The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Complementary and Integrative MedicineFull Access

Popular Herbal and Natural Remedies Used in Psychiatry

Abstract

Natural and herbal remedies, also known as “alternative” or “complementary” medicines, have grown tremendously in popularity over the past two decades, becoming a major component of health care and general wellness in the United States and worldwide. The ready availability of these remedies over the counter and their generally good tolerability and safety contribute to this popularity, and many people have benefited from them, often in cases when conventional treatments have failed or caused intolerable side effects. Despite many Food and Drug Administration (FDA)–approved psychotropic medications on the market, efficacy has been inconsistent for some, and many treatment responders will eventually relapse. Continued research on the efficacy and safety of these alternative therapies is, therefore, important. This article reviews six of the most commonly used natural remedies for psychiatric conditions, including the antidepressants St. John’s wort, omega-3 fatty acids, and S-adenosyl methionine (SAMe); the sedative-hypnotics valerian and melatonin; and the nootropic ginkgo biloba. We outline the general indications for use, suggested doses, possible mechanisms, and adverse effects to give clinicians a good summary of the benefits and liabilities of each. Although there is growing evidence of efficacy and safety to support the use of these remedies, clinicians must be aware of the limitations of the evidence base and take that into account with all the other factors that contribute to clinical decision making.

Natural and herbal remedies, also known as “complementary” or “alternative” medicines (CAMs), have grown tremendously in popularity over the past two decades, becoming a major component of health care and general wellness in the United States and worldwide. Many people certainly benefit from them, often in cases when conventional treatments have failed or caused side effects. A 2007 National Health Interview Survey found that 38% of adults and 12% of children had used CAM practices and products in the past year, representing about $33.9 billion in out-of-pocket costs (1). Although there is growing evidence of efficacy and safety to support the use of these remedies, it is important for clinicians to be aware of the limitations of the evidence base and to take that into account with all the other factors that contribute to clinical decision making (2). In psychiatry, we have about 40 FDA-approved antidepressants on the market, yet their efficacy has been inconsistent (3), and many treatment responders will eventually relapse (4). Continued research on natural therapies is called for, partly because they are readily available over the counter and widely used, and also because of their generally good tolerability and safety.

This article reviews six of the most commonly used natural remedies for psychiatric conditions, such as mood disorders, insomnia, and dementia. Here, I cover the general indications for use, suggested doses, possible mechanisms, and adverse effects to give clinicians a good summary of the benefits and liabilities of each. A summary of key points of information for clinicians is presented in Table 1.

TABLE 1. Natural and Herbal Remedies: Indications, Doses, and Adverse Effectsa

RemedyIndicationsDosingAdverse Effects/Interactions
Ginkgo bilobaDementia (Alzheimer’s and vascular); antidepressant-induced sexual dysfunction120–240 mg/day, BID–TIDMild GI upset, headache, irritability, dizziness, possible bleeding in individuals with bleeding disorders or who take anticoagulants
MelatoninInsomnia (particularly if secondary to circadian disturbance); safe in children and elderly individuals0.1–10.0 mg/daySedation, confusion, inhibition of fertility, decreased sex drive, hypothermia, retinal damage, immunosuppression
Omega-3 fatty acids (EPA and DHA)Depression (including perinatal depression); bipolar disorder; psychotic disorders; borderline personality disorder; attention-deficit disorders; safe in children and elderly individuals1,000–2,000 mg/day (some studies administered up to 10 g/day)GI upset; cycling to mania in patients with bipolar disorder; no adverse interactions
S-adenosyl methionine (SAMe)Depression (including with comorbid medical conditions such as Parkinson’s disease, osteoarthritis and fibromyalgia, sexual dysfunction, neurocognitive disorders, psychotic disorders, and liver disease)200–3,200 mg/day (sometimes higher)Mania in bipolar patients; GI upset, insomnia, anorexia, dry mouth, sweating, dizziness, and nervousness; no adverse interactions
St. John’s wort (Hypericum perforatum L.)Depression, menopausal symptoms300–1,800 mg/day, BID–TIDDry mouth, dizziness, constipation, phototoxicity, serotonin syndrome when combined with SSRIs; adverse interactions with warfarin, cyclosporin, oral contraceptives, theophylline, fenprocoumon, digoxin, camptosar, indinavir, zolpidem, irinotecam, and olanzapine; mania in patients with bipolar disorder
Valerian (Valeriana officinalis)Insomnia; possibly OCD; safe in children, elderly individuals, and menopausal women450–600 mg before bedtimeBlurry vision, dystonias, hepatotoxicity, headaches; some concern about malformations if used during pregnancy

aBID, twice daily; TID, three times daily; GI, gastrointestinal; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; SSRI, selective serotonin reuptake inhibitor; OCD, obsessive-compulsive disorder.

TABLE 1. Natural and Herbal Remedies: Indications, Doses, and Adverse Effectsa

Enlarge table

St. John’s Wort

The herbal remedy St. John’s wort (Hypericum perforatum L. [SJW]) has been used for centuries to treat depression (5). There are about 40 published clinical trials, including many comparisons with tricyclic antidepressants (TCAs) and selective serotonin reuptake inhibitors (SSRIs), and various systematic reviews and meta-analyses.

Early clinical studies in depressed samples, conducted mostly in Europe, supported SJW as more effective than placebo and comparable to TCAs, particularly for milder forms of depression (6). Some reports have supported efficacy for seasonal affective disorder (7, 8) and for menopausal symptoms (9). Many early studies, however, were limited by short duration and a lack of standardized diagnostic practices or instruments. More recent studies have been more rigorous, including comparisons between SJW and SSRIs. In some studies, SJW performed comparably to fluoxetine (10) and sertraline (11). Other trials comparing SJW to sertraline, fluoxetine, and placebo have suggested no advantage for either medication in samples with moderately severe major depressive disorder (MDD) (1215). However, upon closer scrutiny, the data from these studies suggest that remission rates may, in fact, be higher for SJW and that SJW may be more effective for individuals with less severe depression.

Various systematic reviews and meta-analyses of SJW have been published over the past decade. A Cochrane review by Linde et al. (6) examined 29 trials including 5,489 patients; 18 trials compared SJW with placebo, and 17 compared it with standard antidepressants. Results showed heterogeneity, but overall, SJW was found to be superior in efficacy to placebo and equivalent to standard antidepressants, with better tolerability. A meta-analysis by Ng et al. (16) examined 27 clinical trials including 3,808 patients, comparing SJW with SSRIs for treating mild-to-moderate depression. SJW and SSRIs had comparable response and remission rates, and SJW had a significantly lower discontinuation rate than SSRIs. The pooled standard mean difference from baseline scores on the Hamilton Depression Rating Scale (HAM-D) also supports SJW as efficacious. A systematic review by Apaydin and colleagues (17) examined 35 studies comprising 6,993 patients with mild-to-moderate depression. SJW was associated with more treatment responders than placebo and comparable response rates against standard antidepressants, with a milder side-effect profile than the latter. However, the authors cautioned about heterogeneity in the findings, limited data on severe depression, and poor reporting of adverse effects, particularly rare ones. SJW may also be effective in treating menopausal symptoms, as supported by a meta-analysis by Liu et al. (18).

SJW contains more than 150 chemicals, some of which have been proposed as the main psychotropic ingredients (19). Hypericin and hyperforin are the best understood of these (5, 20), and SJW preparations are typically standardized to one of these chemicals. SJW’s mechanism of action is probably multifactorial (5), involving an interaction with the hypothalamus-pituitary-adrenal (HPA) axis that results in decreased cortisol production (21). Other proposed mechanisms include decreased serotonin receptor density, decreased synaptic neurotransmitter reuptake, and direct serotonergic activity (21). SJW also has very slight monoamine oxidase inhibitor (MAOI) activity. Taking SJW does not require the patient to follow an MAOI diet, but combinations of SJW and SSRIs have resulted in serotonin syndrome (21). It is, therefore, not advisable to combine SJW with SSRIs.

