You are on page 1of 16

Eur J Drug Metab Pharmacokinet

DOI 10.1007/s13318-015-0296-z

REVIEW

Pharmacokinetic Herb-Drug Interactions:


Insight into Mechanisms and Consequences
Enoche F. Oga1 • Shuichi Sekine2 • Yoshihisa Shitara3 • Toshiharu Horie4

Ó Springer International Publishing Switzerland 2015

Abstract Herbal medicines are currently in high demand, needed. The authors further suggest the need to standardize
and their popularity is steadily increasing. Because of their and better regulate herbal medicines in order to ensure their
perceived effectiveness, fewer side effects and relatively safety and efficacy when used alone or in combination with
low cost, they are being used for the management of conventional drugs.
numerous medical conditions. However, they are capable
of affecting the pharmacokinetics and pharmacodynamics
of coadministered conventional drugs. These interactions
Key Points
are particularly of clinically relevance when metabolizing
enzymes and xenobiotic transporters, which are responsible
Herbal medicines are usually a complex mixture of
for the fate of many drugs, are induced or inhibited,
chemical constituents whose bioactive components
sometimes resulting in unexpected outcomes. This article
in many instances are yet to be fully characterized,
discusses the general use of herbal medicines in the man-
but are believed to be medicinally efficacious
agement of several ailments, their concurrent use with
conventional therapy, mechanisms underlying herb-drug Metabolizing enzymes and xenobiotic transporters,
interactions (HDIs) as well as the drawbacks of herbal which are responsible for fate of many drugs, could
remedy use. The authors also suggest means of surveillance also be induced or inhibited by these herbal
and safety monitoring of herbal medicines. Contrary to medicines
popular belief that ‘‘herbal medicines are totally safe,’’ we Clinically relevant herb-drug interactions may occur
are of the view that they are capable of causing significant following their coadministration, sometimes
toxic effects and altered pharmaceutical outcomes when resulting in fatal consequences, informing the need
coadministered with conventional medicines. Due to the for caution in the use of these herbal medicines as
paucity of information as well as sometimes conflicting well as more controlled clinical studies in order to
reports on HDIs, much more research in this field is fully clarify the underlying mechanisms for these
altered drug effects
& Enoche F. Oga
eoga@uclan.ac.uk
1 1 Overview of Herbal Medicine Use
School of Pharmacy and Biomedical Sciences, University of
Central Lancashire, Preston, Lancashire PR1 2HE, UK
2 Herbal medicine comprises the use of plant parts for
Department of Biopharmaceutics, Chiba University, Chiba,
Japan medicinal purposes. Unlike conventional drugs, most her-
3 bal medicines are a complex mixture of chemical con-
Pharmacokinetics Laboratory, Meiji Seika Pharma Co., Ltd,
Yokohama, Japan stituents whose bioactive components in many instances
4 are yet to be fully characterized, but have been reported to
Department of Biopharmaceutics and Molecular Toxicology,
Teikyo Heisei University, Tokyo, Japan be medicinally efficacious [1]. There is a long history of
E. F. Oga et al.

the use of these phytomedicines, with herbalism having a of UK herbal medicine and its practitioners [10, 11]. It is
long tradition of use outside of conventional medicine. worth noting that approximately 30–50 % of conventional
Their use has become more established as improvements in medicines in use evolved from natural products from which
analysis and quality control as well as advances in clinical they are synthesized as pharmaceutical preparations [12–
research have shown their value in treating and preventing 14]. For instance, the medicines Catharanthus roseus
disease [2]. In some developing countries, up to 80 % of (vincristine), Atropa belladonna (atropine), Cinchona sp.
the indigenous populace is known to depend on traditional (quinine), Cephaelis ipecacuanha (emetine), Digitalis
systems of medicine and medicinal plants as their primary purpurea (digitoxin and digoxin), Ephedra sinica (ephe-
source of healthcare [3, 4]. This occurrence is not signifi- drine), (Artemisia annua (artemisinin), Rauwolfia serpen-
cantly different across the globe. For instance, reports show tine (reserpine) and Podophyllum peltatum (etoposide)
that over 70 % of German physicians prescribe herbal among several others.
medicines and over 75 % of the German populace has used The main focus of this article is evidence for pharma-
complementary or natural medicine [5]. In countries like cokinetic herb-drug interactions. A review of the litera-
the USA, an increase in their use has been reported because ture reporting herbal medicines, their effects and
of dissatisfaction with the cost of prescription medications, documented interactions with conventional drugs was
combined with an interest in returning to natural or organic conducted between January 2012 and June 2015 on
medicines [6]. In Japan, about 148 kinds of Japanese herbal research databases. Relevant studies were identified
medicines (Kampo medicines) have been approved and are through searches of electronic databases, citations, refer-
listed on the National Health Insurance Drug Tariff. These ence lists, comprehensive pearl-growing and handsearch-
medicines are reportedly prescribed by 72–78 % of Japa- ing-related journals, and conference proceedings. The
nese physicians [7, 8]. These plant-based medicines are relevant online databases (including PubMed, Cochrane
believed to be increasingly utilized because of their low database, Google Scholar and Web of Science) were uti-
cost, perceived safety, efficacy and lower incidence of lized using online keyword searches, with searches limited
adverse effects when compared to orthodox medicines. by date to reports published from 1985 onwards.
While their use is on the increase in some countries,
stringent regulatory requirements in other countries make it
difficult to register herbal medicines, therefore their 2 Mechanisms of Herb-Drug Interactions
reported limited use [9]. In the UK, herbal medicines have
been recognized as substances that pose particular chal- Similar to conventional pharmaceutical products, pharma-
lenges for public health, and effective regulation is being cokinetic and pharmacodynamic alterations are the two
sought that would safeguard the public. To this end, there mechanistic pathways through which herb-drug interac-
have been deliberations on the need for statutory regulation tions (HDIs) occur, resulting in null, beneficial or toxic

Fig. 1 Illustration of the main Herb-Drug Interacons


mechanisms of herb-drug
interactions with some
examples (OATP organic anion
transporting polypeptides, P-gp Pharmacodynamic Pharmacokinec
permeability glycoprotein)

Drug Transporters Metabolizing enzymes


• Addive
• Efflux (Eg. P-gp)
• Synergisc
• Uptake (Eg. OATP) Phase I Phase II
• Antagonisc

Changes in absorpon, distribuon,


metabolism, excreon, protein binding
Eg.
leading to a change in the level of drug
• Kava (Piper methyscum)+ Benzodiazepines
and/or metabolite(s)
• Guarana (Paullinia cupana)+ Sedaves
• Comfrey (Symphytum officinale ) +
Acetaminophen Eg.
• St John’s Wort + Ritonavir
• Milk thistle + Warfarin
• St John’s Wort + Indinavir
Pharmacokinetic Herb-Drug Interactions

responses (Fig. 1). In addition to the cytochrome enzymes, 2.2 Transporter-Mediated HDIs
membrane transporters are known to play an important role
in the modulation of absorption, distribution, metabolism Although most studies have focused on CYP-based drug
and excretion of drugs [15]. These HDIs arise from chan- interactions, the influence of transporters as a mechanism
ges in the function and expression of transporters or for HDIs is increasingly being documented, as it has been
enzymes that mediate the absorption and elimination of revealed that they can play an important role in modulating
drugs in the small intestine, kidney and liver [16]. drug absorption, distribution, metabolism and elimination.
Figure 2 illustrates the distribution of relevant transporters
2.1 Enzyme-Mediated HDIs in the main organs responsible for drug disposition.
Xenobiotic transporters are generally categorized into the
One of the most important causes of clinically relevant ATP-binding cassette (ABC) and solute-carrier (SLC)
HDIs is the inhibition or induction of the activity of superfamilies.
cytochrome P450 (CYP), a superfamily of enzymes cat-
alyzing extremely diverse and often complex reactions in 2.2.1 ATP-Binding Cassette-Based HDIs
the metabolism of numerous drugs, phytomedicines and
xenobiotics. CYPs are the main enzymes involved in drug ABC transporters function as barrier proteins extruding
metabolism, with the majority of drugs interacting with the toxins and xenobiotics out of cells. They include p-glyco-
CYP3A isoform [17, 18]. For instance, several intestinal protein (P-gp), breast cancer resistance protein (BCRP),
CYP3A inhibitors (including the triterpenes, maslinic acid, bile salt export pump (BSEP) as well as several multidrug-
corosolic acid and ursolic acid) have been isolated from associated resistance proteins (MRPs). They are commonly
Vaccinium macrocarpon (cranberry) [19]. Cranberry is expressed on barrier epithelia where they mediate transport
now commonly taken as a treatment for urinary tract across cell membranes [15]. These transporters are signif-
infections because of a group of proanthocyanidins that icant determinants of the pharmacokinetics, efficacy and
exhibit bacterial antiadhesion activity against both antibi- toxicity of xenobiotics (including phytomedicines), and
otic-susceptible and -resistant strains of uropathogenic coadministered drugs may inactivate, inhibit or induce
Escherichia coli bacteria [19, 20]. Another study revealed these transporters. Of all the transporter-mediated HDIs,
that cranberry juice inhibits the CYP3A-mediated meta- P-gp-based interactions are the most studied. There are
bolism of nifedipine, altering its pharmacokinetics by reports on the modulation of P-gp by herbs such as
increasing the concentration of nifedipine in rat plasma Hypericum perforatum (St. John’s wort), Vernonia amyg-
[21]. The study demonstrated that the oxidative activities dalina (Bitter leaf), Tapinanthus sessilifolius (African
of nifedipine in rat intestinal and human hepatic micro- mistletoe), Coptis chinensis (Chinese goldthread), Ginkgo
somes were inhibited after preincubation with cranberry biloba (Maidenhair tree), Piper nigrum (Black pepper) and
juice [21]. However, a clinical study in ten healthy vol- Glycorrhiza glabra (Licorice) [16, 27, 28].
unteers involving the CYP3A probe substrate, midazolam
and a cranberry juice product suggested that an interaction 2.2.2 Solute Carrier-Based HDIs
was unlikely [22]. Interestingly, in a more recent study that
utilized a systematic approach, from in vitro assay (Caco-2 The solute carrier (SLC) transporters mainly comprise
cell and human intestinal microsomes) to a clinical study in uptake transporters including the organic anion transport-
16 healthy volunteers, it was observed that a cranberry ing polypeptides (OATPs), organic cation transporters
juice product revealed a pharmacokinetic interaction with (OCTs), organic anion transporters (OATs), peptide
midazolam [23]. The difference in the finding of these transporters (PEPT) and multidrug and toxic compound
studies suggests the importance of utilizing clinical regi- extrusion transporters (MATEs) [15]. These SLC trans-
mens and taking into consideration the interbrand variation porters are known to facilitate absorption of phytomedici-
of natural products in the design of pharmacokinetic nes and xenobiotics into the systemic circulation. Because
studies. Similarly, the widely used herbal remedy Echi- transporters are mainly expressed in the intestinal epithelial
nacea purpurea (Echinacea) has been reported to signifi- cells and organs of elimination (kidney proximal tubules
cantly induce CYP3A [24]. It has been shown to selectively and liver hepatocytes), they can significantly influence the
modulate the catalytic activity of CYP3A at hepatic and disposition of herbal medicines [29]. Fewer reports inves-
intestinal sites, suggesting that CYP3A substrates with tigate the influence of SLC-mediated herb-drug interac-
relatively high bioavailability may be more susceptible tions when compared to ABC transporters, where the
to Echinacea-mediated interactions [25, 26]. As CYP3A is majority of the studies examine the interaction with P-gp
the main drug-metabolizing enzyme, the potential of HDIs [30]. However, several interesting drug-food interactions
occurring with Echinacea purpurea may be high. with fruit juices involving uptake transporters have been
E. F. Oga et al.

