Planta Med 2012; 78(13): 1428-1457
DOI: 10.1055/s-0031-1298536
Mechanism of Herb-Drug Interactions
Reviews
Georg Thieme Verlag KG Stuttgart · New York

Phenotyping Studies to Assess the Effects of Phytopharmaceuticals on In Vivo Activity of Main Human Cytochrome P450 Enzymes

Gregor Zadoyan
1   ITECRA GmbH & Co. KG, Cologne, Germany
,
Uwe Fuhr
1   ITECRA GmbH & Co. KG, Cologne, Germany
2   Department of Pharmacology, Clinical Pharmacology Unit, University of Cologne, Cologne, Germany
› Institutsangaben
Weitere Informationen

Correspondence

Prof. Dr. med. Uwe Fuhr
Department of Pharmacology, Clinical Pharmacology Unit, University of Cologne
Gleueler Str. 24
50931 Cologne
Germany
Telefon: +49 22 14 78 52 30   
Fax: +49 22 14 78 70 11   

Publikationsverlauf

received 25. Januar 2012
revised 11. April 2012

accepted 15. April 2012

Publikationsdatum:
15. Mai 2012 (online)

 

Abstract

The extensive use of herbal drugs and their multiple components and modes of action suggests that they may also cause drug interactions by changing the activity of human cytochrome P450 enzymes. The purpose of the present review is to present the available data for the top 14 herbal drug sales in the U. S. Studies describing the effects of herbal drugs on phenotyping substrates for individual CYPs were identified by a comprehensive MEDLINE search. Drugs included Allium sativum (Liliaceae), Echinacea purpurea (Asteraceae), Serenoa repens (Arecaceae), Ginkgo biloba (Ginkgoaceae), Vaccinium macrocarpon (Ericaceae), Glycine max (Fabaceae), Panax ginseng (Araliaceae), Actea racemosa (Ranunculaceae), Hypericum perforatum (Hypericaceae), Silybum marianum (Asteraceae), Camellia sinensis (Theaceae), Valeriana officinalis (Valerianaceae), Piper methysticum (Piperaceae), and Hydrastis canadensis (Ranunculaceae) preparations. We identified 70 clinical studies in 69 publications. The majority of the herbal drugs appeared to have no clear effects on most of the CYPs examined. If there was an effect, there was mild inhibition in almost all cases, as seen with garlic or kava effects on CYP2E1 and with soybean components on CYP1A2. The most pronounced effects were induction of CYP3A and other CYPs by St. Johnʼs wort and the inhibitory effect of goldenseal on CYP3A and CYP2D6, both being borderline between mild and moderate in magnitude. With the exceptions of St. Johnʼs wort and goldenseal, the information currently available suggests that concomitant intake of the herbal drugs addressed here is not a major risk for drugs that are metabolized by CYPs.


#

Abbreviations

AUC(0-∞): area under the concentration-time curve from 0 to infinity

Cmax: maximum serum concentration

CI: confidence interval

CYP: cytochrome P450 enzyme

EGCG: epigallocatechin gallate

EMA: European Medicines Agency

FDA: Food and Drug Administration

GABA: gamma-aminobutyric acid

GBE: Ginkgo biloba extract

(d)GTE: (decaffeinated) green tea extract

INR: international normalized ratio of prothrombin time

LSS: limited sampling strategy

p: p value

Poly E: Polyphenon E®

rac: racemic

SJW: St. Johnʼs wort

t 1/2: elimination half-life


#

Introduction

Herbal drugs are claimed to exert their unique mode of action by the complex interplay of many different constituents. Beyond actions of various constituents on more than a single target, it is postulated to also arise from mutual interactions of related chemical moieties at both the pharmacokinetic and pharmacodynamic level. It is therefore not surprising that herbal drugs may also have an effect on the activity of cytochrome P450 enzymes (CYPs), the most important enzyme family to mediate phase I metabolism of small molecule drugs and other human xenobiotics. This became particularly obvious when St. Johnʼs wort was recognized to be a potent inducer of CYP3A [1] and other proteins involved in pharmacokinetics, and the detection of the inhibitory effects of grapefruit juice on gut wall CYP3A was a reminder that natural products may be involved in drug interactions [2].

Nowadays, there is general agreement that the potential of herbal drugs to cause drug-drug interactions needs to be assessed as thoroughly as for non-herbal drugs, including interactions at the level of CYP-mediated metabolism. In vitro studies with herbal drugs probably are less reliable than those with individual chemicals because both effective concentrations in vitro as well as in vivo processing of the complex mixtures can hardly be predicted. Because not all potential combinations with other drugs can be tested, the effect of drugs on individual enzymes is usually assessed using the phenotyping approach. Phenotyping for a CYP enzyme, i.e., quantification of its actual in vivo activity in an individual, is performed by administration of a selective substrate for this enzyme and subsequent determination of appropriate pharmacokinetic metrics closely reflecting enzyme activity [3]. Phenotyping substrates are usually marketed drugs with therapeutic indications, which are chosen mainly based on their selectivity for the CYP to be examined, on tolerability, and on availability. The standard design of a respective clinical study is a crossover design with administration of the phenotyping agents in both periods, combined with coadministration of the drug to be tested at its highest chronic therapeutic dose in one period [3]. Current regulatory guidance has adopted this procedure and also provides recommendations on individual phenotyping agents [4], [5].

It is beyond the scope of this review to evaluate all individual substances and metrics used to quantify the effects of herbal drugs on individual CYPs in detail; for a respective assessment, see [3]. CYP phenotyping drugs recommended by the FDA [4], [5] include the following: CYP1A2, theophylline, caffeine; CYP2B6, efavirenz, bupropion; CYP2C8, amodiaquine, cerivastatin, repaglinide, rosiglitazone; CYP2C9, warfarin, tolbutamide; CYP2C19, omeprazole, esoprazole, lansoprazole, pantoprazole; CYP2D6, metoprolol, desipramine, dextromethorphan, atomoxetine; CYP2E1, chlorzoxazone; CYP3A4, midazolam, buspirone, felodipine, lovastatin, eletriptan, sildenafil, simvastatin, triazolam. The EMA list is more limited, comments on the lack of exhaustive validation of some agents, and also occasionally asks for monitoring of specific reactions. The FDA created and posted an additional extensive list in 2011 [6] presenting sensitive substrates (as mentioned in the 1999 document “Guidance for Industry: In Vivo Drug Metabolism/Drug Interaction Studies – Study Design, Data Analysis, and Recommendations for Dosing and Labeling”, which formally is still valid today) and substrates with a narrow therapeutic range, but provides little advice on the selection of the individual agent. While caffeine, warfarin, tolbutamide, and midazolam, and most respective metrics derived from plasma concentrations may be considered as fully validated (with the limitations described above) [3], caveats apply for the other listed substances, as additional metabolic pathways are known and/or validation data are limited. Several substances present in EMA and FDA lists had been withdrawn from the market prior to generation of the lists.

A combination of substances in a phenotyping cocktail in order to address a panel of CYPs in a single clinical study has been studied extensively [3], [7], [8]. There should be no mutual interaction of the drugs within a cocktail; an excellent example how to study this in detail has been published [9]. The EMA guidance addresses cocktail studies, while for the FDA, only semiofficial information is available (see [10], [11]), showing that the FDA also accepts data from cocktail studies. While in general the cocktail approach appears to be appropriate, the inclusion of “historical” substances with limited validation and/or affinity to several CYPs such as dapsone or quinidine may compromise the results for other cocktail components.

Pharmacokinetic metrics, which reflect CYP activity best, may be metabolic ratios of a metabolite over a parent drug in plasma or urine, clearance of the parent drug, or partial clearance via a specific pathway, depending on the individual phenotyping substrate used. It is desirable to use metrics without the need of drawing many blood samples during the entire concentration vs. time profile of the phenotyping drug in order to keep burdens for both study participants and investigators as low as possible. Some metrics can indeed be derived using a limited sampling strategy (LSS) or even from a single sample [3]. However, the most recent 2010 EMA (draft) guidelines on the investigation of drug interactions (CPMP/EWP/560/95/Rev. 1 – Corr.) as well as the respective FDA guidance website [12] ask explicitly for the determination of complete area under the curve (AUC) values of phenotyping substrates, requiring at least 8–10 samples per study period, and discourages the use of single concentrations and metabolic ratios. The reason for this reluctance to accept metrics derived using an LSS probably is that their validation is limited to the quantification of baseline activity in individuals without major diseases with effects on pharmacokinetics and in the absence of factors with a major impact on pharmacokinetics. However, any effects of interacting drugs may not be limited to the activity of the CYP to be investigated, but may also affect absorption, secondary metabolism, and other pharmacokinetic processes of the phenotyping substrate. Because of their multiple ingredients, herbal drugs would be at risk to unexpectedly exert such additional effects. LLS-based metrics presumably are less robust to such effects, with the risk that these would either be erroneously identified as an effect on CYP activity, or that existing effects on CYPs would be hidden. While the confounding mechanism could then be identified by a change in the entire concentration vs. time profile of the phenotyping drug, these may well go unnoticed with LLS-based metrics only. Furthermore, LLS and complete sampling may also reach different results in interaction studies [13] because the concentration of interacting moieties and, thus, the extent of interaction change during a dosing interval – which is only partially covered by an LLS.

A second caveat concerns the estimation of the sample size required in interaction studies with herbal drugs. We repeatedly observed that the apparent intraindividual variability of phenotyping agent pharmacokinetics was clearly higher than reported in previous studies with small molecule drugs [14], [15]. The reason for this observation is unclear; one could speculate that nonspecific effects on gastrointestinal motility are involved. A higher safety margin when calculating the sample size appears to be the only way to handle this problem as long as the mechanisms are unclear.

The regulatory (i.e., safety) purpose of interaction studies with any drugs including herbal drugs is to investigate the maximal extent of interaction for maximal exposure towards the “perpetrator”. To this end, effects on phenotyping metrics should be larger rather than smaller than the true effects on CYP activity (sensitive metrics). In contrast, true effects on CYP activity for a given dose would theoretically be required to adapt the dose of concomitantly given drugs subject to this interaction. For the latter approach, changes in enzyme activity could then be incorporated in simulations to predict the extent of interactions with other drugs [11]; however, as these methods are at an early stage, the former approach currently still seems to be more appropriate.

In summary, the phenotyping approach is the method of choice to study the potential of drugs including herbal drugs to change CYP activity and thus the pharmacokinetics of their drug substrates, but much expertise and very close attention to the details of individual studies is required in order to understand the consequences of respective results. More research is needed especially to achieve better transferability of results to quantitatively predict effects of other drug substrates in treated patients.


#

Methods

The aim of the present review was to give a concise overview on herb-drug interactions in human clinical studies, especially phenotyping and phenotyping cocktail studies, with respect to the major human drug metabolizing cytochrome P450 enzymes.

A literature search was conducted in MEDLINE (as accessed via PubMed on December 2, 2011) using the search terms of the plantʼs name in combination with “cytochrome P450′′, plus “drug interaction” or “herb-drug interaction”. The search was limited to English and German language papers. For further information, reviews and additional publications from all reference lists were read and relevant data were extracted. The discussion of case reports, animal data as well as in vitro experiments were not the primary objective of the present review and were not included.


#

Results

We identified 70 clinical studies in 69 publications. An overview of the data is presented in [Table 1] (at the end of the paper). Herb-drug interactions were present for 10 of the top 14 selling botanicals in the U. S. for 2006, including garlic with 2 drugs (chlorzoxazone, saquinavir), echinacea with 3 drugs (caffeine, midazolam, tolbutamide), ginkgo with 3 drugs (midazolam, omeprazole, tolbutamide), soy with 2 drugs (caffeine, theophylline), ginseng with 1 drug (debrisoquine), black cohosh with 1 drug (debrisoquine), St. Johnʼs wort with 19 drugs (alprazolam, atorvastatin, chlorzoxazone, cortisol, cyclosporine, desogestrel, ethinyl estradiol, gliclazide, imatinib, indinavir, ivabradine, mephenytoin, midazolam, norethindrone, omeprazole, quazepam, verapamil, voriconazole, and warfarin), kava with 1 drug (chlorzoxazone), and goldenseal with 2 drugs (debrisoquine, midazolam).

Table 1 Reported clinical studies on the effects of herbal drugs and/or respective components on cytochrome P450 enzymes in humans.

Prescribed drug

Actually used probe drug for CYP isoform

Sample size

Dosage and duration of treatment

Preparation and content

Study design

Clinical result of interaction, magnitude of change in % of PK parameters (p value)

Possible mechanism, involved CYP

Ref.

Garlic

Alprazolam

Alprazolam

14

3 × 600 mg twice daily for 14 days

Coated tablets with 600 µg allicin

Phenotyping at baseline vs. end of herb treatment

No effect on alprazolam pharmacokinetics

None, CYP3A4

[25]

Caffeine

Caffeine

12

500 mg thrice daily for 28 days

Garlic oil

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on caffeine pharmacokinetics

None, CYP1A2

[27]

Chlorzoxazone

Chlorzoxazone

12

500 mg thrice daily for 28 days

Garlic oil

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

Decreased 6-hydroxychlorzoxazone/chlorzoxazone serum ratios, 22 % (p = 0.005)

Inhibition of CYP2E1

[27]

Debrisoquine

Debrisoquine

12

500 mg thrice daily for 28 days

Garlic oil

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on debrisoquine urinary recovery ratio

None, CYP2D6

[27]

Dextromethorphan

Dextromethorphan

14

3 × 600 mg twice daily for 14 days

Coated tablets with 600 µg allicin

Phenotyping at baseline vs. end of herb treatment

No effect on dextromethorpan pharmacokinetics

None, CYP2D6

[25]

Docetaxel

Docetaxel

10

600 mg twice daily for 12 days

Garlic tablets with 3600 µg allicin

Prospective pharmacokinetic study; garlic coadministration in 1st cycle of docetaxel treatment

No effect on docetaxel pharmacokinetics

None, CYP3A4

[23]

Midazolam

Midazolam

12

500 mg thrice daily for 28 days

Garlic oil

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on midazolam pharmacokinetics

None, CYP3A4

[27]

Ritonavir

Ritonavir

10

2 × 5 mg twice daily for 4 days

Garlic powder in liquid-filled soft gelatin capsules

Open, two-treatment, two-period, two-sequence, randomized, crossover study; ritonavir administration at end of supplement phase

No effect on ritonavir pharmacokinetics

None, CYP3A4

[21]

Saquinavir

Saquinavir

10

4000 mg twice daily for 21 days

Garlic caplets with 3.6 mg/caplet allicin and 4.8 mg/caplet allin

Two-treatment, three-period, single-sequence, longitudinal study; saquinavir administration at end of garlic supplementation in period two

Decreased mean saquinavir AUC 51 % (p = 0.007), mean C8 49 % (p = 0.002) and mean Cmax 54 % (p = 0.006)

Not known, CYP3A4

[26]

Rac-warfarin

Rac-warfarin

12

2 × 500 mg thrice daily for 14 days

Enteric-coated garlic tablets containing 2000 mg of fresh garlic bulb equivalent to 3.71 mg of allicin per tablet

Open-label, three-treatment, randomized crossover study; warfarin administration alone vs. after 2 weeks of pretreatment with garlic

No effect on either S-warfarin or R-warfarin pharmacokinetics

None, CYP2C9

[67]

Rac-warfarin

48

5 mL twice daily for 12 weeks

Aged garlic extract with 305 g/L of extracted solids

Double-blind, randomized, placebo-controlled pilot study

No effect on warfarin pharmacokinetics and pharmacodynamics

None, CYP2C9

[24]

Echinacea

Caffeine

Caffeine

12

400 mg 4 times daily for 8 days

Echinacea purpurea root

Two-period, open-label, fixed-schedule study; phenotyping before vs. after a short course of echinacea

Reduced oral clearance of caffeine by 27 % (p = 0.49)

Inhibition of CYP1A2

[31]

Caffeine

12

800 mg twice daily for 28 days

Echinacea purpurea (no standardization)

Open-label study randomized for supplementation sequence

No effect on caffeine pharmacokinetics

None, CYP1A2

[35]

Chlorzoxazone

Chlorzoxazone

12

800 mg twice daily for 28 days

Echinacea purpurea (no standardization)

Open-label study randomized for supplementation sequence

No effect on chlorzoxazone pharmacokinetics

None, CYP2E1

[35]

Debrisoquine

Debrisoquine

12

800 mg twice daily for 28 days

Echinacea purpurea (no standardization)

Open-label study randomized for supplementation sequence

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[35]

Debrisoquine

12

800 mg twice daily for 28 days

Echinacea purpurea extract, standardized to 2.2 mg isobutylamides per capsule

Open-label study randomized for supplementation sequence

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[36]

Dextromethorphan

Dextromethorphan

12

400 mg 4 times daily for 8 days

Echinacea purpurea root

Two-period, open-label, fixed-schedule study; phenotyping before vs. after a short course of echinacea

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[31]

Lopinavir

Midazolam

13

500 mg thrice daily for 28 days

Echinacea purpurea fresh liquid extract 8 : 1 (250 mg) softgel capsules

Open-label study on the steady-state pharmacokinetics of lopinavir and ritonavir

No effect on lopinavir pharmacokinetics

None, CYP3A4

[32]

Midazolam

Midazolam

12

400 mg 4 times daily for 8 days

Echinacea purpurea root

Two-period, open-label, fixed-schedule study; phenotyping before vs. after a short course of echinacea

No effect on midazolam pharmacokinetics

None, CYP3A4

[31]

Midazolam

13

500 mg thrice daily for 28 days

Echinacea purpurea fresh liquid extract 8 : 1 (250 mg) softgel capsules

Open-label study on the steady-state pharmacokinetics of lopinavir and ritonavir

Modest decrease in midazolam AUC(0-∞) by 27 % (p = 0.008)

Not known, CYP3A4

[32]

Midazolam

12

800 mg twice daily for 28 days

Echinacea purpurea (no standardization)

Open-label study randomized for supplementation sequence;

No effect on midazolam pharmacokinetics

None, CYP3A4

[35]

Ritonavir

Midazolam

13

500 mg thrice daily for 28 days

Echinacea purpurea fresh liquid extract 8 : 1 (250 mg) softgel capsules

Open-label study on the steady-state pharmacokinetics of lopinavir and ritonavir

No effect on ritonavir pharmacokinetics

None, CYP3A4

[32]

Tolbutamide

Tolbutamide

12

400 mg 4 times daily for 8 days

Echinacea purpurea root

Two-period, open-label, fixed-schedule study; phenotyping before vs. after a short course of echinacea

Reduced oral clearance of tolbutamide by 11 % (p = 0.001)

Inhibition of CYP2C9

[31]

Warfarin

Warfarin

12

675 mg 4 times daily for 7 days

A mixture of 600 mg of Echinacea angustifolia root and 675 mg of Echinacea purpurea root standardized to 5.75 mg of total alkamides per tablet

Open-label, randomized, three-treatment, crossover study; administration of single dose of warfarin vs. after two weeks of pretreatment with echinacea

No effect on warfarin pharmacokinetics

None, CYP2C9

[34]

Saw palmetto

Alprazolam

Alprazolam

12

320 mg once daily for 14 days

ProstActive capsules containing 197.7 mg of nonesterified fatty acids, representing 62 % of the total extact

Phenotyping before vs. after pretreatment of saw palmetto

No effect on alprazolam pharmacokinetics

None, CYP3A4

[38]

Caffeine

Caffeine

12

160 mg twice daily for 28 days

Standardized to 85 % to 95 % fatty acids and sterols

Open-label study randomized for supplementation sequence

No effect on caffeine pharmacokinetics

None, CYP1A2

[35]

Chlorzoxazone

Chlorzoxazone

12

160 mg twice daily for 28 days

Standardized to 85 % to 95 % fatty acids and sterols

Open-label study randomized for supplementation sequence

No effect on chlorzoxazone pharmacokinetics

None, CYP2E1

[35]

Debrisoquine

Debrisoquine

12

160 mg twice daily for 28 days

Standardized to 85 % to 95 % fatty acids and sterols

Open-label study randomized for supplementation sequence

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[35]

Dextromethorphan

Dextromethorphan

12

320 mg once daily for 14 days

ProstActive capsules containing 197.7 mg of nonesterified fatty acids, representing 62 % of the total extact

Phenotyping before vs. after pretreatment of saw palmetto

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[38]

Midazolam

Midazolam

12

160 mg twice daily for 28 days

Standardized to 85–95 % fatty acids and sterols

Open-label study randomized for supplementation sequence

No effect on midazolam pharmacokinetics

None, CYP3A4

[35]

Ginkgo

Alprazolam

Alprazolam

12

120 mg twice daily for 14 days

EGb 761 tablets standardized to 24 % ginkgo flavonol glycosides and 6 % terpene lactones as well as bilobalide

Phenotyping at baseline vs. treatment with ginkgo

No effect on alprazolam pharmacokinetics

None, CYP3A4

[54]

Bupropion

Bupropion

14

2 × 60 mg twice daily for 14 days

Ginkgo biloba capsules standardized with a minimum of 24 % ginkgo flavone glycosides and 6 % terpene lactones

Randomized, 2-phase crossover study; voriconzole administration before vs. after pretreatment with ginkgo

No effect on bupropion pharmacokinetics

None, CYP2B6

[53]

Caffeine

Caffeine

12

60 mg 4 times daily for 28 days

Ginkgo biloba standardized to 24 % flavone glycosides and 6 % terpene lactones

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on caffeine pharmacokinetics

None, CYP1A2

[27]

Caffeine

18

120 mg twice daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo vs. phenotyping at the end of treatment

No effect on caffeine pharmacokinetics

None, CYP1A2

[15]

Caffeine

18

240 mg once daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo and placebo vs. phenotyping at the end of treatment

No effect on caffeine pharmacokinetics

None, CYP1A2

[15]

Chlorzoxazone

Chlorzoxazone

12

60 mg 4 times daily for 28 days

Ginkgo biloba standardized to 24 % flavone glycosides and 6 % terpene lactones

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on chlorzoxazone pharmacokinetics

None, CYP2E1

[27]

Cortisol

Omeprazole

18

2 × 70 mg twice daily for 12 days

Each tablet contains 70 mg of standardized Ginkgo biloba leaf extract (16.04 mg of flavonol glycosides and 4.77 mg of terpene lactones)

Open-label, sequential design; phenotyping at baseline vs. end of 12-day treatment period

No effect on cortisol pharmacokinetics

None, CYP3A

[50]

Debrisoquine

Debrisoquine

12

60 mg 4 times daily for 28 days

Ginkgo biloba standardized to 24 % flavone glycosides and 6 % terpene lactones

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[27]

Dextromethorphan

Dextromethorphan

12

120 mg twice daily for 14 days

EGb 761 tablets

Phenotyping at baseline vs. treatment with ginkgo

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[54]

