Monday, June 1, 2020

Kava ( Piper methysticum G. Forster. f. (Piperaceae) ++ )


HERBAL
MEDICINAL
PLANT
----------------------------------------------------------

 KAVA


 



Piper methysticum  G.  Forster. f. (Piperaceae) ++


BY



RETTODWIKART THENU











KAVA
(kah’vah)  



Piper methysticum  G. Forster. f. (Piperaceae)



SUMMARY AND PHARMACEUTICAL COMMENT

The chemistry of kava is well documented (see Constituents) and there is strong evidence that the kavalactone constituents are responsible for the observed pharmacological activities. Randomised, double-blind, placebo-controlled clinical trials of certain standardised kava preparations have shown beneficial effects on measures of anxiety, although because of methodological limitations of some studies, further welldesigned trials are required to confirm the anxiolytic effects.
Also, most trials have been carried out with one particular standardised kava extract (containing 70% kavalactones) and it cannot be assumed that the effects shown in these studies will be produced by other kava extracts. Clinical trials involving patients with anxiety have also compared welldefined standardised kava preparations with certain standard anxiolytic agents. While these studies have suggested that the kava extracts tested may be as effective as certain standard anxiolytic agents, further investigation is necessary. Data from pharmacological studies provide supporting evidence for the anxiolytic effects of kava, although many of the other traditional uses of kava (see Herbal use) have not been tested scientifically. Many pharmacological studies involving individual kavalactones have investigated the effects of the synthetic kavalactone (_)-kavain, rather than the natural compound (þ)-kavain. Some studies have used both the natural compound and the synthetic racemate and have reported a lack of stereospecific effect.
In placebo-controlled clinical trials, standardised kava extracts generally have been well tolerated; reported adverse events have been mild and transient and similar in nature and frequency to those reported for placebo. Clinical trials, however, can provide only limited information on the safety profile of a medicine. Spontaneous reports of hepatotoxicity associated with the use of kava preparations have arisen since the year 2000. Although the risk of serious liver toxicity is thought to be low, the reaction is idiosyncratic. Against this background, kava was prohibited in unlicensed medicines in the UK in 2003, and in the EU, all licensed kava products were removed from the market. In 2005 in the UK, evidence relating to hepatotoxicity associated with kava was reviewed and the Expert Working Group’s report concluded that there was insufficient new evidence to support a change in the regulatory position, i.e. the inclusion of kava in unlicensed medicines remains prohibited. Regulatory action has also been taken in Canada and Australia (voluntary recall), and in the USA, consumers were warned of the risk of liver toxicity with use of kava-containing products.
Other adverse reactions documented for kava preparations include an ichthyosiform (scaly, non-inflammatory) skin condition, termed ’kava dermopathy’, usually associated with the traditional method of preparing and ingesting kava (see Side-effects, Toxicity, Skin reactions).
In view of the documented pharmacological actions of kava and in view of the reported inhibitory activity against certain cytochrome P450 drug metabolising enzymes, the potential for preparations of kava root/rhizome to interfere with other medicines administered concurrently, particularly those with similar or opposing effects, should be considered. Use of kava should be avoided during pregnancy and breastfeeding.
Although kava is prohibited in the UK and several other countries, individuals may obtain kava preparations over the Internet. Healthcare professionals should be aware that patients may be taking herbal medicinal products containing kava. Healthcare professionals should enquire about use of kava in patients presenting with symptoms of hepatotoxicity  (see Side-effects, Toxicity, Hepatotoxicity). Adverse reactions have been reported in association with use of ’herbal ecstasy’ tablets, which often contain ephedrine alkaloids, although healthcare professionals should be aware that some products have been stated to contain kava. or many centuries, Pacific Islanders have used the kava kava root to prepare a beverage used in welcoming ceremonies for important visitors. Drinking kava kava is not only done to induce pleasant mental states but also to reduce anxiety and promote socialising. It is believed that the first report about kava kava came to the West from Captain James Cook during his voyages through the Pacific region.



TRADE NAMES
Kava Kava (available from numerous manfacturers) Alcohol Free Kava Kava, Kava Kava Power,
Kava Kava Premium, Kava Kava Root

DESCRIPTION
MEDICINAL PARTS: The medicinal parts are the peeled, dried, cut rhizome, which has normally been freed from the roots, and the fresh rhizome with the roots.
FLOWER AND FRUIT: The plant has numerous small flowers in spike-like inflorescences 3 to 9 cm long.
LEAVES, STEM AND ROOT: The plant is a 2 to 3 m high, erect dioecious bush. The leaves are very large, measuring 13 to 28 cm by 10 to 22 cm. They have a deeply cordate base and 9 to 13 main ribs that are slightly soft on the undersurface. The stipules are large. The plant has a massive, 2 to 10 kg, branched and very juicy rhizome with many roots. They are blackish-gray on the outside and whitish on the inside. The fracture is mealy and somewhat splintery. The central portion is porous with irregularly twisted thin woody bundles, separated by broad medullary rays, forming meshes beneath the bark.
CHARACTERISTICS: The taste is pungent and numbing, and the odor is reminiscent of lilac.
HABITAT: The plant is indigenous to the South Sea Islands and is mainly cultivated there.
PRODUCTION: Kava Kava rhizome consists of the dried rhizomes of Piper methysticum.
OTHER NAMES: Ava, Ava Pepper, Intoxicating Pepper, Kawa, Kawa Pepper, Tonga, Kew, Sakau, Yagona



SPECIES (FAMILY)
Piper methysticum Forst. f. (Piperaceae) Fourteen different varieties are used throughout Oceania
(Polynesia, Melanesia, Micronesia).(1)
RELATED SPECIES
Cultivars of P. methysticum have been developed in some Pacific Islands from Piper wichmannii C. DC
(syn: Piper erectum C. DC, Piper arbuscula Trelease).(2, 3)

SYNONYM(S)
Intoxicating Pepper, Kava-kava, Kawa, Kawa-kawa, Macropiper methysticum (G. Forst.) Hook. & Arn., M. latifolium Miq., Waghi, Wati, Bari (Irian Jaya), Koniak, Keu, Oyo (Papua New Guinea)

ORIGIN
Kava is a shrub found on the South Sea Islands. Kava is indigenous to the islands of the South Pacific.
The native Polynesians use the rhizome to make a mildly intoxicating beverage.
Kava rhizome extract has anxiolytic and sedative effects.

PHARMACOPODIAL AND OTHER MONOGRAPHS
BHMA 2003(G66)
BHP 1996(G9)
Complete German Commission E 1998(G3)
Expanded German Commission E 2000(G4)
Martindale 35th edition(G85)
WHO volume 2 2002(G70)

LEGAL CATEGORY (LICENSED PRODUCTS)
Prohibited in unlicensed medicines in the UK.(4)

CONSTITUENTS
Kavalactones Kawalactones, kavapyrones, 2-pyrones, d-lactones with styryl or dihydrostyryl substituents.(1–3, 5–9,G56) Dried rhizomes should contain at least 3.5% kavalactones(G56) and good-quality material 5.5–8.3%.(10) Ethanol–water extracts contain 30% kavalactones, whereas acetone–water extracts contain 70%.(G56) The kavalactones occur as a complex mixture of at least 18 compounds,(5) which are of three main types: styryl enolide pyrones (e.g. kawain (= kavain), dimethoxykawain, methysticin), styryl dienolide pyrones (e.g. yangonin, desmethoxyyangonin), and dihydrostyryl enolide pyrones (e.g. dihydrokawain, dimethoxydihydrokawain, dihydromethysticin). The four major kavalactones of the rhizome are kawain (1–2%), dihydrokawain (0.6–1%), methysticin (1.2–2%) and dihydromethysticin (0.5–0.8%).(G56) Smaller quantities (<0.1%) of dimeric kavalactones (e.g. truxyangonins I, II, III) have also been isolated.(2, 3)
Alkaloids/Amides   Cepharadione A (aporphine-type) is a minor component (4 kg yielded 1 mg).(11) Small quantities of Ncinnamoylpyrrolidine and its O-methoxy analogue are also present.(6, 12, 13)
Chalcones   Flavokawains A, B and C.(6, 9, 14)
Flavonoids   Pinostrobin, 5,7-dimethoxyflavanone.(14)
Steroids   Sitosterol, stigmasterol, stigmastanol.(6, 13)
Esters   Bornyl cinnamate(13) and bornyl 3,4-methylenedioxycinnamate.(14)
Aliphatic Alcohols   Docosan-1-ol, dodecan-1-ol, eicosan-1-ol, hexacosan-1-ol, hexadecan-1-ol, octadecan-1-ol, n tetradecanol, transphytol.(6)
Other Constituents   Cinnamylideneacetone,(5) long-chain fatty acids.(6)

OTHER PARTS OF THE PLANT
Stem peelings may be included as raw material in kava commerce due to the high demand for the rhizome; leaves and branches are used in folk medicine. Pipermethystine (a piperidone amide) is present in stem peelings (traces to 0.85%).(15) 3a,4a-Epoxy-5bpipermethysticin (0.93%) was isolated from stem peelings of one cultivar, but was absent from 10 other cultivars, and the related alkaloid awaine was present in the unopened leaves of 11 cultivars (0.16–2.67%). 7,8-Dihydrokawain, 7,8-dihydromethysticin and 5,6,7,8-tetrahydroyangonin are present in stem peelings.

OTHER SPECIES
Kavalactones occur in P. wichmanni and P. sanctum. The latter species contains several cinnamoyl butenolides (piperolides), e.g. methylenedioxypiperolide and 7,8-epoxypiperolide.(2, 3)

PHARMACOLOGY
·         HERB: Kava rhizome (Piperis methystici rhizoma). The herb consists of the  peeled, cut and dried rhizomes (usually with the root parts removed) of Piper methysticum G. F. and preparations of the same.
·         IMPORTANT Constituents: Kava lactones (kava pyrones, 5–12 %) consisting mainly of ()-kavain (1.8 %), ()-methysticin (1.2 %), desmethoxyyangonin (1 %), and yangonin (1 %).
·         PHARMACOLOGICAL PROPERTIES: Kava pyrones have sedative and central muscle relaxant effects. Anticonvulsive, neuroprotective, narcosis-enhancing, central muscle relaxant, spasmolytic, analgesic, and local anesthetic effects were observed in animals. The herb has anxiolytic and soporific effects in humans.

USES
USES
Kava is used as an anxiolytic, antiepileptic, antidepressant, antipsychotic, and for anxiety, attention defi cit–hyperactivity disorder, insomnia, restlessness, and headaches. It is also used as a muscle relaxant and to promote wound healing.
INVESTIGATIONAL USE
Research is underway for use in cancer.

FOOD USE
Kava is used as an intoxicant drink, on either informal or ceremonial occasions, by Pacific Islanders e.g. from Fiji, Samoa and Tonga.(1, 7) Some claim that it has a pleasant, cooling, aromatic taste with numbing on the tongue and is stimulating, while others refer to great bitterness with a burning sensation in the mouth. It is reputed to reduce fatigue, allay anxieties and produce a cheerful and sociable attitude. Unpleasant effects reported include dizziness, sleeping disorders, stomach pains, lethargy and skin reactions. These reported effects are taken from a wide geographical area and any differences may be due to a number of reasons including plant varieties or growing conditions.

HERBAL USE
In many parts of the Pacific it is believed that kava is beneficial to health by soothing nervous conditions, inducing relaxation and sleep, counteracting fatigue, and reducing weight. Medicinal uses also include treatment of urinary tract infections, asthma, rheumatism, headache, fever, gonorrhoea and syphilis, and use as a diuretic and stomachic.(1, 7, G34) The medicinal use of kava is now widespread, e.g. across Europe, North America and Australia, where it is used to treat anxiety, nervous tension, restlessness, mild depression and menopausal symptoms.(G9, G32, G50, G56, G60, G67) It has also been adopted by the Aboriginal community in parts of Australia as an intoxicating drink.(16) The German Commission E recommended kava for the treatment of nervous anxiety, stress and restlessness.(G3) Traditional uses listed for kava rhizome in other standard herbal and pharmaceutical reference texts include cystitis, urethritis, infection or inflammation of the genitourinary tract, rheumatism and, topically, for joint pains.(G66)

CLINICAL USE
Kava kava extracts are popular in Europe and have been investigated in numerous clinical trials, primarily in European countries. As a result, many research papers have been published in languages other than English. In order to provide a more complete description of the evidence available, secondary sources have been used where necessary. The extract which has been most studied is kava kava extract WS®1490.


Kava Plants Photograph by Inga Spence

Figure 1. Kava (Piper methysticum).


Dikenal Bisa Usir Rasa Cemas, Apa Risiko dan Manfaat Kava Kava?
Figure 2. Kava – dried drug substance (rhizome).


