+ All Categories
Home > Documents > Hypericum perforatum : Pharmacokinetic, Mechanism of Action, Tolerability, and Clinical Drug-Drug...

Hypericum perforatum : Pharmacokinetic, Mechanism of Action, Tolerability, and Clinical Drug-Drug...

Date post: 26-Nov-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
13
REVIEW Hypericum perforatum: Pharmacokinetic, Mechanism of Action, Tolerability, and Clinical DrugDrug Interactions Emilio Russo, 1,2 * Francesca Scicchitano, 1,2 Benjamin J. Whalley, 3 Carmela Mazzitello, 1,2 Miriam Ciriaco, 1,2 Stefania Esposito, 1,2 Marinella Patanè, 1,2 Roy Upton, 4 Michela Pugliese, 5 Serafina Chimirri, 1,2 Maria Mammì, 1,2 Caterina Palleria 1,2 and Giovambattista De Sarro 1,2 1 Science of Health Department, School of Medicine, University of Catanzaro, Catanzaro, Italy 2 Pharmacovigilances Center Region Calabria, University Hospital Mater Domini, Catanzaro, Italy 3 School of Chemistry, Food and Nutritional Sciences, and Pharmacy, The University of Reading, Whiteknights, Reading, Berkshire RG6 6AP, UK 4 American Herbal Pharmacopoeia, Scotts Valley, CA, USA 5 Department of Veterinary Science, University of Messina, Messina, Italy Hypericum perforatum (HP) belongs to the Hypericaceae family and is one of the oldest used and most extensively investigated medicinal herbs. The medicinal form comprises the leaves and flowering tops of which the primary ingredients of interest are naphthodianthrones, xanthones, flavonoids, phloroglucinols (e.g. hyperforin), and hypericin. Although several constituents elicit pharmacological effects that are consistent with HPs antidepres- sant activity, no single mechanism of action underlying these effects has thus far been found. Various clinical trials have shown that HP has a comparable antidepressant efficacy as some currently used antidepressant drugs in the treatment of mild/moderate depression. Interestingly, low-hyperforin-content preparations are effective in the treatment of depression. Moreover, HP is also used to treat certain forms of anxiety. However, HP can induce various cytochrome P450s isozymes and/or P-glycoprotein, of which many drugs are substrates and which are the main origin of HPdrug interactions. Here, we analyse the existing evidence describing the clinical consequence of HPdrug interactions. Although some of the reported interactions are based on findings from in vitro studies, the clinical importance of which remain to be demonstrated, others are based on case reports where causality can, in some cases, be determined to reveal clinically significant interactions that suggest caution, consideration, and disclosure of potential interactions prior to informed use of HP. Copyright © 2013 John Wiley & Sons, Ltd. Keywords: Hypericum perforatum; St. Johns wort; drugs; interaction; cytochrome P450; P-glycoprotein. INTRODUCTION A growing percentage of the population uses herbal products for preventative and therapeutic purposes. Several reasons for this wide usage have been proposed and include (i) the erroneous notion that, being of natural origin, herbals are safer than conventional med- icines; (ii) the philosophical belief that naturally derived drugs are more consistent with individual personal values; (iii) individualsdesire for personal control over their health; and (iv) hard-to-treat conditions for which conventional therapies are unavailable (Cheung et al., 2007; Cauffield, 2000). However, such lay beliefs belie the evidence that herbal drugs cause significant side effects either individually or due to herbdrug interactions (Bent, 2008; De Smet, 2007; Ernst, 1998). Hypericum perforatum (HP), or St Johns wort, is a plant whose extract, one of the best- characterised herbal medicines, is widely sold in health food stores and pharmacies in Europe and the USA (annual US sales increased from $20m in 1995 to $200m in 1997) for the treatment of depression (Lecrubier et al., 2002; Kasper et al., 2006). Preclinical studies on the central nervous system activity of HP extracts suggest that it also displays anxiolytic, sedative, nootropic, antischizophrenic, anti- convulsant, antidiabetic, and analgesic activities and that it may be beneficial in the treatment of alcohol, nicotine, and caffeine addiction in experimental animals (Can et al., 2011; Can and Ozkay, 2012). Recent clinical studies found HP extracts to be efficacious and well tolerated in the treatment of mild to moderate depressive episodes and for the short-term treatment of symptoms in mild depressive disorders (Chen et al., 2011; Kasper et al., 2010). Moreover, HP extracts can be effective in the treatment of generalised anxiety disorder, somatoform disorders, sleep disorders, obsessivecompulsive disorder, and seasonal affective disorder (Can et al., 2009). Although >150 discrete ingredients that exhibit many additive, synergistic, and partly antagonistic effects have thus far been identified in this plant (Butterweck and Schmidt, 2007), HPs most characteristic constituents are naphthodianthrones (hypericin and pseudohypericin) and phloroglucinols (hyperforin and adhyperforin) (Butterweck and Schmidt, 2007; Nahrstedt and Butterweck, 1997), where hypericin is considered to be the most important * Correspondence to: Prof. Emilio Russo, Ph.D., Chair of Pharmacology, Department of Science of Health, School of Medicine, University of Catanzaro, Via T. Campanella, 115, 88100 Catanzaro, Italy. E-mail: [email protected] PHYTOTHERAPY RESEARCH Phytother. Res. (2013) Published online in Wiley Online Library (wileyonlinelibrary.com) DOI: 10.1002/ptr.5050 Copyright © 2013 John Wiley & Sons, Ltd. Received 05 February 2013 Revised 03 July 2013 Accepted 05 July 2013
Transcript

* CorrespDepartmeCatanzaroE-mail: eru

PHYTOTHERAPY RESEARCHPhytother. Res. (2013)Published online in Wiley Online Library(wileyonlinelibrary.com) DOI: 10.1002/ptr.5050

Copyright

REVIEW

Hypericum perforatum: Pharmacokinetic,Mechanism of Action, Tolerability, and ClinicalDrug–Drug Interactions

Emilio Russo,1,2* Francesca Scicchitano,1,2 Benjamin J. Whalley,3 Carmela Mazzitello,1,2Miriam Ciriaco,1,2 Stefania Esposito,1,2 Marinella Patanè,1,2 Roy Upton,4 Michela Pugliese,5Serafina Chimirri,1,2 Maria Mammì,1,2 Caterina Palleria1,2 and Giovambattista De Sarro1,21Science of Health Department, School of Medicine, University of Catanzaro, Catanzaro, Italy2Pharmacovigilance’s Center Region Calabria, University Hospital Mater Domini, Catanzaro, Italy3School of Chemistry, Food and Nutritional Sciences, and Pharmacy, The University of Reading, Whiteknights, Reading, BerkshireRG6 6AP, UK4American Herbal Pharmacopoeia, Scotts Valley, CA, USA5Department of Veterinary Science, University of Messina, Messina, Italy

Hypericum perforatum (HP) belongs to the Hypericaceae family and is one of the oldest used and mostextensively investigated medicinal herbs. The medicinal form comprises the leaves and flowering tops of whichthe primary ingredients of interest are naphthodianthrones, xanthones, flavonoids, phloroglucinols (e.g. hyperforin),and hypericin. Although several constituents elicit pharmacological effects that are consistent with HP’s antidepres-sant activity, no single mechanism of action underlying these effects has thus far been found. Various clinical trialshave shown that HP has a comparable antidepressant efficacy as some currently used antidepressant drugs in thetreatment of mild/moderate depression. Interestingly, low-hyperforin-content preparations are effective in thetreatment of depression. Moreover, HP is also used to treat certain forms of anxiety. However, HP can inducevarious cytochrome P450s isozymes and/or P-glycoprotein, of which many drugs are substrates and which are themain origin of HP–drug interactions. Here, we analyse the existing evidence describing the clinical consequenceof HP–drug interactions. Although some of the reported interactions are based on findings from in vitro studies,the clinical importance of which remain to be demonstrated, others are based on case reports where causality can,in some cases, be determined to reveal clinically significant interactions that suggest caution, consideration, anddisclosure of potential interactions prior to informed use of HP. Copyright © 2013 John Wiley & Sons, Ltd.

Keywords: Hypericum perforatum; St. John’s wort; drugs; interaction; cytochrome P450; P-glycoprotein.

INTRODUCTION

A growing percentage of the population uses herbalproducts for preventative and therapeutic purposes.Several reasons for this wide usage have been proposedand include (i) the erroneous notion that, being ofnatural origin, herbals are safer than conventional med-icines; (ii) the philosophical belief that naturally deriveddrugs are more consistent with individual personalvalues; (iii) individuals’ desire for personal control overtheir health; and (iv) hard-to-treat conditions for whichconventional therapies are unavailable (Cheung et al.,2007; Cauffield, 2000). However, such lay beliefs beliethe evidence that herbal drugs cause significant sideeffects either individually or due to herb–drug interactions(Bent, 2008; De Smet, 2007; Ernst, 1998).Hypericum perforatum (HP), or St John’s wort, is a

plant whose extract, one of the best- characterised herbalmedicines, is widely sold in health food stores andpharmacies in Europe and the USA (annual US salesincreased from $20m in 1995 to $200m in 1997) for the

ondence to: Prof. Emilio Russo, Ph.D., Chair of Pharmacology,nt of Science of Health, School of Medicine, University of, Via T. Campanella, 115, 88100 Catanzaro, [email protected]

© 2013 John Wiley & Sons, Ltd.

treatment of depression (Lecrubier et al., 2002; Kasperet al., 2006). Preclinical studies on the central nervoussystem activity of HP extracts suggest that it also displaysanxiolytic, sedative, nootropic, antischizophrenic, anti-convulsant, antidiabetic, and analgesic activities andthat it may be beneficial in the treatment of alcohol,nicotine, and caffeine addiction in experimental animals(Can et al., 2011; Can and Ozkay, 2012).

Recent clinical studies found HP extracts to beefficacious and well tolerated in the treatment of mildto moderate depressive episodes and for the short-termtreatment of symptoms in mild depressive disorders(Chen et al., 2011; Kasper et al., 2010). Moreover, HPextracts can be effective in the treatment of generalisedanxiety disorder, somatoform disorders, sleep disorders,obsessive–compulsive disorder, and seasonal affectivedisorder (Can et al., 2009).

