ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Hepatotoxicity due to herbal medications and dietary supplements

Hepatotoxicity due to herbal medications and dietary supplements
Literature review current through: Jan 2024.
This topic last updated: Nov 15, 2023.

INTRODUCTION — Complementary and alternative medical (CAM) therapies encompass a diverse group of practices that include the use of herbal and dietary supplements (HDS) (table 1) [1]. The use of CAM therapies in the United States and worldwide has been increasing steadily; however, the true prevalence is unknown [2]. In the United States, sales of CAMs in 1994 were $4 billion and increased to $14.8 billion (2008), to $30.2 billion (2012), and over $40 billion by 2019 [3-7]. The most commonly used CAM therapies are herbal and dietary supplements. Survey data have suggested that use of HDS ranges from 10 to over 75 percent depending on the country [8-13]. Up to 20 percent of adults report regular consumption of HDS [5,7,14]. In the United States in 2019, consumers spent over $9.6 billion on herbal products [2]. The most common reason reported for their use is for health improvement and health maintenance [15]. Women, persons younger than age 65 years, and individuals with a higher annual household income have been reported to be more likely to use CAM therapies [1,3,5,16,17].

Use of herbal preparations can be traced back thousands of years to ancient Egypt, China, India, and Sumeria, and formulations have been expanded upon over the centuries [18,19]. Many patients consider herbal remedies to be safe and completely free of unwanted side effects [20]. This is concerning since herbal products have biologic activity that can lead to severe hepatotoxicity or that interact with one another or with prescription medications. Fewer than 40 percent of patients disclose to their clinician that they are using these products, either alone or in conjunction with conventional drugs [1,3,21].

DATABASE OF DRUGS, HERBS, AND SUPPLEMENTS ASSOCIATED WITH HEPATOTOXICITY — It is estimated the number of herbal and dietary supplements (HDS) on the market is over 80,000 [6]. Over 1000 medications and herbal products have been implicated in the development of drug-induced liver injury, and the list continues to grow [13,22]. A searchable database of drugs, herbal medications, and dietary supplements associated with hepatotoxicity has been developed by the National Institutes of Health (NIH) [23]. Additionally, the NIH has developed a dietary supplement label database, containing over 165,000 product labels [24].

REGULATION — Regulation of herbal and dietary supplements (HDS) varies by country. In 1962, the US Kefauver-Harris Drug Amendment required that all over-the-counter and prescription products show proof of safety and efficacy [25]. Complementary and alternative medicines (CAMs) were assigned to the food supplement category and had a lower threshold of required evidence for safety [25]. An attempt by the US Food and Drug Administration (FDA) to develop stricter regulations was met with significant opposition by supplement manufacturers, lobbyists, and consumers.

The Dietary Supplement Health and Education Act (DSHEA) of 1994 was an amendment to the US Federal Food, Drug, and Cosmetic Act (FD&C) [26]. The DSHEA defines a dietary ingredient as a vitamin; mineral; herb or other botanical; amino acid; enzyme, organ or glandular tissue; dietary substance for use by humans to supplement the diet by increasing the total dietary intake; or a concentrate, metabolite, constituent, extract, or combination of the preceding substances [18,27]. These products may be in the form of tablets, capsules, soft gels, gel caps, liquids, teas, or powders. Medical foods are also covered under this regulation. The United States Pharmacopeia–National Formulary (USP–NF) is the official reference for herbal preparations [26]. The USP is an official public standards-setting authority for all prescription and over-the-counter medicines and other health care products manufactured or sold in the United States [28]. The USP also sets standards for food ingredients and dietary supplements.

Products in distribution prior to 1994 were allowed to remain on the market; their use was justified based upon experience from trial and error ("generally recognized as safe"), not rigorous scientific scrutiny. The FDA requires that manufacturers and distributors who wish to market products with "new dietary ingredients" notify the FDA about those ingredients. A new dietary ingredient is one that was not marketed prior to passage of the DSHEA in 1994. Manufacturers are responsible for reporting any side effects and ensuring that their product has not been adulterated. The FDA monitors product information, such as labeling and claims, and also monitors safety through a voluntary dietary supplement adverse event reporting system. Over the years, the FDA has issued warnings on products that have been reported to have hepatotoxicity [29]. The Federal Trade Commission (FTC) regulates advertising of dietary supplements [30].

The DSHEA requires that manufacturers establish product safety before marketing. Manufacturers must also ensure that the product label is truthful and not misleading. The label must contain a complete list of all ingredients contained in the product and the identity of the manufacturer. Manufacturers must also limit the claims that can be made (health claims, nutrient content claims, and structure/function claims) [31]. Structure/function claims are those which claim that the nutrient or dietary ingredient is intended to affect normal structure or function in humans (ie, "calcium builds strong bones"). They may also describe the means by which a nutrient or dietary ingredient acts to maintain such structure/function (ie, "fiber maintains bowel regularity"), or they may describe general wellbeing from consumption of the nutrient or dietary ingredient. Structure/function claims may also express a benefit related to a nutrient deficiency disease (ie, "vitamin C prevents scurvy"). In this last setting, they must include a statement telling how widespread the disease is in the United States. If a manufacturer makes a structure/function claim, the DSHEA requires a disclaimer: "This statement has not been evaluated by the FDA. This product is not intended to diagnose, treat, cure, or prevent any disease."