Dextromethorphan

18

120 mg twice daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo vs. phenotyping at end of treatment

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[15]

Dextromethorphan

18

240 mg once daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo and placebo vs. phenotyping at end of treatment

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[15]

Diazepam

Diazepam

12

120 mg twice daily for 28 days

Each tablet contains 40 mg of standardized Ginkgo biloba extract (9.6 mg flavonol glycosides and 2.4 mg terpene lactones)

Open-label, sequential design; phenotyping at baseline vs. at end of treatment with ginkgo

No effect on diazepam pharmacokinetics

None, CYP3A4

[57]

Flurbiprofen

Flurbiprofen

11

2 × 60 mg thrice a day for 1 day

EGb 761 tablets

Randomized, two-way crossover study; phenotyping at baseline vs. at end of treatment with ginkgo or placebo

No effect on flurbiprofen pharmacokinetics

None, CYP2C9

[48]

Lopinavir

Midazolam

14

120 mg twice daily for 14 days

Capsules containing 29.2 % flavonol glycosides and 5.1 % terpene lactones of total content

Open-label investigation on the steady-state pharmcokinetics of lopinavir and ritonavir; phenotyping at baseline vs. at end of treatment of ginkgo administration

No effect on lopinavir pharmacokinetics

None, CYP3A4

[52]

Midazolam

Midazolam

12

60 mg 4 times daily for 28 days

Ginkgo biloba standardized to 24 % flavone glycosides and 6 % terpene lactones

Open-label, randomized study; phenotyping at baseline vs. at end of supplement phase

No effect on midazolam pharmacokinetics

None, CYP3A4

[27]

Midazolam

18

120 mg twice daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo vs. phenotyping at end of treatment

No effect on midazolam pharmacokinetics

None, CYP3A4

[15]

Midazolam

18

240 mg once daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo and placebo vs. phenotyping at end of treatment

No effect on midazolam pharmacokinetics

None, CYP3A4

[15]

Midazolam

15

120 mg twice daily for 28 days

Capsules containing 29.2 % flavonol glycosides and 5.1 % terpene lactones of total content

Single-sequence, longitudinal investigation; phenotyping at baseline vs. at end of treatment with Ginkgo biloba extract

Mild-moderate reduced AUC(0-∞) for midazolam

Not known, CYP3A4

[55]

Midazolam

Midazolam

14

120 mg twice daily for 14 days

Capsules containing 29.2 % flavonol glycosides and 5.1 % terpene lactones of total content

Open-label investigation on the steady-state pharmcokinetics of lopinavir and ritonavir; phenotyping at baseline vs. end of treatment of ginkgo administration

Decreased midazolam AUC(0-∞) by 34 % (p = 0.03) and Cmax by 31 % (p = 0.03)

Induction of CYP3A

[52]

Midazolam

10

2 × 60 mg thrice daily for 28 days

EGb 761 tablets

Phenotyping before vs. after administration of Ginkgo biloba extract

Increased AUC(0-∞) by 25 % and decreased oral clearance by 26 % for midazolam

Inhibition of CYP3A4

[56]

Omeprazole

Omeprazole

18

120 mg twice daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo vs. phenotyping at end of treatment

No effect on omeprazole pharmacokinetics

None, CYP2C19

[15]

Omeprazole

18

240 mg once daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo and placebo vs. phenotyping at end of treatment

No effect on omeprazole pharmacokinetics

None, CYP2C19

[15]

Omeprazole

18

2 × 70 mg twice daily for 12 days

Each tablet contains 70 mg of standardized Ginkgo biloba leaf extract (16.04 mg of flavonol glycosides and 4.77 mg of terpene lactones)

Open-label, sequential design; phenotyping at baseline vs. end of 12-day treatment period

Decrease in the AUC ratio of omeprazole to 5-hydroxyomeprazole by 54.4 % (p < 0.01)

Induction of CYP2C19

[50]

Ritonavir

Midazolam

14

120 mg twice daily for 14 days

Capsules containing 29.2 % flavonol glycosides and 5.1 % terpene lactones of total content

Open-label investigation on the steady-state pharmcokinetics of lopinavir and ritonavir; phenotyping at baseline vs. end of treatment of ginkgo administration

No effect on ritonavir pharmacokinetics

None, CYP3A4

[52]

Tolbutamide

Tolbutamide

18

120 mg twice daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo vs. phenotyping at end of treatment

No effect on tolbutamide pharmacokinetics

None, CYP2C9

[15]

Tolbutamide

18

240 mg once daily for 8 days

EGb 761 tablets

Open-label, randomized, threefold crossover, cocktail study; pretreatment with ginkgo and placebo vs. phenotyping at end of treatment

No effect on tolbutamide pharmacokinetics

None, CYP2C9

[15]

Tolbutamide

10

2 × 60 mg thrice daily for 28 days

EGb 761 tablets

Phenotyping before vs. after administration of Ginkgo biloba extract

Slightly decreased AUC(0-∞) by 16 % for tolbutamide

Induction of CYP2C9

[56]

Voriconazole

Voriconazole

14

120 mg twice daily for 12 days

Ginkgo biloba capsules standardized with a minimum of 24 % flavone glycosides and 6 % terpene lactones

Randomized, two-phase crossover design; phenotyping at baseline (without pretreatment) vs. after pretreatment with Ginkgo biloba

No effect on voriconazole pharmacokinetics

None, CYP3A4

[51]

Rac-warfarin

Rac-warfarin

12

2 × 40 mg thrice daily for 7 days

EGb 761 tablets

Randomized, open-label, three-treatment, three-period, three-
sequence, crossover study; phenotyping alone vs. after 7 days pretreatment with ginkgo

No effect on either S- or R-warfarin pharmacokinetics

None, CYP2C9

[49]

Cranberry

Cyclosporine

Cyclosporine

12

240 mL once a day for 1 day

Concentrated cranberry juice

Open-label, randomized, three-
way crossover study with three sequences; phenotyping with water intake (baseline) vs. with juice intake (treatment)

No effect on the overall disposition of cyclosporine

None, CYP3A

[69]

Diclofenac

Diclofenac

8

180 mL once a day for 1 day

Concentrated cranberry juice containing 27 % cranberry

Open-label, two-period, crossover design; phenotyping with water intake (baseline) vs. pretreatment with cranberry juice

No change in pharmacokinetics of diclofenac

None, CYP2C9

[65]

Flurbiprofen

Flurbiprofen

14

8 oz. a day for 1 day

Concentrated cranberry juice

Crossover design; phenotyping with water (baseline) vs. cranberry juice placebo vs. cranberry juice

No effect on flurbiprofen pharmacokinetics

None, CYP2C9

[66]

Midazolam

Midazolam

10

200 mL thrice daily for 10 days

Concentrated cranberry juice was diluted with tap water (1 : 4 vol/vol)

Randomized, two-phase crossover study; phenotyping with water (baseline) vs. pretreatment with cranberry juice

No effect on midazolam pharmacokinetics

None, CYP3A4

[68]

Tizanidine

Tizanidine

10

200 mL thrice daily for 10 days

Concentrated cranberry juice was diluted with tap water (1 : 4 vol/vol)

Randomized, two-phase crossover study; phenotyping with water (baseline) vs. pretreatment with cranberry juice

No effect on tizanidine pharmacokinetics

None, CYP1A2

[68]

Rac-warfarin

Rac-warfarin

10

200 mL thrice daily for 10 days

Concentrated cranberry juice was diluted with tap water (1 : 4 vol/vol)

Randomized, two-phase crossover study; phenotyping with water (baseline) vs. pretreatment with cranberry juice

No effect on either S-warfarin or R-warfarin pharmacokinetics and pharmacodynamics

None, CYP2C9

[68]

Rac-warfarin

12

2 × 500 mg thrice daily for 14 days

Two capsules with cranberry juice concentrate are equivalent to 57 g of fruit per day

Open-label, three-treatment, randomized crossover study; warfarin administration alone or after two weeks of pretreatment with cranberry

No effect on either S-warfarin or R-warfarin pharmacokinetics but pharmacodynamics (increased mean AUC(INR) by approx. 30 %)

None, CYP2C9

[67]

Soy

Caffeine

Caffeine

18

1000 mg once daily for 14 days

Genistein tablets

Phenotyping once before vs. once at end of treatment with genistein

Decreased urinary paraxanthine ratio by 41 % and increased urinary 1,7-dimethylurate ratio by 47 %

Inhibition of CYP1A2 and induction of CYP2A6

[74]

Cortisol

Cortisol

20

50 mg twice daily for 14 days

Capsules with soy extract containing 50 mg isoflavones (10.9 mg daidzein and 16.5 mg genistein)

Phenotyping at baseline vs. at end of pretreatment

No alteration of urinary 6β-hydroxycortisol/cortisol ratio

None, CYP3A4

[76]

Losartan

Losartan

18

2 × 1000 mg twice daily for 14 days

Genistein Soy Complex tablets

Open-label, two-phase study; phenotyping at baseline vs. at end of pretreatment

No effect on pharmacokinetics of losartan or its metabolite E-3174

None, CYP2C9

[75]

Theophylline

Theophylline

10

200 mg twice daily for 10 days

200 mg daidzein

Single-blind, placebo-controlled, parallel study; phenotyping at baseline vs. after daidzein or placebo coadministration

Increased theophilline AUC(0–48) by ~ 34 % (p < 0.05), AUC(0-∞) by ~ 34 % (p < 0.05), Cmax by ~ 24 % (p < 0.05) and t 1/2 by ~ 41 % (p = 0.011)

Inhibition of CYP1A2

[73]

Ginseng

Caffeine

Caffeine

12

500 mg thrice daily for 28 days

Panax ginseng standardized to 5 % ginsenosides

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on caffeine pharmacokinetics

None, CYP1A2

[27]

Chlorzoxazone

Chlorzoxazone

12

500 mg thrice daily for 28 days

Panax ginseng standardized to 5 % ginsenosides

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on chlorzoxazone pharmacokinetics

None, CYP2E1

[27]

Cortisol

Cortisol

20

100 mg twice daily for 14 days

Panax ginseng 100 mg standardized to 4 % ginsenosides

Phenotyping at baseline vs. at end of pretreatment

No alteration of urinary 6β-hydroxycortisol/cortisol ratio

None, CYP3A

[76]

Debrisoquine

Debrisoquine

12

500 mg thrice daily for 28 days

Panax ginseng standardized to 5 % ginsenosides

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

Decreased debrisoquine urinary recovery ratios of 7 % (p = 0.003)

Inhibition of CYP2D6

[27]

Midazolam

Midazolam

12

500 mg thrice daily for 28 days

Panax ginseng standardized to 5 % ginsenosides

Open-label, randomized, phenotyping at baseline vs. end of each supplement phase

No effect on midazolam pharmacokinetics

None, CYP3A4

[27]

Warfarin

Warfarin

12

500 mg thrice daily for 14 days

Aqueous extract of 9.2 % Panax ginseng equivalent to 100 mg crude root

Randomized, open-label, controlled study; warfarin intake with Panax ginseng vs. warfarin intake only

No effect on warfarin pharmacokinetics

None, CYP2C9

[80]

Rac-warfarin

Rac-warfarin

12

2 × 500 mg thrice daily for 7 days

Capsules containing extract equivalent to 0.5 g Panax ginseng root and 8.93 mg ginsenosides

Open-label, three-treatment, three-period, three-sequence, randomized, crossover study; warfarin intake alone vs. after 7 daysʼ pretreatment with ginseng

No effect on warfarin pharmacokinetics

None, CYP2C9

[83]

Black cohosh

Caffeine

Caffeine

12

1090 mg twice daily for 28 days

Black cohosh root extract capsules standardized to 0.2 % triterpene glycosides

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on caffeine pharmacokinetics

None, CYP1A2

[85]

Chlorzoxazone

Chlorzoxazone

12

1090 mg twice daily for 28 days

Black cohosh root extract capsules standardized to 0.2 % triterpene glycosides

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on chlorzoxazone pharmacokinetics

None, CYP2E1

[85]

Debrisoquine

Debrisoquine

12

1090 mg twice daily for 28 days

Black cohosh root extract capsules standardized to 0.2 % triterpene glycosides

Open-label study; phenotyping at baseline vs. at the end of supplementation

Decreased debrisoquine urinary recovery ratios of 7 %

Inhibition of CYP2D6

[85]

Debrisoquine

18

40 mg twice daily for 14 days

Black cohosh extract standardized to 2.5 % triterpene glycosides per tablet

Open-label study randomized for supplementation sequence

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[36]

Midazolam

Midazolam

12

1090 mg twice daily for 28 days

Black cohosh root extract capsules standardized to 0.2 % triterpene glycosides

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on midazolam pharmacokinetics

None, CYP3A4

[85]

Midazolam

19

40 mg twice daily for 14 days

Black cohosh extract standardized to 2.5 % triterpene glycosides

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on midazolam pharmacokinetics

None, CYP3A4

[86]

St. Johnʼs wort

Alprazolam

Alprazolam

28

240 mg twice daily with low hyperforin for 10 days

Capsules containing 60 mg SJW extract, with 0.25 mg of total hypericins and 0.88 mg hyperforin

Double-blind, placebo-controlled, parallel-grouped study; phenotyping at baseline vs. at end of treatment with SJW or placebo

No effect on alprazolam pharmacokinetics

None, CYP3A4

[96]

Alprazolam

Alprazolam

7

300 mg thrice daily for 4 days

Capsules standardized to 0.3 % hypericin

Two-phase study; phenotyping at baseline vs. at end of treatment

No effect on alprazolam pharmacokinetics

None, CYP3A4

[88]

Alprazolam

12

300 mg thrice daily for 14 days

Tablets containing 300 mg of SJW extract standardized to 0.12 % to 0.3 % hypericin

Open-label crossover study with fixed treatment order; phenotyping at baseline vs. at end of treatment

A twofold decrease in AUC for alprazolam plasma concentration vs. time (p < 0.001) and a twofold increase in alprazolam clearance (p < 0.001)

Induction of CYP3A4

[92]

Alprazolam

16

300 mg twice daily for 28 days

Tablets containing 300 mg of active substance

Randomized, open, crossover study; SJW (active treatment) vs. vitamin product (control)

Increased serum level of LDL cholesterol by appr. 30 % (p = 0.02) and increase in total cholesterol by approx. 30 % (p = 0.02)

Induction of CYP3A4

[104]

Caffeine

Caffeine

28

240 mg twice daily with low hyperforin for 10 days

Capsules containing 60 mg SJW extract, corresponding to 0.25 mg of total hypericins and 0.88 mg hyperforin

Double-blind, placebo-controlled, parallel-grouped study; phenotyping at baseline vs. at end of treatment with SJW or placebo

No effect on caffeine pharmacokinetics

None, CYP1A2

[96]

Caffeine

12

300 mg thrice daily for 14 days

Tablets containing 0.3 % hypericin and 4 % hyperforin

Two-phase, randomized, placebo-controlled crossover study; pretreatment with SJW or placebo vs. phenotyping at end of intake

No effect on caffeine pharmacokinetics

None, CYP1A2

[93]

Caffeine

12

3 × 300 mg once daily for 2 days

Capsules containing 900 µg hypericin

Three-period, open-label, fixed schedule study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP1A2

[89]

Caffeine

12

300 mg thrice daily for 14 days

Capsules containing 900 µg hypericin

Three-period, open-label, fixed schedule study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP1A2

[89]

Caffeine

16

300 mg thrice daily for 14 days

SJW extract containing 900 µg hypericin

Open study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP1A2

[90]

Caffeine

12

300 mg thrice daily for 28 days

Hypericum perforatum standardized to 0.3 % hypericin

Open-label, randomized study; phenotyping at baseline vs. at end of supplement phase

No effect on caffeine pharmacokinetics

None, CYP1A2

[27]

Chlorzoxazone

Chlorzoxazone

12

300 mg thrice daily for 28 days

Hypericum perforatum standardized to 0.3 % hypericin

Open-label, randomized study; phenotyping at baseline vs. at end of supplement phase

A 26 % (p = 0.006) rise in the 6-hydroxychlorzoxazone/chlorzoxazone serum ratio

Induction of CYP2E1

[27]

Cortisol

Cortisol

16

300 mg thrice daily for 14 days

SJW extract containing 900 µg hypericin

Open study; phenotyping at baseline vs. at end of treatment

Increased 6β-hydroxycortisol/cortisol molar concentration ratio in urine of 85 %

Induction of CYP3A4

[90]

Cortisol

13

300 mg thrice daily for 14 days

Tablets standardized to 0.3 % hypericin

Unblinded, multiple-dose, single-treatment study; phenotyping at baseline vs. at end of treatment

Increased urinary 6β-hydroxycortisol/cortisol ratio of 114 % (p = 0.003)

Induction of CYP3A4

[91]

Cyclosporine

Cyclosporine

10

300 mg thrice daily with low hyperforin for 14 days

Capsule containing 0.1 mg total hyperforin, 0.45 mg total hypericin and 15.6 mg total flavonoids

Randomized crossover study; phenotyping at baseline vs. at end of treatment

No effect on cyclosporine pharmacokinetics

None, CYP3A4

[95]

Cyclosporine

10

300 mg thrice daily with high hyperforin for 14 days

Capsule containing 7.0 mg total hyperforin, 0.45 mg total hypericin and 16.16 mg total flavonoids

Randomized crossover study; phenotyping at baseline vs. at end of treatment

Decreased AUC(0–12) for cyclosporine by 52 % (p < 0.05)

Induction of CYP3A4

[95]

Debrisoquine

Debrisoquine

18

300 mg thrice daily for 14 days

St. Johnʼs wort extract standardized to 3 % hyperforin

Open-label study randomized for supplementation sequence

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[36]

Debrisoquine

12

300 mg thrice daily for 28 days

Hypericum perforatum standardized to 0.3 % hypericin

Open-label, randomized study; phenotyping at baseline vs. end of supplement phase

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[27]

Desogestrel

Desogestrel

18

300 mg thrice daily for 28 days

SJW extract standardized to 0.3 % hypericin

Oral contraceptive intake before (control) vs. cotreatment with SJW extract

Decreased 3-ketodesogestrel AUC(0–24) by ~ 44 % (p = 0.001) and Cmax by ~ 18 % (p = 0.005) during cycle A and by ~ 42 % (p = 0.001) and by ~ 23 % (p < 0.001) during cycle B, respectively

Inhibition of CYP2C9/CYP2C19 and/or induction of CYP3A4

[101]

Dextromethorphan

Dextromethorphan

7

300 mg thrice daily for 4 days

Capsules standardized to 0.3 % hypericin

Two-phase study; phenotyping at baseline vs. at end of treatment

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[88]

Dextromethorphan

12

300 mg thrice daily for 14 days

Tablets containing 300 mg of an SJW extract standardized to 0.12 % to 0.3 % hypericin

Open-label crossover study with fixed treatment order; phenotyping at baseline vs. at end of treatment

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[92]

Dextromethorphan

12

3 × 300 mg once daily for 2 days

Capsules containing 900 µg hypericin

Three-period, open-label, fixed schedule study; phenotyping at baseline vs. at end of treatment

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[89]

Dextromethorphan

Dextromethorphan

12

300 mg thrice daily for 14 days

Capsules containing 900 µg hypericin

Three-period, open-label, fixed schedule study; phenotyping at baseline vs. at end of treatment

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[89]

Dextromethorphan

16

300 mg thrice daily for 14 days

SJW extract containing 900 µg hypericin

Open study; phenotyping at baseline vs. at end of treatment

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[90]

Ethinyl estradiol

Midazolam

12

300 mg thrice daily for 28 days

SJW extract

Oral contraceptive intake before (control) vs. cotreatment with SJW extract

Increased mean CLoral of ethinyl estradiol by 47 %, increased midazolam CLoral by 50 %

Induction of CYP3A4

[99]

Ethinyl estradiol

16

300 mg thrice daily for 28 days

SJW extract

Placebo-controlled, single-blind sequential study; oral contraceptive intake before (control) vs. cotreatment with SJW extract

Reduced AUC of ethinyl estradiol by 14 % (p = 0.016)

Induction of CYP3A4

[100]

Ethinyl estradiol

18

300 mg thrice daily for 28 days

SJW extract standardized to 0.3 % hypericin

Oral contraceptive intake before (control) vs. cotreatment with SJW extract

No effect on ethinyl estradiol pharmacokinetics

None, CYP3A4

[101]

Gliclazide

Gliclazide

21

300 mg thrice daily for 15 days

SJW extract

Sequential crossover, two-treatment study; gliclazide alone vs. at end of treatment with SJW

Reduced gliclazide AUC(0-∞) by 33 % and decreased Cmax by 22 %

Not known, CYP2C9

[109]

Imatinib

Imatinib

10

300 mg thrice daily for 14 days

SJW

Open-label, complete crossover, fixed-sequence, pharmacokinetic study; imatinib administration before vs. at end of treatment with SJW

Reduction of AUC(0-∞) of imatinib by 32 % (p = 0.0001) and reduced Cmax by 29 % (p = 0.005)

Induction of CYP3A4

[102]

Imatinib

12

300 mg thrice daily for 14 days

SJW extract

Open-label, two-period, fixed-
sequence study; imatinib administration before vs. at end of treatment with SJW

Increased imatinib clearance by 43 % (p < 0.001), decreased AUC(0-∞) by 30 % (p < 0.001)

Induction of CYP3A4

[103]

Indinavir

Indinavir

8

300 mg thrice daily for 14 days

SJW preparation standardized to 0.3 % hypericin

Open-label study; indinavir administration at baseline vs. at end of treatment with SJW

Reduced AUC(0–8) of indinavir by 57 % (p = 0.0008)

Induction of CYP3A4

[110]

Ivabradine

Ivabradine

12

300 mg thrice daily for 14 days

SJW tablets

Open-label, two-period, nonrandomized, phase I, pharmacokinetic study; ivabradine administration at baseline vs. at end of treatment with SJW

Decreased Cmax by 51 % (p < 0.01) and AUC by 61 % (p < 0.01) for ivabradine

Induction of CYP3A4

[105]

Mephenytoin

Mephenytoin

12

300 mg thrice daily for 14 days

Tablets containing 0.3 % hypericin and 4 % hyperforin

Two-phase, randomized, placebo-controlled crossover study; pretreatment with SJW or placebo vs. phenotyping at end of intake

Raised urinary 4′-hydroxymephenytoin excretion by 151.5 % (p = 0.0156)

Induction of CYP2C19

[93]

Midazolam

Midazolam

42

6 different concentrations of hyperforin for 14 days

Coated tablets with 300 mg of dried Hypericum herba containing 0.13–41.25 mg hyperforin, 1.08–4.86 mg hypericins and 17.92–80.64 mg flavonoids

Open-label, randomized, interaction study with six parallel SJW medication groups; phenotyping at baseline vs. at end of treatment with SJW