Anxiety
Evidence from double-blind clinical studies indicates that kava kava is an effective treatment for anxiety, repeatedly shown to be as effective as benzodiazepines.
A 2000 Cochrane review of the scientific literature assessed the results from seven, double-blind, randomised, placebo-controlled trials and concluded that kava kava extract has significant anxiolytic activity and is superior to placebo for the symptomatic treatment of anxiety (Pittler & Ernst 2000). An update of this review was published in 2003 and analysed results from 12 clinical studies involving 700 subjects (Pittler & Ernst 2003). The results of seven studies that used the Hamilton Anxiety Scale (HAM-A) score were pooled and a significant reduction in anxiety was observed for kava kava treatment compared with placebo. The results of the five studies that were not submitted to metaanalysis largely support these findings. The extract most commonly tested was WS 1490 at a dose of up to 300 mg daily. According to the authors of the review, none of the trials reported any hepatotoxic events and seven trials measured liver enzyme levels as safety parameters and reported no clinically significant changes.
In 2005, Witte et al published results of another meta-analysis which included data from six placebo- controlled, randomised trials with the kava kava extract WS®1490. The endpoints were the change in HAMA during treatment (continuous and binary). Kava kava significantly improved anxiety with a mean improvement of 5.94 points on the HAMA scale better than placebo. Interestingly, kava kava seemed to be more effective in females and in younger patients. The rigorous meta-analysis found no evidence of publication bias, no remarkable heterogeneity amongst the studies and concluded that trials had high methodological standards. Based on this impressive result, authors concluded that kava kava remains as an effective alternative to benzodiazepines, selective serotonin re-uptake inhibitors (SSRIs) and other antidepressants in the treatment of non-psychotic anxiety disorders.
GENERALISED ANXIETY DISORDER
An 8-week randomised, double-blind, multicentre clinical trial involving 129 outpatients with GAD showed that kava kava LI 150 (400 mg/day) was as effective as buspirone in the acute treatment of GAD, with about 75% of patients responding to treatment (Boerner et al 2003).

COMPARATIVE STUDIES
Comparative studies suggest the absence of significant differences between benzodiazepines and kavain or kava kava extract as treatments for anxiety. A 1993 double-blind, comparative study involving 174 subjects over 6 weeks demonstrated that 300 mg/day of a 70% kava lactone extract produced a similar improvement in anxiety level, as measured by HAM-A scores, to 15 mg oxazepam or 9 mg bromazepam taken daily (Woelk et al 1993). D,Lkavain produced equivalent anxiolytic effects to oxazepam in 38 outpatients with neurotic or psychosomatic disturbances, under double-blind study conditions (Lindenberg & Pitule-Schodel 1990). Benzodiazepine with drawal Kava kava may have a role in reducing anxiety and improving subjective wellbeing during benzodiazepine withdrawal, according to a 2001 randomised, double-blind, placebo-controlled study (Malsch & Kieser 2001). During the first 2 weeks of that study, kava kava dose was increased from 50 mg/day to 300 mg/day while benzodiazepine use was tapered off during the same period. Kava kava extract was superior to placebo in reducing anxiety as measured by the HAM-A scale and improved subjects’ feelings of wellbeing according to a subjective wellbeing scale (Bf-S total scores). Lack of tolerance The results from a randomised, double-blind trial conducted over 25 weeks have found that physical tolerance does not develop to kava kava extract and it is well tolerated (Volz & Kieser 1997). Evidence from a randomised, double-blind study conducted with 84 patients has shown that treatment with kavain (one of the active constituents of kava kava) produces continuous improvements in parameters such as memory function, vigilance, fluency of mental functions and reaction time. Interestingly, these effects were reported over a relatively short period of 3 weeks (Scholing & Clausen 1977). Another randomised, double-blind trial conducted with 52 patients over 28 days not only confirmed anxiolytic activity but also found that kavain promoted subjective vitality-related performance (Lehmann et al 1989). Commission E approves the use of kava kava in conditions of nervous anxiety and restlessness (Blumenthal et al 2000).
Menopausal and perimenopausal anxiety
A randomised, placebo-controlled study conducted with 40 menopausal women found that using kava kava extract, together with hormone replacement therapy (HRT), led to significant reductions in anxiety, as measured by the HAM-A scale at both 3- and 6-month follow-up (De et al 2000). A 3-month, randomised, open study of 68 perimenopausal women showed that treatment with kava kava (100 mg/day) significantly reduced anxiety (P < 0.001) at 1 month and 3 months. This was significantly greater than that spontaneously occurring in controls (P < 0.009) (Cagnacci et al 2003).
Insomnia
The hypnotic activity of kava kava extract was confirmed in a RCT in which a single dose of 300 mg kava kava extract was found to improve the quality of sleep significantly (Emser & Bartylla 1991, as reported by Ernst et al 2001). In vivo experiments with D,L-kavain have shown that it reduces active wakefulness and significantly prolongs sleep, compared with placebo (Holm et al 1991).

OTHER USES
Traditionally, the herb has been used to treat urinary tract infections, asthma, conditions associated with pain, gonorrhoea and syphilis, and to assist with weight reduction, muscle relaxation and sleep. Topically, it has been used as a local anaesthetic and to treat pruritus.



ACTIONS
PHARMACOLOGICAL ACTIONS
Kava has been investigated mostly for its anxiolytic effects, although other central nervous system activities, such as anticonvulsant and analgesic properties, and other effects have been documented following preclinical studies. The kavalactones are believed to be the major active constituents of kava.
IN VITRO AND ANIMAL STUDIES
Pharmacokinetics Uptake of the kavalactones kavain, dihydrokavain, yangonin and desmethoxyyangonin into brain tissue has been documented following intraperitoneal administration of each of these compounds at a dose of 100 mg/kg to mice.(17) Maximum concentrations of kavain and dihydrokavain were noted five minutes after administration, and these compounds were rapidly eliminated. In contrast, yangonin and desmethoxyyangonin were eliminated more slowly. All four compounds were also detected in mouse brain tissue following intraperitoneal administration of kava resin 120 mg/kg (containing kavain 36.7%, dihydrokavain 19.2%, yangonin 15% and desmethoxyyangonin 13.3%), although the concentrations of kavain and yangonin were higher than was noted following individual administration of these constituents.


Central Nervous System Activities
Anxiolytic properties for kava extract and isolated kavalactones have been documented in an experimental model of anxiety, the chick social separation–stress procedure. In a series of experiments, kava extract (containing 30% kavalactones; 30 mg/mL per kg body weight), dihydrokavain (30 mg/mL per kg body weight) and chlordiazepoxide (5 mg/mL per kg body weight) administered intraperitoneally 30 minutes before testing significantly reduced the separation–stress effect (p < 0.05 for each substance).(18) However, the isolated kavalactones kavain, methysticin, dihydromethysticin, yangonin and desmethoxyyangonin administered according to the same regimen did not have a statistically significant effect. Further work using the same experimental model confirmed these findings and found that total kavalactone content was not predictive of outcome, but that a dihydrokavain content of at least 15% was necessary for anxiolytic activity.(19) In this study, the kava samples and fractions that demonstrated anxiolytic activity were reported to be without sedative effects.
In contrast, previous studies have reported sedative effects for kava extract. In mice, kava extract (containing 7% kavalactones) at doses of at least 50 mg/kg body weight (by intraperitoneal injection) reduced spontaneous motility to a greater extent than did control.(20) The effect was enhanced by the addition of (_)- kavain (ratio of kava extract to (_)-kavain, 1 : 0.12), although this compound had no sedative effect when administered alone. In another experimental model, kava extract 100 mg/kg body weight and (_)-kavain 12 mg/kg body weight, each administered alone, had no sedative effect, whereas a combination of the two substances significantly reduced amphetamine (5 mg/kg body weight subcutaneously)-induced hypermotility. Sedative effects have also been documented for an ethanolic extract of kava rhizome (containing 50% kavalactones) 100 mg/kg body weight administered by gastric tube and 200 mg/kg body weight intraperitoneally in the amphetamine-induced hypermotility test and barbiturate-induced sleeping time, respectively.(21)
In studies utilising the conditioned avoidance response test in rats, an experimental model used to test for antipsychotic activity, aqueous (kavalactone-free) kava extract 30–500 mg/kg body weight intraperitoneally had no statistically significant effect.(22) However, administration of kava resin at doses of 125 mg/kg intraperitoneally significantly inhibited the conditioned avoidance response, although to a lesser extent than did chlorpromazine and haloperidol. In cats, a kava extract in arachis oil (50–100 mg kavalactones per kg body weight intraperitoneally) and the individual kavalactone (_)-kavain (10–50 mg per kg body weight intraperitoneally) were reported to be active in the amygdala complex region of the brain.(23)
Receptor binding studies with kava extracts and individual kavalactones have reported conflicting results. One series of experiments found that kava resin and individual kavalactones displayed only weak activity on GABAA- and no activity on GABAB-binding sites in rat brain membranes in vitro, and that there was no significant effect on benzodiazepine receptors following intraperitoneal administration of kava resin 150 mg/kg body weight to mice.(24) A marked effect of kavain on GABA has also been stated to be unlikely.(25) By contrast, a kavalactoneenriched ethanol/aqueous extract of kava rhizome (containing 58% kavalactones and 42% other lipid-soluble compounds) increased the density of GABA-binding sites in certain brain regions.(26) Other experiments have shown concentration- and structure-dependent effects of kavalactones on binding of bicuculline methochloride (BMC) to GABAA receptors from rat cortex preparations.(27) (þ)-Kavain, (þ)-methysticin and (þ)-dihydromethysticin enhanced BMC binding by 18–28% at a concentration of 0.1 mmol/L, whereas (þ)-dihydrokavain did so only at a concentration of 10 mmol/L, and yangonin at a concentration of 1 mmol/L; desmethoxyyangonin had no effect. Further radioreceptor assays demonstrated that these six kavalactones had no effect on the binding of flunitrazepam to benzodiazepine receptors in rat cortex preparations, indicating that the influence of kavalactones on GABAA receptors was not based upon an
interaction with benzodiazepine receptors.(27)
Other in vitro studies have investigated the effects of kava extracts and individual kavalactones on other transmitters in the central nervous system (CNS). A kavalactone-rich kava rhizome extract (containing 68% kavalactones) was a reversible inhibitor of monoamine-oxidase B (MAO-B) in intact and disrupted platelets (inhibitory concentration IC50 24 mmol/L and 1.2 mmol/ L, respectively), although there were differences in MAO-B inhibition among the different synthetic kavalactones with desmethoxyyangonin and (_)-methysticin being the most potent inhibitors.(28) Differences between kavalactones in inhibition of noradrenaline (norepinephrine) uptake in synaptosomes prepared from rat cerebral cortex and hippocampus have also been documented: (_)- and (þ)-kavain gave approximately equal values and both were more potent inhibitors than (þ)-methysticin, although none of the compounds inhibited serotonin uptake.(29) It has been suggested, following in vitro studies involving ipsapirone (a serotonin-1A receptor agonist)-induced field potential changes in guinea-pig hippocampal slices, that kavain and dihydromethysticin may modulate serotonin-1A receptor activity, although further work is needed to identify the precise mechanism for this.(30)
In vivo studies in rats administered a single oral dose of (þ)- dihydromethysticin 100 mg/kg body weight or fed (_)-kavain in the diet over a 78-day period showed that neither kavalactone regimen affected brain tissue concentrations of dopamine and serotonin, although since extracellular neurotransmitter concentrations were not measured in this study, receptor-mediated effects of kavalactones on dopaminergic and serotonergic neurons could not be excluded.(31) In vivo (rats), kava extract 20 and 120 mg/kg body weight intraperitoneally increased dopamine concentrations in the nucleus accumbens, although a dose of 220 mg/kg body weight led to an initial decrease followed by an increase above baseline values.(32) It was suggested that this ceiling effect may be due to yangonin which may have dopamine antagonist activity. The development of physiological tolerance to an aqueous extract of kava administered intraperitoneally to mice has been documented, although there was no clear evidence of development of physiological or learned tolerance to kava resin.(33)
Anticonvulsant And Neuroprotective Activities
Studies described in the older literature have documented anticonvulsant effects for kavalactones in several experimental models.(34, G50) The anticonvulsant properties of (þ)-methysticin in vitro may arise from a direct membrane action on the excitability of neurons,(34) and in vitro assays have shown that (þ)-methysti-cin,(35) (þ)-kavain(36) and the synthetic kavalactone (_)-kavain(34–36) appear to interact with voltage-dependent sodium channels, and that (_)-kavain also interacts with voltage-dependent calcium channels.(37) Inhibition by (_)-kavain of veratridine-activated voltage-dependent sodium ion channels in synaptosomes from rat cerebral cortex,(38) and veratridine-induced increase in intracellular calcium ion concentrations has been described following in vitro studies utilising rat cerebrocortical synaptosomes.(39) Reduction in veratridine-induced glutamate release following (_)-kavain administration has been reported both in vitro(39) and in vivo in freely moving rats.(40) Substances which reduce extracellular glutamate concentrations are of interest for their potential as anticonvulsant agents. Some of the mechanisms described above documented for certain kavalactones may also be important in neuroprotective effects reported for the synthetic kavalactone (_)-kavain. For example, the role of sodium-ion channel blockade in the neuroprotective effect of (_)-kavain against anoxia in vitro has been described,(41) and (_)-kavain (50, 100 or 200 mg/kg intraperitoneally) has been shown to protect nigrostriatal dopaminergic neurons against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced toxicity in mice, an experimental model of Parkinson's disease.(42) A neuroprotective effect against ischaemic brain damage in mice and rats has been demonstrated for kava extract (WS-1490 containing 70% kavalactones) and the individual kavalactones methysticin and dihydromethysticin, but not for kavain, dihydrokavain and yangonin. Kava extract 150 mg/ kg given orally as an emulsion (polyethyleneglycol 400 and water; 20 : 80) one hour before experimentally induced ischaemia, and methysticin and dihydromethysticin (both 10 and 30 mg/kg intraperitoneally 15 minutes before induction of ischaemia), compared with control, significantly reduced the size of the infarct area in mice brains (p < 0.05).(43) In rats, kava extract administered according to the same regimen as used in mice significantly reduced infarct volume compared with control (p <0.05).
Analgesic Activity
Antinociceptive activity in vivo (mice) in the tail immersion test has been documented for kava resin (150 mg/kg intraperitoneally) and for the individual kavalactones dihydrokawain, dihydromethysticin, kavain and methysticin at doses of 150, 275, 300 and 360 mg/kg (intraperitoneally), respectively, compared with controls.(44) Yangonin, tetrahydroyangonin, desmethoxyyangonin and dehydroyangonin had no or only a weak effect. Both kava resin (200 mg/kg, orally) and aqueous kava extract (250 mg/ kg, intraperioneally) displayed antinociceptive activity in the acetic acid-induced writhing test, also in mice. In further tests using both models, naloxone failed to reverse the antinociceptive effects of kava resin or aqueous kava extract, indicating that analgesic activity of kava is achieved via non-opiate pathways.(44) Analgesic activity of dihydrokawain and dihydromethysticin has been reported previously.(45)

