MEDICAL JOURNALS/PROFESSIONAL RESOURCES: HERBAL REMEDIES IN PSYCHIATRIC PRACTICE

MEDICAL JOURNALS/PROFESSIONAL RESOURCES:
HERBAL REMEDIES IN PSYCHIATRIC PRACTICE

Albert H. C. Wong, MD, FRCPC; Michael Smith, MRPharmS, ND; Heather S. Boon, BScPhm, PhD

Patients' use of alternative and complementary health services has created a need for physicians to become informed about the current literature regarding these treatments. Herbal remedies may be encountered in psychiatric practice when they are used to treat psychiatric symptoms; produce changes in mood, thinking, or behavior as a side effect; or interact with psychiatric medications. English-language articles and translated abstracts or articles (where available) found on MEDLINE and sources from the alternative/complementary health field were reviewed. Each herb was assessed for its safety, side effects, drug interactions, and efficacy in treating target symptoms or diagnoses. A synopsis of the information available for each herb is presented. In many cases the quantity and quality of data were insufficient to make definitive conclusions about efficacy or safety. However, there was good evidence for the efficacy of St John's wort for the treatment of depression and for ginkgo in the treatment of memory impairment caused by dementia. More research is required for most of the herbs reviewed, but the information published to date is still of clinical interest in diagnosing, counseling, and treating patients who may be taking botanical remedies.

(Arch Gen Psychiatry. 1998;55:1033-1044)

It is currently estimated that alternative/complementary medicine is used by 20% to 30% of the general North American population, and use appears to be most common in patients with chronic conditions.1,2 It is estimated that North Americans spend more than $11 billion dollars for chiropractic, naturopathic, and herbal therapies not covered by health plans each year,1,3 and the current annual growth rate of the alternative/complementary medicine industry is estimated to be 20%.4 Patients' growing interest in alternative/complementary medicine has created the need for accurate information that is accessible to physicians. While this review cannot be comprehensive, especially in covering remedies used in less developed countries, the botanicals commonly encountered in North America that have particular relevance to psychiatry will be discussed. The herbal remedies reviewed herein will be divided into 3 sections: (1) herbs that are commonly used to treat psychiatric symptoms, (2) herbs that have psychotropic effects, and (3) herbs that may interact with either psychiatric illnesses or the drugs used to treat these illnesses.

Information included in this review was drawn from comprehensive MEDLINE (1986-1997) searches, frequently cited or landmark articles, sources commonly used by the alternative health care community in North America (eg, The Canadian College of Naturopathic Medicine reference library), and consensus reports from expert committees (eg, The German Commission E5 and the European Scientific Cooperative on Phytotherapy). The efficacy of herbal interventions was rated according to criteria derived from the 1994 Canadian Guide to Clinical Preventive Health Care by the Canadian Task Force on the Periodic Health Examination6 and the 1996 Guide to Clinical Preventive Services by the US Preventive Services Task Force.7 This rating system was designed to provide clinically relevant guidance regarding the use of these herbs by patients.

Many of the herbs discussed in this article have a variety of constituents and putative therapeutic indications. Only the information relevant to psychiatric illness, symptoms, or treatments will be considered because of space limitations. This review focuses primarily on the English-language literature. A further caveat concerns the lack of standardization, quality control, and regulation of commercial herbal products in much of the world.8,9 The studies cited herein report results of a wide range of herbal preparations, some of which may not be applicable to other preparations of the same herb.

Herbal Remedies Commonly Used to Treat Psychiatric Symptoms

Black Cohosh
Black cohosh (Cimicifuga racemosa [L] Nutt) has a history of use among North American aboriginal peoples as a treatment for the hot flashes, anxiety, and dysphoria associated with menopause; as an analgesic; and to promote lactation and menses.10 Its putative action is on the gonadotropin system, through direct estrogen ligands that suppress luteinizing hormone release, and through nonestrogen ligands that appear to decrease luteinizing hormone secretion with long-term use.11 A variety of uncontrolled studies have demonstrated some clinical benefit in the treatment of ovarian insufficiency symptoms.12-15

A randomized study comparing a commercial product of C racemosa (Remifemin) with conventional hormonal therapy for the treatment of ovarian insufficiency symptoms showed comparable efficacy with both treatments (n=60).16 A randomized, double-blind, placebo-controlled trial with the same product (n=80) found superior efficacy to placebo or conjugated estrogen therapy for the treatment of both physical and mental menopausal symptoms.17 The long-term benefits of postmenopausal hormone treatment have not been compared with those of C racemosa. The dose of black cohosh ranges from 40 to 200 mg daily, and the onset of action is reported to be up to 2 weeks.18

Potential side effects include gastric upset, throbbing headaches, dysphoria, and cardiovascular depression. Caution should be exercised if the herb is taken with other hormonal therapies, since C racemosa likely interacts with the sex hormone system, although such interaction has not been documented. It should be avoided during pregnancy and lactation.18,19

German Chamomile
German chamomile (Matricaria recutita L) has been used for the treatment of gastrointestinal tract discomfort, peptic ulcer disease, mouth and skin irritation, pediatric colic and teething, and mild insomnia and anxiety.18,20 The herb contains the flavonoid apigenin, which may have an affinity for the benzodiazepine receptor21 and may also interact with the histamine system.22 A mild hypnotic effect has been reported in mice21 and in humans,23-25 but no randomized or controlled clinical studies were identified. Doses commonly range from 2 to 4 g of dried flower heads 3 times daily, normally prepared as a tea.5,18 Chamomile is also commercially available as a liquid extract (1:1, 45% ethanol), which is dosed as 1 to 4 mL 3 times daily,18 and as a tincture (1:5, 45% ethanol), of which 3 to 10 mL is taken 3 times daily.18 Potential adverse reactions are rare and mainly allergic in nature.19,23,24,26-28

Evening Primrose
Some authors have proposed the use of evening primrose (Oenothera biennis L) in the treatment of schizophrenia, childhood hyperactivity, and dementia, apparently on the basis of isolated reports of prostaglandin abnormalities in schizophrenia29 and attention-deficit/hyperactivity disorder.30-32 There is little scientific evidence or cultural tradition to support this usage. The pharmacological constituents of interest are the essential and nonessential fatty acids: cis-linoleic acid, cis-?-linolenic acid, oleic acid, palmitic acid, and stearic acid.19,33,34 However, the empirical evidence for changes in fatty acid levels after oral administration is sparse,35 and the literature on the efficacy of fatty acid administration in the treatment of schizophrenia and attention-deficit/hyperactivity disorder is contradictory.36-38 In addition, there is currently insufficient information to recommend evening primrose in the treatment of dementia.19,39 The majority of evening primrose oil supplements contain 8% cis-?-linolenic acid, and the daily adult dose ranges from 6 to 8 g, normally given in divided doses.19

In general, evening primrose oil is relatively safe,40-42 but it should be used with caution in mania43 and epilepsy.19,41,43 There have been cases in which evening primrose oil appears to have exacerbated epilepsy. Drugs that may interact adversely with evening primrose include phenothiazines,37,39,44 nonsteroidal anti-inflammatory drugs, corticosteroids,45?-blockers,39 and anticoagulants.43

Ginkgo
The ginkgo tree (Ginkgo biloba L) is one of the oldest deciduous tree species on earth.46 It has been used for medicinal purposes extensively in Europe and has a minor role in traditional Chinese medicine.47-49 Its indications are varied and include dementia, "chronic cerebrovascular insufficiency," and "cerebral trauma."47,48 Standardized commercial preparations are widely available and usually contain the active constituents flavone glycosides (24%) and terpenoids (6%).50,51 Most clinical studies have used specific extracts of Ginkgo (EGb 761 and LI 1370) that are equivalent to the commercial preparations. The dose for most indications is 40 mg of standardized extract 3 times daily,20,48 which must be given for 1 to 3 months before the full therapeutic effects are apparent.48,52

There is considerable evidence that Ginkgo extracts can improve vascular perfusion53-59 by modulating vessel wall tone60-69 and can decrease thrombosis70 through antagonism of platelet activating factor.71-75 Ischemic sites may benefit in particular from ginkgo treatment.76-84 The antioxidant properties that have been attributed to the flavonoid components found in ginkgo are believed to play an important role in its postulated neuroprotective and ischemia-reperfusion-protective effects.85-91 The extract EGb 761 has been shown to have both hydroxyl radical scavenging activity and superoxide dismutase-like activity.68,92

Kleijnen and Knipschild53 reviewed 40 controlled trials on the use of ginkgo in the treatment of "chronic cerebral insufficiency." Although only 8 of the studies were deemed to be of good quality, all but 1 found clinically significant improvement in symptoms, such as memory loss, concentration difficulties, fatigue, anxiety, and depressed mood. Studies investigating the use of ginkgo to augment memory or treat memory loss have produced conflicting results. Generally, some improvement is reported in patients with moderate to severe memory impairment, but no significant improvement is seen in those with mild to no memory impairment.93-103

There is also evidence, derived from randomized, controlled trials, that ginkgo extracts are effective in the treatment of psychopathological conditions and memory impairment caused by Alzheimer and vascular dementia.104-107 For example, 1 multicenter, randomized, double-blind, placebo-controlled trial (N=216) assessed the use of ginkgo in the treatment of outpatients diagnosed with primary degenerative dementia of the Alzheimer type or multi-infarct dementia of mild to moderate severity (DSM-III-R108 criteria). The participants were given either 120 mg of G biloba extract (EGb 761) or placebo twice daily. Response to treatment was defined as response to a minimum of 2 of the 3 primary outcome variables: the Clinical Global Impressions (item 2) to assess psychopathology, the Syndrom-Kurztest to assess memory and attention, and the Nurnberger Alters-Beobachtungsskala rating scale to assess the ability to perform the activities of daily life. The investigators reported that the frequency of response was significantly (P<.005) higher in the ginkgo-treated group, which was confirmed by intention-to-treat analyses.104

A recent North American multicenter, randomized, controlled trial in a similar patient population (outpatients with mild to severe Alzheimer disease or multi-infarct dementia) followed 309 patients for 52 weeks and reported similar results. Patients in this study received 40 mg of G biloba extract (EGb 761) or placebo 3 times daily, and the following outcome measures were used: the Alzheimer's Disease Assessment Scale-Cognitive subscale, the Geriatric Evaluation by Relative's Rating Instrument, and the Clinical Global Impression of Change. With an intention-to-treat analysis, those taking ginkgo scored significantly higher on both the Alzheimer's Disease Assessment Scale-Cognitive subscale (P=.04) and the Geriatric Evaluation by Relative's Rating Instrument (P=.005). There was no difference reported between the 2 groups in the Clinical Global Impression of Change scores. The investigators concluded that, although the changes in the ginkgo-treated groups were modest, they were of sufficient magnitude to be recognized by caregivers.107

Ginkgo has been used by patients in an attempt to treat impotence, including antidepressant-induced sexual dysfunction. In one open trial (N=60), patients with proved arterial erectile dysfunction who had not previously responded to papaverine ingested 60 mg of G biloba extract daily for 12 to 18 months. Fifty percent of the men had gained potency after 6 months of therapy; however, the role of Ginkgo in this recovery is difficult to determine, given the large psychological component of impotence and the fact that this trial was not blinded. There are no reports cited in the MEDLINE literature that investigated the use of ginkgo for antidepressant-induced sexual dysfunction.

