 = 12–23 μM.
Letrozole
Tamoxifen
Increase concentrations[67]
IfosfamideDecrease levels of active
metabolites
CYP1A2 inhibition
In vitro: Flavonoid chrysoeriol inhibits CYP1A2 with Ki = 3.39 ± 0.21 μM.
Dasatinib
Imatinib
Increase concentrations[67]
Dacarbazine
Flutamide
Decrease levels of active
metabolites
CYP2D6 inhibition
In vitro: Hirsutinolides inhibits CYP2D6 with IC50 = 15–41 μM.
Doxorubicin
Imatinib
Increase concentrations[67]
TamoxifenDecrease levels of active
metabolites
Foeniculum vulgareCYP2C9 inhibition
In vitro: 100 µg/mL of Foeniculum vulgare extract inhibits CYP2C9 with percent inhibition more than 75%.
Dasatinib
Imatinib
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
CYP2D6 inhibition
In vitro: Water extract of Foeniculum vulgare inhibits CYP2D6 with IC50 = 23 ± 2 µg/mL.
Doxorubicin
Imatinib
Increase concentrations[70]
TamoxifenDecrease levels of active
metabolites
CYP2E1 inhibition
In vitro: Water extract of Foeniculum vulgare inhibits CYP2E1 with IC50 = 23 ± 4 µg/mL.
Dacarbazine
Tamoxifen
Decrease levels of active
metabolites
[71]
CYP3A4 inhibition
In vitro: 100 µg/mL of Foeniculum vulgare extract inhibits CYP3A4 with percent inhibition more than 75%,
water extract of Foeniculum vulgare inhibits CYP3A4 with IC50 = 40 ± 4 µg/mL.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[19,70]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Gynostemma
pentaphyllum
CYP2D6 inhibition
In vitro: Gypenosides inhibit CYP2D6 with IC50 = 1.61 µg/mL.
Doxorubicin
Imatinib
Increase concentrations[72]
TamoxifenDecrease levels of active
metabolites
CYP2C8 inhibition
In vitro: Gypenosides inhibit CYP2C8 with IC50 = 20.06 µg/mL.
Nilotinib
Paclitaxel
Tamoxifen
Increase concentrations[72]
Ifosfamide
Imatinib
Decrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Gypenosides inhibit CYP3A4 with IC50 = 34.76 µg/mL.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[72]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
CYP2C9 inhibition
In vitro: Gypenosides inhibit CYP2C9 with IC50 = 54.52 µg/mL.
Dasatinib
Imatinib
Tamoxifen
Increase concentrations[72]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
Kaempferia
parviflora
CYP1A2 inhibition
Patients who used extract from Kaempferia parviflora showed CYP1A2 inhibition. It also showed interaction with fluoxetine.
Dasatinib
Imatinib
Increase concentrations[75]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
CYP2D6 inhibition
In vitro: Ethanolic extract of Kaempferia parviflora inhibits CYP2D6 with
IC50 = 77 ± 9.54 µg/mL.
Doxorubicin
Imatinib
Increase concentrations[74]
TamoxifenDecrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Ethanolic extract of Kaempferia parviflora inhibits CYP3A4 with
IC50 = 28 ± 19.5 µg/mL.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[74]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Moringa oleiferaCYP1A2 inhibition
In vitro: Ethanolic extract inhibits CYP1A2 with IC50 = 13.8 ± 9.8 µg/mL.
Dasatinib
Imatinib
Increase concentrations[80]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
Nelumbo nuciferaCYP2C9 inhibition
In vitro: Alkaloid fraction of Nelumbo nucifera inhibits CYP2C9 with IC50 = 52.58 µg/mL.
Dasatinib
Imatinib
Increase concentrations[84]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
CYP2C19 inhibition
In vitro: Ethanolic extract of Nelumbo nucifera inhibits CYP2C19 with IC50 = 77.38 µg/mL.
Alkaloid fraction of Nelumbo nucifera inhibits CYP2C19 with IC50 = 40.79 µg/mL.
ImatinibIncrease concentrations[84]
TamoxifenDecrease levels of active
metabolites
CYP2D6 inhibition
In vitro: Extract of Nelumbo nucifera inhibits CYP2D6 with IC50 = 12.05 µg/mL.
Alkaloid fraction of Nelumbo nucifera
inhibits CYP2D6 with IC50 = 0.96 µg/mL.
In vivo: Alkaloid fraction of Nelumbo nucifera inhibits CYP2D6 in rat.
Doxorubicin
Imatinib
Increase concentrations[84,108]
TamoxifenDecrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Extract of Nelumbo nucifera inhibits CYP3A4 with IC50 = 15.7 ± 2.1 µg/mL.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[85]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Nigella sativaCYP1A2 inhibition
In vitro: Thymoquinone inhibits CYP1A2 with IC50 26.5 ± 2.9 µM
Dasatinib
Imatinib