Typical doses of SJW range from 300 to 1,800 mg/day, usually divided on a three-times-daily basis, with 900 mg/day considered as a standard therapeutic dose. Different manufacturing methods may produce variability in efficacy (21). The most common side effects are dry mouth, dizziness, and constipation (21). Less common side effects include phototoxicity (hypersensitivity to sunlight), cycling to mania in patients with bipolar disorder, and drug–drug interactions via the liver enzyme CYP-450–3A4 (21). These interactions may result in decreased activity of warfarin, cyclosporin, oral contraceptives, theophylline, fenprocoumon, digoxin, indinavir, camptosar, verapamil, benzodiazepines, zolpidem, irinotecam, and olanzapine (2227), and possibly others. Therefore, extreme caution is required for patients with HIV who are taking protease inhibitors, as well as for patients with cancer who are receiving chemotherapy and transplant patients taking immunosuppressive drugs (21).

The evidence thus far supports SJW for treatment of depressive disorders, although it may be less effective in chronic and/or severe depression. The use of SJW also appears to be cost effective, compared with the use of standard antidepressants (28). More large-scale, controlled trials are needed to better characterize SJW’s place in the psychopharmacologist’s armamentarium.

Omega-3 Fatty Acids

Omega-3 fatty acids are a family of polyunsaturated lipids, including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), derived primarily from fish and fish oil preparations, and alpha linolenic acid (ALA), which is derived from vegetable sources such as flaxseed oil (29). Their role in the treatment of mood disorders has been extensively studied over the past two decades, with generally positive, although mixed, results (2935). Fewer clinical trials have examined omega-3s in other psychiatric conditions, including psychotic disorders and developmental disorders (32).

Most psychiatric investigations into omega-3 fatty acids have typically used EPA or a combination of EPA and DHA, mostly as an adjunct therapy for unipolar depression (36) but also as monotherapy. Evidence for DHA therapy alone is more limited (3739), despite its important role in the development of the human brain (40). The optimal dose of omega-3 fatty acids remains unclear. Early studies have used doses as high as 9–10 g/day, but more recent studies suggest benefit with as little as 1–2 g/day (2935). The differential efficacy of EPA and DHA remains unclear, but some recent comparative studies have favored EPA as the stronger antidepressant (4143).

The data overall are difficult to interpret because of heterogeneity among studies, particularly regarding omega-3 preparations, doses, and study design. Several meta-analyses examined their efficacy for unipolar depression, with mixed results (2931, 33, 34). Doses frequently range from 1 to 2 g/day. Use of preparations with ≥60% EPA in combinations (29) has been recommended. Recent work by Mischoulon and colleagues suggests that omega-3s may be more effective in individuals who are overweight and/or have high levels of inflammatory markers (42, 43).

Other potential applications include treatment of perinatal depression (35) and bipolar disorder (44, 45), although preferably for the condition’s depressive phase rather than mania (46). Omega-3s have also been studied for treatment of schizophrenia and other psychotic disorders, with less encouraging results (47, 48), although there is some evidence of preventive effects (4951). Some benefits have been reported in borderline personality disorder (32, 52). Recent reports support beneficial mood effects in children and adolescents (53), as well as some benefit in attention deficit disorders (54, 55). Evidence of efficacy in dementia is not as convincing (56, 57). They are likely safe for pregnant women, as eating fish is considered safe during pregnancy (58), and studies of supplements have suggested benefits for the fetus (59), but caution should still be exercised in this population. These reports collectively suggest a broad range of applications for omega-3s.

The mechanism of action of omega-3s with regard to psychiatric symptoms is unclear but probably multifactorial. Proposed mechanisms include inhibiting G-protein signal transduction, membrane stabilization, and anti-inflammatory effects, among others (60, 61).

Omega-3 preparations typically contain 500–1,000 mg of omega-3, usually constituting a mixture of DHA, EPA, and other essential fatty acids such as ALA. Preparations of purified individual omega-3 fatty acids are also available. As with most nutraceuticals, there are no published head-to-head studies comparing different brands or preparations of omega-3s, which limits the ability to make specific recommendations.

Omega-3 fatty acids are a promising treatment, given their tolerability (gastrointestinal [GI] distress is the main complaint reported in most studies) and potential benefits in various psychiatric conditions. There have been anecdotal cases of cycling to mania in bipolar patients who were depressed (40); individuals with bipolar disorder should, therefore, take omega-3s with concurrent mood stabilizers. Previous concerns about elevated risk of bleeding in postsurgical patients have been mostly disproven (62), despite the fact that caution is often recommended in patients taking anticoagulants (63). More placebo-controlled studies of effectiveness with large samples, comparisons of safety and effectiveness between the different types of omega-3 fatty acids, and further investigations into the various possible indications and mechanisms of action of the omega-3s are needed.

S-Adenosyl Methionine (SAMe)

SAMe is an intermediate in the metabolism of folate and B12 and donates methyl groups used in the synthesis of brain neurotransmitters such as serotonin, acetylcholine, and dopamine (64), with potential indirect effects on norepinephrine synthesis (65). The literature thus far generally supports parenteral (intravenous [i.v.] or intramuscular [i.m.]) and oral SAMe preparations as more effective than placebo and comparable with tricyclic antidepressants at doses up to 3,200 mg/day (6570). There are more than 50 clinical trials of SAMe for depressive disorders, including 17 open ones, 19 randomized controlled trials, and 21 comparisons with other antidepressants. Doses of SAMe have ranged from 200 to 3,200 mg/day (70). Most comparisons have been with TCAs, and, surprisingly, only one clinical trial has compared SAMe with SSRIs. A study by Mischoulon et al. (71) with 189 MDD subjects found no advantage for SAMe or escitalopram over placebo but found that male subjects seemed to improve more robustly than female subjects in this sample (72).

There has been only one major meta-analysis of comparisons of SAMe with placebo published in the past 15 years (68). This analysis covered 28 of 47 studies. SAMe’s effect was found to be superior to that of placebo for efficacy measures; however, significance was attained only for effect size analysis. The pooled estimate of effect reflected a 5- to 6-point drop on the HAM-D, which represents solid improvement but certainly not full resolution. Comparisons with other antidepressants suggested no significant differences in risk ratios and effect sizes, which supports equivalence between SAMe and other agents, but most of these studies were limited by the lack of a placebo arm. Concerns were raised about publication bias, small samples, and heterogeneity of doses and delivery systems; for these reasons, the authors were cautious in their interpretation of the results. The encouraging findings, overall, support further study, including comparisons of cost-effectiveness, tolerability, side-effect profiles, and patient preference. A recent review by Sharma and colleagues (70) noted that relatively few new studies of SAMe have emerged since 2002, suggesting that the meta-analysis by Hardy et al (68) remains highly relevant.

SAMe may be combined with conventional antidepressants, in some cases, accelerating onset of action (73, 74). Alpert et al. (75) found a benefit of SAMe augmentation in 30 SSRI nonresponders after 6 weeks of open-label SAMe at 800–1,600 mg/day. Papakostas et al. (76) followed up this work with a study of 73 SSRI/SNRI (serotonin norepinephrine reuptake inhibitor) nonresponders who were randomized for 6 weeks to SAMe at 800 mg BID (twice a day) or placebo BID; results showed a significant advantage for SAMe. Apart from these studies, there is a surprising lack of augmentation trials combining SAMe with newer agents.

SAMe may be a particularly good choice for people with the methylene tetrahydrofolate reductase (MTHFR) deficiency, as it would theoretically bypass the steps in folate metabolism that are dependent on this enzyme (65). SAMe may have a faster onset of action than conventional antidepressants, with some patients improving within 2 weeks (67).