a Enterocytes b Hepatocytes
BCRP
Efflux BSEP
Uptake OATP MRP3 MATE1
PEPT1 MRP2
OAT2
Uptake OATP1B1 P-gp
OATP1B3 Bile
BCRP OATP2B1
Efflux Uptake MRP2 Efflux
MRP2 OCT OCT1
MRP4
P-gp

Apical membrane Basolateral membrane

c Renal tubular cell

MATE1
OAT1 MATE2-K
Uptake OAT2 MRP2 Efflux
OAT3 MRP4
OATP4C1 OCTN1
OCT2 OCTN2
P-gp

Blood Renal lumen

Fig. 2 Transporter expression in the main organs influencing drug export pump, MATE multidrug and toxic compound extrusion
disposition (a). Uptake and efflux transporters at the apical and transporters, MRP multidrug-associated resistance proteins, OAT
basolateral membranes of enterocytes (b). Uptake and efflux trans- organic anion transporters, OATP organic anion transporting polypep-
porters in hepatic cells (c). Uptake and efflux transporters in the renal tides, OCT organic cation transporters, OCTN carnitine/organic cation
epithelial cells (BCRP breast cancer resistance protein, BSEP bile salt transporter, P-gp permeability glycoprotein)

reported [31, 32]. For instance, grapefruit juice inhibition miltiorrhiza (Red sage), a traditional Chinese herbal med-
of OATP resulted in reduced blood concentrations of fex- icine that is commonly used for the prevention and treat-
ofenadine, celiprolol, talinolol and acebutolol [33]. On the ment of cerebrovascular and cardiovascular disorders.
contrary, concurrent intake of grapefruit juice has been Likewise, herbs modulating OAT1, a transporter actively
shown to increase the in vivo plasma concentrations of involved in renal active secretion, have been reported [37].
many drugs. This has been attributed to various factors, Among several Chinese herbal medicines examined in the
including, the inhibition of P-gp and an inhibition of study, Gui Zhi Fu Ling Wan (commonly formulated as a
intestinal CYP3A4, resulting in an increase in the fraction mixture of Ramulus cinnamomi, Poria cocos, Cotex mou-
of drug absorbed [34]. Other clinical reports on SLC tan, Radix paeonia and Semen persicae) and Chia Wei
transporters include a study in healthy volunteers on bai- Hsiao Yao San (commonly formulated as a mixture of
calin from Radix scutellariae (Baikal skullcap), which Radix bupleuri, Radix glycyrrhizae, Angelica sinensis and
showed a reduction in the plasma concentration of coad- Paeonia alba) exhibited significant inhibitions on hOAT1-
ministered rosuvastatin as a result of OATP modulation mediated [3H]-p-amino hippuric acid uptake in vitro as
[35]. In the study, the observed decrease in the plasma well as p-amino hippuric acid clearance and net secre-
concentration of rosuvastatin in the presence of baicalin tion in vivo [37].
was thought to be partially mediated by baicalin’s induc-
tion of hepatic rosuvastatin uptake through OATP1B1 [35]. 2.3 Dual Enzyme- and Transporter-Mediated HDIs
A study in rats reported that the pharmacokinetic disposi-
tion of salvianolic acid B was altered by rifampicin because Some phytomedicines are known to influence both trans-
of the inhibition of Oatp-mediated influx [36]. Salvianolic porter and CYP function. P-gp and CYP3A4 both consti-
acid B is one of the most bioactive components of Salvia tute a highly efficient barrier for many orally absorbed
Pharmacokinetic Herb-Drug Interactions

drugs with a wide overlap in their substrates [38]. Rhodiola with their conventional chemotherapeutic treatment [49].
rosea (Golden root), a herbal medicine used in the man- Among cancer patients in general, between 7 and 48 %
agement of depression, has shown potent inhibition of both have reported taking herbal medicines [50]. These herbal
P-gp and CYP3A4 [39]. Also, St. John’s wort (Hypericum medicines are used by cancer patients with the belief that
perforatum), one of the most widely used herbal medicines, they are capable of killing tumor cells, improving cancer-
with several medicinal effects including its recognized related symptoms as well as reducing the adverse drug
antidepressant properties, induces both CYP3A and the effects posed by the therapy [51]. However, in order to
efflux transporter, P-gp [40]. A recent study revealed that appropriately integrate herbal medicine use into conven-
quercetin and rutin, which are common herbal flavonoids, tional cancer therapy, pharmacological and clinical studies
induced the functions of both P-gp and CYP3A4 by must be carried out on these herbals, with relevant moni-
decreasing the bioavailability of coadministered cyclos- toring for the emergence of adverse effects [52]. This is of
porin [41]. These alterations of normal P-gp efflux and particular importance as several chemotherapeutic agents
CYP activity have an impact on the pharmacokinetic dis- are known to have considerable interindividual pharma-
position of CYP3A and P-gp substrate drugs when coad- cokinetic variability, which coupled with their narrow
ministered, leading to changes including lower efficacy therapeutic index may pose a higher risk for the occurrence
and/or the emergence of toxicity [42]. of toxic HDIs. For instance, St. John’s wort has been
reported to induce the metabolism of imatinib, an oncolytic
2.4 Influence on Transcriptional Regulators used in the management of chronic myelogenous leukemia
and gastrointestinal stromal tumors. These studies showed
Several CYPs and transporters that influence drug dispo- an alteration in the pharmacokinetics of imatinib when
sition can be induced by xenobiotics and herbs [43]. The coadministered with St. John’s wort due to an induction of
nuclear receptors pregnane X receptor (PXR) and consti- CYP3A [53, 54]. This induction of CYP isoenzymes and
tutive androstane receptor (CAR), which are present in the drug transporters would often lead to therapeutic failure as
small intestine and liver, have emerged as transcriptional a result of a lower plasma concentration of the anticancer
regulators of CYPs (especially CYP3A, -2B6, -2C8, -2C9 drugs [49]. Likewise, a pharmacodynamic interaction
and -2C19) and drug transporters: P-gp, MRP2 and OATP involving irinotecan (a potent anticancer drug for the
[44, 45]. Besides the various xenobiotics that have been management of advanced colon cancer), which may cause
reported to activate PXR, herbals including St. John’s Wort severe diarrhea as an adverse effect, has been reported to
are known to potently induce it [46]. These nuclear have worsened diarrhea on coadministration of St. John’s
receptors have also been reported to enhance the expres- wort through downregulation of intestinal proinflammatory
sion of phase II-conjugating enzymes such as uridine 50 - cytokines and inhibition of intestinal epithelial apoptosis
diphospho-glucuronosyltransferase (UDP-glucuronosyl- [55]. In another study, the metabolism and toxicity of
transferase, UGT), sulfotransferase and glutathione-S- irinotecan was altered on coadministration with St. John’s
transferase enzymes [47, 48]. As a result of an increased wort with a compromise in overall antitumor activity [56].
induction of these nuclear receptors, there is an enhanced Grapefruit juice intake has been reported to alter the
expression of P-gp and CYPs, which is likely to reduce the pharmacokinetics of the cytotoxic drug, etoposide, with a
rate of absorption and increase the rate of elimination of clinical study revealing a 26.2 % decrease in the area under
drug substrates. the plasma concentration-time curve (AUC) of etoposide
after oral administration. It was postulated that the alter-
ation of intestinal P-gp-mediated transport was a possible
3 Herbal Medicine Use and Interactions explanation for the observed effect [57]. Several in vitro
with Conventional Drugs studies have been reported with pointers to the possibility
of relevant herb-drug interactions. For example, Gin-
Several evidence-based studies on clinically relevant HDIs senoside Rh2 from Panax ginseng significantly enhanced
have been documented. Table 1 summarizes some clini- the cytotoxicity of daunomycin and vinblastine in adri-
cally significant herb-drug interactions. amycin-resistant P388 leukaemia cells [58]. Another
pharmacologically active constituent from Panax ginseng,
3.1 Cancer Ginsenoside Rg3, inhibited the efflux of vinblastine and
reversed resistance to doxorubicin and vincristine in drug-
Because of the high global incidence of cancer, the use of resistant KBV20C cells [59]. Competition of Ginsenoside
herbal medicines by cancer patients is quite common with Rg3 for binding to P-gp was demonstrated as the mecha-
an increasing number of cancer patients making use of nism for the inhibition of drug efflux. The study showed
complementary and alternative medicines in combination that ginsenoside Rg3 was an effective modulator in
Table 1 Clinically relevant herb-drug interactions
Disorder Conventional drug Herbal medicine Effect in human subjects References