The Final Rule for Current Good Manufacturing Practices for Dietary Supplements was passed in 2007 as a further step in attempting to ensure the safety of HDS [32]. This rule requires that manufacturers follow certain standards in the production of HDS. Manufacturers are required to verify the contents of the product (ie, identify the ingredients, strength) and ensure that the product is contamination free. However, the Final Rule does not address the safety or efficacy of the product ingredient itself.

The DSHEA requires that a product's label accurately reflect its contents. However, there may be significant discrepancies between the ingredients listed on the label and the actual contents of the preparation. This is especially true of preparations containing multiple plants or herbs (ie, Chinese remedies), which may be adulterated either unintentionally or intentionally by cheaper, more toxic herbs, heavy metals, microbials, and pharmaceutical medicines (such as acetaminophen, aspirin, benzodiazepines, sildenafil, or steroids) [20,33-48]. As many as one-third or more of products can be adulterated [37,49-51]. There can also be variations between batches of the same herbal product from the same manufacturer [33,52-54]. It can be difficult to identify the agent responsible for the hepatotoxicity [55].

The widespread use of mixed remedies and the lack of randomized trials make any review of the safety and efficacy of herbal remedies difficult. In addition, conformity to the USP–NF standards is voluntary and there are no regulatory standards for HDS. There has been a push in the United States to force major policy changes in oversight of the HDS industry as well as grant the FDA the ability to act against manufacturers providing misleading information. Both proposals have been met with resistance from the HDS industry. The European Food Safety Authority (EFSA) monitors claims made on herbal products [56]. Directive 2004/24/EC in the European Union requires that manufacturers register products and comply with specific manufacturing and quality standards [57]. Across the world, regulation and safety monitoring remain limited, and the World Health Organization (WHO) has developed a Traditional Medicine Strategy [58].

FREQUENCY OF HEPATOTOXICITY — Precise estimates of the frequency of hepatotoxicity attributable to herbal and dietary supplements (HDS) are unknown. Patients often do not report the use of herbal products to their clinicians [3,45]. As a result, hepatotoxicity associated with herbal use may be missed. The United States Drug-Induced Liver Injury Network (DILIN) found that 20 percent of cases of drug-induced liver injury (DILI) were attributed to HDS and HDS are the second most common class of agent to cause DILI [6,59-62]. Additionally, this proportion of DILI caused by HDS increased from 7 to 20 percent between 2004 and 2013 [59]. Other studies, depending on the geographic location and type of data collection, have implicated HDS in up to 73 percent of cases of DILI [49,57,59,63-73]. It has been estimated that fewer than 1 percent of adverse reactions to dietary supplements are reported [74].

DIAGNOSIS — Patients are often reluctant to discuss use of herbal and dietary supplements (HDS)/complementary and alternative medicines (CAMs) or do not recognize them as medications [45]. It is important for the clinician to discuss the use of these products with the patient in a nonjudgmental manner. Patients can then be educated on the potential adverse effects and herb-herb or drug-herb interactions that may occur. This may prevent the development of hepatotoxicity. However, the unreported use of herbal remedies must be considered as a possible etiology in any setting of clinical manifestations of liver injury.

The diagnosis of herb-induced liver injury (HILI) can be difficult. Clinical history may be helpful but is not always reliable. The relationship between the exposure and hepatic toxicity (latency) is not always clear and is quite variable. Patients may be taking multiple preparations or multi-ingredient products, making identification of a single offending agent impossible. Due to poor regulation of these products, HDS are prone to intentional or inadvertent adulteration with other ingredients, making identification of the responsible agent difficult [75]. Poor adherence to good manufacturing practices and to quality control contribute to risk of hepatotoxicity [76-78]. Many cases of hepatotoxicity resulted following ingestion of herbal products that contained untested combinations of products, adulterated products, or both [57]. Ingredients found in some HDS products have included unwanted chemicals, prescription medications, heavy metals, and pesticides [75]. HDS products may also vary in composition from batch to batch depending on manufacturing practices. These products are also frequently mislabeled [79]. Patients may also have confounding concomitant liver disease, such as alcohol-associated liver disease or metabolic dysfunction-associated steatohepatitis (MASH), which can produce similar clinical and laboratory features. There is no gold standard, and no specific tests or serum biomarkers exist to confirm a diagnosis of drug-induced liver injury (DILI) from either a prescription medication or an herbal remedy. Proving that an HDS causes HILI relies on both chronologic and clinical criteria [80,81].

A diagnosis of HILI should be suspected in patients with nonspecific symptoms developing after introduction of an HDS (such as nausea, anorexia, malaise, fatigue, right upper quadrant pain, or pruritus), and such symptoms prompt further evaluation. Features suggesting HILI include lack of illness prior to ingesting the HDS, clinical illness or biochemical abnormalities developing after beginning the offending agent, and improvement after the product is withdrawn. Patients may present with asymptomatic liver enzyme elevations, acute or chronic hepatitis (symptomatic malaise, nausea, vomiting, abdominal pain, etc), acute liver failure with coagulopathy and encephalopathy, or with signs and symptoms of cirrhosis. Jaundice is the most common presentation (46 percent) followed by abdominal pain and nausea [82]. Serum aminotransferases may be markedly elevated (up to >20 times the upper limit of normal). The clinical patterns of liver injury are similar to those seen with other forms of DILI. These patterns include hepatocellular, cholestatic, a mixture of the two, or vascular (ie, sinusoidal obstruction syndrome).