Other potential applications of SAMe include those for depression with comorbid medical conditions such as Parkinson’s disease, osteoarthritis and fibromyalgia, sexual dysfunction, neurocognitive disorders, psychotic disorders, and liver disease (70), but evidence is less robust in these cases. It also appears to be safe in pregnancy, but evidence is too preliminary to make definitive recommendations (70).

SAMe is well tolerated and very safe (65). Its primary reported side effect is GI upset, but other adverse effects may include insomnia, anorexia, dry mouth, sweating, dizziness, and nervousness (65). Individuals with bipolar depression have reported increased anxiety, mania, or hypomania (66, 70, 77). SAMe should, therefore, be used with a concurrent mood stabilizer in this population.

While reported doses of SAMe range from 400 to 3,200 mg/day (70, 71), some patients with depression who were observed in clinical practice may require even higher doses, which appear safe to administer if the patient tolerates the higher doses well. So far, no serious drug–drug interactions have been reported. SAMe appears very promising, and further research will clarify its role in the psychiatrist’s armamentarium.

Valerian

Valerian (Valeriana officinalis) is a popular herbal sedative and mild hypnotic that has been used worldwide for over 1,000 years (78). Its soporific effect is likely due to its active ingredients, including valepotriates and sesquiterpenes, which may function similarly to benzodiazepines or barbiturates (78), with GABA-ergic activity (78).

There are more than 40 published controlled trials of valerian that include healthy participants and symptomatic individuals (7881). Many studies suggest efficacy comparable with that of benzodiazepines, with fewer side effects and no apparent tolerance (78). There is also evidence that valerian is beneficial in children (82) and the elderly (8385), as well as in menopausal women (86). However, a few meta-analyses and systematic reviews are not very supportive of efficacy (7981). One small study suggests its benefits in treating obsessive–compulsive disorder (OCD) (87). It may be effective for treating insomnia in postmenopausal women (86, 88).

One possible reason for the inconsistent findings in clinical trials may be the powerful smell of valerian that can result in unmasking (78), but this can be overcome by adding some of valerian’s inactive ingredients into placebos, a more recent practice. Recommended doses of valerian are 450–600 mg before bedtime (78), and there is no apparent increased benefit from higher doses. Valerian appears to promote natural sleep after a few weeks of regular use, rather than treating insomnia acutely (78).

Valerian is well tolerated. Headaches and GI complaints are the ones most often reported (78). There is no evidence of a hangover effect in the morning after use. It appears safe in overdose and has shown no significant adverse interactions (89). Some rare toxic reactions have been reported, including blurry vision, dystonias, hepatotoxicity (9092), withdrawal, and delirium (93). There is no evidence of dependence or daytime drowsiness, and it appears safe for children (94) and elderly individuals (95). Retrospective studies suggest safety in pregnancy, but valerian use in pregnant women appears uncommon. There is some concern about rare outcomes such as malformations (96); for this reason, its use should probably be avoided during pregnancy. In summary, valerian appears to be a promising hypnotic that decreases sleep latency and improves sleep quality, with potential niches in various patient populations.

Melatonin

Melatonin is a hormone derived from serotonin in the pineal gland that helps to regulate circadian rhythms (97). It is often used by people who travel across time zones to reset their biological clock and prevent or alleviate jet lag. Melatonin appears to be an effective hypnotic that works fairly quickly upon administration. It may be more effective for generally healthy people with insomnia due to circadian disturbances (98, 99) as opposed to those with diagnosed psychiatric disorders. Melatonin may function by interacting with the suprachiasmatic nucleus to reset the circadian pacemaker and attenuate the alerting process (100), and it may have a direct sedative effect (101).

There are about 20 clinical studies of melatonin for insomnia, including some with children and elderly individuals. Studies in children with sleep disorders (102) and neurodevelopmental disorders (103) have produced encouraging results.

A recent meta-analysis by Auld and colleagues (104) examined 12 double- or single-blind randomized and controlled studies. Results supported significant advantages for melatonin over placebo at reducing sleep onset latency in primary insomnia, delayed sleep-phase syndrome, and regulating the sleep–wake patterns in patients who are blind. A meta-analysis by Ferracioli-Oda et al. (105) of 19 trials found improvement in sleep onset latency, total sleep time, and overall sleep quality. McCleery et al. (106) found no benefits of melatonin for sleep disorders in patients with dementia. Likewise, minimal benefits were found for treating patients with Parkinson’s disease (107). Other meta-analyses and reviews also offer mixed results regarding the degree of efficacy of melatonin versus placebo (108).

Recommended doses range widely, from 0.5 mg/day to 10 mg/day (109), with commercial preparations reflecting this range. There is a long-acting version of melatonin dosed at 2 mg/day that has been shown to be effective, particularly in older individuals (110113).

Side effects from melatonin appear to be few and benign. High doses may cause daytime sedation or confusion (114). Serious adverse effects, although rare, may include decreased fertility and sex drive (115), hypothermia (116), and retinal damage (117, 118). Because of potential interactions with the HPA axis and thymus gland, melatonin may cause immunosuppression and should, therefore, be used with caution in individuals taking steroids or those who are HIV positive (119, 120).

In summary, melatonin is a promising hypnotic, generally accepted as safe and effective (121). Apart from the aforementioned concerns about immunosuppressants, there appear to be no adverse interactions with other drugs. It is likely safe to combine with most prescription psychotropics. Because of encouraging data in children with sleep disorders (102) and neurodevelopmental disorders (103), as well as in older patients (110113) these may represent particularly good niches for melatonin, since stronger sedative-hypnotics could be avoided in these vulnerable populations.

Ginkgo Biloba

Ginkgo biloba, the seed from the ginkgo tree, has been used in traditional Chinese medicine for thousands of years (122). Ginkgo’s primary indication is for improving cognitive function (including memory, abstract thinking, and psychomotor function) in individuals with organic brain diseases such as Alzheimer’s dementia.

Over 30 placebo-controlled double-blind trials in populations with dementia have been published (122). These trials suggest that dementia improves with ginkgo treatment. However, the standards for testing the efficacy of nootropic drugs have changed over the years. Early studies examined cognitive symptoms but not the patient’s functioning in daily activities and need for care (122). Many meta-analyses and systematic reviews have suggested its efficacy (123127).

Zhang and colleagues (125) recently reviewed ten systematic reviews that included applications of ginkgo in Alzheimer’s disease (AD), vascular dementia (VaD), mixed dementia, and mild cognitive impairment (MCI). Overall ginkgo demonstrated a dose-dependent improvement in cognition, neuropsychiatric symptoms, and daily activities, with optimal results at doses of 240 mg/day. Tolerability was comparable to that of placebo, and its application for AD did particularly well, with fewer incidences of vertigo, tinnitus, angina pectoris, and headache.

A similar review of 12 reviews by Yuan and colleagues (126) found varied quality of the studies but generally supported the efficacy of ginkgo compared with placebo regarding cognitive performance, activities of daily living, and clinical global impression, also when dosed at about 240 mg/day, with good tolerability and safety. Limitations of studies included insufficient evidence for periods of less than 22 weeks. The authors also pointed out that doses below 200 mg daily are less likely to be effective.

A meta-analysis by Hashiguchi et al. (127) examined nine of 13 studies lasting 12 to 52 weeks with doses >120 mg/day in 2,381 patients. Ginkgo outperformed placebo in seven of the nine studies. The advantage was maintained when examining by type of dementia, including AD, VaD, and combined AD and VaD. Doses of 240 mg/day were deemed the most effective, with good tolerability.