Although >150 discrete ingredients that exhibit manyadditive, synergistic, and partly antagonistic effects havethus far been identified in this plant (Butterweck andSchmidt, 2007), HP’s most characteristic constituents arenaphthodianthrones (hypericin and pseudohypericin) andphloroglucinols (hyperforin and adhyperforin) (Butterweckand Schmidt, 2007; Nahrstedt and Butterweck, 1997),where hypericin is considered to be the most important

Received 05 February 2013Revised 03 July 2013

Accepted 05 July 2013

E. RUSSO ET AL.

active ingredient although its concentration varies be-tween different parts of the plant (0.02–2.5%). However,hypericin has only been shown to have activity (e.g.monoamine oxidase inhibitor) in vitro and has failed toexert detectable effects in animal models. In Germany,HP has been approved by the Commission E since 1984for the treatment of anxiety, depression, and insomniaalthough interest in HP has primarily focused upon itsantidepressant effects. Additionally, numerous studieshave supported wound-healing effects (Negrash andPochinok, 1972; Rao et al., 1991; Šaljić, 1975), antimicro-bial properties (one of the oldest historical uses of HP,predominantly as St John’s wort oil) against a number ofbacterial and fungal strains (Saddiqe et al., 2010; Gaindand Ganjoo, 1959; Khosa and Bhatia, 1982; Shakirovaet al., 1970; Negrash and Pochinok, 1972; Brondz et al.,1982; Gibbons et al., 2002; Saroglou et al., 2007), andantiviral activity (Meruelo et al., 1988).Experimentally,HP extracts and one isolated constituent,

hyperforin, inhibit reuptake of several neurotransmitterssuch as serotonin, noradrenaline, dopamine, glutamate,and gamma-aminobutyric acid in vitro and in vivo(Capasso et al., 2003), many of which are involved in theregulation of mood, motivation, and reward. Subchronictreatment with an HP extract caused downregulation ofβ-adrenergic receptors (De Marchis et al., 2006), and theextract alters animal behaviour not only in several antide-pressant models (e.g. forced-swimming test), consistentwith the effects of conventional antidepressants (e.g.fluoxetine; De Marchis et al., 2006; Butterweck, 2003),but also in a chronic stress, mouse model (Crupi et al.,2011) and exerts anxiolytic and antipanic effects upon ratstested using the elevated T-maze, an animal model ofinnate (panic) and learned (generalised) anxiety (Beijaminiand Andreatini, 2003). However, extracts almost devoidof hyperforin have also demonstrated clinical efficacy(Schrader, 2000), suggesting that hyperforin is not solelyresponsible for the extract’s effects.Mild adverse effects attributed to HP have occasionally

been reported and include gastrointestinal complaints,tiredness, dizziness, and allergic and photosensitivityreactions (typically only at doses above 11.25mg of totalhypericin; Nahrstedt and Butterweck, 1997; Hendersonet al., 2002; Zhou and Lai, 2008).Furthermore and particularly given its nonprescrip-

tion nature, HP is often taken in combination with otherconventional medicines, creating the potential for herb–drug interactions (Hu et al., 2005; Izzo, 2012), theprevalence of which may be hard to quantify given thelay belief that ‘natural products are safe’ and so may leadto under-reporting of their use to clinicians. Commondrugs that interact with HP include anticoagulants, anti-convulsants, anti-HIV drugs, antidepressants, immuno-suppressant agents, antimicrobial drugs, hypoglycaemicdrugs, and oral contraceptives (Borrelli and Izzo, 2009;He et al., 2012) where their areas under the plasmaconcentration–time curve (AUC) and mean/peak bloodconcentrations are decreased (Borrelli and Izzo, 2009).However, from extant case reports, despite noted changesin the pharmacokinetic parameters of some of these drugs(e.g. digoxin and oral contraceptives), no changes topharmacodynamic effects have been found.Hypericum perforatum extracts are powerful activa-

tors of the enzyme CYP3A4 (Hemeryck and Belpaire,2002) and also increase the activity of the P-glycoprotein(P-gp), an adenosine triphosphate-dependent drug

Copyright © 2013 John Wiley & Sons, Ltd.

transporter responsible for increased drug excretion(Hemeryck and Belpaire, 2002; Müller, 2003; Izzo, 2004).Most commercially available drugs are substrates forCYPs and/or P-gp, and some have low therapeutic indices,making interactions affecting available drug potentiallydangerous via either a loss of therapeutic effect or theinduction of toxicity.

In this review, we summarise HP’s known pharmacoki-netic and pharmacodynamic properties before criticallyconsidering its tolerability and the potential or knownHP–drug interactions.

PHARMACODYNAMICS ANDPHARMACOKINETICS OF HYPERICUMPERFORATUM

As previously stated, HP is most widely used for eitherthe treatment of depressive syndromes or antiviral/antiretroviral effects, although the literature reveals usesof this plant for other pharmacological and medicalpurposes (Table 1).

The pharmacokinetics of hyperforin, hypericin, andpseudohypericin, the components of HP extracts thatare presumed to be responsible for its pharmacologicaleffects, has been studied in detail in both animals andhumans (Butterweck, 2003; Caccia and Gobbi, 2009;Wurglics and Schubert-Zsilavecz, 2006). Despite struc-tural similarity (Fig. 1), hypericin, pseudohypericin,and hyperforin exhibit pharmacokinetic differences(Table 2). The pharmacokinetics of these componentswas studied in healthy volunteers after administrationof HP dry extract. After oral administration of singledoses (range = 300–1800mg ) of HP extract (0.3% ofhypericin), hypericin was detectable in the blood after1.3 h, and peak plasma concentrations were reachedafter ~4.6 h and steady-state levels by 4days (Brochmolleret al., 1997; Staffeldt et al., 1994; Upton, 1997). The plasmahalf-life of hypericin is reported to be ~25h (Nangia et al.,2000), the prolonged duration of which is due tohypericin’s affinity for albumin and lipoproteins (Bianchiet al., 1997). Although the bioavailability of hypericin is~14%, the therapeutic concentration of hypericin in brainremains unknown, and although it has been suggested thatbrain levels reach only 5% of plasma levels, hypericin’shalf-life in the brain may be weeks (Upton, 1997).

Pharmacokinetic studies have also been conductedusing a well-characterised pharmaceutical HP extract(LI 160; Lichtwer Pharma, Berlin, Germany) that con-tains 300-mg dry HP extract standardised to 0.24–0.32%hypericins (hypericin and pseudohypericin). Single oraldoses of 300, 900, and 1800mg were administered orallyto 12 healthy subjects, and peak plasma concentrationsof pseudohypericin of 2.7, 11.7, and 30.6 ng/ml, respec-tively, were found after 0.4–0.6 h (Staffeldt, 1994). Subse-quently, it was reported that after oral administration of300, 600, and 1200mg of two different ethanolic HPextracts (containing 5%and 0.5%hyperforin), hyperforinpeak plasma levels were reached after 2.8–3.6 h (Table 2).Here, hyperforin pharmacokinetics was linear fordoses up to 600mg whereas higher doses producedlower-than-expected plasma concentrations after linearextrapolation. In a study employing repeated doses,hyperforin accumulation was not be detected; steady-state plasma concentration after 900mg/day of extract

Phytother. Res. (2013)

Table 1. Suggested pharmacological mechanisms of actions of Hypericum

Activity of Hypericum Mechanism of action Reference

Antidepressant Inhibition of monoamine oxidase (MAO) by hypericin Suzuki et al., 1984; Bladt and Wagner,1994; Bombardelli and Morazzoni, 1995;Chatterjee et al., 1998a, 1998b; Thiedeand Walper, 1994

Inhibition of MAO by the fraction of flavonoids in theextract of Hypericum (39%) structurally similar tothe MAO inhibitorsSuppression of the release of interleukin 6, asubstance related to depression as it modulates therelease of cortisolInhibition of reuptake of serotonin, norepinephrine,dopamine, and gamma-aminobutyric acid by hyperforinInhibition of the enzyme catechol-O-methyltransferase

Antiviral The hypericin to act effectively requires an intactmembrane or cell surface (viral envelope)

Bianchi et al., 1997; Bombardelli andMorazzoni, 1995; Yip et al., 1996; Degaret al., 1992; Bianchi et al., 1997;Takahashi et al., 1989

The mechanism of action includes the photo-activation of hypericinAction at the level of the anchor assembly or virusBinding of hypericin to the lipid membrane of thevirus by inhibition of viral fusion and syncytiumformationDirect action against the capsid of the virus byinhibiting mobilityInhibition of protein kinase C phosphorylation of thereceptor involved in CD4Inhibition of tyrosine kinase

Anticancer Molecule inhibition of nuclear factor κB, the switchcircuits of inflammation and inflammatory angiogenesis

Vandebogaerde and de Witte, 1995

Blocking of the endothelium migration in response toinflammatory cytokines

Vandebogaerde and de Witte, 1995;Thomas et al., 1992a, 1992b

Prevention of Alzheimer’sdisease

Hypericin may interfere with the processes ofpolymerization of beta-amyloid peptide responsiblefor the onset of Alzheimer’s disease

Griffith et al., 2010

Antibacterial Presence of substances with antimicrobial,antibacterial, and antifungal (essential oils,phloroglucinols, flavonoids, and tannins) activity

Saddiqe et al., 2010

Antigastritis and gastric ulcer Soothing and healing. Zdunić et al., 2009Topical Antibacterial properties Schempp et al., 1999

HYPERICUM PERFORATUM: PK, MOA, SAFETY, AND DDI

was ~100ng/ml (Biber et al., 1998) with a half-life of 9 h(Nangia et al., 2000) (Table 2).Although the poor pharmacokinetic profile of HP

extracts [i.e. low bioavailability (15–20%) and poorblood–brain barrier penetration] has been correlatedwith the 4- to 6-week treatment period typicallyrequired to achieve a therapeutic benefit in patients(Bennett et al., 1998), it should be noted that conventionalantidepressants with better pharmacokinetic profilesalso typically require a similar treatment period beforetherapeutic effects are seen (Woelk, 2000).Hypericum perforatum’s ability to act as a substrate of

both CYPs and P-gp has been widely confirmed in vitro(Perloff et al., 2001), in vivo (Imai et al., 2008), and inclinical studies (Xie et al., 2005). In the latter case, HPinduces the activity of several CYP isozymes includingCYP3A4, CYP2E1, and CYP2C19, thereby increasingthe metabolism of many drugs (Gurley et al., 2005;Wang et al., 2004). Moreover, HP’s effect upon CYP3A4has been studied in greater detail where HP administra-tion via the oral route affected midazolam metabolism

Copyright © 2013 John Wiley & Sons, Ltd.

to a greater extent than via the intravenous route,which suggests that HP primarily acts via intestinal,and not hepatic, CYP3A4 (Wang et al., 2001; Dresseret al., 2003).

P-glycoprotein, one of the most clinically importanttransmembrane transporters in humans, is extensivelydistributed and expressed in the intestinal epithelium,hepatocytes, renal proximal tubular cells, adrenalgland, and capillary endothelial cells comprising theblood–brain barrier (Thiebaut et al., 1987), a specificlocalisation that suggests an active role in drug elimina-tion and absorption (Zhou, 2008). HP induces synthesisof P-gp in the intestines to reduce drug absorption(Perloff et al., 2001), and in kidneys, resulting inincreased drug excretion. The former effect has alsobeen reported in the gut of healthy volunteers(Hennessy et al., 2002) and may therefore reduce orincrease plasma concentrations of known P-gp sub-strates such as digoxin (Dürr et al., 2000), fexofenadine(Dresser et al., 2003; Xie et al., 2005), and talinolol(Schwarz et al., 2007).

Phytother. Res. (2013)

Figure 1. Chemical structures of hypericin, pseudohypericin, andhyperforin. Structures of the main chemical constituents ofHypericum: the naphthodianthrone hypericin and pseudohypericinand phloroglucinol hyperforin.