Hepatocellular injury is more frequently seen with HILI than with DILI, secondary to conventional medications [6,29,65,83]. If an immunologic reaction is suspected, the illness will generally recur upon reintroduction of the offending substance. For patients with suspected HILI, rechallenge is not advised. Unlike the more stereotypic presentations seen with hepatotoxicity due to most drugs, the clinical presentations of injury secondary to HDS are less typical and have been less well-defined [29]. Exceptions to this are hepatotoxicity related to anabolic steroids and pyrrolizidine alkaloid-containing products. The most common presentation is hepatocellular injury [84]. Female patients and patients presenting with a hepatocellular pattern of injury have a poorer prognosis [67,85,86]. Bile duct loss in the setting of cholestatic pattern of injury is also predictive of a poor outcome [84,87]. (See "Drug-induced liver injury".)

Liver biopsy may be useful if the diagnosis of HILI is uncertain. However, liver histology will not identify a specific product. Studies have reported that among patients who underwent liver biopsy, over 70 percent of patients have hepatocellular injury, followed by sinusoidal obstruction syndrome (approximately 10 percent) and cholestatic liver injury (9 percent) [82].

Continued use of the offending product increases morbidity once hepatotoxicity has developed [34,88]. The patient may even increase the use of the herbal preparation to help manage the new symptoms, further worsening the liver injury. For patients with elevated liver enzymes, other causes of liver disease must be ruled out. Patients often use herbal remedies to treat their chronic disease and may, in fact, exacerbate it [4,5,52]. Continued ingestion of the herbal preparation in the face of ongoing hepatotoxicity may lead to acute liver failure, sinusoidal obstruction syndrome, or cirrhosis [65,89].

The Council for International Organizations of Medical Sciences (CIOMS) developed a series of standard designations for drug-induced liver disorders and classification of injury [90]. The US Food and Drug Administration Drug Hepatotoxicity Steering Committee proposed modifications to the CIOMS classification scheme for classifying hepatotoxicity in clinical trials (table 2) [91]. These guidelines are used as markers of hepatotoxicity in clinical trials.

A number of scales have been developed that attempt to codify causality of drug toxicity into objective criteria [92]. The best-known scale is the Roussel Uclaf Causality Assessment Method (RUCAM) scale. Other scales, including the Maria & Victorino scale and the Naranjo scale, are simpler clinical diagnostic scales [81,93-95]. Studies that have compared these models suggest that the RUCAM scale may have better discriminative ability [80,92,96]. While these scales have not been validated in the setting of herbal medications, they may be useful (table 3). Another scale in development, the Revised Electronic Causality Assessment Method (RECAM), updates and expands upon the RUCAM [97].

None of these scales address all risk factors, and none are used routinely in clinical practice [81]. In addition, none were developed specifically for HDS-induced hepatotoxicity. One of the most important diagnostic criteria remains the exclusion of other causes of liver injury. RUCAM has been validated independently in the setting of drug rechallenge [94]. The Drug-Induced Liver Injury Network (DILIN) developed the DILIN Causality Scoring System to adjudicate the causality of drug-induced injury for patients enrolled into its prospective clinical trial [98]. This model relies on structured expert opinion, and when compared with the RUCAM, it produces higher agreement rates and likelihood scores. However, intraobserver variability remains significant. Unfortunately, the DILIN scale is not a clinically viable option for determining causality since it relies on expert opinion.

Key elements for attributing liver injury to an HDS are similar to those for other drug products and include:

Exposure must precede the onset of liver injury (although the latent period is highly variable).

Underlying liver disease should be excluded.

Injury may improve when the HDS is stopped (although, in some cases, injury may initially worsen for days or weeks while, in cases of acute liver failure, declining liver biochemical tests may indicate deterioration rather than improvement).

Liver injury may have recurred more rapidly and severely after repeated exposure.

None of the causality assessments models have been validated in the setting of HDS hepatotoxicity. They are less reliable given the possibility of product variability and contamination [84].

TREATMENT — Early recognition of toxicity is important to permit assessment of severity and monitoring for acute liver failure. The mainstay of therapy for herb-induced liver injury is withdrawal of the offending toxin. (See "Acute liver failure in adults: Etiology, clinical manifestations, and diagnosis".)

Glucocorticoids are of uncertain benefit for most forms of herbal and dietary supplement (HDS) hepatotoxicity, although they may have a role for treating patients with hypersensitivity reactions [99]. Our practice is to give glucocorticoids to patients with hypersensitivity reactions who have progressive cholestasis despite stopping HDS and who have biopsy features that resemble those seen in autoimmune hepatitis. In addition, we give glucocorticoids to patients with extrahepatic manifestations of a hypersensitivity reaction that warrant glucocorticoid treatment (eg, severe pulmonary involvement in patients with drug reaction with eosinophilia and systemic symptoms). (See "Overview of autoimmune hepatitis", section on 'Histology' and "Drug reaction with eosinophilia and systemic symptoms (DRESS)", section on 'Management'.)