Gingko has been compared with synthetic nootropic drugs. Cholinesterase inhibitors are somewhat more effective but not as well tolerated, and may be combined with ginkgo (128132). Ginkgo’s strongest advantage may lie in its lower incidence of side effects (133); consequently, many physicians have favored ginkgo over the synthetic nootropics as a first-line treatment (134). Ginkgo appears to have no clear preventive effects on dementia (135).

Gingko contains many active components, such as flavonoids (quercetin, kaempferol, and isorhamnetin) and terpene lactones (ginkgolides, bilobalide, and ginkgolic acids) (122). It appears to have multiple mechanisms of action. It is thought to stimulate and protect functional nerve cells from hypoxia and ischemia and free radical damage (136), and it may stabilize neuronal membranes (122). Other functions may include preservation of muscarinic choline receptors and alpha-2 adrenergic receptors and promotion of choline uptake in the hippocampus (122).

The suggested dose of ginkgo is 120–240 mg/day, dosed two to three times a day. Assessment of its full effect may require at least one year (122, 123). Long-term benefits are unclear. Alzheimer’s dementia may respond better than VaD (123). Side effects may include mild GI upset, headache, irritability, dizziness, or allergic reactions. There are no established interactions with other drugs (123, 137), but caution is recommended with patients who have bleeding disorders or are taking anticoagulant medications such as coumadin, because ginkgo inhibits platelet-activating factor (PAF) (136, 137). Nonetheless, a recent meta-analysis of 18 trials (138) including healthy volunteers and patients with dementia, peripheral artery disease, or diabetes mellitus did not find any increased risk of bleeding, based on hemostatic outcomes.

Some studies have suggested that ginkgo may alleviate antidepressant-induced sexual dysfunction (139, 140). In an open trial of ginkgo in 63 patients with antidepressant-induced sexual dysfunction (from SSRIs, SNRIs, TCAs, and MAOIs) (140), 91% of women and 76% of men reported improvement in all aspects of the sexual cycle with doses of 60–180 mg BID. On the other hand, an open trial by Ashton and colleagues (141) found limited effect on antidepressant-induced sexual dysfunction in a small sample of 22 subjects. Wheatley (142) likewise found no significant advantage to ginkgo over placebo in a small study. A controlled study by Kang and colleagues (143) found benefit with ginkgo, but a high placebo response rate prevented a significant difference after 8 weeks. A study of women with sexual arousal disorder found that ginkgo may have an augmentative effect with sex therapy but not when used alone (144). The mechanism for improvement in sexual functioning may involve ginkgo’s interaction with PAF, prostaglandins, peripheral vasodilatation, and central serotonin and norepinephrine receptor activity.

In summary, ginkgo appears to alleviate dementia symptoms, or at least to slow down the course of the illness, with a benign side-effect profile. The full extent of its role as a nootropic and as a treatment for sexual dysfunction remains to be clarified. Apart from the risk of hemorrhage in people with bleeding disorders or who take anticoagulants, ginkgo appears to be safe to combine with other medications.

Recommendations for Clinicians

Natural remedies remain a very popular alternative or complement for individuals with psychiatric disorders. Therefore, it is important for practitioners to be reasonably well versed in their use, particularly with regard to safety. The best candidates for natural treatments include patients with mild illness, who would be unlikely to have a catastrophic outcome if the treatment did not work. Likewise, patients with refractory illness who have not benefited from approved treatments (or suffered from side effects) may have nothing to lose by trying a natural remedy, under supervision by a physician (145).

Clinicians should routinely ask their patients whether they are using any “alternative” remedies, since many individuals do not inform those who are treating them about their use of these agents (145). Then the practitioner should explain the risks and benefits of each natural therapy under consideration and compare these to the risks and benefits of available conventional treatments. This would help the patient make an informed decision. Likewise, while more is being learned about the risks and benefits of combinations with standard medications, clinicians should be aware of what other medications their patients are taking and be knowledgeable about potential adverse interactions.

Dr. Mischoulon is the director of the Depression Clinical and Research Program, Massachusetts General Hospital; and professor of psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston.
Send correspondence to Dr. Mischoulon (e-mail: ).

Dr. Mischoulon reports receiving research/grant funding from the National Center for Complementary and Integrative Health (NCCIH), the National Institute for Mental Health (NIMH), and research support from Nordic Naturals. He has provided unpaid consulting for Pharmavite, LLC, and Gnosis USA, Inc.

References

1 National Institutes of Health: Fact Sheet: Complementary and Alternative Medicine. 2010, October. https://report.nih.gov/NIHfactsheets/Pdfs/ComplementaryandAlternativeMedicine(NCCAM).pdf Accessed 8/21/2017Google Scholar

2 Gerbarg PL, Brown RP, Muskin PR: Complementary and integrative medicine DSM-5, in Clinical Decision Making in Complementary and Integrative Treatment in Psychiatric Practice. Edited by Gerbarg PL, Brown RP, Muskin PR. Washington, DC, American Psychiatric Association Publishing, 2017Google Scholar

3 Kirsch I, Deacon BJ, Huedo-Medina TB, et al.: Initial severity and antidepressant benefits: a meta-analysis of data submitted to the Food and Drug Administration. PLoS Med 2008; 5:e45CrossrefGoogle Scholar

4 Safer DJ: Differing antidepressant maintenance methodologies. Contemp Clin Trials 2017; 61:87–95CrossrefGoogle Scholar

5 Schulz V, Hansel R, Blumenthal M, et al.: St. John's wort as an antidepressant; in Rational Phytotherapy: A Reference Guide for Physicians and Pharmacists, 5th ed. Berlin, Springer, 2011, pp. 62–85Google Scholar

6 Linde K, Berner MM, Kriston L: St John’s wort for major depression. Cochrane Database of Systematic Reviews 2008; 4:CD000448Google Scholar

7 Kasper S: Treatment of seasonal affective disorder (SAD) with hypericum extract. Pharmacopsychiatry 1997; 30(Suppl 2):89–93CrossrefGoogle Scholar

8 Wheatley D: Hypericum in seasonal affective disorder (SAD). Curr Med Res Opin 1999; 15:33–37CrossrefGoogle Scholar

9 Grube B, Walper A, Wheatley D: St. John’s wort extract: efficacy for menopausal symptoms of psychological origin. Adv Ther 1999; 16:177–186Google Scholar

10 Schrader E: Equivalence of St John’s wort extract (Ze 117) and fluoxetine: a randomized, controlled study in mild-moderate depression. Int Clin Psychopharmacol 2000; 15:61–68CrossrefGoogle Scholar

11 Brenner R, Azbel V, Madhusoodanan S, et al.: Comparison of an extract of hypericum (LI 160) and sertraline in the treatment of depression: a double-blind, randomized pilot study. Clin Ther 2000; 22:411–419CrossrefGoogle Scholar

12 Shelton RC, Keller MB, Gelenberg A, et al.: Effectiveness of St John’s wort in major depression: a randomized controlled trial. JAMA 2001; 285:1978–1986CrossrefGoogle Scholar

13 Lecrubier Y, Clerc G, Didi R, et al.: Efficacy of St. John’s wort extract WS 5570 in major depression: a double-blind, placebo-controlled trial. Am J Psychiatry 2002; 159:1361–1366CrossrefGoogle Scholar

14 Hypericum Depression Trial Study Group: Effect of Hypericum perforatum (St John’s wort) in major depressive disorder: a randomized controlled trial. JAMA 2002; 287:1807–1814CrossrefGoogle Scholar

15 Fava M, Alpert J, Nierenberg AA, et al.: A double-blind, randomized trial of St John’s wort, fluoxetine, and placebo in major depressive disorder. J Clin Psychopharmacol 2005; 25:441–447CrossrefGoogle Scholar