E. RUSSO ET AL.

These HP effects on P-gp or CYP enzymes are typi-cally observed after treatment periods of at least 10 days(Wang et al., 2002), and although data following acute(1–3 days ) or shorter (i.e. 4–9 days ) treatment periodsare not available, no relevant clinical effects have beenreported (Rengelshausen et al., 2005). Furthermore, thedegree of CYP3A4 and P-gp induction is comparablebetween ethnic groups, specifically Caucasians, AfricanAmericans, Hispanics, Chinese, Indians, and Malays(Xie et al., 2005). Finally, hyperforin appears to be respon-sible for the increased expression of CYPs and P-gp(Will-Shahab et al., 2009). Indeed, hyperforin is a pow-erful inducer of CYP3A4 and P-gp (Zhou et al., 2007),and clinical results indicate that hyperforin contentdetermines the extent of HP–drug interactions becauseextracts containing little hyperforin exert only weakeffects on CYP and P-gp (Will-Shahab et al., 2009).

Table 2. Pharmacokinetics of hyperforin, hypericin, and pseudohyperic

Hypericum perforatum extractcomponents Doses (mg)

Peak plasma le(ng/ml)

Hypericin (0.3%) 300 1.5900 4.1

1800 14.2Pseudohypericin (0.24–0.32%) 300 2.7

900 11.71800 30.6

Hyperforin (5%) 3×300mg/day 100.0

Copyright © 2013 John Wiley & Sons, Ltd.

EFFICACY OF HYPERICIM PERFORATUM

In recent years, many studies have proven the efficacy ofsome HP extracts in the treatment of mild to moderatedepression, leading to its successful use as an antidepres-sant in both Europe and the USA (Kasper et al., 2006;Schrader, 2000; Linde et al., 1996; Linde and Knüppel,2005; Schrader et al., 1998; Woelk, 2000; Uebelhacket al., 2004; Kasper et al., 2008). The average daily dosetypically required to such effects is 900mg (range:600–1200mg ) of dry extract as three divided doses perday. Its efficacy and tolerability are well established withclinical studies demonstrating activity comparable withconventional antidepressants, while lacking major sideeffects. As described earlier, suspected HP-related ad-verse events are primarily limited to mild gastrointestinalor cutaneous reactions although the overall incidence ofsuch events is lower than side effects typically reportedfor tricyclic antidepressants (Kim et al., 1999) or selectiveserotonin reuptake inhibitors (SSRIs) (Lecrubier et al.,2002; Schrader, 2000; Woelk, 2000; Szegedi et al., 2005;Gastpar et al., 2006). In clinical studies, HP has showneffectiveness as an antidepressant that is higher thanplacebo in mild or moderate depression, and at leastone study has suggested that HP is superior to paroxetinein the treatment of moderate to severe depression(Szegedi et al., 2005). More than 50 randomised con-trolled trials and more than 15 larger observationalstudies have now investigated the effectiveness of HPextracts in the acute treatment of depressive disorders.Results of the first trial became available in 1979(Hoffmann and Kühl, 1979), and until the mid-1990s,about 25 trials were published. Furthermore, clinicalstudies have compared the effectiveness of HP with thatof tricyclic antidepressants (Linde et al., 2008; Schulz,2006) and found their effects to be largely comparable.

A recent, meta-analysis of randomised controlledtrials reported similar efficacy for the acute use of HPextracts as with SSRIs (Rahimi et al., 2009) although nocomparative studies of efficacy associated with medium-term and long-term use exist, making it impossible toascertain HP’s effectiveness as a maintenance treatmentfor patients with recurrent depression or other distur-bances (dysthymia and bipolar disorder). Also, identifica-tion of the optimal therapeutic dose of HP remainsdifficult because different concentrations have been usedin clinical trials, but none have been specifically under-taken to determine either a minimum effective or optimaldose. Broadly, the majority of studies have used HPextracts standardised to 0.2–0.3% hypericin with dosesranging from 400- to 900-mg HP extract daily. Although

in, components of Hypericum perforatum extracts.

velsHalf-life (h) Bioavailability (%) Reference

24.8–26.5 14 Staffeldt et al., 1994

16.3–36 21 Staffeldt et al., 1994

9 Nangia et al., 2000

Phytother. Res. (2013)

HYPERICUM PERFORATUM: PK, MOA, SAFETY, AND DDI

several large studies have shown that HP can be an effec-tive treatment for major depression, others have foundcontrary results (Montgomery et al., 2000; Shelton et al.,2001; Hypericum Depression Trial Study Group, 2002).Finally, more limited research suggests that HP has

antimicrobial properties that may be of use in thetreatment of burns and wounds (Öztürk et al., 2006;Rao et al., 1991), whereas other preclinical in vitro andin vivo studies have suggested that HP may hold valuein the treatment of other ailments, including cancer,inflammatory disorders, and bacterial and viral diseases,as well as potential uses as an antioxidant andneuroprotective agent (Klemow et al., 2011). However,the extent to which these suggestions might translateto clinical practice remains unrealised.

SAFETY OF HYPERICUM PERFORATUM

Although HP extracts appear to be well tolerated, themost frequently reported side effects are nausea, rash,fatigue, restlessness, and photosensitivity (Table 3), whichhas been largely attributed to naphthodianthrones(hypericin and pseudohypericin) (Rodríguez-Landa andContreras, 2003; Schulz et al., 2006) and typicallymanifests at higher-than-recommended doses. Althoughsevere phototoxic reactions seem to be very rare events(only two cases of severe photo-allergic reactions follow-ing ingestion of HP preparations have been reported inthe literature; Schulz, 2006), patients should be informedthat HP extracts, like some SSRIs, can increase light sen-sitivity (Doffoel-Hantz et al., 2009). In an open study of3250 patients, the most commonly reported side effectswere gastrointestinal symptoms (0.6%), allergic reactions(0.5%), and fatigue (0.4%) (Woelk et al., 1994), whereas,in a meta-analysis of 23 studies, 19.8% of patients usingHP preparations reported side effects, as compared with35.9% of subjects using standard antidepressant medica-tions with 4% of patients in the HP group dropping outin comparison with 7.7% receiving standard antidepres-sants (De Smet and Nolen, 1996).The safety of using HP during pregnancy is important,

not least because women in their reproductive years arefrequent users of natural health products (Eisenberget al., 1998; Fugh-Berman and Kronenberg, 2003) andmay develop depression during pregnancy (Bennettet al., 2004). There are very limited data to support thesafety of HP use during pregnancy or breastfeeding;Lee et al. (2003) compared 30 breastfeeding womentaking different HP preparations for postnatal depression

Table 3. Overview of Hypericum perforatum extract-relatedadverse drugs reactions

Adverse drugs reactions References

Nausea, rash, asthenia,and restlessness

Schrader, 2000; Woelk, 2000; Rodríguez-Landa and Contreras, 2003; Woelk et al.,1994

Photosensitivity Schulz, 2006; Rodríguez-Landa andContreras, 2003; Golsch et al., 1997;Bove, 1998; Nierenberg, et al., 1999;Moses and Mallinger, 2000

Serotonin syndrome Evans, 2008; Bonetto et al., 2007

Copyright © 2013 John Wiley & Sons, Ltd.

with other breastfeeding mothers not taking the com-pound and reported several adverse events in infantsnursed by mothers taking HP (two cases of colic, two ofdrowsiness, and one of lethargy).

A prospective, observational, controlled cohort studycompared 54 women exposed to HP during pregnancywith 108 others who were also pregnant and eithertaking conventional antidepressants (n= 54) or wereotherwise healthy (n= 54). Results showed no signifi-cant difference (p= 0.26) in rates of major postnatalmalformations across groups (5%, 4%, and 0%), andthe authors noted that these rates are comparable withthe risk expected in the general population (Morettiet al., 2009). A small number of animal studies haveshown that HP does not affect cognitive development,long-term growth, or physical maturation and causesno long-term behavioural deficits. However, otheranimal studies have reported lower birth weights inoffspring when HP was consumed during pregnancyand that HP may increase uterine tone (Rayburn et al.,2000, 2001a, 2001b; Dugoua et al., 2006). Furthermore,Gregoretti et al. (2004) demonstrated that chronic HPtreatment during pregnancy or lactation can be respon-sible for histological alterations in the liver and kidneyof rats.

Other HP side effects include confusion, restlessness,lethargy, and dryness of the mouth (Klemow et al.,2011), and in some cases, HP use has been associatedwith psychotic events (Shimizu et al., 2004; Stevinsonand Ernst, 2004).

HYPERICUM PERFORATUM–DRUGINTERACTIONS

Central nervous system drugs

Major drug classes with central nervous system activityinclude antidepressants, antipsychotics, antiepileptics,and anxiolytics but also anaesthetics and analgesics.They act differently to exert their therapeutic effectbut can be grouped by virtue of a common metabolicpathway involving CYP. Co-administration of HP withthese substances induces either reduced plasma concen-trations due to enzyme induction or a summation ofeffects, depending on the case (Table 4).

A primary concern associated with the use of HParises via its putative action as an SSRI leading to aconsequential risk of serotonin syndrome (Lantz et al.,1999), a potentially life-threatening condition arisingfrom excessive serotonergic stimulation. Serotonin syn-drome is characterised by at least three of the following:confusion, agitation, hyperreflexia, diaphoresis, shiveringor tremor, nausea, diarrhoea, lack of coordination, fever,coma, flushing, or rhabdomyolysis (Cookson, 1993). It isusually caused by use of therapeutic drugs (e.g. SSRIs),drug–drug interactions, intentional self-poisoning, ordeliberate poisoning (Boyer and Shannon, 2005; Mathewet al., 1996). In fact, many drug combinations have beenassociated with serotonin syndrome; for example, aclinical study reported seizures in a patient with serotoninsyndrome symptoms followed by acute rhabdomyolysis,a condition apparently precipitated by concurrenttriptan use with long-term fluoxetine and HP treatment

Phytother. Res. (2013)

Table 4. HP–drug interactions

Prescribed drug Clinical results of the interaction with HP Possible mechanism References

AntihistamineFexofenadine Increased the maximum plasma

concentration and decreased the oral clearance

Wang et al., 2002; Di et al.,2008

BronchodilatorTheophylline Decreased plasma concentration Induction of hepatic

cytochromesChen et al., 2012

CardiovascularWarfarin A loss of the anticoagulant effect; significant

reduction in the pharmacological effect ofracemic warfarin

Particle formation inaqueous solution with HP;induction of CYP3A4

Gröning et al., 2003; Jiang et al.,2004

Phenprocoumon Decreased plasma levels Induction of CYP3A4 Chen et al., 2011Nifedipine Induced metabolism with increased

plasma concentrations of dehydronifedipineInduction of CYP3A4 andCYP2C19

Wang et al., 2007

Verapamil Reduced bioavailability Induction of first-passCYP3A4 metabolism

Tannergren et al., 2004

Digoxin Decreased intestinal absorption;reduction of plasma AUC and Cmax

Induction of the P-gp Gottesman et al., 1996; Johneet al., 1999

HypolipidemicAtorvastatin Increased LDL Increases CYP3A4 and P-

gp activityHoltzman et al., 2006;Markowitz et al., 2003Increased total cholesterol