Patients should be followed by serial biochemical measurements. Hepatology consultation may be warranted.

Recovery should be expected in the majority of patients after discontinuing the HDS. More advanced disease requires supportive therapy, with acute liver failure often leading to death or need for liver transplantation. In this setting, mortality can be over 80 percent without transplantation. The development of jaundice (bilirubin greater than two times the upper limit of normal, in the setting of an alanine aminotransferase greater than three times the upper limit of normal) following introduction of an HDS potentially portends a poor outcome and should also prompt immediate referral to a center with expertise in hepatology [67,100,101]. Patients who recover from HDS-induced hepatotoxicity generally have a favorable prognosis.

In the United States, the US Food and Drug Administration (FDA) records HDS toxicity. Cases of HDS hepatotoxicity should be reported to 1-800-332-1088 or https://www.fda.gov/safety/reporting-serious-problems-fda/reporting-health-professionals.

SPECIFIC HEPATOTOXIC HERBS — The many reports in the literature of the toxic effects of herbal remedies underscore the view that not all natural products are harmless. Hepatotoxicity may be the most frequent adverse reaction to herbal remedies [102,103]. It is often difficult to pinpoint the product that has caused the injury since most products do not contain a single ingredient. Plant extracts are regarded as multi-ingredient products since the product is a concentration of phytochemicals from the plant [7]. Some of the more frequently reported offending herbs include Chinese remedies and teas (Jin Bu Huan [104,105], Ma-Huang [106-108]), germander [89,109-114], valerian [115], mistletoe [116], skullcap [115-117], chaparral [118-122], comfrey [123-127], herbal teas containing toxic alkaloids [125,128-130], pennyroyal oil [131], and kava (table 4) [103,132-136]. Although liver abnormalities are frequently transient, cases of chronic liver disease and acute liver failure have been described [20,136,137].

The list that follows includes some of the herbs that have been associated with hepatotoxicity. However, the list of herbal medications, dietary supplements, and medications associated with hepatic injury is long. A more complete listing can be found in the searchable database of drugs, herbal medications, and dietary supplements developed by the National Institutes of Health [138].

Pyrrolizidine alkaloids — Pyrrolizidine alkaloids (PAs) are a group of naturally occurring alkaloids based on the structure of pyrrolizidine. More than 660 PAs and PA N-oxides have been identified in over 6000 plant species throughout the world (3 to 5 percent of the world's flowering plants) (table 5) [139,140]. They are used for a variety of conditions, including arthritis, gout, and infections. Over 50 percent of PAs exhibit hepatotoxicity [129,140]. One of the historical examples of hepatotoxicity was reported following the use of "bush tea." Bush tea was brewed predominantly from the Senecio plant, which is high in PAs.

Other plant families that have shown hepatotoxicity after exposure include: Boraginaceae (Heliotropium, Trichodesma, Symphytum officinale [Comfrey]), Compositae (Senecio [Bush Teas], Eupatorium), Crotalaria (Leguminosae), Echinacea (Echinacea purpurea) [141], Germander (Teucrium chamaedrys), Greater Celandine (Chelidonium majus), and Scrophul-ariaceae (Castilleja) [123-125,128-130,142-144]. PA poisoning is common in areas where traditional remedies are frequently used (such as Africa and India) [145,146]. In Western countries, it is seen more sporadically. Comfrey has been removed from the market in France following numerous reports of liver damage. It remains widely available in the United States despite US Food and Drug Administration (FDA) requests to remove comfrey products from the market [125,147].

Alkaloid toxicity usually results in moderate to severe liver damage, particularly to the hepatic central vein and sinusoidal endothelium. Hemorrhagic necrosis, hepatomegaly, ascites, and endothelial cell proliferation are also seen. This leads to occlusion of the hepatic veins, resulting in hepatic sinusoidal obstruction syndrome (formerly veno-occlusive disease), a progressive form of portal hypertension, which often progresses to hepatic failure [148,149]. Sinusoidal obstruction syndrome is the characteristic histologic feature of PA hepatotoxicity [144]. (See "Hepatic sinusoidal obstruction syndrome (veno-occlusive disease) in adults".)

The precise mechanism of hepatic injury is unknown but appears to result from accumulation of highly reactive electrophilic metabolites produced via the cytochrome P450 (CYP450) enzyme system [20,123,131]. The increased concentration of P450 enzymes within the hepatic centrilobular region correlates with the changes seen histologically. The pyrrolic and pyrrole-like metabolites are highly reactive and lead to the formation of deoxyribonucleic acid (DNA) and protein adducts as well as glutathione conjugates [144,150]. The damage is compounded by depletion of glutathione or induction of CYP450 3A4 [18,151]. PAs have also been shown to induce apoptosis [151,152].

Nonthrombotic luminal occlusion of the small centrilobular veins leads to hepatic congestion and subsequent hemorrhagic parenchymal necrosis [20,153]:

With acute injury, patients typically present with sudden onset abdominal pain, jaundice, conjugated hyperbilirubinemia, and elevated aminotransferases. Hepatomegaly and ascites are common.