16 Ng QX, Venkatanarayanan N, Ho CY: Clinical use of Hypericum perforatum (St John’s wort) in depression: A meta-analysis. J Affect Disord 2017; 210:211–221CrossrefGoogle Scholar

17 Apaydin EA, Maher AR, Shanman R, et al.: A systematic review of St. John’s wort for major depressive disorder. Syst Rev 2016; 5:148CrossrefGoogle Scholar

18 Liu YR, Jiang YL, Huang RQ, et al.: Hypericum perforatum L. preparations for menopause: a meta-analysis of efficacy and safety. Climacteric 2014; 17:325–335CrossrefGoogle Scholar

19 Seifritz E, Hatzinger M, Holsboer-Trachsler E: Efficacy of Hypericum extract WS(®) 5570 compared with paroxetine in patients with a moderate major depressive episode - a subgroup analysis. Int J Psychiatry Clin Pract 2016; 20:126–132CrossrefGoogle Scholar

20 Brockmöller J, Reum T, Bauer S, et al.: Hypericin and pseudohypericin: pharmacokinetics and effects on photosensitivity in humans. Pharmacopsychiatry 1997; 30(Suppl 2):94–101CrossrefGoogle Scholar

21 Nierenberg AA, Lund HG, Mischoulon D: St. John’s wort: a critical evaluation of the evidence of antidepressant effects; in Natural Medications for Psychiatric Disorders: Considering the Alternatives. Edited by Mischoulon D, Rosenbaum J. Philadelphia, Lippincott, Williams & Wilkins, 2008Google Scholar

22 Moore LB, Goodwin B, Jones SA, et al.: St. John’s wort induces hepatic drug metabolism through activation of the pregnane X receptor. Proc Natl Acad Sci USA 2000; 97:7500–7502CrossrefGoogle Scholar

23 Miller LG: Herbal medicinals: selected clinical considerations focusing on known or potential drug-herb interactions. Arch Intern Med 1998; 158:2200–2211CrossrefGoogle Scholar

24 Miller JL: Interaction between indinavir and St. John’s wort reported. Am J Health Syst Pharm 2000; 57:625–626CrossrefGoogle Scholar

25 Piscitelli SC, Burstein AH, Chaitt D, et al.: Indinavir concentrations and St John’s wort. Lancet 2000; 355:547–548CrossrefGoogle Scholar

26 Davis SA, Feldman SR, Taylor SL: Use of St. John’s wort in potentially dangerous combinations. J Altern Complement Med 2014; 20:578–579CrossrefGoogle Scholar

27 Russo E, Scicchitano F, Whalley BJ, et al.: Hypericum perforatum: pharmacokinetic, mechanism of action, tolerability, and clinical drug-drug interactions. Phytother Res 2014; 28:643–655CrossrefGoogle Scholar

28 Solomon D, Adams J, Graves N: Economic evaluation of St. John’s wort (Hypericum perforatum) for the treatment of mild to moderate depression. J Affect Disord 2013; 148:228–234CrossrefGoogle Scholar

29 Sublette ME, Ellis SP, Geant AL, et al.: Meta-analysis of the effects of eicosapentaenoic acid (EPA) in clinical trials in depression. J Clin Psychiatry 2011; 72:1577–1584CrossrefGoogle Scholar

30 Grosso G, Pajak A, Marventano S, et al.: Role of omega-3 fatty acids in the treatment of depressive disorders: a comprehensive meta-analysis of randomized clinical trials. PLoS One 2014; 9:e96905CrossrefGoogle Scholar

31 Appleton KM, Sallis HM, Perry R, et al.: Omega-3 fatty acids for depression in adults. Cochrane Database Syst Rev 2015; (11):CD004692Google Scholar

32 Bozzatello P, Brignolo E, De Grandi E, et al.: Supplementation with omega-3 fatty acids in psychiatric disorders: a review of literature data. J Clin Med 2016; 5:E67CrossrefGoogle Scholar

33 Appleton KM, Sallis HM, Perry R, et al.: ω-3 Fatty acids for major depressive disorder in adults: an abridged Cochrane review. BMJ Open 2016; 6:e010172CrossrefGoogle Scholar

34 Mocking RJ, Harmsen I, Assies J, et al.: Meta-analysis and meta-regression of omega-3 polyunsaturated fatty acid supplementation for major depressive disorder. Transl Psychiatry 2016; 6:e756CrossrefGoogle Scholar

35 Lin PY, Chang CH, Chong MF, et al.: Polyunsaturated fatty acids in perinatal depression: a systematic review and meta-analysis. Biol Psychiatry 2017; 82: 560–569CrossrefGoogle Scholar

36 Sarris J, Murphy J, Mischoulon D, et al.: Adjunctive nutraceuticals for depression: a systematic review and meta-analyses. Am J Psychiatry 2016; 173:575–587CrossrefGoogle Scholar

37 Marangell LB, Martinez JM, Zboyan HA, et al.: A double-blind, placebo-controlled study of the omega-3 fatty acid docosahexaenoic acid in the treatment of major depression. Am J Psychiatry 2003; 160:996–998CrossrefGoogle Scholar

38 Mischoulon D, Best-Popescu C, Laposata M, et al.: A double-blind dose-finding pilot study of docosahexaenoic acid (DHA) for major depressive disorder. Eur Neuropsychopharmacol 2008; 18:639–645CrossrefGoogle Scholar

39 Lewis MD, Hibbeln JR, Johnson JE, et al.: Suicide deaths of active-duty US military and omega-3 fatty-acid status: a case-control comparison. J Clin Psychiatry 2011; 72:1585–1590CrossrefGoogle Scholar

40 Freeman MP, Hibbeln JR, Wisner KL, et al.: Omega-3 fatty acids: evidence basis for treatment and future research in psychiatry. J Clin Psychiatry 2006; 67:1954–1967CrossrefGoogle Scholar

41 Mozaffari-Khosravi H, Yassini-Ardakani M, Karamati M, et al.: Eicosapentaenoic acid versus docosahexaenoic acid in mild-to-moderate depression: a randomized, double-blind, placebo-controlled trial. Eur Neuropsychopharmacol 2013; 23:636–644CrossrefGoogle Scholar

42 Mischoulon D, Nierenberg AA, Schettler PJ, et al.: A double-blind, randomized controlled clinical trial comparing eicosapentaenoic acid versus docosahexaenoic acid for depression. J Clin Psychiatry 2015; 76:54–61CrossrefGoogle Scholar

43 Rapaport MH, Nierenberg AA, Schettler PJ, et al.: Inflammation as a predictive biomarker for response to omega-3 fatty acids in major depressive disorder: a proof-of-concept study. Mol Psychiatry 2016; 21:71–79CrossrefGoogle Scholar

44 Stoll AL, Severus WE, Freeman MP, et al.: Omega 3 fatty acids in bipolar disorder: a preliminary double-blind, placebo-controlled trial. Arch Gen Psychiatry 1999; 56:407–412CrossrefGoogle Scholar

45 Keck PE Jr, Mintz J, McElroy SL, et al.: Double-blind, randomized, placebo-controlled trials of ethyl-eicosapentanoate in the treatment of bipolar depression and rapid cycling bipolar disorder. Biol Psychiatry 2006; 60:1020–1022CrossrefGoogle Scholar

46 Sarris J, Mischoulon D, Schweitzer I: Omega-3 for bipolar disorder: meta-analyses of use in mania and bipolar depression. J Clin Psychiatry 2012; 73:81–86CrossrefGoogle Scholar

47 Peet M, Brind J, Ramchand CN, et al.: Two double-blind placebo-controlled pilot studies of eicosapentaenoic acid in the treatment of schizophrenia. Schizophr Res 2001; 49:243–251CrossrefGoogle Scholar