Other Activities
Kava extract has been reported to have a muscle relaxant effect in isolated frog muscles, thought to be due to a direct effect on muscle contractility.(46) Reductions in contractions of isolated guinea-pig ileum induced by carbachol and by raised extracellular potassium ion concentrations have been documented for the synthetic kavalactone (_)-kavain, although the compound had no effect on caffeine-induced contractions of ileum strips or on calcium ion-induced contractions of skinned muscles.(47) (_)- Kavain has also been reported to relax maximally contracted murine airway smooth muscle and to reduce carbachol- and potassium chloride-induced airway smooth muscle contraction.(48)
Further investigation indicated that nitric oxide and cyclooxygenase- mediated events did not play a role in kavain-induced relaxation. In contrast, a previous study found that (þ)-kavain inhibited human platelet aggregation in a concentration-dependent manner in vitro.(49) The formation of prostaglandin E2 and thromboxane B2 was also inhibited in a concentration-dependent manner, suggesting that (þ)-kavain is an inhibitor of cyclooxygenase.
An ethanol extract of kava and the isolated kavalactones dehydrokavain, dihydrokavain, kavain, yangonin and methysticin inhibited tumour necrosis factor alpha (TNFa) release in vitro from BALB/3T3 cells incubated with okadaic acid and suppressed lipopolysaccharide-induced TNFa production in vivo in diabetic mice following intraperitoneal administration.(50) Antifungal activity against several microorganisms, including Candida albicans, has been described for a 10% aqueous kava extract.(51)

CLINICAL STUDIES
Clinical trials of kava preparations have focused on investigating anxiolytic effects in various patient groups. Several trials have assessed effects in healthy volunteers, and others have explored the effects of kavain, a constituent of kava. A Cochrane systematic review of monopreparations of kava for the treatment of anxiety included 12 randomised, double-blind, placebo-controlled trials involving a total of 700 participants.(52) All but one of these trials tested the effects of a standardised (70% kavalactones) preparation of kava rhizome (WS-1490) at various dosages but typically equivalent to 60–240 mg kavalactones daily for four weeks. Only two trials were conducted for longer than four weeks; both used a dose equivalent to 210 mg kavalactones daily given for eight weeks in one study(53) and 24 weeks in the other.(54)
Seven of the 12 trials, involving a total of 380 participants, used the total score on the Hamilton Anxiety Scale as their primary outcome measure, and provided data suitable for meta-analysis. Meta-analysis showed a reduction in anxiety scale scores in kava recipients, compared with placebo recipients (weighted mean difference: 3.9; 95% confidence interval (CI), 0.1–7.7; p = 0.05). All except one of these trials included participants with nonpsychotic anxiety; one study involved women with anxiety associated with the climacteric (perimenopausal period). Removing this trial and the trial that did not assess the kava extract WS-1490 from the meta-analysis indicated a statistically significant reduction in anxiety scores for kava recipients compared with placebo recipients (weighted mean difference: 3.4; 95% CI, 0.5– 6.4; p = 0.02).(52)
The five studies not included in the meta-analysis reported statistically significant improvements for kava recipients, compared with placebo recipients, on outcomes (e.g. response rates, reduction in scores on various anxiety scales).(52) These five studies were heterogeneous in that they involved different patient groups, such as women with anxiety associated with the perimenopausal period, individuals with preoperative anxiety, and outpatients with neurotic anxiety. Consequently, dosage regimens of kava varied widely (e.g. equivalent to kavalactones 60 mg in the evening and 1 hour preoperatively, to 140 mg kavalactones daily for four weeks). The conclusions of the review were that kava extract is an effective symptomatic treatment for anxiety, but that limitations of the studies included meant that further rigorous trials were needed.(52) A similar meta-analysis, which included six rando-mised, double-blind, placebo-controlled trials assessing the effects of the kava extract WS-1490 in patients with non-psychotic anxiety disorders reached a similar conclusion.(55)
Some other research has been published, but has added little to the evidence base because of methodological issues. In a randomised, double-blind, placebo-controlled, multicentre trial, 61 patients with sleep disturbances associated with anxiety, tension and restlessness states of non-psychotic origin received a kava extract (WS-1490) 200 mg daily, or placebo, for four weeks.(56) At the end of the study, statistically significant improvements in the two primary outcome measures quality of sleep and recuperative effect after sleep were observed for kava recipients, compared with the placebo group (p = 0.007 and 0.018, respectively; intention-to-treat analysis).
In a randomised, double-blind, placebo-controlled trial conducted entirely over the Internet, 391 adults who scored at least 40 points on the State-Trait Anxiety Inventory (STAI) State scale and who reported having sleeping problems on at least two occasions received capsules containing kava extract (each containing total kavalactones 100 mg) one three times daily, two capsules containing valerian extract (each containing valerenic acids 3.2 mg; no further details of preparation provided) one hour before bedtime, or placebo, for 28 days.(57) At the end of the study, there were no
statistically significant differences between kava and placebo with respect to the primary outcome measures changes from baseline in STAI-state anxiety scores and Insomnia Severity Index scores.
For example, a study involving 68 perimenopausal women reported that kava extract 100 mg (containing 55% kavain) or 200 mg daily improved anxiety compared with no treatment, but participants were not masked as to their treatment allocation.(58) Results from some clinical studies have suggested that kava extracts may be as effective as certain standard anxiolytic agents, although this requires further investigation and confirmation. In a six-week, randomised, double-blind trial involving 172 patients with non-psychotic anxiety, the standardised kava extract WS- 1490 (containing 70% kavalactones) 100 mg three times daily was as effective as oxazepam 5mg and bromazepam 3 mg, each taken three times daily.(59) Another randomised, double-blind, multicentre trial, involving 129 outpatients with generalised anxiety disorder, reported that the kava extract LI-150 400 mg (standardised to 30% kavapyrones = 120 mg) each morning for eight weeks was as effective as buspirone 5mg twice daily and opipramol (a tricyclic antidepressant) 50 mg twice daily.(60)
A randomised, placebo-controlled study involving 40 postmenopausal women with anxiety assessed the effects of a kava extract 100 mg daily (containing 55% kavain) given in addition to hormone replacement therapy (oestrogens plus progestogens or oestrogens alone).(61) It was reported that women who received kava showed greater reductions in anxiety scores than women who received placebo. However, the study had various methodological limitations.
In a randomised, controlled study involving 54 healthy volunteers, kava extract (LI-150, equivalent to 120 mg kavalactones; n = 18) and valerian extract 600 mg (LI-156; n = 18), taken daily for one week, significantly reduced systolic blood pressure following mental stress tests, compared with baseline values, whereas no such reduction was observed in the no-treatment control group (p < 0.001 for both kava and valerian).(62) No effect on diastolic blood pressure was recorded for either herbal preparation, and valerian, but not kava, appeared to reduce heart rate following mental stress tests. These findings require confirmation in placebo-controlled studies, and their relevance to everyday stress needs to be investigated.(62)
Several other randomised, double-blind, controlled trials involving patients with anxiety have compared the effects of the synthetic kavalactone, (_)-kavain, administered at a dose of 200 mg three times daily for 3–4 weeks, with those of placebo,(63) or benzodiazepines, such as oxazepam.(64) One trial assessed the effects of kavain, or placebo, 200 mg three times daily for three weeks in 83 outpatients who had been treated with benzodiazepines for at least six weeks and who were undergoing benzodiazepine withdrawal.(65) Generally, these studies have reported beneficial effects for kavain, but typically have involved only small numbers of patients.
A preliminary study involving healthy individuals assessed the effects of a single 300 mg dose of a kava extract on cognitive performance and mood in a randomised, double-blind, placebocontrolled trial.(66) Some improvements in mood (in trait cheerful participants only) and cognitive performance were observed during the study, although further assessment of the effects of kava on these parameters is required. A case series described improvements after administration of a kava extract (WS-1490) in extrapyramidal side-effects experienced by 42 patients with various psychiatric diagnoses who were receiving neuroleptic drugs.(67) The hypothesis that kava extract can reduce extrapyramidal side-effects requires testing in rigorous randomised controlled trials.
Pharmacokinetics Little is known about the clinical pharmacokinetics of kava preparations. Several metabolites of kavalactones have been detected and identified in human urine following ingestion of around 1 L of kava (prepared by the traditional method of aqueous extraction of kava rhizome) over 1 hour by healthy male volunteers before sleeping.(68) Urine samples were collected before sleeping and on rising in the morning. Kawain, dihydrokawain, desmethoxyyangonin, tetrahydroyangonin, dihydromethysticin, 11-methoxytetrahydroyangonin, yangonin, methysticin and dehydromethysticin were detected unchanged in human urine; metabolic transformations observed included reduction of the 3,4-double bond and/or demethylation of the 4 methoxyl group of the kavalactone ring. The C12 hydroxy analogue of yangonin (12-hydroxy-12-desmethoxyyangonin) was also detected, and it may have been formed by demethylation of yangonin and/or C12 hydroxylation of desmethoxyyangonin. Dihydroxylated metabolites of the kavalactones and products from ring opening of the kavalactone ring were not detected.(68)


MAIN ACTION 
Central Nervous System (CNS) Effects
The kava lactones reach a large number of targets that influence CNS activity and act centrally and peripherally. They interact with dopaminergic, serotonergic, GABA-ergic and glutamatergic neurotransmission, seem to inhibit monoamine oxidase B and exert multiple effects on ion channels, according to in vitro and in vivo research (Grunze et al 2001). Additionally, animal studies show that kava lactones are chiefly responsible for these effects that give rise to many of the herb’s clinical actions (Cairney et al 2002).
Hypnotic
Although the exact mechanism of action is not yet understood, it has been observed that sleep promotion may be due to the preferential activity of D,L-kavain and kava kava extract on the limbic structures and, in particular, the amygdalar complex (Holm et al 1991) in the brain. In an electroencephalogram (EEG) brainmapping study it was demonstrated that D,Lkavain could induce a dose-dependent increase in delta-, theta- and alpha-1 power, as well as a decrease in alpha-2 and beta power. These results indicate a sedative effect at the higher dose range (Frey 1991).
Anxiolytic Effects
A recent study showed that kava kava extract produces a statistically significant dose-dependent anxiolytic-like behavioural change in rat models of anxiety (Garrett et al 2003). The effect is not mediated through the benzodiazepine binding site on the GABA-A receptor complex, as flumazenil, a competitive benzodiazepine receptor antagonist, did not block this effect. Meta-analyses confirm that the anxiolytic effect is clinically significant and comparable to benzodiazepines.
Analgesic And Local Anaesthetic
Both the aqueous and lipid-soluble extracts of kava kava exhibit antinociceptive properties in experimental animal models (Jamieson & Duffield 1990). The effect is not mediated by an opiate pathway, as naloxone does not reduce the effects when administered in doses that reverse the effects of morphine. More recently, in vitro research has identified several compounds found in kava kava that have the ability to inhibit COX-1 and to a lesser extent COX-2 enzyme activities (Wu et al 2002). The local anaesthetic effect of kava kava is well known for topical use and has been described as similar to procaine and cocaine (Mills & Bone 2000).
Antispasmodic Activity
Antispasmodic activity for skeletal muscle has been observed in vitro and in vivo for both kava kava extract and kava lactones (Mills & Bone 2000). In vivo research suggests that kavain impairs vascular smooth muscle contraction, likely through inhibition of calcium channels (Martin et al 2002).