The subacute and chronic forms are more insidious and may mimic cirrhosis and portal hypertension from other causes. However, histology shows a distinctive pattern of centrilobular injury [148].

Hepatotoxicity from PAs is dose and time dependent and reproducible [154]. Approximately one-half of patients with hepatotoxicity develop limited lesions and generally proceed to complete recovery once the offending herb has been discontinued [148,154]. More extensive disease can lead to cirrhosis, hepatic failure, and death [20,154]. The acute form is rapidly fatal in 20 to 40 percent of patients (with worse prognosis in adults compared with children). Approximately 15 percent with acute disease will progress to subacute or chronic injury, succumbing within several years to end-stage liver disease. Of the remainder, most develop cirrhosis and portal hypertension [34,148,154,155]. Death may occur two weeks to more than two years after poisoning, but patients may recover almost completely if the herb intake is discontinued and liver damage is not severe.

Ayurvedic herbs — Ayurvedic medicine originated in ancient India. These herbal and dietary supplement (HDS) products have been used most commonly on the Indian subcontinent in the practice of traditional medicine (Ayurveda), but their use is expanding worldwide. Ayurveda uses many medicinal plants that contain toxic pyrrolizidine alkaloids [156,157]. They include Psoralea corylifolia, Centella asiatica, Acacia catechu, Eclipta alba, and Vetivexia [66,157-159]. Both acute and chronic hepatitis have been reported. Additionally, many of these products have been found to be adulterated with heavy metals, including lead, mercury, or arsenic [46,160]. Ashwagandha (Withania somnifera) is popular in Ayurvedic medicine. Although generally considered safe, there have been reports of cholestatic liver injury following its ingestion [161,162].

Germander — The blossoms of germander (Teucrium chamaedrys) have been used for thousands of years for a variety of symptoms [163]. It is used in tea form or alcohol-based extracts. The herb is said to have choleretic properties, although this has never been scientifically proven. It is also used for dyspepsia, hypertension, diabetes, gout, and obesity [139]. Liver injury can present as acute hepatitis, chronic hepatitis, or acute liver failure [89,109,110,163,164]. The herb has been removed from the market in France because of its hepatotoxicity [112,113]. The majority of cases of hepatotoxicity have occurred at the manufacturers' recommended doses (600 to 1600 mg/day). Clinical symptoms generally develop after approximately two to three months of ingestion and include a nonspecific hepatitis, which generally runs a benign course [20]. Typical features include anorexia, nausea, abdominal pain, and jaundice associated with a marked elevation in serum aminotransferases. Complete recovery is usually seen within two to six months following discontinuation of the herb. Fatal cases have been described, as have more insidious presentations with progression to cirrhosis [18,20,89,111]. Cirrhosis generally develops in patients who have ingested the offending herb for longer periods of time or in larger quantities.

Germander contains toxic alkaloids, the furanic neoclerodane diterpenes (NCDs), which are oxidized by the CYP450 3A4 enzyme system into hepatotoxic reactive metabolites [109,165]. The toxic metabolites accumulate and covalently bind to cellular proteins, which in association with the depletion of the free-radical scavenger glutathione, leads to acute centrilobular hepatocyte necrosis in vivo. In vitro studies have revealed progressive apoptosis and cytoskeletal disorganization [165].

Poley (Teucrium polium) is in the same genus as germander and is used for diabetes (in Israel), gastric complaints (in North Africa), fever (in Italy), and as a poultice (in Spain). It has also been linked to hepatotoxicity and acute liver failure [114,166,167]. Teucrium capitatum and T. viscidum have also been reported to cause hepatotoxicity [168,169].

Greater celandine — Greater celandine (Chelidonium majus) is a member of the poppy family that grows mainly in Europe and Asia but has been introduced into North America. It is considered a cleansing herb and is said to stimulate pancreatic enzymes and bile. In fact, it has been shown to increase biliary flow in experimental models [170]. Therefore, it is often used for hepatobiliary (jaundice, hepatitis, and gallbladder) and digestive tract (dyspepsia, gastroenteritis, irritable bowel syndrome, constipation, anorexia, stomach cancer, intestinal polyps) complaints [171]. The plant contains over 20 alkaloids [171]. When used orally, it has been implicated in numerous cases of liver injury [143,171-178]. Injury generally develops after one to six months of use. Jaundice is generally the predominant symptom, but anorexia, nausea, vomiting, and abdominal pain and itching have been reported. Liver enzyme elevations range from 100 to over 4000 units/L, and injury is hepatocellular, resembling acute viral hepatitis in the majority of cases [178]. However, cholestatic hepatitis has also been reported [143,172]. The mechanism of hepatotoxicity is unclear. Liver injury appears to show a female predominance [178,179]. Histologic examination of the involved liver has suggested metabolic hepatotoxicity [143,172,178]. There have been no reports of fatalities and discontinuing generally leads to resolution of liver injury over two to six months. Greater celandine has been banned from use in many European countries.

Chaparral — Chaparral (Larrea tridentata) is indigenous to the southwestern United States and is derived from the leaves of the creosote bush (greasewood). It is said to have antioxidant, antiamebic, antibacterial, antifungal, and antiviral properties. It is used for diarrhea, cramps, chest pain, upper respiratory tract infections, rheumatism, cutaneous venereal lesions, chronic skin disorders, and weight loss [154]. It has been popular among patients with human immunodeficiency virus (HIV) infection [180]. It is available in the form of tea, capsules, tablets, and salves [118]. Scientific evidence regarding chaparral's effectiveness is lacking.

Patients with chaparral-induced hepatotoxicity typically present with acute hepatitis [118]. Marked elevation in the serum aminotransferase and bilirubin values may be seen. Alkaline phosphatase may be mildly elevated. Acute hepatocellular injury of varying degrees of severity has been reported, including mild cholestatic hepatitis, severe collapse of parenchymal architecture, hepatic fibrosis, and massive liver failure requiring transplantation [118-122,181]. The onset of disease occurs 3 to 52 weeks after the ingestion and resolves in 1 to 17 weeks in most cases [118]. Progression to cirrhosis or acute liver failure requiring liver transplantation has been reported [118]. There have been no reported cases of chaparral hepatotoxicity published in over 10 years [138].

Atractylis gummifera and callilepis laureola — Atractylis gummifera grows in the Mediterranean and is used as an antipyretic, emetic, abortifacient, and diuretic, and hepatotoxicity continues to be reported [20,182,183]. Toxicity is secondary to two diterpenoid glucosides: atractyloside and carboxyatractyloside [184]. These toxins inhibit mitochondrial oxidative phosphorylation by interacting with a mitochondrial adenine nucleotide translocator, inducing oxidative stress [184,185]. The onset of toxicity is rapid, occurring within hours after ingestion. It often leads to acute liver and kidney failure and subsequent death.

Callilepis laureola (Impila) is native to South Africa [186,187]. Like A. gummifera, it contains atractyloside. Its toxicity has been linked to depletion of cellular glutathione. It has a similar rapid onset of action, and mortality rates are over 90 percent by five days after ingestion. There have been reports that supplementation with N-acetylcysteine may reduce its toxicity [186].

Pennyroyal — Pennyroyal tea and leaf extracts are generally considered to be safe. However, pennyroyal oil (Mentha pulegium or Hedeoma pulegoides) is a highly toxic agent that can cause both hepatic and neurologic injury even in small doses (ie,15 mL) [138]. It is frequently used to flavor herbal remedies and teas (particularly mint teas) and is often used for digestive disorders, liver disorders, amenorrhea, gout, colds, increased micturition, and for skin diseases. It also continues to be used as an abortifacient. Acute toxicity leads to gastrointestinal distress and central nervous system effects within one to two hours of ingestion, and fatal ingestions can occur with ingestion of >15 mL of the oil [188].

Pennyroyal oil contains 85 percent pulegone. CYP450 1A2 and 2E1 produce oxidative metabolites (menthofuran) of pulegone that are associated with severe hepatic toxicity with centrilobular degeneration and massive hepatocellular necrosis [154]. These metabolites bind to cellular proteins and deplete hepatic glutathione [131,189,190]. The use of N-acetylcysteine may help minimize hepatic injury [188].

Mistletoe — Mistletoe is used in traditional medication and refers to two unrelated plants: American mistletoe (Phoradendron leucarpum) and European mistletoe (Viscum album). They are purported to be effective in treating asthma, epilepsy, arteriosclerosis, cardiac arrhythmia, cramps, blood loss, nervous conditions, high blood pressure, joint pain, dizziness, and infertility [116]. There have been case reports of hepatitis [116]. Lectins in mistletoe products induce apoptosis and can also stimulate the immune system [29]. However, controversy remains as to whether mistletoe actually causes liver disease. Both appear to be safe at conventional doses and are an unlikely cause of symptomatic liver injury [138]. In reported cases, the ingested capsules were also laced with Skullcap (Scutellaria galericulata). Skullcap and Valerian (Valeriana officinalis) have been implicated in liver injury, although there are no experimental data supporting their toxicity [20,115].

Kava-kava — Kava (Piper methysticum) is used as a dietary supplement and has been used in the South Pacific for centuries for complaints of anxiety, sleeplessness, and menopausal symptoms. Numerous reports of severe hepatotoxicity and liver failure have been described in Europe and the United States [103,132-136,191-193]. Hepatotoxicity has been reported within a few weeks to up to two years (average 2 to 24 weeks) following ingestion [191].

Clinically, reports of toxicity range from transient elevation in aminotransferases to acute liver failure and death [103,138]. Liver injury has been characterized histologically with hepatic necrosis, cholestasis, and lobular inflammation. The mechanism of hepatotoxicity is unclear, but multiple hepatotoxic components have been identified, including pipermethystine and flavokavain B [194,195]. The aerial portions of the plant contain the toxic alkaloid pipermethysticin and are generally not consumed [194,195].

Based upon case reports of serious toxicity, the FDA issued a consumer advisory regarding this toxic herbal product. Details of the advisory are available online. The product has been banned from many European markets (including Germany, France, Great Britain, and Switzerland). The link of toxicity to kava remains debated [194]. Some propose that the aqueous preparation is safe [196] while others have shown that even this preparation leads to hepatotoxicity [197].

Green tea extracts — Green tea is widely consumed, is safe in moderation, and may have beneficial effects. Concentrated extracts of green tea (Camellia sinensis L.) have been associated with hepatocellular injury, particularly in high doses (>800 mg) or prolonged exposure [7,198-201]. Numerous supplement extracts are available and lead to heightened exposure to the catechin polyphenols. These extracts have been used in multiple weight-loss products, many of which have been removed from the market due to toxicity.

The precise mechanism of hepatotoxicity remains unclear. The extraction process increases the catechin content as well as potential contaminants [7]. Catechins such as epigallocatechin-3-gallate (EGCG) are likely to be involved. EGCG constitutes over 50 percent of the catechin content. EGCG induces reactive oxygen species similar to other herbal products [202]. This may overwhelm hepatic drug-metabolizing capability [201]. A genetic predisposition may also predispose to injury (HLA-B*35:01, C*04:01), and females are more susceptible than males [198,203,204]. Hepatotoxicity is increased in those who are fasting [141,205]. The pattern of injury is predominantly hepatocellular, but mixed and cholestatic presentations have been reported [6,202,206]. Liver injury generally develops within three months of use and usually resolves with discontinuation of the product; however, fatalities have been reported. The United States Pharmacopeia–National Formulary has proposed a cautionary labeling requirement on products with green tea extract, which has not been implemented [201].

Anabolic (muscle-building) supplements — Anabolic steroid use has been associated with many types of liver injury, including hepatitis, intrahepatic cholestasis, peliosis hepatis, hepatocellular adenoma, and hepatocellular carcinoma [207,208]. Selective androgen receptor modulators (SARMs) are found in some supplements marketed for muscle-building, and they mimic the effects of testosterone and anabolic steroids [209]. Despite regulation and known adverse effects, including hepatotoxicity, use of these agents remains widespread, and patients continue to present with liver injury [210,211]. The classic presentation is one of hepatocellular injury with severe jaundice at a median of approximately 70 days following use [212]. Patients who present with markedly elevated bilirubin levels are also prone to acute kidney injury [29]. The FDA has issued public health advisories warning consumers to avoid body-building products containing anabolic steroids or steroid-like compounds [213,214]. (See "High-risk dietary supplements: Patient evaluation and counseling", section on 'Sports supplements'.)

Black cohosh — Black cohosh (Cimicifuga racemose syn. Actaea racemosa) is indigenous to eastern North America. It is widely available, and the rhizomes and roots are commonly used for the treatment of peri- and postmenopausal symptoms. The product contains many compounds, including phenolic acids, flavonoids, volatile oils, triterpene glycosides, and tannins [141]. There have been multiple reports of hepatotoxicity and adulteration (particularly with Asian Cimicifuga species) [215,216]. Reported toxicity has been an autoimmune-like liver injury, and liver failure necessitating transplantation has been reported [141,215,217-220]. However, the causality between black cohosh and hepatotoxicity remains uncertain. Subsequent meta-analyses have suggested that this product was effective and safe [221,222].

Flavocoxid — Flavocoxid is a prescription medical food that is a blend of two flavonoids, baicalin and catechins (derived from the plants Scutellaria baicalensis and Acacia catechu, respectively). It has been used to treat osteoarthritis. The FDA recommended discontinuation of flavocoxid because of reports of serious adverse events, including drug-induced liver injury (DILI) [223]. In a prospective study of 877 patients, four patients (0.4 percent) developed liver injury [224]. The association between flavocoxid use and liver injury was deemed highly likely in three cases and possible in one. In all four cases, liver tests returned to normal within 3 to 12 weeks of discontinuing the flavocoxid.

Kratom — Kratom (Mitragyna speciosa) is native to Southeast Asia and has been traditionally used to treat diarrhea or for its stimulant effect. In the United States, most users take the product for pain, depression, or as an anxiolytic [225]. Its effects are secondary to mitragynine and 7-hydroxymitragynine effects on the mu-opioid (agonist) receptors. (See "Opioid withdrawal: Medically supervised withdrawal during treatment for opioid use disorder", section on 'Kratom'.)

Kratom has been associated with liver injury [226,227]. Liver injury may be severe; both hepatocellular and mixed patterns have been reported. Despite discontinuation of the product, bilirubin may continue to rise before normalizing. Kratom products have been banned in some countries [7]. The FDA attempted to classify Kratom as a schedule I drug, but lobbyists prevented this [227,228]. This has resulted in increasing reports of injury.

Cannabidiol (CBD) — CBD is a non-psychotropic, biologically active component of marijuana (Cannabis sativa). There has been little oversight into the manufacture of CBD products, and sale of both substandard and adulterated products have been reported [7]. Clinical trials with CBD noted that up to 20 percent of those taking it developed elevated liver enzymes, generally after several months of chronic use. The mechanism of hepatic injury remains unclear.