There is one randomized, controlled trial showing improvement in resistant depression with ginkgo as an augmenting agent with conventional antidepressants.109 One in vitro study reported that compounds present in both dried and fresh Ginkgo leaves have monoamine oxidase (MAO) (both A and B) inhibitory activity;110 however, there is currently no evidence that G biloba extracts ingested in normal dosages by humans will inhibit MAO activity in the brain. Finally, there is evidence from animal studies that ginkolide B may have a neuroprotective effect in brain injury.84,111,112

Side effects from ginkgo appear to be relatively uncommon, but include headache, gastrointestinal tract upset, and skin allergy to the Ginkgo fruit.48,49,53,61,113-115 Of these, headache is the most common, and it is best prevented by starting with a low dose and gradually titrating to the required dose. Many researchers have suggested that ginkgo theoretically may potentiate other anticoagulants or increase bleeding time; however, these effects rarely have clinically significant implications. Millions of people take ginkgo every year, yet only 2 reported cases of bleeding problems (neither a confirmed drug interaction) may be found in the literature.116,117 Caution should still be exercised when ginkgo is taken in conjunction with anticoagulant treatment (including aspirin) or where there is a risk of bleeding (eg, peptic ulcer disease, subdural hematoma). Safety in pregnancy and lactation has not been established.

Hops
Although hops (Humulus lupulus L) is used by the brewing industry to produce beer, the female flowers of the plant also have a long medicinal history as a mild sedative.25,118 Hops is also currently used as a mild hypnotic agent18,33,119-121; however, there are no clinical studies of its use as a single agent to treat specific symptoms or illnesses, such as insomnia or anxiety disorders. The sedating effects of hops may be mediated by one of its constituent volatile oils, 2-methyl-3-butene-2-ol,20,121,122 but there is insufficient information to confirm this. Adverse effects include allergy and disruption of menstrual cycles.19,25,123,124 Hops is commonly given 3 times daily and before bed in the following doses: 0.5 to 1g of dried flowers, 0.5 to 1 mL of liquid extract (1:1, 45% ethanol), or 1 to 2 mL of tincture (1:5, 60% ethanol).18 The use of hops should be avoided in depression, in pregnancy, and during lactation.18,19,25 Although there are currently no documented case examples, care should be taken when hops is used with sedative-hypnotic agents and alcohol, as a potentiation of their effect may be seen.

Kava
Preparations made from the roots of kava (Piper methysticum, Forst) have been used extensively by the peoples of the South Pacific for both medicinal and cultural purposes.125 Medicinally, it is reputed to have anxiolytic, anticonvulsant, sedative, and muscle relaxant properties.125,126 While a number of pharmacologically active agents have been identified, most interest has centered on the ?-pyrones commonly referred to as kavalactones. Conflicting evidence exists regarding the affinity of kava pyrones for various ?-aminobutyric acid (GABA) or benzodiazepine-binding sites.127,128 Kavain, a kavalactone, has been shown to block the voltage-dependent sodium ion channel.129 In animals, kava has been reported to exhibit neuroprotective effects against ischemia.130 Anticonvulsive effects have also been noted.131-133 Many published works regarding kava are in German, but a number of reviews are available in English.125,134

Kava has been reported to produce changes on the electroencephalogram similar to those seen with diazepam.135 Several human clinical trials suggest that kava products standardized for kavalactone content (70%) may be beneficial in the management of anxiety and tension of nonpsychotic origin.136,137 Kava appears not to adversely affect cognitive function, mental acuity, or coordination125,138 in comparison with oxazepam, as measured by event-related potentials during cognitive testing.139,140 Clinical trials with kava have used doses of standardized preparations that range from 100 to 200 mg of kavalactones daily in divided doses or a single dose at bedtime.126

Long-term administration with higher doses (eg, 400 mg of kavalactones) may result in scaling of the skin on the extremities.125,126 It has been hypothesized that kava produces a vitamin B deficiency that results in the scaling. However, administration of nicotinamide (100 mg daily for 3 weeks) did not resolve the condition.141 Kava may interact with benzodiazepines; there is 1 controversial case report involving alprazolam.142 While the consumption of large amounts of alcohol has been noted to potentiate the actions of kava in mice,143 administration of a standardized kava extract in a placebo-controlled, double-blind study showed few adverse effects.144 The possibility exists that concomitant administration may potentiate the action of other centrally mediated agents.126

Lemon Balm
An aromatic member of the mint family, lemon balm (Melissa officinalis L) has a history of use as an anxiolytic.25,121 Although 1 article has reported hypnotic and analgesic effects in mice,145 there are currently no clinical studies demonstrating hypnotic or anxiolytic effects in humans, even though some authors endorse this use.25,118,119 One study using a combination product containing valerian and lemon balm showed a sleep-promoting effect, but it is difficult to conclude what role lemon balm played in this effect.146 Doses of lemon balm range from 1 to 4g daily.25 No side effects have been reported from ingestion of lemon balm; however, safety in pregnancy and lactation has not been established. Lemon balm may potentiate the effects of other central nervous system (CNS) depressants, including alcohol,145 and may interact with thyroid medications or thyroid disease.33,147-150

Passion Flower
Passion flower (Passiflora incarnata L) is native to the Americas, where its perennial vine leaves have been used as a sedative by indigenous peoples such as the Aztecs.19,33,151 Its current use as a sedative-hypnotic118,119,151-153 is supported by the findings of some animal studies153-157; however, the active ingredients118,151,158,159 and mechanism of action remain obscure. No clinical studies of P incarnata alone have been found, although one randomized, controlled trial that used a commercial preparation containing P incarnata in addition to valerian showed benefit in the treatment of adjustment disorder with anxious mood.160 Passion flower is often given 3 times daily in the following doses: 0.25 to 1 g of dried herb (commonly taken as a tea); 0.5 to 1 mL of liquid extract (1:1; 25% alcohol); or 0.5 to 2.0 mL of tincture (1:8; 45% alcohol).18,19

Hypersensitivity vasculitits161 and "altered consciousness"162 have been reported with products containing passion flower. Passion flower may cause sedation, and so the usual precautions regarding operation of a motor vehicle or machinery should be observed.153 Excessive use during pregnancy and lactation should be avoided.19 Interactions with other psychotropic medications have not been adequately studied.

Skullcap
Members of the genus Scutellaria have a long history of medicinal use; the roots in traditional Chinese medicine and the aerial parts in western herbalism.19,163 Skullcap (Scutellaria laterifolia L) has been used as a sedative and anticonvulsant.119,164 The active ingredients and pharmacology are not well documented. In addition, existing studies are not necessarily applicable to preparations that patients may be taking, because different species and parts of the plant are used.113 Skullcap is available in several dosage forms that are commonly taken 3 times daily: 1 to 2 g of dried herb or 2 to 4 mL of liquid extract (1:1; 25% alcohol).18 Adverse reactions include giddiness, confusion, sedation, seizures,165,166 and possibly hepatotoxic effects166 (the hepatotoxic effect in this case was later attributed to another ingredient of the preparation167). Although there is insufficient information to make specific recommendations regarding safety, skullcap should be avoided in pregnancy and lactation, and may interact with other CNS drugs.168

St John's Wort
The use of St John's wort (Hypericum perforatum L) may be traced back to the texts of the ancient Greek physicians Hippocrates, Pliny, and Galen, and continued through the Classical, Renaissance, and Victorian eras.169,170 Its contemporary usage has been as an antidepressant, for which there is more rigorous evidence than for any other herbal remedy.171,172 The active ingredients responsible for antidepressant action have been investigated (mainly hypericin and pseudohypericin),173-177 but the putative mechanism of action of St John's wort extracts remains controversial.

The hypericins were found to be absorbed within 2 hours, in a dose-dependent manner. These compounds are widely distributed and have a plasma half-life of 24 hours, allowing steady-state concentrations to be reached in 4 days.175,176 It is estimated that 14% to 21% of the compounds are systemically available.176 In vitro experiments found hypericin mainly in the cytoplasmic membrane and cytoplasm, with smaller amounts found in the nucleus.178

Hypericum extracts show affinity for a variety of neurotransmitter receptors, including: adenosine, GABAA, GABAB, serotonin (5-HT)1, central benzodiazepine, forskolin, inositol triphosphate, and the MAO A and B enzymes.179 Hypericin by itself has an affinity for the N-methyl-D-aspartate receptor.180 However, the concentrations required for in vivo activity are unlikely to be attained after oral administration, except for activity at the GABA receptors.180 Various authors have proposed serotonin reuptake inhibition,181,182 decreased serotonin receptor expression,183 altered receptor regulation,182 inhibition of benzodiazepine binding,184 increased excretion of adrenergic metabolites,185 and inhibition of MAO186,187 to explain the clinically observed antidepressant effects. Although MAO inhibitor activity is an attractive explanation for the antidepressant actions of St John's wort, studies are unable to confirm that putative MAO inhibition is responsible for antidepressant effects.187-189

Animal studies show changes, similar to those seen with other antidepressants, on behavioral tests.190 Changes are seen in assays of motor activity, exploratory behavior, analgesia, ketamine sleeping time, and temperature.191,192 Rats treated for 6 months with LI 160 (a commercial hypericum extract) were found to have significantly increased numbers (50% more) of both 5-HT1A and 5-HT2A serotonin receptors, without changes in affinity.193 Human studies show electroencephalographic changes with St John's wort that are different from those seen with tricyclic antidepressants: shortening of evoked potential latencies and enhancement of theta and beta-2 regions of the resting electroencephalogram in the absence of sleep changes.194,195

There is strong evidence of efficacy in mild to moderate depression, as reviewed by Linde et al.171 That meta-analysis included 23 randomized trials with a total of 1757 outpatients, in which extracts of St John's wort alone (20 of 23 trials) or in combination with other herbs (3 of 23) were tested against placebo (15 trials) or antidepressant drugs (8 trials). Outcome was assessed by means of a pooled estimate of the "responder rate ratio" (response rate in the hypericum group vs the control group). St John's wort was reported to be clearly superior to placebo and comparable with conventional drug treatment, with lower side-effect and dropout rates in the hypericum group. Concerns raised in the article by Linde et al include the heterogeneity of patients, interventions, extract preparations, and diagnostic classifications among the various trials. A more recent review (but not meta-analysis) of 12 randomized trials (11 of which were included in the meta-analysis by Volz172) expressed similar concerns regarding the methods of the original studies, the possibility of subtherapeutic control drug dosing, and the variability of hypericum preparations. Overall, there are inadequate data regarding long-term use and efficacy in severe depression. There are concerns regarding the standardization and quality control of commercial preparations.196,197 Clearly, more research is needed to address these shortcomings in the literature.