OTHER ACTIONS
Cytochrome Inhibition
In vitro studies published in 2002 suggested that whole kava kava extract and kava lactones have widespread inhibitory effects on various cytochrome enzymes, such as CYP3A4 (Unger et al 2002), whereas in vivo tests found no effects on CYP3A4/5, CYP1A2 or CYP2D6, but did demonstrate significant inhibition (approximately 40%) of CYP2E1 (Gurley et al 2005). Clinical tests confirm no significant effect on CYP2D6 (Gurley et al 2008).
Animal studies found that kava kava extract decreased the expression of CYP2D1 (human CYP2D6 homolog) at a dose of 2.0 g/kg in females and increased the expression of CYP1A2, 2B1 and 3A1 in 1.0 and 2.0 g/kg groups of both sexes (Clayton et al 2007). The clinical relevance of these findings is unclear, as the doses used were extremely large and not clinically relevant.  Kava acts as a sedative, an analgesic, and an anxiolytic. It has been used for ceremonial purposes in Micronesia and Polynesia for thousands of years in the place of alcoholic beverages, which have not always been available.
Sedative Action
The sedative action of kava is unlike any other. It appears to act directly on the limbic system. Kava lactones may actually modify receptor areas rather than bind to receptor binding sites (Holm et al, 1991).
Anxiolytic Action
There appears to be no lack of effectiveness, even at large doses over time. Several studies confi rm the ability of kava to decrease anxiety. One study used 84 volunteers with anxiety conditions who received kavain, a kava lactone, in doses of 400 mg/day. In the experimental group, the result was an increase in memory and reaction time (Scholing et al, 1977). A more recent study showed a signifi cant reduction of anxiety symptoms with the use of kava (Pittler et al, 2000). One group of volunteers was given 100 mg of kava extract three times daily, while the other received a placebo. After 4 weeks, when the subjects were evaluated using the Hamilton Anxiety Scale, the kava group reported a signifi cant decrease in anxiety symptoms (Kinzler et al, 1991).
Analgesic, Antiinfl ammatory Action
The analgesic effect of kava appears to be unrelated to that of other pain relievers. Kava does not bind to opiate receptors and does not block pain impulses in the central nervous system. Its mechanism of action is unknown at present. One study (Folmer et al, 2006) identifi ed kava as possessing TNF-alpha–induced activation of a nuclear factor. This information leads the researcher to believe that kava could be used for antiinfl ammatory conditions.

ACTIVITIES
Allergenic (1; CRC); Analgesic (1; APA; FNF; WAM); Anaphrodisiac (f; MAD); Anesthetic (1; BGB; CRC; MAB; MAD; PH2); Antiaggregant (1; MAB); Antibacterial (1; MAB; MAD); Anticonvulsant (1; FNF; KOM; PH2; SHT); Antidepressant (1; APA); Antidopamine (1; MAB); Antiepileptic (1; BGB); Antifatigue (f; PNC); Antiischemic (1; MAB); Antipyretic (1; MAB); Antirheumatic (1; FNF); Antiseptic (1; FNF; MAD); Antispasmodic (1; APA; BGB; CRC; PH2; WAM); Antithrombic (1; PH2); Anxiolytic (1; KOM; MAB; PHR; PH2; WAM); Aperitif (1; MAD); Aphrodisiac (f; APA; CRC); Climacteric (f; BGB); CNS-Depressant (1; APA); Contraceptive (f; MAB); Cyclooxygenase Inhibitor (1; PH2); Diaphoretic (f; CRC; MAD); Diuretic (1; APA; MAB; MAD; PNC); Dopaminergic (1; PH2); Expectorant (f; CRC); Fungicide (1; CRC; MAB); Hypnotic (1; MAB; PH2); Hyporeflexic (1; BGB); Lactagogue (f; CRC); Memorigenic (1; MAB); Myorelaxant (1; APA; FNF; KOM; PH2; SKY); Narcotic (1; CRC); Neuroprotective (1; HH2); Psychotropic (f; PH2); Sedative (2; FNF; KOM; PH2; WAM); Serotoninergic (1; PH2); Sobering (1; MAB); Stimulant (f; CRC; PNC); Tonic (f; CRC; MAD; PNC); Tranquilizer (1; APA).


INDICATIONS
Anorexia (1; MAB; MAD); Anxiety (2; APA; KOM; MAB; PHR; PH2; WAM); Arthrosis (f; MAD); Asthma (f; BGB; PH2); Backache (f; CRC); Bacteria (1; MAB; MAD); Blennorrhea (f; MAD); Bronchosis (f; PNC); Catarrh (f; MAB); Chill (f; CRC); Cholecystosis (f; MAB); Cold (f; CRC; MAB); Colic (f; MAB); Congestion (f; MAD); Convulsion (1; FNF; KOM; PH2; SHT); Cough (f; CRC; MAB); Cramp (1; APA; BGB; CRC; MAB; PH2; WAM); Cystosis (f; MAD; PH2); Debility (f; CRC; MAB); Depression (1; APA; BGB); Dermatosis (f; CRC; MAB; MAD); Despondency (f; MAB); Dizziness (1; APA; MAB; MAD); Dysmenorrhea (1; FNF; SHT; WAM); Dyspepsia (1; APA; PH2); Dysuria (1; WAM); Earache (1; MAB; MAD); Eczema (f; MAD); Elephantiasis (f; CRC); Encephalosis (f; MAD); Enterosis (1; WAM); Enuresis (f; MAB); Epilepsy (1; BGB; MAB); Fatigue (1; MAB); Fever (1; CRC; MAD; MAB); Filariasis (f; MAB); Fungus (1; CRC; MAB); Gastrosis (f; PH2); Gonorrhea (f; CRC, MAB; MAD; PH2); Gout (f; APA; PNC); Headache (1; APA; CRC; FNF; MAD); Heart (f; CRC); Hemorrhoid (f; MAB); Herpes (f; MAD); Hot Flash (f; BGB); Hyperactivity (1; APA; WAM); Ichthyosis (f; MAD); Incontinence (f; MAB); Infection (1; CRC; MAB); Insomnia (2; APA; FNF; KOM; MAB; PHR; PH2; WAM); Leprosy (f; MAB); Leukorrhea (f; CRC; MAB); Menopause (1; APA; BGB; MAB); Menstrual Cramp (1; FNF); Migraine (1; APA); Myalgia (1; MAB); Mycosis (1; CRC; MAB); Nephrosis (f; CRC; MAB); Nervousness (2; APA; FNF; KOM; PHR; PH2; WAM); Neuralgia (f; MAB); Neurasthenia (f; CRC; MAD); Obesity (f; PH2); OCD (1; WAF); Ophthalmia (f; MAB); Pain (1; APA; BGB; CRC; FNF; MAB; MAD; PH2; WAM); Palpitation (1; APA); Prolapse (f; MAB); Prostatosis (f; MAD); Psoriasis (f; MAD); Pulmonosis (f; CRC); Restlessness (2; APA; KOM); Rheumatism (1; CRC; FNF; PH2; PNC); Sore Throat (f; MAB); Stomachache (1; APA); Stress (2; APA; KOM; PH2; SHT); Syphilis (f; PH2); Thrombosis (1; PH2); Toothache (1; MAB; MAD); Tuberculosis (f; CRC); Urethrosis (f; CRC; MAD; PH2); UTI (f; BGB; MAB); Vaginosis (f; CRC; MAB); VD (f; APA; CRC; MAD; PH2); Vertigo (f; MAB); Water Retention (1; APA; MAB; MAD; PNC); Wet Dream (f; CRC).

INDICATIONS AND USAGE
• Nervousness and insomnia
Kava Kava is used for nervous tension, stress and agitation.
Unproven Uses: In folk medicine, the herb is used as a sleeping agent and sedative; for asthma, rheumatism, dyspeptic symptoms, chronic cystitis, syphilis, gonorrhea and weight reduction.
Homeopathic Uses: Kava Kava is used for states of excitement and exhaustion. It is also used for gastritis and pain in the urethra. 

INDICATIONS
Nervous tension, states of tension and anxiety.

PRODUCT AVAILABILITY
Capsules, beverage, extract, tablets, tincture
PLANT PARTS USED: Dried rhizome, dried roots  (sometimes referred to incorrectly as the root)


DOSAGES
DOSAGES
Anxiolytic
·         Adult PO extract, standardized: 45-70 mg kava lactones tid (Murray, Pizzorno, 1998)
Depression
·         Adult PO extract, standardized: 45-70 mg kava lactones tid (Murray, Pizzorno, 1998)
General Use
·         Adult PO extract, standardized: 70 mg kava lactones tid (Foster, 1998)
·         Adult PO capsules/tablets: 400-500 mg up to 6 times/day (Foster, 1998)
·         Adult PO tincture: 15-30 drops (dilution 1:2 ) taken tid in water (Foster, 1998)
Sedative
·         Adult PO extract, standardized: 190-200 mg kava lactones 60 min at bedtime

DOSAGES
Dosages for oral administration (adults) for treatment of anxiety recommended in older and contemporary standard herbal reference texts are given below.
·         Dried Rhizome   1.5–3 g per day.(G50) Equivalent to 60–120 mg kavalactones per day.(G3)
·         Liquid Extract   3–6mL per day (1 : 2 liquid extract, unspecified solvent).(G50)
Standardised Preparations
·         100–200 mg kavalactones per day.(G50)
·         60 mg kavalactones 2–4 times per day in tablet form.(G50)
·         60–120 mg kavalactones per day.(G3)
Kava exists in numerous varieties of differing potency (7) and only preparations with standardised kavalactone content should be used for medicinal purposes. Medicinal extracts prepared with ethanol–water yield dry extracts with about 30% kavalactones content, whereas acetone–water prepared dry extracts contain about 70% kavalactones.(G56)
Dosages (adults) used in clinical trials have varied widely, but typically are those equivalent to 60–240 mg kavalactones daily by oral administration in divided doses (see Pharmacological Actions, Clinical studies). Duration of use of kava extracts generally should not exceed three months.(G3, G4, G56)

DOSAGES
·         1 tsp cup/night (JAD); 1.5–3 g dry root/day (MAB); 100–300 mg root several x/day (MAD); 2–4 g powdered root 1–3 x /day (AHP; PNC); 2–4 ml liquid root extract (PNC);
·         3–6 ml fluid extract (1:2)/day (MAB); 1–3 ml tincture/day (SKY); 60–600 mg kavalactones/day (AHP);
·         ca 250 ml kavalactones/day (24–70 mg 3 x /day) (APA); 180–210 kavalactones 1 hour before bedtime (APA);
·         1 (525 mg) capsule (StX with 250 mg certified potency kava-kava root extract with at least 75 mg kavalactone) 3 x /day (NH).

DOSAGES RANGE
·         Cut Rhizome: 1.7–3.4 g/day.
·         Dried Rhizome: 1.5–3 g/day in divided doses or equivalent to 60–120 mg kavapyrones daily.
·         Fluid Extract (1:2): 3–8.5 mL/day in divided doses.
Ideally, ethanolic extracts should contain > 20 mg/ mL kava lactones.
According to clinical studies
·         Anxiety: generally doses up to 300 mg daily of kava kava extract WS 1490 providing 105–210 mg kavalactones. A kava kava extract LI 150 (400 mg/day) was used successfully in generalised anxiety disorder.
·         Insomnia — a single dose of 300 mg kava kava extract.
·         Benzodiazepine with drawal — 300 mg/day of kava kava extract.

DOSAGES AND DURATION OF USE
Daily dose: 60–120 mg of herb preparations.

DOSAGES
Mode of Administration: Comminuted rhizome and other galenic preparations for oral use.
How Supplied:
Capsules — 100 mg, 125 mg, 128 mg, 150 mg, 250 mg, 390 mg, 400 mg, 425 mg, 455 mg, 500 mg
Liquid — 1:1, 1:2
Preparation: There are a number of different extraction recipes depending on the pharmaceutical companies.
Daily Dosage:
Capsules — The root extract is taken 150 mg to 300 mg twice daily, with a daily dosage of kava pyrones 50 to 240 mg (Herberg, 1996; Lehmann, 1996).
Tincture — The tincture is taken as 30 drops with water three times daily (Chavallier, 1996).
Infusion — Take 1/2 cup twice daily (Chavallier. 1996).
Note: The drug should be administered with food or liquid due to its lipid solubility (Fachinfo Antares 120 (R). 1996). The activity of the herb is enhanced when mixtures of the kava pyrones are taken instead of a single pyrone (Jamieson,1989).
Homeopathic  Dosage: The herb is taken as 5 to 10 drops, 1 tablet or 5 to 10 globules 1 to 3 times daily, or 1 ml injection solution sc twice weekly (HAB1).
Storage: The herb should be stored away from direct light, moisture and heat at room temperature.

PRECAUTIONS AND ADVERSE REACTIONS

GENERAL: No health hazards are known in conjunction with the proper administration of designated therapeutic dosages. Administration of the herb leads to rare cases of allergic reactions and gastrointestinal complaints. Slight morning tiredness can appear at the beginning of the therapy. Motor reflexes and judgment when driving may be reduced while taking the herb.
CENTRAL NERVOUS SYSTEM: Dyskinesia and choreoathetosis of the limbs, trunk, neck and facial musculature have been reported secondary to the administration of kava (Schelosky, 1995; Spillane, 1997).
ENDOCRINE: Following long-term use of high doses of Kava extract, weight loss was reported (Mathews, 1988).
HEPATOTOXICITY: Increase in gamma-glutamyl transferase (GGT) levels have been associated with high doses of Kava extract (Mathews, 1988). Two cases of acute hepatitis with an increase of liver enzymes were reported. Necrotizing hepatitis was determined after a liver biopsy, and upon discontinuation of Kava, liver tests normalized (Stahl, 1998).
MUSCULOSKELETAL: Minor inhibition of movement and impaired motor reflexes have been observed with the use of Kava (Jamieson, 1990).
OCULAR: Increase in pupil diameter, reduction of the near point of accommodation and near point of convergence, and disturbance to the oculomotor balance have been reported with Kava (Garner, 1985). Eye irritation has been reported with the heavy consumption of Kava (Ruze, 1990).
SKIN: Heavy chronic consumption of Kava is associated with a peculiar, scaly rash suggestive of ichthyosis (Ruze, 1990). A reversible, slight yellowing of the skin has been associated with long-term use of Kava. Sebotropic drug reactions resulting from Kava-Kava extract therapy has been reported (Jappe, 1998). The drug should not be taken for longer than three months without a doctor's supervision.

DRUG INTERACTIONS:
ALCOHOL — Concomitant use of Kava Kava with alcohol results in an increase in each other's hypnotic action. The alcohol also increases the possibility for kava toxicity (Jamieson, 1990).
ALPRAZOLAM —- Kava used simutaneously with alprazolam has resulted in coma (Almeida, 1996).
CNS Depressants, such as Barbituates — The herb may potentiate the effectiveness of substances that act on the central nervous system.
PSYCHOACTIVE AGENTS — The intensity of psychoactive agents may be intensified with kava (Jamieson, 1990).
DOPAMINE — Kava Kava has been reported to antagonize the effect of dopamine. Patients with Parkinson's Disease taking levodopa should avoid the use of the herb (Baum, 1998; Cupp, 1999; Schelosky, 1995).
PREGNANCY: the drug is contraindicated during pregnancy.
NURSING MOTHERS: The drug is contraindicated in nursing mothers.

ADVERSE EFFECTS: The prolonged use of high doses of kava can, in rare cases, lead to gastrointestinal complaints, oculomotor equilibrium disorders, pupil dilation, and insufficiency of accommodation. Slight morning fatigue can occur in the initial phase of treatment. Disorders of complex movement with otherwise unimpaired consciousness are initial signs of overdose, followed by fatigue and a tendency to fall asleep. Kava increases the action of substances that affect the central nervous system, e. g., alcohol, barbiturates, and other psychoactive drugs. A few studies yielded some indication of hepatotoxicity in relation to administration of kava. Though this information is limited to date and still awaits scientific evaluation, it is recommended to consider the following when using kava products.
WARNING: Kava should not be taken on a daily basis for more than 4 weeks.
WARNING: Use of kava should be discontinued if symptoms of jaundice appear.
WARNING: Patients with a history of liver problems or who suspect possible liver problems or who are taking pharmaceutical drugs should use kava only with the advice of a professional health care provider.
WARNING: See p. 212 for cautions in the use of kava in disorders of the nervous system.
HERB–DRUG INTERACTIONS: Kava should not be used by anyone who has liver problems, is taking any drugs with known adverse effects on the liver, such as NSAIDS, or is a regular consumer of alcohol.

ADVERSE REACTIONS
In RCT, the incidence of adverse effects to kava kava has been found to be similar to placebo. Two postmarketing surveillance studies involving more than 6000 patients found adverse effects in 2.3% and 1.5% of patients taking 120–240 mg standardised extract (Ernst 2002). The most common side effects appear to be gastrointestinal upset and headaches when used in recommended doses.
Hepatotoxicity
A systematic review assessing the safety of kava kava which included a total of 7078 patients taking kava kava extract equivalent to 10 mg to 240 mg kava lactones per day for 5–7 weeks identified no cases of hepatotoxicity (Stevinson et al 2002). Considering that case reports of hepatotoxicity exist, they should be considered a very rare event based on the evidence. In 2008, a quantitative causality assessment of 26 critical cases came to a similar conclusion stating kava kava taken as recommended is associated with rare hepatotoxicity, whereas overdose, prolonged treatment, and co-medication may carry an increased risk (Teschke et al 2008). Importantly, a recent World Health Organization report identified that liver toxicity risk is associated with kava kava acetonic and ethanolic extracts, whereas the traditional kava kava preparation which has been prepared for centuries in water does not have the same risk of liver injury. To put the risk into perspective, estimates from case reports and the sales figures of kava kava extracts (using data from Germany) show an incidence rate of one potential case in 60–125 million patients for liver toxicity (Sorrentino et al 2006).
Possible Mechanisms for Kava-Kava-Induced Hepatotoxicity
The exact cause remains elusive; however, several mechanisms have been proposed. Genetic polymorphism of cytochrome enzymes, leading to interindividual variation in drug metabolism, may be one important factor in the marked discrepancy in hepatotoxic response to kava kava (Singh 2005). Other possible mechanisms are inhibition of cytochrome P450 by kava kava, reduction in liver glutathione content and, more remotely, inhibition of cyclooxygenase enzyme activity. The direct toxicity of kava kava extracts is quite small under any analysis; yet, the potential for drug interactions and/or the potentiation of the toxicity of other compounds is larger (Clouatre 2004). Recent animal tests with three different kava kava extracts (a methanolic and an acetonic root and a methanolic leaf extract) indicate that the these kava kava extracts are toxic to mitochondria, leading to inhibition of the respiratory chain, increased reactive oxygen species (ROS) production, a decrease in the mitochondrial membrane potential and eventually to apoptosis of exposed cells. In predisposed patients, mitochondrial toxicity of kava kava extract may provide another explanation for hepatotoxic reactions (Lude et al 2008). It has also been suggested that reactions are immunologically mediated (Schulze et al 2003). Considering that the toxic reactions are limited to methanolic and ethanolic extracts, it is possible that chemicals other than kava lactones, such as alkaloids not bioavailable in water extracts, may be responsible for hepatotoxicity.
Long-Term Use
Heavy kava kava drinkers acquire a reversible ichthyosiform eruption, known as kanikani in Fijian or ‘kava dermopathy’ in English-speaking countries. This condition is characterised by yellow discolouration of the skin, hair and nails. This temporary condition reverses once kava kava use is discontinued. A 2003 report found no evidence of brain dysfunction in heavy and long-term kava kava users (Cairney et al 2003).

SIGNIFICANT INTERACTIONS
Alcohol
Potentiation of CNS sedative effects has been reported in an animal study; however, one doubleblind, placebo controlled study found no additive effects on CNS depression or safety related performance (Herberg 1993). Alternatively, a study of 10 subjects found that when alcohol and kava kava were combined, kava kava potentiated both the perceived and measured impairment compared to alcohol alone (Foo & Lemon 1997). Caution is advised when taking this combination together.
Barbiturates
Additive effects are theoretically possible. Use with caution and monitor drug dosage. However, interaction may be beneficial under professional supervision.
Benzodiazepines
Additive effects are theoretically possible. Use with caution and monitor drug dosage. However, interaction may be beneficial under professional supervision. The combination has been used successfully to ease symptoms of benzodiazepine withdrawal. Antagonistic effects are theoretically possible, thereby reducing the effectiveness of l-dopa. Avoid concurrent use unless under professional supervision until safety is confirmed.
Methadone and morphine
Additive effects with increased CNS depression are theoretically possible, so use with caution, although interactions may be beneficial under professional supervision.
Substrates for CYP2E1
Inhibition of CYP2E1 has been demonstrated in vivo — serum levels of CYP2E1 substrates may become elevated — use caution.


OVERDOSAGE
Overdosage can result in disorders of complex movement, accompanied by undisturbed consciousness, later tiredness and tendency to sleep.


CONTRAINDICATIONS, INTERACTIONS, AND SIDE EFFECTS
CLASS 2B, 2C, 2D. Contraindicated for endogenous depression (AHP). Maximum tolerated doses for dogs was 60 mg/kg, for rats 320 mg/kg StX (70% kavapyrones). Perversely, if the authors didn’t misspeak, the dogs tolerated 24 mg/kg/day. Of >4000 patients taking 105 mg/day StX (70% kavapyrones), 1.5% had objectionable side effects (allergy, dizziness, GI distress, and headache). At levels 100 times the therapeutic dose (roughly 13 liters kava beverage a day or 300–400 mg rhizome per week) caused anorexia, ataxia, dyspnea, hair loss, red eyes, skin rash, visual problems, and yellow skin. “There is no potential for physical or psychological dependency. Use should not exceed 3 months.” (AHP) Germans limit use to 1–3 months (AHP). Commission E reports contraindications: esophageal and gastrointestinal stenoses; adverse effects: allergic reactions (rarely). Other sources report intestinal obstruction (AEH). Many reports suggest a yellowing of the skin in chronic users. “Chronic ingestion may lead to ‘kawism’ characterized by dry, flaking, discolored skin, and reddened eyes” (LRNP, May 1987). Persistent rumors suggest that overdoses can cause intoxication. Commission E warns against the concomitant use of kava with barbituates, antidepressant medications, and CNS agents. Lactating or pregnant women should not use kava (WAM). “Not permitted as a non-medicinal ingredient in oral use products in Canada” (Michols, 1995). Abuse by Australian Aborigines suggest links to hematuria, infectious disease, neurological abnormalities, pulmonary hypotension, nephrosis, visual disturbances, ischemic heart disease, thrombosis, and sudden heart attacks (MAB). The following quote might scare abusers, as it should, “Full consciousness is maintained with even fatal doses” (APA, quoting Weiss, 1988).

CONTRA-INDICATIONS, WARNINGS
It has been stated that kava is contra-indicated in endogenous depression.(G3, G4) Even when administered in accordance with recommended dosage regimens, kava may adversely affect motor reflexes, and may affect ability to drive and/or operate machinery.( G3,G4) It has been reported that there is no evidence that use of kava extracts has the potential for physical or psychological dependency to develop.(G56) However, as most clinical studies of kava extracts have been of short duration, typically around four weeks (maximum 24 weeks) and/or usually have involved only small numbers of participants, further study is required before definitive statements are made on the potential for dependency with kava.
Drug interactions There is an isolated report of a 54-year-old man who was taking alprazolam, cimetidine and terazosin and who became lethargic and disoriented three days after he began taking kava purchased from a health-food store (no further details of the kava preparation were provided).(106) The man was hospitalised and his symptoms resolved after several hours. He tested negatively for alcohol, and positively for benzodiazepines; the man stated he had not taken overdoses of either alprazolam or kava. The clinical importance and role of kava in this reaction is not known, although there is a view that concurrent use of kava  and substances with central nervous system effects could lead to enhanced activity.(G3, G4)
The effects on performance of a kava extract given in combination with bromazepam have been explored in a randomised, double-blind, controlled crossover trial involving 18 healthy volunteers. Participants received a kava extract (Antares) equivalent to 120 mg kavalactones twice daily, or bromazepam 4.5 mg twice daily, or both agents, for 14 days.(107) Significant reductions in indicators of performance, such as motor coordination, were reported for recipients of both kava and bromazepam, compared with recipients of kava alone, but there was no difference between kava plus bromazepam compared with bromazepam alone. Isolated case reports of extrapyramidal symptoms associated with use of kava extracts have led to the suggestion that constituents of kava may have dopamine antagonist effects (see Side-effects, Toxicity, Central nervous system effects).(88) On this basis, the potential for kava to interact with dopamine agonists or antagonists should be considered.
Inhibition of certain cytochrome P450 (CYP) drug metabolising enzymes has been shown in vitro and in vivo for kava extracts and individual kavalactones. In a randomised, open-label, crossover study, 12 healthy volunteers received kava root extract 1000 mg twice daily (subsequently found to be equivalent to total kavalactones 138 mg daily) for 28 days; probe drugs were administered before and after kava administration to assess effects on CYP enzymes.(108) Administration of the kava extract according to this dosage regimen inhibited CYP2E1, but not CYP3A4/5, CYP2D6 and CYP1A2. In in vitro studies, methanolic, acetone and ethyl acetate extracts of kava rhizome significantly inhibited CYP3A4 activity, compared with control, at concentrations as low as 10 mg/mL (ethyl acetate extract).(109) In other in vitro experiments, several individual kavalactones were tested for their effects on the activities of CYP1A2, CYP2C9, CYP2C19 and CYP2D6, as well as CYP3A4. Desmethoxyyangonin, dihydromethysticin and methysticin produced a concentration-dependent inhibition of one or more of the CYP isoforms at concentrations of <10 mmol/ L, considered as 'potent' inhibition (e.g. IC50 values for desmethoxyyangonin, dihydromethysticin and methysticin for CYP2C19 were 0.51, 0.43 and 0.93 mmol/L, respectively, and for dihydromethysticin and methysticin for CYP3A4 under certain assay conditions were 2.49 and 1.49 mmol/L, respectively).(110) In several cases, this degree of inhibition was greater than that shown by positive controls which are known to produce clinically significant drug interactions. Similar results have been reported for an ethanolic kava root extract and the individual kavalactones desmethoxyyangonin, dihydromethysticin and yangonin: in two in vitro models, both the extract and the individual kavalactones inhibited CYP1A2, CYP2C9, CYP2C19, CYP2E1 and CYP3A4 with IC50 values of around 10 mmol/L.(104)
Differences in CYP enzyme inhibition have been described for commercial preparations (acetone, methanol and ethanol extracts) and traditional aqueous extracts, with commercial preparations having a greater inhibitory effect than traditional preparations on CYP3A4, CYP1A2, CYP2C9 and CYP2C19.(111) Desmethoxyyangonin and dihydromethysticin induced the expression of CYP3A23 by approximately seven-fold in an in vitro system; other experimental results suggested that the inductive effect of these kavalactones is additively or synergistically enhanced by the presence of other kavalactones.(112) Other in vitro experiments have demonstrated that kava root extract and the individual kavalactones kavain, dihydrokavain, methysticin, dihydromethysticin and desmethoxyyangonin inhibit the efflux transporter Pglycoprotein.( 113)
The clinical relevance of these findings is not known, although the potential for kava extracts to interact with concurrently administered drugs metabolised mainly by the CYP enzymes mentioned above should be considered. There are conflicting results from in vitro studies regarding the effects of the kavalactone (þ)-kavain on cyclooxygenase activity.(48, 49) One study reported that (þ)-kavain inhibited human platelet aggregation in vitro (see In vitro and animal studies, Other activities], although the clinical relevance of this, if any is not known. At present, there is insufficient evidence to warn against the concurrent use of kava preparations and antiplatelet agents. The kavalactones kavain, methysticin, yangonin and desmethoxyyangonin did not inhibit alcohol dehydrogenase in vitro when applied to the system at concentrations of 1, 10 and 100 mmol/L.(114)
ALCOHOL The effects of concurrent use of kava extract and alcohol have undergone some investigation. In a randomised, doubleblind, controlled trial, 20 healthy participants received kava extract (WS-1490; Laitan) 300 mg daily (equivalent to 210 mg kavalactones), or placebo, for eight days.(115) Alcohol was ingested on days one, four and eight in quantities sufficient to achieve a blood alcohol concentration of 50 mg%; participants underwent a series of tests designed to assess psychomotor performance before and after alcohol consumption. The results indicated that there was no difference in performance between the kava and placebo groups, apart from one test (concentration) in which the kava group was reported to be superior to the placebo group.(115)
A small study involving 40 healthy participants found that the concurrent ingestion of a kava beverage (350mL of aqueous extract of Fijian kava) and alcohol 0.75 g/kg led to a greater reduction in cognitive performance, as assessed by a series of tests, compared with that observed with ingestion of alcohol alone; ingestion of kava alone did not affect cognitive performance.(116) Studies in mice given ethanol (3.5 and 4 g/kg, intraperitoneally) and kava resin 200 or 300 mg/kg orally have demonstrated a prolongation of hypnotic effects.(117)
Pregnancy and lactation There is a lack of information on the use of kava preparations during pregnancy and breastfeeding. Given the lack of data, kava should be avoided during these periods.

CONTRAINDICATIONS AND PRECAUTIONS
Endogenous depression — according to Commission E (Blumenthal et al 2000). Although clinical studies indicate no adverse effects on vigilance, the herb’s CNS effects may slow some individuals’ reaction times, thereby affecting ability to drive a car or operate heavy machinery.
Additionally, it should not be used by people with preexisting liver disease and long-term continuous use should be avoided unless under medical supervision. It should be used with caution in the elderly and in those with Parkinson’s disease.
PREGNANCY USE
Safety is unknown.

CONTRAINDICATIONS
Pregnancy category is 2; Breastfeeding category is 3A.
Kava should not be given to children younger than 12 years of age. This herb should not be used by persons with major depressive disorder or Parkinson’s disease, or by those with hypersensitivity to it.
                                       
SIDE EFFECTS/ADVERSE REACTIONS
Most side effects and adverse reactions occur when high doses are taken for a long period.
CNS: Increased refl exes, drowsiness
EENT: Blurred vision, red eyes
GI: Nausea, vomiting, anorexia, weight loss, hepatic damage

SIDE EFFECTS/ADVERSE REACTIONS—CONT’D
GU: Hematuria
HEMA: Decreased platelets, lymphocytes, bilirubin, protein, and albumin; increased red blood cell volume
INTEG: Hypersensitivity reactions; skin yellowing and scaling (high doses)
RESP: Shortness of breath, pulmonary hypertension

INTERACTIONS
Drug
Antiparkinsonians (carbidopa, levodopa): Antiparkinsonian drugs may increase symptoms of parkinsonism when used with kava; do not use concurrently.
Antipsychotics (chlorpromazine, fl uphenazine, loxapine, mesoridazine, molindone, perphenazine, prochlorperazine, promazine, thioridazine, thiothixene, trifl uoperazine, trifl upromazine): Antipsychotics taken with kava may result in neuroleptic movement disorders.
Barbiturates (amobarbital, aprobarbital, butabarbital, phenobarbital, secobarbital): Barbiturates taken with kava may result in increased sedation.
Benzodiazepines: Increased sedation and coma (theoretical) may result when kava is used with benzodiazepines, including alprazolam; do not use concurrently.
CNS depressants: CNS depressants such as alcohol, benzodiazepines, and barbiturates may cause increased sedation when used with kava; avoid concurrent use.
Cytochrome P450 1A2, 2C9, 2C19, 2D6, 3A4 substrates: Kava signifi cantly decreases these substrates; use cautiously in patients taking these agents.
Food
Increased absorption of kava occurs when it is taken with food.
Lab Test
AST, ALT, LDH, bilirubin: Kava may increase hepatic function tests.

SIDE-EFFECTS, TOXICITY
In randomised, placebo-controlled trials involving different patient groups with anxiety, kava extracts generally have been well tolerated; adverse events reported, and their frequencies, are similar to those reported for placebo. However, clinical trials have the statistical power only to detect common, acute adverse effects.
Spontaneous reports of suspected adverse drug reactions associated with kava preparations have raised concerns over hepatotoxic reactions (see Hepatotoxicity). A systematic review of eight placebo-controlled trials of kava extracts administered at doses equivalent to 55–240 mg kavalactones daily for two days to 24 weeks found that adverse events
reported for both kava and placebo were most commonly gastrointestinal symptoms, tiredness, restlessness, tremor and headache.(69) Three trials included in the review, one of which tested kava extract 100 mg daily (equivalent to 55 mg kavalactones) for 24 weeks,(61) reported that adverse events were not observed in either the kava or placebo groups.
A similar finding was reported by a more recent Cochrane systematic review of monopreparations of kava for the treatment of anxiety (see Clinical studies).(52) This review comprised seven of the eight placebo-controlled trials from the earlier review(69) (the other trial was excluded from the Cochrane review because it tested kava extract in addition to hormone replacement therapy)(61) and five new trials.
Four of the trials, involving 30% of the total number of participants in the trials included in the review, reported that adverse events were not observed during treatment with kava extract.(52) Randomised, double-blind clinical trials comparing kava extracts with certain benzodiazepines and other anxiolytic agents have also found kava to be well tolerated. In a six-week trial involving 172 patients with non-psychotic anxiety, gastrointestinal disturbances occurred in one of 57 participants who received WS-1490 100 mg three times daily (equivalent to 210 mg kavalactones daily), whereas tiredness, vertigo and pruritus occurred in seven of the remaining 115 participants who received oxazepam 5mg or bromazepam 3 mg, both taken three times daily.(59) In an eight-week trial involving 129 outpatients with generalised anxiety disorder, 14 of 43 (33%) participants who received the kava extract LI-150 400 mg (standardised to 30% kavalactones = 120 mg) each morning experienced adverse events, compared with 10 (24%) and 11 (26%) participants who received buspirone 5mg twice daily and opipramol 50 mg twice daily, respectively.(60) A total of 27 adverse events was reported in the kava group, compared with 16 and 14 for buspirone and opipramol, respectively. Adverse events reported for kava included upper respiratory tract infections, gastrointestinal disorders, weight changes, skin reactions and tachycardia, all of which were also reported for buspirone and/or opipramol. These clinical trials and systematic reviews, however, provide only limited evidence to support the safety of kava extracts since they involved only small numbers of participants, involved different patient groups, tested different doses of kava extract (typically equivalent to 60–240 mg kavalactones daily), and most were of relatively short duration, usually around four weeks.
Further, most trials investigated preparations of the kava extract WS-1490, and other standardised extracts of kava and kava preparations supplied by herbal medicine practitioners have undergone considerably less assessment. Two post-marketing surveillance studies published in the early to mid-1990s involving patients treated in one study with WS-1490 150 mg daily (equivalent to 105 mg kavalactones; n = 4049) and in the other with Antares 120 (equivalent to 120 mg kavalactones; n = 3029) reported that the frequencies of adverse events were 1.5% and 2.3%, respectively.(69) In both studies, adverse events commonly reported were mild gastrointestinal disorders and allergic reactions and, in the latter study, headaches and vertigo, which stopped when kava treatment was discontinued. A rather higher frequency of adverse events was reported during a postmarketing surveillance study carried out in Brazil.(70) Among 850 participants with anxiety who received WS-1490 100 mg three times daily (equivalent to 210 mg kavalactones daily), 16.7% reported adverse events, most commonly fatigue/tiredness, nausea, confusion and gastrointestinal upset.
The World Health Organization's Uppsala Monitoring Centre (WHO-UMC; Collaborating Centre for International Drug Monitoring) receives summary reports of suspected adverse drug reactions from national pharmacovigilance centres of over 70 countries worldwide. To the end of the year 2005, the WHOUMC's Vigisearch database contained a total of 91 reports, describing a total of 189 adverse reactions, for products reported to contain P. methysticum only as the active ingredient (see Table 1).(71) This number may include some of the case reports described in the sections below. Reports originated from nine different countries. The total number of reactions included reports describing a total of 55 reactions associated with liver and biliary system disorders (see Table 1), including three cases of hepatic failure and two of hepatic coma. (These data were obtained from the Vigisearch database held by the WHO Collaborating Centre for International Drug Monitoring, Uppsala, Sweden. The information is not homogeneous at least with respect to origin or likelihood that the pharmaceutical product caused the adverse reaction. Any information included in this report does not represent the opinion of the World Health Organization.)

Table 1 Summary of spontaneous reports (n = 91) of suspected adverse drug reactions associated with single-ingredient Piper methysticum preparations held in the Vigisearch database of the World Health Organization’s Uppsala Monitoring Centre for the period up to end of 2005(71, a, b)

a Specific reactions described where n = 3 or more
bCaveat statement. These data were obtained from the Vigisearch database held by the WHO Collaborating Centre for International Drug Monitoring, Uppsala, Sweden. The information is not homogeneous at least with respect to origin or likelihood that the pharmaceutical product caused the adverse reaction. Any information included in this report does not represent the opinion of the World Health Organization

Hepatotoxicity
Clinical data None of the clinical trials and post-marketing surveillance studies described above reported hepatotoxicity as an observed adverse event, although not all studies carried out liver function tests on participants. Seven of the 11 trials included in the Cochrane review of the kava extract WS-1490 for the treatment of anxiety (see Clinical studies) did involve monitoring participants' liver function (e.g. serum aspartate transaminase and alanine  transaminase concentrations) and did not report any abnormalities or clinically significant changes in values obtained.(52)
Over the years 2000 and 2001, a safety concern arose regarding cases of hepatoxicity reported in association with the use of kava extracts. The signal first emerged in Switzerland, following a cluster of spontaneous reports to the medicines' regulatory authority, and was strengthened a year or so later following further spontaneous reports from Switzerland and Germany. By July 2002, a total of 68 reports of liver toxicity associated with use of kava had been received by regulatory authorities in Canada, France, the UK and USA, as well as in Switzerland and Germany,(72) and by the end of January 2005, 79 cases of liver damage associated with use of kava had been identified worldwide.(73) The severity of the liver damage described in the reports
varied from abnormal liver function test results to irreversible liver failure and death; at least six patients received liver transplants, one of whom, as well as at least two other individuals, subsequently died. Cases reported in the UK included two of raised liver function test values in men aged 40 and 48 who had taken unspecified kava preparations for three months and eight years, respectively.(74) Both stopped taking kava and their liver function test values normalised. Another UK case related to a woman (age not stated) who had taken kava 150 mg three times daily for two months, in addition to fluoxetine, and who experienced jaundice and raised liver function test values and was hospitalised for seven weeks.
In the UK, evidence relating to the hepatotoxicity associated with kava was reviewed in 2005 in a public consultation and later that year by the Expert Working Group set up to consider the evidence. The Expert Working Group's report was published in July 2006 and concluded that there was insufficient new evidence to support a change in the regulatory position, hence the inclusion of kava in unlicensed medicines in the UK remains prohibited.(75) The report also identified several new questions and issues that may be important with respect to hepatotoxicity of kava, including the possibility that other alkaloid and/or amide constituents may be present, and their possible contribution to hepatotoxicity, and the need for a systematic evaluation of all marketed kava products and their source material, and of the variation in the phytochemistry of kava cultivars.(75) Earlier reviews of German data relating to hepatotoxicity associated with kava have produced conflicting opinions on causality. One review emphasised that there was no dose–response relationship for kava-associated hepatotoxicity, and that crude estimates of incidence based on primary care data suggest that any risk of hepatotoxicity is similar to that of benzodiazepines.(76)
However, this conclusion is questionable since estimates of this nature can be inaccurate and misleading. By contrast, a review of seven previously published and 29 unpublished case reports of kava-associated hepatotoxicity concluded that these data clearly showed the potential for severe, unpredictable kava-related hepatotoxicity.(77) Cases included nine individuals who developed fulminant hepatic failure, of whom six underwent successful liver transplantation (one only after retransplantation), two died after transplantation due to postoperative infectious complications, and one who was too old to undergo transplantation also died. All other cases, which comprised mostly cholestatic or necrotising hepatitis, underwent full recovery after withdrawal of kava treatment. Among these 36 reports, the relationship between kava ingestion and hepatotoxicity was considered 'certain' in three cases and 'probable' in 21.(77) Most individuals were concurrently using other medication and several were regular consumers of alcohol.
A case report from Australia describes a 56-year-old woman who developed fatigue, nausea and jaundice after taking a preparation named 'Kava 1800 Plus' one tablet three times daily for around 10 weeks.(78) Each tablet was stated to contain kavalactones 60 mg, Passiflora incarnata 50 mg and Scutellaria lateriflora 100 mg, although the latter ingredient was not identified in the product, so the precise composition of the product is unknown. She presented two weeks after first experiencing these symptoms and was hospitalised. Five days later, a biopsy revealed non-specific severe acute hepatitis with pan-acinar necrosis and collapse of hepatic lobules. She underwent liver transplantation on day 17 after admission, but the procedure was complicated and she died from progressive blood loss and circulatory failure. Subsequent examinations confirmed massive hepatic necrosis. Until the year 2003, it was thought that kava-associated hepatoxicity occurred only with ethanolic and acetonic kava extracts. However, recent reports described hepatoxic effects associated with consumption of traditional aqueous extracts of kava root.
Two cases in New Caledonia involved women aged in their fifties who started consuming kava prepared in the traditional manner as an aqueous preparation.(79) Both women developed signs and symptoms of hepatotoxicity, including icterus and raised liver function test values four or five weeks after starting kava. Neither patient consumed alcohol. Kava consumption was stopped; both patients recovered and liver function test values normalised over the following three months.(79) Subsequently, in a cross-sectional study, blood samples were collected from 27 individuals who were chronic kava drinkers, recruited from kava bars in New Caledonia. Participants had been consuming kava regularly for at least five years and had a mean intake equivalent to around 32 g kavalactones weekly or 70 mg/kg daily; 12 participants also consumed alcohol.(79) Transaminase concentrations were more than 1.5 times the upper limit of normal in three participants, and 23 participants had increases in concentration of GGT (which is not necessarily a specific marker of liver injury); alkaline phosphatase and bilirubin concentrations were within normal ranges for all subjects. Another crosssectional study, involving indigenous people from an Arnhem Land community (Northern Territory, Australia), included 98 participants, of whom 36 had never used kava.(80) Of the 62 kava users, 14 had ingested kava within the previous 24 hours, and 10 and 15 had used kava within the previous one or two weeks, or one or two months, respectively. Investigation of liver function test values indicated that changes appeared to be reversible and started to normalise after one or two weeks of kava abstinence.(80)
Inhibition of certain cytochrome P450 (CYP) drug metabolising enzymes has been shown in vitro and in vivo for kava extracts and individual kavalactones (see Contra-indications, Warnings). The relevance of this for the hepatotoxic effects described for kava is not known; further work is needed to determine whether the inhibition of CYP enzymes by kava can lead to raised plasma concentrations of concurrently ingested drugs with hepatotoxic effects.

Skin Reactions
An ichthyosiform (scaly, non-inflammatory), usually yellowish or whitish, skin condition termed kava dermopathy has been documented among kava users in Polynesia, Micronesia and Melanesia where powdered kava rhizome is prepared as a drink with cold water or coconut milk.(81) The condition is reversible on stopping kava. Initially, it was thought that the condition was related to niacin deficiency, but this hypothesis was rejected following a small randomised, placebo-controlled trial of nicotinamide 100 mg daily for three weeks which showed no difference between groups.(82)
Measures of health among 39 users of kava (prepared as a cold water infusion of powdered kava rhizome) were compared with those of 34 age-matched non-users of kava in an Aboriginal community in the Northern Territory in Australia.(83) Most (n = 35) were 'heavy' or 'very heavy' users of kava (310 g or more per week). It was reported that kava users were more likely to complain of poor health, and to have a scaly skin rash. However, the study had several methodological limitations (e.g. no correction for multiple statistical tests) and potential biases.
Several cases of allergic skin reactions have been reported in association with kava use. One case described a man who presented with oedema and severe non-pruritic erythema involving his upper body, head and neck, the morning after drinking several cups of 'kava tea'.(84) It was reported that the man had previously had a similar reaction to kava tea three months earlier whilst overseas and for which he was hospitalised and treated with intravenous corticosteroids. A case of systemic contact-type dermatitis following several weeks' use of kava extract (Antares), chlorprothixene (an antipsychotic agent with properties similar to those of chlorpromazine) and diazepam has been described.(85)
Two further cases described a 70-year-old man and a 52-year-old woman who experienced skin eruptions (erythematous plaques and/or papules) in sebaceous gland-rich areas after using kava extract (no further details provided) for two to three weeks.(86) Both patients were reported to display reactions to kava in diagnostic allergy or skin patch tests. Generalised erythema and papules with severe itching were described in a 36-year-old woman who had taken kava extract (Antares) 120 mg daily for three weeks.(87) The rash, but not the itching, responded to short-term treatment with systemic corticosteroids, and six weeks later, patch test results for Antares were positive one day after application.

Central Nervous System Effects
Four cases of involuntary movements and dyskinesia associated with use of kava extracts have been reported, although causality has not been established; it has been stated that these symptoms suggest that constituents of kava may have antagonistic effects on central dopaminergic pathways.(88) In three cases, involuntary movements involving the neck, head and/or trunk, and involuntary oral and lingual dyskinesia began within a few minutes to 4 hours after ingestion of kava extracts (Laitan 100 mg or Kavasporal forte 150 mg) for anxiety. One of these cases involved a 28-year-old man who had previously experienced three episodes of acute dystonic reactions following exposure to promethazine and fluspirilene, although he denied having used these medicines in relation to the current episode. The fourth case report described a 76-year-old woman being treated with levodopa 500 mg and benserazide 125 mg for Parkinson's disease and who experienced an increase in the duration and frequency of her 'off' periods 10 days after starting Kavasporal forte 150 mg twice daily, prescribed by her physician for tension. (The 'on–off' phenomenon – sudden swings in mobility–immobility – occurs with long-term use of levodopa.) In all four cases, symptoms resolved on stopping kava or following treatment with biperiden administered intravenously.
Two other cases describe neurological symptoms following excessive use of traditional preparations of kava, i.e. as a beverage. A 27-year-old Aboriginal Australian man experienced generalised severe choreoathetosis (characterised by chorea and athetosis, a form of dyskinesia) without impairment of consciousness on three occasions after drinking large amounts of kava (precise quantity not specified).(89) Routine investigations were normal, apart from raised liver function test values (serum alkaline phosphatase 162 IU/L, normal range 35–135 IU/L; gamma-glutamyltransferase 426 IU/L, normal range <60 IU/L). His symptoms responded to treatment with diazepam administered intravenously. Disorientation was reported in a 34-year-old Tongan man, a heavy user of kava (40 bowls daily for 14 years), who had ingested further excessive amounts of kava over the previous 12 hours.(90) The man was treated in hospital with Plasmalyte intravenously and intramuscular thiamine and five hours after admission his symptoms had resolved. In a controlled study, individuals intoxicated following kava consumption (205 g powder) experienced ataxia, tremors, sedation, blepharospasm and reduced accuracy performing a visual search task, when compared with control subjects who had not ingested kava. These results suggest that kava intoxication results in specific abnormalities of movement co-ordination and visual attention, but normal performance of complex cognitive functions.(91)

Effects On Mental Performance
The effects of kava extracts and the synthetic kavalactone (_)- kavain on mental performance have been explored in studies involving healthy volunteers. Preliminary studies involving small numbers of volunteers have suggested that kava extract (WS-1490 200 mg three times daily for five days) did not appear to impair memory as assessed by certain tests (e.g. word recognition) carried out under laboratory conditions.(92) In another series of tests, designed to assess mental alertness, volunteers received kava extract, Antares 120 (standardised to 120 mg kavalactones per tablet), one tablet daily, diazepam 10 mg daily, or placebo.(93) It was reported that the experiments provided evidence that kava did
not cause drowsiness or lack of concentration, for example, reaction time was reduced in placebo recipients, but not kava recipients. Other research involving volunteers found that a single dose of kava extract 600 mg (LI-158; drug–extract ratio, 12.5 : 1) led to a 'moderate' increase in tiredness, compared with placebo, and as assessed using visual analogue scale scores, although statistical analysis was not reported.(94) Confirmation of these findings is required. In a battery of psychometric and other tests following administration of a range of single doses of the synthetic kavalactone (_)-kavain (200, 400 and 600 mg) and clobazam 30 mg to healthy volunteers, (_)-kavain appeared to have a sedative effect which was stated to be different to that observed with clobazam.(95) Compared with placebo, (_)-kavain, but not clobazam, improved intellectual performance, attention, concentration and reaction time.