Increase concentrations[88]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
CYP2C9 inhibition
In vitro: Thymoquinone inhibits CYP2C9 with IC50 0.5 ± 0.4 µM
Dasatinib
Imatinib
Increase concentrations[88]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Thymoquinone inhibits CYP3A4 with IC50 25.2 ± 3.1 µM
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[88]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
CYP2C19 inhibition
In vitro: Thymoquinone inhibits CYP2C19 with IC50 3.6 ± 0.9 µM
ImatinibIncrease concentrations[91]
TamoxifenDecrease levels of active
metabolites
Orthosiphon aristatus
(Orthosiphon stamineus)
CYP2C19 inhibition
In vitro: Petroleum ether extract of Orthosiphon aristatus inhibits CYP2C19 with IC50 = 67.1 μg/mL.
Sinensetin and eupatorin, active compounds of Orthosiphon aristatus, inhibit CYP2C19 with IC50 = 71.6 and 12.1 µg/mL, respectively.
ImatinibIncrease concentrations[38]
TamoxifenDecrease levels of active
metabolites
CYP2D6 inhibition
In vitro: Ethanolic extract of Orthosiphon aristatus inhibits CYP2D6 with IC50 = 31.0 ± 19.5 µg/mL.
Eupatorin, an active compound of Orthosiphon aristatus, inhibits CYP2D6 with IC50 = 3.8 µg/mL.
Doxorubicin
Imatinib
Increase concentrations[74,96]
TamoxifenDecrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Dichloromethane and petroleum ether extracts of Orthosiphon aristatus inhibit CYP3A4 with IC50 = 96.5 and 46.3 µg/mL, respectively.
Ethanolic extract of Orthosiphon aristatus inhibits CYP3A4 with
IC50 = 40 ± 8.7 µg/mL.
Rosmarinic acid and eupatorin, active compounds of Orthosiphon aristatus, inhibit CYP3A4 with IC50 = 86.9 and 5.0 µg/mL, respectively.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[74,96]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active metabolites
UGT1A1 inhibition
In vitro: Spray-dried 50% methanolic powder of Orthosiphon aristatus inhibits UGT1A1 with IC50 = 24.65 µg/mL.
Etoposide
Dasatinib
Increase concentrations[32]
Pimpinella anisumCYP3A4 inhibition
In vitro: 100 µg/mL of Pimpinella anisum extract inhibits CYP3A4 with percent inhibition more than 50%.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Piper nigrumCYP2C9 inhibition
In vitro: Black pepper and white pepper extracts inhibit CYP2C9 with
IC50 = 12.1 and 3.2 µg/mL, respectively.
Dasatinib
Imatinib
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Decrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Black pepper and white pepper extracts inhibit CYP3A4 with IC50 = 4.1 and 1.0 µg/mL, respectively.
Methanolic extract from Piper nigrum leaves and fruits inhibit CYP3A4 with IC50 = 25 and 29 µg/mL, respectively.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[19,42]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Senna alata
(Casssia alata)
CYP1A2 inhibition
In vitro: Water extract powder of Senna alata inhibits CYP1A2 with IC50 = 28.3 ± 2.42 µg/mL.
Dasatinib
Imatinib
Increase concentrations[77]
Dacabarzine
Flutamide
Decrease levels of active
metabolites
CYP2D6 inhibition
In vitro: Ethanolic extract of Senna alata inhibits CYP2D6 with IC50 = 33.0 ± 25.6 µg/mL.
Doxorubicin
Imatinib
Increase concentrations[74,77]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Ethanolic extract of Senna alata inhibits CYP3A4 with IC50 = 24.3 ± 14.3 µg/mL.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[74]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active metabolites
Trachyspermum ammiCYP3A4 inhibition
In vitro: 100 µg/mL of Trachyspermum ammi extract inhibits CYP3A4 with percent inhibition more than 50%.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[19]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Thunbergia laurifoliaCYP2D6 inhibition
In vitro: Ethanolic extract of Thunbergia laurifolia inhibits CYP2D6 with IC50 = 45.0 ± 5.0 µg/mL.
Doxorubicin
Imatinib
Increase concentrations[74]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Zingiber montanum
(Zingiber cassumunar)
CYP2D6 inhibition
In vitro: Extract of Zingiber montanum inhibits 25% of CYP2D6 when compare with Quinidine.
Doxorubicin
Imatinib
Increase concentrations[42]
TamoxifenDecrease levels of active
metabolites
CYP3A4 inhibition
In vitro: Extract of Zingiber montanum inhibits 50% of CYP3A4 when compare with Ketoclonazole.