Cancer Imatinib St. John’s wort (Hypericum 43 % : in imatinib CL [49, 53,


perforatum) ; of up to 32 % mean AUC 54]
Significant ; in Cmax and t1/2
Irinotecan St. John’s wort 42 % ; in Cp of the active metabolite of irinotecan [49, 55,
56]
Etoposide Echinacea (Echinacea purpurea) Median number of leucocytes : significantly in comparison to the control [126]
Leucovirin group
5-Fluoro uracil
Etoposide Echinacea One case reporting a possible interaction between etoposide, and [127]
echinacea which resulted in trombocytopenia requiring a platelet
transfusion
HIV/AIDS Saquinavir Garlic (Allium sativum) Significantly decreases the systemic exposure and [71]
mean saquinavir AUC by 51 %
; Cmax by 54 %
Indinavir St. John’s wort AUC0–8 of indinavir decreased by a mean of 57 % [68]
49–99 % ; in concentration 8 h post-dosing
; in the mean Cmax of indinavir from 12.3 to 8.9 lg/mL
Nevirapine St. John’s wort : in CL of nevirapine by 35 %a [128]
Protease inhibitors SP-303 (an extract of Croton Significant reduction of stool weight and stool frequency in AIDS [129]
lechler) patients who had diarrhea
Cardiovascular disease Nifedipine Ginseng (Panax ginseng) : Cmax of nifedipine by 29 % [130]
Simvastatin St. John’s wort 52 % ; in AUC and 28 % ; in Camax [131]
Debrisoquin Goldenseal (Hydrastis canadensis) Goldenseal strongly inhibited CYP2D6 and CYP3A4/5 activity [132]
Talinolol Curcumin (Curcuma longa) The consumption of curcumin ; AUC and Cmax of talinolol [133]
Digoxin St. John’s wort 10 days of treatment with St. John’s wort ; digoxin AUC(0–24) by 25 % [102]
Diabetes Chlorpropamide Garlic : hypoglycaemia [134]
Asthma and allergy Fexofenadine St. John’s wort Single dose significantly : Cmax of fexofenadine by 45 % and [98]
significantly ; oral CL by 20 %b
Long-term dose significantly ; Cmax of fexofenadine by 35 % and
significantly : oral CL by 47 %c
Fexofenadine Grape fruit juice (Citrus paradisi) Consumption of grapefruit juice concomitantly or 2 h before [135]
fexofenadine administration was associated with ; oral fexofenadine Cp
Fexofenadine Orange juice (Citrus sinensis) Orange juice ; the AUC, Cmax and the urinary excretion values [136]
fexofenadine
Thiazides Gingko (Gingko biloba) : blood pressure when combined with thiazide diuretics [134]
E. F. Oga et al.
Table 1 continued
Disorder Conventional drug Herbal medicine Effect in human subjects References

Depression Midazolam Echinacea Significantly : systemic CL of midazolam by 34 % [25]


Significantly ; midazolam AUC by 23 %
Significantly : oral availability of midazolam after echinacea dosing
Midazolam Goldenseal Goldenseal strongly inhibited CYP3A4/5 activity [132]
Alprazolam St. John’s wort Significantly ; alprazolam AUC by 54 % [42]
SSRIs (Citalopram, fluoxetine, St. John’s wort Increased serotonergic effects with risk of increased incidence of adverse [137]
fluvoxamine, paroxetine, reactions
sertraline)
Pharmacokinetic Herb-Drug Interactions

Deep vein thrombosis Warfarin Ginkgo Potently inhibits platelet activating factor-mediated platelet aggregation [138–140]
Ibuprofen : the fluidity of blood
Warfarin Garlic Garlic : clotting time and international normalized ratio (INR) of warfarin [131]
Warfarin St. John’s wort ; anticoagulant effect of warfarin [141]
Warfarin Ginseng Ginseng significantly ; warfarin’s anticoagulant effect by ; the INR and [105]
reducing plasma warfarin concentrations
CNS stimulation Caffeine Echinacea Echinacea dosing significantly ; the oral CL of caffeine [25]
Immunosuppression Cyclosporine St. John’s wort 46 % ; in AUC and 42 % ; in Ca,c
max of cyclosporine [142]
Tacrolimus ; in AUC by 58 %a,c of cyclosporine [143]
Vitamin supplementation Folic acid Green tea (Camellia sinensis) Green and black tea extracts ; folic acid bioavailability [144]
Mineral supplementation Iron Chilli (Capsicum annuum) Inhibition of iron absorption in young women [145]
Antitussive Dextromethorphan Bitter orange (Citrus Bioavailability of dextromethorphan increased significantly with grapefruit and [146]
aurantium) seville orange juicea,d
Grapefruit (Citrus paradisi)
Oral Contraception Norethindrone St. John’s wort Significant : in oral CL of norethindrone, : risk of unintended pregnancy and [147]
breakthrough bleeding
Ethinyl estradiol St. John’s wort Significant ; in the half-life of ethinyl estradiol, : risk of unintended pregnancy [147]
and breakthrough bleeding
Headaches Triptans (sumatriptan, naratriptan, St. John’s wort Increased serotonergic effects with risk of increased incidence of adverse [137]
rizatriptan, zolmitriptan) reactions
Gastric ulcers Omeprazole Gingko Significant ; in plasma concentrations of omeprazole and omeprazole sulphone [148]
and significant : of 5-hydroxyomeprazole
AUC area under the plasma concentration-time curve, AUC(0–t) area under the plasma concentration-time curve from time zero to time t, CL clearance, Cmax maximum plasma concentration, Cp
plasma concentration, INR international normalized ratio, t1/2 half life, SSRI selective serotonin reuptake inhibitor
a
Suggested mechanism, induction of CYP 3A4
b
Suggested mechanism, inhibition of intestinal p-gp
c
Suggested mechanism, induction of p-gp
d
Suggested mechanism, inhibition of intestinal p-gp
E. F. Oga et al.

restoring the sensitivity of resistant cells to doxorubicin, they are coadministered with conventional medicines as
vincristine, etoposide and colchicine in human P-gp MDR their concurrent use has resulted in beneficial and/or
cells at concentrations of 5–40 lM [59]. Although no detrimental effects. For instance, an in vitro study revealed
clinical report on drug interaction studies involving gin- that Sutherlandia frutescens (Cancer bush) inhibited the
seng and P-gp substrates are known, these in vitro studies metabolism of atazanavir in human liver microsomes and
on its P-gp blockade role may point to its beneficial may have important implications for the absorption and
function as a natural multidrug resistance reversal agent metabolism and the overall oral bioavailability of ataza-
[60]. navir [66]. Also the HIV protease inhibitor ritonavir on
Also on a positive note, because of the hepatotoxicity coadministration with some herbal components has been
caused by some conventional anticancer drugs, herbal shown to modulate both P-gp and CYP3A4. In particular,
medicines including Punica granatum (pomegranate), the herbal constituents kaempferol and quercetin (from
Phyllanthus emblica (Indian gooseberry), Mangifera indica several plants), hypericin (from Hypericum perforatum)
(mango), Acacia catechu (Black cutch) and Camellia and allicin (from Allium sativum) have been shown to
sinensis (Tea) have been used for their hepatoprotective and inhibit the in vitro efflux and CYP3A4-mediated metabo-
antioxidant properties when hepatotoxic chemotherapeutic lism of xenobiotics and may interact with HIV antiretro-
agents are utilized [61]. Other herbal medicines used fre- virals that are P-gp or CYP3A4 substrates, such as ritonavir
quently by cancer patients include Zingiber officinale (gin- [67]. Also, a clinical study in healthy volunteers demon-
ger), Ginkgo biloba (ginkgo), Piper methysticum (kava- strated that St. John’s wort potently altered the pharma-
kava), quercetin (from several plants and also honey), Panax cokinetics of indinavir (a protease inhibitor and known
sp (ginseng), Curcuma longa (curcumin), Viscum album CYP substrate) via induction of CYP3A by St. John’s wort.
(European mistletoe), beta-carotene (especially from Dau- As illustrated in Fig. 3, the study revealed a large reduction
cus carota), Glycyrrhiza glabra (licorice), Astragalus in indinavir concentrations by concomitant St. John’s wort.
membranaceus (astragalus), Viscum album (mistletoe), This finding has important clinical implications for HIV-
Echinacea sp. (echinacea), Berberis aristata (Indian bar- infected patients receiving the two agents since low plasma
berry) and Allium sativum (garlic). These medications are concentrations of protease inhibitors are a cause of
perceived to be nontoxic, alleviate the symptoms of cancer, antiretroviral resistance and treatment failure [68]. Garlic is
boost the immune system, manage the adverse effects of another herbal remedy commonly utilized by HIV/AIDS
chemotherapeutics or even tackle the cancer itself [27, 62]. patients. Although some studies have shown that garlic has
However, there is limited scientific evidence for their the potential to induce CYP isoenzymes, consequently
interaction with conventional drugs. On the whole, further reducing the effectiveness of antiretroviral drugs, in vitro
research is required to prevent therapeutic failure and toxi- assessments of its effect on CYPs are conflicting [69, 70].
city in cancer patients when herbal medicines are concur-
rently taken with conventional medicines. This would aid in
establishing guidelines for their concomitant use.