All SJW preparations tested resulted in a decrease in midazolam AUC. The extent of midazolam AUC decrease correlated significantly with increasing the HYF dose (r = 0.765, p < 0.001)

Induction of CYP3A4

[97]

Midazolam

12

3 × 300 mg once daily for 2 days

Capsules containing 900 µg hypericin

Three-period, open-label, fixed schedule study; phenotyping at baseline vs. at end of treatment

No effect on midazolam pharmacokinetics

None, CYP3A4

[89]

Midazolam

12

300 mg thrice daily for 14 days

Capsules containing 900 µg hypericin

Three-period, open-label, fixed schedule study; phenotyping at baseline vs. at end of treatment

> 50 % (p < 0.05) decrease in midazolam AUCoral and Cmax, a 21 % (p < 0.05) decrease in midazolam AUCintravenous

Induction of CYP3A4

[89]

Midazolam

12

300 mg thrice daily for 28 days

Hypericum perforatum standardized to 0.3 % hypericin

Open-label, randomized, phenotyping at baseline vs. end of each supplement phase

A 141 % (p < 0.001) increase in mean 1-hydroxymidazolam/midazolam serum ratio

Induction of CYP3A4

[27]

Midazolam

20

500 mg twice daily with low hyperforin for 14 days

SJW capsules containing 500 mg Hyperici herba powder

Open-label, one-sequence crossover study; phenotyping at baseline vs. at end of treatment with SJW

No effect on midazolam pharmacokinetics

None, CYP3A4

[98]

Midazolam

12

300 mg thrice daily for 28 days

SJW extract

Oral contraceptive intake before (control) vs. cotreatment with SJW extract

Reduced oral AUC(0-∞) by 35 % (p = 0.076), reduced oral Cmax by 16 % (p = 0.214) and increased oral clearance of midazolam by 45 % (p = 0.007)

Induction of CYP3A4

[99]

Norethindrone

Midazolam

12

300 mg thrice daily for 28 days

SJW extract

Oral contraceptive intake before (control) vs. cotreatment with SJW extract

Reduced AUC(0–24) by 12 % (p = 0.15), reduced Cmax by 7 % (p = 0.045) and increased oral clearance of norethindrone by 14 % (p = 0.042)

Induction of CYP3A4

[99]

Norethindrone

16

300 mg thrice daily for 28 days

SJW extract

Placebo-controlled, single-blind sequential study; oral contraceptive intake before (control) vs. cotreatment with SJW extract

Reduced AUC of norethindrone by 14 % (p = 0.021)

Induction of CYP3A4

[100]

Omeprazole

Omeprazole

12

300 mg thrice daily for 14 days

SJW extract tablets containing 0.3 % total hypericin and 4 % hyperforin

Two-phase, randomized, placebo-controlled crossover study; pretreatment with SJW or placebo vs. omeprazole intake at end of treatment

Increased Cmax and AUC(0-∞) of 5-hydroxyomeprazole by 38.1 % (p = 0.028) and by 37.2 % (p = 0.005); increased Cmax and AUC(0-∞) of omeprazole sulfone by 155.5 % (p = 0.001) and by 158.7 % (p = 0.017)

Induction of CYP2C19 and CYP3A4

[94]

Quazepam

Quazepam

13

300 mg thrice daily for 14 days

SJW caplets standardized to 0.3 % hypericin

Randomized, double-blind, crossover study; pretreatment with SJW or placebo vs. quazepam intake at end of treatment

Reduced AUC(0–48) by 55 % (p < 0.05) and Cmax by 8.7 % (p < 0.05)

Induction of CYP3A4

[107]

Tolbutamide

Tolbutamide

28

240 mg twice daily with low hyperforin for 10 days

Capsules containing 60 mg SJW extract, corresponding to 0.25 mg of total hypericins and 0.88 mg hyperforin

Double-blind, placebo-controlled, parallel-grouped study; phenotyping at baseline vs. at end of treatment with SJW or placebo

No effect on tolbutamide pharmacokinetics

None, CYP2C9

[96]

Tolbutamide

12

3 × 300 mg once daily for 2 days

Capsules containing 900 µg hypericin

Three-period, open-label, fixed schedule study; phenotyping at baseline vs. at end of treatment

No effect on tolbutamide pharmacokinetics

None, CYP2C9

[89]

Tolbutamide

12

300 mg thrice daily for 14 days

Capsules containing 900 µg hypericin

Three-period, open-label, fixed schedule study; phenotyping at baseline vs. at end of treatment

No effect on tolbutamide pharmacokinetics

None, CYP2C9

[89]

Rac-verapamil

Rac-verapamil

8

300 mg thrice daily for 14 days

SJW tablets containing 3 % to 6 % hyperforin

Phenotyping at baseline vs. at end of treatment

Decreased AUC by 78 % (p < 0.0001) for R-verapamil and by 80 % (p < 0.0001) for S-verapamil and decreased Cmax by 76 % (p < 0.0001) for R-verapamil and by 78 % (p < 0.0001) for S-verapamil

Induction of CYP3A4

[106]

Voriconazole

Voriconazole

16

300 mg thrice daily for 15 days

SJW extract

Open-label, controlled, fixed-dose schedule study; phenotyping at baseline vs. at end of treatment

Reduced AUC(0-∞) by 59 % (p = 0.0004) of voriconazole after 15 days

Induction of CYP2C19

[108]

Rac-warfarin

Rac-warfarin

12

1000 mg thrice daily for 14 days

Tablets containing standardized dry extract equivalent to 1 g Hypericum perforatum, 0.825 mg hypericin and 12.5 mg hyperforin

Open-label, three-treatment, three-period, three-sequence, randomized, crossover study; warfarin intake alone vs. after 14 daysʼ pretreatment with St. Johnʼs wort

Reduced AUC(0-∞) by 27 % (p < 0.05) for S-warfarin and by 23 % (p < 0.05) for R-warfarin, induced apparent clearance of both S- and R-warfarin by 29 % (p < 0.05) and by 23 % (p < 0.05), respectively

Induction of CYP1A2 and/or CYP3A4 and CYP2C9

[83]

Milk thistle

Caffeine

Caffeine

12

175 mg twice daily for 28 days

Standardized to 80 % silymarins

Open-label study randomized for supplementation sequence

No effect on caffeine pharmacokinetics

None, CYP1A2

[35]

Chlorzoxazone

Chlorzoxazone

12

175 mg twice daily for 28 days

Standardized to 80 % silymarins

Open-label study randomized for supplementation sequence

No effect on chlorzoxazone pharmacokinetics

None, CYP2E1

[35]

Debrisoquine

Debrisoquine

12

175 mg twice daily for 28 days

Standardized to 80 % silymarins

Open-label study randomized for supplementation sequence

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[35]

Debrisoquine

18

300 mg thrice daily for 14 days

Milk thistle extract standardized to 80 % silymarin per capsule

Open-label study randomized for supplementation sequence

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[36]

Irinotecan

Irinotecan

6

200 mg thrice daily for 14 days

Standardized capsules containing 200 mg milk thistle seed extract (containing 80 % silymarin)

Irinotecan administration before vs. after treatment with milk thistle capsules

No effect on irinotecan clearance

None, CYP3A4

[114]

Midazolam

Midazolam

12

175 mg twice daily for 28 days

Standardized to 80 % silymarins

Open-label study randomized for supplementation sequence

No effect on midazolam pharmacokinetics

None, CYP3A4

[35]

Midazolam

19

300 mg thrice daily for 14 days

Milk thistle standardized to 80 % silymarin

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on midazolam pharmacokinetics

None, CYP3A4

[86]

Nifedipine

Nifedipine

16

2 × 140 mg twice a day for 1 day

Silymarin capsules containing 173.0–186.7 mg dry extract from milk thistle fruits [36–44 : 1], equivalent to 140 mg silymarin, calculated as silibinin

Open, within-subject crossover design with period-balanced randomly allocated sequences: nifedipine administration at baseline vs. at end of treatment

No effect on nifedipine pharmacokinetics

None, CYP3A4

[14]

Green tea

Alprazolam

Alprazolam

11

211 mg green tea catechins for 14 days

Decaffeinated green tea capsules containing 2 mg catechin, 11 mg epicatechin, 18 mg epigallocatechin, 126 mg epigallocatechin gallate, 0.9 mg caffeine

Phenotyping at baseline vs. treatment with ginkgo

No effect on alprazolam pharmacokinetics

None, CYP3A4

[126]

Buspirone

Buspirone

42

800 mg EGCG daily for 28 days

Polyphenon E (decaffeinated green tea extract) capsules with 200 mg EGCG/capsule

Cocktail study; phenotyping at baseline vs. at end of treatment

No effect on buspirone pharmacokinetics

None, CYP3A4

[127]

Caffeine

Caffeine

42

800 mg EGCG daily for 28 days

Polyphenon E

Cocktail study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP1A2

[127]

Dextromethorphan

Dextromethorphan

11

211 mg green tea catechins for 14 days

Decaffeinated green tea capsules containing 2 mg catechin, 11 mg epicatechin, 18 mg epigallocatechin, 126 mg epigallocatechin gallate, 0.9 mg caffeine

Phenotyping at baseline vs. treatment with ginkgo

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[126]

Dextromethorphan

42

800 mg EGCG daily for 28 days

Polyphenon E

Cocktail study; phenotyping at baseline vs. at end of treatment

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[127]

Losartan

Losartan

42

800 mg EGCG daily for 28 days

Polyphenon E

Cocktail study; phenotyping at baseline vs. at end of treatment

No effect on losartan pharmacokinetics

None, CYP2C9

[127]

Valerian

Alprazolam

Alprazolam

12

2 × 500 mg nightly for 14 days

Tablets containing 500 mg dry valerian root extract with 5.51 mg valerenic acid

Open-label, fixed treatment order, crossover study; phenotyping at baseline vs. pretreatment with valerian extract

No effect on alprazolam pharmacokinetics

None, CYP3A4

[135]

Caffeine

Caffeine

12

125 mg thrice daily for 28 days

Valerian root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on caffeine pharmacokinetics

None, CYP1A2

[85]

Chlorzoxazone

Chlorzoxazone

12

125 mg thrice daily for 28 days

Valerian root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on chlorzoxazone pharmacokinetics

None, CYP2E1

[85]

Debrisoquine

Debrisoquine

12

125 mg thrice daily for 28 days

Valerian root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[85]

Dextromethorphan

Dextromethorphan

12

2 × 500 mg nightly for 14 days

Tablets containing 500 mg dry valerian root extract with 5.51 mg valerenic acid

Open-label, fixed treatment order, crossover study; phenotyping at baseline vs. pretreatment with valerian extract

No effect on dextromethorphan pharmacokinetics

None, CYP2D6

[135]

Midazolam

Midazolam

12

125 mg thrice daily for 28 days

Valerian root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on midazolam pharmacokinetics

None, CYP3A4

[85]

Kava

Caffeine

Caffeine

12

1000 mg twice daily for 28 days

Kava kava root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on caffeine pharmacokinetics

None, CYP1A2

[85]

Chlorzoxazone

12

1000 mg twice daily for 28 days

Kava kava root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

Reduced 6-hydroxychlorzoxazone/chlorzoxazone serum ratios by ~ 40 % (p = 0.009)

Inhibition of CYP2E1

[85]

Debrisoquine

Debrisoquine

12

1000 mg twice daily for 28 days

Kava kava root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[85]

Debrisoquine

18

136.3 mg thrice daily for 14 days

Kava kava rhizome extract standardized to 75 mg kavalactones per capsule

Open-label study randomized for supplementation sequence

No effect on debrisoquine pharmacokinetics

None, CYP2D6

[36]

Midazolam

Midazolam

12

1000 mg twice daily for 28 days

Kava kava root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on midazolam pharmacokinetics

None, CYP3A4

[85]

Midazolam

16

1227 mg thrice daily for 14 days

Kava kava rhizome extract standardized to 75 mg kavalactones per capsule

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on midazolam pharmacokinetics

None, CYP3A4

[139]

Goldenseal

Caffeine

Caffeine

12

900 mg thrice daily for 28 days

Goldenseal root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on caffeine pharmacokinetics

None, CYP1A2

[85]

Chlorzoxazone

Chlorzoxazone

12

900 mg thrice daily for 28 days

Goldenseal root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

No effect on chlorzoxazone pharmacokinetics

None, CYP2E1

[85]

Debrisoquine

Debrisoquine

12

900 mg thrice daily for 28 days

Goldenseal root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

Reduced debrisoquine urinary recovery ratios by 40 % (p < 0.05)

Inhibition of CYP2D6

[85]

Debrisoquine

18

1070 mg thrice daily for 14 days

Root extract standardized to 24.1 mg isoquinoline alkaloids per capsule

Open-label study randomized for supplementation sequence

Reductions in debrisoquine urinary recovery ratios by 47 % (p < 0.05)

Inhibition of CYP2D6

[36]

Midazolam

Midazolam

12

900 mg thrice daily for 28 days

Goldenseal root extract (no standardization claim)

Open-label study; phenotyping at baseline vs. at the end of supplementation

Reduced 1-hydroxymidazolam/midazolam serum ratios by 40 % (p < 0.05)

Inhibition of CYP3A4/5

[85]

Midazolam

16

1323 mg thrice daily for 14 days

Goldenseal root extract standardized to 24.1 mg isoquinoline alkaloids

Open-label study; phenotyping at baseline vs. at the end of supplementation

Increase (p < 0.05) in midazolam AUC(0-∞) by 62 %, elimination half-life by 57 %, and Cmax by 41 %

Inhibition of CYP3A

[139]

Ginger

Rac-warfarin

Rac-warfarin

12

3 × 400 mg thrice daily for 7 days

Capsules containing extract equivalent to 0.4 g of ginger rhizome powder

Randomized, open-label, three-treatment, three-period, three-
sequence, crossover study; phenotyping alone vs. after 7 days pretreatment with ginkgo

No effect on S- and R-warfarin pharmacokinetics

None, CYP2C9

[49]

Angelica tenuissima

Caffeine

Caffeine

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP1A2

[143]

Chlorzoxazone

Chlorzoxazone

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP2E1

[143]

Dextromethorphan

Dextromethorphan

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP2D6

[143]

Losartan

Losartan

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP2C9

[143]

Midazolam

Midazolam

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP3A4

[143]

Omeprazole

Omeprazole

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on caffeine pharmacokinetics

None, CYP2C19

[143]

Angelica dahurica

Caffeine

Caffeine

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

Decreased metabolic ratio of caffeine to 10 % of baseline activity (p < 0.001)

Inhibition of CYP1A2

[143]

Chlorzoxazone

Chlorzoxazone

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on chlorzoxazone metabolic ratio

None, CYP2E1

[143]

Dextromethorphan

Dextromethorphan

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

Slightly increase in dextromethorphan metabolic ratio

None, CYP2D6

[143]

Losartan

Losartan

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on losartan metabolic ratio

None, CYP2C9

[143]

Midazolam

Midazolam

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No changes in plasma concentration of midazolam

None, CYP3A4

[143]

Omeprazole

Omeprazole

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract equivalent to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on omeprazole metabolic ratio

None, CYP2C19

[143]

Scutellaria baicalensis

Bupropion

Bupropion

17

500 mg thrice daily for 14 days

Baicalin capsules

Two-phase, two-treatment, sequential study; bupropion intake at baseline vs. at end of treatment

Increased hydroxybupropion AUC(0-∞) by 87 % (p < 0.01) and Cmax by 73 % (p < 0.01)

Induction of CYP2B6

[144]

Caffeine

Caffeine

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract eq. to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on caffeine metabolic ratio

None, CYP1A2

[143]

Chlorzoxazone

Chlorzoxazone

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract eq. to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

A 1.42-fold (p = 0.039) increase in metabolic ratio of chlorzoxazone after multiple administration

Induction of CYP2E1

[143]

Dextromethorphan

Dextromethorphan

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract eq. to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on dextromethorphan metabolic ratio

None, CYP2D6

[143]

Losartan

Losartan

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract eq. to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

Decreased metabolic ratio of losartan to 71 % (p = 0.024) of baseline value

Inhibition of CYP2C9

[143]

Midazolam

Midazolam

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract eq. to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No changes in plasma concentration of midazolam

None, CYP3A4

[143]

Omeprazole

Omeprazole

8

2000 mg thrice daily for 13 days

Encapsuled aqueous extract eq. to 2 g of herb

Open-label, parallel group study; phenotyping at baseline vs. at end of treatment

No effect on omeprazole metabolic ratio

None, CYP2C19

[143]

Grapes/red wine

Buspirone

Buspirone

42

2 × 500 mg once daily for 28 days

Caplets containing 500 mg resveratrol plus inert pharmaceutical excipients

Phenotyping at baseline vs. at end of treatment

A 33 % (p = 0.01) increase in buspirone AUC

Inhibition of CYP3A4

[146]

Caffeine

Caffeine

42

2 × 500 mg once daily for 28 days

Caplets containing 500 mg resveratrol plus inert pharmaceutical excipients

Phenotyping at baseline vs. at end of treatment

A 16 % (p = 0.0005) decrease in caffeine/paraxanthine ratio

Induction of CYP1A2

[146]

Dextromethorphan

Dextromethorphan

42

2 × 500 mg once daily for 28 days

Caplets containing 500 mg resveratrol plus inert pharmaceutical excipients

Phenotyping at baseline vs. at end of treatment

A 70 % (p = 0.01) increase in post-
intervention dextromethorphan/dextrorphan molar ratio

Inhibition of CYP2D6

[146]

Losartan

Losartan

42

2 × 500 mg once daily for 28 days

Caplets containing 500 mg resveratrol plus inert pharmaceutical excipients

Phenotyping at baseline vs. at end of treatment

A 171 % (p < 0.0001) increase in CYP2C9 phenotypic index

Inhibition of CYP2C9

[146]

Curcuma

Caffeine

Caffeine

16

2 × 500 mg once daily for 14 days

Curcumin capsules

A two-phase, crossover design; caffeine administration at baseline vs. at end of treatment

Decreased plasma AUC(0-∞) of paraxanthine by 46.6 % (p = 0.032) and decreased urinary excretion by 36.4 % (p < 0.000), increased urinary excretion of 1,7-dimethylurate by 77.3 % (p = 0.036)

Inhibition of CYP1A2 and induction of CYP2A6

[147]

In addition to the 14 plants described in detail (see discussion), we found positive herb-drug interactions for Angelica dahurica with 1 drug (caffeine), Scutelleria baicalensis with 3 drugs (bupropion, chlorzoxazone, losartan), grapes/red wine with 4 drugs (buspirone, caffeine, dextromethorphan, losartan), and curcuma with 1 drug (caffeine).

The following detailed description of the respective botanicals is sorted in descending order of their U. S. sales ranking according to Blumenthal et al. [16].

Garlic [Allium sativum (L.) (Liliaceae)]

Garlic is the most popular herbal remedy. Postulated pharmacological actions of garlic include antibacterial, antiviral, antifungal, antihypertensive, blood glucose lowering, antithrombotic, antimutagenic, and antiplatelet actions [17]. Responsible for these activities are organosulfur compounds like alliin, allicin, diallyl disulfide, ajoene, and many others [18]. When the bulb is processed, alliin reacts with the enzyme allinase to produce the active constituent of garlic, allicin. Allicin is the compound often used to standardize many garlic formulations [19]. Garlic is available in different forms of pharmaceutical preparations, such as dry powder products, oil-macerated, volatile garlic oil, and juices of fresh garlic [20].

A clinical study assessed the effect of short-term administration of garlic supplements on single-dose ritonavir [21], a protease inhibitor that is mainly metabolized by CYP3A4 [22]. Acute dosing of this garlic extract over 4 days did not significantly alter the pharmacokinetics of ritonavir. In another trial, 11 female patients with metastatic breast cancer were treated with the CYP3A4 substrate docetaxel and garlic for 12 consecutive days. Garlic supplementation had no statistically significant effects on the pharmacokinetic profile of docetaxel when administered over the short term (4 days) or long term (12 days) [23]. A further study investigated the effect of garlic (over 12 days) on CYP2C9 activity by using warfarin as the probe substrate and showed no effect on pharmacokinetics or pharmacodynamics of the probe [24]. Another clinical study with 14 normal volunteers found no influence of garlic on the activity of CYP3A4 and CYP2D6 by a consecutive intake of garlic for 14 days [25]. In contrast, other studies showed an effect of garlic preparations by long-term use longer than 14 days: A clinical study with 10 healthy volunteers demonstrated that long-term (21 days) use of garlic caplets led to a significant decline in the plasma concentrations of saquinavir [26], an HIV-1 protease inhibitor that is metabolized by CYP3A4 [22]. The similarity in the magnitude of the decreases in AUC (− 51 %), Cmax (− 54 %), and concentration at 8 hours (C8) (− 49 %) suggested that garlic affected the bioavailability of saquinavir rather than its systemic clearance [26]. A cocktail interaction study in 12 healthy elderly subjects (mean = 67 years) who took garlic oil for 28 days showed no significant effects on CYP1A2, CYP2D6, and CYP3A4 activity, but it produced a significant decrease in CYP2E1 activity [27].

In summary, garlic appears to have only a minor potential to cause herb-drug interactions, with CYP2E1 as the only identifiable target. The saquinavir case needs further investigation.


#

Echinacea [Echinacea purpurea (L.) Moench (Asteraceae)]

Echinacea has anti-inflammatory and immunomodulating properties and is widely used for the treatment of the upper respiratory tract [28]. Echinacea preparations are made from the roots and/or other parts of the plant by juicing, alcohol extraction, infusion, decoction, or consumed as tablets or capsules [29]. Echinacea preparations are not chemically standardized. The constituent base is complex, consisting of phenols (cichoric and caftaric acid), polysaccharides, and alkylamides. The immunomodulatory effect was reported to be caused by alkylamides, which bind to human cannabinoid receptors 1 and 2, and inhibit tumor necrosis factor α [30].

With a sales ranking of #2 in the U. S. market [16], it is very important to determine the ability of echinacea to effect metabolic drug-drug interactions. Clinical studies indicate conflicting data about the effect of echinacea on the hepatic drug oxidation system. In a cocktail interaction study with 12 healthy volunteers, CYP1A2 was inhibited as assessed by a reduced oral clearance of the CYP1A2 probe caffeine (− 27 %, p = 0.049), there was a minor inhibitory effect on CYP2C9 (− 11 %, p = 0.001), while there was no effect on CYP2D6 or CYP3A4 [31]. Two further clinical studies determined the influence of echinacea on CYP3A4. In one of these studies, 13 healthy volunteers were dosed with lopinavir-ritonavir and the probe substrate midazolam. Neither lopinavir nor ritonavir pharmacokinetic parameter values were significantly altered after 14 days of echinacea administration [32]. The other study in 12 healthy male volunteers conducted with warfarin, a substrate of CYP2C9 and CYP3A4 for S-warfarin, and CYP3A4 and CYP1A2 for R-warfarin [33], found no clinically significant pharmacokinetic and pharmacodynamic interaction [34]. In a cocktail interaction study with 12 healthy volunteers determined by Gurley et al. [35], long-term supplementation of echinacea had no effect on CYP1A2, CYP2D6, CYP2E1, or CYP3A4 activity [35]. The same research group assessed the influence of echinacea on the activity of CYP2D6 in a clinical study with 18 normal volunteers, again with no effects [36].