Simvastatin Increased LDL Decreased plasmaconcentrations

Sugimoto et al., 2001

GastrointestinalOmeprazole,esomeprazole,andpantoprazole

Decrease plasma concentration ofproton pump inhibitors

Induction of CYP2C19 Wang et al., 2004

Loperamide Brief episode of delirium Theoretically induces amonoamine oxidaseinhibitor–drug reaction

Khawaja et al., 1999

Oral contraceptivesEtinilestradiol anddesogestrel

Reduction of plasmatic concentration,bleeding, and pregnancies

Induction of CYP3A4 Zhou et al., 2004; Hall et al.,2003; Borrelli and Izzo, 2009;Dresser et al. 2003; Izzo, 2004Increased clearance of noretindrone

and decreased half-time of etinilestradiolEtinilestradiol andnoretindrone

Increased metabolism of noretindroneand etinilestradiol

Non-steroidal antiinflammatory drugsIbuprofen Reduction of plasmatic concentration Increase expression of

glycoprotein GBell et al., 2007b; Zhou et al.,2004; Izzo, 2004; Dresser et al.,2003

CorticosteroidsDexamethasone,prednisone, andbudesonide

Reduction of plasmatic concentration Induction of CYP3A4 Izzo, 2004; Bell et al., 2007a

OpioidsMethadone andpethidine

Reduction of plasmatic concentrationandabstinence syndrome

Induction of CYP2D2 Dostalek et al., 2005

DextromethorphanOxycodone Reduction of plasmatic concentration Induction of CYP3A4 Nieminen et al., 2010

Reduction of plasmatic concentrationAntimicrobialVoriconazole Decreased AUC Induction of CYP3A4,

CYP2C19, and CYP2C9Borrelli and Izzo, 2009

Erythromycin Increased metabolism of erythromycin(decreased AUC)

InductionofCYP3A4 (40%) Borrelli and Izzo, 2009

Indinavir Decrease in AUC of 57% Induction of CYP3A4 Borrelli and Izzo, 2009; Chenet al., 2012

(Continues)

E. RUSSO ET AL.

Copyright © 2013 John Wiley & Sons, Ltd. Phytother. Res. (2013)

Table 4. (Continued)

Prescribed drug Clinical results of the interaction with HP Possible mechanism References

Antineoplastic

Imatinib Decreased plasma concentration Induction CYP3A4 andP-gpIrinotecan Altered hepatic metabolism Caraci et al., 2011

Docetaxel Decreased clinical efficacy Izzo and Ernst, 2009ImmunosuppressantsCyclosporine Decreased plasma concentration Induction enzymes

cytochrome and P-gpHe et al., 2012; Hu et al., 2005

Tacrolimus Organ rejection Mai et al., 2003Hypoglycaemic agentsGliclazide Decreased plasma concentration Induction enzymes

cytochrome and P-gpIzzo and Ernst, 2009

Tolbutamide Di et al., 2008

AUC, area under the curve; HP, Hypericum perforatum; LDL, low-density lipoprotein; P-gp, P-glycoprotein.

HYPERICUM PERFORATUM: PK, MOA, SAFETY, AND DDI

(Evans, 2008). Therefore, triptan use with concurrent flu-oxetine and HP may be inappropriate (Bonetto et al.,2007). Moreover, HP and SSRIs or buspirone orbupropion can exert additive effects on serotonin reuptakeinhibition that are responsible for serotonin syndrome(Borrelli and Izzo, 2009; Caraci et al., 2011).Additional potential pharmacokinetic interactions are

associated with hypericin’s induction of CYP3A4 andCYP2C19 and include reductions of the AUC and half-life and a significant increase of oral clearance of benzodi-azepines (alprazolam and midazolam) (Hall et al., 2003;Markowitz et al., 2003) in addition to reducing the AUCof amitriptyline and nortriptyline. Furthermore, theinduction of CYP2C19 is responsible for increasedurinary excretion of phenytoin metabolites (Borrelli andIzzo, 2009) and decreased plasma concentration ofzolpidem, probably by enhancing CYP3A4 activity (Hojoet al., 2011), whereas induction of CYP1A2 and CYP3A4was responsible for HP-induced decreases in clozapine’sAUC (Van Strater and Bogers, 2012). With respect toanticonvulsant drug–HP interactions, no significant influ-ence of HP on the pharmacokinetics of carbamazepine,sodium valproate, or phenobarbital have been reported(Rahimi and Abdollahi, 2012; Dasgupta et al., 2006).However, although HP modulation of CYP enzymes

has been quite well described, the mechanism underly-ing HP-induced delay in onset of the effects of thegeneral anaesthetics fentanyl, propofol, and sevofluranedescribed by Borrelli and Izzo (2009) has not beenelucidated. It should however be noted that pharmaco-kinetic changes reported for HP cannot be extrapolatedto clinically relevant events although the potential forclinically relevant interactions is greatest for drugsexhibiting very narrow therapeutic indices.

Bronchodilators

The only reported interaction for this drug category iswith theophylline where HP significantly reduces AUCand blood concentrations through CYP3A4 inhibitionto cause a loss of asthma control (Chen et al., 2012)(Table 4). A single case of decreased plasma levels oftheophylline following HP treatment (300mg daily) hasbeen reported (Nebel et al., 1999) and necessitated anincreased dosage of theophylline to restore therapeuticeffects. However, this remains a single case because acontrolled population study demonstrated that HP

Copyright © 2013 John Wiley & Sons, Ltd.

administration (300mg/day) for 15 days did not affectplasma or urine levels of theophylline (Morimoto et al.,2004). Therefore, there is limited evidence to supportwidespread concern about this interaction althoughmonitoring of theophylline dose and blood levels inpatients using HP may be necessary if poor symptomcontrol is evident in individual patients.

Cardiovascular drugs

Oral anticoagulants. Oral anticoagulants are vitamin Kantagonists and act in the liver to inhibit synthesis offactors II, VII, IX, and X of the coagulation and theanticoagulant proteins C and S by competitive inhibitionof the enzyme epoxide reductase. Warfarin is the mostwidely used molecule of this class, the metabolism ofthe two enantiomers of which is undertaken by differentcytochromes, whose activity is susceptible to modulationby concomitant intake of other cytochrome substrates.Thus, R-warfarin is mainly metabolised by CYP1A2and CYP3A4 and S-warfarin, the more potent form,predominantly by CYP2C19A.

A clinical trial showed that HP induces the apparentclearance of both S-warfarin and R-warfarin, which, inturn, resulted in a significant reduction in racemicwarfarin’s pharmacological effect (Jiang et al., 2004).Similarly, another trial revealed HP-induced decreasesin plasma levels of phenprocoumon (a coumarinic anti-coagulant chemically related to warfarin) (Chen et al.,2012) (Table 4). Together, these results provide a basisto explain a number of case reports (~29 as of 2005) ofincreased prothrombin time or corresponding decreasesin international normalised ratio associated with warfarin(Yue et al., 2000) or phenprocoumon (Bon et al., 1999)with HP use. Despite these sometimes significant changesin pharmacokinetic parameters, no cases of bleeding epi-sodes or thrombotic events attributable to concomitantHP use have been reported (Schulz and Johne, 2005).

Gröning et al. (2003) have investigated the physical–chemical interactions between HP extracts and severaldrugs, more precisely, the formation of nanoparticles,microparticles, and precipitates in aqueous drug-freeand drug-containing preparations of HP. In aqueousinfusions of HP, only small amounts of nanoparticlesand microparticles were observed using photon correla-tion spectroscopy and scanning electron microscopy.Warfarin has a negligible effect on the particle formation

Phytother. Res. (2013)

E. RUSSO ET AL.

in aqueous infusions. However, in further studies withHPdry extracts reconstituted in water, nanoparticles and pre-cipitates are already formed in other drug-free prepara-tions. In this case, the amount of free warfarin in thesolution is maximally reduced by 36.6%. The authorssuggested that the loss of anticoagulant effect describedin literature may partly be caused by particle formationduring drug administration (Gröning et al., 2003).

Calcium channel blockers. Nifedipine, a calcium channelantagonist, is primarily metabolised by CYP3A4 todehydronifedipine. As such, nifedipine is often used asa probe drug for determining CYP3A4-mediateddrug–drug interactions in human studies. A rapid andsensitive liquid chromatography/tandem mass spec-trometry method was developed and validated to deter-mine nifedipine and dehydronifedipine simultaneouslyin human plasma. This method has been successfullyused by Wang et al. (2007) to study nifedipine pharma-cokinetic interactions with HP in healthy volunteerswhere HP was shown to induce nifedipine metabolismto increase plasma concentrations of dehydronifedipine(Wang et al., 2007). HP also interacts with verapamil toreduce its bioavailability by induction of first-passCYP3A4 metabolism, most likely in the gut (Tannergrenet al., 2004) (Table 4).

Cardiac inotropic drugs. Digoxin is a digitalis drug usedto increase the force of contraction of both atrial andventricular myocardial fibres (positive inotropic effect).Drug interaction studies with digoxin are particularlyimportant because of its low therapeutic index. It hasbeen shown that chronic HP use lowers the bioavailabilityof digoxin, potentially via reduced intestinal absorptionarising from induction of the multidrug resistance trans-porter, P-gp. Actually, the concurrent action of intestinalCYP3A4 and P-gp creates an absorption barrier, whichinfluences the oral bioavailability of many medications(Wacher et al., 1996; Watkins, 1997). Digoxin is a P-gpsubstrate and is largely unmetabolised in humans; thus,after 10days of HP treatment, oral digoxin exhibited a25% reduction of both plasmaAUC andmaximum bloodconcentration (Cmax) (Johne et al., 1999; Di et al., 2008).Moreover, consistent with these results, two trials havealso shown decreased plasma digoxin concentrationsfollowing 10days of HP administration (Johne et al.,1999; Mueller et al., 2004). Consistent with intestinalinduction of P-gp, this interaction was characterised by areduction in AUC0–24 and Cmax, which led to a reductionin Ctrough, whereas in the parallel study of Mueller et al.(2004), a 25% decrease in steady-state digoxin concentra-tion was observed in a group of healthy volunteers.Conversely, in the same study, administration of othercrude preparations (teas, oil, and alcohol extract) pro-duced no (or only marginal) effects on digoxin plasmalevels. Thus, the extent of HP–digoxin interactions varybetween HP preparations and doses but does appearparticularly well correlated with hyperforin levels(Mueller et al., 2004) (Table 4). Although the precedingresults suggest that HP-mediated decreases in digoxinlevels are clinically relevant, no cases of therapeuticinteractions between HP and digoxin have been reportedto date.