48 Fenton WS, Dickerson F, Boronow J, et al.: A placebo-controlled trial of omega-3 fatty acid (ethyl eicosapentaenoic acid) supplementation for residual symptoms and cognitive impairment in schizophrenia. Am J Psychiatry 2001; 158:2071–2074CrossrefGoogle Scholar

49 Amminger GP, Schäfer MR, Papageorgiou K, et al.: Long-chain omega-3 fatty acids for indicated prevention of psychotic disorders: a randomized, placebo-controlled trial. Arch Gen Psychiatry 2010; 67:146–154CrossrefGoogle Scholar

50 Amminger GP, Schäfer MR, Schlögelhofer M, et al.: Longer-term outcome in the prevention of psychotic disorders by the Vienna omega-3 study. Nat Commun 2015; 6:7934CrossrefGoogle Scholar

51 Pawełczyk T, Grancow M, Kotlicka-Antczak M, et al.: Omega-3 fatty acids in first-episode schizophrenia - a randomized controlled study of efficacy and relapse prevention (OFFER): rationale, design, and methods. BMC Psychiatry 2015; 15:97CrossrefGoogle Scholar

52 Zanarini MC, Frankenburg FR: Omega-3 fatty acid treatment of women with borderline personality disorder: a double-blind, placebo-controlled pilot study. Am J Psychiatry 2003; 160:167–169CrossrefGoogle Scholar

53 Trebatická J, Hradečná Z, Böhmer F, et al.: Emulsified omega-3 fatty-acids modulate the symptoms of depressive disorder in children and adolescents: a pilot study. Child Adolesc Psychiatry Ment Health 2017; 11:30CrossrefGoogle Scholar

54 Chang JC, Su KP, Mondelli V, et al.: Omega-3 polyunsaturated fatty acids in youths with attention deficit hyperactivity disorder (ADHD): a systematic review and meta-analysis of clinical trials and biological studies. Neuropsychopharmacology (Epub ahead of print July 25, 2017). doi: 10.1038/npp.2017.160CrossrefGoogle Scholar

55 Tan ML, Ho JJ, Teh KH: Polyunsaturated fatty acids (PUFAs) for children with specific learning disorders. Cochrane Database Syst Rev 2016; 9:CD009398Google Scholar

56 Burckhardt M, Herke M, Wustmann T, et al.: Omega-3 fatty acids for the treatment of dementia. Cochrane Database Syst Rev 2016; 4:CD009002Google Scholar

57 Song C, Shieh CH, Wu YS, et al.: The role of omega-3 polyunsaturated fatty acids eicosapentaenoic and docosahexaenoic acids in the treatment of major depression and Alzheimer’s disease: acting separately or synergistically? Prog Lipid Res 2016; 62:41–54CrossrefGoogle Scholar

58 Advice about eating fish: what pregnant women & parents should know. Washington, DC: U.S. Food and Drug Administration, n.d. https://www.fda.gov/downloads/Food/ResourcesForYou/Consumers/UCM536321.pdfGoogle Scholar

59 Ostadrahimi A, Salehi-Pourmehr H, Mohammad-Alizadeh-Charandabi S, et al.: The effect of perinatal fish oil supplementation on neurodevelopment and growth of infants: a randomized controlled trial. Eur J Nutr (Epub ahead of print, July 27, 2017). doi: 10.1007/s00394-017-1512-1CrossrefGoogle Scholar

60 Stoll AL: Omega-3 fatty acids in mood disorders: a review of neurobiological and clinical actions; in Natural Medications in Psychiatric Disorders. Edited by Mischoulon D, Rosenbaum J. Philadelphia, Lippincott, Williams & Wilkins, 2008Google Scholar

61 Mazza M, Marano G, Traversi G, et al.: The complex interplay of depression, inflammation and omega-3: state of the art and progresses in research. Clin Ter 2015; 166:e242–e247Google Scholar

62 Begtrup KM, Krag AE, Hvas AM: No impact of fish oil supplements on bleeding risk: a systematic review. Dan Med J 2017; 64:A5366Google Scholar

63 Gross BW, Gillio M, Rinehart CD, et al.: Omega-3 fatty acid supplementation and warfarin: a lethal combination in traumatic brain injury. J Trauma Nurs 2017; 24:15–18CrossrefGoogle Scholar

64 Baldessarini RJ: Neuropharmacology of S-adenosyl-L-methionine. Am J Med 1987; 83(5A):95–103CrossrefGoogle Scholar

65 Alpert JE, Papakostas GI, Mischoulon D: One-carbon metabolism and the treatment of depression: roles of S-adenosyl nethionine (SAMe) and folate; in Natural Medications for Psychiatric Disorders: Considering the Alternatives. Edited by Mischoulon D, Rosenbaum J. Philadelphia, Lippincott Williams & Wilkins, 2008Google Scholar

66 Spillman M, Fava M: S-adenosyl-methionine (ademethionine) in psychiatric disorders. CNS Drugs 1996; 6:416–425CrossrefGoogle Scholar

67 Mischoulon D, Fava M: Role of S-adenosyl-L-methionine in the treatment of depression: a review of the evidence. Am J Clin Nutr 2002; 76(Suppl):1158S–1161SCrossrefGoogle Scholar

68 Hardy ML, Coulter I, Morton SC, et al.: S-adenosyl-L-methionine for treatment of depression, osteoarthritis, and liver disease. (Evidence Reports/Technology Assessments, No. 64). Rockville, MD, Agency for Healthcare Research and Quality, 2002Google Scholar

69 Papakostas GI: Evidence for S-adenosyl-L-methionine (SAM-e) for the treatment of major depressive disorder. J Clin Psychiatry 2009; 70(Suppl 5):18–22CrossrefGoogle Scholar

70 Sharma A, Gerbarg P, Bottiglieri T, et al.: as Work Group of the American Psychiatric Association Council on Research: S-adenosylmethionine (SAMe) for Neuropsychiatric Disorders: A Clinician-Oriented Review of Research. J Clin Psychiatry 2017; 78:e656–e667CrossrefGoogle Scholar

71 Mischoulon D, Price LH, Carpenter LL, et al.: A double-blind, randomized, placebo-controlled clinical trial of S-adenosyl-L-methionine (SAMe) versus escitalopram in major depressive disorder. J Clin Psychiatry 2014; 75:370–376CrossrefGoogle Scholar

72 Sarris J, Price LH, Carpenter LL, et al.: Is S-adenosyl methionine (SAMe) for depression only effective in males? A re-analysis of data from a randomized clinical trial. Pharmacopsychiatry 2015; 48:141–144CrossrefGoogle Scholar

73 Alvarez E, Udina C, Guillamat R: Shortening of latency period in depressed patients treated with SAMe and other antidepressant drugs. Cell Biol Rev 1987; S1:103–110Google Scholar

74 Berlanga C, Ortega-Soto HA, Ontiveros M, et al.: Efficacy of S-adenosyl-L-methionine in speeding the onset of action of imipramine. Psychiatry Res 1992; 44:257–262CrossrefGoogle Scholar

75 Alpert JE, Papakostas G, Mischoulon D, et al.: S-adenosyl-L-methionine (SAMe) as an adjunct for resistant major depressive disorder: an open trial following partial or nonresponse to selective serotonin reuptake inhibitors or venlafaxine. J Clin Psychopharmacol 2004; 24:661–664CrossrefGoogle Scholar

76 Papakostas GI, Mischoulon D, Shyu I, et al.: S-adenosyl methionine (SAMe) augmentation of serotonin reuptake inhibitors for antidepressant nonresponders with major depressive disorder: a double-blind, randomized clinical trial. Am J Psychiatry 2010; 167:942–948CrossrefGoogle Scholar

77 Carney MWP, Chary TK, Bottiglieri T, et al.: Switch mechanism in affective illness and oral S-adenosylmethionine (SAM). Br J Psychiatry 1987; 150:724–725CrossrefGoogle Scholar