Many commercial St John's wort products are standardized extracts (0.3% hypericin) of which 300 to 900 mg are given daily in 3 divided doses.171 This is approximately equivalent to 2 to 4 g of the dried herb.19 In general, fewer adverse effects are seen with hypericum than with conventional antidepressants,171,198 but they may include photodermatitis,199 delayed hypersensitivity, gastrointestinal tract upset, dizziness, dry mouth, sedation, restlessness, and constipation.19,171,200,201 There do not appear to be significant adverse effects on cardiac conduction with hypericum extracts.202 The use of St John's wort is contraindicated in pregnancy, lactation, exposure to strong sunlight, and pheochromocytoma.19 Because of the lack of information regarding the mechanism of action of H perforatum extracts, the potential for MAO inhibitor-like drug interactions cannot be excluded.19,203

Valerian
Valerian (Valeriana officalis L and Valeriana species) has a rich history of use throughout the world for a variety of indications, including as a sedative.20 Research on the mechanism of action has yielded contradictory findings. Extracts of valerian have affinity for GABAA receptors,204,205 likely because of the relatively high content of GABA itself that has been documented to be a constituent of valerian.206,207 The amount of GABA present in aqueous extracts of valerian is sufficient to induce release of GABA in synaptosomes and may also inhibit GABA reuptake.208,209 However, since GABA does not readily cross the blood-brain barrier, the relevance of these findings to central sedating effects is questionable. Other postulated mechanisms of action include inhibition of the catabolism of GABA by valerenolic acid and acetylvalerenolic acid210 and affinity for the 5-HTA receptor by another constituent of valerian, hydroxypinoresinal.211 Adenosine receptors may also be a target of valerian extracts.212

Animal behavioral tests with valerian show results consistent with other hypnotic agents such as the benzodiazepines,211, 213-222 as well as contradictory reports of anticonvulsant activity223,224 and possible antidepressant effects.219,225 Imaging with cerebral nuclear medicine scans in rats showed CNS depressant effects.211,226,227

Human clinical studies of valerian confirm a mild sedative effect,228 although the exact effects on sleep architecture, quality, and the electroencephalogram are inconsistent.229-235 These results are based on a relatively small number of subjects and do not evaluate the efficacy of valerian as a treatment for primary or secondary insomnia. There is no evidence to suggest that valerian is superior to existing hypnotic medications or other treatments for insomnia. This review found only 1 English-language report of a subjective anxiolytic effect.229

The dosage for valerian ranges from 2 to 3 g of the dried root given 3 times daily or at bedtime.211 Adverse effects include reports of hepatotoxic effects, although the offending preparations often contained a mixture of ingredients, making it difficult to draw definitive conclusions.236-240 There is currently insufficient information to recommend valerian in pregnancy and during lactation,241-243 although no reports of teratogenicity were found. The sedative effects of valerian may potentiate the effects of other CNS depressants,213, 221, 244, 245 and the usual precautions taken with other sedating agents also apply to valerian.

Herbal Remedies With CNS Effects

Capsicum
Commonly encountered in the form of chili or cayenne pepper, capsicum (Capsicum annuum L) has been used topically for pain relief in a variety of healing traditions.25,246 The active ingredients form a class referred to as the capsaicinoids, of which the most important is capsaicin.247,248 Capsaicin depletes substance P, thus inhibiting substance P-mediated pain transmission.249-252 Numerous trials have concluded that 0.075% capsaicin cream is a safe and effective treatment for painful diabetic neuropathy.253-258 There is also some evidence that 0.025% capsaicin cream may be useful in relieving the pain of postherpetic neuralgia.259-264 Several proprietary creams containing capsaicin (eg, Axsain and Zostrix) are available in North America. Traditionally, Capsicum has been ingested as a treatment for gastrointestinal tract complaints, such as colic and dyspepsia. In addition, it is thought to improve peripheral circulation in patients with cardiovascular conditions.19 These indications have not been investigated scientifically. Capsicum products used internally may elevate the secretion of catecholamines,265 thus caution with concurrent MAO inhibitor treatment is recommended.

Chaste Tree
Found in the Mediterranean and central Asia, chaste tree (Vitex Agnus-castus L) has been used medicinally since the times of ancient Greece and Rome to treat symptoms of premenstrual syndrome, mastodynia, menopause, hyperprolactinemia, and menstrual irregularity.266-269 The exact mechanism of action is unclear but likely involves modulating the prolactin axis.118,270,271 Affinity for D1 and D2 dopamine receptors has also been reported.272,273 These data point to a potential interaction with other dopaminergic drugs, such as the antipsychotics and metoclopramide.270

Siberian Ginseng
Siberian ginseng (Eleuthrococcus senticosus [Rupr and Maxim] Maxim), a member of the Araliaceae family and also known as eleuthero, is native to the northern parts of China, Japan, Korea, and eastern Russia. Used in traditional Chinese medicine for more than 400 years, the roots are thought to help fatigue and stress and to improve endurance.274,275 This type of "ginseng" must be distinguished from plants of the genus Panax (eg, Panax ginseng, Panax quinquefolius), which are discussed below. The eleutherosides contained in Siberian ginseng are thought to mediate the antifatigue and immunostimulatory properties attributed to this plant.19,274,276 Eleuthero should be used cautiously with sedative-hypnotic agents, as some studies report alteration of barbiturate-induced sleeping time.18,274,277

Herbal Remedies That May Cause Psychiatric Symptoms

Ginseng
Often confused with E senticosus (Siberian ginseng), P ginseng CA Meyer (ie, ginseng, Chinese ginseng, Korean ginseng) and P quinquefolius L (ie, Canadian ginseng, American ginseng) have a long list of indications, including treating stress and fatigue and improving endurance. Although many mechanisms of action have been postulated, it probably affects the hypothalamic-pituitary-adrenal axis, resulting in elevated plasma corticotropin and corticosteroid levels.278-284 One of the most common side effects is insomnia,285 while others include hypertension, diarrhea, restlessness, anxiety, and euphoria.286,287 Ginseng should be used with caution in patients with hypertension and diabetes and in conjunction with centrally acting medications.25,276 In addition, ginseng may potentiate the effect of MAO inhibitors,45,288-290 stimulants (including caffeine), and haloperidol.291

Yohimbe
Yohimbe (Pausinystalia yohimbe [K Schum]) is a botanical medicine derived from the bark of the P yohimbe (K Schum) tree.20 The active constituents are the alkaloids, notably one of the yohimbane derivatives called yohimbine.292 Yohimbine appears to act as an ?2-adrenoceptor antagonist,292,293 and the hydrochloride salt is used in the treatment of erectile dysfunction.294,295 Yohimbe bark is reputed to have aphrodisiac properties and is widely sold for this purpose.20

When administered to humans, yohimbine causes a variety of symptoms, including anxiety, nervousness, palpitations, and restlessness, as well as signs such as elevated 3-methoxy-4-hydroxyphenylglycol and cortisol levels.292, 296, 297 In fact, yohimbine is one of the agents commonly used to provoke panic attacks and anxiety in studies of the pathophysiology, psychopharmacology, and treatment of anxiety disorders.298, 299 Tricyclic antidepressants, medications with central a-adrenergic blocking properties, centrally acting sympathomimetics, MAO inhibitors, and antimuscarinic agents are all known to potentiate the action of yohimbine.292 Yohimbine also may contribute to psychotic symptoms,296 mania,300 and seizures,301 but these effects are not well documented.

Clearly, yohimbine-containing products have the potential to produce psychiatric symptoms, primarily anxiety or panic, especially in patients with preexisting panic disorder.302 While it is important to be aware of this in examining patients with anxiety who are also taking herbal remedies, many commercial products containing yohimbe bark actually have little or no yohimbine.303

Conclusions

We have reviewed the most common herbal products used in North America that are likely to be encountered in psychiatric practice. With the exception of St John's wort for depression and ginkgo for dementia, there is insufficient evidence to recommend the use of herbal medicines in the treatment of psychiatric illness. None of these herbal remedies is clearly superior to current conventional treatments. Because these products are widely available and often used by the general public, more clinical research is needed to establish safety and efficacy. A working knowledge of the pharmacological data and clinical literature is necessary to properly counsel, diagnose, and treat patients who may be using herbal products. The advances of modern medicine, science, and technology are greater than at any other time in history, and the rate of discovery of new knowledge and techniques is accelerating. However, the experience and healing traditions of other cultures, whether in less developed countries or in history, should not be ignored. Contemporary medical research may finally allow us to separate the traditional remedies that can effectively treat disease from those that are superstition and myth. In addition, research into the biochemical and pharmacological effects of these herbs may uncover novel treatments for psychiatric illness or yield fresh insights into basic disease mechanisms.

Author/Article Information

From the Clarke Institute of Psychiatry, Faculty of Medicine, University of Toronto (Dr Wong), Canadian College of Naturopathic Medicine (Mr Smith), and Faculty of Pharmacy, University of Toronto (Dr Boon), Toronto, Ontario; and Centre for Studies in Family Medicine, Department of Family Medicine, The University of Western Ontario, London (Dr Boon).

Excerpted from Alternative Medicine: An Objective Assessment, American Medical Association 2000

If you would like to purchase the book, Alternative Medicine: An Objective Assessment, click here.

References

1. Eisenberg DM, Kessler RC, Foster C, Norlock FE, Calkins DR, Delbanco TL. Unconventional medicine in the United States: prevalence, costs and patterns of use. N Engl J Med. 1993; 328:246-252.

2. Berger E. Canada Health Monitor Survey #9. Toronto, Ontario: Price Waterhouse; 1993.

3. The rise of alternative medicine. Pharm Pract. 1997;13:50.

4. Khaliq Y. Alternative medicine: what pharmacists need to know. Pharm Pract. 1997; 13:44-50, 83-85.

5. Blumenthal M, Brusse WR, Goldbert A, Gruenwald J, Hall T, Riggins CW, Rister RS, eds. The Complete German Commission E Monographs. Austin, Tex: American Botanical Council; 1998.

6. The Canadian Task Force on the Periodic Health Examination. The Canadian Guide to Clinical Preventive Health Care. Ottawa, Ontario: Health Canada; 1994.

7. US Preventive Services Task Force. Guide to Clinical Preventive Services. 2nd ed. Baltimore, Md: Williams & Wilkins; 1996.

8. O'Neill A. Danger and safety in medicines. Soc Sci Med. 1994;38:497-507.

9. Mills S. Safety awareness in complementary medicine. Complement Ther Med. 1996; 4:48-51.

10. Brinker F. An overview of conventional, experimental and botanical treatments of non-malignant prostate conditions. Br J Phytother. 1993/1994;3:154-176.

11. Ducker E-M, Kopanski L, Jarry H, Wuttke W. Effects of extracts from Cimicifuga racemosa on gonadotropin release in menopausal women and ovariectomized rats. Planta Med. 1991; 57:420-424.

12. Warnecke G. Beeinflussing klimakterisch beschwerden durch ein phytotherapeutikum. Med Welt. 1985;36:871-874.

13. Daiber W. Menopause symptoms: success without hormones. Arztliche Prax. 1983; 35:1946-1947.

14. Vorberg G. Treatment of menopause symptoms. Z Allgemeinmedizin. 1984;60:626-629.

15. Petho A. Menopause symptoms: is it possible to switch from hormone treatment to a botanical gynecologicum? Arztliche Prax. 1987; 47:1551-1553.