Hypocholesterolaemic drugs. ‘Statins’ or inhibitors of3-hydroxy-3-methylglutaryl coenzyme A reductase, the

Copyright © 2013 John Wiley & Sons, Ltd.

key enzyme in the biosynthetic pathway of cholesterol,are the drugs of choice for the treatment ofhypercholesterolaemia and can reduce the risk ofcardiovascular morbidity and mortality (ScandinavianSimvastatin Survival Study Group, 1994; West of ScotlandCoronary Prevention Study Group, 1998) in subjects withcardiovascular disease. Statins are hepatically metabolisedthrough catalysis induced by CYP2C9 (fluvastatin) orCYP3A4 (simvastatin, lovastatin, and atorvastatin)(Shitara and Sugiyama, 2006). Furthermore, atorvastatinmight inhibit P-gp and may also be a substrate for thistransporter (Holtzman et al., 2006). With regard to HP,controlled clinical studies revealed that mean low-densitylipoprotein cholesterol level and total cholesterol are sig-nificantly higher after 4weeks of treatment with productscontaining HP despite the use of statins (atorvastatin andsimvastatin) (Andrén et al., 2007; Eggertsen et al., 2007)(Table 4). HP lowers simvastatin plasma levels, whereasno interaction with pravastatin was observed (Sugimotoet al., 2001). Furthermore, in patients with hypercholester-olemia receiving simvastatin, HP can increase low-densitylipoprotein levels (Eggertsen et al., 2007) (Table 4).Moreover, it is very likely that products containing HPmay interact with atorvastatin (Andrén et al., 2007).

Drugs acting on the gastrointestinal tract

Proton pump inhibitors (PPIs) are hepatically metabolisedby two isozymes of CYP (CYP2C19 and CYP3A4).Pantoprazole is also metabolised by a cytosolicsulfotransferase, esomeprazole primarily by CYP2C19,and omeprazole by CYP3A4. In particular, caution shouldbe exercised when taking omeprazole and esomeprazolewith HP as reduced plasma concentrations of PPIs havebeen reported (Wang et al., 2004).

Although no single scientific study yet publisheddescribes interactions between HP and antidiarrhoealmedications, Khawaja et al. (1999) documented a briefepisode of acute delirium, possibly induced by exposureto HP, valerian root, and loperamide. It was suggestedthat the observed delirium was related to the concomi-tant use of these medications, although the mechanismwas not elucidated (Khawaja et al., 1999). Loperamidealone can cause delirium, but it remains possible thatHP and/or valerian contributed to the precipitation ofthis event (Schwartz and Rodrigues, 1991) (Table 4).

Oral contraceptives

Clinical studies have demonstrated a higher incidence ofintracyclic bleeding episodes after co-administration ofHP and oral contraceptives (Hall et al., 2003; Murphyet al., 2005). The intermenstrual bleeding is believed toarise from HP-induced reductions in plasma concentra-tions of oral contraceptive tablet components (Table 4).Furthermore, such bleeding irregularities could adverselyaffect compliancewith oral contraceptive use and, togetherwith HP-induced decreases in serum 3-ketodesogestrelconcentrations, enhance the risk of unintended pregnan-cies (Will-Shahab et al., 2009). Conversely, from thereported extensive use of a leading European HP extractby women of child-bearing age (~4 million between 1991and 1999), eight reports of interim bleeding equate to anincidence of approximately 1 report per 500 000 women

Phytother. Res. (2013)

HYPERICUM PERFORATUM: PK, MOA, SAFETY, AND DDI

treated, which is an incidence lower than that of interimbleeding associated with oral contraceptive use alone(Rosenberg et al., 1996).

Antiinflammatory drugs, corticosteroids, and opioids

Among the non-steroidal antiinflammatory drugs,ibuprofen interacts with HP; increased P-gp expressionreduces plasma levels of the drug and, consequently,decreases its effects (Bell et al., 2007a) (Table 4). TheCYP3A4 pathway also mediates the metabolism ofcorticosteroids: prednisone, prednisolone, dexamethasone,and budesonide; HP co-administration reduces their AUC(Bell et al., 2007b).The induction of CYP3A4 and CYP2D2 by HP also

reduces the plasma concentration of several opioids suchasoxycodone, dextromethorphan, andpethidine (Table4)(Dostalek et al., 2005; Nieminen et al., 2010); interactionsthat may be of clinical significance when consideringpatients with chronic pain (Nieminen et al., 2010).

Antimicrobial drugs (antibacterial, antifungal,and antiviral)

A number of clinically significant interactions of HPwith antibacterial, antifungal, and antiviral drugs havebeen identified (Di et al., 2008), and thus, concomitantadministration of HP should be avoided in order toreduce treatment failures, particularly in high-risk cases.Antimicrobial drugs are substrates of CYP3A4, and it isknown that HP-mediated induction of CYP3A4 reducesthe AUC of voriconazole, erythromycin, and indinavir(Borrelli and Izzo, 2009; Chen et al., 2012) (Table 4).

Antineoplastic drugs

Hypericum perforatum extracts, via induction of bothCYP3A4 and P-gp, may reduce plasma concentrationsof several antineoplastic agents (e.g. imatinib, irinotecan,and docetaxel) and hence their clinical efficacy (Izzo andErnst, 2009) (Table 4). Specifically, an unblinded,randomised crossover study showed that patients treatedwith irinotecan should abstain from taking HP becauseplasma levels of this drug were dramatically reduced, aninteraction with the potential to exert a deleteriousimpact on treatment outcome (Mathijssen et al., 2002).Moreover, two trials found increased imatinib clearancefollowing HP administration, confirming that the HP co-administration can affect the therapeutic action of theantineoplastic drug (Frye et al., 2004; Smith et al., 2004).

Immunosuppressant drugs

A 2003 study confirmed previous clinical trials and casereports describing interaction between HP and immuno-suppressive drugs (Alscher and Klotz, 2003) where HPsignificantly reduced both AUC and plasma concentra-tions of cyclosporine and tacrolimus (Table 4) (Hu et al.,2005; Chen et al., 2012). An important consequence ofthese interactions may be the risk of organ rejection(Mai et al., 2003).

Copyright © 2013 John Wiley & Sons, Ltd.

Oral hypoglycaemic drugs

Clinical trials indicate that HP, via CYP and/or P-gpinduction, can reduce the plasma concentrations (and/orincrease the clearance) of gliclazide (Izzo and Ernst,2009). Although HP does not alter the pharmacokineticsof tolbutamide, it does increase the incidence ofhypoglycaemia (Zhou et al., 2004), with a more recentclinical study demonstrating that both pharmacokineticand pharmacodynamic components may play a role inthese interactions (Table 4) (Di et al., 2008).

DISCUSSION

Here, we have summarised the safety and efficacy of HPand classified the potential clinical effects of known orevidence-based potential interactions between conven-tional drugs and HP. HP is one of the oldest and best-investigated medicinal herbs, and substantial evidencesupports its effectiveness in the treatment of variousforms of depression and anxiety (Rahimi and Abdollahi,2012) although the most clearly relevant safety issue withHP extracts lies with HP–drug interactions. Notably,extracts containing low or negligible amounts ofhyperforin can be effective in the treatment of depressionbut lack interactive potential (Mueller et al., 2009). HPextracts are potent activators of the enzymes CYP3A4and CYPC19 (Hemeryck and Belpaire, 2002), enzymesthat play a relevant role in the metabolism of many drugs.Furthermore, HP extracts also increase the activity ofP-gp, which is responsible for increased excretion ofdrugs. Thus, pharmacokinetic interactions can occurwhen HP is combined with drugs that are metabolisedthrough CYP3A4, CYP2C19, and/or are P-gp substrates(Chen et al., 2011). Pharmacodynamic interactions canoccur when HP is combined with drugs that increase theconcentration of 5-hydroxytryptamine in the brain (Chenet al., 2011). HP interactions include reduced bloodconcentration of anticoagulants, cyclosporine, gastroin-testinal drugs, and antineoplastic drugs (e.g. imatinib);serotonin syndrome; or lethargy when HP was given con-comitantly with SSRIs. However, many others that arepredictable might have not been reported or studied.More generally, HP interaction with drugs metabolisedby CYPs might be easily controlled, whereas the role ofP-gp appears to be more complex (Ott et al., 2010).

CONCLUSIONS

Herb–drug interactions associated with HP have beeninvestigated to a greater extent than that for any otherbotanical. The fact that HP affects CYP and P-gp, theprimary substrates associated with conventional drugmetabolism, raises significant concerns for HP–druginteractions. Despite reports of changes in pharmacoki-netics of a variety of conventional medications and thewidespread use of HP worldwide, the incidence ofclinically relevant interactions based on clinical studies,case reports, and pharmacovigilance data remains verysmall. The greatest risk for HP–drug interactions is fordrugs that have a narrow therapeutic and safety index.Additionally, aside from the potential of interactions,

Phytother. Res. (2013)

E. RUSSO ET AL.

there is a very low incidence of adverse effects with HPuse. The low incidence of side effects and the low cost ofHP represent an interesting therapeutic alternative tosynthetic antidepressants when used alone. There isgood evidence that HP and preparations with lowhyperforin content can be effective as monotherapiesfor the treatment of mild to moderate depression(Mueller et al., 2009). However, potential herb–drug

Copyright © 2013 John Wiley & Sons, Ltd.

interactions should be considered and monitored, andtherapies adjusted as necessary.

Conflict of Interest

The authors have declared that there is no conflict of interest.

REFERENCES

Alscher DM, Klotz U. 2003. Drug interaction of herbal teacontaining St. John’s wort with cyclosporine. Transpl Int16: 543–544.

Andrén L, Andreasson A, Eggertsen R. 2007. Interaction betweena commercially available St. John’s wort product (Movina) andatorvastatin in patients with hypercholesterolemia. Eur J ClinPharmacol 63: 913–916.

Beijamini V, Andreatini R. 2003. Effects of Hypericum perforatumand paroxetine on rat performance in the elevated T-maze.Pharmacol Res 48: 199–207.

Bell EC, Ravis WR, Chan HM, Lin YJ. 2007a. Lack of pharmacoki-netic interaction between St. John’s wort and prednisone. AnnPharmacother 41: 1819–1824.

Bell EC, Ravis WR, Lloyd KB, Stokes TJ. 2007b. Effects of St.John’s wort supplementation on ibuprofen pharmacokinetics.Ann Pharmacother 41: 229–234.

Bennett HA, Einarson A, Taddio A, Koren G, Einarson TR. 2004.Prevalence of depression during pregnancy: systematicreview. Obstet Gynecol 103: 698–709.

Bennett DA, Phun L Jr, Polk JF, Voglino SA, Zlotnik V, Raffa RB.1998. Neuropharmacology of St. John’s wort (Hypericum).Ann Pharmacother 32: 1201–1208.

Bent S. 2008. Herbal medicine in the United States: review of effi-cacy, safety, and regulation: grand rounds at University ofCalifornia, San Francisco Medical Center. J Gen Intern Med23: 854–859.

Bianchi A, Adamoli R, Durante A, Saibene A. 1997. PianteMedicinali e AIDS. Ed. Tecniche Nuove: Milan.