3.2 HIV/AIDS

Around the world, the HIV epidemic rages on, emerging


as the greatest challenge to global health. However, AIDS-
related deaths are decreasing largely because of increased
access to treatment. Herbal medicines are widely used by
HIV patients to complement conventional therapy, with an
increasing number of studies investigating their use in HIV
management [63]. Common herbal medicines used during
HIV management include Allium sativum (garlic), Euca-
lyptus globulus (Blue gum), Aloe vera, Trigonostema
Fig. 3 Mean concentration-time profile of indinavir alone (solid line)
xyphophylloides, Vatica astrotricha, Vernonia amygdalina
and with concomitant St. John’s wort (dotted line), showing a large
(Bitterleaf), Lippia javanica (Fever tree), Bidens pilosa reduction in indinavir concentrations by concomitant St. John’s wort.
(Blackjack), Peltoforum africanum (Weeping wattle), In the study, fasting participants received indinavir 800 mg orally.
Hypoxis hemerocallidea (African star grass) and Moringa They then received two more doses at 8-h intervals to achieve steady-
state. On the morning of day 2, an 800 mg dose was given. On day 3,
oleifera (Drumstick tree) [16, 64, 65]. However, it is of
participants began treatment with St. John’s wort (300 mg three times
importance to thoroughly investigate the potential alter- daily) for 14 days. Piscitelli et al. [68]. (Reproduced with permission
ations in pharmacokinetic and toxicological profiles when from Elsevier)
Pharmacokinetic Herb-Drug Interactions

In a clinical study, garlic was shown to reduce the plasma are known to exhibit an antimalarial effect, antagonistic
pharmacokinetic concentrations of saquinavir by altering antimalarial properties when used in combination with
the CYP3A4 isoform of the CYP system, the isozyme artesunic acid in Plasmodium berghei-infected mice have
through which saquinavir is metabolized, recommending been reported [78]. In the study, the extracts of Carica
patient caution on concomitant administration [71]. papaya when solely administered had good antimalarial
Another study in healthy volunteers examining the effect of activity. In a similar study, the influence of Eurycoma
an odorless garlic product indicated an insignificant longifolia extract (commonly known as Tongkat ali, a
reduction in the AUC and plasma concentrations of riton- herbal remedy commonly used in malaria therapy) was
avir following short-term garlic consumption [72]. This is investigated on coadministration with artemisinin (WHO’s
thought to arise from transitory induction followed by recommended first-line antimalarial) in experimental mice.
inhibition on the various drug disposition pathways of Findings from that study revealed the suppression of par-
ritonavir, especially as the treatment duration of garlic asitemia in Plasmodium yoelii-infected mice. This is sug-
(twice daily for 4 days) was too short to observe a signif- gestive of a promising, potential antimalarial candidate by
icant change in ritonavir plasma concentrations [72]. This both oral and subcutaneous routes [79]. Similar synergistic
informs the need for caution, especially if large quanti- herb-drug interactions involving goniothalamin (Gonio-
ties of raw or processed garlic are concomitantly taken thalamus scortechinii)-chloroquine, Vernonia amyg-
with any protease inhibitor. Another study in healthy dalina—chloroquine and curcumin (isolated from
human subjects indicated a significant increase in the Curcuma longa)-artemisinin combinations have been doc-
bioavailability and maximum plasma concentration of umented with the suggestion that they be considered for
nevirapine following 6 days of piperine intake [73]. future trials in the search for malaria combination therapy
Piperine is a major component of Piper nigrum and Piper [80–82]. Also, herbs can be combined for their additive
longum (black pepper and long pepper, respectively) and effect as shown by the mixture of a Khaya ivorensis
has been reported to inhibit drug-metabolizing enzymes (African mahogany)-Alstonia boonei (English alstonia)
and increase the plasma concentrations of several drugs, extract mixture as an antimalarial prophylactic remedy
including P-gp substrates [74]. [83].

3.3 Malaria 3.4 Liver Diseases

Malaria is an important global public health issue with high Hepatic disorders (including alcoholic liver disease, hep-
morbidity and mortality rates. The majority of malarial atitis, cirrhosis and steatosis) are still a cause for global
endemic regions are from the world’s developing econo- concern [84]. Unfortunately, several orthodox drugs use-
mies. As a result of the relatively high costs for conven- d in the management of liver diseases are inade-
tional antimalarials, many patients are known to take quate and sometimes can have serious side effects.
herbal medicines for its prevention and treatment [75]. However, herbal medicines have been used in the treat-
Herbal medicines commonly used in the management of ment of liver diseases since ancient times. Silybum mari-
malaria include Vernonia amygdalina (Bitterleaf), Piper anum (milk thistle) is a widely used herbal remedy
longum (Long pepper), Tapinanthus sessilifolius (African especially for the management of liver and gallbladder
mistletoe), leaves of Carica papaya (Pawpaw), leaves and disorders [85]. Although preclinical evidence strongly
bark of Azadiractha indica (Neem) and rhizomes of Cur- supports its use as a hepatoprotectant, further well-de-
cuma longa (turmeric) [76]. Sometimes these herbs are signed clinical trials may be necessary to confirm this [86].
used alone or in combination with orthodox medicines. A In-depth research has been conducted on its most active
study based on an in vitro (Caco-2 cell), ex vivo (Ussing component, silymarin, a flavonoid complex. Silymarin and
chamber) and in vivo rat model revealed that extracts of its active constituent, silybin, are believed to act as
Vernonia amygdalina, Tapinanthus sessilifolius and Carica antioxidants, scavenging free radicals and inhibiting lipid
papaya, which are herbals commonly used in traditional peroxidation, thus finding use in chronic liver disorders
malaria, cancer and diabetes therapy, inhibited P-gp [87, 88]. It is believed to be safe and well tolerated, with
mediated digoxin transport [28, 77]. Findings from that very minor side effects reported when taken within the
study suggested that caution should be observed when recommended dose range (gastrointestinal upset, mild
those herbs are concomitantly used with P-gp substrate laxative effect and rare allergic reaction). Because of its
drugs as they may enhance their absorptive transport [28]. vast use, significant work has been carried out, examining
Frequently, antagonistic effects have been reported on the potential for herb-drug interactions. In a study by
coadministration of herbals and orthodox medicines. For Gurley et al. [89], insignificant changes in the disposition
instance, although the leaves of Carica papaya (Pawpaw) of digoxin were observed on coadministration with
E. F. Oga et al.

silymarin, posing no clinically significant risk for P-gp- administration with St. John’s wort has resulted in reduced
mediated herb-drug interactions. On the other hand, a study plasma concentration of theophylline [96]. However, in
has shown that silybin A and silybin B at clinically relevant another clinical study in healthy Japanese male volunteers,
concentrations inhibit CYP2C9-mediated metabolism of no significant alteration in the plasma pharmacokinetics of
warfarin [90]. This HDI needs to be further evaluated theophylline was observed when coadministered with St.
because of the narrow therapeutic index of warfarin. In John’s wort [97]. Likewise there are reports on herb-drug
addition, there are several reports on the hepatoprotective interactions involving fexofenadine, a well-known P-gp
function of glycyrrhizin (a major constituent of licorice). substrate probe that is commonly used in the management
Besides inhibiting liver cell injury caused by many chem- of allergy. A clinical study in healthy volunteers indicated
icals, it is also used in the treatment of chronic hepatitis and a significant increase in the maximum plasma concentra-
cirrhosis [91]. Glycyrrhizin has been shown to be a strong tion of fexofenadine and a significant decrease in its oral
inhibitor of 11-b-hydroxysteroid dehydrogenase (the clearance following the administration of a single dose of
enzyme responsible for catalyzing the conversion of cor- St. John’s wort. In this study, no change in the half-life or
tisol to the inactive steroid cortisone) and is thought to alter renal clearance was observed [98]. This observation was
the pharmacokinetics of prednisolone by inhibiting its also confirmed by another clinical study in healthy volun-
metabolism. In one study, oral glycyrrhizin increased the teers that demonstrated that pretreatment with St. John’s
plasma prednisolone concentrations in six healthy males wort significantly enhanced the oral clearance of fexofe-
[92]. nadine by 1.6-fold. Here also no alteration in the half-life
was observed. The authors suggested the predominant
3.5 Asthma and Allergic Diseases inductive effect of St. John’s wort on P-gp in the intestinal
epithelium, which consequently caused a decrease in the
There are several reports on the use of herbal medicines in absorbed fraction of oral fexofenadine [99]. As inhibition
the treatment and management of asthma and allergic and induction of P-gp may significantly influence drug
diseases (including allergic rhinitis, food allergy and atopic disposition, caution may need to be exercised in cases of
dermatitis), which affect a high percentage of the popula- their coadministration.
tion. Some of these phytomedicines are used alone, while
some others are used in combination with conventional 3.6 Depression
medicines. Corticosteroids are known to form a key com-
ponent in the management of asthma and allergy. A study Several herbs have been used in the management of
investigated the interaction potential of cortisol on con- depression. Of particular note is Hypericum perfora-
comitant administration with Glycyrrhiza glabra (licorice) tum (St. John’s wort), which has gained widespread pop-
and grapefruit juice. The findings showed significantly ularity as ‘‘nature’s Prozac.’’ It has been used for centuries
increased cortisol AUC and mean serum concentrations as a natural remedy for the treatment of a number of dis-
following the intake of licorice and grapefruit juice. eases [100]. Some clinical studies have provided evidence
Coadministration with grapefruit juice gave rise to a more that it is as effective as conventional antidepressants.
complete intestinal absorption of cortisol during the first Although the antidepressive mechanism is not fully
hours, indicative that interactions between various con- understood, its therapeutic effects have been confirmed in
stituents and P-gp in the intestinal walls are implicated several studies when compared with placebo or standard
[93]. One study examined the relationship between the use antidepressant agents [101]. Even though it is widely
of herbal medicines and adherence to inhaled corticos- obtained as an over-the-counter remedy, knowledge about
teroids. It was revealed that utilizing herbal medicines was the pharmacokinetics of ingredients and drug interactions
associated with lower adherence to inhaled corticosteroids of St. John’s wort is not commensurate. Because of its
and poor outcomes among asthmatic patients, probably ability to induce CYP3A4/P-gp, there are some reports on
because of patients’ worry over the adverse effects of the its interaction with other CYP3A/P-gp substrates leading to
corticosteroids [94]. Petasites hybridus (Butterbur), a pharmacokinetic interaction with drugs known to have
herbal remedy with antihistamine and anti-leukotriene narrow therapeutic windows (e.g. Digoxin), for which
activity, conferred complementary antiinflammatory therapeutic drug monitoring may be necessary [102].
activity in asthmatic patients who were receiving inhaled Because several drugs are cosubstrates of CYP3A and
corticosteroids, suggesting the potential benefit in asthma P-gp, their disposition is markedly affected by concomitant
management [95]. Although theophylline is commonly administration of St. John’s wort through an activation of
utilized in asthma therapy, its use complicated by its nuclear receptors, resulting in enhanced metabolism and
interaction with several other drugs and its narrow thera- efflux transport of the coadministered substrate drug to
peutic index. For instance, in a study its concomitant different extents depending on the relative contributions of
Pharmacokinetic Herb-Drug Interactions