16. Lehmann-Willenbrock E, Riedel H. Klinische und endokrinologische untersuchungen zur therapie ovarieller ausfallserscheinungen nach hysterekomie unter belassung der adnexe. Zentralbl Gynakol. 1988;110:611-618.

17. Stoll W. Phytotherapeutikum beeinflust atrophisches Vaginalepithel. Therapeutikon. 1987;1:23-31.

18. Bradley P. British Herbal Compendium. Bournemouth, England: British Herbal Medicine Association; 1992.

19. Newall C, Anderson L, Phillipson J. Herbal Medicines: A Guide for Health Care Professionals. London, England: Pharmaceutical Press; 1996:296.

20. Tyler V. Herbs of Choice: The Therapeutic Use of Phytomedicinals. Binghamton, NY: Pharmaceutical Products Press; 1994:209.

21. Viola H, Wasowski C, Levi de Stein M, Wolfman C, Silveira R, Dajas F, Medina JH, Paladini AC. Apigenin, a component of Matricaria recutita flowers, is a central benzodiazepine receptors-ligand with anxiolyic effects. Planta Med. 1995;61:213-216.

22. Miller T, Wittstock U, Lindequist U, Teuscher E. Effects of some components of the essential oil of chamomile, Chamomilla recutita on histamine release from rat mast cells. Planta Med. 1996;62:60-61.

23. Berry M. The chamomiles. Pharm J. 1995;254:191-193.

24. Mann C, Staba EJ. The chemistry, pharmacology, and commercial formulations of chamomile. Herbs Spices Med Plants. 1984;1:235-280.

25. Mills S. The Essential Book of Herbal Medicine. 2nd ed. London, England: Penguin Books Ltd; 1991:677.

26. Paulsen E, Andersen K, Hausen B. Compositae dermatitis in a Danish dermatology department in one year. Contact Dermatitis. 1993;29:6-10.

27. Hausen B. The sensitizing capacity of Compositae plants, III: test results and cross reactions in Compositae-sensitive patients. Dermatologica. 1979;159:1-11.

28. Hausen B, Busker E, Carle R. The sensitizing capacity of Compositae plants, VII: experimental investigations with extracts of Chamomilla recutita (L.) Rauschert and Anthemis cotula L. Planta Med. 1984;50:229–234.

29. Vaddadi K. The use of gamma-linoleic acid and linoleic acid to differentiate between temporal lobe epilepsy and schizophrenia. Prostaglandins Med. 1981;27:313-323.

30. Stevens L, Zentall S, Abate M, Kuczek A, Burgess J. Omega-3 fatty acids in boys with behaviour, learning and health problems. Physiol Behav. 1996;59:915-920.

31. Stevens LJ, Zentall SS, Deck JL, Abate ML, Watkins BA, Lipp SR, Burgess JR. Essential fatty acid metabolism in boys with attention-deficit hyperactivity disorder. Am J Clin Nutr. 1995; 62:761-768.

32. Mitchell EA, Aman MG, Turbott SS, Manku M. Clinical characteristics and serum essential fatty acid levels in hyperactive children. Clin Paediatr. 1987;26:406-411.

33. Leung AY, Foster S. Encyclopedia of Common Natural Ingredients Used in Food, Drugs, and Cosmetics. 2nd ed. Toronto, Ontario: John Wiley & Sons Inc; 1996:649.

34. Briggs CJ. Evening primrose. Rev Pharm Can. 1986;119:249-254.

35. Manku M, Morse-Fischer N, Horrobin D. Changes in human plasma essential fatty acid levels as a result of administration of linoleic acid and gamma-linolenic acid. Eur J Clin Nutr. 1988;42:55-60.

36. Vaddadi K, Courtney P, Gilleard C, Manku M, Horrobin D. A double-blind trial of essential fatty acid supplementation in patients with tardive dyskinesia. Psychiatry Res. 1989; 27:313-323.

37. Holman C, Bell A. A trial of evening primrose oil in the treatment of chronic schizophrenia. J Orthomol Psychiatry. 1983;12:302-304.

38. Blackburn M. Use of efamol (oil of evening primrose) for depression and hyperactivity in children. In: Liss AR, ed. Pathophysiology and Roles in Clinical Medicine. New York, NY: Alan R. Liss Inc; 1990:345-349.

39. Horrobin DF. Gammalinolenic acid: an intermediate in essential fatty acid metabolism with potential as an ethical pharmaceutical and as a food. Rev Contemp Pharmacother. 1990;1:1–45.

40. Chenoy R, Hussain S, Tayob Y, O'Brien P, Moss M, Morse P. Effect of oral gamalinolenic acid from evening primrose oil on menopausal flushing. BMJ. 1994;38:501-503.

41. Joe LA, Hart LL. Evening primrose oil in rheumatoid arthritis. Ann Pharmacother. 1993;27:1475-1477.

42. Kleijnen J. Evening primrose oil [see comments]. BMJ. 1994;309:824-825.

43. Barber HJ. Evening primrose oil: a panacea? Pharm J. 1988;240:723-725.

44. Cant A, Shay J, Horrobin DF. The effect of maternal supplementation with linolenic and gammalinolenic acids on the fat composition and content of human milk. J Nutr Sci Vitaminol. 1991;37:573-579.

45. Stockley I. Drug Interactions: A Sourcebook of Adverse Interactions, Their Mechanisms, Clinical Importance, and Management. 3rd ed. Cambridge, England: Blackwell Scientific Press; 1994.

46. Major RT. The ginkgo, the most ancient living tree. Science. 1967;157:1270-1273.

47. Foster S. Ginkgo, Ginkgo biloba. Austin, Tex: American Botanical Council; 1990.

48. Gaby AR. Ginkgo biloba extract: a review. Altern Med Rev. 1996;1:236-242.

49. Houghton PJ. Ginkgo. Pharm J. 1994; 253:122-123.

50. Sticher O. Ginkgo biloba -- analysis and dosage forms [in German]. Pharm Unserer Zeit. 1992;21:253-265.

51. Sticher O. Quality of ginkgo preparations. Planta Med. 1993;59:2-11.

52. Kleijnen J, Knipschild P. Ginkgo biloba for cerebral insufficiency. Br J Clin Pharmacol. 1992;34:352-358.

53. Kleijnen J, Knipschild P. Ginkgo biloba. Lancet. 1992;340:1136-1139.

54. Jung F, Mrowietz C, Kiesewetter H, Wenzel E. Effect of Ginkgo biloba on fluidity of blood and peripheral microcirculation in volunteers. Arzneimittelforsch. 1990;40:589-593.

55. Liberti L. Ginkgo. Lawrence Rev Nat Products. February 1988.

56. Le Poncin Lafitte M, Rapin J, Rapin JR. Effects of Ginkgo biloba on changes induced by quantitative cerebral microembolization in rats. Arch Int Pharmacodyn Ther. 1980;243:236-244.

57. Gaby AR. Ginkgo biloba extract: a review. Alternative Med Rev. 1996;1:236-242.

58. Krieglstein J, Beck T, Seibert A. Influence of an extract of Ginkgo biloba on cerebral blood flow and metabolism. Life Sci. 1986;39:2327-2334.

59. Tighilet B, Lacour M. Pharmacological activity of the Ginkgo biloba extract (EGb 761) on equilibrium function recovery in the unilateral vestibular neurectomized cat. J Vestib Res. 1995;5:187-200.

60. Stucker O, Pons C, Duverger JP, Drieu K. Effects of Ginkgo biloba extract (EGb 761) on arteriolar spasm in a rat cremaster muscle preparation. Int J Microcirc Clin Exp. 1996;16:98-104.

61. DeFeudis FV. Ginkgo biloba Extract (EGb 761): Pharamacological Activities and Clinical Applications. Paris, France: Elsevier; 1991:3-5.

62. Vilain B, DeFeudis FV, Clostre F. Effect of an extract of Ginkgo biloba on the isolated ileum of the guinea pig. Gen Pharmacol. 1982; 13:401-405.

63. Puglisi L, Salvadori S, Gabrielli G, Pasargiklian R. Pharmacology of natural compounds, I: smooth muscle relaxant activity induced by a Ginkgo biloba L. extract on guinea-pig trachea. Pharmacol Res Commun 1988;20:573-589.

64. Delaflotte S, Auguet M, DeFeudis FV, Baranes J, Clostre F, Drieu K, Braquet P. Endothelium-dependent relaxations of rabbit isolated aorta produced by carbachol and by Ginkgo biloba extract. Biomed Biochim Acta. 1984;43(suppl 8–9):S212-S216.

65. Auguet M, DeFeudis FV, Clostre F. Effects of Ginkgo biloba on arterial smooth muscle responses to vasoactive stimuli. Gen Pharmacol. 1982;13:169-171.

66. Auguet M, DeFeudis FV, Clostre F, Deghenghi R. Effects of an extract of Ginkgo biloba on rabbit isolated aorta. Gen Pharmacol. 1982;13:225-230.

67. Hellegouarch A, Baranes J, Clostre F, Drieu K, Braquet P, DeFeudis FV. Comparison of the contractile effects of an extract of Ginkgo biloba and some neurotransmitters on rabbit isolated vena cava. Gen Pharmacol. 1985;16:129-132.

68. Pincemail J, Deby C. Antiradical properties of Ginkgo biloba extract [in French]. Presse Med. 1986;15:1475-1479.

69. Robak J, Gryglewski RJ. Flavonoids are scavengers of superoxide anions. Biochem Pharmacol. 1988;37:837-841.

70. Smith PF, Maclennan K, Darlington CL. The neuroprotective properties of the Ginkgo biloba leaf: a review of the possible relationship to platelet-activating factor (PAF). J Ethno-phar-macol. 1996;50:131-139.

71. Braquet P. The ginkgolides: potent platelet-activating factor antagonists isolated from Ginkgo biloba L.: chemistry, pharmacology and clinical applications. Drugs Future. 1987;12:643-699.

72. Guinot P, Caffrey E, Lambe R, Darragh A. Tanakan inhibits platelet-activating-factor-induced platelet aggregation in healthy male volunteers. Haemostasis. 1989;19:219-223.

73. Bourgain RH, Maes L, Andries R, Braquet P. Thrombus induction by endogenic PAF-acether and its inhibition by Ginkgo biloba extracts in the guinea pig. Prostaglandins. 1986;32:142-144.

74. Bourgain RH, Andries R, Braquet P. Effect of ginkgolide PAF-acether antagonists on arterial thrombosis. Adv Prostaglandin Thromboxane Leukot Res. 1987;17B:815-817.

75. Belougne E, Aguejouf O, Imbault P, Azougagh Oualane F, Doutremepuich F, Droy-LeFaix MT, Doutremepuich C. Experimental thrombosis model induced by laser beam: application of aspirin and an extract of Ginkgo biloba: EGb 761. Thromb Res. 1996;82:453-458.