Other Reactions
There are isolated reports of myopathy and myoglobinuria associated with the use of kava preparations, although causality in these cases has not been established. One report described dermatomyositis associated with use of kava for anxiety by a 47-year-old woman.(96) The woman, who had also been taking valproic acid for 18 months and sertraline occasionally over two years for bipolar disorder, developed a rash involving her back, neck and face, as well as muscle weakness, two weeks after taking kava (dosage not specified). She improved initially following treatment with methylprednisolone, but then developed a fever which prompted her to attend a hospital emergency department. Investigations revealed a raised serum creatine kinase concentration (8654 U/L, normal values stated as 24–170 U/L) and myopathic patterns in various muscles, and biopsy samples showed changes indicative of dermatomyositis.
The woman was treated initially with parenteral prednisone, after which her creatine kinase concentration returned to normal, and also received methotrexate for five months and hydroxychloroquine. Prednisone treatment was reduced over the following year, and at one year of follow-up the woman remained symptom-free.
Another isolated report describes a 29-year-old man who experienced severe muscle pain and passed dark urine one morning a few hours after having taken a herbal product said to contain kava 100 mg, Ginkgo biloba extract 200 mg and guarana (which contains methylxanthines) 500 mg (daily dosage was not stated), for the first time.(97) The man was admitted to an intensive care unit and was found to have highly elevated serum creatine kinase (100 500 IU/L, normal range given as 0–195 IU/L) and myoglobin (10 000 ng/mL, normal range stated as 0–90 ng/ mL) concentrations, but no renal complications. Investigations excluded metabolic myopathy as a possible cause; his signs and symptoms subsided over six weeks.
Disturbances of visual function have been reported following a study involving a 30-year-old kava-naïve male volunteer who ingested 600 mL of aqueous extract of pulverised kava 'root' (rhizome).(98) Measurements involving the man's right eye only indicated reductions in near point of accommodation and convergence, an increase in pupil diameter, and disturbance of oculomotor balance, but no effects on visual or stereoacuity or ocular refractive error. The experiment was not carried out according to a double-blind, controlled design and, therefore, the findings require further investigation.
A case of hypokalaemic renal tubular acidosis due to Sjögren's syndrome (a symptom complex of unknown aetiology, marked by keratoconjunctivitis sicca, xerostamia, with or without lachrymal and salivary gland enlargement, respectively, and presence of connective tissue disease, usually rheumatoid arthritis, but sometimes systemic lupus erythematosus, scleroderma or polymyositis) has been reported in a 36-year-old woman.(99) She was stated to have begun taking kava, echinacea and St John's wort two weeks before becoming ill, but the report does not provide any further details of the echinacea species contained in the product(s), nor of the types of preparations, formulations, dosages and routes of administration of any of the herbal medicines listed.
The woman was hospitalised with severe generalised muscle weakness and tests revealed she had a serum potassium ion concentration of 1.3 mEq/L. She was given electrolyte replacement for four days after which the muscle weakness resolved, and was started on hydroxychloroquine 200 mg daily for 'probable' Sjögren's syndrome. The authors suggested that ingestion of echinacea may have aggravated an autoimmune disorder (see Echinacea, Side-effects, Toxicity) although causality has not been established.(99)
No clear evidence of an association between kava consumption and ischaemic heart disease (IHD) was found in a case–control study (using up to four randomly selected control subjects) involving 83 individuals from Aboriginal communities in Arnhem Land (Northern Territory, Australia) who were diagnosed with IHD for the first time during 1992–1997.(100) In a similar case– control study involving 115 individuals (and 415 control subjects) with pneumonia, no association between kava use and pneumonia was found.(101)

Toxicology
Incubation of the kava alkaloid pipermethystine, which occurs mostly in kava leaves and stem peelings, with human hepatoma cells resulted in a 90% loss in cell viability within 24 hours when applied at a concentration of 100 mmol/L and 65% cell death at a concentration of 50 mmol/L.(102) Further experiments indicated that pipermethystine causes cell death in part by disrupting mitochondrial function. It has been suggested that pipermethysticine could be involved in hepatoxicity associated with kava root extracts,(102) although since hepatotoxicity has been associated both with authorised commerical products (therefore made according to the principles of good manufacturing practice) and traditional aqueous extracts (which might be more easily contaminated with leaves and stem peelings) this suggestion requires further examination.
A study involving small numbers of rats administered an aqueous (water) extract of kava 'root' equivalent to kavalactones 200 or 500 mg/kg/day for 2–4 weeks found that serum concentrations of the enzymes alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase and lactate dehydrogenase were not elevated following kava administration, compared with control.(103) The clinical relevance of these findings is not known.
In other toxicological studies, LD50 values for a standardised kava extract containing 70% kavalactones have been reported as 370 mg/kg and 16 g/kg for intraperitoneal and oral administration, respectively, in rats, and 380 mg/kg and 1.8 g/kg for intraperitoneal and oral administration, respectively, in mice.(G50)
An ethanolic extract of kava root was reported to be cytotoxic (EC50 approximately 50 mmol/L) in an in vitro system assessing viability of human hepatocytes. The kavalactones methysticin, desmethoxyyangonin and yangonin also displayed cytotoxicity in this system, with methysticin having the greatest cytotoxic activity.(104) In vitro cytotoxicity of kavalactones has also been described in a human lymphoblastoid cell line; the cytotoxic effect appears to be due to the parent compound and not as a result of activation of kava constituents to toxic metabolites.(105)


CLIENT CONSIDERATIONS
ASSESS
·         Assess the reason the client is using kava.
·         Assess for hypersensitivity reactions. If present, discontinue the use of kava and administer an antihistamine or other appropriate therapy.
·         Assess for use of other central nervous system depressants, including alcohol, barbiturates, benzodiazepines, antianxiety medications, and sedatives/hypnotics (see Interactions).
ADMINISTER
·         Instruct the client to store kava products in a cool, dry place, away from heat and moisture.
·         Instruct the client not to use kava for longer than 3 months unless under the direction of an herbalist. This herb may be habit forming.
·         Inform the client that kava absorption is increased when kava is taken with food.
TEACH CLIENT/FAMILY
·         Inform the client that pregnancy category is 2 and breastfeeding category is 3A.
·         Caution the client not to give kava to children younger than 12 years of age.
·         Inform the client that excessive doses may result in daytime drowsiness. Advise the client not to operate heavy machinery or engage in hazardous activities if drowsiness occurs.
·         Caution the client not to use kava with other central nervous system depressants (see Interactions).


PRACTICE POINTS/PATIENT COUNSELLING
·         Kava kava is a scientifically proven treatment for the symptoms of anxiety and stress states. Its anxiety-reducing effects are similar to those of 15 mg oxazepam or 9 mg bromazepam; yet, physical tolerance and reduced vigilance have not been observed.
·         It also reduces symptoms of anxiety related to menopause when used together with HRT, and reduces withdrawal symptoms associated with benzodiazepine discontinuation.
·         It has anxiolytic, sedative, antispasmodic, analgesic and local anaesthetic activities.
·         Although the herb is considered to have a low incidence of adverse effects, long-term use should be carefully supervised because of the possibility of developing adverse reactions.
·         Rare hepatotoxic effects have been reported for methanolic and ethanolic extracts, whereas traditional aqueous extracts are considerably safer. The mechanism responsible remains elusive.

PATIENTS’ FAQs

What will this herb do for me?
Kava kava is an effective herbal relaxant that reduces symptoms of anxiety and restlessness. It is also used to relieve anxiety in menopause, insomnia and symptoms of benzodiazepine withdrawal.
When will it start to work?
Anxiety-relieving effects are usually seen within the first few weeks of use.
Are there any safety issues?
Taking high doses long term has been associated with a number of side effects and should be avoided.

CLINICAL NOTE

COMMERCIAL KAVA KAVA PRODUCTS AND LINKS TO HEPATOTOXICITY
Conflicting reports abound. On 15 August 2002, the Therapeutic Goods Administration (TGA) initiated a voluntary recall of all products containing kava kava. The response was undertaken due to incoming details from European countries of case reports of hepatotoxicity apparently associated with the use of commercial kava kava products.
The decision to remove kava kava from the market has been viewed as controversial and questioned by many people. Toxicological and clinical studies have shown that kava kava extracts are virtually devoid of toxic effects and, when assessed primarily by the British regulatory authority (MCA) and a German research group, a critical analysis of the suspected cases in Germany reveals that a very probable causal relationship could be established in only one patient (Teschke et al 2003). It is suspected that a rare, immunologically mediated, idiosyncratic mechanism may be responsible (Schulze et al 2003) and the extraction process used to produce kava kava products also had an influence. It now appears that the aqueous method results in extraction of glutathione, in addition to kava lactones, an important factor for protecting the liver from potential damage, whereas the acetone extraction method does not (Whitton et al 2003). As a result, kava kava products made with the acetone extraction process are more toxic than those produced via aqueous extraction methods. This is an important distinction to make, as most European products were made using acetone extraction, whereas Australian products were chiefly made via aqueous extraction.
Interestingly, fulminant hepatic failure has not been documented with traditional use either in Pacific countries or in the Northern Territory, where Aboriginal kava kava drinkers consume kava lactones in doses estimated to be 10–50-fold the recommended levels (Currie & Clough 2003). Several reports published in 2003 have found no evidence of aqueous kava kava extracts inducing irreversible liver toxicity in vivo (Singh & Devkota 2003) or in humans (Clough et al 2003). One study involving long-term users of aqueous kava kava extracts found that although changes to liver function could occur at moderate levels of consumption, they are reversible and begin to return to baseline after 1–2 weeks’ abstinence from kava kava. A recent animal study testing large concentrations of kava lactones (7.3 or 73 mg/kg of kava lactones/day) over 3 months and 6 months found no signs of toxicity. In addition, no behavioural or physiological changes were observed on discontinuation of kava lactone feeding after 3 months (Sorrentino et al 2006).
Australia was not alone, and other countries also issued health advisory cautions or banned kava kava-containing products from sale. Although these actions effectively removed kava kava products from the market, the traditional kava kava beverage continued to be consumed in the Pacific Islands and the kava kava-producing countries of the Pacific found the controversy surprising, given the long history of apparent safe use in the Pacific. The impact of European and UK withdrawal of kava kava was devastating to the South Pacific economies. In January 2003, the Kava Evaluation Group was established in Australia to review the accumulating safety data and by August that same year the Complementary Medicine Evaluation Committee recommended to the TGA that certain forms of kava kava could be considered safe. The TGA accepted these recommendations and amended the regulations accordingly. Currently in Australia, there is a maximum limit of 125 mg kava lactones allowable per tablet or capsule, 3 g of dried rhizome per tea bag and all products containing kava kava must not provide more than 250 mg kava lactones in the recommended daily dose. The World Health Organization published a report in May 2007 entitled Assessment of the risk of hepatotoxicity with kava products (WHO 2007).
It evaluated data from 93 case reports of which 8 were determined to have a close association between the use of kava kava and liver dysfunction; 53 cases were classified as having a possible relationship, but they could not be fully assessed due to insufficient data or other potential causes of liver damage; 5 cases had a positive rechallenge. Most of the other case reports could not be evaluated due to lack of information. It concluded that there is ‘significant concern’ for a cause and effect relationship between kava kava products and hepatotoxity, especially for organic extracts. Other risk factors appear to include heavy alcohol intake, preexisting liver disease, genetic polymorphisms of cytochrome P450 enzymes, excessive dosage and co-medication with other potentially hepatotoxic drugs and potentially interacting drugs.

PREPARATIONS

PROPRIETARY SINGLE-INGREDIENT PREPARATIONS
Brazil: Ansiopax; Calmonex; Farmakava; Kavakan; Kavalac; Kavasedon; Laitan; Natuzilium. Czech Republic: Antares; Kavasedon; Leikan. Venezuela: Kavasedon.
PROPRIETARY MULTI-INGREDIENT PREPARATIONS
USA: Calming Aid.

EXTRACTS
Increase GABA in the synaptic cleft by increasing GABA secretion and inhibiting its reuptake (SHT). LD50 dihydrokavain = 920 mg/kg orl mouse (MAB), LD50 dihydromethysticin = 1050 mg/kg orl mouse (MAB), LD50 StX (70% kavalactones) = 16,000 mg/kg orl rat, 1800 mg/kg orl mouse, 370 mg/kg ipr rat, 380 mg/kg ipr mouse (MAB). This indicates that the mix is safer than the individual lactone, at least orally in rats and mice (MAB). Kava slows hyperactivity in mice, but not as much as antipsychotic drugs. When chewed, the root produces numbness in the mouth similar to what one would experience with cocaine and longer-lasting than what one would experience with benzocaines (APA). In a traditional Hawaiian remedy, leaves were chewed and given to anxious or restless children for its calming effect, and to induce sleep. And for the old reprobates “kava tends to lower one’s interest in sexual activities.” ( = ) lactones are 10 times more anticonvulsant than mephenesin against strychnine; the mixture of lactones was synergistic; the potency of the mix was equal to that of pure dihydromethysticin; synergy more pronounced with oral than ivn administration; lactones better absorbed in mix than as isolated silver bullets (MAB). Lactones = cocaine and procaine as analgesic and anesthetic; dihydromethysticin better than aspirin but inferior to morphine as analgesic (MAB).





REFERENCE


Barnes, J., Anderson, L. A., and Phillipson, J. D. 2007. Herbal Medicines Third Edition. Pharmaceutical Press. Auckland and London.

Braun, L and Cohen, M. 2010. Hebs and Natural Supplements An Evidence Based Guide 3R D Edition. Elsevier Australia. Australia.

Duke, J. A. with Mary Jo Bogenschutz-Godwin, Judi duCellier, Peggy-Ann K. Duke. 2002. Handbook of Medicinal Herbs 2nd Ed. CRC Press LLC. USA.

Gruenwald, J., Brendler, T., Jaenicke, Ch. 2000.  PDR for Herbal Medicines.  Medical Economics Company, Inc. at Montvale, NJ 07645-1742. USA

Kraft, K and Hobbs, C. 2004 . Pocket Guide to Herbal Medicine. Thieme. Stuttgart New York.

Linda S-Roth. 2010. Mosby’s Handbook Of Herbs & Natural Supplements, Fourth Edition. Mosby Elsevier. USA






No comments:

Post a Comment