In summary, the results of all described studies propose that echinacea has a fairly low capability for causing herb-drug interactions with human cytochrome P450 enzymes.


#

Saw palmetto [Serenoa repens (W. Bartram) Small (Arecaceae)]

Saw palmetto is the most popular herbal remedy used to alleviate the symptoms related to benign prostatic hyperplasia [37]. It has an ability to improve urologic symptoms and urine flow measures comparable to that of finasteride [35]. Saw palmetto extracts gained from the ripe, dried fruit have anti-inflammatory and spasmolytic properties. Most saw palmetto extracts are composed of mixtures of fatty acids including capric, caprylic, lauric, linoleic, linolenic, myristic, oleic, palmitic, and stearic acids, which typically account for 80–90 % of the extract [38].

There are only two clinical studies that have assessed the impact of saw palmetto on human cytochrome P450 enzymes. A cocktail interaction study with 12 healthy volunteers demonstrated that saw palmetto had no significant modulatory effects on CYP1A2, CYP2D6, CYP2E1, and CYP3A4 [35]. Another clinical study with 12 normal volunteers determined whether saw palmetto affects the activity of CYP2D6 and CYP3A4. For the probe substrates dextromethorphan (CYP2D6 activity) and alprazolam (CYP3A4 activity), the results indicated a lack of effect [38].

Overall, saw palmetto extract at generally recommended doses seems to be an unlikely candidate for CYP-mediated herb-drug interactions.


#

Ginkgo [Ginkgo biloba (L.) (Ginkgoaceae)]

Ginkgo extract is a popular herbal remedy used for a variety of disorders. EGb 761® special extract, for example, is a dry extract from Ginkgo biloba leaves (drug-extract ratio 35–67 : 1) that has been adjusted to 22–27 % ginkgo flavonoids and 5.0–7.0 % terpene lactones consisting of 2.8–3.4 % ginkgolides A, B, C and 2.6–3.2 % bilobalides, with a ginkgolic acid content less than 5 ppm [39]. EGb 761® interferes with various pathomechanisms relevant to dementing disorders [40], [41], [42]. A large number of clinical studies suggest that Ginkgo biloba extract (GBE) may have beneficial effects on memory, cognition, and the vascular system [43], [44], [45], [46], [47].

For an herbal drug to be taken by elderly people, often with several chronic diseases, the ability of GBE to cause metabolic drug-drug interactions should be known. The literature exhibits conflicting data, particularly on long-term treatment of 12 days and more. A clinical study in 11 healthy volunteers assessed the effect of GBE on the activity of CYP2C9 using flurbiprofen as a substrate. The subjects took 2 × 60 mg EGb 761® tablets thrice daily for 1 day. There was no significant difference between ginkgo or placebo treatment in any of flurbiprofenʼs kinetic parameters [48]. Another research group also could not find any effect of ginkgo, neither inducing nor inhibiting, on CYP1A2, CYP3A4 and CYP2C9 activity using racemic warfarin as a probe drug in 12 healthy male subjects, who took ginkgo for 7 consecutive days [49]. Using a cocktail phenotyping approach, a study with 18 healthy volunteers provided evidence that EGb 761® had no clinically relevant inhibitory or inducing effects towards human CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 when administered at chronic therapeutic doses for 8 days [15]. A clinical study with 18 healthy Chinese subjects investigated the potential herb-drug interaction between ginkgo and omeprazole, a widely used CYP2C19 substrate [50]. The volunteers had taken ginkgo tablets 2 × 70 mg twice daily for 12 days. In this study, plasma concentrations of omeprazole were significantly decreased and 5-hydroxyomeprazole as its metabolite significantly increased following ginkgo administration in comparison to baseline indicating an induction of CYP2C19 [50]. In contrast, a clinical study performed in 14 Chinese volunteers examined the possible effects of ginkgo as an inducer of CYP2C19 on single-dose pharmacokinetics of voriconazole [51]. Ginkgo pretreatment for 12 days did not significantly affect pharmacokinetic parameters of voriconazole [51]. A clinical study evaluated as the primary objective the effect of GBE on the exposure of lopinavir, an HIV protease inhibitor metabolized by CYP3A4, in 14 healthy volunteers [52]. The secondary objectives were to compare ritonavir exposure pre- and post-GBE, and assess the effect of GBE on single doses of the probe drug midazolam. Ginkgo biloba extract decreased midazolam AUC(0-∞) by 34 % (p = 0.03) and Cmax by 31 % (p = 0.03) but did not significantly affect the exposures of lopinavir and ritonavir [52]. A clinical study performed in 14 healthy male volunteers determined the effects of GBE on the pharmacokinetics of bupropion, a substrate of CYP2B6. Ginkgo biloba extract administration of 240 mg · day−1 (two 60-mg capsules taken twice daily) for 14 days did not significantly alter the basic pharmacokinetic parameters of bupropion [53]. Another clinical study with 12 healthy volunteers assessed the influence of GBE on the activity of CYP2D6 and CYP3A4 for about 14 days. For the probe substrates dextromethorphan (CYP2D6 activity) and alprazolam (CYP3A4 activity), no statistical differences were observed between baseline and post-GBE treatment indicating a lack of effect on CYP2D6 and CYP3A4 [54]. In a cocktail interaction study with 12 healthy volunteers and treatment duration of 28 days, no significant effect on CYP1A2, CYP2D6, CYP2E1, and CYP3A4 activity for Ginkgo biloba was found [27]. But other studies observed that ginkgo inhibits CYP3A4 [55], [56]. A study with 15 subjects showed that the geometric mean midazolam AUC(0-∞) prior to GBE administration was reduced by 34 % after GBE administration [55]. The outcome of the study of Uchida and colleagues performed in 10 male healthy volunteers was an inhibiting effect of GBE on CYP3A4 activity as shown by an increased AUC(0-∞) by 25 % and decreased oral clearance by 26 % of midazolam, and an inducing effect on CYP2C9 activity according to a reduced AUC(0-∞) by 16 % of tolbutamide [56]. A pharmacokinetic study in healthy volunteers conducted with diazepam as a substrate of CYP2C19 did not suggest the presence of an herb-drug interaction [57].

Altogether, it appears that GBE may have some effect on the activity of CYP enzymes when applied in patients, probably depending on the preparation used.


#

Cranberry [Vaccinium macrocarpon (Aiton) (Ericaceae)]

Cranberries are primarily cultivated for consumption as foods and beverages [58]. Products of the cranberry industry include fresh fruit (5 %), juices (60 %), sauces, dried fruit, and ingredients (35 %), such as frozen fruit, juice concentrates, and spray-dried powders [59], [60]. Cranberry juice contains phytochemicals such as proanthocyanidins, flavonols, and quercetin [61]. The juice and concentrated extracts of cranberries are increasingly popular among consumers because of its use for the prevention and adjunctive treatment of urinary tract infections [58]. Moreover, cranberry juice shows efficacy in reducing urinary tract infections by acidifying the urine [62] and in reducing bacteriuria in elderly persons [63]. It also features positive effects against drug-resistant bacteria [64].

The large popularity of cranberry juice should give reason to examine its ability to cause herb-drug interactions. There are only a few data available. A clinical study in 8 healthy volunteers investigated the potential interaction between cranberry juice and diclofenac, a substrate of CYP2C9. An intake of 180 mL of cranberry juice twice a day for 5 days did not change the pharmacokinetics of diclofenac [65]. Another clinical study evaluated the effect of cranberry juice and other beverages on CYP2C9 activity. Fourteen healthy volunteers received flurbiprofen as a probe substrate for CYP2C9 in combination with 8 oz. of cranberry juice. None of the beverages altered CYP2C9-mediated clearance of flurbiprofen in humans [66]. Further investigations on CYP2C9 in clinical studies showed that cranberry juice also had no interaction potential on the pharmacokinetics of warfarin [67], [68]. In addition to CYP2C9, Lilja and colleagues assessed the effect of cranberry juice on the activities of CYP1A2 and CYP3A4. Ten healthy volunteers took 200 mL cranberry juice thrice daily for 10 days. They observed no effects of cranberry juice on the pharmacokinetics of tizanidine (as CYP1A2 probe) and midazolam (as the CYP3A4 probe) [68]. Another clinical study in 12 healthy male volunteers revealed that an intake of 240 mL of cranberry juice did not affect the pharmacokinetics of cyclosporine as a substrate of CYP3A [69].

In summary, daily ingestion of more than 1 glass of cranberry juice seems not to alter the activities of cytochrome P450 enzymes.


#

Soy [Glycine max (L.) Merr. (Fabaceae)]

Soy is a plant native to East Asia, commonly grown for its bean, which has been reported to have various health benefits. Soybeans provide ample amounts of α-linolenic acid [70] and significant amounts of isoflavones. Three soybean isoflavones, genistein, daidzein, and glycitein, and their various glycoside forms account for roughly 50, 40, and 10 %, respectively, of total isoflavone content [71]. Epidemiological and experimental researchers have provided extensive information on the anti-estrogenic effects of soy isoflavones on human health [72]. In China, Japan, Korea, and other countries in the Far East as well as lately in Western countries, the bean and products made from it such as soy sauce, soy flour, soy milk, and tofu are a popular part of the diet.

Hence, it is very important to take the ability of soy for an herb-drug interaction into account. A clinical study performed in 20 healthy volunteers examined the potential effect of daidzein on CYP1A2 activity and on the pharmacokinetics of theophylline as a probe substrate [73]. Plasma concentrations and derived parameters including AUC(0–48), Cmax, and t 1/2 were significantly increased by approximately one-third [73]. Another clinical study investigated the effect of 1 g genistein once daily for 14 days on the caffeine-based metrics of CYP1A2 and CYP2A6 in 18 healthy female volunteers [74]. Genistein decreased the urinary caffeine metabolite ratio used to assess CYP1A2 activity by 41 %, whereas the urinary ratio for CYP2A6 activity increased by 47 %, suggesting that genistein inhibited CYP1A2 and induced CYP2A6 [74]. A clinical study in 18 healthy Chinese female volunteers provided no effects of soy extract on the pharmacokinetics of losartan as a substrate of CYP2C9 and CYP3A4 [75]. A clinical study assessed the drug interaction potential of soy extract on CYP3A using the urinary excretion of the 6β-hydroxycortisol/cortisol ratio as a marker of enzyme induction. Twenty healthy subjects received a soy extract containing 50 mg isoflavones twice daily for 14 days. The soy extract had no effect [76].

Altogether, soybean constituents may be weak inhibitors of CYP1A2, which translates into clinical relevance only in extraordinary situations.


#

Asian ginseng [Panax ginseng (L.) (Araliaceae)]

Asian ginseng (ginseng) is marketed for a wide range of indications, which include erectile dysfunction, cancer prevention, enhanced physical function, and improved cognitive functions [77]. Ginseng extracts are generally standardized to ginsenosides. Ginsenosides are a class of steroid glycosides and triterpene saponins [78]. The roots of Panax ginseng contain at least 25 different triterpene saponins [79]. Ginsenosides Rb1, Rb2, Rc, Rd, Re, Rf, and Rg1 are reported as major constituents and each ginsenoside has been shown to have different pharmacological effects, including immune system modulation, antistress activities, and antihyperglycemic activities, anti-inflammatory, antioxidant, and anticancer effects, antiplatelet, antithrombotic, vasodilatory, cardiotonic, angiogenic, and neuroprotective effects [79], [80], [81], [82].

For an herbal drug to be widely used by the elderly community for a variety of indications, the ability of ginseng to cause metabolic drug-drug interactions should be determined. Clinical studies in humans have shown that Panax ginseng has no effect on a number of CYP enzymes [77]. In a cocktail interaction study with 12 healthy elderly volunteers (mean = 67 years), Panax ginseng inhibited CYP2D6 activity assessed using debrisoquine urinary recovery ratios significantly, but only by 7.0 % [27]. No significant effect on CYP1A2, CYP3A4, or CYP2E1 activity for Panax ginseng was found [27]. One trial investigated the interaction between warfarin and Panax ginseng in ischemic stroke patients. Twenty-five patients were enrolled in the study; twelve patients in the ginseng group received Panax ginseng and warfarin for 2 weeks, and the control group (n = 13) received only warfarin for the same duration of time. There were no statistically significant differences between the ginseng group and control group [80]. Another study in 12 healthy male subjects conducted to investigate the effect of ginseng on the pharmacokinetics and pharmacodynamics of warfarin found no clinically significant changes in AUC, t 1/2, and apparent total clearance [83]. This study confirmed that ginseng had no effect on the activity of CYP1A2, CYP3A4, or CYP2C9 in healthy volunteers [83]. Another clinical study could also not show any effect of Panax ginseng on CYP3A. Twenty healthy subjects received 100 mg Panax ginseng standardized to 4 % ginsenosides twice daily for 14 days. The urinary excretion of the 6β-hydroxycortisol/cortisol ratio was used as a marker of CYP3A induction but no significant alteration was observed for Panax ginseng [76].

In summary, it appears that Asian ginseng has no effect on the activity of CYP enzymes in vivo.


#

Black cohosh [Actaea racemosa (L.) Nutt. (Ranunculaceae)]

Extracts of black cohosh are made from the roots and rhizomes of the plant. Black cohosh preparations (tinctures or tablets of dried materials) are commonly used as an alternative to hormone therapy in perimenopausal women to treat symptoms such as hot flashes, vaginal dryness, and mood swings [19], [84].

A few clinical studies conducted the effect of black cohosh on human cytochrome P450 enzymes. A cocktail interaction study with 12 healthy volunteers showed no significant effects of black cohosh on CYP1A2, CYP2E1, and CYP3A4 activity, but it did have an effect on CYP2D6 [85]. Black cohosh exhibited a statistically significant decrease in the CYP2D6 phenotype (p = 0.02), but the magnitude of the result (~ 7.0 % reduction) is not clinically relevant [85]. In another clinical study with 18 normal volunteers, Gurley et al. assessed the influence of black cohosh only on the activity of CYP2D6 to corroborate their earlier findings. This time, comparisons of pre- and post-supplementation of 8-hour debrisoquine urinary recovery ratios revealed no statistically significant effects on CYP2D6 [36]. Another pharmacokinetic study in 19 healthy volunteers conducted with midazolam as a substrate of CYP3A also did not suggest the presence of an herb-drug interaction [86].

In summary, black cohosh appears to have no clinically relevant effects on cytochrome P450 activity.


#

St. Johnʼs wort [Hypericum perforatum (L.) (Hypericaceae)]

St. Johnʼs wort (SJW) is an herbaceous perennial plant native to Europe. Extracts obtained from the aerial parts of Hypericum perforatum have been recommended traditionally for a wide range of medical conditions [87]. St. Johnʼs wort is commonly used to treat mild-to-moderate depression but is also used to treat anxiety, obsessive-compulsive disorder, and premenstrual syndrome [19]. The extracts of SJW contain numerous pharmacologically active ingredients, including naphthodianthrones (e.g., hypericin and its derivatives) and phloroglucinols derivatives (e.g., hyperforin, which inhibits the reuptake of a number of neurotransmitters, including serotonin) [87]. The most common recommended dose is 900 mg per day standardized to 0.3 % hypericin to treat depression [19].

Several clinical studies have clearly revealed that SJW may alter CYP activity. Many of the interaction studies indicate that SJW is a potent inducer of CYP3A4, CYP2E1, and CYP2C19, with no effect on CYP1A2, CYP2D6, or CYP2C9 [20], [77]. The effect of SJW on CYP3A4 is seen after long-term treatment. Markowitz et al. assessed the effects of SJW on CYP2D6 and CYP3A4 activity. Seven normal subjects received 3 times daily a commercial SJW formulation (Solaray®) 300 mg, standardized to 0.3 % hypericin for 4 days. Dextromethorphan (CYP2D6 activity) and alprazolam (CYP3A4 activity) were administered as probe substrates. No statistically significant differences were found in any estimated pharmacokinetic parameter for alprazolam or dextromethorphan for this short-term treatment [88]. A cocktail interaction study with 12 healthy volunteers examined the effect of SJW on CYP activity. Tolbutamide (CYP2C9), caffeine (CYP1A2), dextromethorphan (CYP2D6), oral midazolam (intestinal wall and hepatic CYP3A), and intravenous midazolam (hepatic CYP3A) were administered before, with short-term SJW dosing (900 mg), and after 2 weeks of intake (300 mg 3 times a day) to determine CYP activities [89]. Short-term administration of SJW had no effect on CYP activities. Long-term SJW administration caused a significant (p < 0.05) increase in oral clearance of midazolam and a corresponding significant decline in oral bioavailability [89]. There are many other clinical studies that confirm these findings. Another clinical cocktail interaction study assessed the influence of SJW on the activity of CYP1A2, CYP2D6, and CYP3A4. Eight healthy male and 8 healthy female subjects were treated with SJW extract (3 × 300 mg · day−1) for 14 days [90]. After 2 weeks of treatment with SJW, the mean increase in the 6β-hydroxycortisol/cortisol molar concentration ratio in urine, used as an index of activity of CYP3A4, was 85 %. Additionally, the authors found a slight (but not significant) increase in the paraxanthine/caffeine ratio in saliva after SJW. However, most of the subjects exhibiting an apparent induction of CYP1A2 were females [90]. No influence of SJW on CYP2D6 activity was found [90]. A clinical cocktail interaction study performed in 12 elderly subjects (mean = 67 years) assessed the pre- and post-supplementation phenotypic ratios for CYP3A4, CYP1A2, CYP2E1, and CYP2D6 [27]. Twenty-eight days of SJW supplementation resulted in a 141 % increase in the mean one-hour 1-hydroxymidazolam/midazolam serum ratio (p < 0.001) [27]. Similar to its effect on CYP3A4, SJW produced significant increases in CYP2E1 activity (p = 0.006). No statistically significant differences in mean values were noted for CYP1A2 and CYP2D6 [27]. In a later clinical study performed by the same researchers [36], no significant differences were observed among the mean baseline debrisoquine urinary recovery ratio by supplementation of 14 days of SJW. In contrast to Wenk and colleagues, no sex-related changes in CYP phenotypes were noted. Two further clinical studies suggested that SJW is an inducer of CYP3A4 following 14 days of treatment [91], [92]. There are also two clinical studies which determined the effect of SJW on CYP2C19 activity. In each study, 12 healthy volunteers received a 300-mg SJW tablet 3 times daily for 14 days. In one study, the activities of CYP2C19 and CYP1A2 were measured using mephenytoin and caffeine, respectively [93], and in the other study, the activities of CYP2C19 and CYP3A4 were measured using omeprazole [94]. In both studies, it was found that SJW treatment significantly increased CYP2C19 activity.

Furthermore, clinical data imply that hyperforin content affects the extent of SJW interactions, since extracts with a low hyperforin amount had a weak or no effect on CYP activity. A clinical study compared the effects of 2 SJW preparations with high and low hyperforin content on the pharmacokinetics of cyclosporine. In a crossover study, 10 renal transplant patients were randomized into 2 groups and received SJW extract 300 mg (two 150 mg capsules) 3 times a day (total dose, 900 mg/d) containing low or high concentrations of hyperforin for 14 days in addition to their regular regimen of cyclosporine [95]. The study showed a significant difference between the effects of the 2 SJW preparations on cyclosporine pharmacokinetics. The area under the plasma concentration-time curve, within one dosing interval (AUC0–12; p < 0.0001), values with high hyperforin SJW comedication were 45 % lower (95 % CI − 37 % to − 54 %; p < 0.05) than for low hyperforin SJW [95]. Arold and colleagues performed two clinical interaction studies with 28 healthy volunteers in each study. In study A, alprazolam (CYP3A4) and caffeine (CYP1A2), and in study B, tolbutamide (CYP2C9) and digoxin (p-glycoprotein), were given as probe substrates, respectively. The participants received SJW with a low hyperforin content (Esbericum® capsules; 240 mg · day−1, 3.5 mg hyperforin) or placebo for 10 days. No statistically significant differences were found in the primary kinetic parameters between the placebo group and the SJW group at the end of both studies [96]. Another two clinical studies conducted by Mueller and colleagues evaluated the effect on CYP3A function of SJW preparations with a wide range from very low to high hyperforin content. In the first study, 42 healthy volunteers were randomized into 6 different SJW medication groups for 14 days. A single oral dose of midazolam was used as a probe substrate. All SJW preparations tested resulted in a decrease in midazolam AUC, although the extent of the effect differed [97]. St. Johnʼs wort extract with a hyperforin content of 41 mg/day decreased midazolam AUC0–12 by 79.4 % (95 % CI − 88.6 to − 70.1). St. Johnʼs wort powder tablets with a hyperforin content of 12 mg/day resulted in a decrease of 47.9 % (95 % CI − 59.7 to − 36.2), while SJW powder tablets with an amount of 0.13 mg/day of hyperforin reduced midazolam AUC0–12 by only 21.1 % (95 % CI − 33.9 to − 8.3) [97]. The second study evaluated the effect of an SJW powder only with a low hyperforin content on CYP3A function. Twenty healthy male volunteers received SJW as capsules containing 500 mg Hyperici herba powder with 0.06 mg total hyperforin per capsule and had to take two capsules per day, for 14 days. Midazolam AUC0-∞ was reduced by 11.3 % (95 % CI − 22.8 to 0.21) indicating a significant but mild induction of CYP3A function [98]. No significant changes were observed after SJW treatment regarding midazolam Cmax, t max, and t 1/2 (p > 0.05) [98].

Apart from using probe drugs in clinical interaction studies to show that SJW is a potential inducer of CYP3A4, CYP2E1, and CYP2C19, SJW has also been shown to have the ability to clinically interact with a number of frequently used drugs. St. Johnʼs wort may reduce the efficacy of oral contraceptives (e.g., induction of ethinyl estradiol-norethindrone metabolism [99], [100]; decrease in serum 3-ketodesogestrel concentrations [101]), may reduce the pharmacokinetics of imatinib by increasing its clearance [102], [103], may interact with cardiovascular drugs (e.g., decreased plasma concentrations of atorvastatin [104], ivabradine [105], and R- and S-verapamil [106]), may induce the apparent clearance of both S- and R-warfarin, which in turn resulted in a significant reduction in the pharmacological effect of rac-warfarin [83], may interact with drugs acting on the central nervous system (e.g., decreased plasma concentration of quazepam [107], alprazolam [92], and midazolam [27], [89], [97], [99]), may reduce plasma voriconazole concentrations after long-term but not short-term administration [108], may alter gliclazide pharmacokinetics [109], and may reduce the AUC of the HIV-1 protease inhibitor indinavir [110].