76. Clostre F. From the body to the cell membrane: the different levels of pharmacological action of Ginkgo biloba extract [in French]. Presse Med. 1986;15:1529-1538.

77. Rapin JR, Le Poncin Lafitte M. Cerebral glucose consumption: the effect of Ginkgo biloba extract [in French]. Presse Med. 1986;15:1494-1497.

78. Spinnewyn B, Blavet N, Clostre F. Effects of Ginkgo biloba extract on a cerebral ischemia model in gerbils [in French]. Presse Med. 1986; 15:1511-1515.

79. Spinnewyn B, Blavet N, Clostre F, Bazan N, Braquet P. Involvement of platelet-activating factor (PAF) in cerebral post-ischemic phase in Mongolian gerbils. Prostaglandins. 1987; 34:337-349.

80. Koc RK, Akdemir H, Kurtsoy A, Pasaoglu H, Kavuncu I, Pasaoglu A, Karakucuk I. Lipid peroxidation in experimental spinal cord injury: comparison of treatment with Ginkgo biloba, TRH and methylprednisolone. Res Exp Med. 1995;195:117-123.

81. Otamiri T, Tagesson C. Ginkgo biloba extract prevents mucosal damage associated with small-intestinal ischaemia. Scand J Gastroenterol. 1989;24:666-670.

82. Seif-El-Nasr M, El-Fattah AA. Lipid peroxide, phospholipids, glutathione levels and superoxide dismutase activity in the rat brain after ischemia: effect of Ginkgo biloba extract. Pharmacol Res. 1995;32:273-278.

83. Birkle DL, Kurian P, Braquet P, Bazan NG. Platelet-activating factor antagonist BN52021 decreases accumulation of free polyunsaturated fatty acid in mouse brain during ischemia and electroconvulsive shock. J Neurochem. 1988;51:1900-1905.

84. Panetta T, Marcheselli VL, Braquet P, Spinnewyn B, Bazan NG. Effects of a platelet activating factor antagonist (BN 52021) on free fatty acids, dicylglycerols, polyphosphoinositides and blood flow in the gerbil brain: inhibition of ischemia-reperfusion induced cerebral injury. Biochem Biophys Res Commun. 1987;149:580-587.

85. Haramaki N, Aggarwal S, Kawabata T, Droy-Lefaix MT, Packer L. Effects of natural antioxidant Ginkgo biloba extract (EGB 761) on myocardial ischemia-reperfusion injury. Free Radical Biol Med. 1994;16:789-794.

86. Oyama Y, Chikahisa L, Ueha T, Kanemaru K, Noda K. Ginkgo biloba extract protects brain neurons against oxidative stress induced by hydrogen peroxide. Brain Res. 1996;712:349-352.

87. Oyama Y, Fuchs PA, Katayama N, Noda K. Myricetin and quercetin, the flavonoid constituents of Ginkgo biloba extract, greatly reduce oxidative metabolism in both resting and Ca(2+)-loaded brain neurons. Brain Res. 1994;635:125-129.

88. Shen JG, Zhou DY. Efficiency of Ginkgo biloba extract (EGb 761) in antioxidant protection against myocardial ischemia and reperfusion injury. Biochem Mol Biol Int. 1995;35:125-134.

89. Szabo ME, Droy-Lefaix MT, Doly M, Braquet P. Free radical-mediated effects in reperfusion injury: a histologic study with superoxide dismutase and EGb 761 in rat retina. Ophthalmic Res. 1991;23:225-234.

90. Maitra I, Marcocci L, Droy-Lefaix MT, Packer L. Peroxyl radical scavenging activity of Ginkgo biloba extract EGb 761. Biochem Pharmacol. 1995;49:1649-1655.

91. Rong Y, Geng Z, Lau BH. Ginkgo biloba attenuates oxidative stress in macrophages and endothelial cells. Free Radical Biol Med. 1996; 20:121-127.

92. Pincemail J, Dupuis M, Nasr C, Hans P, Haag-Berrurier M, Anton R, Deby C. Superoxide anion scavenging effect and superoxide dismutase activity of Ginkgo biloba extract. Experientia. 1989;45:708-712.

93. Stoll S, Scheuer K, Pohl O, Muller WE. Ginkgo biloba extract (EGb 761) independently improves changes in passive avoidance learning and brain membrane fluidity in the aging mouse. Pharmacopsychiatry. 1996; 29:144-149.

94. Gesser B, Voelp A, Klasser M. Study of the long-term action of a Ginkgo biloba extract on vigilance and mental performance as determined by means of quantitative pharmaco-EEG and psychometric measurements. Arzneimittelforschung. 1985;35:1459-1465.

95. Rai GS, Shovlin C, Wesnes KA. A double-blind, placebo controlled study of Ginkgo biloba extract ("tanakan") in elderly outpatients with mild to moderate memory impairment. Curr Med Res Opin. 1991;12:350-355.

96. Allain H, Raoul P, Lieury A, LeCoz F, Gandon JM, d'Arbigny P. Effect of two doses of Ginkgo biloba extract (EGb 761) on the dual-coding test in elderly subjects. Clin Ther. 1993;15:549-558.

97. Hindmarch I. Activity of Ginkgo biloba extract on short-term memory [in French]. Presse Med. 1986;15:1592-1594.

98. Subhan Z, Hindmarch I. The psychopharmacological effects of Ginkgo biloba extract in normal healthy volunteers. Int J Clin Pharmacol Res. 1984;4:89-93.

99. Semlitsch HV, Anderer P, Saletu B, Binder GA, Decker KA. Cognitive psychophysiology in nootropic drug research: effects of Ginkgo biloba on event-related potentials (P300) in age-associated memory impaiment. Pharmacopsychiatry. 1995;28:134-142.

100. Warburton DM. Clinical psychopharmacology of Ginkgo biloba extract [in French]. Presse Med. 1986;15:1595-1604.

101. Winter E. Effects of an extract of Ginkgo biloba on learning and memory in mice. Pharmacol Biochem Behav. 1991;38:109-114.

102. Wesnes K, Simmons D, Rock M, Simpson P. A double-blind placebo-controlled trial of Gingko biloba extract in the treatment of idiopathic cognitive impairment in the elderly. Hum Psychopharmacol. 1987;2:159-169.

103. Gessner B, Voelp A, Klasser M. Study of the long-term action of a Ginkgo biloba extract on vigilance and mental performance as determined by means of quantitative pharmaco-EEG and psychometric measurements. Arzneimittelforschung. 1985;35:1459-1465.

104. Kanowski S, Herrmann WM, Stephan K, Wierich W, Horr R. Proof of efficacy of the Ginkgo biloba special extract EGb 761 in outpatients suffering from mild to moderate primary degenerative dementia of the Alzheimer type or multi-infarct dementia. Phytomedicine. 1997;4:3-13.

105. Hofferberth B. The efficacy of EGb 761 in patients with senile dementia of the Alzheimer type: a double-blind, placebo-controlled study on different levels of investigation. Hum Psychopharmacol. 1994;9:215-222.

106. Taillandier J, Ammar A, Rabourdin JP, Ribeyre JP, Pichon J, Niddam S, Pierart H. Treatment of cerebral aging disorders with Ginkgo biloba extract: a longitudinal multicenter double-blind drug vs placebo study [in French]. Presse Med. 1986;15:1583-1587.

107. Le Bars PL, Katz MM, Berman N, Turan M, Freedman AM, Schatzberg AF. A placebo-controlled, double-blind, randomized trial of an extract of Ginkgo biloba for dementia. JAMA. 1997;278:1327-1332.

108. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Revised Third Edition. Washington, DC: American Psychiatric Association; 1987.

109. Schubert H, Halama P. Depressive episode primarily unresponsive to therapy in elderly patients: efficacy of Ginkgo biloba extract (EGb 761) in combination with antidepressants. Geriatr Forschung. 1993;3:45-53.

110. White HL, Scates PW, Cooper BR. Extracts of Ginkgo biloba leaves inhibit monoamine oxidase. Life Sci. 1996;58:1315-1321.

111. Attella MJ, Hoffman SW, Stasio MJ, Stein DG. Ginkgo biloba extract facilitates recovery from penetrating brain injury in adult male rats. Exp Neurol. 1989;105:62-71.

112. Rodriguez de Turco EB, Droy-Lefaix MT, Bazan NG. Decreased electroconvulsive shock-induced diacylglycerols and free fatty acid accumulation in the rat brain by Ginkgo biloba extract (EGb 761): selective effect in hippocampus as compared with cerebral cortex. J Neurochem. 1993;61:1438-1444.

113. Tyler VE. The Honest Herbal. 3rd ed. Philadelphia, Pa: George F. Stickley Co; 1993.

114. Lepoittevin JP, Benezra C, Asakawa Y. Allergic contact dermatitis to Ginkgo biloba L.: relationship with urushiol. Arch Dermatol Res. 1989;281:227-230.

115. Mitchell JC, Maibach HI, Guin J. Leaves of Ginkgo biloba not allergenic for Toxicodendron-sensitive subjects. Contact Dermatitis. 1981;7:47-48.

116. Rowin J, Lewis SL. Spontaneous bilateral subdural hematoma associated with chronic Gingko biloba ingestion. Neurology. 1996;46:1775-1776.

117. Rosenblatt M, Mindel J. Spontaneous hyphema associated with ingestion of Ginkgo biloba extract. N Engl J Med. 1997;336:1108.

118. Weiss RF. Herbal Medicine. 6th ed. Gothen-burg, Sweden: AB Arcanum; 1988:362.

119. Hoffmann D. Holistic Herbal. Rockport, Calif: Element Books; 1996:256.

120. Chevalier A. The Encyclopedia of Medicinal Plants. London, England: Readers Digest; 1996:336.

121. Bove M. An Encyclopedia of Natural Healing for Children and Infants. New Canaan, Conn: Keats; 1996:303.

122. Hansel R, Wohlfart R. Narcotic action of 2-methyl-3-butene-2-ol contained in the exhalation of hops. Z Naturforschung. 1980; 35:1096-1097.

123. O'Donovan W. Hops dermatitis. Lancet. 1924;2:597.

124. Newmark FM. Hops allergy and terepene sensitivity: an occupational disease. Ann Allergy. 1978;41:311-312.

125. Singh Y, Blumenthal M. Kava: an overview. Herbalgram. 1997;39:34-54.

126. Bone K. Kava: a safe herbal treatment for anxiety. Br J Phytother. 1993/1994;3:147-153.

127. Jussogie A, Scmiz A, Heimke C. Kavapyrone extract enriched from Piper methysticum as modulator of the GABA binding site in different regions of the rat brain. Psychopharmacology. 1994;116:469-474.

128. Davies L, Drew C, Duffield P, Johnston GA, Jamieson DD. Kava pyrones and resin: studies on GABA(A), GABA (B), and benzodiazepine binding sites in the rodent brain. Pharmacol Toxicol. 1992;71:120-126.

129. Glietz J, Beile A, Peters T. Kawain inhibits vetradine-activated voltage-dependent Na+ channels in synaptosomes prepared from rat cerebral cortex. Neuropharmacology. 1995;24:1133-1138.