Biber A, Fischer H, Römer A, Chatterjee SS. 1998. Oral bioavail-ability of hyperforin from Hypericum extracts in rats andhuman volunteers. Pharmacopsychiatry 31: 36–43.

Bladt S, Wagner H. 1994. Inhibition of MAO by fractions andconstituents of Hypericum extract. J Geriatr Psychiatry Neurol7: 57–59.

Bombardelli E, Morazzoni P. 1995. Hypericum perforatum.Fitoterapia 66: 43–68.

Bon S, Hartmann K, Kubn M. 1999. Johanniskraut: EnzyminduktorEin? Schweitzer Apothekerzeitung 16: 535–536.

Bonetto N, Santelli L, Battistin L, Cagnin A. 2007. Serotonin syn-drome and rhabdomyolysis induced by concomitant use oftriptans, fluoxetine andHypericum.Cephalalgia27: 1421–1423.

Borrelli F, Izzo AA. 2009. Herb–drug interactions with St John’swort (Hypericum perforatum): an update on clinical observa-tions. AAPS J 11: 710–727.

Bove GM. 1998. Acute neuropathy after exposure to sun in apatient treated with St John’s wort. Lancet 352: 1121–1122.

Boyer EW, Shannon M. 2005. The serotonin syndrome. N Engl JMed 352: 1112–1120.

Brochmoller J, Rheum T, Bauer S, Kerb R, Hubner WD, Roots I.1997. Hypericin and pseudohypericin: pharmacokinetics andeffects on photosensitivity in humans. Pharmacopsychiatry30: 94–101.

Brondz I, Greibrokk T, Groth PA, Aasen AJ. 1982. The relativestereochemistry of hyperforin—an antibiotic from Hypericumperforatum L. Tetrahehedron Lett 23: 1299–1300.

Butterweck V, Schmidt M. 2007. St. John’s wort: role of activecompounds for its mechanism of action and efficacy. WienMed Wochenschr 157: 356–361.

Butterweck V. 2003. Mechanism of action of St John’s wort indepression: what is known? CNS Drugs 17: 539–562.

Caccia S, Gobbi M. 2009. St. John’s wort components and thebrain: uptake, concentrations reached and the mechanismsunderlying pharmacological effects. Curr Drug Metabolism10: 1055–1065.

Can OD, Ozkay UD. 2012. Effects of Hypericummontbretti extracton the central nervous system and involvement of GABA

(A)/benzodiazepine receptors in its pharmacological activity.Phytother Res 26: 1695–1700.

Can ÖD, Öztürk Y, Demir Özkay Ü. 2009. A natural antidepressantHypericum perforatum L.: review. Turk Klinikleri J Med Sci29: 708–715.

Can ÖD, Öztürk Y, Öztürk N, et al. 2011. Effects of treatment withSt. John’s wort on blood glucose levels and pain perceptionsof streptozotocin-diabetic rats. Fitoterapia 82: 576–584.

Capasso F, Gaginella TS, Grandolini G, Izzo AA. 2003.Phytotherapy: A Quick Reference to Herbal Medicine. SpringerVerlag: Berlin.

Caraci F, Crupi R, Drago F, Spina E. 2011. Metabolic drug interac-tions between antidepressants and anticancer drugs: focuson selective serotonin reuptake inhibitors and Hypericumextract. Curr Drug Metabol 12: 570–577. Review.

Cauffield JS. 2000. The psychosocial aspects of complementary andalternative medicine. Pharmacotherapy 20(11): 1289–1294.

Chatterjee SS, Bhattacharya SK, Wonnemann M, Singer A, MullerWE. 1998a. Hyperforin as a possible antidepressant componentof Hypericum extracts. Life Sci 63: 499–510.

Chatterjee SS, Noldner M, Koch E, Erdelmeier C. 1998b. Antide-pressant activity of Hypericum perforatum and hyperforin:the neglected possibility. Pharmacopsychiatry 31: 7–15.

Chen XW, Serag ES, Sneed KB, et al. 2011. Clinical herbal interac-tions with conventional drugs: from molecules to maladies.Curr Mel Chem 8: 4836–4850. Review.

Chen XW, Sneed KB, Pan SY, et al. 2012. Herb–drug interactionsand mechanistic and clinical considerations. Curr DrugMetabol 13: 640–651.

Cheung CK, Wyman JF, Halcon LL. 2007. Use of complementaryand alternative therapies in community-dwelling older adults. JAltern Complement Med 13(9): 997–1006.

Cookson J. Side-effects of antidepressants. 1993. Br J Psychiatry163: 20–24.

Crupi R, Mazzon E, Marino A, et al. 2011. Hypericum perforatumtreatment: effect on behaviour and neurogenesis in a chronicstress model in mice. BMC Complement Altern Med 11: 7.

Dasgupta A, Tso G, Szelei-Stevens K. 2006. St. John’s wort doesnot interfere with therapeutic drug monitoring of 12 com-monly monitored drugs using immunoassays. J Clin Lab Anal20: 62–67.

De Marchis GM, Bürgi S, Kientsch U, Honegger UE. 2006. VitaminE reduces antidepressant-related beta-adrenoceptor down-regulation in cultured cells. Comparable effects on St. John’swort and tricyclic antidepressant treatment. Planta Med72: 1436–1437.

De Smet PA, Nolen WA. 1996. St John’s wort as an antidepressant.BMJ 313: 241–242.

De Smet PA. 2007. Clinical risk management of herb–drug interac-tions. Br J Clin Pharmacol 63: 258–267.

Degar S, Prince AM, Pascual D, et al. 1992. Inactivation of thehuman immunodeficiency virus by hypericin: evidence forphotochemical alteration of p24 and block in uncoating. AIDSRes Hum Retrovirus 8: 1929–1936.

Di YM, Li CG, Xue CC, Zhou SF. 2008. Clinical drugs that interactwith St. John’s wort and implication in drug development.Curr Pharm Design 14: 1723–1742.

Doffoel-Hantz V, Boulitrop-Morvan C, Sparsa A, Bonnetblanc JM,Dalac S, Bédane C. 2009. Photosensitivity associated with selec-tive serotonin reuptake inhibitors.Clin ExpDermatol34: 763–765.

Dostalek M, Pistovcakova J, Jurica J, et al. 2005. Effect ofSt John’s wort (Hypericum perforatum) on cytochrome P-450activity in perfused rat liver. Life Sci 78: 239–244.

Dresser GK, Schwarz UI, Wilkinson GR, Kim RB. 2003. Coordinateinduction of both cytochrome P4503A and MDR1 by St John’swort in healthy subjects. Clin Pharmacol Ther 73: 41–50.

Phytother. Res. (2013)

HYPERICUM PERFORATUM: PK, MOA, SAFETY, AND DDI

Dugoua JJ, Mills E, Perri D, Koren G. 2006. Safety and efficacy ofSt. John’s wort (Hypericum) during pregnancy and lactation.Can J Clin Pharmacol 13: 268–276. Review.

Dürr D, Stieger B, Kullak-Ublick GA, et al. 2000. St John’s wortinduces intestinal P-glycoprotein/MDR1 and intestinal andhepatic CYP3A4. Clin Pharmacol Ther 68: 598–604.

Eggertsen R, Andreasson A, Andrén L. 2007. Effects of treatmentwith a commercially available St John’s wort product (Movina)on cholesterol levels in patients with hypercholesterolemiatreatedwith simvastatin.Scand JPrimHealthCare25: 154–159.

Eisenberg DM, Davis RB, Ettner SL, et al. 1998. Trends in alterna-tive medicine use in the United States, 1990–1997: results ofa follow-up national survey. JAMA 280: 1569–1575.

Ernst E. 1998. Harmless herbs? A review of the recent literature.Am J Med 104: 170–178. Review.

Evans RW. 2008. Triptans and serotonin syndrome. Cephalalgia28: 573–574.

Frye RF, Fitzgerald SM, Lagattuta TF, Hruska MW, Egorin MJ.2004. Effect of St John’s wort on imatinib mesylate pharma-cokinetics. Clin Pharmacol Ther 76: 323–329.

Fugh-BermanA, Kronenberg F. 2003. Complementary and alternativemedicine (CAM) in reproductive-age women: a review of ran-domized controlled trials. Reprod Toxicol 17: 137–152. Review.

Gaind KN, Ganjoo TN. 1959. Antibacterial principle of Hypericumperforatum Linn. Indian J Pharm 21: 172.

Gastpar M, Singer A, Zeller K. 2006. Comparative efficacy andsafety of a once-daily dosage of Hypericum extract STW3-VIand citalopram in patients with moderate depression: adouble-blind, randomised, multicentre, placebo-controlledstudy. Pharmacopsychiatry 39: 66–75.

Gibbons S, Ohlendorf B, Johnsen I. 2002. The genus Hypericum—a valuable resource of anti-staphylococcal leads. Fitoterapia73: 300–304.

Golsch S, Vocks E, Rakoski J, Brockow K, Ring J. 1997. Reversibleincrease in photosensitivity to UV-B caused by St. John’s wortextract. Hautarzt 48: 249–252.

Gottesman MM, Pastan I, Ambudkar SV. 1996. P-glycoprotein andmultidrug resistance. Curr Opin Genet Dev 6: 610–617.

Gregoretti B, Stebel M, Candussio L, Crivellato E, Bartoli F, DecortiG. 2004. Toxicity of Hypericum perforatum (St. John’s wort)administered during pregnancy and lactation in rats. ToxicolAppl Pharmacol 200: 201–205.

Griffith TN, Varela-Nallar L, Dinamarca MC, Inestrosa NC. 2010.Neurobiological effects of Hyperforin and its potential inAlzheimer’s disease therapy. Curr Med Chem 17: 391–406.

Gröning R, Breitkreutz J, Müller RS. 2003. Physico-chemicalinteractions between extracts of Hypericum perforatum L.and drugs. Eur J Pharm Biopharm 56: 231–236.

Gurley BJ, Gardner SF, Hubbard MA, et al. 2005. Clinical assess-ment of effects of botanical supplementation on cytochromeP450 phenotypes in the elderly: St John’s wort, garlicoil, Panax ginseng and Ginkgo biloba. Drugs Aging22: 525–539.

Hall SD, Wang Z, Huang SM, et al. 2003. The interaction betweenSt John’s wort and an oral contraceptive. Clin Pharmacol Ther74: 525–535.

He SM, Sneed KB, Chen XW, Cao C, Zhou SF. 2012. Herb–druginteractions and mechanistic and clinical considerations. CurrDrug Metabol 13: 640–651.

Hemeryck A, Belpaire FM. 2002. Selective serotonin reuptakeinhibitors and cytochrome P-450 mediated drug–drug interac-tions: an update. Curr Drug Metabl 3: 13–37. Review.

Henderson L, Yue QY, Bergquist C, Gerden B, Arlett P. 2002.St John’s wort (Hypericum perforatum): drug interactions andclinical outcomes. Br J Clin Pharmacol 54: 349–356. Review.

Hennessy M, Kelleher D, Spiers JP, et al. 2002. St Johns wortincreases expression of P-glycoprotein: implications for druginteractions. Br J Clin Pharmacol 53: 75–82.