In summary, clinical evidence of the effects of SJW on CYP enzymes is undisputed. There are numerous studies which have shown that the inducing effect of SJW depends on treatment duration and the preparation, primarily the amount of hyperforin. Patients and physicians should be well informed about the interaction potential of St. Johnʼs wort.


#

Milk thistle [Silybum marianum (L.) Gaertn. (Asteraceae)]

Extracts of milk thistle are recognized for the treatment of liver injury. The active principle is a mixture of flavolignans called Silymarin [20]. Silymarin is made from the seeds of milk thistle and is composed of six closely related flavonolignans (silibinin, isosilybin A, isosilybin B, silychristin, isosilychristin, silydianin) and one flavonoid (taxifolin) [111]. Silibinin, the major active constituent of silymarin, consists of 2 diasteroisomers, silybin A and silybin B [111], [112]. Silymarin has cytoprotective, antioxidative, and radical scavenging as well as anti-inflammatory and antifibrotic properties [14]. It is used to self-treat hepatic disorders, including hepatitis C and cirrhosis, and as a hepatoprotectant, particularly for mushroom poisoning [113].

For a popular herbal product to be taken for hepatoprotection and chemoprevention, the ability of milk thistle extract to cause metabolic drug-drug interactions should be known. Several in vivo studies in humans indicate that milk thistle has no effect on the hepatic drug oxidation system. A clinical study was conducted to examine the effect of silymarin on cytochrome P450 3A4. Sixteen healthy male volunteers were administered with immediate release nifedipine as a CYP3A4 test drug either alone or with the coadministration of silymarin. The coadministration of silymarin for 1 day did not considerably change the extent of absorption or metabolism of nifedipine but might decrease the absorption rate. Silymarin was not a potent CYP3A4 inhibitor in vivo [14]. Van Erp and colleagues investigated the effect of milk thistle on the pharmacokinetics of irinotecan, a substrate for CYP3A4. Neither short-term intake (4 days) nor long-term intake (12 days) of milk thistle showed significant effects on irinotecan clearance [114]. Gurley et al. investigated the in vivo effect of milk thistle on human cytochrome P450 three times. A clinical study with 19 normal subjects assessed the clinical significance of milk thistle supplementation on human cytochrome P450 3A activity. The studyʼs purpose was to compare the effect of milk thistle on CYP3A to a clinically recognized inducer, rifampin, and inhibitor, clarithromycin. In contrast to rifampin and clarithromycin, no significant changes in the probe substrate midazolam pharmacokinetics were observed as a result of milk thistle supplementation [86]. In a similar study, 18 healthy volunteers were administered with a standardized milk thistle extract to assess the effect on cytochrome P450 2D6. The study conducted with debrisoquine as a substrate of CYP2D6 also did not suggest the presence of an herb-drug interaction [36]. In a cocktail interaction study with 12 healthy volunteers, he and his colleagues determined long-term supplementation of milk thistle extracts on CYP1A2, CYP2D6, CYP2E1, or CYP3A4 activity. They found no statistically significant differences in mean CYP1A2, CYP2D6, CYP2E1, or CYP3A4 phenotypic ratios [35].

In summary, current data suggest that milk thistle has no major effects on the activity of CYP enzymes when applied in patients.


#

Green tea [Camellia sinensis (L.) Kuntze (Theaceae)]

After withering the freshly picked leaves, a brief heating, roasting, or steaming prevents the fermentation of the tea leaves. For this reason, almost all active ingredients such as catechins (70 %), minor flavonols (10 %), and polymeric flavonoids (20 %) contained in the fresh leaves will remain [19]. The main catechin component of green tea is epigallocatechin gallate (EGCG) that accounts for 50–80 % of the catechins in green tea [115], [116]. Polyphenon E® (Poly E), for example, is a widely used concentrated green tea extract (GTE) from green tea leaves. It contains a total catechin fraction of 89 % with EGCG as the main component accounting for 65 % of the material followed by 9.0 % epicatechin, 6.6 % epicatechin gallate, 3.8 % epigallate catechin, 1.0 % catechin, 0.2 % gallocatechin, and 0.2 % catechin gallate [117]. Green tea, GTE, and its major active compound ECGC demonstrated antioxidant, anticarcinogenic, anti-inflammatory, antiatherogenic, immunomodulatory, and chemopreventive properties [118], [119], [120], [121]. It has antiatherosclerotic effects on dysfunctional vessels in smokers through increasing the level of nitric oxide and reducing oxidative stress [122], [123]. Green tea may have cardiovascular protecting effects through inhibition of angiotensin-converting enzyme activity [124]. Standardized green tea compounds are effective for decreasing blood pressure, low-density lipoprotein cholesterol, and oxidative stress [125].

The large variety of assumed medical uses of green tea imply that the potential for drug interactions could be high. A clinical study with 11 healthy volunteers assessed the influence of decaffeinated GTE (dGTE) on the activity of CYP2D6 and CYP3A4. The probe substrates dextromethorphan (CYP2D6 activity) and alprazolam (CYP3A4 activity) were administered orally at baseline, and again after treatment with four dGTE capsules (< 1 mg caffeine) per day for 14 days. No significant differences in dextromethorphan and alprazolam pharmacokinetics were observed at baseline and after treatment with dGTE indicating a lack of effect on CYP2D6 and CYP3A4 [126]. In a clinical cocktail interaction study with 42 healthy volunteers, no clinically significant effects on CYP1A2, CYP2C9, CYP2D6, and CYP3A4 activity for GTE were found [127]. The study participants received a cocktail of CYP metabolic probe drugs, including caffeine, losartan, dextromethorphan, and buspirone for assessing the activity of CYP1A2, CYP2C9, CYP2D6, and CYP3A4, respectively. The subjects underwent 4 weeks of green tea catechin intervention at a dose that contains 800 mg EGCG daily. The intervention did not alter the phenotypic indices of CYP1A2, CYP2C9, and CYP2D6, but resulted in a 20 % increase (p = 0.01) in the area under the plasma buspirone concentration-time profile, suggesting a small reduction in CYP3A4 activity [127].

Overall, both authors concluded that repeated green tea catechin administration is unlikely to modify the disposition of medications metabolized by CYPs.


#

Valerian [Valeriana officinalis (L.) (Valerianaceae)]

Valerian is a popular remedy prepared from its roots, rhizomes (underground stems), and stolons (horizontal stems). The root is chiefly used for medicinal benefits. It can be found in capsule, tea, tablet, or liquid extract forms. The most abundant constituents of valerian are monoterpenes and sesquiterpenes, including the genus-specific valepotriates and valerenic acid [128]. Valerian root also contains appreciable levels of gamma-aminobutyric acid (GABA) [129] and has sedative, anxiolytic, and hypnotic properties [130]. It is often taken to help alleviate insomnia. There are several clinical studies to evaluate the evidence of efficacy of valerian as a treatment for insomnia [131], [132], [133], [134].

A clinical study with 12 normal volunteers assessed the influence of a valerian supplement on the activity of CYP2D6 and CYP3A4. The probe substrates dextromethorphan (CYP2D6 activity) and alprazolam (CYP3A4 activity) were administered orally at baseline, and again after exposure to two valerian tablets nightly for 14 days. Valerian showed no clinically relevant effects on the disposition of medications primarily dependent on the CYP2D6 or CYP3A4 pathways for metabolism [135]. In a clinical interaction study with 12 healthy volunteers, the study participants received valerian for 28 days. Probe drug cocktails of caffeine for CYP1A2 activity and midazolam for CYP3A4 activity, followed 24 hours later by debrisoquine for CYP2D6 activity and chlorzoxazone for CYP2E1 activity to avoid potential interference, were administered before (baseline) and at the end of supplementation. Valerian had no significant effect on any CYP phenotypes [85].

In summary, valerian appears unlikely to produce CYP-mediated herb-drug interactions.


#

Kava [Piper methysticum (G.) Forst. (Piperaceae)]

Kava is still a popular herbal beverage. The commercial products, if not withdrawn from the market for hepatotoxicity, are prepared from dried rhizomes of the kava plant, and the more contemporary dosage form is a capsule, which usually contain a standard 30 % of kavalactones. The constituents of kava extract are kavalactones, kawain, methysticin, dihydromethysticin, desmethoxyyangonin, and dihydrokawain [19]. Kavalactones, the assumed active principles, are predominantly concentrated in the plantʼs rhizome rather than in its upper stems or leaves [136]. Kavalactones effects are a slight numbing of the gums and mouth, and vivid dreams. Kava has been reported to improve cognitive performance and promote a cheerful mood [137]. Kava has anxiolytic and sedative properties and is often suggested to alleviate the symptoms of anxiety [138].

There are only a few clinical studies which revealed the clinical influence of potential interactions mediated by cytochrome P450 enzymes. A clinical study performed by Gurley and colleagues assessed the kava supplementation on human CYP3A activity using midazolam as a phenotypic probe. Sixteen healthy volunteers received kava for 14 days. Midazolam disposition was not affected by kava supplementation [139]. The same research group assessed the influence of kava on the activity of CYP2D6 in a clinical study with 18 healthy volunteers. Kava was not a potent modulator of human CYP2D6 in vivo [36]. A cocktail interaction study in 12 healthy subjects for 28 days showed no significant effects on CYP1A2, CYP2D6, and CYP3A4 activity for kava, but significantly reduced phenotypic ratios for CYP2E1 (~ 40 %, p = 0.009) [85].

In summary, kava may interact with CYP2E1 substrates. Thus, concomitant ingestion of kava and drugs that are CYP2E1 substrates may increase their therapeutic and adverse effects.


#

Goldenseal [Hydrastis canadensis (L.) (Ranunculaceae)]

Goldenseal is used as a versatile herbal remedy and has many different medicinal properties. Its roots and rhizomes, which internally are bright yellow in color, have been used as a traditional medicine for the treatment of infection, inflammation, and as an immune system booster. It is taken orally to treat upper respiratory infections and gastrointestinal tract disorders [140]. Modern herbalists consider it an alternative anticatarrhal, anti-inflammatory, antiseptic, astringent, bitter tonic, laxative, and muscular stimulant [140]. Commercial preparations of goldenseal may be purchased in tincture form or as a liquid extract [141]. Goldenseal extract contains isoquinoline alkaloids, including berberine, (+)- and (−)-hydrastine, and lesser amounts of hydrastinine [142]. Chemically, these three goldenseal alkaloids possess a methylenedioxyphenyl moiety, which, in studies of cytochrome P450 (P450) -dependent drug metabolism, frequently give rise to inhibition [142].

Gurley et al. determined the effects of goldenseal supplementation only on human CYP3A activity. Sixteen healthy volunteers received goldenseal for 14 days. Statistically significant increases (p < 0.05) in midazolam AUC(0-∞) (62 %), elimination half-life (57 %), and Cmax (41 %) were observed after goldenseal extract supplementation. Goldenseal reduced midazolam apparent oral clearance by 36 % (p < 0.001) [139]. The same research group conducted a clinical assessment in 18 healthy volunteers on the effects of goldenseal only on human CYP2D6 activity. Pre- and post-supplementation phenotypic trait measurements were determined for CYP2D6 using 8-hour debrisoquine urinary recovery ratios. Comparisons of pre- and post-supplementation 8-hour debrisoquine urinary recovery ratios revealed significant inhibition (~ 50 %) of CYP2D6 activity for goldenseal [36]. In a cocktail interaction study, 12 healthy volunteers received goldenseal for 28 days. The probe drug cocktail of midazolam (for CYP3A4/5 activity) and caffeine (CYP1A2 activity), followed 24 hours later by chlorzoxazone (CYP2E1 activity) and debrisoquine (CYP2D6 activity), were administered before (baseline) and at the end of supplementation. Goldenseal produced significant reductions in CYP2D6 (p < 0.0001) and CYP3A4/5 (p < 0.0001) phenotypes [85].

Overall, goldenseal seems to be a mild to moderate inhibitor of CYP3A4/5 and CYP2D6. Accordingly, patients should refrain from taking goldenseal supplements concomitantly with prescriptive medications, particularly those extensively metabolized by CYP2D6 and CYP3A4/5.


#

Other herb-drug interactions

In addition to the herbs described above, there are further plants with therapeutic benefits, which have been investigated with regard to effects on the human cytochrome P450 drug metabolizing system. A clinical study in 12 healthy male subjects showed no effect of ginger [Zingiber officinale (R.) (Zingiberaceae)] on the pharmacokinetic parameters of S- or R-warfarin [49]. A cocktail interaction study investigated the effects of multiple doses of three herbal medicines on metabolic activities of CYP1A2, 2C9, 2C19, 2D6, 2E1, and 3A4 [143]. The roots of Angelica tenuissima (L.) [Apiaceae], Angelica dahurica (L.) [Apiaceae], and Scutellaria baicalensis (L.) [Lamiaceae] were administered to 24 healthy male volunteers. Angelicae tenuissimae radix had no influences on CYP activities. Angelicae dahuricae radix significantly decreased CYP1A2 activity to 10 % of baseline activity (95 % CI 0.05–0.21). Scutellariae radix showed significant changes in CYP2C9 and CYP2E1 activities. Baseline values for losartan as a CYP2C9 probe were decreased to 71 % (0.54–0.94) and the metabolic activity of chlorzoxazone as a CYP2E1 probe showed a 1.42-fold (1.03–1.97) increase [143]. Furthermore, in another study with 17 healthy male subjects, baicalin, a flavone glucuronide of baicalein extracted from Scutellariae radix, significantly induced CYP2B6 activity as measured by bupropion hydroxylation (an average 63 % increase in the AUC ratio of hydroxybupropion over bupropion and an 87 % increase in the AUC of hydroxybupropion) [144]. There are also several other single constituents of plants which may have effects on cytochrome P450 enzymes. For example, resveratrol as the main non-flavonoid polyphenol found in red wine and grapes [Vitis vinifera (L.) (Vitaceae)] has a wide range of biological and pharmacological activities including antioxidant, anti-inflammatory, antimutagenic, and anticarcinogenic effects [145]. A cocktail interaction study in 42 healthy volunteers determined the effect of pharmacological doses of resveratrol on CYP1A2, CYP2C9, CYP2D6, and CYP3A4. Resveratrol intervention was found to inhibit the phenotypic indices of CYP3A4, 2D6, and 2C9, and to induce the phenotypic index of 1A2 [146]. Curcumin, a yellow curry spice extracted from the rhizome of Curcuma longa (L.) [Zingiberaceae], is a polyphenolic non-flavonoidic that displays anti-inflammatory and antioxidant activities [145]. A clinical study with 16 unrelated, healthy Chinese men investigated the effect of curcumin on the activities of CYP1A2 and CYP2A6 using caffeine as a probe drug. Cytochrome P450 1A2 activity was decreased by 28.6 % (95 % CI 15.6–41.8; p < 0.000), while increases were observed in CYP2A6 (by 48.9 %; 95 % CI 25.3–72.4; p < 0.000) [147].


#

Limitations

Reported drug-herb interaction studies used a broad range of specific preparations of the respective herbs. The information on these preparations is sparse in many of the studies, and even if commercial products have been used, other batches will have different compositions and thus may have different potentials do cause drug-drug interactions. Phytopharmaceuticals composed of different fractions from parts of a plant (leaves, roots, seeds, fruit, other parts) may have completely different compositions. Thus, any extrapolation of the data gathered here to other products is flawed to an unknown extent. Furthermore, many studies used invalid phenotyping metrics and/or had small numbers of participants without proper estimation of the sample size required to answer the scientific question. The published literature thus provides only a rough estimate to which extent marketed drugs prepared from specific plants or other preparations of these plants, such as infusions, would indeed cause clinically relevant drug interactions in patients.


#
#

Conclusions

Our article provides a brief description of clinical interaction studies between phytopharmaceuticals and human cytochrome P450 enzymes for the top 14 common botanicals sales in the U. S. The majority of the herbal drugs appeared to have no clear effects on most of the CYPs examined. If there were an effect, the herbal drugs would qualify as mild inhibitors (less than a 2-fold change in enzyme activity) in almost all cases, e.g., in the case of inhibition of CYP2E1 by garlic and by kava or for inhibitory effects of soybean components on CYP1A2. The most pronounced effects were the well-known induction of several members of the CYP family by St. Johnʼs wort and the inhibitory effect of goldenseal on CYP3A and CYP2D6, both being borderline between mild and moderate (more than 2-fold but less than 5-fold) in magnitude. With these two exceptions, concomitant intake of herbal drugs is not a major risk for drugs that are metabolized by CYPs.


#
#

Conflict of Interest

No conflict of interest is to be declared.