130. Backhauss C, Krieglstein J. Extract of kava and its methysticin constituents protect brain tissues against ischaemic damage in rodents. Eur J Pharmacol. 1992;215:265-269.

131. 0Kretzschmar R, Meyer HJ. Comparative experiments on the anticonvulsant efficacy of Piper methysticum pyrone bonds [in German]. Arch Int Pharmacodyn. 1969;177:261-277.

132. Kretzschmar R, Meyer HJ, Teschendorf HJ, Zollner B. Antagonistic effect of natural 5,6-hydrated kava pyrones on strychnine poisoning and experimental local tetanus [in German]. Arch Int Pharmacodyn. 1969;182:251-268.

133. Meyer HJ, Kretzschmar R. Research on the relationship between molecular structure and pharmacological effect of aryl-substituted 4-methoxy pyrones of the kava pyrone type [in German]. Arzneimittelforschung. 1969; 19:617-623.

134. Hansel R. Kava-kava in modern drug research: portrait of a medicinal plant. Q Rev Nat Med. Winter 1996:259-274.

135. Gebner B, Cnota P. Extract of kava-kava rhizome in comparison with diazepam and placebo. Z Phytother. 1994;15:30-37.

136. Volz HP, Kieser M. Kava-kava extract WS 1490 versus placebo in anxiety disorders: a randomized placebo-controlled 25-week outpatient trial. Pharmacopsychiatry. 1997;30:1-5.

137. Lehmann E, Kinzler E, Friedemann J. Efficacy of a special kava extract (Piper methysticum) in a patients with states of anxiety, tension and excitedness of non-mental origin: a double-blind placebo-controlled study of four weeks treatment. Phytomedicine. 1996;3:113-119.

138. Russell P, Bakker D, Singh N. The effects of kava on alerting and speed of access of information from long-term memory. Bull Psychonom Soc. 1987;25:236-237.

139. Munte T, Heinze H, Matzke M, Steitz J. Effects of oxazepam and an extract of kava roots (Piper methysticum) on event-related potentials in a word recognition task. Neuropsychobiology. 1993;27:46-53.

140. Heinze HJ, Munthe TF, Stelz J, Matzke M. Pharmacopsychological effects of oxazepam and kava extract in a visual search paradigm assessed with event related potentials. Pharmacopsychiatry. 1994;27:224-230.

141. Ruze P. Kava-induced dermapathy. Lancet. 1990;335:1142-1145.

142. Almeida JC, Grimsley EW. Coma from the health food store: interaction between kava kava and alprazolam. Annu Int Med. 1996;125:940-941.

143. Jamieson D, Duffield P. Positive interaction to kava in mice. Clin Exp Pharmacol Physiol. 1990;17:509-514.

144. Herberg K. Effect of kava-special extract ws 1490 combined with ethyl alcohol on safety-relevant performance parameters. Blutalkohol. 1993;30:96-105.

145. Soulimani R, Fleurentin J, Mortier F, Misslin R, Derrieu G, Pelt JM. Neurotropic action of the hydroalcoholic extract of Melissa officianalis in the mouse. Planta Med. 1991;57:105-109.

146. Dressing H, Riemann D, Low M. Insomnia: are valerian/balm combinations of equal value to benzodiazepine? Therapiewoche. 1992;42:726-736.

147. Brinker F. Inhibition of endocrine function by botanical agents. J Naturopath Med. 1990;1:10-18.

148. Auf'mkolk M, Ingbar JC, Amir SM, Winterhoff H, Sourgens H, Hesch RD, Ingbar SH. Inhibition by certain plant extracts of the binding and adenylate cyclase stimulatory effect of bovine thyrotropin in human thyroid membranes. Endocrinology. 1984;115:527-534.

149. Auf'mkolk M, Kohrle J, Gumbinger H, Winterhoff H, Hesch RD. Antihormonal effects of plant extracts: iodothyronine deiodinase of rat liver is inhibited by extracts and secondary metabolites of plants. Horm Metab Res. 1984;16:188-192.

150. Auf'mkolk M, Ingbar J, Kubota K, Amir SM, Ingbar SH. Extracts and auto-oxidized constituents of certain plants inhibit the receptor-binding and the biological activity of Graves' immunoglobulin. Endocrinology. 1985; 116:1687-1693.

151. Bergner P. Passion flower. Med Herbalism. 1995;7:13–14, 26.

152. Flynn J. The herbal management of stress. Aust J Med Herbalism. 1996;8:15-18.

153. European Scientific Cooperative on Phytotherapy. Passiflora Herba/Passiflora: ESCOP Monographs: Fascicule 4. Exeter, England: European Scientific Cooperative on Phytotherapy; 1997:58.

154. Soulimani R, Younos C, Jarmouni S, Bousta D, Misslin R, Mortier F. Behavioural effects of Passiflora incarnata L. and its indole alkaloid and flavonoid derivatives and maltol in the mouse. J Ethnopharmacol, 1997;57:11-20.

155. Della Loggia R, Tubaro A, Redaelli C. Evaluation of the activity on the mouse CNS of several plant extracts and a combination of them [in Italian]. Riv Neurol. 1981;51:297-310.

156. Speroni E, Minghetti A. Neuropharmacological activity of extracts from Passiflora incarnata. Planta Med. 1988;54:488-491.

157. Wolfman C, Viola H, Paladini A, Dajas F, Medina JH. Possible anxiolytic effects of chrysin, a central benzodiazepine receptor ligand isolated from Passiflora coerulea. Pharmacol Biochem Behav. 1994;47:1-4.

158. Meier B. Passiflora incarnata L. -- passion flower: portrait of a medicinal plant. Q Rev Nat Med.1985;3:191-202.

159. Aoyagi N, Kimura R, Murata T. Studies on Passiflora incarnata dry extract, I: isolation of maltol and pharmacological action of maltol and ethyl maltol. Chem Pharm Bull. 1974; 22:1008-1013.

160. Bourin M, Bougerol T, Guitton B, Broutin E. A combination of plant extracts in the treatment of outpatients with adjustment disorder with anxious mood: controlled study versus placebo. Fundam Clin Pharmacol. 1997;11:127-132.

161. Smith GW, Chalmers TM, Nuki G. Vasculitis associated with herbal preparation containing Passiflora extract. Br J Rheumatol. 1993; 32:87?88.

162. Solbakken AM, Rorbakken G, Gundersen T. Nature medicine as intoxicant [in Norwegian]. Tidsskr Nor Laegeforen. 1997;117:1140-1141.

163. Foster S, Chongxi Y. Herbal Emissaries: Bringing Chinese Herbs to the West. Rochester, Vt: Healing Arts Press; 1992:356.

164. Wren R. Potter's New Encyclopedia of Botanical Drugs and Preparations. Saffron Walden, England: CW Daniel Co; 1988:362.

165. De Smet P. Scutellaria species. In: De Smet P, Keller K, Hansel R, Chandler R, eds. Adverse Effects of Herbal Drugs. Heidelberg, Germany: Springer-Verlag; 1993:289–296.

166. Perharic L, Shaw D, Colbridge M, House I, Leon C, Murray V. Toxicological problems resulting from exposure to traditional remedies and food supplements. Drug Safety. 1994;11:284-294.

167. De Smet P. Notes added in proof. In: De Smet P, Keller K, Hansel R, Chandler R, eds. Adverse Effects of Herbal Drugs. Heidelberg, Germany: Springer-Verlag; 1996:229-240.

168. D'Arcy P. Adverse reactions and interactions with herbal medicines, part 2: drug interactions. Adverse Drug React Toxicol Rev. 1993;12:147-162.

169. Murray MT. The Healing Power of Herbs. Rocklin, Calif: Prima Publishing; 1992:246.

170. Upton R. St. John's wort (Hypericum perforatum). In: Upton R, ed. American Herbal Pharmacopoeia and Therapeutic Compendium. Santa Cruz, Calif: American Herbal Pharmacopoeia; 1997.

171. Linde K, Ramirez G, Mulrow CD, Pauls A, Weidenhammer W, Melchart D. St John's wort for depression: an overview and meta-analysis of randomised clinical trials [see comments]. BMJ. 1996;313:253-258.

172. Volz H-P. Controlled clinical trials of hypericum extracts in depressed patients: an overview. Pharmacopsychiatry. 1997;30:72-76.

173. Wagner H, Bladt S. Pharmaceutical quality of hypericum extracts. J Geriatr Psychiatry Neurol. 1994;7(suppl 1):S65-S68.

174. Kartnig T, Gobel I, Heydel B. Production of hypericin, pseudohypericin and flavonoids in cell cultures of various Hypericum species and their chemotypes. Planta Med. 1996;62:51–53.

175. Staffeldt B, Kerb R, Brockmoller J, Ploch M, Roots I. Pharmacokinetics of hypericin and pseudohypericin after oral intake of the hypericum perforatum extract LI 160 in healthy volunteers. J Geriatr Psychiatry Neurol. 1994;7(suppl 1):S47-S53.

176. Kerb R, Brockmoller J, Staffeldt B, Ploch M, Roots I. Single-dose and steady-state pharmacokinetics of hypericin and pseudohypericin. Antimicrob Agents Chemother. 1996;40:2087–2093.

177. Nahrstedt A, Butterweck V. Biologically active and other chemical constitents of the herb of Hypericum perforatum L. Pharmacopsychiatry. 1997;30:129-134.

178. Miskovsky P, Sureau F, Chinsky L, Turpin P-Y. Subcellular distribution of hypericin in human cancer cells. Photochem Photobiol. 1995;62:546-549.

179. Cott JM. In vitro receptor binding and enzyme inhibition by Hypericum perforatum extract. Pharmacopsychiatry. 1997;30:108-112.

180. Cott J. NCDEU update: natural product formulations available in Europe for psychotropic indications. Psychopharmacol Bull. 1995;31:745-751.

181. Perovic S, Muller WE. Pharmacological profile of hypericum extract: effect on serotonin uptake by postsynaptic receptors. Arzneimittelforschung. 1995;45:1145-1148.

182. Muller WE, Rolli M, Schafer C, Hafner U. Effects of hypericum extract (LI 160) in biochemical models of antidepressant activity. Pharmacopsychiatry. 1997;30:102-107.

183. Muller WE, Rossol R. Effects of hypericum extract on the expression of serotonin receptors. J Geriatr Psychiatry Neurol. 1994;7(suppl 1):S63-S64.

184. Baureithel KH, Buter KB, Engesser A, Burkard W, Schaffner W. Inhibition of benzodiazepine binding in vitro by amentoflavone, a constituent of various species of 186 Hypericum. Pharm Acta Helv. 1997;72:153-157.

185. Von HM, Zoller M. Antidepressive effect of a Hypericum extract. Drug Res. 1984;34:918-920.

186. Suzuki O, Katsumata Y, Oya M, Bladt S, Wagner H. Inhibition of monoamine oxidase by hypericin. Planta Med. 1984;50:272-274.

187. Bladt S, Wagner H. Inhibition of MAO by fractions and constituents of hypericum extract. J Geriatr Psychiatry Neurol. 1994;7(suppl 1):S57-S59.