CYP3A and P-gp. For instance a study reported that the sinesis) is a popular beverage and dietary supplement with
extent of induction measured by oral clearance was dif- some reported interactions with conventional medicines. A
ferent with CYP3A activity (midazolam, which is solely study in healthy subjects examined its interaction potential
metabolized by CYP3A), which showed more increase with buspirone (a CYP3A4 substrate). The extract con-
than P-gp function (fexofenadine, a nonmetabolized drug), taining 800 g epigallocatechin gallate was administered
whereas with cyclosporine (CYP3A and P-gp are both daily for 4 weeks and was shown to significantly increase
importantly involved in its disposition), the change in oral the bioavailability of buspirone [106]. Of note, however, is
clearance appeared to be more closely associated with the the fact that this CYP isozyme inhibition is unlikely to be
increase in MDR1 than with CYP3A [99]. In general, most of clinical relevance [106]. Another human study further
of these studies support the need for caution as well as revealed that a decaffeinated extract of green tea did not
stricter regulations of herbal medicines for safer drug alter the pharmacokinetics of either dextromethorphan or
therapy in medicine [103]. Panax ginseng is a widely used alprazolam, indicating the improbability of green tea to
herbal medicine, notably used for its antidepressant effect. alter the disposition of CYP2D6 or CYP3A4 substrates
A study investigated the influence of Panax ginseng on [107].
CYP3A function using the probe midazolam as a substrate
probe [104]. Comparison of pharmacokinetic parameter 3.7 Other Conditions
values of midazolam was calculated and compared before
and following the administration of Panax ginseng. As Herbal medicines are also widely used in the management
illustrated in Fig. 4, the findings revealed a significant colds, infections and inflammation. For instance, echinacea
reduction for the midazolam area under the concentration- is commonly used for the treatment of the common cold,
time curve from zero to infinity, half-life and maximum coughs, bronchitis, influenza, and inflammation of the mouth
concentration [104]. Also, American ginseng (Panax and pharynx, and it is one of the most sold herbal medicines
quinquefolius) reportedly reduces the effect of warfarin in in use, reportedly consumed by 10–20 % of herbal users
healthy patients. This was observed in a clinical study in [25]. Because of its immunostimulatory effect, it is also
healthy volunteers following ginseng consumption for commonly used for HIV and upper respiratory tract infec-
2 weeks in which changes in the international normalized tions [108, 109]. In a study investigating the influence of
ratio, peak plasma warfarin concentration and warfarin Echinacea purpurea on the in vivo activity of CYP iso-
AUC were found to be statistically significantly greater in zymes (including CYP3A4), after oral and intravenous
the group [105]. Here, the effect was postulated to be as a midazolam administration, modulation of the catalytic
result of the inductive effect of ginseng on the hepatic drug activity of CYP3A4 at both hepatic and intestinal sites were
metabolizing enzyme system, as warfarin is known to be reported. Inhibition of intestinal CYP3A4 activity was
metabolized by the CYP system. Green tea (from Camelia observed, with a potent increase in midazolam’s oral
bioavailability as well as significant enhancement of mida-
zolam’s systemic clearance by inducing hepatic CYP3A. In
addition, the study advises the exercise of caution when it is
coadministered with drugs that are dependent on CYP3A or
CYP1A2 for their elimination [25].
Similarly, studies investigating interactions of Ginkgo
biloba (Maidenhair tree)—a herbal remedy commonly
used for memory enhancement—have been reported. For
instance, a human study revealed a significant increase in
the bioavailability and peak plasma concentration of tali-
nolol as a result of P-gp efflux inhibition following long-
term administration of ginkgo [110, 111]. This effect was
suggested to be as a result of the P-gp inhibitory effect of
some ginkgo flavonol constituents, including quercetin,
kaempferol and isorhamnetin, which have been docu-
Fig. 4 Concentration-time profiles for midazolam (±SEM) before mented [112]. In another study, the effect of the extract of
and after Panax ginseng administration. In this study, the participants Ginkgo biloba on midazolam (CYP3A4 substrate) and
were administered a single 8-mg oral dose of midazolam syrup. They tolbutamide (CYP2C9 substrate) was investigated [113].
then began taking P. ginseng at a dose of 500 mg twice daily for
There, the AUC of tolbutamide following ginkgo intake
28 days. On day 28 of P. ginseng administration, participants
returned to the clinic for a repeat dose of midazolam. Malati et al. was significantly reduced. Conversely, the AUC of mida-
[104]. (Reproduced with permission from John Wiley & Sons, Inc) zolam was significantly enhanced, while its oral clearance
E. F. Oga et al.

was decreased. However, another study reported the not the case in many instances and has resulted in the
intestinal induction of CYP3A4 following midazolam emergence of toxic responses.
administration to healthy humans. After administration for Although the use of herbal products is rapidly increasing,
4 weeks, the bioavailability and maximum plasma con- there are only few national surveillance systems monitoring
centration of midazolam significantly reduced with no and evaluating adverse reactions associated with their use
change in the half-life suggestive of intestinal, but not [118]. In order to identify and assess the possible risks
hepatic, induction [113]. Another clinical study investi- associated with herbal medicine use, pharmacovigilance
gated the intake of the same dose of Ginkgo biloba extract studies are essential. For this, the establishment of phar-
after 2-week administration in combination with lopinavir, macovigilance centers would play a significant role in pro-
fexofenadine and midazolam [114]. The CYP3A4 induc- moting awareness of herbal medicine safety and the need to
tive effect of ginkgo was indicated by a significant decrease report the observance of adverse drug reactions [119].
in the AUC and maximum plasma concentration of mida- Pharmacovigilance encompasses the totality of monitoring
zolam. However, neither lopinavir nor ritonavir pharma- drug safety including identifying plausible adverse drug
cokinetic parameter values were significantly altered. This interactions, assessing risks and benefits as well as con-
is likely due to ritonavir’s more potent inhibition of veying concerns over drug safety [120]. It is essential to
CYP3A4. It was therefore suggested that Ginkgo biloba regulate herbal medicine use as well as ensure appropriate
extract may unlikely reduce the exposure of ritonavir- quality control measures including quality specification;
boosted protease inhibitors, while concentrations of good manufacturing practices for herbal medicines, labeling
unboosted protease inhibitors may be affected [114]. and licensing schemes for manufacturing, imports and
marketing should be enforced [121]. Also of importance is
the need to extend knowledge on drug safety rather than the
4 Drawbacks, Surveillance and Safety Monitoring general approach of demonstrating toxicity. With an
of Herbal Medicines increased awareness and increasing number of the populace
utilizing herbal medicines, more and more toxicological
Despite the fact that herbal medicines are widely used, the assessments need to be being conducted. This is expected to
safety and efficacy profile of several of them are in doubt include an investigation into the potential for genotoxicity,
and unproven. Moreover, many consumers misinterpret the reproductive toxicity, hepatotoxicity, nephrotoxicity and
natural origin of herbal medicines as a sign of safety, neurotoxicity of the phytomedicines [122, 123]. There are
without appreciating that herbal ingredients can cause recommendations concerning the need to strengthen exper-
serious adverse effects [115]. A major concern regarding tise in toxicology as well as determining newer approaches
the use of herbal medicines is their safety and toxicity toward solving the present toxicological issues [124]. For
profile. They may pose harm to patients under different example, the Caenorhabiditis elegans model has been pro-
circumstances through idiosyncratic or allergic reactions or ven to be a reliable and invaluable tool in toxicological
the risk of herb-drug interaction occurring when taken assessments, especially with respect to neurotoxic evalua-
concomitantly with conventional medicine. Because rig- tions. In addition to its sharing a high sequence identity with
orous testing and regulatory agency approval are not rou- several human genes as well as its anatomical and physio-
tinely considered for many herbal medicines, they may be logical characteristics, its genome has been fully sequenced
prone to easy adulteration or contamination, and thus and the nervous system has been extensively studied,
harmful. Worldwide, several instances of toxicity have thereby making it of immense benefit in toxicological
been reported on using herbal medicines. Of significance is assessments [125].
the development of kidney failure by several women in
Belgium after taking slimming pills containing the
herb Aristolochic fangchi. This further resulted in transi- 5 Conclusion
tional cell carcinoma in some of these patients [116]. In
addition, standardization of herbal medicines sometimes The use of herbal medicines is increasing, possibly because
becomes challenging as their chemical makeup differs of their widespread promotion in the media as well as
depending on the part of the plant used, growing condi- unsubstantiated health care claims. Although herbal
tions, periods of harvest as well as storage conditions. medicines are beneficial, despite popular belief, they are
Combination products composed of multiple natural not completely harmless, and HDIs may occur on con-
products complicate matters further. In using herbal comitant use with conventional drugs, but many possibly
medicines, standardization, regulation and the scientific go unnoticed because of various factors. It is safer to view
proof of patients’ safety are of paramount importance, them as unrefined pharmaceuticals, capable of producing
especially in the development phase [117]. However, this is physiologic change, for better or worse.
Pharmacokinetic Herb-Drug Interactions