Dasatinib
Docetaxel
Doxorubicin
Etoposide
Imatinib
Letrozole
Megestrol
Nilotinib
Paclitaxel
Vinblastine
Vincristine
Vinorelbine
Increase concentrations[42]
Cyclophosphamide
Ifosfamide
Tamoxifen
Decrease levels of active
metabolites
Table 3. Lists of anticancer drugs and Thai herbs utilized for the determination of potential DHIs.
Table 3. Lists of anticancer drugs and Thai herbs utilized for the determination of potential DHIs.
Anticancers in 2020 Thailand NLEMThai Herbs in 2020 THP
Alkylating drugs
  • Busulfan
  • Chlorambucil
  • Cyclophosphamide
  • Melphalan
  • Carmustine
  • Ifosfamide
  • Procarbazine
Cytotoxic antibiotics
8.
Bleomycin
9.
Dactinomycin
10.
Doxorubicin hydrochloride
11.
Idarubicin hydrochloride
12.
Mitomycin
13.
Mitoxantrone hydrochloride
Antimetabolites
14.
Cytarabine
15.
Fluorouracil
16.
Mercaptopurine
17.
Methotrexate
18.
Capecitabine
19.
Fludarabine phosphate
20.
Gemcitabine hydrochloride
21.
Oxaliplatin
22.
Tegafur + uracil
23.
Tioguanine
Vinca alkaloids and etoposide
24.
Etoposide
25.
Vinblastine
26.
Vincristine
27.
Vinorelbine
Other antineoplastic drugs
28.
Asparaginase
29.
Cisplatin
30.
Carboplatin
31.
Hydroxycarbamide
32.
Arsenic trioxide
33.
Leucovorin calcium
34.
Dacarbazine
35.
Mitotane
36.
Tretinoin
37.
Paclitaxel
38.
Topotecan
39.
Docetaxel
40.
Erlotinib
41.
Imatinib
42.
Nilotinib
43.
Dasatinib
44.
Rituximab
45.
Trastuzumab
Sex hormones and hormone antagonists in malignant diseases
46.
Tamoxifen
47.
Letrozole
48.
Megestrol
49.
Flutamide
50.
Ketoconazole
51.
Leuprorelin
52.
Triptorelin
1.
Acorus calamus
2.
Aegle marmelos
3.
Albizia procera
4.
Allium ascalonicum
5.
Allium sativum
6.
Andrographis paniculata
7.
Anethum graveolens
8.
Angelica dahurica
9.
Angelica sinensis
10.
Arcangelisia flava
11.
Areca catechu
12.
Artemisia annua
13.
Atractylodes lancea
14.
Aucklandia lappa
15.
Caesalpinia bonduc
16.
Capsicum annuum
17.
Carum carvi
18.
Cassia fistula
19.
Centella asiatica
20.
Cissus quadrangularis
21.
Citrus hystrix
22.
Clerodendrum indicum
23.
Clinacanthus nutans
24.
Cuminum cyminum
25.
Curcuma longa
26.
Curcuma spp.
27.
Cyanthillium cinereum
28.
Dracaena cochinchinensis
29.
Eurycoma longifolia
30.
Ficus racemosa
31.
Foeniculum vulgare
32.
Gynostemma pentaphyllum
33.
Harrisonia perforata
34.
Hibiscus sabdariffa
35.
Hyptis suaveolens
36.
Kaempferia parviflora
37.
Lepidium sativum
38.
Ligusticum sinense
39.
Mesua ferrea
40.
Mimusops elengi
41.
Momordica charantia
42.
Moringa oleifera
43.
Morus alba
44.
Murdannia loriformis
45.
Nardostachys jatamansi
46.
Nelumbo nucifera
47.
Neopicrorhiza scrophulariiflora
48.
Nigella sativa
49.
Ocimum sanctum
50.
Orthosiphon aristatus
51.
Phyllanthus emblica
52.
Pimpinella anisum
53.
Piper betle
54.
Piper nigrum
55.
Piper retrofractum
56.
Piper sarmentosum
57.
Piper wallichii
58.
Plantago ovata
59.
Pterocarpus santalinus
60.
Santalum album
61.
Senna alata
62.
Senna garrettiana
63.
Senna tora
64.
Solanum trilobatum
65.
Solori scandens
66.
Tarlmounia elliptica
67.
Terminalia bellirica
68.
Terminalia chebula
69.
Thunbergia laurifolia
70.
Tiliacora triandra
71.
Tinospora crispa
72.
Trachyspermum ammi
73.
Zingiber montanum
74.
Zingiber officinale
75.
Zingiber zerumbet
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jiso, A.; Khemawoot, P.; Techapichetvanich, P.; Soopairin, S.; Phoemsap, K.; Damrongsakul, P.; Wongwiwatthananukit, S.; Vivithanaporn, P. Drug-Herb Interactions among Thai Herbs and Anticancer Drugs: A Scoping Review. Pharmaceuticals 2022, 15, 146. https://doi.org/10.3390/ph15020146

AMA Style

Jiso A, Khemawoot P, Techapichetvanich P, Soopairin S, Phoemsap K, Damrongsakul P, Wongwiwatthananukit S, Vivithanaporn P. Drug-Herb Interactions among Thai Herbs and Anticancer Drugs: A Scoping Review. Pharmaceuticals. 2022; 15(2):146. https://doi.org/10.3390/ph15020146

Chicago/Turabian Style

Jiso, Apisada, Phisit Khemawoot, Pinnakarn Techapichetvanich, Sutinee Soopairin, Kittiphong Phoemsap, Panrawee Damrongsakul, Supakit Wongwiwatthananukit, and Pornpun Vivithanaporn. 2022. "Drug-Herb Interactions among Thai Herbs and Anticancer Drugs: A Scoping Review" Pharmaceuticals 15, no. 2: 146. https://doi.org/10.3390/ph15020146

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop