Mistletoe tree/ leaf for treatment of cardiovascular diseased
Adverse Drug Events Anticoagulation Cardiology Clinical Pharmacy and Pharmacology Complementary and Alternative Medicine Heart Failure Ischemic Heart Disease Pain Medicine Rhythm Disorders S
Herbal Medicine for the Treatment of Cardiovascular Disease
Clinical Considerations
Nick H. Mashour, MD; George I. Lin, MD; William H. Frishman, MD
Author Affiliations Article Information
Arch Intern Med. 1998;158(20):2225-2234. doi:10.1001/archinte.158.20.2225
Abstract
Herbs have been used as medical treatments since the beginning of civilization and some derivatives (eg, aspirin, reserpine, and digitalis) have become mainstays of human pharmacotherapy. For cardiovascular diseases, herbal treatments have been used in patients with congestive heart failure, systolic hypertension, angina pectoris, atherosclerosis, cerebral insufficiency, venous insufficiency, and arrhythmia. However, many herbal remedies used today have not undergone careful scientific assessment, and some have the potential to cause serious toxic effects and major drug-to-drug interactions. With the high prevalence of herbal use in the United States today, clinicians must inquire about such health practices for cardiac disease and be informed about the potential for benefit and harm. Continuing research is necessary to elucidate the pharmacological activities of the many herbal remedies now being used to treat cardiovascular diseases.
Since the beginning of human civilization, herbs have been an integral part of society, valued for both their culinary and medicinal properties. Herbal medicine has made many contributions to commercial drug preparations manufactured today including ephedrine from Ephedra sinica (ma-huang), digitoxin from Digitalis purpurea (foxglove), salicin (the source of aspirin) from Salix alba (willow bark), and reserpine from Rauwolfia serpentina (snakeroot), to name just a few. A naturally occurring β-adrenergic blocking agent with partial agonism has been identified in an herbal remedy.1 The recent discovery of the antineoplastic drug paclitaxel from Taxus brevifolia (pacific yew tree) stresses the role of plants as a continuing resource for modern medicine.
However, with the development of patent medicines in the early part of the 20th century, herbal medicine has been losing ground to new synthetic medicines touted by scientists and physicians to be more effective and reliable. Nevertheless, about 3% of English-speaking adults in the United States still report having used herbal remedies in the preceding year.2 This figure is probably much higher for non–English-speaking Americans. Despite this heavy use of herbal medicines in the United States, health practitioners often fail to ask about their use when taking clinical histories. It is imperative that physicians become more aware of the wide array of herbal medicines available, as well as learning more about their beneficial and adverse effects.3
Part of the problem for both consumers and physicians has been the paucity of scientific data on herbal medicines used in the United States.4 As a result, those who wish to obtain factual information regarding the therapeutic use or potential harm of herbal remedies would have to obtain it from books and pamphlets, most of which base their information on traditional reputation rather than relying on existing scientific research. One may wonder why the herbal industry never chose to simply prove its products safe and effective. The answer is primarily economical. With the slim chance of patent protection for the many herbs that have been in use for centuries, pharmaceutical companies have not provided financial support for research on the merits of herbal medicine.5 At the same time, the National Institutes of Health have only been able to offer limited funding for this purpose.
This review examines herbal medicines that affect the cardiovascular system both in terms of efficacy and safety as gleaned from the scientific literature that is available. These herbs are categorized under the primary diseases they treat. However, most herbal medicines have multiple cardiovascular effects that frequently overlap. The purpose of this organization is to simplify, not to pigeonhole herbs under specific diseases. In general, the dilution of active components in herbal medicines results in fewer adverse and toxic effects in comparison with the concentration of active components in the allopathic medicines. However, these adverse effects and drug interactions should not be overlooked; cardiovascular disease is a serious health hazard and no herbal remedy regimen should be initiated without careful consideration of its potential impact (Table 1).
Congestive heart failure
A number of herbs contain potent cardioactive glycosides, which have positive inotropic actions on the heart. The drugs digitoxin, derived from either D purpurea (foxglove) or Digitalis lanata, and digoxin, derived from D lanata alone, have been used in the treatment of congestive heart failure for many decades. Cardiac glycosides have a low therapeutic index, and the dose must be adjusted to the needs of each patient. The only way to control dosage is to use standardized powdered digitalis, digitoxin, or digoxin. When 12 different strains of D lanata plants were cultured and examined, their total cardenolide yield ranged from 30 to almost 1000 nmol/1 g.6 As is evident, treating congestive heart failure with nonstandardized herbal drugs would be dangerous and foolhardy.
Some common plant sources of cardiac glycosides include D purpurea (foxglove, already mentioned), Adonis microcarpa and Adonis vernalis (adonis), Apocynum cannabinum (black Indian hemp), Asclepiascurassavica (redheaded cotton bush), Asclepias friticosa (balloon cotton), Calotropis precera (king's crown), Carissa spectabilis (wintersweet), Cerebra manghas (sea mango), Cheiranthus cheiri (wallflower), Convallaria majalis (lily of the valley, convallaria), Cryptostegia grandiflora (rubber vine), Helleborus niger (black hellebore), Helleborus viridus, Nerium oleander (oleander), Plumeria rubra (frangipani), Selenicerus grandiflorus (cactus grandiflorus), Strophanthus hispidus and Strophanthus kombe (strophanus), Thevetia peruviana (yellow oleander), and Urginea maritima (squill).5,7-15 Even the venom glands of the animal Bufo marinus (cane toad) contain cardiac glycosides.8 Recently, the digitalislike steroid in the venom of the B marinus toad was identified as a previously described steroid, marinobufagenin. Marinobufagenin demonstrated high digoxinlike immunoreactivity and was antagonized with an antidigoxin antibody.16
Accidental poisonings and even suicide attempts with ingestion of cardiac glycosides are abundant in the medical literature.17-21 Some herbal remedies (eg, Siberian ginseng) can elevate synthetic digoxin drug levels and cause toxic effects.22 In the United States, there are about 15,000 intoxications due to accidental or intentional ingestion of poisonous plants annually.23 In 1993, 2388 toxic exposures in the United States were reported to be due to plant glycosides. Of these, the largest percentage were attributed to oleander (ie, 25%).24 In the case of oleander, all plant tissues, including the seeds, roots, stems, leaves, berries, and blossoms, are considered extremely toxic.19 In fact, death in humans has been reported following ingestion of as little as 1 oleander leaf.25 The clinical manifestations of oleander intoxication, as well as other natural glycosides, is virtually identical to digoxin overdose. Morbidity and mortality are mainly related to cardiotoxic adverse effects that usually include life-threatening ventricular tachyarrhythmias, bradycardia, and heart block. The diagnosis should rely on the clinical presentation of unexplained hyperkalemia, and cardiac, neurologic, and gastrointestinal symptoms.19
The diagnosis can be further supported by the detection of the substance digoxin in a radioimmunoassay for digoxin. However, the extent of cross-reactivity between the cardiac glycosides from herbal sources and antibodies used in the radioimmunoassays has not been clearly defined.26 For this reason, digoxin assays may serve to confirm the suspected diagnosis but not to quantify the severity. Once the diagnosis has been established, the use of digoxin-specific Fab antibody fragments may be helpful in the treatment of severe intoxication. Other modalities, such as dialysis, cannot be easily facilitated because, like digoxin, natural glycosides are distributed extensively into peripheral tissues.
Hypertension
The root of R serpentina (snakeroot), the natural source of the alkaloid reserpine, has been a Hindu Ayurvedic remedy since ancient times. In 1931, Indian literature first described the use of R serpentina root for the treatment of hypertension and psychoses; however, the use of Rauwolfia alkaloids in Western medicine did not begin until the mid1940s.27 Both standardized whole root preparations of R serpentina and its reserpine alkaloid are officially monographed in the United States Pharmacopeia.28 A powdered whole root of 200 to 300 mg orally is equivalent to 0.5 mg of reserpine.29
Reserpine was one of the first drugs used on a large scale to treat systemic hypertension. It acts by irreversibly blocking the uptake of biogenic amines (norepinephrine, dopamine, and serotonin) in the storage vesicles of central and peripheral adrenergic neurons, thus leaving the catecholamines to be destroyed by the intraneuronal monoamine oxidase in the cytoplasm. The depletion of catecholamines accounts for reserpine's sympatholytic and antihypertensive actions. Reserpine's effects are long lasting, since recovery of sympathetic function requires synthesis of new storage vesicles, which takes days to weeks. Reserpine lowers blood pressure by decreasing cardiac output, peripheral vascular resistance, heart rate, and renin secretion. With the introduction of other antihypertensive drugs with fewer central nervous system adverse effects, the use of reserpine has diminished. The daily oral dose of reserpine should be 0.25 mg or less, and as little as 0.05 mg if given with a diuretic. Using the whole root, the usual adult dose is 50 to 200 mg/d administered once daily or in 2 divided doses.27-29
Rauwolfia alkaloids are contraindicated for use in patients with previously demonstrated hypersensitivity to these substances, in patients with a history of mental depression (especially with suicidal tendencies), in patients with active peptic ulcer disease or ulcerative colitis, and in patients receiving electroconvulsive therapy. The most common adverse effects are sedation and inability to concentrate and perform complex tasks. Reserpine may cause mental depression, sometimes resulting in suicide, and its use must be discontinued at the first sign of depression. Reserpine's sympatholytic effect and its enhancement of parasympathetic actions account for its well-described adverse effects: nasal congestion, increased gastric secretion, and mild diarrhea.27-30
Stephania tetrandra is an herb sometimes used in traditional Chinese medicine to treat hypertension. Tetrandrine, an alkaloid extract of S tetrandra, has been shown to be a calcium ion channel antagonist, paralleling the effects of verapamil. Tetrandrine blocks T and L calcium channels, interferes with the binding of diltiazem and methoxyverapamil at calcium-channel binding sites, and suppresses aldosterone production.31,32 A parenteral dose (15 mg/kg) of tetrandrine in conscious rats decreases mean, systolic, and diastolic blood pressures for more than 30 minutes; however, an intravenous 40-mg/kg dose killed the rats by myocardial depression. In stroke-prone hypertensive rats, an oral dose of 25 or 50 mg/kg produced a gradual and sustained hypotensive effect after 48 hours without affecting plasma renin activity.33 In addition to its cardiovascular actions, tetrandrine has reported antineoplastic, immunosuppressive, and mutagenic effects.31
Tetrandrine is 90% protein-bound with an elimination half-life of 88 minutes, according to dog studies; however, rat studies have shown a sustained hypotensive effect for more than 48 hours after a 25- or 50-mg oral dose. Tetrandrine causes liver necrosis in dogs orally administered 40 mg/kg of tetrandrine 3 times weekly for 2 months, reversible swelling of liver cells with a 20-mg/kg dose, and no observable changes with a 10-mg/kg dose.31 Given the evidence of hepatotoxicity, many more studies are necessary to establish a safe dosage of tetrandrine in humans.
More recently, tetrandrine has been implicated in an outbreak of rapidly progressive renal failure, termed Chinese herb nephropathy. Numerous individuals developed the condition after using a combination of several Chinese herbs as part of a dieting regimen. It has been hypothesized that the cause may be attributed to misidentification of S tetrandra; nonetheless, questions still remain as to the role of tetrandra in the development of this serious toxic effect.34-37
The root of Lingusticum wallichii is used in traditional Chinese medicine as a circulatory stimulant, hypotensive drug, and sedative.38 Tetramethylpyrazine, the active constituent extracted from L wallichii, inhibits platelet aggregation in vitro and lowers blood pressure by vasodilation in dogs. With its actions independent of the endothelium, tetramethylpyrazine's vasodilatory effect is mediated by calcium channel antagonism and nonselective antagonism of α-adrenergic receptors. Some evidence suggests that tetramethylpyrazine acts on the pulmonary vasculature.31 Currently, there is insufficient information to evaluate the safety and efficacy of this herbal medicinal.
Uncaria rhynchophylla is sometimes used in traditional Chinese medicine to treat hypertension. Its indole alkaloids, rhynchophylline and hirsutine, are thought to be the active principles of U rhynchophylla's vasodilatory effect. The mechanism of U rhynchophylla's actions is unclear. Some studies point to an alteration in calcium ion flux in response to activation, whereas others point to hirsutine's inhibition of nicotine-induced dopamine release.31 One in vitro study has shown U rhynchophylla extract relaxes norepinephrine-precontracted rat aorta through endothelium-dependent and -independent mechanisms. For the endothelium-dependent component, U rhynchophylla extract appears to stimulate endothelium-derived relaxing factor and/or nitric oxide release without involving muscarinic receptors.39 Also, in vitro and in vivo studies have shown that rhynchophylline can inhibit platelet aggregation and reduce platelet thromboses induced with collagen or adenosine diphosphate plus epinephrine.31 Safety and efficacy cannot be evaluated at this time because of a lack of clinical data.
Veratrum (hellebore) is a perennial herb grown in many parts of the world. Varieties include Veratrumviride from Canada and the eastern United States, Veratrumcalifornicum from the western United States, Veratrumalbum from Alaska and Europe, and Veratrum japonicum from Asia. All Veratrum plants contain poisonous alkaloids known to cause vomiting, bradycardia, and hypotension. Most cases of Veratrum poisonings are due to misidentification with other plants. Although once a treatment for hypertension, the use of Veratrum alkaloids has lost favor owing to a low therapeutic index and unacceptable toxicity, as well as the introduction of safer antihypertensive drug alternatives.40
Veratrum alkaloids enhance nerve and muscle excitability by increasing sodium ion conductivity. They act on the posterior wall of the left ventricle and the coronary sinus baroreceptors, causing reflex hypotension and bradycardia via the vagus nerve (Bezold-Jarisch reflex). Nausea and vomiting are secondary to the alkaloids' actions on the nodose ganglion.40
The diagnosis of Veratrum toxicity is established by history, identification of the plant, and strong clinical suspicion. Clinical symptoms usually occur quickly, often within 30 minutes.41 Treatment is mainly supportive and directed at controlling bradycardia and hypotension. Veratrum-induced bradycardia usually responds to treatment with atropine; however, the blood pressure response to atropine is more variable and requires the addition of pressors. Other electrocardiographic changes, such as atrioventricular dissociation, may also be reversible with atropine.42 Seizures are a rare complication and may be treated with conventional anticonvulsants. For patients with preexisting cardiac disease, the use of β-agonists or pacing may be necessary. Nausea may be controlled with phenothiazine antiemetics. Recovery usually occurs within 24 to 48 hours.40
Evodia rutaecarpa (wu-chu-yu) is a Chinese herbal drug that has been used as a treatment for hypertension. It contains an active vasorelaxant component called rutaecarpine that can cause endothelium-dependent vasodilation in experimental models.43
Angina pectoris
Crataegus hawthorn, a name encompassing many Crataegus species (such as Crataegus oxyacantha and Crataegus monogyna in the West and Crataegus pinnatifida in China) has acquired the reputation in modern herbal literature as an important tonic for the cardiovascular system that is particularly useful for angina. Crataegus leaves, flowers, and fruits contain a number of biologically active substances, such as oligomeric procyanins, flavonoids, and catechins. From current studies, Crataegus extract appears to have antioxidant properties and can inhibit the formation of thromboxane as well.44,45
Also, Crataegus extract antagonizes the increases in cholesterol, triglyceride, and phospholipid levels in low-density lipoprotein (LDL) and very low-density lipoprotein in rats fed a hyperlipidemic diet; thus, it may inhibit the progression of atherosclerosis.46 This hypocholesterolemic action may be due to an up-regulation of hepatic LDL receptors resulting in greater influx of plasma cholesterol into the liver. Crataegus also prevents cholesterol accumulation in the liver by enhancing cholesterol degradation to bile acids, as well as suppressing cholesterol biosynthesis.47
According to another study, Crataegus extract, in high concentrations, has a cardioprotective effect on ischemic-reperfused hearts without causing an increase in coronary blood flow.48 On the other hand, oral and parenteral administration of oligomeric procyanins of Crataegus has been shown to lead to an increase in coronary blood flow in both cats and dogs.49,50 Double-blind clinical trials have demonstrated simultaneous cardiotropic and vasodilatory actions of Crataegus.51 In essence, Crataegus increases coronary perfusion, has a mild hypotensive effect, antagonizes atherogenesis, and has positive inotropic and negative chronotropic actions.46,52 In a recent multicenter, placebo-controlled, double-blind study, an extract of Crataegus was shown to clearly improve the cardiac performance of patients with New York Heart Association class II heart failure. In this study, the primary parameter analyzed was the heart rate product (systolic blood pressure × heart rate).53 Recent studies have suggested that the mechanism of cardiac action for Crataegus species may be due to the inhibition of the 3‘, 5‘-cyclic adenosine monophosphate phosphodiesterase.54
Hawthorn is relatively devoid of adverse effects. In fact, in comparison with other inotropic drugs such as epinephrine, amrinone, milrinone, and digoxin, Crataegus has a potentially reduced arrhythmogenic risk because of its ability to prolong the effective refractory period, while the other drugs mentioned previously all shorten this parameter.55,56 Also, it should be noted that concomitant use of hawthorn with digitalis can markedly enhance the activity of digitalis.5,57 Undoubtedly, more studies are needed to show that hawthorn can be used safely and effectively.
Because of its resemblance to Panax ginseng (Asian ginseng), Panax notoginseng has acquired the common name of pseudoginseng, especially since it is often an adulterant of P ginseng preparations. In traditional Chinese medicine, the root of P notoginseng is used for analgesia and hemostasis. It is also often used in the treatment of patients with angina and coronary artery disease.38Panax notoginseng has been described as a calcium ion channel antagonist in vascular tissue. More specifically, its pharmacological action may be as a novel and selective calcium ion antagonist that does not interact with the L-type calcium ion channel but rather may interact with the receptor-operated calcium ion channel.58
Although clinical trials are lacking, in vitro studies using P notoginseng suggest possible cardiovascular effects. One study that used purified notoginsenoside R1, extracted from P notoginseng, on human left umbilical vein endothelial cells showed a dose- and time-dependent synthesis of tissue-type plasminogen activating factor without affecting the synthesis of plasminogen activating inhibitor. Thus, fibrinolytic parameters were enhanced.59 Another study suggests that P notoginseng saponins may inhibit atherogenesis by interfering with the proliferation of smooth muscle cells.60 In vitro and in vivo studies using rats and rabbits demonstrate that P notoginseng may be useful as an antianginal drug, since it dilates coronary arteries in all concentrations. The role of P notoginseng in the treatment of hypertension is less certain, since P notoginseng causes vasodilation or vasoconstriction depending on the concentration and target vessel.61 The results of these in vitro and in vivo studies are encouraging; however, clinical trials will be necessary to make a more informed decision regarding the use of P notoginseng.
Salvia miltiorrhiza (dan-shen), a relative of the Western sage Salvia officinalis, is native to China. In traditional Chinese medicine, the root of S miltiorrhiza is used as a circulatory stimulant, sedative, and cooling drug.38Salvia miltiorrhiza may be useful as an antianginal drug because it has been shown to dilate coronary arteries in all concentrations, similar to P notoginseng. Also, S miltiorrhiza has variable action on other vessels depending on its concentration, so it may not be as helpful in treating hypertension.61 In vitro, S miltiorrhiza, in a dose-dependent fashion, inhibits platelet aggregation and serotonin release induced by either adenosine diphosphate or epinephrine, which is thought to be mediated by an increase in platelet cyclic adenosine monophosphate caused by S miltiorrhiza's inhibition of cyclic adenosine monophosphate phosphodiesterase.62Salvia miltiorrhiza appears to have a protective action on ischemic myocardium, enhancing the recovery of contractile force on reoxygenation.63 More recently, S miltiorrhiza has been shown to protect myocardial mitochondrial membranes from ischemia-reperfusion injury and lipid peroxidation because of its free radical–scavenging effects.64 Qualitatively and quantitatively, a decoction of S miltiorrhiza was as efficacious as the more expensive isolated tanshinones.59
Clinical trials will be necessary to evaluate the safety and efficacy of S miltiorrhiza. Of note, it has been observed clinically that when S miltiorrhiza and warfarin sodium are coadministered, there is an increased incidence in warfarin-related adverse effects; in rats S miltiorrhiza was shown to increase the plasma concentrations of warfarin as well as the prothrombin time.65
Atherosclerosis
In addition to its use in the culinary arts, garlic (Allium sativum) has been valued for centuries for its medicinal properties. Garlic is one of the herbal medicines that has been examined more closely by the scientific community. In recent decades, research has focused on garlic's use in preventing atherosclerosis. Garlic, like many of the other herbal medicines discussed previously, has demonstrated multiple beneficial cardiovascular effects. A number of studies have demonstrated these effects that include lowering blood pressure, inhibiting platelet aggregation, enhancing fibrinolytic activity, reducing serum cholesterol and triglyceride levels, and protecting the elastic properties of the aorta.
Consumption of large quantities of fresh garlic (0.25 to 1.0 g/kg or about 5-20 average sized 4-g cloves in a person weighing 78.7 kg) has been shown to produce the beneficial effects mentioned earlier.66 In support of this, a recent double-blind cross-over study was conducted on moderately hypercholesterolemic men that compared the effects of 7.2 g of aged garlic extract with placebo on blood lipid levels. This study found that there was a maximal reduction of 6.1% in total serum cholesterol levels and 4.6% in LDL cholesterol levels with garlic compared with placebo.67
However, despite positive evidence from numerous trials, some investigators have been hesitant to outright endorse the routine use of garlic for cardiovascular disease because many of the published studies had methodological shortcomings,66,68-72 perhaps because constituent trials were small, lacking statistical power. Also, inappropriate methods of randomization, lack of dietary run-in period, short duration, or failure to undertake intention-to-treat analysis may explain the cautious acceptance of previous meta-analyses.73 In fact, one recent study found no demonstrable effect of garlic ingestion on lipid and lipoprotein levels. This study used a cross-over design protected by a washout period to reduce between-subject variability as well as close assessment and reporting of dietary behavior, which had been lacking in previous trials.74 Another study found no effect of garlic on cholesterol absorption, cholesterol synthesis, or cholesterol metabolism.71 As is evident, the precise extent of garlic's impact on atherosclerosis remains controversial; larger, more rigorously designed trials may be necessary to better determine its utility in preventing cardiovascular disease.
Garlic has also been studied in hypertensive patients as a blood pressure–lowering agent. Similar to its lipid effects, no conclusive studies have been conducted and many methodological shortcomings exist in study designs. The results of one meta-analysis that considered 8 different trials suggest some clinical use for patients with mild hypertension, but there is insufficient evidence to recommend its use as routine clinical therapy.68 Garlic has also been shown to possess antiplatelet activity. In the past, this action was mostly documented in vitro.75 A new study examined the effect of the consumption of a fresh clove of garlic on platelet thromboxane production and showed that after 26 weeks, serum thromboxane levels were reduced about 80%.76 This may prove to be beneficial in the prevention of thrombosis in the future. Recently, the effect of long-term garlic intake on the elastic properties of the aorta was also studied. Participants in the trial (limited to those aged 50-80 years) consumed 300 mg/d of standardized garlic powder for more than 2 years. The results showed that the pulse-wave velocity and standardized elastic vascular resistance of the aorta were lower in the garlic group than in the control group. Consequently, long-term garlic powder intake may have a protective effect on the elastic properties of the aorta related to aging.77 In these ways, garlic has shown numerous beneficial cardiovascular effects that need to be investigated further to determine its therapeutic utility.
Intact cells of garlic bulbs include an odorless, sulfur-containing amino acid known as allinin. When garlic is crushed, allinin comes into contact with allinase, which converts allinin to allicin. Allicin has potent antibacterial properties, but it is also highly odoriferous and unstable. Ajoenes, self-condensation products of allicin, appear to be responsible for garlic's antithrombotic activity. Most authorities now agree that allicin and its derivatives are the active constituents of garlic's physiological activity. Fresh garlic releases allicin in the mouth during the chewing process. Dried garlic preparations lack allicin but contain allinin and allinase. Since allinase is inactivated in the stomach, dried garlic preparations should be coated with enteric so that they pass through the stomach into the small intestine where allinin can be enzymatically converted to allicin. Few commercial garlic preparations are standardized for their allicin yield based on allinin content, hence making their effectiveness less certain.5 However, one double-blind, placebo-controlled study involving 261 patients for 4 months using one 800-mg tablet of garlic powder daily, standardized to 1.3% allinin content, demonstrated significant reductions in total cholesterol (12%) and triglyceride levels (17%).78
Aside from a garlic odor on the breath and body, moderate garlic consumption causes few adverse effects. However, consumption in excess of 5 cloves daily may result in heartburn, flatulence, and other gastrointestinal disturbances. Some people have reported allergic reactions to garlic, most commonly allergic contact dermatitis. Patch testing with 1% diallyl disulfide is recommended when garlic allergy is suspected.79 Because of its antithrombotic activity, garlic should be used with caution in people taking oral anticoagulants concomitantly.5,80
The resin of Commiphora mukul (gugulipid), a small, thorny tree native to India, has long been used in Ayurvedic medicine to treat lipid disorders. The primary mechanism of action of gugulipid is through an increase in the uptake and metabolism of LDL cholesterol by the liver.81 In a double-blind, cross-over study completed in 125 patients taking gugulipid compared with 108 patients taking clofibrate, the average decrease in serum cholesterol and triglyceride levels was 11% and 16.8%, respectively, with gugulipid compared with 10% and 21.6%, respectively, with clofibrate. In general, hypercholesterolemic patients responded more favorably to gugulipid therapy than hypertriglyceridemic patients.82 Moreover, it was shown in another randomized, double-blind trial that C mukul also decreased LDL cholesterol levels by 12.5% and the total cholesterol–high-density lipoprotein cholesterol ratio by 11.1%, whereas the levels were unchanged in the placebo group.83
Besides being potentially as effective in lowering blood lipid levels as modern hyperlipidemic drugs, gugulipid may even be safer. In the trial mentioned previously, compliance was greater than 96%, with only the adverse effects of headache, mild nausea, and hiccups noted.83 However, it has been shown that gugulipid may affect the bioavailability of other cardiovascular drugs, namely, propranolol hydrochloride and diltiazem hydrochloride. Gugulipid significantly reduced the peak plasma concentration and area under the curve of both these drugs, which may lead to diminished efficacy or nonresponsiveness.84 Undoubtedly, gugulipid is a natural lipid-lowering drug with potential for therapeutic use, but rigorous, larger clinical trials will be necessary to further evaluate its safety and efficacy before it can be endorsed as an alternative therapy for hyperlipidemia and prevention of atherosclerosis.
Maharishi amrit kalash-4 and Maharishi amrit kalash-5 are 2 complex herbal mixtures with significant antioxidant properties that have been shown to inhibit LDL oxidation in patients with hyperlipidemia. In experimental studies, the herbal mixtures have also been shown to inhibit enzymatic- and nonenzymatic-induced microsomal lipid peroxidation and platelet aggregation.85
Cerebral and peripheral vascular disease
Having existed for more than 200 million years, Ginkgo biloba (maidenhair tree) was apparently saved from extinction by human intervention, surviving in Far Eastern temple gardens while disappearing for centuries in the West. It was reintroduced to Europe in 1730 and became a favorite ornamental tree.38,86 Although the root and kernels of G biloba have long been used in traditional Chinese medicine, the tree gained attention in the West during the 20th century for its medicinal value after a concentrated extract of G biloba leaves was developed in the 1960s. At least 2 groups of substances within G biloba extract (GBE) demonstrate beneficial pharmacological actions. The flavonoids reduce capillary permeability as well as fragility and serve as free radical scavengers. The terpenes (ie, ginkgolides) inhibit platelet-activating factor, decrease vascular resistance, and improve circulatory flow without appreciably affecting blood pressure.57,87 Continuing research appears to support the primary use of GBE for treating cerebral insufficiency and its secondary effects on vertigo, tinnitus, memory, and mood; also, GBE appears to be useful for treating peripheral vascular disease, including diabetic retinopathy and intermittent claudication.5,57,87-91
In a randomized, placebo-controlled, double-blind study, EGb 761, which is a standardized extract of G biloba with respect to its flavonol glycoside and terpene lactone content, was shown to significantly decrease the areas of ischemia as measured by transcutaneous partial pressure of oxygen during exercise. Because of its rapid anti-ischemic action, EGb 761 may be valuable in the treatment of intermittent claudication and peripheral artery disease in general.92
Also, studies have been examining the cardioprotective efficacy of EGb 761 in regard to its anti–free radical action in myocardial ischemia–reperfusion injury. In vitro studies with animal models have shown that this compound may exert such an effect.93,94 A clinical study of 15 patients undergoing coronary bypass surgery demonstrated that oral EGb 761 therapy may limit free radical–induced oxidative stress occurring in the systemic circulation and at the level of the myocardium during these operations.95 It remains to be studied whether extracts of G biloba may be used as pharmacological adjuvants to limit tissue damage and metabolic alterations following coronary bypass surgery, coronary angioplasty for acute myocardial infarctions, or even in managing coronary thrombosis.
Although approved as a drug in Europe, Ginkgo is not approved in the United States and is instead marketed as a food supplement, usually supplied as 40-mg tablets of extract. Since most of the investigations examining the efficacy of GBEs used preparations such as EGb 761 or LI 1370, the bioequivalence of other GBE products has not been established. The recommended dosage in Europe is one 40-mg tablet taken 3 times daily with meals (120 mg/d).5,87 Adverse effects due to GBE are rare but can include gastrointestinal disturbances, headache, and allergic skin rash.5,87
Known mostly as a culinary spice and flavoring agent, Rosmarinus officinalis (rosemary) is listed in many herbal sources as a tonic and all-around stimulant. Traditionally, rosemary leaves are said to enhance circulation, aid digestion, elevate mood, and boost energy. When applied externally, the volatile oils are supposedly useful for arthritic conditions and baldness.5
Although research on rosemary is scant, some studies have focused on antioxidant effects of diterpenoids, especially carnosic acid and carnosol, isolated from rosemary leaves. In addition to having antineoplastic effects, antioxidants in rosemary have been credited with stabilizing erythrocyte membranes and inhibiting superoxide generation and lipid peroxidation.96,97 Essential oils of rosemary have demonstrated antimicrobial, hyperglycemic, and insulin-inhibiting properties.98,99 Rosemary leaves contain high amounts of salicylates, and its flavonoid pigment diosmin is reported to decrease capillary permeability and fragility.57,100,101
Despite the conclusions derived from in vitro and animal studies, the therapeutic use of rosemary for cardiovascular disorders remains questionable, because few, if any, clinical trials have been conducted using rosemary. Because of the lack of studies, no conclusions can be reached regarding the use of the antioxidants of rosemary in inhibiting atherosclerosis. Although external application may cause cutaneous vasodilation from the counterirritant properties of rosemary's essential oils, there is no evidence to support any prolonged improvement in peripheral circulation.5 While rosemary does have some carminative properties, it may also cause gastrointestinal and kidney disturbances in large doses.5,101 Until more studies are done, rosemary should probably be limited to its use as a culinary spice and flavoring agent rather than as a medicine.
Venous insufficiency
The seeds of horse chestnut, Aesculus hippocastanum, have long been used in Europe to treat venous disorders such as varicose veins. The saponin glycoside aescin from horse chestnut extract (HCE) inhibits the activity of lysosomal enzymes thought to contribute to varicose veins by weakening vessel walls and increasing permeability, which result in dilated veins and edema.5 In fact, recent research has shown that A hippocastanum inhibits only against hyaluronidase but not elastase, and this activity is linked mainly to the saponin escin.102 In animal studies, HCE, in a dose-dependent fashion, increases venous tone, venous flow, and lymphatic flow. It also antagonizes capillary hyperpermeability induced by histamine, serotonin, or chloroform. This extract has been shown to decrease edema formation of lymphatic and inflammatory origin. Horse chestnut extract has antiexudative properties, suppressing experimentally induced pleurisy and peritonitis by inhibiting plasma extravasation and leukocyte emigration, and its dose-dependent antioxidant properties can inhibit in vitro lipid peroxidation.103,104 Randomized, double-blind, placebo-controlled trials with HCE show are eduction in edema, measured using plethysmography.105,106
In another recent randomized, placebo-controlled study, the efficacy and safety of class 2 compression stockings and dried HCE were compared. Both HCE and the compression stockings decreased lower leg edema after 12 weeks of therapy; the results showed an average 43.8-mL reduction with HCE and 46.7-mL with compression stockings, while the placebo group showed an increase of 9.8 mL. Both HCE and compression therapy were well tolerated, with no serious adverse effects. This study may indicate that both of these modalities are reasonable alternatives for the effective treatment of patients with chronic venous insufficiency.107 Also, HCE has been shown to markedly improve other symptoms associated with chronic venous insufficiency, such as pain, tiredness, itching, and tension in the swollen leg, in a case-observation study.108 Aside from effects on venous insufficiency, prophylactic use of HCE has been thought to decrease the incidence of thromboembolic complications of gynecological surgery. However, since this issue is still controversial,109 this does not appear to be the case.109
Standardized HCE is prepared as an aqueous alcohol extract of 16% to 21% of triterpene glycosides, calculated as aescin. The usual initial dosage is 90 to 150 mg/d of aescin, which may be reduced to 35 to 70 mg/d if clinical benefit is seen.5 Standardized HCE preparations are not available in the United States, but nonstandardized products may be available.
Some manufacturers promote the use of topical preparations of HCE for treatment of varicose veins as well as hemorrhoids; however, at least one study has demonstrated poor aescin distribution at sites other than the skin and muscle tissues underlying the application site.110 Moreover, the involvement of arterioles and veins in the pathophysiology of hemorrhoids makes the effectiveness of HCE doubtful, since HCE has no known effects on the arterial circulation. For now, research studies have yet to confirm any clinical effectiveness of topical HCE preparations.
Although adverse effects are uncommon, HCE may cause gastrointestinal irritation. Parenteral aescin has produced isolated cases of anaphylactic reactions, as well as hepatic and renal toxic effects.5,111-113 In the event of toxicity, aescin can be eliminated via dialysis, with elimination dependent on protein-binding.114 Horse chestnut extract is also one of the components of venocuran, a drug marketed as a treatment for venous disorders. In 1975, venocuran was determined to cause a pseudolupus syndrome characterized by recurrent fever, myalgia, arthralgia, pleuritis, pulmonary infiltrates, pericarditis, myocarditis, and mitochondrial antibodies in the absence of nuclear antibodies after prolonged treatment.115,116 Venocuran has since been withdrawn from the market; however, the nature of its pathophysiologic action is still unknown.
Like A hippocastanum, Ruscus aculeatus (butcher's broom) is also known for its use in treating venous insufficiency. Ruscus aculeatus is a short evergreen shrub found commonly in the Mediterranean region. Two steroidal saponins, ruscogenin and neurogenin, extracted from the rhizomes of R aculeatus are thought to be its active components.101 In vivo studies on hamster cheek pouch reveal that topical Ruscus extract dose dependently antagonizes histamine-induced increases in vascular permeability.117 Moreover, topical Ruscus extract causes dose-dependent constriction of venules without appreciably affecting arterioles.118 Topical Ruscus extract's vascular effects are also temperature dependent and appear to counter the sympathetic nervous system's temperature-sensitive vascular regulation: venules dilate at a lower temperature (25°C), constrict at near physiologic temperatures (36.5°C), and further constrict at higher temperatures (40°C); arterioles dilate at 25°C, are unaffected at 36.5°C, and remain unaffected or constrict at 40°C, depending on Ruscus concentration.119 Based on the influence of prazosin, diltiazem, and rauwolscine, the peripheral vascular effects of Ruscus extract appear to be selectively mediated by effects on calcium channels and α1-adrenergic receptors with less activity at α2-adrenergic receptors.117,118 Also, R aculeatus exhibits strong antielastase activity and has little effect on hyaluronidase in direct contrast to A hippocastanum. This activity may contribute to their efficacy in the treatment of venous insufficiency since these enzyme systems are involved in the turnover of the main components of the perivascular amorphous substance.102
Several small clinical trials using topical Ruscus extract support its role in treating venous insufficiency. One randomized, double-blind, placebo-controlled trial involving 18 volunteers showed a beneficial decrease in femoral vein diameter (median decrease, 1.25 mm) using duplex B-scan ultrasonography. The decrease was measured 2.5 hours after applying 4 to 6 g of a cream containing 64 to 96 mg of Ruscus extract.120 In another small trial (N = 18) it was shown that topical Ruscus extract may be helpful in reducing venous dilation during pregnancy.121 Oral agents may be useful as topical drugs for venous insufficiency, although the evidence is less convincing.122
Although capsule, tablet, ointment, and suppository (for hemorrhoids) preparations of Ruscus extract are available in Europe, only capsules are available in the United States. These capsules contain 75 mg of Ruscus extract and 2 mg of rosemary oil.101 Aside from occasional nausea and gastritis, adverse effects from using R aculeatus have rarely been reported, even in high doses.57 Nevertheless, one should be wary of any drug that has not been thoroughly tested. Although there is ample evidence to support the pharmacological activity of R aculeatus, there is still a relative deficiency of clinical data to establish its actual safety and efficacy. Until more studies are completed, no recommendations regarding dosage can be offered.
Arrhythmia
In traditional Chinese medicine, arrhythmias are categorized by the characteristic symptoms of palpitations and abnormal pulse. Numerous Chinese herbal medicines are identified to have antiarrhythmic effects, such as xin bao, ci zhu wan, bu xin dan, and several others.123 However, few clinical trials have been conducted to study their effects and safety. Xin bao is one agent that has begun to be examined. The mechanism of action of xin bao is thought to be through its stimulation and increased excitability of the sinuatrial node.124 In one observational study, the effects of xin bao were documented in 87 patients with sick sinus syndrome. Xin bao was administered orally 2 to 3 times per day for 2 months. Patients with major symptoms of sick sinus syndrome, which included dizziness, palpitations, and chest pressure, improved significantly after treatment.124 No serious adverse effects were noted. This study suggests a possible role of xin bao in the treatment of sick sinus syndrome. However, more scientific research on xin bao and other antiarrhythmic Chinese herbs mentioned previously are necessary before any recommendations can be made for their routine use in patients with sick sinus syndrome or other arrhythmias.
Comment
With the high prevalence of herbal medicine use in the United States, health practitioners should remember to inquire about such health practices when taking clinical histories and remain informed of the beneficial or harmful effects of these treatments. Continuing research is necessary to elucidate the pharmacological activities of the many cardiopotent herbal medicines and to stimulate future pharmaceutical development of therapeutically beneficial herbal drugs. However, such research is currently lacking in the United States and requires more support from government agencies before the full potential of these types of treatments can be determined. At the same time, legal surveillance of herbal medicine use with low safety margins should be instituted for the sake of public health; this is especially imperative for those herbs with adverse cardiovascular reactions125 and drug interactions. As more information becomes available regarding the safety and efficacy of herbal medicines through new clinical trials, research-supported claims may one day become available to consumers and physicians in a manner similar to the allopathic medicines.
Accepted for publication July 23, 1998.
Reprints: William H. Frishman, MD, New York Medical College, Munger Pavilion, Valhalla, NY 10595.
References
1.
Wu B-NHuang Y-CWu H-MHong S-JChiang L-CChen I-J A highly selective (α1-adrenergic blocker with partial β2 agonist activity derived from ferulic acid, an active component of Ligusticum wallichii Franch. J Cardiovasc Pharmacol. 1998;31750- 757Google ScholarCrossref
2.
Eisenberg DMKessler RCFoster C et al. Unconventional medicine in the United States. N Engl J Med. 1993;328246- 252Google ScholarCrossref
3.
Astin JA Why patients use alternative medicine: results of a national study. JAMA. 1998;2791548- 1553
ArticleGoogle ScholarCrossref
4.
Ernst E Harmless herbs? a review of the recent literature. Am J Med. 1998;104170- 178Google ScholarCrossref
5.
Tyler VE Herbs of Choice: The Therapeutic Use of Phytomedicinals. New York, NY Pharmaceutical Product Press1994;
6.
Stuhlemmer UKreis WEisenbeiss MReinhard E Cardiac glycosides in partly submerged shoots of Digitalis lanata. Planta Med. 1993;59539- 545Google ScholarCrossref
7.
Dickstein ESKunkel FW Foxglove tea poisoning. Am J Med. 1980;69167- 169Google ScholarCrossref
8.
Radford DJGillies ADHinds JADuffy P Naturally occurring cardiac glycosides. Med J Aust. 1986;144540- 544Google Scholar
9.
Cheung KHinds JADuffy P Detection of poisoning by plant-origin cardiac glycoside with the Abbott Tdx analyzer. Clin Chem. 1989;35295- 297Google Scholar
10.
Moxley RASchneider NRSteinegger DHCarlson MP Apparent toxicosis associated with lily-of-the-valley (Convallaria majalis) ingestion in a dog. J Am Vet Med Assoc. 1989;195485- 487Google Scholar
11.
Bossi MBrambilla GCavilla A et al. Threatening arrhythmia by uncommon digitalic toxicosis [in Italian]. G Ital Cardiol. 1981;112254- 2257Google Scholar
12.
Hayes BEBessen HAWightman WD Oleander tea: herbal draught of death. Ann Emerg Med. 1985;14350- 353Google ScholarCrossref
13.
Ansford AJMorris H Fatal oleander poisoning. Med J Aust. 1981;1360- 361Google Scholar
14.
Shaw DPearn J Oleander poisoning. Med J Aust. 1979;2267- 269Google Scholar
15.
Tumcok YKozan OCadvar C et al. Urginea maritima (squill) toxicity. J Toxicol Clin Toxicol. 1995;3383- 86Google ScholarCrossref
16.
Bagrov AYRoukoyatkina NIPinaev AGDmitrieva RI Effects of two endogenous Na+, K(+)-ATPase inhibitors, marinobufagenin and ouabain, on isolated rat aorta. Eur J Pharmacol. 1995;274151- 158Google ScholarCrossref
17.
Nishoka SAResende ES Transitory complete atrioventricular block associated to ingestion of Nerium oleander. Rev Assoc Med Bras. 1995;4160- 62Google Scholar
18.
Rich SALibera JMLocke RJ Treatment of foxglove extract poisoning with digoxin-specific Fab fragments. Ann Emerg Med. 1993;221904- 1907Google ScholarCrossref
19.
Safadi RLevy IAmitai YCaraco Y Beneficial effect of digoxin-specific Fab antibody fragments in oleander intoxication. Arch Intern Med. 1995;1552121- 2125
ArticleGoogle ScholarCrossref
20.
Galey FDHolstege DMPlumlee KH et al. Diagnosis of oleander poisoning in livestock. J Vet Diagn Invest. 1996;8358- 364Google ScholarCrossref
21.
Langford SDBoor PJ Oleander toxicity: an examination of human and animal toxic exposure. Toxicology. 1996;1091- 13Google ScholarCrossref
22.
McRae S Elevated serum digoxin levels in a patient taking digoxin and Siberian ginseng. CMAJ. 1996;155293- 295Google Scholar
23.
Geehr E Common toxic plant ingestions. Emerg Med Clin North Am. 1984;2553- 563Google Scholar
24.
Litovitz TLClark LRSoloway RA 1993 Annual Report of the American Association of Poison Control Centers Toxic Exposure Surveillance System. Am J Emerg Med. 1993;12546- 584Google ScholarCrossref
25.
Szabuniewicz MMcGrady JDCamp BJ Treatment of experimentally induced oleander poisoning. Arch Int Pharmcodyn. 1971;18912- 21Google Scholar
26.
Osterloh JHerold SPond S Oleander interference in the digoxin radioimmunoassay in a fatal ingestion. JAMA. 1982;2471596- 1597
ArticleGoogle ScholarCrossref
27.
Oates JA Antihypertensive agents and the drug therapy of hypertension. Hardman JGLimbird LEMolinoff PB et al. eds Goodman and Gilman's The Pharmacological Basis of Therapeutics. 9th ed. New York, NY McGraw-Hill Book Co1996;781- 808Google Scholar
28.
Not Available, Rauwolfia alkaloids USP DI Drug Information for the Health Care Professional. Vol 118th ed. Rockville, Md US Pharmacopeial Convention1998;2486Google Scholar
29.
Not Available, Physician's Desk Reference. 43rd ed. Montvale, NJ Medical Economics Co1989;1648
30.
Brunton LL Agents affecting gastrointestinal water flux, emesis and antiemetics, bile acids and pancreatic enzymes. Hardman JGLimbird LEMolinoff PB et al. eds Goodman and Gilman's The Pharmacological Basis of Therapeutics 9th ed. New York, NY McGraw-Hill Book Co1996;917- 936Google Scholar
31.
Sutter MCWang YX Recent cardiovascular drugs from Chinese medicinal plants. Cardiovasc Res. 1993;271891- 1901Google ScholarCrossref
32.
Rossier MFPython CPCapponi AM et al. Blocking T-type calcium channels with tetrandrine inhibits steroidogenesis in bovine adrenal glomerulosa cells. Endocrinology. 1993;1321035- 1043Google Scholar
33.
Kawashima KHayakawa TMiwa Y et al. Structure and hypotensive activity relationships of tetrandrine derivatives in stroke-prone spontaneously hypertensive rats. Gen Pharmacol. 1990;21343- 347Google ScholarCrossref
34.
Vanherweghem JL A new form of nephropathy secondary to the absorption of Chinese herbs [in French]. Bull Mem Acad R Med Belg. 1994;149128- 135Google Scholar
35.
Schmeiser HHBieler CAWiessler MVan Ypersele DEStrihou CCosyns JP Detection of DNA adducts formed by aristolochic acid in renal tissue from patients with Chinese herbs nephropathy. Cancer Res. 1996;562025- 2028Google Scholar
36.
Bieler CAStiborova MWiessler M et al. 32P-post-labelling analysis of DNA adducts formed by aristolochic acid in tissues from patients with Chinese herbs nephropathy. Carcinogenesis. 1997;181063- 1067Google ScholarCrossref
37.
Violin C Belgian (Chinese herb) nephropathy: why? J Pharm Belg. 1997;527- 27Google Scholar
38.
Ody P The Complete Medicinal Herbal. New York, NY Dorling Kindersley1993;
39.
Kuramochi TChu JSuga T Gou-teng (from Uncaria rhynchophylla Miquel)-induced endothelium-dependent and -independent relaxations in the isolated rat aorta. Life Sci. 1994;542061- 2069Google ScholarCrossref
40.
Jaffe AMGephardt DCourtemanche L Poisoning due to ingestion of Veratrum viride (false hellebore). J Emerg Med. 1990;8161- 167Google ScholarCrossref
41.
Quatrehomme GBertrand FChauvet COllier A Intoxication from Veratrum album. Hum Exp Toxicol. 1993;12111- 115Google ScholarCrossref
42.
Festa MAndreeto BBallaris MAPanio APiervittori R Un caso di avvelenamento da Veratro. Minerva Anestesiol. 1996;62195- 196Google Scholar
43.
Chiou W-FShum AY-CLiao J-FChen C-F Studies of the cellular mechanisms underlying the vasorelaxant effects of rutaecarpine, a bioactive component extracted from an herbal drug. J Cardiovasc Pharmacol. 1997;29490- 498Google ScholarCrossref
44.
Bahorun TTrotin FPommery J et al. Antioxidant activities of Crataegus monogyna extracts. Planta Med. 1994;60323- 328Google ScholarCrossref
45.
Vibes JLasserre BGleye J et al. Inhibition of thromboxane A2 biosynthesis in vitro by the main components of Crataegus oxyacantha (Hawthorn) flower heads. Prostaglandins Leukot Essent Fatty Acids. 1994;50173- 175Google ScholarCrossref
46.
Shanthi SParasakthy KDeepalakshmi PDDevaraj SN Hypolipidemic activity of tincture of Crataegus in rats. Indian J Biochem Biophys. 1994;31143- 146Google Scholar
47.
Rajerdan SDeepalakshmi PDParasakthy KDevaraj HDevaraj SN Effect of tincture of Crataegus on the LDL-receptor activity of hepatic plasma membrane of rats fed an atherogenic diet. Atherosclerosis. 1996;123235- 241Google ScholarCrossref
48.
Nasa YHashizume HHoque ANAbiko Y Protective effect of Crataegus extract on the the cardiac mechanical dysfunction in isolated perfused working rat heart. Arzneimittelforschung. 1993;43945- 949Google Scholar
49.
Roddweig CHensel H Reaction of local myocardial blood flow in nonanesthesized dogs and anesthesized cats to the oral and parenteral administration of a Crataegus fraction (oligomere procyanidines) [in German]. Arzneimittelforschung. 1977;271407- 1410Google Scholar
50.
Taskov M On the coronary and cardiotonic action of crataegus. Acta Physiol Pharmacol Bulg. 1977;353- 57Google Scholar
51.
Blesken R Crataegus in cardiology [in German]. Fortschr Med. 1992;110290- 292Google Scholar
52.
Petkov V Plants and hypotensive, antiatheromatous and coronarodilating action. Am J Chin Med. 1979;7197- 236Google ScholarCrossref
53.
Weikl AAssmus KDNeukum-Schmidt A et al. Crataegus special extract WS 1442: assessment of objective effectiveness in patients with heart failure (NYHA II). Fortschr Med. 1996;114291- 296Google Scholar
54.
Schussler MHolzl JFricke U Myocardial effects of flavonoids from Crataegus species. Arzneimittelforschung. 1995;45842- 845Google Scholar
55.
Joseph GZhao YKlaus W Pharmacologic action profile of Crataegus extract in comparison to epinephrine, amirinone, milrinone, and digoxin in the isolated perfused guinea pig heart [in German]. Arzneimittelforschung. 1995;451261- 1265Google Scholar
56.
Popping SRose HIonescu IFischer YKammermeier H Effect of a hawthorn extract on contraction and energy turnover of isolated rat cardiomyocytes. Arzneimittelforschung. 1995;451157- 1161Google Scholar
57.
Mawrey DB Herbal Tonic Therapies. New Canaan, Conn Keats Publishing Inc1993;
58.
Kwan CY Vascular effects of selected antihypertensive drugs derived from traditional medicinal herbs. Clin Exp Pharmacol Physiol. 1995;22297- 299Google ScholarCrossref
59.
Zhang WWojta JBinder BR Effect of notoginsenoside R1 on the synthesis of tissue-type plasminogen activator and plasminogen activator inhibitor-1 in cultured human umbilical vein endothelial cells. Arterioscler Thromb Vasc Biol. 1994;141040- 1046Google ScholarCrossref
60.
Lin SGZheng XLChen QYSun JJ Effect of Panax notoginseng saponins on increased proliferation of cultured aortic smooth muscle cells stimulated by hypercholestoremic serum. Chung Kuo Yao Li Hsueh Pao. 1993;14314- 316Google Scholar
61.
Lei XLChiou GC Cardiovascular pharmacology of Panax notoginseng (Burk) FH Chen and Salvia miltiorrhiza. Am J Chin Med. 1986;14145- 152Google ScholarCrossref
62.
Wang ZRoberts JMGrant PG et al. The effect of a medicinal Chinese herb on platelet function. Thromb Haemost. 1982;48301- 306Google Scholar
63.
Yagi AFujimoto KTanonaka K et al. Possible active components of tan-shen (Salvia miltiorrhiza) for protection of the myocardium against ischemia-induced derangements. Planta Med. 1989;5551- 54Google ScholarCrossref
64.
Zhao BLJiang WZhao YHou JWXin WJ Scavenging effects of Salvia miltiorrhiza on free radicals and its protection for myocardial mitochondrial membranes from ischemia-reperfusion injury. Biochem Mol Biol Int. 1996;381171- 1182Google Scholar
65.
Chan KLo ACYeung JHWoo KS The effects of Danshen (Salvia miltiorrhiza) on warfarin pharmacodynamics and pharmacokinetics of warfarin enantiomers in rats. J Pharm Pharmacol. 1995;47402- 406Google ScholarCrossref
66.
Kleijnen JKnipschild PTer Riet G Garlic, onions and cardiovascular risk factors: a review of the evidence from human experiments with emphasis on commercially available preparations. Br J Clin Pharmacol. 1989;28535- 544Google ScholarCrossref
67.
Steiner MKhan AHHolbert DLin RI A double-blind cross-over study in hypercholesterolemic men that compared the effect of aged garlic extract and placebo administration on blood lipids. Am J Clin Nutr. 1996;64866- 870Google Scholar
68.
Silagy CANeil HA A meta-analysis of the effect of garlic on blood pressure. J Hypertens. 1994;12463- 468Google Scholar
69.
Silagy CNeil A Garlic as a lipid lowering agent: a meta-analysis. J R Coll Physicians Lond. 1994;2839- 45Google Scholar
70.
Kendler BS Garlic (Allium sativum) and onion (Allium cepa): a review of their relationship to cardiovascular disease. Prev Med. 1987;16670- 685Google ScholarCrossref
71.
Isaacsohn JLMoser MStein EA et al. Garlic powder and plasma lipids and lipoproteins: a multicenter, randonized, plcebo-controlled trial. Arch Intern Med. 1998;1581189- 1194
ArticleGoogle ScholarCrossref
72.
Jain AKVargas RGotzkowsky SMcMahon FG Can garlic reduce levels of serum lipids? a controlled clinical study. Am J Med. 1993;94632- 635Google ScholarCrossref
73.
Neil HASilagy CALancaster T et al. Garlic powder in the treatment of moderate hyperlipidaemia: a controlled trial and meta-analyses. J R Coll Physicians Lond. 1996;30329- 334Google Scholar
74.
Simons LABalaSubramaniam SVon Konigsmark MParfitt ASimons JPeters W On the effect of garlic on plasma lipids and lipoproteins in mild hypercholesterolemia. Atherosclerosis. 1995;113219- 225Google ScholarCrossref
75.
Bordia AVerma SKSrivastava KC Effect of garlic on platelet aggregation in humans: a study in healthy subjects and patients with coronary artery disease. Prostaglandins Leukot Essent Fatty Acids. 1996;55201- 205Google ScholarCrossref
76.
Ali MThomson M Consumption of a garlic clove a day could be beneficial in preventing thrombosis. Prostaglandins Leukot Essent Fatty Acids. 1995;53211- 212Google ScholarCrossref
77.
Breithaupt-Grogler KLing MBoudoulas HBeliz GG Protective effect of chronic garlic intake on elastic properties of aorta in the elderly. Circulation. 1997;962649- 2655Google ScholarCrossref
78.
Mader FH Treatment of hyperlipidaemia with garlic powder tablets: evidence from the German Association of General Practitioners' Multicentric Placebo-Controlled Double-Blind Study. Arzneimittelforschung. 1990;401111- 1116Google Scholar
79.
Delaney TADonnelley AM Garlic dermatitis. Australas J Dermatol. 1996;37109- 110Google ScholarCrossref
80.
Rose KDCroissant PDParliament CFLevin MB Spontaneous spinal epidural hematoma with associated liver dysfunction from excessive garlic ingestion: a case report. Neurosurgery. 1990;26880- 882Google ScholarCrossref
81.
Singh VKaul SChander RKapoor NK Stimulation of low density lipoprotein receptor activity in liver membrane of guggulsterone treated rats. Pharmacol Res. 1990;2237- 44Google ScholarCrossref
82.
Nityanand SSrivastava JSAsthana OP Clinical trials with gugulipid: a new hypolipidemic agent. J Assoc Physicians India. 1989;37323- 328Google Scholar
83.
Sing RBNiaz MAGhosh S Hypolipidemic and antioxidant effects of Commiphora mukul as an adjunct to dietary therapy in patients with hypercholesterolemia. Cardiovasc Drugs Ther. 1994;8659- 664Google ScholarCrossref
84.
Dalvi SSNayak VKPohujani SMDesai NKKshirsagar NAGupta KC Effect of gugulipid on bioavailability of diltiazem and propranolol. J Assoc Physicians India. 1994;42454- 455Google Scholar
85.
Sundaram VHanna ANLubow GPKoneru LFalko JMSharma HM Inhibition of low-density lipoprotein oxidation by oral herbal mixtures Maharishi Amrit Kalash-4 and Maharishi Amrit Kalash-5 in hyperlipidemic patients. Am J Med Sci. 1997;314303- 310Google ScholarCrossref
86.
Huxtable RJ The pharmacology of extinction. J Ethnopharmacol. 1992;371- 11Google ScholarCrossref
87.
Z'Brun A Ginkgo: myth and reality [in German]. Schweiz Rundsch Med Prax. 1995;841- 6Google Scholar
88.
Kleinjen JKnipschild P Ginkgo biloba for cerebral insufficiency. Br J Clin Pharmacol. 1992;34352- 358Google ScholarCrossref
89.
Allard M Treatment of the disorders of aging with Ginkgo biloba extract [in French]. Presse Med. 1986;151540- 1545Google Scholar
90.
LeBars PLKatz MMBerman N et al. North American Study Group, A placebo-controlled, double-blind, randomized trial of an extract of Ginkgo biloba for dementia JAMA. 1997;2781327- 1332
ArticleGoogle ScholarCrossref
91.
Doly MDroy-Lefaix MTBraquet P Oxidative stress in diabetic retina. EXS. 1992;62299- 307Google Scholar
92.
Mouren XCalliard PSchwartz F Study of the antiischemic action of EGb 761 in the treatment of peripheral arterial occlusive disease by TcPO2 determination. Angiology. 1994;45413- 417Google ScholarCrossref
93.
Tosaki ADroy-Lefaix MTPali TDas DK Effects of SOD, catalase, and a novel antiarrhythmic drug, Egb 761, on reperfusion-induced arrhythmias in isolated rat hearts. Free Radic Biol Med. 1993;14361- 370Google ScholarCrossref
94.
Haramaki NAggarwal SKawabata TDroy-Lefaix MTPacker L Effects of natural antioxidant EGb 761 on myocardial ischemia-reperfusion injury. Free Radic Biol Med. 1994;16789- 794Google ScholarCrossref
95.
Pietri SSeguin JRD'Arbigny PDrieu KCulcasi M Egb 761 pretreatment limits free radical-induced oxidative stress in patients undergoing coronary bypass surgery. Cardiovasc Drugs Ther. 1997;11121- 131Google ScholarCrossref
96.
Offord EAMace KRuffieux C et al. Rosemary components inhibit benzo[a]pyrene induced genotoxicity in human bronchial cells. Carcinogenesis. 1995;162057- 2062Google ScholarCrossref
97.
Haraguchi HSaito TOkamura NYagi A Inhibition of lipid peroxidation and superoxide generation by diterpenoids from Rosmarinus officinalis. Planta Med. 1995;61333- 336Google ScholarCrossref
98.
Larrondo JVAgut MCalvo-Torras MA Antimicrobial activity of essences from labiates. Microbios. 1995;82171- 172Google Scholar
99.
Al-Hader AAHasan ZAAqel MB Hyperglycemic and insulin release inhibitory effects of Rosmarinus officinalis. J Ethnopharmacol. 1994;43217- 221Google ScholarCrossref
100.
Swan ARDutton SPTruswell AS Salicylates in foods. J Am Diet Assoc. 1985;85950- 960Google Scholar
101.
Tyler VE The Honest Herbal: A Sensible Guide to the Use of Herbs and Related Remedies. 3rd ed. New York, NY Pharmaceutical Product Press1993;
102.
Facino RMCarini MStefani RAldini GSaibene L Anti-elastase and anti-hyaluronidase activities of saponins and sapogenins from Hedera helix, Aesculus hippocastanum, and Ruscus aculeatus: factors contributing to their efficacy in the treatment of venous insufficiency. Arch Pharm (Weinheim). 1995;328720- 724Google ScholarCrossref
103.
Guillaume MPadioleau F Veinotonic effect, vascular protection, antiinflammatory and free radical scavenging properties of horse chestnut extract. Arzneimittelforschung. 1994;4425- 35Google Scholar
104.
Rothkopf MVogel G New findings on the efficacy and mode of action of the horse chestnut saponin escin [in German]. Arzneimittelforschung. 1976;26225- 235Google Scholar
105.
Diehm CVollbrecht Not AvailableAmendt KComberg HU Medical edema protection: clinical benefit in patients with chronic deep vein incompetence: a placebo controlled double-blind study. Vasa. 1992;21188- 192Google Scholar
106.
Bisler HPfeifer RKluken NPauschinger P Effects of horse chestnut seed extract on transcapillary filtration in chronic venous insufficiency [in German]. Dtsch Med Wochenschr. 1986;1111321- 1329Google ScholarCrossref
107.
Diehm CTrampisch HJLange SSchmidt C Comparison of leg compression stocking and oral horse chestnut seed extract therapy in patients with chronic venous insufficiency. Lancet. 1996;347292- 294Google ScholarCrossref
108.
Greeske KPohlmann BK Horse chestnut seed extract: an effective therapy principle in general practice: drug therapy in chronic venous insufficiency [in German]. Fortschr Med. 1996;114196- 200Google Scholar
109.
Schorr DMGruber UF Prophylaxis of thromboembolic complications in gynecological surgery [in German]. Geburtshilfe Frauenheilkd. 1977;37291- 296Google Scholar
110.
Lang W Studies on the percutaneous absorption of 3H-aescin in pigs. Res Exp Med (Berl). 1977;169175- 187Google ScholarCrossref
111.
Takegoshi KTohyama TOkunda K et al. A case of Venoplant-induced hepatic injury. Gastroenterol Jpn. 1986;2162- 65Google Scholar
112.
Voigt EJunger H Acute posttraumatic renal failure following therapy with antibiotics and beta-aescin [in German]. Anaesthesist. 1978;2781- 83Google Scholar
113.
Hellberg KRuschewski WDe Vivie R Drug induced acute renal failure after heart surgery [in German]. Thoraxchir Vask Chir. 1975;23396- 399Google Scholar
114.
Lang W Dialysability of aescin [in German]. Arzneimittelforschung. 1984;34221- 223Google Scholar
115.
Grob PJMuller-Schoop JWHacki MAJoller-Jemelka HI Drug-induced pseudolupus. Lancet. 1975;2144- 148Google ScholarCrossref
116.
Walli FGrob PJMuller-Schoop J Pseudo-(venocuran-) lupus: a minor episode in the history of medicine [in German]. Schweiz Med Wochenschr. 1981;1111398- 1405Google Scholar
117.
Bouskela ECyrino FZMarcelon G Possible mechanisms for the inhibitory effect of Ruscus extract on increased microvascular permeability induced by histamine in hamster cheek pouch. J Cardiovasc Pharmacol. 1994;24281- 285Google ScholarCrossref
118.
Bouskela ECyrino FZMarcelon G Possible mechanisms for the venular constriction elicited by Ruscus extract on hamster cheek pouch. J Cardiovasc Pharmacol. 1994;24165- 170Google ScholarCrossref
119.
Bouskela ECyrino FZMarcelon G Effects of Ruscus extract on the internal diameter of arterioles and venules of the hamster cheek pouch microcirculation. J Cardiovasc Pharmacol. 1993;22221- 224Google ScholarCrossref
120.
Berg D Venous constriction by local administration of ruscus extract [in German]. Fortschr Med. 1990;108473- 476Google Scholar
121.
Berg D Venous tonicity in pregnancy varicose veins [in German]. Fortschr Med. 1992;11067- 6871- 72Google Scholar
122.
Weindorf NSchultz-Ehrenburg U Controlled study of increasing venous tone in primary varicose veins by oral administration of Ruscus aculeatus and trimethylhespiridinchalcone [in German]. Z Hautkr. 1987;6228- 38Google Scholar
123.
Zhou ZYJin HD Clinical Manual of Chinese Herbal Medicine. New York, NY Churchill Livingstone Inc1997;
124.
Chen ZY Use of Xin Bao in the treatment of 87 patients with sick sinus syndrome. Chung Hsi I Chieh Ho Tsa Chih. 1990;10529- 531Google Scholar
125.
Jones TKLawson BM Profound neonatal congestive heart failure caused by maternal concumption of blue cohosh herbal medication. J Pediatr. 1998;132550- 552Google ScholarCrossref
JAMA Internal Medicine
JAMA Network
Help
© 2022 Amer
Comments
Post a Comment