Most herbal medicines, unlike conventional drugs, 2. Calixto JB. Efficacy, safety, quality control, marketing and
comprise a complex mixture of chemical constituents. In regulatory guidelines for herbal medicines (phytotherapeutic
agents). Braz J Med Biol Res. 2000;33(2):179–89.
order to better understand the detection and handling of 3. Mahady GB. Global harmonization of herbal health claims.
HDIs, an expansion of knowledge on phytochemicals in J Nutr. 2001;131(3s):1120S–3S.
herbal medicines is essential. Presently, in most cases, 4. World Health Organization. Traditional medicine. 2008.
complete characterization of the bioactive compounds is 5. Tuffs A. Three out of four Germans have used complementary
or natural remedies. BMJ. 2002;325(7371):990.
not well defined, and information on toxicity and adverse 6. Tachjian A, Maria V, Jahangir A. Use of herbal products and
effects are insufficient. It is expected that standardization, potential interactions in patients with cardiovascular diseases.
including chemical fingerprinting, would become potent J Am Coll Cardiol. 2010;55(6):515–25.
tools for quality control of herbal medicines. However, 7. Komiya A, Watanabe A, Fuse H. Herbal medicine in Japan.
J Men’s Health. 2011;8(1):S15–8.
because most of the available HDI information is based on 8. Moschik EC, et al. Usage and attitudes of physicians in Japan
individual case reports, animal studies and in vitro data, concerning traditional Japanese medicine (kampo medicine): a
extensive research is required to confirm and assess the descriptive evaluation of a representative questionnaire-based
clinical significance of these potential interactions. HDIs survey. Evid Based Complement Altern Med. 2012;2012:139818.
9. Zhang X. Regulatory situation of herbal medicines: a worldwide
may be underreported, and appropriate pharmacovigilance review. WHO Traditional Medicine Programme; 1998. p. 45.
requires the collective responsibility of the patient, health 10. Walker DR. Report on the regulation of herbal medicines and
practitioner and researchers. practitioners. 2015.
In this review, the pharmacokinetic (drug-metabolizing 11. Boyle SP, et al. Evaluation of quality control strategies in Scutel-
laria herbal medicines. J Pharm Biomed Anal. 2011;54(5):951–7.
enzymes and drug transporter systems) mechanisms have 12. Newman DJ, Cragg GM. Natural products as sources of new
been considered to play a role in these interactions. There drugs over the 30 years from 1981 to 2010. J Nat Prod.
are reports on the inductive role of PXR as a nuclear 2012;75(3):311–35.
receptor; however, it is of importance to research other 13. Lahlou M. The success of natural products in drug discovery.
Pharmacol Pharm. 2013;4:17–31.
receptors, constitutive androstane receptor (CAR), and 14. Katiyar C, et al. Drug discovery from plant sources: an inte-
vitamin D-binding receptor (VDR), as these may also play grated approach. Ayu. 2012;33(1):10–9.
an integral role in the mechanism of inductive processes 15. Giacomini KM, et al. Membrane transporters in drug develop-
involved in HDIs. Future perspectives for the application ment. Nat Rev Drug Discov. 2010;9(3):215–36.
16. Muller AC, Kanfer I. Potential pharmacokinetic interactions
of HDIs are in new drug development and use of herbal between antiretrovirals and medicinal plants used as comple-
medicines as adjuvants to conventional drugs. They may mentary and African traditional medicines. Biopharm Drug
also be used for their additive or synergistic effect when Dispos. 2011;32(8):458–70.
coadministered with modern medicine. This may require a 17. Guengerich FP. Cytochrome P450 s and other enzymes in drug
metabolism and toxicity. AAPS J. 2006;8(1):E101–11.
decreased dose of the conventional drug and possibly result 18. Paine MF, et al. The human intestinal cytochrome P450 ‘‘pie’’.
in a reduction in the manifestation of side effects. In par- Drug Metab Dispos. 2006;34(5):880–6.
ticular, patients taking drugs with a narrow therapeutic 19. Kim E, et al. Isolation and identification of intestinal CYP3A
index should be discouraged from using herbal products inhibitors from cranberry (Vaccinium macrocarpon) using
human intestinal microsomes. Planta Med. 2011;77(3):265–70.
because of the ease of toxicity or ineffectiveness. 20. Jepson RG, Williams G, Craig JC. Cranberries for preventing
In summary, HDIs certainly occur and may have serious urinary tract infections. Cochrane Database Syst Rev.
consequences. However because they are under-re- 2012;10:CD001321.
searched, the present knowledge is incomplete. It is worth 21. Uesawa Y, Mohri K. Effects of cranberry juice on nifedipine
pharmacokinetics in rats. J Pharm Pharmacol.
mentioning that further research and more controlled 2006;58(8):1067–72.
clinical studies are needed to clarify and determine the 22. Lilja JJ, Backman JT, Neuvonen PJ. Effects of daily ingestion of
underlying mechanisms for these altered drug effects. cranberry juice on the pharmacokinetics of warfarin, tizanidine,
and midazolam–probes of CYP2C9, CYP1A2, and CYP3A4.
Compliance with Ethical Standards Clin Pharmacol Ther. 2007;81(6):833–9.
23. Ngo N, et al. Identification of a cranberry juice product that
Funding No financial support was received for conducting this inhibits enteric CYP3A-mediated first-pass metabolism in
study. humans. Drug Metab Dispos. 2009;37(3):514–22.
24. Penzak SR, et al. Echinacea purpurea significantly induces cyto-
Conflict of interest The authors report no competing interests. chrome P450 3A activity but does not alter lopinavir-ritonavir
exposure in healthy subjects. Pharmacotherapy. 2010;30(8):797–805.
25. Gorski JC, et al. The effect of echinacea (Echinacea purpurea
root) on cytochrome P450 activity in vivo. Clin Pharmacol Ther.
References 2004;75(1):89–100
26. Hansen TS, Nilsen OG. In vitro CYP3A4 metabolism: inhibition
1. Sasidharan S, et al. Extraction, isolation and characterization of by Echinacea purpurea and choice of substrate for the evaluation
bioactive compounds from plants’ extracts. Afr J Tradit Com- of herbal inhibition. Basic Clin Pharmacol Toxicol.
plement Altern Med. 2011;8(1):1–10. 2008;103(5):445–9.
E. F. Oga et al.