78 Mischoulon D: The herbal anxiolytics kava and valerian for anxiety and insomnia. Psychiatr Ann 2002; 32:55–60CrossrefGoogle Scholar

79 Fernández-San-Martín MI, Masa-Font R, Palacios-Soler L, et al.: Effectiveness of valerian on insomnia: a meta-analysis of randomized placebo-controlled trials. Sleep Med 2010; 11:505–511CrossrefGoogle Scholar

80 Nunes A, Sousa M: [Use of valerian in anxiety and sleep disorders: what is the best evidence?]. Acta Med Port 2011; 24(Suppl 4):961–966Google Scholar

81 Sarris J, Byrne GJ: A systematic review of insomnia and complementary medicine. Sleep Med Rev 2011; 15:99–106CrossrefGoogle Scholar

82 Müller SF, Klement S: A combination of valerian and lemon balm is effective in the treatment of restlessness and dyssomnia in children. Phytomedicine 2006; 13:383–387CrossrefGoogle Scholar

83 Schulz H, Stolz C, Müller J: The effect of valerian extract on sleep polygraphy in poor sleepers: a pilot study. Pharmacopsychiatry 1994; 27:147–151CrossrefGoogle Scholar

84 Francis AJ, Dempster RJ: Effect of valerian, Valeriana edulis, on sleep difficulties in children with intellectual deficits: randomised trial. Phytomedicine 2002; 9:273–279CrossrefGoogle Scholar

85 Glass JR, Sproule BA, Herrmann N, et al.: Acute pharmacological effects of temazepam, diphenhydramine, and valerian in healthy elderly subjects. J Clin Psychopharmacol 2003; 23:260–268CrossrefGoogle Scholar

86 Taavoni S, Ekbatani N, Kashaniyan M, et al.: Effect of valerian on sleep quality in postmenopausal women: a randomized placebo-controlled clinical trial. Menopause 2011; 18:951–955CrossrefGoogle Scholar

87 Pakseresht S, Boostani H, Sayyah M: Extract of valerian root (Valeriana officinalis L.) vs. placebo in treatment of obsessive-compulsive disorder: a randomized double-blind study. J Complement Integr Med 2011; 8. pii: /j/jcim.2011.8.issue-1/1553-3840.1465/1553-3840.1465.xml. https://doi.org/10.2202/1553-3840.1465CrossrefGoogle Scholar

88 Taavoni S, Nazem Ekbatani N, Haghani H: Valerian/lemon balm use for sleep disorders during menopause. Complement Ther Clin Pract 2013; 19:193–196CrossrefGoogle Scholar

89 Kelber O, Nieber K, Kraft K: Valerian: no evidence for clinically relevant interactions. Evid Based Complement Alternat Med 2014; 2014:879396CrossrefGoogle Scholar

90 MacGregor FB, Abernethy VE, Dahabra S, et al.: Hepatotoxicity of herbal remedies. BMJ 1989; 299:1156–1157CrossrefGoogle Scholar

91 Chan TY, Tang CH, Critchley JA: Poisoning due to an over-the-counter hypnotic, Sleep-Qik (hyoscine, cyproheptadine, valerian). Postgrad Med J 1995; 71:227–228CrossrefGoogle Scholar

92 Farrell RJ, Lamb J: Herbal remedies. BMJ 1990; 300:47–48CrossrefGoogle Scholar

93 Garges HP, Varia I, Doraiswamy PM: Cardiac complications and delirium associated with valerian root withdrawal. (letter) JAMA 1998; 280:1566–1567CrossrefGoogle Scholar

94 Ross SM: Valerian root and lemon balm extracts: a phytomedicine compound improves symptoms of hyperactivity, attention deficits, and impulsivity in children. Holist Nurs Pract 2015; 29:391–395CrossrefGoogle Scholar

95 Diaper A, Hindmarch I: A double-blind, placebo-controlled investigation of the effects of two doses of a valerian preparation on the sleep, cognitive and psychomotor function of sleep-disturbed older adults. Phytother Res 2004; 18:831–836CrossrefGoogle Scholar

96 Holst L, Nordeng H, Haavik S: Use of herbal drugs during early pregnancy in relation to maternal characteristics and pregnancy outcome. Pharmacoepidemiol Drug Saf 2008; 17:151–159CrossrefGoogle Scholar

97 Sack RL, Hughes RJ, Edgar DM, et al.: Sleep-promoting effects of melatonin: at what dose, in whom, under what conditions, and by what mechanisms? Sleep 1997; 20:908–915CrossrefGoogle Scholar

98 Sack RL, Brandes RW, Kendall AR, et al.: Entrainment of free-running circadian rhythms by melatonin in blind people. N Engl J Med 2000; 343:1070–1077CrossrefGoogle Scholar

99 Monti JM, Cardinali DP: A critical assessment of the melatonin effect on sleep in humans. Biol Signals Recept 2000; 9:328–339CrossrefGoogle Scholar

100 Vanecek J, Watanabe K: Mechanisms of melatonin action in the pituitary and SCN. Adv Exp Med Biol 1999; 460:191–198CrossrefGoogle Scholar

101 Cajochen C, Kräuchi K, Wirz-Justice A: The acute soporific action of daytime melatonin administration: effects on the EEG during wakefulness and subjective alertness. J Biol Rhythms 1997; 12:636–643CrossrefGoogle Scholar

102 Jan JE, Freeman RD, Fast DK: Melatonin treatment of sleep-wake cycle disorders in children and adolescents. Dev Med Child Neurol 1999; 41:491–500Google Scholar

103 Blackmer AB, Feinstein JA: Management of sleep disorders in children with neurodevelopmental disorders: a review. Pharmacotherapy 2016; 36:84–98CrossrefGoogle Scholar

104 Auld F, Maschauer EL, Morrison I, et al.: Evidence for the efficacy of melatonin in the treatment of primary adult sleep disorders. Sleep Med Rev 2017; 34:10–22CrossrefGoogle Scholar

105 Ferracioli-Oda E, Qawasmi A, Bloch MH: Meta-analysis: melatonin for the treatment of primary sleep disorders. PLoS One 2013; 8:e63773CrossrefGoogle Scholar

106 McCleery J, Cohen DA, Sharpley AL: Pharmacotherapies for sleep disturbances in dementia. Cochrane Database Syst Rev 2016; 11:CD009178Google Scholar

107 Trotti LM, Karroum EG: Melatonin for sleep disorders in patients with neurodegenerative diseases. Curr Neurol Neurosci Rep 2016; 16:63CrossrefGoogle Scholar

108 Ooms S, Ju YE: Treatment of sleep disorders in dementia. Curr Treat Options Neurol 2016; 18:40CrossrefGoogle Scholar

109 Arendt J: Jet-lag and shift work: (2). Therapeutic use of melatonin. J R Soc Med 1999; 92:402–405CrossrefGoogle Scholar

110 Luthringer R, Muzet M, Zisapel N, et al.: The effect of prolonged-release melatonin on sleep measures and psychomotor performance in elderly patients with insomnia. Int Clin Psychopharmacol 2009; 24:239–249CrossrefGoogle Scholar

111 Wade AG, Ford I, Crawford G, et al.: Nightly treatment of primary insomnia with prolonged release melatonin for 6 months: a randomized placebo controlled trial on age and endogenous melatonin as predictors of efficacy and safety. BMC Med 2010; 8:51CrossrefGoogle Scholar

112 Lemoine P, Garfinkel D, Laudon M, et al.: Prolonged-release melatonin for insomnia - an open-label long-term study of efficacy, safety, and withdrawal. Ther Clin Risk Manag 2011; 7:301–311Google Scholar