  • References

  • 1 Moore LB, Goodwin B, Jones SA, Wisely GB, Serabjit-Singh CJ, Willson TM, Collins JL, Kliewer SA. St. Johnʼs wort induces hepatic drug metabolism through activation of the pregnane X receptor. Proc Natl Acad Sci USA 2000; 97: 7500-7502
  • 2 Bailey DG, Spence JD, Munoz C, Arnold JM. Interaction of citrus juices with felodipine and nifedipine. Lancet 1991; 337: 268-269
  • 3 Fuhr U, Jetter A, Kirchheiner J. Appropriate phenotyping procedures for drug metabolizing enzymes and transporters in humans and their simultaneous use in the “cocktail” approach. Clin Pharmacol Ther 2007; 81: 270-283
  • 4 Huang SM, Zhang L. Guidance for industry: drug interaction studies – study design, data analysis, implications for dosing, and labeling recommendations. Available at http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM072101.htm Accessed December 02, 2011
  • 5 European Medicines Agency. Guideline on the investigation of drug interactions. Available at http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2010/05/WC500090112.pdf Accessed December 02, 2011
  • 6 US Food and Drug Administration. Drug development and drug interactions: table of substrates, inhibitors and inducers. Available at http://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/ucm093664.htm Accessed December 02, 2011
  • 7 Tanaka E, Kurata N, Yasuhara H. How useful is the “cocktail approach” for evaluating human hepatic drug metabolizing capacity using cytochrome P450 phenotyping probes in vivo?. J Clin Pharm Ther 2003; 28: 157-165
  • 8 Zhou H, Tong Z, McLeod JF. “Cocktail” approaches and strategies in drug development: valuable tool or flawed science?. J Clin Pharmacol 2004; 44: 120-134
  • 9 Turpault S, Brian W, Van Horn R, Santoni A, Poitiers F, Donazzolo Y, Boulenc X. Pharmacokinetic assessment of a five-probe cocktail for CYPs 1A2, 2C9, 2C19, 2D6 and 3A. Br J Clin Pharmacol 2009; 68: 928-935
  • 10 Zhang L. A regulatory perspective on the utilization of cocktail approach in assessing drug interactions. Available at http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/UCM237160.htm Accessed December 02, 2011
  • 11 Zhang L, Reynolds KS, Zhao P, Huang SM. Drug interactions evaluation: an integrated part of risk assessment of therapeutics. Toxicol Appl Pharmacol 2010; 243: 134-145
  • 12 US Food and Drug Administration. Guidances (Drugs). Available at http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm Accessed December 02, 2011
  • 13 Ma JD, Nguyen ET, Tsunoda SM, Greenberg HE, Gorski JC, Penzak SR, Lee LS. Assessment of oral midazolam limited sampling strategies to predict area under the concentration time curve (AUC) during cytochrome P450 (CYP) 3A baseline, inhibition and induction or activation. Int J Clin Pharmacol Ther 2010; 48: 847-853
  • 14 Fuhr U, Beckmann-Knopp S, Jetter A, Luck H, Mengs U. The effect of silymarin on oral nifedipine pharmacokinetics. Planta Med 2007; 73: 1429-1435
  • 15 Zadoyan G, Rokitta D, Klement S, Dienel A, Hoerr R, Gramatte T, Fuhr U. Effect of Ginkgo biloba special extract EGb 761(R) on human cytochrome P450 activity: a cocktail interaction study in healthy volunteers. Eur J Clin Pharmacol 2012; 68: 553-560
  • 16 Blumenthal M, Ferrier GKL, Cavaliere C. Total sales of herbal supplements in United States show steady growth. Herbal Gram 2006; 71: 64-66
  • 17 Borrelli F, Capasso R, Izzo AA. Garlic (Allium sativum L.): adverse effects and drug interactions in humans. Mol Nutr Food Res 2007; 51: 1386-1397
  • 18 Lanzotti V. The analysis of onion and garlic. J Chromatogr A 2006; 1112: 3-22
  • 19 Shord SS, Shah K, Lukose A. Drug-botanical interactions: a review of the laboratory, animal, and human data for 8 common botanicals. Integr Cancer Ther 2009; 8: 208-227
  • 20 Butterweck V, Derendorf H. Potential of pharmacokinetic profiling for detecting herbal interactions with drugs. Clin Pharmacokinet 2008; 47: 383-397
  • 21 Gallicano K, Foster B, Choudhri S. Effect of short-term administration of garlic supplements on single-dose ritonavir pharmacokinetics in healthy volunteers. Br J Clin Pharmacol 2003; 55: 199-202
  • 22 Hsu A, Granneman GR, Cao G, Carothers L, el-Shourbagy T, Baroldi P, Erdman K, Brown F, Sun E, Leonard JM. Pharmacokinetic interactions between two human immunodeficiency virus protease inhibitors, ritonavir and saquinavir. Clin Pharmacol Ther 1998; 63: 453-464
  • 23 Cox MC, Low J, Lee J, Walshe J, Denduluri N, Berman A, Permenter MG, Petros WP, Price DK, Figg WD, Sparreboom A, Swain SM. Influence of garlic (Allium sativum) on the pharmacokinetics of docetaxel. Clin Cancer Res 2006; 12: 4636-4640
  • 24 Macan H, Uykimpang R, Alconcel M, Takasu J, Razon R, Amagase H, Niihara Y. Aged garlic extract may be safe for patients on warfarin therapy. J Nutr 2006; 136: 793S-795S
  • 25 Markowitz JS, Devane CL, Chavin KD, Taylor RM, Ruan Y, Donovan JL. Effects of garlic (Allium sativum L.) supplementation on cytochrome P450 2D6 and 3A4 activity in healthy volunteers. Clin Pharmacol Ther 2003; 74: 170-177
  • 26 Piscitelli SC, Burstein AH, Welden N, Gallicano KD, Falloon J. The effect of garlic supplements on the pharmacokinetics of saquinavir. Clin Infect Dis 2002; 34: 234-238
  • 27 Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Cui Y, Ang CY. Clinical assessment of effects of botanical supplementation on cytochrome P450 phenotypes in the elderly: St Johnʼs wort, garlic oil, Panax ginseng and Ginkgo biloba . Drugs Aging 2005; 22: 525-539
  • 28 Taylor JA, Weber W, Standish L, Quinn H, Goesling J, McGann M, Calabrese C. Efficacy and safety of echinacea in treating upper respiratory tract infections in children: a randomized controlled trial. JAMA 2003; 290: 2824-2830
  • 29 Freeman C, Spelman K. A critical evaluation of drug interactions with Echinacea spp. Mol Nutr Food Res 2008; 52: 789-798
  • 30 Gertsch J, Schoop R, Kuenzle U, Suter A. Echinacea alkylamides modulate TNF-alpha gene expression via cannabinoid receptor CB2 and multiple signal transduction pathways. FEBS Lett 2004; 577: 563-569
  • 31 Gorski JC, Huang SM, Pinto A, Hamman MA, Hilligoss JK, Zaheer NA, Desai M, Miller M, Hall SD. The effect of echinacea (Echinacea purpurea root) on cytochrome P450 activity in vivo . Clin Pharmacol Ther 2004; 75: 89-100
  • 32 Penzak SR, Robertson SM, Hunt JD, Chairez C, Malati CY, Alfaro RM, Stevenson JM, Kovacs JA. Echinacea purpurea significantly induces cytochrome P450 3A activity but does not alter lopinavir-ritonavir exposure in healthy subjects. Pharmacotherapy 2010; 30: 797-805
  • 33 Wittkowsky AK. Warfarin and other coumarin derivatives: pharmacokinetics, pharmacodynamics, and drug interactions. Semin Vasc Med 2003; 3: 221-230
  • 34 Abdul MI, Jiang X, Williams KM, Day RO, Roufogalis BD, Liauw WS, Xu H, Matthias A, Lehmann RP, McLachlan AJ. Pharmacokinetic and pharmacodynamic interactions of echinacea and policosanol with warfarin in healthy subjects. Br J Clin Pharmacol 2010; 69: 508-515
  • 35 Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Carrier J, Khan IA, Edwards DJ, Shah A. In vivo assessment of botanical supplementation on human cytochrome P450 phenotypes: Citrus aurantium, Echinacea purpurea, milk thistle, and saw palmetto. Clin Pharmacol Ther 2004; 76: 428-440
  • 36 Gurley BJ, Swain A, Hubbard MA, Williams DK, Barone G, Hartsfield F, Tong Y, Carrier DJ, Cheboyina S, Battu SK. Clinical assessment of CYP2D6-mediated herb-drug interactions in humans: effects of milk thistle, black cohosh, goldenseal, kava kava, St. Johnʼs wort, and Echinacea . Mol Nutr Food Res 2008; 52: 755-763
  • 37 Gerber GS. Saw palmetto for the treatment of men with lower urinary tract symptoms. J Urol 2000; 163: 1408-1412
  • 38 Markowitz JS, Donovan JL, Devane CL, Taylor RM, Ruan Y, Wang JS, Chavin KD. Multiple doses of saw palmetto (Serenoa repens) did not alter cytochrome P450 2D6 and 3A4 activity in normal volunteers. Clin Pharmacol Ther 2003; 74: 536-542
  • 39 Diamond BJ, Shiflett SC, Feiwel N, Matheis RJ, Noskin O, Richards JA, Schoenberger NE. Ginkgo biloba extract: mechanisms and clinical indications. Arch Phys Med Rehabil 2000; 81: 668-678
  • 40 Abdel-Kader R, Hauptmann S, Keil U, Scherping I, Leuner K, Eckert A, Muller WE. Stabilization of mitochondrial function by Ginkgo biloba extract (EGb 761). Pharmacol Res 2007; 56: 493-502
  • 41 Tchantchou F, Xu Y, Wu Y, Christen Y, Luo Y. EGb 761 enhances adult hippocampal neurogenesis and phosphorylation of CREB in transgenic mouse model of Alzheimerʼs disease. FASEB J 2007; 21: 2400-2408
  • 42 Tchantchou F, Lacor PN, Cao Z, Lao L, Hou Y, Cui C, Klein WL, Luo Y. Stimulation of neurogenesis and synaptogenesis by bilobalide and quercetin via common final pathway in hippocampal neurons. Int J Alzheimers Dis 2009; 18: 787-798
  • 43 Elovic EP, Zafonte RD. Ginkgo biloba: applications in traumatic brain injury. J Head Trauma Rehabil 2001; 16: 603-607
  • 44 Cohen-Salmon C, Venault P, Martin B, Raffalli-Sebille MJ, Barkats M, Clostre F, Pardon MC, Christen Y, Chapouthier G. Effects of Ginkgo biloba extract (EGb 761) on learning and possible actions on aging. J Physiol 1997; 91: 291-300
  • 45 Thiagarajan G, Chandani S, Harinarayana Rao S, Samuni AM, Chandrasekaran K, Balasubramanian D. Molecular and cellular assessment of Ginkgo biloba extract as a possible ophthalmic drug. Exp Eye Res 2002; 75: 421-430
  • 46 Rioufol G, Pietri S, Culcasi M, Loufoua J, Staat P, Pop C, Drieu K, Ovize M. Ginkgo biloba extract EGb 761 attenuates myocardial stunning in the pig heart. Basic Res Cardiol 2003; 98: 59-68
  • 47 DeFeudis FV. A brief history of EGb 761 and its therapeutic uses. Pharmacopsychiatry 2003; 36: S2-S7
  • 48 Greenblatt DJ, von Moltke LL, Luo Y, Perloff ES, Horan KA, Bruce A, Reynolds RC, Harmatz JS, Avula B, Khan IA, Goldman P. Ginkgo biloba does not alter clearance of flurbiprofen, a cytochrome P450-2C9 substrate. J Clin Pharmacol 2006; 46: 214-221
  • 49 Jiang X, Williams KM, Liauw WS, Ammit AJ, Roufogalis BD, Duke CC, Day RO, McLachlan AJ. Effect of ginkgo and ginger on the pharmacokinetics and pharmacodynamics of warfarin in healthy subjects. Br J Clin Pharmacol 2005; 59: 425-432
  • 50 Yin OQ, Tomlinson B, Waye MM, Chow AH, Chow MS. Pharmacogenetics and herb-drug interactions: experience with Ginkgo biloba and omeprazole. Pharmacogenetics 2004; 14: 841-850
  • 51 Lei HP, Wang G, Wang LS, Ou-yang DS, Chen H, Li Q, Zhang W, Tan ZR, Fan L, He YJ, Zhou HH. Lack of effect of Ginkgo biloba on voriconazole pharmacokinetics in Chinese volunteers identified as CYP2C19 poor and extensive metabolizers. Ann Pharmacother 2009; 43: 726-731
  • 52 Robertson SM, Davey RT, Voell J, Formentini E, Alfaro RM, Penzak SR. Effect of Ginkgo biloba extract on lopinavir, midazolam and fexofenadine pharmacokinetics in healthy subjects. Curr Med Res Opin 2008; 24: 591-599
  • 53 Lei HP, Ji W, Lin J, Chen H, Tan ZR, Hu DL, Liu LJ, Zhou HH. Effects of Ginkgo biloba extract on the pharmacokinetics of bupropion in healthy volunteers. Br J Clin Pharmacol 2009; 68: 201-206
  • 54 Markowitz JS, Donovan JL, Lindsay DeVane C, Sipkes L, Chavin KD. Multiple-dose administration of Ginkgo biloba did not affect cytochrome P-450 2D6 or 3A4 activity in normal volunteers. J Clin Psychopharmacol 2003; 23: 576-581
  • 55 Penzak SR, Busse KH, Robertson SM, Formentini E, Alfaro RM, Davey Jr. RT. Limitations of using a single postdose midazolam concentration to predict CYP3A-mediated drug interactions. J Clin Pharmacol 2008; 48: 671-680
  • 56 Uchida S, Yamada H, Li XD, Maruyama S, Ohmori Y, Oki T, Watanabe H, Umegaki K, Ohashi K, Yamada S. Effects of Ginkgo biloba extract on pharmacokinetics and pharmacodynamics of tolbutamide and midazolam in healthy volunteers. J Clin Pharmacol 2006; 46: 1290-1298
  • 57 Zuo XC, Zhang BK, Jia SJ, Liu SK, Zhou LY, Li J, Zhang J, Dai LL, Chen BM, Yang GP, Yuan H. Effects of Ginkgo biloba extracts on diazepam metabolism: a pharmacokinetic study in healthy Chinese male subjects. Eur J Clin Pharmacol 2010; 66: 503-509
  • 58 Aston JL, Lodolce AE, Shapiro NL. Interaction between warfarin and cranberry juice. Pharmacotherapy 2006; 26: 1314-1319
  • 59 Zuo Y, Wang C, Zhan J. Separation, characterization, and quantitation of benzoic and phenolic antioxidants in American cranberry fruit by GC-MS. J Agric Food Chem 2002; 50: 3789-3794
  • 60 McKay DL, Blumberg JB. Cranberries (Vaccinium macrocarpon) and cardiovascular disease risk factors. Nutr Rev 2007; 65: 490-502
  • 61 Pham DQ, Pham AQ. Interaction potential between cranberry juice and warfarin. Am J Health Syst Pharm 2007; 64: 490-494
  • 62 Griffiths P. The role of cranberry juice in the treatment of urinary tract infections. Br J Community Nurs 2003; 8: 557-561
  • 63 Ngo N, Yan Z, Graf TN, Carrizosa DR, Kashuba AD, Dees EC, Oberlies NH, Paine MF. Identification of a cranberry juice product that inhibits enteric CYP3A-mediated first-pass metabolism in humans. Drug Metab Dispos 2009; 37: 514-522
  • 64 Howell AB, Foxman B. Cranberry juice and adhesion of antibiotic-resistant uropathogens. JAMA 2002; 287: 3082-3083
  • 65 Ushijima K, Tsuruoka S, Tsuda H, Hasegawa G, Obi Y, Kaneda T, Takahashi M, Maekawa T, Sasaki T, Koshimizu TA, Fujimura A. Cranberry juice suppressed the diclofenac metabolism by human liver microsomes, but not in healthy human subjects. Br J Clin Pharmacol 2009; 68: 194-200
  • 66 Greenblatt DJ, von Moltke LL, Perloff ES, Luo Y, Harmatz JS, Zinny MA. Interaction of flurbiprofen with cranberry juice, grape juice, tea, and fluconazole: in vitro and clinical studies. Clin Pharmacol Ther 2006; 79: 125-133
  • 67 Mohammed Abdul MI, Jiang X, Williams KM, Day RO, Roufogalis BD, Liauw WS, Xu H, McLachlan AJ. Pharmacodynamic interaction of warfarin with cranberry but not with garlic in healthy subjects. Br J Pharmacol 2008; 154: 1691-1700
  • 68 Lilja JJ, Backman JT, Neuvonen PJ. Effects of daily ingestion of cranberry juice on the pharmacokinetics of warfarin, tizanidine, and midazolam – probes of CYP2C9, CYP1A2, and CYP3A4. Clin Pharmacol Ther 2007; 81: 833-839
  • 69 Grenier J, Fradette C, Morelli G, Merritt GJ, Vranderick M, Ducharme MP. Pomelo juice, but not cranberry juice, affects the pharmacokinetics of cyclosporine in humans. Clin Pharmacol Ther 2006; 79: 255-262
  • 70 Wu Z, Rodgers RP, Marshall AG. Characterization of vegetable oils: detailed compositional fingerprints derived from electrospray ionization fourier transform ion cyclotron resonance mass spectrometry. J Agric Food Chem 2004; 52: 5322-5328
  • 71 Messina M, Messina V. The role of soy in vegetarian diets. Nutrients 2010; 2: 855-888
  • 72 Kishida T, Nagamoto M, Ohtsu Y, Watakabe M, Ohshima D, Nashiki K, Mizushige T, Izumi T, Obata A, Ebihara K. Lack of an inducible effect of dietary soy isoflavones on the mRNA abundance of hepatic cytochrome P-450 isozymes in rats. Biosci Biotechnol Biochem 2004; 68: 508-515
  • 73 Peng WX, Li HD, Zhou HH. Effect of daidzein on CYP1A2 activity and pharmacokinetics of theophylline in healthy volunteers. Eur J Clin Pharmacol 2003; 59: 237-241
  • 74 Chen Y, Xiao CQ, He YJ, Chen BL, Wang G, Zhou G, Zhang W, Tan ZR, Cao S, Wang LP, Zhou HH. Genistein alters caffeine exposure in healthy female volunteers. Eur J Clin Pharmacol 2011; 67: 347-353
  • 75 Wang G, Xiao CQ, Li Z, Guo D, Chen Y, Fan L, Qian RH, Peng XJ, Hu DL, Zhou HH. Effect of soy extract administration on losartan pharmacokinetics in healthy female volunteers. Ann Pharmacother 2009; 43: 1045-1049
  • 76 Anderson GD, Rosito G, Mohustsy MA, Elmer GW. Drug interaction potential of soy extract and Panax ginseng . J Clin Pharmacol 2003; 43: 643-648
  • 77 Izzo AA, Ernst E. Interactions between herbal medicines and prescribed drugs: an updated systematic review. Drugs 2009; 69: 1777-1798
  • 78 Lee MH, Jeong JH, Seo JW, Shin CG, Kim YS, In JG, Yang DC, Yi JS, Choi YE. Enhanced triterpene and phytosterol biosynthesis in Panax ginseng overexpressing squalene synthase gene. Plant Cell Physiol 2004; 45: 976-984
  • 79 Shibata S. Chemistry and cancer preventing activities of ginseng saponins and some related triterpenoid compounds. J Korean Med Sci 2001; 16: S28-S37
  • 80 Lee SH, Ahn YM, Ahn SY, Doo HK, Lee BC. Interaction between warfarin and Panax ginseng in ischemic stroke patients. J Alternat Complement Med 2008; 14: 715-721
  • 81 Briskin DP. Medicinal plants and phytomedicines. Linking plant biochemistry and physiology to human health. Plant Physiol 2000; 124: 507-514
  • 82 Vogler BK, Pittler MH, Ernst E. The efficacy of ginseng. A systematic review of randomised clinical trials. Eur J Clin Pharmacol 1999; 55: 567-575
  • 83 Jiang X, Williams KM, Liauw WS, Ammit AJ, Roufogalis BD, Duke CC, Day RO, McLachlan AJ. Effect of St Johnʼs wort and ginseng on the pharmacokinetics and pharmacodynamics of warfarin in healthy subjects. Br J Clin Pharmacol 2004; 57: 592-599
  • 84 Rhyu MR, Lu J, Webster DE, Fabricant DS, Farnsworth NR, Wang ZJ. Black cohosh (Actaea racemosa, Cimicifuga racemosa) behaves as a mixed competitive ligand and partial agonist at the human mu opiate receptor. J Agric Food Chem 2006; 54: 9852-9857
  • 85 Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Khan IA, Shah A. In vivo effects of goldenseal, kava kava, black cohosh, and valerian on human cytochrome P450 1A2, 2D6, 2E1, and 3A4/5 phenotypes. Clin Pharmacol Ther 2005; 77: 415-426
  • 86 Gurley B, Hubbard MA, Williams DK, Thaden J, Tong Y, Gentry WB, Breen P, Carrier DJ, Cheboyina S. Assessing the clinical significance of botanical supplementation on human cytochrome P450 3A activity: comparison of a milk thistle and black cohosh product to rifampin and clarithromycin. J Clin Pharmacol 2006; 46: 201-213
  • 87 Borrelli F, Izzo AA. Herb-drug interactions with St Johnʼs wort (Hypericum perforatum): an update on clinical observations. AAPS J 2009; 11: 710-727
  • 88 Markowitz JS, DeVane CL, Boulton DW, Carson SW, Nahas Z, Risch SC. Effect of St. Johnʼs wort (Hypericum perforatum) on cytochrome P-450 2D6 and 3A4 activity in healthy volunteers. Life Sci 2000; 66: PL133-PL139
  • 89 Wang Z, Gorski JC, Hamman MA, Huang SM, Lesko LJ, Hall SD. The effects of St Johnʼs wort (Hypericum perforatum) on human cytochrome P450 activity. Clin Pharmacol Ther 2001; 70: 317-326
  • 90 Wenk M, Todesco L, Krahenbuhl S. Effect of St Johnʼs wort on the activities of CYP1A2, CYP3A4, CYP2D6, N-acetyltransferase 2, and xanthine oxidase in healthy males and females. Br J Clin Pharmacol 2004; 57: 495-499
  • 91 Roby CA, Anderson GD, Kantor E, Dryer DA, Burstein AH. St Johnʼs Wort: effect on CYP3A4 activity. Clin Pharmacol Ther 2000; 67: 451-457
  • 92 Markowitz JS, Donovan JL, DeVane CL, Taylor RM, Ruan Y, Wang JS, Chavin KD. Effect of St Johnʼs wort on drug metabolism by induction of cytochrome P450 3A4 enzyme. JAMA 2003; 290: 1500-1504
  • 93 Wang LS, Zhu B, Abd El-Aty AM, Zhou G, Li Z, Wu J, Chen GL, Liu J, Tang ZR, An W, Li Q, Wang D, Zhou HH. The influence of St Johnʼs Wort on CYP2C19 activity with respect to genotype. J Clin Pharmacol 2004; 44: 577-581
  • 94 Wang LS, Zhou G, Zhu B, Wu J, Wang JG, Abd El-Aty AM, Li T, Liu J, Yang TL, Wang D, Zhong XY, Zhou HH. St Johnʼs wort induces both cytochrome P450 3A4-catalyzed sulfoxidation and 2C19-dependent hydroxylation of omeprazole. Clin Pharmacol Ther 2004; 75: 191-197
  • 95 Mai I, Bauer S, Perloff ES, Johne A, Uehleke B, Frank B, Budde K, Roots I. Hyperforin content determines the magnitude of the St Johnʼs wort-cyclosporine drug interaction. Clin Pharmacol Ther 2004; 76: 330-340
  • 96 Arold G, Donath F, Maurer A, Diefenbach K, Bauer S, Henneicke-von Zepelin HH, Friede M, Roots I. No relevant interaction with alprazolam, caffeine, tolbutamide, and digoxin by treatment with a low-hyperforin St Johnʼs wort extract. Planta Med 2005; 71: 331-337
  • 97 Mueller SC, Majcher-Peszynska J, Uehleke B, Klammt S, Mundkowski RG, Miekisch W, Sievers H, Bauer S, Frank B, Kundt G, Drewelow B. The extent of induction of CYP3A by St. Johnʼs wort varies among products and is linked to hyperforin dose. Eur J Clin Pharmacol 2006; 62: 29-36
  • 98 Mueller SC, Majcher-Peszynska J, Mundkowski RG, Uehleke B, Klammt S, Sievers H, Lehnfeld R, Frank B, Thurow K, Kundt G, Drewelow B. No clinically relevant CYP3A induction after St. Johnʼs wort with low hyperforin content in healthy volunteers. Eur J Clin Pharmacol 2009; 65: 81-87
  • 99 Hall SD, Wang Z, Huang SM, Hamman MA, Vasavada N, Adigun AQ, Hilligoss JK, Miller M, Gorski JC. The interaction between St Johnʼs wort and an oral contraceptive. Clin Pharmacol Ther 2003; 74: 525-535
  • 100 Murphy PA, Kern SE, Stanczyk FZ, Westhoff CL. Interaction of St. Johnʼs Wort with oral contraceptives: effects on the pharmacokinetics of norethindrone and ethinyl estradiol, ovarian activity and breakthrough bleeding. Contraception 2005; 71: 402-408
  • 101 Pfrunder A, Schiesser M, Gerber S, Haschke M, Bitzer J, Drewe J. Interaction of St Johnʼs wort with low-dose oral contraceptive therapy: a randomized controlled trial. Br J Clin Pharmacol 2003; 56: 683-690
  • 102 Smith PF, Bullock JM, Booker BM, Haas CE, Berenson CS, Jusko WJ. Induction of imatinib metabolism by Hypericum perforatum . Blood 2004; 104: 1229-1230
  • 103 Frye RF, Fitzgerald SM, Lagattuta TF, Hruska MW, Egorin MJ. Effect of St Johnʼs wort on imatinib mesylate pharmacokinetics. Clin Pharmacol Ther 2004; 76: 323-329
  • 104 Andren L, Andreasson A, Eggertsen R. Interaction between a commercially available St. Johnʼs wort product (Movina) and atorvastatin in patients with hypercholesterolemia. Eur J Clin Pharmacol 2007; 63: 913-916
  • 105 Portoles A, Terleira A, Calvo A, Martinez I, Resplandy G. Effects of Hypericum perforatum on ivabradine pharmacokinetics in healthy volunteers: an open-label, pharmacokinetic interaction clinical trial. J Clin Pharmacol 2006; 46: 1188-1194
  • 106 Tannergren C, Engman H, Knutson L, Hedeland M, Bondesson U, Lennernas H. St Johnʼs wort decreases the bioavailability of R- and S-verapamil through induction of the first-pass metabolism. Clin Pharmacol Ther 2004; 75: 298-309
  • 107 Kawaguchi A, Ohmori M, Tsuruoka S, Nishiki K, Harada K, Miyamori I, Yano R, Nakamura T, Masada M, Fujimura A. Drug interaction between St Johnʼs Wort and quazepam. Br J Clin Pharmacol 2004; 58: 403-410
  • 108 Rengelshausen J, Banfield M, Riedel KD, Burhenne J, Weiss J, Thomsen T, Walter-Sack I, Haefeli WE, Mikus G. Opposite effects of short-term and long-term St Johnʼs wort intake on voriconazole pharmacokinetics. Clin Pharmacol Ther 2005; 78: 25-33
  • 109 Xu H, Williams KM, Liauw WS, Murray M, Day RO, McLachlan AJ. Effects of St Johnʼs wort and CYP2C9 genotype on the pharmacokinetics and pharmacodynamics of gliclazide. Br J Pharmacol 2008; 153: 1579-1586
  • 110 Piscitelli SC, Burstein AH, Chaitt D, Alfaro RM, Falloon J. Indinavir concentrations and St Johnʼs wort. Lancet 2000; 355: 547-548
  • 111 Kroll DJ, Shaw HS, Oberlies NH. Milk thistle nomenclature: why it matters in cancer research and pharmacokinetic studies. Integr Cancer Ther 2007; 6: 110-119
  • 112 Hogan FS, Krishnegowda NK, Mikhailova M, Kahlenberg MS. Flavonoid, silibinin, inhibits proliferation and promotes cell-cycle arrest of human colon cancer. J Surg Res 2007; 143: 58-65
  • 113 Brantley SJ, Oberlies NH, Kroll DJ, Paine MF. Two flavonolignans from milk thistle (Silybum marianum) inhibit CYP2C9-mediated warfarin metabolism at clinically achievable concentrations. J Pharmacol Exp Ther 2010; 332: 1081-1087
  • 114 van Erp NP, Baker SD, Zhao M, Rudek MA, Guchelaar HJ, Nortier JW, Sparreboom A, Gelderblom H. Effect of milk thistle (Silybum marianum) on the pharmacokinetics of irinotecan. Clin Cancer Res 2005; 11: 7800-7806
  • 115 Lu H, Meng X, Li C, Sang S, Patten C, Sheng S, Hong J, Bai N, Winnik B, Ho CT, Yang CS. Glucuronides of tea catechins: enzymology of biosynthesis and biological activities. Drug Metab Dispos 2003; 31: 452-461
  • 116 Feng WY. Metabolism of green tea catechins: an overview. Curr Drug Metab 2006; 7: 755-809
  • 117 Zhang Q, Fu H, Pan J, He J, Ryota S, Hara Y, Wang Y, Lubet RA, You M. Effect of dietary Polyphenon E and EGCG on lung tumorigenesis in A/J Mice. Pharm Res 2010; 27: 1066-1071
  • 118 Yang CS, Chen L, Lee MJ, Balentine D, Kuo MC, Schantz SP. Blood and urine levels of tea catechins after ingestion of different amounts of green tea by human volunteers. Cancer Epidemiol Biomarkers Prev 1998; 7: 351-354
  • 119 Yang TT, Koo MW. Chinese green tea lowers cholesterol level through an increase in fecal lipid excretion. Life Sci 2000; 66: 411-423
  • 120 Yang TT, Koo MW. Inhibitory effect of Chinese green tea on endothelial cell-induced LDL oxidation. Atherosclerosis 2000; 148: 67-73
  • 121 Zhong Z, Froh M, Connor HD, Li X, Conzelmann LO, Mason RP, Lemasters JJ, Thurman RG. Prevention of hepatic ischemia-reperfusion injury by green tea extract. Am J Physiol Gastrointest Liver Physiol 2002; 283: G957-G964
  • 122 Oyama J, Maeda T, Kouzuma K, Ochiai R, Tokimitsu I, Higuchi Y, Sugano M, Makino N. Green tea catechins improve human forearm endothelial dysfunction and have antiatherosclerotic effects in smokers. Circ J 2010; 74: 578-588
  • 123 Oyama J, Maeda T, Sasaki M, Kozuma K, Ochiai R, Tokimitsu I, Taguchi S, Higuchi Y, Makino N. Green tea catechins improve human forearm vascular function and have potent anti-inflammatory and anti-apoptotic effects in smokers. Intern Med 2010; 49: 2553-2559
  • 124 Persson IA, Persson K, Hagg S, Andersson RG. Effects of green tea, black tea and Rooibos tea on angiotensin-converting enzyme and nitric oxide in healthy volunteers. Public Health Nutr 2010; 13: 730-737
  • 125 Nantz MP, Rowe CA, Bukowski JF, Percival SS. Standardized capsule of Camellia sinensis lowers cardiovascular risk factors in a randomized, double-blind, placebo-controlled study. Nutrition 2009; 25: 147-154
  • 126 Donovan JL, Chavin KD, Devane CL, Taylor RM, Wang JS, Ruan Y, Markowitz JS. Green tea (Camellia sinensis) extract does not alter cytochrome p 450 3A4 or 2D6 activity in healthy volunteers. Drug Metab Dispos 2004; 32: 906-908
  • 127 Chow HH, Hakim IA, Vining DR, Crowell JA, Cordova CA, Chew WM, Xu MJ, Hsu CH, Ranger-Moore J, Alberts DS. Effects of repeated green tea catechin administration on human cytochrome P450 activity. Cancer Epidemiol Biomarkers Prev 2006; 15: 2473-2476
  • 128 Kennedy DO, Wightman EL. Herbal extracts and phytochemicals: plant secondary metabolites and the enhancement of human brain function. Adv Nutr 2011; 2: 32-50
  • 129 Houghton PJ. The scientific basis for the reputed activity of Valerian. J Pharm Pharmacol 1999; 51: 505-512
  • 130 Houghton PJ. The biological activity of Valerian and related plants. J Ethnopharmacol 1988; 22: 121-142
  • 131 Leathwood PD, Chauffard F, Heck E, Munoz-Box R. Aqueous extract of valerian root (Valeriana officinalis L.) improves sleep quality in man. Pharmacol Biochem Behav 1982; 17: 65-71
  • 132 Leathwood PD, Chauffard F. Aqueous extract of valerian reduces latency to fall asleep in man. Planta Med 1985; 144-148
  • 133 Dorn M. [Efficacy and tolerability of Baldrian versus oxazepam in non-organic and non-psychiatric insomniacs: a randomised, double-blind, clinical, comparative study]. Forsch Komplementarmed Klass Naturheilkd 2000; 7: 79-84
  • 134 Donath F, Quispe S, Diefenbach K, Maurer A, Fietze I, Roots I. Critical evaluation of the effect of valerian extract on sleep structure and sleep quality. Pharmacopsychiatry 2000; 33: 47-53
  • 135 Donovan JL, DeVane CL, Chavin KD, Wang JS, Gibson BB, Gefroh HA, Markowitz JS. Multiple night-time doses of valerian (Valeriana officinalis) had minimal effects on CYP3A4 activity and no effect on CYP2D6 activity in healthy volunteers. Drug Metab Dispos 2004; 32: 1333-1336
  • 136 Anke J, Ramzan I. Pharmacokinetic and pharmacodynamic drug interactions with Kava (Piper methysticum Forst. f.). J Ethnopharmacol 2004; 93: 153-160
  • 137 Thompson R, Ruch W, Hasenohrl RU. Enhanced cognitive performance and cheerful mood by standardized extracts of Piper methysticum (Kava-kava). Hum Psychopharmacol 2004; 19: 243-250
  • 138 Kennedy DA, Seely D. Clinically based evidence of drug-herb interactions: a systematic review. Expert Opin Drug Saf 2010; 9: 79-124
  • 139 Gurley BJ, Swain A, Hubbard MA, Hartsfield F, Thaden J, Williams DK, Gentry WB, Tong Y. Supplementation with goldenseal (Hydrastis canadensis), but not kava kava (Piper methysticum), inhibits human CYP3A activity in vivo . Clin Pharmacol Ther 2008; 83: 61-69
  • 140 Brown PN, Roman MC. Determination of hydrastine and berberine in goldenseal raw materials, extracts, and dietary supplements by high-performance liquid chromatography with UV: collaborative study. J AOAC Int 2008; 91: 694-701
  • 141 Lancaster HM, Davidson AL. Commercial pharmaceutical preparations: 4. Hydrastis – golden seal. Can Med Assoc J 1927; 17: 1317-1320
  • 142 Chatterjee P, Franklin MR. Human cytochrome p 450 inhibition and metabolic-intermediate complex formation by goldenseal extract and its methylenedioxyphenyl components. Drug Metab Dispos 2003; 31: 1391-1397
  • 143 Yi S, Cho JY, Lim KS, Kim KP, Kim J, Kim BH, Hong JH, Jang IJ, Shin SG, Yu KS. Effects of Angelicae tenuissima radix, Angelicae dahuricae radix and Scutellariae radix extracts on cytochrome P450 activities in healthy volunteers. Basic Clin Pharmacol Toxicol 2009; in press
  • 144 Fan L, Wang JC, Jiang F, Tan ZR, Chen Y, Li Q, Zhang W, Wang G, Lei HP, Hu DL, Wang D, Zhou HH. Induction of cytochrome P450 2B6 activity by the herbal medicine baicalin as measured by bupropion hydroxylation. Eur J Clin Pharmacol 2009; 65: 403-409
  • 145 Ramassamy C. Emerging role of polyphenolic compounds in the treatment of neurodegenerative diseases: a review of their intracellular targets. Eur J Pharmacol 2006; 545: 51-64
  • 146 Chow HH, Garland LL, Hsu CH, Vining DR, Chew WM, Miller JA, Perloff M, Crowell JA, Alberts DS. Resveratrol modulates drug- and carcinogen-metabolizing enzymes in a healthy volunteer study. Cancer Prev Res (Phila) 2010; 3: 1168-1175
  • 147 Chen Y, Liu WH, Chen BL, Fan L, Han Y, Wang G, Hu DL, Tan ZR, Zhou G, Cao S, Zhou HH. Plant polyphenol curcumin significantly affects CYP1A2 and CYP2A6 activity in healthy, male Chinese volunteers. Ann Pharmacother 2010; 44: 1038-1045