27. Eichhorn T, Efferth T. P-glycoprotein and its inhibition in 48. Tien ES, Negishi M. Nuclear receptors CAR and PXR in the regu-
tumors by phytochemicals derived from Chinese herbs. lation of hepatic metabolism. Xenobiotica. 2006;36(10–11):1152–63.
J Ethnopharmacol. 2012;141(2):557–70. 49. Meijerman I, Beijnen JH, Schellens JH. Herb-drug interactions
28. Oga EF, et al. P-glycoprotein mediated efflux in Caco-2 cell in oncology: focus on mechanisms of induction. Oncologist.
monolayers: the influence of herbals on digoxin transport. 2006;11(7):742–52.
J Ethnopharmacol. 2012;144(3):612–7. 50. Gratus C, et al. The use of herbal medicines by people with
29. Li Y, Lu J, Paxton JW. The role of ABC and SLC transporters in cancer: a qualitative study. BMC Complement Altern Med.
the pharmacokinetics of dietary and herbal phytochemicals and 2009;9:14.
their interactions with xenobiotics. Curr Drug Metab. 51. He SM, et al. Effects of herbal products on the metabolism and
2012;13(5):624–39. transport of anticancer agents. Expert Opin Drug Metab Toxicol.
30. Hermann R, von Richter O. Clinical evidence of herbal drugs as 2010;6(10):1195–213.
perpetrators of pharmacokinetic drug interactions. Planta Med. 52. Fong HH. Integration of herbal medicine into modern medical
2012;78(13):1458–77. practices: issues and prospects. Integr Cancer Ther.
31. Dahan A, Altman H. Food-drug interaction: grapefruit juice 2002;1(3):287–93 (discussion 293).
augments drug bioavailability–mechanism, extent and rele- 53. Smith P, et al. The influence of St. John’s wort on the phar-
vance. Eur J Clin Nutr. 2004;58(1):1–9. macokinetics and protein binding of imatinib mesylate. Phar-
32. Bailey DG, et al. Grapefruit juice-drug interactions. Br J Clin macotherapy. 2004;24(11):1508–14.
Pharmacol. 1998;46(2):101–10. 54. Frye RF, et al. Effect of St John’s wort on imatinib mesylate
33. Bailey DG. Fruit juice inhibition of uptake transport: a new type pharmacokinetics. Clin Pharmacol Ther. 2004;76(4):323–9.
of food-drug interaction. Br J Clin Pharmacol. 55. Hu ZP, et al. St. John’s wort attenuates irinotecan-induced
2010;70(5):645–55. diarrhea via down-regulation of intestinal pro-inflammatory
34. Dresser GK, Bailey DG. The effects of fruit juices on drug cytokines and inhibition of intestinal epithelial apoptosis. Tox-
disposition: a new model for drug interactions. Eur J Clin Invest. icol Appl Pharmacol. 2006;216(2):225–37.
2003;33(Suppl 2):10–6. 56. Mathijssen RH, et al. Effects of St. John’s wort on irinotecan
35. Fan L, et al. The effect of herbal medicine baicalin on phar- metabolism. J Natl Cancer Inst. 2002;94(16):1247–9.
macokinetics of rosuvastatin, substrate of organic anion-trans- 57. Reif S, et al. Effect of grapefruit juice intake on etoposide
porting polypeptide 1B1. Clin Pharmacol Ther. bioavailability. Eur J Clin Pharmacol. 2002;58(7):491–4.
2008;83(3):471–6. 58. Hasegawa H, et al. Reversal of daunomycin and vinblastine
36. Zhao D, et al. Influence of rifampicin on the pharmacokinetics resistance in multidrug-resistant P388 leukemia in vitro through
of salvianolic acid B may involve inhibition of organic anion enhanced cytotoxicity by triterpenoids. Planta Med.
transporting polypeptide (Oatp) mediated influx. Phytother Res. 1995;61(5):409–13.
2012;26(1):118–21. 59. Kim SW, et al. Reversal of P-glycoprotein-mediated multidrug
37. Lin CC, et al. Evaluation of chinese-herbal-medicine-induced resistance by ginsenoside Rg(3). Biochem Pharmacol.
herb-drug interactions: focusing on organic anion transporter 1. 2003;65(1):75–82.
Evid Based Complement Altern Med. 2012;2012:967182. 60. Zhou S, Lim LY, Chowbay B. Herbal modulation of P-glyco-
38. Christians U, Schmitz V, Haschke M. Functional interactions protein. Drug Metab Rev. 2004;36(1):57–104.
between P-glycoprotein and CYP3A in drug metabolism. Expert 61. Hiraganahalli BD, et al. Hepatoprotective and antioxidant
Opin Drug Metab Toxicol. 2005;1(4):641–54. activity of standardized herbal extracts. Pharmacogn Mag.
39. Hellum BH, et al. Potent in vitro inhibition of CYP3A4 and 2012;8(30):116–23.
P-glycoprotein by Rhodiola rosea. Planta Med. 62. Olaku O, White JD. Herbal therapy use by cancer patients: a
2010;76(4):331–8. literature review on case reports. Eur J Cancer.
40. Rahimi R, Abdollahi M. An update on the ability of St. John’s 2011;47(4):508–14.
wort to affect the metabolism of other drugs. Expert Opin Drug 63. Lee SA, et al. Anti-HIV-1 efficacy of extracts from medicinal
Metab Toxicol. 2012;8(6):691–708. plants. J Microbiol. 2010;48(2):249–52.
41. Yu CP, et al. Quercetin and rutin reduced the bioavailability of 64. Liu J. The use of herbal medicines in early drug development for
cyclosporine from Neoral, an immunosuppressant, through the treatment of HIV infections and AIDS. Expert Opin Investig
activating P-glycoprotein and CYP 3A4. J Agric Food Chem. Drugs. 2007;16(9):1355–64.
2011;59(9):4644–8. 65. Bepe N, et al. The impact of herbal remedies on adverse effects
42. Markowitz JS, et al. Effect of St John’s wort on drug metabo- and quality of life in HIV-infected individuals on antiretroviral
lism by induction of cytochrome P450 3A4 enzyme. JAMA. therapy. J Infect Dev Ctries. 2011;5(1):48–53.
2003;290(11):1500–4. 66. Muller AC, et al. Interactions between phytochemical compo-
43. Willson TM, Kliewer SA. PXR, CAR and drug metabolism. Nat nents of Sutherlandia frutescens and the antiretroviral, ataza-
Rev Drug Discov. 2002;1(4):259–66. navir in vitro: implications for absorption and metabolism.
44. Lehmann JM, et al. The human orphan nuclear receptor PXR is J Pharm Pharm Sci. 2012;15(2):221–33.
activated by compounds that regulate CYP3A4 gene expression 67. Patel J, et al. In vitro interaction of the HIV protease inhibitor
and cause drug interactions. J Clin Invest. 1998;102(5):1016–23. ritonavir with herbal constituents: changes in P-gp and CYP3A4
45. Kliewer SA, Goodwin B, Willson TM. The nuclear pregnane X activity. Am J Ther. 2004;11(4):262–77.
receptor: a key regulator of xenobiotic metabolism. Endocr Rev. 68. Piscitelli SC, et al. Indinavir concentrations and St John’s wort.
2002;23(5):687–702. Lancet. 2000;355(9203):547–8.
46. Moore LB, et al. St. John’s wort induces hepatic drug metabo- 69. Foster BC, et al. An in vitro evaluation of human cytochrome
lism through activation of the pregnane X receptor. Proc Natl P450 3A4 and P-glycoprotein inhibition by garlic. J Pharm
Acad Sci. 2000;97(13):7500–2. Pharm Sci. 2001;4(2):176–84.
47. Geick A, Eichelbaum M, Burk O. Nuclear receptor response 70. Hajda J, et al. Garlic extract induces intestinal P-glycoprotein,
elements mediate induction of intestinal MDR1 by rifampin. but exhibits no effect on intestinal and hepatic CYP3A4 in
J Biol Chem. 2001;276(18):14581–7. humans. Eur J Pharm Sci. 2010;41(5):729–35.
Pharmacokinetic Herb-Drug Interactions

71. Piscitelli SC, et al. The effect of garlic supplements on the 91. Kumada H. Long-term treatment of chronic hepatitis C with
pharmacokinetics of saquinavir. Clin Infect Dis. glycyrrhizin [stronger neo-minophagen C (SNMC)] for pre-
2002;34(2):234–8. venting liver cirrhosis and hepatocellular carcinoma. Oncology.
72. Gallicano K, Foster B, Choudhri S. Effect of short-term 2002;62(Suppl 1):94–100.
administration of garlic supplements on single-dose ritonavir 92. Chen MF, et al. Effect of oral administration of glycyrrhizin on
pharmacokinetics in healthy volunteers. Br J Clin Pharmacol. the pharmacokinetics of prednisolone. Endocrinol Jpn.
2003;55(2):199–202. 1991;38(2):167–74.
73. Kasibhatta R, Naidu MU. Influence of piperine on the pharma- 93. Methlie P, et al. Grapefruit juice and licorice increase cortisol
cokinetics of nevirapine under fasting conditions: a randomised, availability in patients with Addison’s disease. Eur J Endocrinol.
crossover, placebo-controlled study. Drugs R D. 2011;165(5):761–9.
2007;8(6):383–91. 94. Roy A, et al. Use of herbal remedies and adherence to inhaled
74. Bhardwaj RK, et al. Piperine, a major constituent of black corticosteroids among inner-city asthmatic patients. Ann
pepper, inhibits human P-glycoprotein and CYP3A4. J Pharma- Allergy Asthma Immunol. 2010;104(2):132–8.
col Exp Ther. 2002;302(2):645–50. 95. Lee DK, et al. Butterbur, a herbal remedy, confers comple-
75. Asase A, Akwetey GA, Achel DG. Ethnopharmacological use of mentary anti-inflammatory activity in asthmatic patients
herbal remedies for the treatment of malaria in the Dangme receiving inhaled corticosteroids. Clin Exp Allergy.
West District of Ghana. J Ethnopharmacol. 2010;129(3):367–76. 2004;34(1):110–4.
76. Wells TN. Natural products as starting points for future anti- 96. Nebel A, et al. Potential metabolic interaction between St.
malarial therapies: going back to our roots? Malar J. John’s wort and theophylline. Ann Pharmacother.
2011;10(Suppl 1):S3. 1999;33(4):502.
77. Oga EF, Sekine S, Horie T. Ex vivo and in vivo investigations of 97. Morimoto T, et al. Effect of St. John’s wort on the pharma-
the effects of extracts of Vernonia amygdalina, Carica papaya cokinetics of theophylline in healthy volunteers. J Clin Phar-
and Tapinanthus sessilifolius on digoxin transport and pharma- macol. 2004;44(1):95–101.
cokinetics: assessing the significance on rat intestinal P-glyco- 98. Wang Z, et al. Effect of St John’s wort on the pharmacokinetics
protein efflux. Drug Metab Pharmacokinet. 2013;28(4):314–20. of fexofenadine. Clin Pharmacol Ther. 2002;71(6):414–20.
78. Onaku LO, et al. Antagonistic antimalarial properties of paw- 99. Dresser GK, et al. Coordinate induction of both cytochrome
paw leaf aqueous extract in combination with artesunic acid in P4503A and MDR1 by St John’s wort in healthy subjects. Clin
Plasmodium berghei-infected mice. J Vector Borne Dis. Pharmacol Ther. 2003;73(1):41–50.
2011;48(2):96–100. 100. Shelton RC. St John’s wort (Hypericum perforatum) in major
79. Mohd Ridzuan MA, et al. Eurycoma longifolia extract-artemi- depression. J Clin Psychiatry. 2009;70(Suppl 5):23–7.
sinin combination: parasitemia suppression of Plasmodium 101. Linde K, et al. St John’s wort for depression–an overview and
yoelii-infected mice. Trop Biomed. 2007;24(1):111–8. meta-analysis of randomised clinical trials. BMJ.
80. Mohd Ridzuan MA, et al. Antimalarial properties of Gonio- 1996;313(7052):253–8.
thalamin in combination with chloroquine against Plasmodium 102. Johne A, et al. Pharmacokinetic interaction of digoxin with an
yoelii and Plasmodium berghei growth in mice. Trop Biomed. herbal extract from St John’s wort (Hypericum perforatum). Clin
2006;23(2):140–6. Pharmacol Ther. 1999;66(4):338–45.
81. Nandakumar DN, et al. Curcumin-artemisinin combination 103. Durr D, et al. St John’s Wort induces intestinal P-glycoprotein/
therapy for malaria. Antimicrob Agents Chemother. MDR1 and intestinal and hepatic CYP3A4. Clin Pharmacol
2006;50(5):1859–60. Ther. 2000;68(6):598–604.
82. Iwalokun BA. Enhanced antimalarial effects of chloroquine by 104. Malati CY, et al. Influence of Panax ginseng on cytochrome
aqueous Vernonia amygdalina leaf extract in mice infected with P450 (CYP)3A and P-glycoprotein (P-gp) activity in healthy
chloroquine resistant and sensitive Plasmodium berghei strains. participants. J Clin Pharmacol. 2012;52(6):932–9.
Afr Health Sci. 2008;8(1):25–35. 105. Yuan CS, et al. Brief communication: American ginseng reduces
83. Tepongning RN, et al. Potential of a Khaya ivorensis—Alstonia warfarin’s effect in healthy patients: a randomized, controlled
boonei extract combination as antimalarial prophylactic remedy. Trial. Ann Intern Med. 2004;141(1):23–7.
J Ethnopharmacol. 2011;137(1):743–51. 106. Chow HH, et al. Effects of repeated green tea catechin admin-
84. Kim WR, et al. Burden of liver disease in the United States: istration on human cytochrome P450 activity. Cancer Epidemiol
summary of a workshop. Hepatology. 2002;36(1):227–42. Biomark Prev. 2006;15(12):2473–6.
85. Verma S, Thuluvath PJ. Complementary and alternative medi- 107. Donovan JL, et al. Green tea (Camellia sinensis) extract does
cine in hepatology: review of the evidence of efficacy. Clin not alter cytochrome p450 3A4 or 2D6 activity in healthy vol-
Gastroenterol Hepatol. 2007;5(4):408–16. unteers. Drug Metab Dispos. 2004;32(9):906–8.
86. Giese LA. Milk thistle and the treatment of hepatitis. Gas- 108. Molto J, et al. Herb-drug interaction between Echinacea pur-
troenterol Nurs. 2001;24(2):95–7. purea and darunavir-ritonavir in HIV-infected patients.
87. Flora K, et al. Milk thistle (Silybum marianum) for the therapy Antimicrob Agents Chemother. 2011;55(1):326–30.
of liver disease. Am J Gastroenterol. 1998;93(2):139–43. 109. Barnes J, et al. Echinacea species (Echinacea angustifolia (DC.)
88. Tamayo C, Diamond S. Review of clinical trials evaluating Hell., Echinacea pallida (Nutt.) Nutt., Echinacea purpurea (L.)
safety and efficacy of milk thistle (Silybum marianum [L.] Moench): a review of their chemistry, pharmacology and clini-
Gaertn.). Integr Cancer Ther. 2007;6(2):146–57. cal properties. J Pharm Pharmacol. 2005;57(8):929–54.
89. Gurley BJ, et al. Effect of milk thistle (Silybum marianum) and 110. Fan L, et al. Effects of Ginkgo biloba extract ingestion on the
black cohosh (Cimicifuga racemosa) supplementation on pharmacokinetics of talinolol in healthy Chinese volunteers.
digoxin pharmacokinetics in humans. Drug Metab Dispos. Ann Pharmacother. 2009;43(5):944–9.
2006;34(1):69–74. 111. Fan L, et al. Effect of Schisandra chinensis extract and Ginkgo
90. Brantley SJ, et al. Two flavonolignans from milk thistle (Sily- biloba extract on the pharmacokinetics of talinolol in healthy
bum marianum) inhibit CYP2C9-mediated warfarin metabolism volunteers. Xenobiotica. 2009;39(3):249–54.
at clinically achievable concentrations. J Pharmacol Exp Ther. 112. Wang Y, Cao J, Zeng S. Involvement of P-glycoprotein in
2010;332(3):1081–7. regulating cellular levels of Ginkgo flavonols: quercetin,
E. F. Oga et al.