188. Demisch L, Holzl J, Gollnick B, Kacmarczyk P. Identification of MAO-type-A inhibitors in Hypericum perforatum L. (Hyperforat). Pharmacopsychiatry. 1989;22:194.

189. Thiede HM, Walper A. Inhibition of MAO and COMT by hypericum extracts and hypericin. J Geriatr Psychiatry Neurol. 1994;7(suppl 1):S54-S56.

190. Von SNO, Weischer ML. Experimental animal studies of the psychotropic activity of a Hypericum extract. Drug Res. 1987;37:10-13.

191. Ozturk Y. Testing the antidepressant effects of Hypericum species on animal models. Pharmacopsychiatry. 1997;30:125-128.

192. Butterwick V, Wall A, Lieflander-Wulf U, Winterhoff H, Nahrstedt A. Effects of the total extract and fractions of Hypericum perforatum in animal assays for antidepressant activity. Pharmacopsychiatry. 1997;30:117-124.

193. Teufel-Mayer R, Gleitz J. Effects of long-term administration of hypericum extracts on the affinity and density of the central serotoninergic 5-HT1A and 5-HT2A receptors. Pharmacopsychiatry. 1997;30:113-116.

194. Schulz H, Jobert M. Effects of hypericum extract on the sleep EEG in older volunteers. J Geriatr Psychiatry Neurol. 1994;7(suppl 1):S39-S43.

195. Johnson D, Ksciuk H, Woelk H, Sauerwein-Giese E, Frauendorf A. Effects of hypericum extract LI 160 compared with maprotiline on resting EEG and evoked potentials in 24 volunteers. J Geriatr Psychiatry Neurol. 1994; 7(suppl 1):S44-S46.

196. De Smet PA, Nolen WA. St John's wort as an antidepressant [see comments]. BMJ. 1996;313:241-242.

197. Jenike MA. Editorial. J Geriatr Psychiatry Neurol. 1994;7(suppl 1):S1.

198. Vorbach EU, Arnoldt KH, Hubner W-D. Efficacy and tolerability of St. John's wort extract LI 160 versus imipramine in patients with severe depressive episodes according to ICD-10. Pharmacopsychiatry. 1997;30:81-85.

199. Brockmoller J, Reum T, Bauer S, Kerb R, Hubner W-D, Roots I. Hypericin and pseudohypericin: pharmacokinetics and effects on photosensitivity in humans. Pharmacopsychiatry. 1997;30:94-101.

200. Harrer G, Hubner WD, Podzuweit H. Effectiveness and tolerance of the hypericum extract LI 160 compared to maprotiline: a multicenter double-blind study. J Geriatr Psychiatry Neurol. 1994;7(suppl 1):S24-S28.

201. Woelk H, Burkard G, Grunwald J. Benefits and risks of the hypericum extract LI 160: drug monitoring study with 3250 patients. J Geriatr Psychiatry Neurol. 1994;7:S34-S38.

202. Czekalla J, Gastpar M, Hubner W-D, Jager D. The effect of hypericum extract on cardiac conduction as seen in the electrocardiogram compared to that of imipramine. Pharmacopsychiatry. 1997;30:86-88.

203. Gillis MC, ed. Compendium of Pharmaceuticals and Specialties. 31st ed. Ottawa, Ontario: Canadian Pharmaceutical Association; 1998.

204. Holzl J, Godau P. Receptor binding studies with Valeriana officinalis on the benzodiazepine receptor. Planta Med. 1989;55:642.

205. Mennini T, Bernasconi P, Bombardelli E, Morazzoni P. In vitro study on the interaction of extracts and pure compounds from Valeriana officinalis roots with GABA, benzodiazepine and barbiturate receptors in rat brain. Fitoterapia. 1993;64:291-300.

206. Cavadas C, Araujo I, Cotrim MD, Amaral T, Cunha AP, Macedo T, Ribeiro CF. In vitro study on the interaction of Valeriana officinalis L. extracts and their amino acids on GABAA receptor in rat brain. Arzneimittelforschung 1995;45:753-755.

207. Bodesheim U, Holzl J. Isolation and receptor binding properties of alkaloids and lignans from V. officinalis L. Pharmazie. 1997; 52:386–391.

208. Santos MS, Ferreira F, Faro C, Pires E, Carvalho AP, Cunha AP, Macedo T. The amount of GABA present in aqueous extracts of valerian is sufficient to account for [3H]GABA release in synaptosomes. Planta Med. 1994;60:475-476.

209. Santos MS, Ferreira F, Cunha AP, Carvalho AP, Ribeiro CF, Macedo T. Synaptosomal GABA release as influenced by valerian root extract: involvement of the GABA carrier. Arch Int Pharmacodyn Ther. 1994;327:220-231.

210. Riedel E, Hansel R, Ehrke G. Inhibition of GABA catabolism by valerenic acid derivatives. Planta Med. 1982;46:219-220.

211. Proposals for a European Monograph on the Medicinal Use of Valerianae radix (Valerian Root). Brussels, Belgium: European Scientific Cooperative for Phytotherapy; 1990.

212. Holzl J. Baldrian: Ein mittel gegen Schlafstorungen und Nervositat. Dtsch Apoth Zeitung. 1996;136:751-759.

213. Hendriks H, Bos R, Woerdenbag HJ, Koster AS. Central nervous system depressant activity of valerenic acid in the mouse. Planta Med. 1985;1:28-31.

214. Hendricks H, Bos R, Allersma DP, Malingre TM, Koster AS. Pharmacological screening of valerenal and some other components of essential oil of Valeriana officinalis. Planta Med. 1981;42:62-68.

215. Veith J, Schneider G, Lemmer B, Willems M. The influence of some degradation products of valpotriates on the motor activity of light-dark synchronized mice. Planta Med. 1986;3:179-183.

216. Wagner H, Jurcic K, Schaette R. Comparative studies on the sedative action of Valeriana extracts, valpotriates and their degradation products. Planta Med. 1980;36:358-365.

217. Hansel R, Keller K, Rimpler H, Schneider G. Valeriana. In: Hagers Handbuch der Parmzeutischen Praxis.5th ed. Berlin, Germany: Springer-Verlag; 1994:1067-1095.

218. Torrent MT, Iglesias J, Adzet T. Valoracion experimental de la actividad sedante de la tinctura de Valeriana officinalis L. Circ Farmaceut. 1972;30:107-112.

219. Sakamoto T, Mitana Y, Nakajima K. Psychotropic effects of Japanese valerian root extracts. Chem Pharm Bull. 1992;40:758-761.

220. Hikino H, Hikino Y, Kobinata H, Aizawa A. Sedative properties of valeriana roots. Shoyakugako Zasshi. 1980;34:19-24.

221. Takamura K, Kawaguchi M, Nabata H. The preparation and pharmacological screening of kessoglycol derivative. Yakugaku Zasshi. 1975;95:1198-1204.

222. Takamura K, Nabata H, Kawaguchi M. The pharmacological action on the kessoglycol-8–monoacetate. Yakugaku Zasshi. 1975;95:1205-1209.

223. Leuschner J, Muller J, Rudmann M. Characterisation of the central nervous depressant activity of a commercially available valerian root extract. Arzneimittelforschung. 1993;43:638-641.

224. Hiller K-O, Zetler G. Neuropharmacological studies on ethanol extracts of Valeriana officinalis L. behavioural and anticonvulsant properties. Phytother Res. 1996;10:145-151.

225. Oshima Y, Matsuoka S, Ohizumi Y. Antidepressant principles of Valeriana fauriei roots. Chem Pharm Bull. 1995;43:169-170.

226. Kriegelstein J, Grusla D. Central depressant constituent in valerian [in German]. Dtsch Apoth Zeitung. 1988;40:2041-2046.

227. Grusla D, Holzl J, Kriegelstein J. Baldrian-wirkungen im Gehirn der Ratte. Dtsch Apoth Zeitung. 1986;126:2249-2253.

228. Houghton PJ. The biological activity of valerian and related plants. J Ethnopharmacol. 1988;22:121-142.

229. Kohnen R, Oswald WD. The effects of valerian, propanolol, and their combination on activation, performance, and mood of healthy volunteers under social stress conditions. Pharmacopsychiatry. 1988;21:447-448.

230. Leathwood PD, Chauffard F, Heck E, Munoz-Box R. Aqueous extract of valerian root (Valeriana officinalis L.) improves sleep quality in man. Pharmacol Biochem Behav. 1982; 17:65-71.

231. Leathwood PD, Chauffard F. Quantifying the effects of mild sedatives. J Psychiatr Res. 1982;17:115-122.

232. Leathwood PD, Chauffard F, Munoz-Box R. Effect of Valeriana officinalis L. on subjective and objective sleep parameters. In: Sixth European Congress on Sleep Research. Zurich, Switzerland: Basel-Karger; 1983:402-405.

233. Braun R, Dittmar W, Hubner GE, Maurer HR. Influence of valtrate/isovaltrate on the hematopoiesis and metabolic liver activity in mice in vivo. Plant Med. 1984;1:1-4.

234. Balderer G, Borbely AA. Effect of valerian on human sleep. Psychopharmacology. 1985; 87:406-409.

235. Schulz H, Stolz C, Muller J. The effect of valerian extract on sleep polygraphy in poor sleepers: a pilot study. Pharmacopsychiatry. 1994; 27:147-151.

236. McGregor FB, Abernethy VE, Dahabra S, Cobden I, Hayes PC. Hepatotoxicity of herbal remedies. BMJ. 1989;299:1156-1157.

237. Shepherd C. Sleep disorders: liver damage warning with insomnia remedy. BMJ. 1993;306:1472.

238. Bos R, Woerdenbag HJ, De Smet PAGM, Scheffer JJC. Valeriana species. In: De Smet PAGM, Keller K, Hansel R, Chandler RF, eds. Adverse Effects of Herbal Drugs. Berlin, Germany: Springer-Verlag; 1997:165-180.

239. Keochanthala-Bounthanh C, Haag-Berrurier M, Beck JP, Anton R. Effects of thiol compounds versus cytotoxicity of valpotriates on cultured hepatoma cells. Plant Med. 1990;56:190-192.

240. Keochanthala-Bounthanh C, Beck JP, Haag-Berrurier M, Anton R. Effects of two monoterpene esters, valtrate and didrovaltrate, isolated from Valeriana wallichii, on the ultrastructure of hepatoma cells in culture. Phytother Res. 1993;7:124-127.

241. Tufik S, Fujita K, Seabra MDL, Lobo LL. Effects of a prolonged administration of valepotriates in rats on the mothers and their offspring. J Ethnopharmacol. 1994;41:39-44.

242. Berglund F, Flodh H, Lundborg P, Prame B, Sannerstedt R. Drug use during pregnancy and lactation: a classification for drug information. Acta Obstet Gynecol Scand Suppl. 1984;126:1-55.

243. Committee ADE. Medicines in Pregnancy: An Australian Categorization of Risk. Canberra, Australia: Australian Government Publishing Service; 1989.