Correspondence

Prof. Dr. med. Uwe Fuhr
Department of Pharmacology, Clinical Pharmacology Unit, University of Cologne
Gleueler Str. 24
50931 Cologne
Germany
Telefon: +49 22 14 78 52 30   
Fax: +49 22 14 78 70 11   

  • References

  • 1 Moore LB, Goodwin B, Jones SA, Wisely GB, Serabjit-Singh CJ, Willson TM, Collins JL, Kliewer SA. St. Johnʼs wort induces hepatic drug metabolism through activation of the pregnane X receptor. Proc Natl Acad Sci USA 2000; 97: 7500-7502
  • 2 Bailey DG, Spence JD, Munoz C, Arnold JM. Interaction of citrus juices with felodipine and nifedipine. Lancet 1991; 337: 268-269
  • 3 Fuhr U, Jetter A, Kirchheiner J. Appropriate phenotyping procedures for drug metabolizing enzymes and transporters in humans and their simultaneous use in the “cocktail” approach. Clin Pharmacol Ther 2007; 81: 270-283
  • 4 Huang SM, Zhang L. Guidance for industry: drug interaction studies – study design, data analysis, implications for dosing, and labeling recommendations. Available at http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM072101.htm Accessed December 02, 2011
  • 5 European Medicines Agency. Guideline on the investigation of drug interactions. Available at http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2010/05/WC500090112.pdf Accessed December 02, 2011
  • 6 US Food and Drug Administration. Drug development and drug interactions: table of substrates, inhibitors and inducers. Available at http://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/ucm093664.htm Accessed December 02, 2011
  • 7 Tanaka E, Kurata N, Yasuhara H. How useful is the “cocktail approach” for evaluating human hepatic drug metabolizing capacity using cytochrome P450 phenotyping probes in vivo?. J Clin Pharm Ther 2003; 28: 157-165
  • 8 Zhou H, Tong Z, McLeod JF. “Cocktail” approaches and strategies in drug development: valuable tool or flawed science?. J Clin Pharmacol 2004; 44: 120-134
  • 9 Turpault S, Brian W, Van Horn R, Santoni A, Poitiers F, Donazzolo Y, Boulenc X. Pharmacokinetic assessment of a five-probe cocktail for CYPs 1A2, 2C9, 2C19, 2D6 and 3A. Br J Clin Pharmacol 2009; 68: 928-935
  • 10 Zhang L. A regulatory perspective on the utilization of cocktail approach in assessing drug interactions. Available at http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/UCM237160.htm Accessed December 02, 2011
  • 11 Zhang L, Reynolds KS, Zhao P, Huang SM. Drug interactions evaluation: an integrated part of risk assessment of therapeutics. Toxicol Appl Pharmacol 2010; 243: 134-145
  • 12 US Food and Drug Administration. Guidances (Drugs). Available at http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm Accessed December 02, 2011
  • 13 Ma JD, Nguyen ET, Tsunoda SM, Greenberg HE, Gorski JC, Penzak SR, Lee LS. Assessment of oral midazolam limited sampling strategies to predict area under the concentration time curve (AUC) during cytochrome P450 (CYP) 3A baseline, inhibition and induction or activation. Int J Clin Pharmacol Ther 2010; 48: 847-853
  • 14 Fuhr U, Beckmann-Knopp S, Jetter A, Luck H, Mengs U. The effect of silymarin on oral nifedipine pharmacokinetics. Planta Med 2007; 73: 1429-1435
  • 15 Zadoyan G, Rokitta D, Klement S, Dienel A, Hoerr R, Gramatte T, Fuhr U. Effect of Ginkgo biloba special extract EGb 761(R) on human cytochrome P450 activity: a cocktail interaction study in healthy volunteers. Eur J Clin Pharmacol 2012; 68: 553-560
  • 16 Blumenthal M, Ferrier GKL, Cavaliere C. Total sales of herbal supplements in United States show steady growth. Herbal Gram 2006; 71: 64-66
  • 17 Borrelli F, Capasso R, Izzo AA. Garlic (Allium sativum L.): adverse effects and drug interactions in humans. Mol Nutr Food Res 2007; 51: 1386-1397
  • 18 Lanzotti V. The analysis of onion and garlic. J Chromatogr A 2006; 1112: 3-22
  • 19 Shord SS, Shah K, Lukose A. Drug-botanical interactions: a review of the laboratory, animal, and human data for 8 common botanicals. Integr Cancer Ther 2009; 8: 208-227
  • 20 Butterweck V, Derendorf H. Potential of pharmacokinetic profiling for detecting herbal interactions with drugs. Clin Pharmacokinet 2008; 47: 383-397
  • 21 Gallicano K, Foster B, Choudhri S. Effect of short-term administration of garlic supplements on single-dose ritonavir pharmacokinetics in healthy volunteers. Br J Clin Pharmacol 2003; 55: 199-202
  • 22 Hsu A, Granneman GR, Cao G, Carothers L, el-Shourbagy T, Baroldi P, Erdman K, Brown F, Sun E, Leonard JM. Pharmacokinetic interactions between two human immunodeficiency virus protease inhibitors, ritonavir and saquinavir. Clin Pharmacol Ther 1998; 63: 453-464
  • 23 Cox MC, Low J, Lee J, Walshe J, Denduluri N, Berman A, Permenter MG, Petros WP, Price DK, Figg WD, Sparreboom A, Swain SM. Influence of garlic (Allium sativum) on the pharmacokinetics of docetaxel. Clin Cancer Res 2006; 12: 4636-4640
  • 24 Macan H, Uykimpang R, Alconcel M, Takasu J, Razon R, Amagase H, Niihara Y. Aged garlic extract may be safe for patients on warfarin therapy. J Nutr 2006; 136: 793S-795S
  • 25 Markowitz JS, Devane CL, Chavin KD, Taylor RM, Ruan Y, Donovan JL. Effects of garlic (Allium sativum L.) supplementation on cytochrome P450 2D6 and 3A4 activity in healthy volunteers. Clin Pharmacol Ther 2003; 74: 170-177
  • 26 Piscitelli SC, Burstein AH, Welden N, Gallicano KD, Falloon J. The effect of garlic supplements on the pharmacokinetics of saquinavir. Clin Infect Dis 2002; 34: 234-238
  • 27 Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Cui Y, Ang CY. Clinical assessment of effects of botanical supplementation on cytochrome P450 phenotypes in the elderly: St Johnʼs wort, garlic oil, Panax ginseng and Ginkgo biloba . Drugs Aging 2005; 22: 525-539
  • 28 Taylor JA, Weber W, Standish L, Quinn H, Goesling J, McGann M, Calabrese C. Efficacy and safety of echinacea in treating upper respiratory tract infections in children: a randomized controlled trial. JAMA 2003; 290: 2824-2830
  • 29 Freeman C, Spelman K. A critical evaluation of drug interactions with Echinacea spp. Mol Nutr Food Res 2008; 52: 789-798
  • 30 Gertsch J, Schoop R, Kuenzle U, Suter A. Echinacea alkylamides modulate TNF-alpha gene expression via cannabinoid receptor CB2 and multiple signal transduction pathways. FEBS Lett 2004; 577: 563-569
  • 31 Gorski JC, Huang SM, Pinto A, Hamman MA, Hilligoss JK, Zaheer NA, Desai M, Miller M, Hall SD. The effect of echinacea (Echinacea purpurea root) on cytochrome P450 activity in vivo . Clin Pharmacol Ther 2004; 75: 89-100
  • 32 Penzak SR, Robertson SM, Hunt JD, Chairez C, Malati CY, Alfaro RM, Stevenson JM, Kovacs JA. Echinacea purpurea significantly induces cytochrome P450 3A activity but does not alter lopinavir-ritonavir exposure in healthy subjects. Pharmacotherapy 2010; 30: 797-805
  • 33 Wittkowsky AK. Warfarin and other coumarin derivatives: pharmacokinetics, pharmacodynamics, and drug interactions. Semin Vasc Med 2003; 3: 221-230
  • 34 Abdul MI, Jiang X, Williams KM, Day RO, Roufogalis BD, Liauw WS, Xu H, Matthias A, Lehmann RP, McLachlan AJ. Pharmacokinetic and pharmacodynamic interactions of echinacea and policosanol with warfarin in healthy subjects. Br J Clin Pharmacol 2010; 69: 508-515
  • 35 Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Carrier J, Khan IA, Edwards DJ, Shah A. In vivo assessment of botanical supplementation on human cytochrome P450 phenotypes: Citrus aurantium, Echinacea purpurea, milk thistle, and saw palmetto. Clin Pharmacol Ther 2004; 76: 428-440
  • 36 Gurley BJ, Swain A, Hubbard MA, Williams DK, Barone G, Hartsfield F, Tong Y, Carrier DJ, Cheboyina S, Battu SK. Clinical assessment of CYP2D6-mediated herb-drug interactions in humans: effects of milk thistle, black cohosh, goldenseal, kava kava, St. Johnʼs wort, and Echinacea . Mol Nutr Food Res 2008; 52: 755-763
  • 37 Gerber GS. Saw palmetto for the treatment of men with lower urinary tract symptoms. J Urol 2000; 163: 1408-1412
  • 38 Markowitz JS, Donovan JL, Devane CL, Taylor RM, Ruan Y, Wang JS, Chavin KD. Multiple doses of saw palmetto (Serenoa repens) did not alter cytochrome P450 2D6 and 3A4 activity in normal volunteers. Clin Pharmacol Ther 2003; 74: 536-542
  • 39 Diamond BJ, Shiflett SC, Feiwel N, Matheis RJ, Noskin O, Richards JA, Schoenberger NE. Ginkgo biloba extract: mechanisms and clinical indications. Arch Phys Med Rehabil 2000; 81: 668-678
  • 40 Abdel-Kader R, Hauptmann S, Keil U, Scherping I, Leuner K, Eckert A, Muller WE. Stabilization of mitochondrial function by Ginkgo biloba extract (EGb 761). Pharmacol Res 2007; 56: 493-502
  • 41 Tchantchou F, Xu Y, Wu Y, Christen Y, Luo Y. EGb 761 enhances adult hippocampal neurogenesis and phosphorylation of CREB in transgenic mouse model of Alzheimerʼs disease. FASEB J 2007; 21: 2400-2408
  • 42 Tchantchou F, Lacor PN, Cao Z, Lao L, Hou Y, Cui C, Klein WL, Luo Y. Stimulation of neurogenesis and synaptogenesis by bilobalide and quercetin via common final pathway in hippocampal neurons. Int J Alzheimers Dis 2009; 18: 787-798
  • 43 Elovic EP, Zafonte RD. Ginkgo biloba: applications in traumatic brain injury. J Head Trauma Rehabil 2001; 16: 603-607
  • 44 Cohen-Salmon C, Venault P, Martin B, Raffalli-Sebille MJ, Barkats M, Clostre F, Pardon MC, Christen Y, Chapouthier G. Effects of Ginkgo biloba extract (EGb 761) on learning and possible actions on aging. J Physiol 1997; 91: 291-300
  • 45 Thiagarajan G, Chandani S, Harinarayana Rao S, Samuni AM, Chandrasekaran K, Balasubramanian D. Molecular and cellular assessment of Ginkgo biloba extract as a possible ophthalmic drug. Exp Eye Res 2002; 75: 421-430
  • 46 Rioufol G, Pietri S, Culcasi M, Loufoua J, Staat P, Pop C, Drieu K, Ovize M. Ginkgo biloba extract EGb 761 attenuates myocardial stunning in the pig heart. Basic Res Cardiol 2003; 98: 59-68
  • 47 DeFeudis FV. A brief history of EGb 761 and its therapeutic uses. Pharmacopsychiatry 2003; 36: S2-S7
  • 48 Greenblatt DJ, von Moltke LL, Luo Y, Perloff ES, Horan KA, Bruce A, Reynolds RC, Harmatz JS, Avula B, Khan IA, Goldman P. Ginkgo biloba does not alter clearance of flurbiprofen, a cytochrome P450-2C9 substrate. J Clin Pharmacol 2006; 46: 214-221
  • 49 Jiang X, Williams KM, Liauw WS, Ammit AJ, Roufogalis BD, Duke CC, Day RO, McLachlan AJ. Effect of ginkgo and ginger on the pharmacokinetics and pharmacodynamics of warfarin in healthy subjects. Br J Clin Pharmacol 2005; 59: 425-432
  • 50 Yin OQ, Tomlinson B, Waye MM, Chow AH, Chow MS. Pharmacogenetics and herb-drug interactions: experience with Ginkgo biloba and omeprazole. Pharmacogenetics 2004; 14: 841-850
  • 51 Lei HP, Wang G, Wang LS, Ou-yang DS, Chen H, Li Q, Zhang W, Tan ZR, Fan L, He YJ, Zhou HH. Lack of effect of Ginkgo biloba on voriconazole pharmacokinetics in Chinese volunteers identified as CYP2C19 poor and extensive metabolizers. Ann Pharmacother 2009; 43: 726-731
  • 52 Robertson SM, Davey RT, Voell J, Formentini E, Alfaro RM, Penzak SR. Effect of Ginkgo biloba extract on lopinavir, midazolam and fexofenadine pharmacokinetics in healthy subjects. Curr Med Res Opin 2008; 24: 591-599
  • 53 Lei HP, Ji W, Lin J, Chen H, Tan ZR, Hu DL, Liu LJ, Zhou HH. Effects of Ginkgo biloba extract on the pharmacokinetics of bupropion in healthy volunteers. Br J Clin Pharmacol 2009; 68: 201-206
  • 54 Markowitz JS, Donovan JL, Lindsay DeVane C, Sipkes L, Chavin KD. Multiple-dose administration of Ginkgo biloba did not affect cytochrome P-450 2D6 or 3A4 activity in normal volunteers. J Clin Psychopharmacol 2003; 23: 576-581
  • 55 Penzak SR, Busse KH, Robertson SM, Formentini E, Alfaro RM, Davey Jr. RT. Limitations of using a single postdose midazolam concentration to predict CYP3A-mediated drug interactions. J Clin Pharmacol 2008; 48: 671-680
  • 56 Uchida S, Yamada H, Li XD, Maruyama S, Ohmori Y, Oki T, Watanabe H, Umegaki K, Ohashi K, Yamada S. Effects of Ginkgo biloba extract on pharmacokinetics and pharmacodynamics of tolbutamide and midazolam in healthy volunteers. J Clin Pharmacol 2006; 46: 1290-1298
  • 57 Zuo XC, Zhang BK, Jia SJ, Liu SK, Zhou LY, Li J, Zhang J, Dai LL, Chen BM, Yang GP, Yuan H. Effects of Ginkgo biloba extracts on diazepam metabolism: a pharmacokinetic study in healthy Chinese male subjects. Eur J Clin Pharmacol 2010; 66: 503-509
  • 58 Aston JL, Lodolce AE, Shapiro NL. Interaction between warfarin and cranberry juice. Pharmacotherapy 2006; 26: 1314-1319
  • 59 Zuo Y, Wang C, Zhan J. Separation, characterization, and quantitation of benzoic and phenolic antioxidants in American cranberry fruit by GC-MS. J Agric Food Chem 2002; 50: 3789-3794
  • 60 McKay DL, Blumberg JB. Cranberries (Vaccinium macrocarpon) and cardiovascular disease risk factors. Nutr Rev 2007; 65: 490-502
  • 61 Pham DQ, Pham AQ. Interaction potential between cranberry juice and warfarin. Am J Health Syst Pharm 2007; 64: 490-494
  • 62 Griffiths P. The role of cranberry juice in the treatment of urinary tract infections. Br J Community Nurs 2003; 8: 557-561
  • 63 Ngo N, Yan Z, Graf TN, Carrizosa DR, Kashuba AD, Dees EC, Oberlies NH, Paine MF. Identification of a cranberry juice product that inhibits enteric CYP3A-mediated first-pass metabolism in humans. Drug Metab Dispos 2009; 37: 514-522
  • 64 Howell AB, Foxman B. Cranberry juice and adhesion of antibiotic-resistant uropathogens. JAMA 2002; 287: 3082-3083
  • 65 Ushijima K, Tsuruoka S, Tsuda H, Hasegawa G, Obi Y, Kaneda T, Takahashi M, Maekawa T, Sasaki T, Koshimizu TA, Fujimura A. Cranberry juice suppressed the diclofenac metabolism by human liver microsomes, but not in healthy human subjects. Br J Clin Pharmacol 2009; 68: 194-200
  • 66 Greenblatt DJ, von Moltke LL, Perloff ES, Luo Y, Harmatz JS, Zinny MA. Interaction of flurbiprofen with cranberry juice, grape juice, tea, and fluconazole: in vitro and clinical studies. Clin Pharmacol Ther 2006; 79: 125-133
  • 67 Mohammed Abdul MI, Jiang X, Williams KM, Day RO, Roufogalis BD, Liauw WS, Xu H, McLachlan AJ. Pharmacodynamic interaction of warfarin with cranberry but not with garlic in healthy subjects. Br J Pharmacol 2008; 154: 1691-1700
  • 68 Lilja JJ, Backman JT, Neuvonen PJ. Effects of daily ingestion of cranberry juice on the pharmacokinetics of warfarin, tizanidine, and midazolam – probes of CYP2C9, CYP1A2, and CYP3A4. Clin Pharmacol Ther 2007; 81: 833-839
  • 69 Grenier J, Fradette C, Morelli G, Merritt GJ, Vranderick M, Ducharme MP. Pomelo juice, but not cranberry juice, affects the pharmacokinetics of cyclosporine in humans. Clin Pharmacol Ther 2006; 79: 255-262
  • 70 Wu Z, Rodgers RP, Marshall AG. Characterization of vegetable oils: detailed compositional fingerprints derived from electrospray ionization fourier transform ion cyclotron resonance mass spectrometry. J Agric Food Chem 2004; 52: 5322-5328
  • 71 Messina M, Messina V. The role of soy in vegetarian diets. Nutrients 2010; 2: 855-888
  • 72 Kishida T, Nagamoto M, Ohtsu Y, Watakabe M, Ohshima D, Nashiki K, Mizushige T, Izumi T, Obata A, Ebihara K. Lack of an inducible effect of dietary soy isoflavones on the mRNA abundance of hepatic cytochrome P-450 isozymes in rats. Biosci Biotechnol Biochem 2004; 68: 508-515
  • 73 Peng WX, Li HD, Zhou HH. Effect of daidzein on CYP1A2 activity and pharmacokinetics of theophylline in healthy volunteers. Eur J Clin Pharmacol 2003; 59: 237-241