Hoffmann J, Kühl ED. 1979. Therapy of depressive states withhypericin. ZFA (Stuttgart) 55: 776–782.

Hojo Y, Echizenyam M, Ohkubo T, Shimizu T. 2011. Drug interac-tion between St John’s wort and zolpidem in healthy subjects.J Clin Pharm Ther 36: 711–715.

Holtzman CW,Wiggins BS, Spinler SA. 2006. Role of P-glycoproteinin statin drug interactions. Pharmacotherapy 26: 1601–1607.

Hu Z, Yang X, Ho PC, et al. 2005. Herb–drug interactions: a literaturereview. Drugs 65: 1239–1282.

HypericumDepression Trial StudyGroup. 2002. Effect ofHypericumperforatum (St John’s wort) in major depressive disorder: arandomized controlled trial. JAMA 287: 1807–1814.

Copyright © 2013 John Wiley & Sons, Ltd.

Imai H, Kotegawa T, Tsutsumi K, et al. 2008. The recovery time-course of CYP3A after induction by St John’s wort administra-tion. Br J Clin Pharmacol 65: 701–707.

Izzo A, Ernst E. 2009. Interactions between herbal medicinesand prescribed drugs: an updated systematic review. Drugs69: 1777–1798.

Izzo AA. 2004. Drug interactions with St. John’s wort (Hypericumperforatum): a review of the clinical evidence. Int J ClinPharmacol Ther 42: 139–148.

Izzo AA. 2012. Interactions between herbs and conventionaldrugs: overview of the clinical data. Med Princ Pract21: 404–428.

Jiang X, Williams KM, Liauw WS, et al. 2004. Effect of St John’swort and ginseng on the pharmacokinetics and pharmacody-namics of warfarin in healthy subjects. Br J Clin Pharmacol57: 592–599.

Johne A, Brockmöller J, Bauer S, Maurer A, Langheinrich M, RootsI. 1999. Pharmacokinetic interaction of digoxin with an herbalextract from St John’s wort (Hypericum perforatum). ClinPharmacol Ther 66: 338–345.

Kasper S, Anghelescu IG, Szegedi A, Dienel A, Kieser M. 2006.Superior efficacy of St John’s wort extract WS 5570compared to placebo in patients with major depression: arandomized, double-blind, placebo-controlled, multi-centertrial [ISRCTN77277298]. BMC Med 23: 4–14.

Kasper S, Caraci F, Forti B, Drago F, Aguglia E. 2010. Efficacyand tolerability of Hypericum extract for the treatment ofmild to moderate depression. Eur Neuropsychopharmacol20: 747–765.

Kasper S, Volz HP, Moller HJ, Dienel A, Kieser M. 2008. Continua-tion and long-term maintenance treatment with Hypericumextract WS 5570 after recovery from an acute episodeof moderate depression—a double-blind, randomized,placebo controlled long-term trial. Eur Neuropsychopharmacol18: 803–813.

Khawaja IS, Marotta RF, Lippmann S. 1999. Herbal medicines as afactor in delirium. Psychiatr Serv 50: 969–970.

Khosa RL, Bhatia N. 1982. Antifungal effect of Hypericumperforatum. J Sci Res Pt Med 3: 49–50.

KimHL, Streltzer J, Goebert D. 1999. St. John’swort for depression:a meta-analysis of well-defined clinical trials. J Nerv Ment Dis187: 532–538.

Klemow KM, Bartlow A, Crawford J, Kocher N, Shah J, Ritsick M.2011. Medical attributes of St. John’s wort (Hypericumperforatum). In Herbal Medicine: Biomolecular and ClinicalAspects, (2nd edn), Benzie IFF, Wachtel-Galor S (eds). CRCPress: Boca Raton, FL; Chapter 11.

Lantz MS, Buchalter E, Giambanco V. 1999. St. John’s wort andantidepressant drug interactions in the elderly. J GeriatrPsychiatry Neurol Spring 12: 7–10.

Lecrubier Y, Clerc G, Didi R, Kieser M. 2002. Efficacy of St. John’swort extract WS 5570 in major depression: a double-blind,placebo-controlled trial. Am J Psychiatry 159: 1361–1366.

Lee A, Minhas R, Matsuda N, Lam M, Ito S. 2003. The safety ofSt. John’s wort (Hypericum perforatum) during breastfeeding.J Clin Psychiatry 64: 966–968.

Linde K, Berner MM, Kriston L. 2008. St John’s wort for majordepression. Cochrane Database Syst Rev 4: CD000448.Review.

Linde K, Knüppel L. 2005. Large-scale observational studies ofHypericum extracts in patients with depressive disorders—asystematic review. Phytomedicine 12: 148–157. Review.

Linde K, Ramirez G, Mulrow CD, Pauls A, Weidenhammer W,Melchart D. 1996. St John’s wort for depression—anoverview and meta-analysis of randomised clinical trials.BMJ 313: 253–258.

Mai I, Störmer E, Bauer S, Krüger H, Budde K, Roots I. 2003.Impact of St John’s wort treatment on the pharmacokineticsof tacrolimus and mycophenolic acid in renal transplantpatients. Nephrol Dial Transplant 18: 819–822.

Markowitz JS, Donovan JL, DeVane CL, et al. 2003. Effects of St.John’s wort on drug metabolism by induction of cytochromeP450 3A4 enzyme. JAMA 290: 1500–1504.

Mathew NT, Tietjen GE, Lucker C. 1996. Serotonin syndromecomplicating pharmacotherapy. Cephalalgia 16: 323.

Mathijssen RH, Verweij J, de Bruijn P, Loos WJ, Sparreboom A.2002. Effects of St. John’s wort on irinotecan metabolism. JNatl Cancer Inst 94: 1247–1249.

Meruelo D, Lavie G, Lavie D. 1988. Therapeutic agents with dra-matic antiretroviral activity and little toxicity at effective doses:

Phytother. Res. (2013)

E. RUSSO ET AL.

aromatic polycyclic diones hypericin and pseudohypericin. ProcNatl Acad Sci U S A 85: 5230–5234.

Montgomery SA, Hübner WD, Grigoleit HG. 2000. Efficacy andtolerability of St John’s wort extract compared with placeboin patients with a mild to moderate depressive disorder.Phytomedicine 7: 107.

Moretti ME, Maxson A, Hanna F, Koren G. 2009. Evaluating thesafety of St. John’s wort in human pregnancy. ReproducToxicol 28: 96–99.

Morimoto T, Kotegawa T, Tsutsumi K, Ohtani Y, Imai H, Nakano S.2004. Effect of St. John’s wort on the pharmacokineticsof theophylline in healthy volunteers. J Clin Pharmacol 44:95–101.

Moses EL, Mallinger AG. 2000. St. John’s wort: three cases of pos-sible mania induction. J Clin Psychopharmacol 20: 115–117.

Mueller SC, Uehleke B, Woehling H, et al. 2004. Effect of St John’swort dose and preparations on the pharmacokinetics ofdigoxin. Clin Pharmacol Ther 75: 546–557.

Mueller SC, Majcher-Peszynska J, Mundkowski RG, et al. 2009.No clinically relevant CYP3A induction after St. John’s wortwith low hyperforin content in healthy volunteers. Eur J ClinPharmacol 65: 81–87.

Müller WE. 2003. Current St. John’s wort research from mode ofaction to clinical efficacy. Phamacol Res 47: 101–109.

Murphy PA, Kern SE, Stanczyk FZ, Westhoff CL. 2005. Interactionof St. John’s wort with oral contraceptives: effects on thepharmacokinetics of norethindrone and ethinyl estradiol,ovarian activity and breakthrough bleeding. Contraception71: 402–408.

Nahrstedt A, Butterweck V. 1997. Biologically active and otherchemical constituents of the herb of Hypericum perforatum.Pharmacopsychiatry 30: 129–134.

Nangia M, Syed W, Doraiswamy PM. 2000. Efficacy and safety ofSt. John’s wort for the treatment of major depression. PublicHealth Nutr 3: 487–494. Review.

Nebel A, Schneider BJ, Baker RK Kroll DJ. 1999. Potential meta-bolic interaction between St. John’s wort and theophylline.Ann Pharmacother 33: 502.

Negrash AK, Pochinok PY. 1972. Comparative study of chemo-therapeutic and pharmacological properties of antimicrobialpreparations from common St. John’s wort. Fitonotsidy Mater,Soveshch 6: 198–200.

Nieminen TH, Hagelberg NM, Saari TI, et al. 2010. St John’s wortgreatly reduces the concentrations of oral oxycodone. Eur JPain 14: 854–859.

Nierenberg AA, Burt T, Matthews J, Weiss AP. 1999. Mania asso-ciated with St. John’s wort. Biol Psychiatry 46: 1707–1708.

Ott M, Huls M, Cornelius MG, Fricker G. 2010. St. John’s wortconstituents modulate P-glycoprotein transport activity atthe blood–brain barrier. Pharm Res 27: 811–822.

Öztürk N, Korkmaz S, Öztürk Y. 2006. Wound-healing activity ofSt. John’swort (Hypericumperforatum L.) on chicken embryonicfibroblasts. J Ethnopharmacol 111: 33–39.

Perloff MD, von Moltke LL, Störmer E, Shader RI, Greenblatt DJ.2001. Saint John’s wort: an in vitro analysis of P-glycoproteininduction due to extended exposure. Br J Pharmacol134: 1601–1608.

Rahimi R, Abdollahi M. 2012. An update on the ability of St. John’swort to affect the metabolism of other drugs. Expert OpinDrug Metab Toxicol 8: 691–708.

Rahimi R, Nikfar S, Abdollahi M. 2009. Efficacy and tolerabilityof Hypericum perforatum in major depressive disorder incomparison with selective serotonin reuptake inhibitors: ameta-analysis. Progr Neuropsychopharmacol Biol Psychiatry33: 118–127.

Rao SG, Udupa AL, Udupa SL, Rao PGM, Rao G, Kulkarni DR.1991. Calendula and Hypericum: two homeopathic drugspromoting wound healing in rats. Fitoterapia 62: 508–510.

Rayburn WF, Christensen HD, Gonzalez CL. 2000. Effect ofantenatal exposure to Saint John’s wort (Hypericum) onneurobehavior of developing mice. Am J Obstet Gyneco183: 1225–1231.

RayburnWF, Gonzalez CL, Christensen HD, Harkins TL, Kupiec TC.2001a. Impact of Hypericum (St.-John’s-wort) given prena-tally on cognition of mice offspring. Neurotoxicol Teratol23: 629–637.

Rayburn WF, Gonzalez CL, Christensen HD, Stewart JD. 2001b.Effect of prenatally administered Hypericum (St John’s wort)on growth and physical maturation of mouse offspring. Am JObstet Gynecol 184: 191–195.

Copyright © 2013 John Wiley & Sons, Ltd.

Rengelshausen J, Banfield M, Riedel KD, et al. 2005. Oppositeeffects of short-term and long-term St John’s wort intake onvoriconazole pharmacokinetics.Clin Pharmacol Ther78: 25–33.