113 Lyseng-Williamson KA: Melatonin prolonged release: in the treatment of insomnia in patients aged ≥55 years. Drugs Aging 2012; 29:911–923CrossrefGoogle Scholar

114 Dollins AB, Lynch HJ, Wurtman RJ, et al.: Effect of pharmacological daytime doses of melatonin on human mood and performance. Psychopharmacology (Berl) 1993; 112:490–496CrossrefGoogle Scholar

115 Partonen T: Melatonin-dependent infertility. Med Hypotheses 1999; 52:269–270CrossrefGoogle Scholar

116 Mishima K, Satoh K, Shimizu T, et al.: Hypnotic and hypothermic action of daytime-administered melatonin. Psychopharmacology (Berl) 1997; 133:168–171CrossrefGoogle Scholar

117 Wiechmann AF, O’Steen WK: Melatonin increases photoreceptor susceptibility to light-induced damage. Invest Ophthalmol Vis Sci 1992; 33:1894–1902Google Scholar

118 Lehman NL, Johnson LN: Toxic optic neuropathy after concomitant use of melatonin, zoloft, and a high-protein diet. J Neuroophthalmol 1999; 19:232–234CrossrefGoogle Scholar

119 Persengiev S, Marinova C, Patchev V: Steroid hormone receptors in the thymus: a site of immunomodulatory action of melatonin. Int J Biochem 1991; 23:1483–1485CrossrefGoogle Scholar

120 Raghavendra V, Kulkarni SK: Melatonin reversal of DOI-induced hypophagia in rats; possible mechanism by suppressing 5-HT(2A) receptor-mediated activation of HPA axis. Brain Res 2000; 860:112–118CrossrefGoogle Scholar

121 Zhdanova V, Friedman L: Therapeutic potential of melatonin in sleep and circadian disorders; in Natural Medications for Psychiatric Disorders: Considering the Alternatives. Edited by Mischoulon D, Rosenbaum J. Philadelphia, Lippincott, Williams & Wilkins, 2008Google Scholar

122 Amri H, Abu-Asab M, LeBars PL, et al.: Ginkgo biloba extract in cognitive disorders; in Natural Medications for Psychiatric Disorders: Considering the Alternatives. Edited by Mischoulon D, Rosenbaum J. Philadelphia, Lippincott, Williams & Wilkins, 2008Google Scholar

123 Weinmann S, Roll S, Schwarzbach C, et al.: Effects of Ginkgo biloba in dementia: systematic review and meta-analysis. BMC Geriatr 2010; 10:14CrossrefGoogle Scholar

124 Brondino N, De Silvestri A, Re S, et al.: A systematic review and meta-analysis of Ginkgo biloba in neuropsychiatric disorders: from ancient tradition to modern-day medicine. Evid Based Complement Alternat Med 2013; 2013:915691CrossrefGoogle Scholar

125 Zhang HF, Huang LB, Zhong YB, et al.: An overview of systematic reviews of Ginkgo biloba extracts for mild cognitive impairment and dementia. Front Aging Neurosci 2016; 8:276CrossrefGoogle Scholar

126 Yuan Q, Wang CW, Shi J, et al.: Effects of Ginkgo biloba on dementia: an overview of systematic reviews. J Ethnopharmacol 2017; 195:1–9CrossrefGoogle Scholar

127 Hashiguchi M, Ohta Y, Shimizu M, et al.: Meta-analysis of the efficacy and safety of Ginkgo biloba extract for the treatment of dementia. J Pharm Health Care Sci 2015; 1:14CrossrefGoogle Scholar

128 Mazza M, Capuano A, Bria P, et al.: Ginkgo biloba and donepezil: a comparison in the treatment of Alzheimer’s dementia in a randomized placebo-controlled double-blind study. Eur J Neurol 2006; 13:981–985CrossrefGoogle Scholar

129 Yancheva S, Ihl R, Nikolova G, et al.: GINDON Study Group: Ginkgo biloba extract EGb 761(R), donepezil or both combined in the treatment of Alzheimer’s disease with neuropsychiatric features: a randomised, double-blind, exploratory trial. Aging Ment Health 2009; 13:183–190CrossrefGoogle Scholar

130 Cornelli U: Treatment of Alzheimer’s disease with a cholinesterase inhibitor combined with antioxidants. Neurodegener Dis 2010; 7:193–202CrossrefGoogle Scholar

131 Nasab NM, Bahrammi MA, Nikpour MR, et al.: Efficacy of rivastigmine in comparison to ginkgo for treating Alzheimer’s dementia. J Pak Med Assoc 2012; 62:677–680Google Scholar

132 Canevelli M, Adali N, Kelaiditi E, et al.: ICTUS/DSA Group: Effects of Gingko biloba supplementation in Alzheimer’s disease patients receiving cholinesterase inhibitors: data from the ICTUS study. Phytomedicine 2014; 21:888–892CrossrefGoogle Scholar

133 Burkard G, Lehrl S: Verhaltnis von Demenzen vom Multiinfarkt- un vom Alzheimertyp in arztlichen Praxen [Ratio of multi-infarct and Alzheimer-type dementia in medical practices]. Munch Med Wochenschr 1991; 133(Supp. 1):38–43Google Scholar

134 Stoppe G, Sandholzer H, Staedt J, et al.: Prescribing practice with cognition enhancers in outpatient care: are there differences regarding type of dementia? Results of a representative survey in lower Saxony, Germany. Pharmacopsychiatry 1996; 29:150–155CrossrefGoogle Scholar

135 Andrade C, Radhakrishnan R: The prevention and treatment of cognitive decline and dementia: An overview of recent research on experimental treatments. Indian J Psychiatry 2009; 51:12–25CrossrefGoogle Scholar

136 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 Ethnopharmacol 1996; 50:131–139CrossrefGoogle Scholar

137 Leite PM, Martins MAP, Castilho RO: Review on mechanisms and interactions in concomitant use of herbs and warfarin therapy. Biomed Pharmacother 2016; 83:14–21CrossrefGoogle Scholar

138 Kellermann AJ, Kloft C: Is there a risk of bleeding associated with standardized Ginkgo biloba extract therapy? A systematic review and meta-analysis. Pharmacotherapy 2011; 31:490–502CrossrefGoogle Scholar

139 Cohen AJ, Bartlik B: Ginkgo biloba for antidepressant-induced sexual dysfunction. J Sex Marital Ther 1998; 24:139–143CrossrefGoogle Scholar

140 Balon R: Ginkgo biloba for antidepressant-induced sexual dysfunction? J Sex Marital Ther 1999; 25:1–2CrossrefGoogle Scholar

141 Ashton AK, Ahrens K, Gupta S, et al.: Antidepressant-induced sexual dysfunction and Ginkgo biloba. Am J Psychiatry 2000; 157:836–837CrossrefGoogle Scholar

142 Wheatley D: Triple-blind, placebo-controlled trial of Ginkgo biloba in sexual dysfunction due to antidepressant drugs. Hum Psychopharmacol 2004; 19:545–548CrossrefGoogle Scholar

143 Kang BJ, Lee SJ, Kim MD, et al.: A placebo-controlled, double-blind trial of Ginkgo biloba for antidepressant-induced sexual dysfunction. Hum Psychopharmacol 2002; 17:279–284CrossrefGoogle Scholar

144 Meston CM, Rellini AH, Telch MJ: Short- and long-term effects of Ginkgo biloba extract on sexual dysfunction in women. Arch Sex Behav 2008; 37:530–547CrossrefGoogle Scholar

145 Mischoulon D, Rosenbaum J: Afterword; in Natural Medications for Psychiatric Disorders: Considering the Alternatives. Edited by Mischoulon D, Rosenbaum J. Philadelphia, Lippincott, Williams & Wilkins, 2008Google Scholar