  • 74 Chen Y, Xiao CQ, He YJ, Chen BL, Wang G, Zhou G, Zhang W, Tan ZR, Cao S, Wang LP, Zhou HH. Genistein alters caffeine exposure in healthy female volunteers. Eur J Clin Pharmacol 2011; 67: 347-353
  • 75 Wang G, Xiao CQ, Li Z, Guo D, Chen Y, Fan L, Qian RH, Peng XJ, Hu DL, Zhou HH. Effect of soy extract administration on losartan pharmacokinetics in healthy female volunteers. Ann Pharmacother 2009; 43: 1045-1049
  • 76 Anderson GD, Rosito G, Mohustsy MA, Elmer GW. Drug interaction potential of soy extract and Panax ginseng . J Clin Pharmacol 2003; 43: 643-648
  • 77 Izzo AA, Ernst E. Interactions between herbal medicines and prescribed drugs: an updated systematic review. Drugs 2009; 69: 1777-1798
  • 78 Lee MH, Jeong JH, Seo JW, Shin CG, Kim YS, In JG, Yang DC, Yi JS, Choi YE. Enhanced triterpene and phytosterol biosynthesis in Panax ginseng overexpressing squalene synthase gene. Plant Cell Physiol 2004; 45: 976-984
  • 79 Shibata S. Chemistry and cancer preventing activities of ginseng saponins and some related triterpenoid compounds. J Korean Med Sci 2001; 16: S28-S37
  • 80 Lee SH, Ahn YM, Ahn SY, Doo HK, Lee BC. Interaction between warfarin and Panax ginseng in ischemic stroke patients. J Alternat Complement Med 2008; 14: 715-721
  • 81 Briskin DP. Medicinal plants and phytomedicines. Linking plant biochemistry and physiology to human health. Plant Physiol 2000; 124: 507-514
  • 82 Vogler BK, Pittler MH, Ernst E. The efficacy of ginseng. A systematic review of randomised clinical trials. Eur J Clin Pharmacol 1999; 55: 567-575
  • 83 Jiang X, Williams KM, Liauw WS, Ammit AJ, Roufogalis BD, Duke CC, Day RO, McLachlan AJ. Effect of St Johnʼs wort and ginseng on the pharmacokinetics and pharmacodynamics of warfarin in healthy subjects. Br J Clin Pharmacol 2004; 57: 592-599
  • 84 Rhyu MR, Lu J, Webster DE, Fabricant DS, Farnsworth NR, Wang ZJ. Black cohosh (Actaea racemosa, Cimicifuga racemosa) behaves as a mixed competitive ligand and partial agonist at the human mu opiate receptor. J Agric Food Chem 2006; 54: 9852-9857
  • 85 Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Khan IA, Shah A. In vivo effects of goldenseal, kava kava, black cohosh, and valerian on human cytochrome P450 1A2, 2D6, 2E1, and 3A4/5 phenotypes. Clin Pharmacol Ther 2005; 77: 415-426
  • 86 Gurley B, Hubbard MA, Williams DK, Thaden J, Tong Y, Gentry WB, Breen P, Carrier DJ, Cheboyina S. Assessing the clinical significance of botanical supplementation on human cytochrome P450 3A activity: comparison of a milk thistle and black cohosh product to rifampin and clarithromycin. J Clin Pharmacol 2006; 46: 201-213
  • 87 Borrelli F, Izzo AA. Herb-drug interactions with St Johnʼs wort (Hypericum perforatum): an update on clinical observations. AAPS J 2009; 11: 710-727
  • 88 Markowitz JS, DeVane CL, Boulton DW, Carson SW, Nahas Z, Risch SC. Effect of St. Johnʼs wort (Hypericum perforatum) on cytochrome P-450 2D6 and 3A4 activity in healthy volunteers. Life Sci 2000; 66: PL133-PL139
  • 89 Wang Z, Gorski JC, Hamman MA, Huang SM, Lesko LJ, Hall SD. The effects of St Johnʼs wort (Hypericum perforatum) on human cytochrome P450 activity. Clin Pharmacol Ther 2001; 70: 317-326
  • 90 Wenk M, Todesco L, Krahenbuhl S. Effect of St Johnʼs wort on the activities of CYP1A2, CYP3A4, CYP2D6, N-acetyltransferase 2, and xanthine oxidase in healthy males and females. Br J Clin Pharmacol 2004; 57: 495-499
  • 91 Roby CA, Anderson GD, Kantor E, Dryer DA, Burstein AH. St Johnʼs Wort: effect on CYP3A4 activity. Clin Pharmacol Ther 2000; 67: 451-457
  • 92 Markowitz JS, Donovan JL, DeVane CL, Taylor RM, Ruan Y, Wang JS, Chavin KD. Effect of St Johnʼs wort on drug metabolism by induction of cytochrome P450 3A4 enzyme. JAMA 2003; 290: 1500-1504
  • 93 Wang LS, Zhu B, Abd El-Aty AM, Zhou G, Li Z, Wu J, Chen GL, Liu J, Tang ZR, An W, Li Q, Wang D, Zhou HH. The influence of St Johnʼs Wort on CYP2C19 activity with respect to genotype. J Clin Pharmacol 2004; 44: 577-581
  • 94 Wang LS, Zhou G, Zhu B, Wu J, Wang JG, Abd El-Aty AM, Li T, Liu J, Yang TL, Wang D, Zhong XY, Zhou HH. St Johnʼs wort induces both cytochrome P450 3A4-catalyzed sulfoxidation and 2C19-dependent hydroxylation of omeprazole. Clin Pharmacol Ther 2004; 75: 191-197
  • 95 Mai I, Bauer S, Perloff ES, Johne A, Uehleke B, Frank B, Budde K, Roots I. Hyperforin content determines the magnitude of the St Johnʼs wort-cyclosporine drug interaction. Clin Pharmacol Ther 2004; 76: 330-340
  • 96 Arold G, Donath F, Maurer A, Diefenbach K, Bauer S, Henneicke-von Zepelin HH, Friede M, Roots I. No relevant interaction with alprazolam, caffeine, tolbutamide, and digoxin by treatment with a low-hyperforin St Johnʼs wort extract. Planta Med 2005; 71: 331-337
  • 97 Mueller SC, Majcher-Peszynska J, Uehleke B, Klammt S, Mundkowski RG, Miekisch W, Sievers H, Bauer S, Frank B, Kundt G, Drewelow B. The extent of induction of CYP3A by St. Johnʼs wort varies among products and is linked to hyperforin dose. Eur J Clin Pharmacol 2006; 62: 29-36
  • 98 Mueller SC, Majcher-Peszynska J, Mundkowski RG, Uehleke B, Klammt S, Sievers H, Lehnfeld R, Frank B, Thurow K, Kundt G, Drewelow B. No clinically relevant CYP3A induction after St. Johnʼs wort with low hyperforin content in healthy volunteers. Eur J Clin Pharmacol 2009; 65: 81-87
  • 99 Hall SD, Wang Z, Huang SM, Hamman MA, Vasavada N, Adigun AQ, Hilligoss JK, Miller M, Gorski JC. The interaction between St Johnʼs wort and an oral contraceptive. Clin Pharmacol Ther 2003; 74: 525-535
  • 100 Murphy PA, Kern SE, Stanczyk FZ, Westhoff CL. Interaction of St. Johnʼs Wort with oral contraceptives: effects on the pharmacokinetics of norethindrone and ethinyl estradiol, ovarian activity and breakthrough bleeding. Contraception 2005; 71: 402-408
  • 101 Pfrunder A, Schiesser M, Gerber S, Haschke M, Bitzer J, Drewe J. Interaction of St Johnʼs wort with low-dose oral contraceptive therapy: a randomized controlled trial. Br J Clin Pharmacol 2003; 56: 683-690
  • 102 Smith PF, Bullock JM, Booker BM, Haas CE, Berenson CS, Jusko WJ. Induction of imatinib metabolism by Hypericum perforatum . Blood 2004; 104: 1229-1230
  • 103 Frye RF, Fitzgerald SM, Lagattuta TF, Hruska MW, Egorin MJ. Effect of St Johnʼs wort on imatinib mesylate pharmacokinetics. Clin Pharmacol Ther 2004; 76: 323-329
  • 104 Andren L, Andreasson A, Eggertsen R. Interaction between a commercially available St. Johnʼs wort product (Movina) and atorvastatin in patients with hypercholesterolemia. Eur J Clin Pharmacol 2007; 63: 913-916
  • 105 Portoles A, Terleira A, Calvo A, Martinez I, Resplandy G. Effects of Hypericum perforatum on ivabradine pharmacokinetics in healthy volunteers: an open-label, pharmacokinetic interaction clinical trial. J Clin Pharmacol 2006; 46: 1188-1194
  • 106 Tannergren C, Engman H, Knutson L, Hedeland M, Bondesson U, Lennernas H. St Johnʼs wort decreases the bioavailability of R- and S-verapamil through induction of the first-pass metabolism. Clin Pharmacol Ther 2004; 75: 298-309
  • 107 Kawaguchi A, Ohmori M, Tsuruoka S, Nishiki K, Harada K, Miyamori I, Yano R, Nakamura T, Masada M, Fujimura A. Drug interaction between St Johnʼs Wort and quazepam. Br J Clin Pharmacol 2004; 58: 403-410
  • 108 Rengelshausen J, Banfield M, Riedel KD, Burhenne J, Weiss J, Thomsen T, Walter-Sack I, Haefeli WE, Mikus G. Opposite effects of short-term and long-term St Johnʼs wort intake on voriconazole pharmacokinetics. Clin Pharmacol Ther 2005; 78: 25-33
  • 109 Xu H, Williams KM, Liauw WS, Murray M, Day RO, McLachlan AJ. Effects of St Johnʼs wort and CYP2C9 genotype on the pharmacokinetics and pharmacodynamics of gliclazide. Br J Pharmacol 2008; 153: 1579-1586
  • 110 Piscitelli SC, Burstein AH, Chaitt D, Alfaro RM, Falloon J. Indinavir concentrations and St Johnʼs wort. Lancet 2000; 355: 547-548
  • 111 Kroll DJ, Shaw HS, Oberlies NH. Milk thistle nomenclature: why it matters in cancer research and pharmacokinetic studies. Integr Cancer Ther 2007; 6: 110-119
  • 112 Hogan FS, Krishnegowda NK, Mikhailova M, Kahlenberg MS. Flavonoid, silibinin, inhibits proliferation and promotes cell-cycle arrest of human colon cancer. J Surg Res 2007; 143: 58-65
  • 113 Brantley SJ, Oberlies NH, Kroll DJ, Paine MF. Two flavonolignans from milk thistle (Silybum marianum) inhibit CYP2C9-mediated warfarin metabolism at clinically achievable concentrations. J Pharmacol Exp Ther 2010; 332: 1081-1087
  • 114 van Erp NP, Baker SD, Zhao M, Rudek MA, Guchelaar HJ, Nortier JW, Sparreboom A, Gelderblom H. Effect of milk thistle (Silybum marianum) on the pharmacokinetics of irinotecan. Clin Cancer Res 2005; 11: 7800-7806
  • 115 Lu H, Meng X, Li C, Sang S, Patten C, Sheng S, Hong J, Bai N, Winnik B, Ho CT, Yang CS. Glucuronides of tea catechins: enzymology of biosynthesis and biological activities. Drug Metab Dispos 2003; 31: 452-461
  • 116 Feng WY. Metabolism of green tea catechins: an overview. Curr Drug Metab 2006; 7: 755-809
  • 117 Zhang Q, Fu H, Pan J, He J, Ryota S, Hara Y, Wang Y, Lubet RA, You M. Effect of dietary Polyphenon E and EGCG on lung tumorigenesis in A/J Mice. Pharm Res 2010; 27: 1066-1071
  • 118 Yang CS, Chen L, Lee MJ, Balentine D, Kuo MC, Schantz SP. Blood and urine levels of tea catechins after ingestion of different amounts of green tea by human volunteers. Cancer Epidemiol Biomarkers Prev 1998; 7: 351-354
  • 119 Yang TT, Koo MW. Chinese green tea lowers cholesterol level through an increase in fecal lipid excretion. Life Sci 2000; 66: 411-423
  • 120 Yang TT, Koo MW. Inhibitory effect of Chinese green tea on endothelial cell-induced LDL oxidation. Atherosclerosis 2000; 148: 67-73
  • 121 Zhong Z, Froh M, Connor HD, Li X, Conzelmann LO, Mason RP, Lemasters JJ, Thurman RG. Prevention of hepatic ischemia-reperfusion injury by green tea extract. Am J Physiol Gastrointest Liver Physiol 2002; 283: G957-G964
  • 122 Oyama J, Maeda T, Kouzuma K, Ochiai R, Tokimitsu I, Higuchi Y, Sugano M, Makino N. Green tea catechins improve human forearm endothelial dysfunction and have antiatherosclerotic effects in smokers. Circ J 2010; 74: 578-588
  • 123 Oyama J, Maeda T, Sasaki M, Kozuma K, Ochiai R, Tokimitsu I, Taguchi S, Higuchi Y, Makino N. Green tea catechins improve human forearm vascular function and have potent anti-inflammatory and anti-apoptotic effects in smokers. Intern Med 2010; 49: 2553-2559
  • 124 Persson IA, Persson K, Hagg S, Andersson RG. Effects of green tea, black tea and Rooibos tea on angiotensin-converting enzyme and nitric oxide in healthy volunteers. Public Health Nutr 2010; 13: 730-737
  • 125 Nantz MP, Rowe CA, Bukowski JF, Percival SS. Standardized capsule of Camellia sinensis lowers cardiovascular risk factors in a randomized, double-blind, placebo-controlled study. Nutrition 2009; 25: 147-154
  • 126 Donovan JL, Chavin KD, Devane CL, Taylor RM, Wang JS, Ruan Y, Markowitz JS. Green tea (Camellia sinensis) extract does not alter cytochrome p 450 3A4 or 2D6 activity in healthy volunteers. Drug Metab Dispos 2004; 32: 906-908
  • 127 Chow HH, Hakim IA, Vining DR, Crowell JA, Cordova CA, Chew WM, Xu MJ, Hsu CH, Ranger-Moore J, Alberts DS. Effects of repeated green tea catechin administration on human cytochrome P450 activity. Cancer Epidemiol Biomarkers Prev 2006; 15: 2473-2476
  • 128 Kennedy DO, Wightman EL. Herbal extracts and phytochemicals: plant secondary metabolites and the enhancement of human brain function. Adv Nutr 2011; 2: 32-50
  • 129 Houghton PJ. The scientific basis for the reputed activity of Valerian. J Pharm Pharmacol 1999; 51: 505-512
  • 130 Houghton PJ. The biological activity of Valerian and related plants. J Ethnopharmacol 1988; 22: 121-142
  • 131 Leathwood PD, Chauffard F, Heck E, Munoz-Box R. Aqueous extract of valerian root (Valeriana officinalis L.) improves sleep quality in man. Pharmacol Biochem Behav 1982; 17: 65-71
  • 132 Leathwood PD, Chauffard F. Aqueous extract of valerian reduces latency to fall asleep in man. Planta Med 1985; 144-148
  • 133 Dorn M. [Efficacy and tolerability of Baldrian versus oxazepam in non-organic and non-psychiatric insomniacs: a randomised, double-blind, clinical, comparative study]. Forsch Komplementarmed Klass Naturheilkd 2000; 7: 79-84
  • 134 Donath F, Quispe S, Diefenbach K, Maurer A, Fietze I, Roots I. Critical evaluation of the effect of valerian extract on sleep structure and sleep quality. Pharmacopsychiatry 2000; 33: 47-53
  • 135 Donovan JL, DeVane CL, Chavin KD, Wang JS, Gibson BB, Gefroh HA, Markowitz JS. Multiple night-time doses of valerian (Valeriana officinalis) had minimal effects on CYP3A4 activity and no effect on CYP2D6 activity in healthy volunteers. Drug Metab Dispos 2004; 32: 1333-1336
  • 136 Anke J, Ramzan I. Pharmacokinetic and pharmacodynamic drug interactions with Kava (Piper methysticum Forst. f.). J Ethnopharmacol 2004; 93: 153-160
  • 137 Thompson R, Ruch W, Hasenohrl RU. Enhanced cognitive performance and cheerful mood by standardized extracts of Piper methysticum (Kava-kava). Hum Psychopharmacol 2004; 19: 243-250
  • 138 Kennedy DA, Seely D. Clinically based evidence of drug-herb interactions: a systematic review. Expert Opin Drug Saf 2010; 9: 79-124
  • 139 Gurley BJ, Swain A, Hubbard MA, Hartsfield F, Thaden J, Williams DK, Gentry WB, Tong Y. Supplementation with goldenseal (Hydrastis canadensis), but not kava kava (Piper methysticum), inhibits human CYP3A activity in vivo . Clin Pharmacol Ther 2008; 83: 61-69
  • 140 Brown PN, Roman MC. Determination of hydrastine and berberine in goldenseal raw materials, extracts, and dietary supplements by high-performance liquid chromatography with UV: collaborative study. J AOAC Int 2008; 91: 694-701
  • 141 Lancaster HM, Davidson AL. Commercial pharmaceutical preparations: 4. Hydrastis – golden seal. Can Med Assoc J 1927; 17: 1317-1320
  • 142 Chatterjee P, Franklin MR. Human cytochrome p 450 inhibition and metabolic-intermediate complex formation by goldenseal extract and its methylenedioxyphenyl components. Drug Metab Dispos 2003; 31: 1391-1397
  • 143 Yi S, Cho JY, Lim KS, Kim KP, Kim J, Kim BH, Hong JH, Jang IJ, Shin SG, Yu KS. Effects of Angelicae tenuissima radix, Angelicae dahuricae radix and Scutellariae radix extracts on cytochrome P450 activities in healthy volunteers. Basic Clin Pharmacol Toxicol 2009; in press
  • 144 Fan L, Wang JC, Jiang F, Tan ZR, Chen Y, Li Q, Zhang W, Wang G, Lei HP, Hu DL, Wang D, Zhou HH. Induction of cytochrome P450 2B6 activity by the herbal medicine baicalin as measured by bupropion hydroxylation. Eur J Clin Pharmacol 2009; 65: 403-409
  • 145 Ramassamy C. Emerging role of polyphenolic compounds in the treatment of neurodegenerative diseases: a review of their intracellular targets. Eur J Pharmacol 2006; 545: 51-64
  • 146 Chow HH, Garland LL, Hsu CH, Vining DR, Chew WM, Miller JA, Perloff M, Crowell JA, Alberts DS. Resveratrol modulates drug- and carcinogen-metabolizing enzymes in a healthy volunteer study. Cancer Prev Res (Phila) 2010; 3: 1168-1175
  • 147 Chen Y, Liu WH, Chen BL, Fan L, Han Y, Wang G, Hu DL, Tan ZR, Zhou G, Cao S, Zhou HH. Plant polyphenol curcumin significantly affects CYP1A2 and CYP2A6 activity in healthy, male Chinese volunteers. Ann Pharmacother 2010; 44: 1038-1045