244. Shrivastava SC, Sisodia CS. Analgetic studies on Vitax negundo and Valeriana wallichii. Ind Vet J. 1970;47:170-175.

245. Takamura K, Kakimoto M, Kawaguchi M, Iwasaki T. Pharmacological studies on the constituents of crude drugs and plants. J Pharm Soc Jpn. 1973;93:599-606.

246. Govindarajan VS. Capsicum--production, technology, chemistry, and quality, I: history, botany, cultivation and primary processing. CRC Crit Rev Food Sci Nutr. 1985;22:109-176.

247. Cordell GA, Araujo OE. Capsaicin: identification, nomenclature, and pharmacotherapy. Ann Pharmacother. 1993;27:330-336.

248. Jentzsch K, Pock H, Kubelka W. Isolation of dihydrocapsaicin and homodihydrocapsaicin form Capsicum fruits. Monatsch Chem. 1968;99:661-663.

249. Locock RA. Capsicum. Can Pharm J. 1985; 118:517–519.

250. Skofitsch G, Donnerer J, Lembeck F. Comparison of nonivamide and capsaicin with regard to their pharmacokinetics and effects on sensory neurons. Arzneimittelforschung. 1984;34:154-156.

251. Szolesanyi J. Capsaicin, irritation, and desensitization. In: Green BG, Mason JR, Kare MR, eds. Chemical Senses. New York, NY: Marcel Dekker; 1990:14-76.

252. Lynn B. Capsaicin: actions on nociceptive C-fibres and therapeutic potential. Pain. 1990; 41:61-69.

253. Anonymous. Treatment of painful diabetic neuropathy with topical capsaicin: a multicentre, double-blind, vehicle-controlled study. Arch Intern Med. 1991;151:2225-2229.

254. Tandan R, Lewis GA, Krusinski PB, Badger GB, Fries TJ. Topical capsaicin in painful diabetic neuropathy: controlled study with long term follow-up. Diabetes Care. 1992;15:8-14.

255. Dailey GE. Effect of treatment with capsaicin on daily activities of patients with diabetic neuropathy. Diabetes Care. 1992;15:159-165.

256. Scheffler NM, Sheitel PL, Lipton MN. Treatment of painful diabetic neuropathy with capsaicin 0.075%. J Am Podiatr Med Assoc. 1991;81:288-293.

257. Basha KM, Whitehouse FW. Capsaicin: a therapeutic option for painful diabetic neuropathy. Henry Ford Hosp Med J. 1991; 39:138-140.

258. Pfeifer MA, Ross DR, Schrage JP, Gelber DA, Schumer MP, Crain GM, Markwell SJ, Jung S. A highly successful and novel model for treatment of chronic painful diabetic peripheral neuropathy. Diabetes Care. 1993;16:1103-1115.

259. Watson CP, Evans RJ, Watt VR. Post-herpetic neuralgia and topical capsaicin. Pain. 1988;35:289-297.

260. Watson CPN, Evans RJ, Watt VR, Birkett N. Post-herpetic neuralgia: 208 cases. Pain. 1988;35:289-297.

261. Bernstein JE, Korman NJ, Bickers DR, Dahl MV, Millikan LE. Topical capsaicin treatment of chronic postherpetic neuralgia. J Am Acad Dermatol. 1989;21:265-270.

262. Bernstein JE, Bickers DR, Dahl MV, Roshal JY. Treatment of chronic postherpetic neuralgia with topical capsaicin: a preliminary study. J Am Acad Dermatol. 1987;17:93-96.

263. Peikert A, Hentrich M, Ochs G. Topical 0.025% capsaicin in chronic post-herpetic neuralgia: efficacy, predictors of response and long-term course. J Neurol. 1991;238:452-456.

264. Bjerring P, Arendt-Nielsen L, Soderberg U. Argon laser induced cutaneous sensory and pain thresholds in post-herpetic neuralgia: quantitative modulation by topical capsaicin. Acta Derm Venereol. 1990;70:121-125.

265. Watanabe T, Kawada T, Yamamoto M, Iwgi K. Capsaicin, a pungent principle of hot red pepper, evokes catecholamine secretion from the adrenal medulla of anesthetized rats. Biochem Biophys Res Commun. 1987;142:259-264.

266. Du Mee C. Vitex Agnus castus. Aust J Med Herbalism. 1993;5:63-65.

267. Houghton P. Agnus castus. Pharm J. 1994; 253:720-721.

268. Bruckner C. In mitteleuropa genutze heilpflanzen mit milchsekretionsfordernder wirkung (galactagoga). Gleditschia. 1989;17:189-210.

269. Milewicz A, Gejdel E, Sworen H, Sienkiewicz K, Jedrzejak J, Teucher T, Schmitz H. Vitex Agnus-castus extract in the treatment of luteal phase defects due to latent hyperprolactinemia: results of a randomised placebo-controlled double-blind study. Arzneimittelforschung Drug Res. 1993;43:752-756.

270. Bohnert K-J. The use of Vitex Agnus castus for hyperprolactinemia. Q Rev Nat Med. Spring 1997:19-21.

271. Jarry H, Leonhardt S, Gorkow C, Wuttke W. In vitro prolactin but not LSH and FSH release is inhibited by compounds in extracts of Agnus castus: direct evidence for a dopaminergic principle by the dopamine receptor assay. Exp Clin Endocrinol. 1994;102:448-454.

272. Sliutz G, Speiser P, Schultz A, Spona J, Zellinger R. Agnus castus extracts inhibit prolactin secretion of rat pituitary cells. Horm Metab. 1993;25:253-255.

273. Jarry H, Leonhardt S, Wuttke W. Agnus castus as dopaminergous effective principle in mastoynon N. Z Phytother. 1991;12:77-82.

274. Farnsworth NR, Kinghorn AD, Soejarto D, Waller DP. Siberian ginseng (Eleuthrococcus senticosus): current status as an adaptogen. In: Wagner H, Hikino H, Farnsworth NR, eds. Economic and Medicinal Plant Research. Orlando, Fla: Academic Press; 1985:155-215.

275. Leung AY. Encyclopedia of Common Natural Ingredients Used in Food, Drugs and Cosmetics. New York, NY: John Wiley & Sons Inc; 1980.

276. Baldwin CA, Anderson LA, Phillipson JD. What pharmacists should know about ginseng. Pharm J. November 8, 1986:583-586.

277. Medon PJ, Ferguson PW, Watson CF. Effects of Eleutherococcus senticosus extracts on hexobarbital metabolism in vivo and in vitro. J Ethnopharmacol. 1984;10:235-241.

278. Chang H-M, But PP-H. Pharmacology and Applications of Chinese Materia Medica. Singapore, Republic of China: World Scientific Press; 1986.

279. Teegarden R. Chinese Tonic Herbs. New York, NY: Japan Publishing Inc; 1985:197.

280. Bensky D, Gamble A. Chinese Herbal Medicine Materia Medica. Seattle, Wash: Eastland Press; 1986.

281. Avakia EV, Evonuk E. Effects of panax ginseng extract on tissue glycogen and adrenal cholesterol depeletion during prolonged exercise. Planta Med. 1979;36:43-48.

282. Fulder SJ. Ginseng and the hypothalamic-pituitary control of stress. Am J Chin Med. 1981;9:112-118.

283. Wagner H, Norr H, Winterhoff H. Plant adaptogens. Phytomedicine. 1994;1:63-76.

284. Hiai S, Yokoyama H, Oura H. Features of ginseng saponin-induced corticosterone secretion. Endocrinol Jpn. 1979;26:737-740.

285. Scaglione F, Cattaneo G, Alessandria M, Cogo R. Efficacy and safety of the standardised ginseng extract G115 for potentiating vaccination against the influenza syndrome and protection against the common cold [published correction appears in Drugs Exp Clin Res. 1996;22:338]. Drugs Exp Clin Res. 1996;22:65-72.

286. Sonnenborn U, Hansel R, De Smit PAGM. Adverse Effects of Herbal Drugs. Berlin, Germany: Springer Verlag; 1992.

287. Siegel RK. Ginseng abuse syndrome. JAMA. 1979;241:1644-1645.

288. Shader RI, Greenblat DJ. Phenelzine and the dream machine: ramblings and reflections. J Clin Psychopharmacol. 1985;5:65.

289. Shader RI, Greenblatt DJ. Bees, ginseng and MAOIs revisited. J Clin Psychopharmacol. 1988;8:235.

290. Jones BD, Runikis AM. Interaction of ginseng with phenelzine. J Psychopharmacol. 1987;7:201-202.

291. Mitra SK, Chakraborti A, Bhattacharya SK. Neuropharmacological studies on Panax ginseng. Indian J Exp Biol. 1996;34:41-47.

292. De Smet PAGM, Keller K, Hansel R, Chandler R. Adverse Effects of Herbal Drugs. Berlin, Germany: Springer, Verlag; 1997:250.

293. Lambert GA, Lang WJ, Friedman E, Meller E, Gershon S. Pharmacological and biochemical properties of isomeric yohimbine alkaloids. Eur J Pharmacol. 1978;49:39-48.

294. Morales A, Condra M, Owen JA, Surridge DH, Fenemore J, Harris C. Is yohimbine effective in the treatment of organic impotence? results of a controlled trial. J Urol.1987;137:1168-1172.

295. Susset JG, Tessier CD, Wincze J, Bansal S, Malhotra C, Schwacha MG. Effect of yohimbine hydrochloride on erectile impotence: a double-blind study. J Urol. 1989;141:1360-1363.

296. Holmeberg G, Gershon S. Autonomic and psychic effects of yohimbine hydrochloride. Psychopharmacologia. 1961;2:93-106.

297. Charney DS, Woods SW, Goodman WK, Heninger GR. Neurobiological mechanisms of panic anxiety: biochemical and behavioural correlates of yohimbine-induced panic attacks. Am J Psychiatry. 1987;144:1030-1036.

298. Charney DS, Woods SW, Krystal JH, Nagy LM, Heninger GR. Noradrenergic neuronal dysregulation in panic disorder: the effects of intravenous yohimbine and clonidine in panic disorder patients. Acta Psychiatr Scand. 1992;86:273-282.

299. Rasmusson AM, Southwick SM, Hauger RL, Charney DS. Plasma neuropeptide Y (NPY) increases in humans in response to the alpha 2 antagonist yohimbine. Neuropsychopharmacology. 1998;19:95-98.

300. Price LH, Charney DS, Heninger GR. Three cases of manic symptoms following yohimbine administration. Am J Psychiatry. 1984;141:1267-1268.

301. Dunn RW, Corbett R. Yohimbine-induced seizures involve NMDA and GABAergic transmission. Neuropharmacology. 1992; 31:389-395.

302. Charney D, Heninger G, Breier A. Noradrenergic function in panic anxiety: effects of yohimbine in healthy subjects and patients with agoraphobia and panic disorder. Arch Gen Psychiatry. 1984;41:751-763.

303. De Smet P, Smeets O. Potential risks of health food products containing yohimbe extracts. BMJ. 1994;309:958.

Автоматический перевод на русский язык


Читать другие статьи на эту тему