Rodríguez-Landa JF, Contreras CM. 2003. A review of clinical andexperimental observations about antidepressant actions andside effects produced by Hypericum perforatum extracts.Phytomedicine 10: 688–699. Review

Rosenberg L, Bégaud B, Bergman U, et al. 1996. What are the risksof third-generation oral contraceptives? Are third-generationoral contraceptives safe? Hum Reprod 11: 687–688. Review.

Saddiqe Z, Naeem I,MaimoonaA. 2010.A reviewof the antibacterialactivity of Hypericum perforatum L. J Ethnopharmacol 131:511–521.

Šaljić J. 1975.Ointment for treatment of burns.GerOffen83: 197797.Saroglou V, Marin PD, Rančić A, Veljić M, Skaltsa H. 2007.

Composition and antimicrobial activity of the essential oil of sixHypericum species fromSerbia.BiochemSyst Ecol35: 146–152.

Scandinavian Simvastatin Survival Study Group. 1994. Random-ized trial of cholesterol lowering in 4444 patients withcoronary heart disease. Lancet 344: 1383–1389.

Schempp CM, Winghofer B, Langheinrich M, Schopf E, Simon JC.1999. Hypericin levels in human serum and interstitial skinblister fluid after oral single-dose and steady-state administra-tion of Hypericum perforatum L. extract. Skin Pharmacol ApplSkin Physiol 12: 299–304.

Schrader E,Meier B, BrattströmA.1998.Hypericum treatment ofmild-moderate depression in a placebo-controlled study. A prospective,double-blind, randomized, placebo-controlled, multicentre study.Hum Psychopharmacol 13: 163–169.

Schrader E. 2000. Equivalence of St John’s wort extract (Ze 117)and fluoxetine: a randomized, controlled study inmild–moderatedepression. Int Clin Psychopharmacol 15: 61–68.

Schulz HU, Schürer M, Bässler D, Weiser D. 2006. Investigationof the effect on photosensitivity following multiple oraldosing of two different Hypericum extracts in healthy men.Arzneimittelforschung 56: 212–221.

Schulz V, Johne A. 2005. Side effects and drug interactions. InMilestones in Drug Therapy: St. John’s Wort and Its ActivePrinciples in Depression and Anxiety, Müller WE (ed.).Birkhaüser-Verlag: Basel; 145–160.

Schulz V. 2006. Safety of St. John’s wort extract compared tosynthetic antidepressants. Phytomedicine 13: 199–204.

Schwartz RH, Rodrigues WJ. 1991. Toxic delirium possibly causedby loperamide. J Pediatr 118: 656–657.

Schwarz UI, Hanso H, Oertel R, et al. 2007. Induction of intestinalP-glycoprotein by St John’s wort reduces the oral bioavailabilityof talinolol. Clin Pharmacol Ther 81: 669–678.

Shakirova KK, Garagulia dC, Khazanovich RL. 1970. Antimicrobicproperties of some species of St. John’s wort cultivated inUzbekistan. Mikrobiol Zh 32: 494–497.

Shelton RC, Keller MB, Gelenberg A, et al. 2001. Effectiveness ofSt John’s wort in major depression: a randomized controlledtrial. JAMA 285: 1978–1986.

Shimizu K, Nakamura M, Isse K, Nathan PJ. 2004. First-episodepsychosis after taking an extract of Hypericum perforatum(St John’s wort). Hum Psychopharmacol 19: 275–276.

Shitara Y, Sugiyama Y. 2006. Pharmacokinetic and pharmacody-namics alterations of 3-hydroxy-3-methylglutaryl coenzymeA (HMG-CoA) reductase inhibitors: drug–drug interactionsand interindividual differences in transporter and metabolicenzyme function. Pharmacol Ther 112: 71–105.

Smith P, Bullock JM, Booker BM, Haas CE, Berenson CS, JuskoWJ. 2004. The influence of St. John’s wort on the pharmaco-kinetics and protein binding of imatinib mesylate. Pharmaco-therapy 24: 1508–1514.

Staffeldt B, Kerb R, Brockmöller J, Ploch M, Roots I. 1994.Pharmacokinetics of hypericin and pseudohypericin after oralintake of the Hypericum perforatum extract LI 160 in healthyvolunteers. J Geriatr Psychiatry Neurol 7: 47–53.

Stevinson C, Ernst E. 2004. Can St. John’s wort trigger psychoses?Int J Clin Pharmacol Ther 42(9): 473–80.

Sugimoto KI, Ohmori M, Tsuruoka S, et al. 2001. Different effectsof St. John’s wort on the pharmacokinetics of simvastatin andpravastatin. Clin Pharmacol Ther 70: 518–524.

Suzuki O, Katsumata Y, Oya M, Bladt S, Wagner H. 1984. Inhibitionof monoamine oxidase by hypericin. Planta Med 50: 272–274.

Szegedi A, Kohnen R, Dienel A, Kieser M. 2005. Acute treatmentof moderate to severe depression with Hypericum extractWS 5570 (St John’s wort): randomised controlled double blindnon-inferiority trial versus paroxetine. BMJ 330: 503.

Phytother. Res. (2013)

HYPERICUM PERFORATUM: PK, MOA, SAFETY, AND DDI

Takahashi I, Nakanishi S, Kobayashi E, Nakano H, Suzuki K,Tamaoki T. 1989. Hypericin and pseudohypericin specificallyinhibit protein kinase C: possible relation to their antiretroviralactivity. Biochem Biophysl Res Commun 165: 1207–1212.

Tannergren C, Engman H, Knutson L, Hedeland M, Bondesson U,Lennernäs H. 2004. St John’s wort decreases the bioavailabilityof R- and S-verapamil through induction of the first-pass metab-olism. Clin Pharmacol Ther 75: 298–309.

Thiebaut F, Tsuruo T, Hamada H, Gottesman MM, Pastan I,Willingham MC. 1987. Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal humantissues. Proc Natl Acad Sci 84: 7735–7738.

ThomasC,MacGill RS,Miller GC, Pardini RS. 1992a. Photoactivationof hypericin generates singlet oxygen in mitochondria andinhibits succinoxidase. Photochem Photobiol 55: 47–53.

Thomas C, MacGill RS, Neill P, Pardini RS. 1992b. The in vitro andin vivo photoinduced antineoplastic activity of hypericin [meetingabstract]. Proc Annu Meet Am Assoc Cancer Res 33: 2989.

Uebelhack R, Gruenwald J, Graubaum HJ, Busch R. 2004. Efficacyand tolerability of Hypericum extract STW 3-VI in patientswith moderate depression: a double-blind, randomized,placebo-controlled clinical trial. Adv Ther 21: 265–275.

Upton R. 1997. St. John’s wort (Hypericum perforatum). AmericanHerbal Pharmacopoeia (AHP). In HerbalGram 40. AmericanHerbal Pharmacopoeia: Santa Cruz; 132.

Van Strater AC, Bogers JP. 2012. Interaction of St John’s wort(Hypericumperforatum)with clozapine. Int Clin Psychopharmacol27: 121–124.

Vandebogaerde A, de Witte P. 1995. Antineoplastic properties ofphotosensitized hypericin (meeting abstract). Anticancer Res15: 1757–1758.

Wacher VJ, Salphati L, Benet LZ. 1996. Active secretion andenterocytic drug metabolism barriers to drug absorption. AdvDrug Deliv Rev 20: 99–112.

Wang LS, Zhu B, Abd El-Aty AM, et al. 2004. The influence ofSt John’s wort on CYP2C19 activity with respect to genotype.J Clin Pharmacol 44: 577–581.

Wang XD, Li JL, Lu Y, et al. 2007. Rapid and simultaneous determi-nation of nifedipine and dehydronifedipine in human plasma byliquid chromatography–tandem mass spectrometry: applica-tion to a clinical herb–drug interaction study. J Chromatogr BAnalyt Technol Biomed Life Sci 852: 534–544.

Wang Z, Gorski JC, Hamman MA, Huang SM, Lesko LJ, Hall SD.2001. The effects of St John’s wort (Hypericum perforatum)on human cytochrome P450 activity. Clin Pharmacol Ther70: 317–326.

Copyright © 2013 John Wiley & Sons, Ltd.

Wang Z, Hamman MA, Huang SM, Lesko LJ, Hall SD. 2002. Effectof St John’s wort on the pharmacokinetics of fexofenadine.Clin Pharmacol Ther 71: 414–420.

Watkins PB. 1997. The barrier function of CYP3A4 and P-glycoproteinin the small bowel. Adv Drug Deliv Rev 27: 161–170.

West of Scotland Coronary Prevention Study Group. 1998. Influ-ence of pravastatin and plasma lipids on clinical events in theWest of Scotland Coronary Prevention Study (WOSCOPS).Circulation 97: 1440–1445.

Will-Shahab L, Bauer S, Kunter U, Roots I, Brattström A. 2009. StJohn’s wort extract (Ze 117) does not alter the pharmacokinet-ics of a low-dose oral contraceptive. Eur J Clin Pharmacol65: 541.

Woelk H, Burkard G, Grunwald J. 1994. Benefits and risks of theHypericum extract LI 160: drug-monitoring study with 3250patients. J Geriatr Psychiatry Neurol 7: 34–38.

Woelk H. 2000. Comparison of St John’s wort and imipraminefor treating depression: randomised controlled trial. BMJ321: 536–539.

Wurglics M, Schubert-Zsilavecz M. 2006. Hypericum perforatum:a ‘modern’ herbal antidepressant: pharmacokinetics of activeingredients. Clin Pharmacokinet 45: 449–468. Review.

Xie R, Tan LH, Polasek EC, et al. 2005. CYP3A and P-glycoproteinactivity induction with St. John’s wort in healthy volunteersfrom 6 ethnic populations. J Clin Pharmacol 45: 352–356.

Yip L, Hudson JB, Kowalik EG, Zalkow LH, Towers GHN. 1996.Antiviral activity of a derivative of the photosensitive compoundhypericin. Phytomedicine 3: 185–190.

Yue QY, Bergquist C, Gerdén B. 2000. Safety of St John’s wort(Hypericum perforatum). Lancet 355: 576–577.

Zdunić G, Godevac D, Milenković M, et al. 2009. Evaluationof Hypericum perforatum oil extracts for an antiinflammatoryand gastroprotective activity in rats. Phytother Res 23:1559–1564.

Zhou S, Chan E, Pan SQ, Huang M, Lee EJ. 2004. Pharmacokineticinteractions of drugs with St John’s wort. J Psychopharmacol18: 262–276.

Zhou SF, Lai X. 2008. An update of clinical drug interactions withthe herbal antidepressant St. John’s wort. Curr Drug Metab 9:394–409.

Zhou SF, Zhou ZW, Li CG, et al. 2007. Identification of drugs thatinteract with herbs in drug development. Drug Discov Today12: 664–673.

Zhou SF. 2008. Structure, function and regulation of P-glycoprotein and its clinical relevance in drug disposition.Xenobiotica 38: 802–832.

Phytother. Res. (2013)


Recommended