kaempferol, and isorhamnetin. J Pharm Pharmacol. 132. Gurley BJ, et al. In vivo effects of goldenseal, kava kava, black
2005;57(6):751–8. cohosh, and valerian on human cytochrome P450 1A2, 2D6,
113. Uchida S, et al. Effects of Ginkgo biloba extract on pharma- 2E1, and 3A4/5 phenotypes. Clin Pharmacol Ther.
cokinetics and pharmacodynamics of tolbutamide and midazo- 2005;77(5):415–26.
lam in healthy volunteers. J Clin Pharmacol. 133. Juan H, et al. Unexpected effect of concomitantly administered
2006;46(11):1290–8. curcumin on the pharmacokinetics of talinolol in healthy Chi-
114. Robertson SM, et al. Effect of Ginkgo biloba extract on lopi- nese volunteers. Eur J Clin Pharmacol. 2007;63(7):663–8.
navir, midazolam and fexofenadine pharmacokinetics in healthy 134. Chen XW, et al. Clinical herbal interactions with conventional
subjects. Curr Med Res Opin. 2008;24(2):591–9. drugs: from molecules to maladies. Curr Med Chem.
115. Singh D, Gupta R, Saraf SA. Herbs-are they safe enough? an 2011;18(31):4836–50.
overview. Crit Rev Food Sci Nutr. 2012;52(10):876–98. 135. Glaeser H, et al. Intestinal drug transporter expression and the
116. Debelle FD, Vanherweghem JL, Nortier JL. Aristolochic acid impact of grapefruit juice in humans. Clin Pharmacol Ther.
nephropathy: a worldwide problem. Kidney Int. 2007;81(3):362–70.
2008;74(2):158–69. 136. Dresser GK, et al. Fruit juices inhibit organic anion transporting
117. Lietman PS. Herbal medicine development: a plea for a rigorous polypeptide-mediated drug uptake to decrease the oral avail-
scientific foundation. Am J Ther. 2012;19(5):351–6. ability of fexofenadine. Clin Pharmacol Ther.
118. Shetti S, et al. Pharmacovigilance of herbal medicines: current 2002;71(1):11–20.
state and future directions. Pharmacogn Mag. 2011;7(25):69–73. 137. Henderson L, et al. St John’s wort (Hypericum perforatum):
119. Skalli S, Soulaymani R. Safety monitoring of herb-drug inter- drug interactions and clinical outcomes. Br J Clin Pharmacol.
actions: a component of pharmacovigilance. Drug Saf. 2002;54(4):349–56.
2012;35(10):785–91. 138. Chung KF, et al. Effect of a ginkgolide mixture (BN 52063) in
120. Barnes J. Pharmacovigilance of herbal medicines : a UK per- antagonising skin and platelet responses to platelet activating
spective. Drug Saf. 2003;26(12):829–51. factor in man. Lancet. 1987;1(8527):248–51.
121. World Health Organization. The Importance of Pharmacovigi- 139. Rowin J, Lewis SL. Spontaneous bilateral subdural hematomas
lance—Safety Monitoring of Medicinal Products. Uppsala: associated with chronic Ginkgo biloba ingestion. Neurology.
Uppsala Monitoring Centre, WHO Collaborating Centre for 1996;46(6):1775–6.
International Drug Monitoring; 2002. p. 52. 140. Meisel C, Johne A, Roots I. Fatal intracerebral mass bleeding
122. Wang CC, et al. Safety evaluation of commonly used Chinese associated with Ginkgo biloba and ibuprofen. Atherosclerosis.
herbal medicines during pregnancy in mice. Hum Reprod. 2003;167(2):367.
2012;27(8):2448–56. 141. Yue QY, Bergquist C, Gerden B. Safety of St John’s wort
123. Steenkamp V, Stewart MJ. Nephrotoxicity associated with (Hypericum perforatum). Lancet. 2000;355(9203):576–7.
exposure to plant toxins, with particular reference to Africa. 142. Bauer S, et al. Alterations in cyclosporin A pharmacokinetics
Ther Drug Monit. 2005;27(3):270–7. and metabolism during treatment with St John’s wort in renal
124. Gulumian M, Savolainen K. Toxicological issues in developed transplant patients. Br J Clin Pharmacol. 2003;55(2):203–11.
and developing countries: the difference is in approach and not 143. Mai I, et al. Impact of St John’s wort treatment on the phar-
in content. Hum Exp Toxicol. 2012;31(3):205–6. macokinetics of tacrolimus and mycophenolic acid in renal
125. Avila D, Helmcke K, Aschner M. The Caenorhabiditis elegans transplant patients. Nephrol Dial Transplant.
model as a reliable tool in neurotoxicology. Hum Exp Toxicol. 2003;18(4):819–22.
2012;31(3):236–43. 144. Alemdaroglu NC, et al. Influence of green and black tea on folic
126. Melchart D, et al. Polysaccharides isolated from Echinacea acid pharmacokinetics in healthy volunteers: potential risk of
purpurea herbal cell cultures to counteract undesired effects of diminished folic acid bioavailability. Biopharm Drug Dispos.
chemotherapy—a pilot study. Phytother Res. 2008;29(6):335–48.
2002;16(2):138–42. 145. Tuntipopipat S, et al. Chili, but not turmeric, inhibits iron
127. Bossaer JB, Odle BL. Probable etoposide interaction with absorption in young women from an iron-fortified composite
Echinacea. J Diet Suppl. 2012;9(2):90–5. meal. J Nutr. 2006;136(12):2970–4.
128. de Maat MM, et al. Drug interaction between St John’s wort and 146. Di Marco MP, et al. The effect of grapefruit juice and seville
nevirapine. Aids. 2001;15(3):420–1. orange juice on the pharmacokinetics of dextromethorphan: the
129. Holodniy M, et al. A double blind, randomized, placebo-con- role of gut CYP3A and P-glycoprotein. Life Sci.
trolled phase II study to assess the safety and efficacy of orally 2002;71(10):1149–60.
administered SP-303 for the symptomatic treatment of diarrhea 147. Hall SD, et al. The interaction between St John’s wort and an
in patients with AIDS. Am J Gastroenterol. oral contraceptive. Clin Pharmacol Ther. 2003;74(6):525–35.
1999;94(11):3267–73. 148. Yin OQ, et al. Pharmacogenetics and herb-drug interactions:
130. Smith M, Lin K, Zheng Y. An open trial of nifedipine-herb experience with Ginkgo biloba and omeprazole. Pharmacoge-
interactions: nifedipine with St. John’s wort, ginseng or Ginkgo netics. 2004;14(12):841–50.
biloba. Clin Pharmacol Ther. 2001;69(2):86.
131. Sugimoto K, et al. Different effects of St John’s wort on the
pharmacokinetics of simvastatin and pravastatin. Clin Pharma-
col Ther. 2001;70(6):518–24.

You might also like