CHINESE HERBAL REMEDIES — Over 11,000 species of medicinal plants are used in China [141]. Use of traditional Chinese medicines (TCMs) has grown globally. Many TCMs have beneficial effects [36]. However, published reports on hepatotoxicity with TCM have been increasing [36,103,104,108,229-232]. Evidence regarding the incidence and prevalence rates of hepatotoxicity with these products is uncertain [233]. Studies of drug-induced liver injury (DILI) that include TCMs report widely varying data, with TCMs accounting for 4 to 55 percent of cases, depending on the patient cohort [232]. Specific products include Ba Jiao Lian (Dysosma pleianthum), Bol Gol Zhee (Fructus psoraleae), Chi R Yun (Breynia officinalis), Jin Bu Huan (Lycopodium serratum), Jing Tian San Qi (Sedum aizoon syn. Stonecrop), Ma-Huang (Ephedra), Shan Chi (Gynura segetum), Sho Saiko To, and Shou Wu Pian (Polygonum multiflorum).

Much of herbal-induced liver injury (HILI) due to TCMs are believed to be due to herbs containing pyrrolizidine alkaloids [234]. TCMs that have been shown to cause sinusoidal obstruction syndrome include Jing Tian San Qi (Sedum aizoon syn. Stonecrop), Shan Chi (Gynura segetum), and Senecio vulgaris [235-237]. The TCM Shi Can contains Teucrium chamaedrys (Germander) or other Teucrium species, which are a known cause of HILI [89].

The clinical course for patients with hepatotoxicity ranges from elevated liver enzymes [229,230] to acute liver failure and death or need for liver transplantation [137]. Up to 70 percent of these products produce reactive metabolites that cause hepatotoxicity [238]. It is often difficult to identify the agent responsible for the hepatotoxicity since most Chinese preparations are either mixtures of multiple herbal preparations (typically four to six different herbs) or have been adulterated with substituted herbs, heavy metals, or pharmaceutical medicines [20,33,35,37-40,50,51,66]. Generally, a single herb is considered primary (the "king" or "sovereign"), while the additional herbs are thought to act synergistically with the main product or to modify its toxicity [13]. Toxicity is generally observed with preparations that contain aconitine, podophyllin, or anticholinergics [230].

Jin Bu Huan — Jin Bu Huan (Lycopodium serratum) has been used for over 1000 years as a sedative and analgesic [104,163,164]. It has morphine-like properties due to the alkaloid levo-tetrahydropalmatine. It was banned for use in the United States in the 1990s following multiple reports of toxicity [104,105,239].

Both acute and chronic hepatotoxicity have been reported [104,105,239]. Patients with hepatotoxicity present at a mean of 20 weeks (7 to 52 weeks) after ingestion with acute hepatitis associated with fever, nausea, vomiting, fatigue, pruritus, abdominal pain, hepatomegaly, and jaundice. Plasma aminotransferase levels are significantly elevated. The liver biopsies that have been performed appear consistent with a DILI, demonstrating eosinophils and focal hepatocellular necrosis [104,105]. Fibrosis and microvesicular steatosis have also been reported [104].

Symptoms resolve over a mean of eight weeks (2 to 30 weeks) with discontinuation of the herb and have promptly recurred when patients were rechallenged [104,105]. The hepatotoxic mechanism is not known but may be immunologic given the histologic presentation [163].

Ma-Huang (ephedra) — Ma-Huang (Ephedra sinica), an ephedra alkaloid, has been used for centuries for cough, bronchitis, fever, joint symptoms, edema, bone pain, to induce weight loss, and as a central nervous system stimulant [106,108]. The active ingredient, ephedrine, has been associated with psychiatric and autonomic system adverse events. Overall, the cardiovascular and neurologic side effects are more common than liver-related effects. Use of the drug is associated with reports of hepatotoxicity, including hepatitis, acute liver failure, and exacerbation of autoimmune hepatitis [106-108,240,241]. The mechanism of toxicity is not known. Affected patients present in similar fashion to those taking Jin Bu Huan [106,108]. Biopsies revealed diffuse severe hepatocellular necrosis and polymorphonuclear cell infiltrates with occasional eosinophils. Abnormal liver tests usually normalize within four months of discontinuation of drug [241]. Ephedra was withdrawn from the United States market in 2004.

Syo-saiko-to — Syo-saiko-to (Dai-saiko-to, xiao-chai-hu-tang, da-chai-hu-tang, TJ-19) is an herbal product used in China to treat fever and gastrointestinal disorders. It has been used in Japan for treating chronic hepatitis, particularly hepatitis C virus infection. This remedy contains numerous herbs (Bupleurium root, Pinelliae tuber, Scutellaria baicalensis root, ginseng root, ginger rhizome, glycyrrhiza root, and jujube fruit). The components in this product differ in their proportion of herbal products. There is some evidence that these products have anti-inflammatory, antioxidant, antifibrotic, and antitumorigenic activity [242]. There have been multiple reports of hepatotoxicity associated with this product; however, some occurring up to three months after ingestion and recurring with rechallenge [243-245]. The offending component is suspected to be the Chinese skullcap (Scutellaria baicalensis). The histologic picture is one of centrilobular confluent or spotty necrosis, microvesicular steatosis, acidophil bodies, and possibly granulomas [243]. Patients should be cautioned against using this herbal remedy.

Chaso or Onshido — Chaso and Onishido are used for weight loss. The toxic ingredient in these formulations is thought to be N-nitroso-fenfluramine, which depletes adenosine triphosphate (ATP) and impairs mitochondrial oxidative phosphorylation [29]. This results in diffuse hepatic necrosis and nonspecific inflammation. Cases of acute liver failure, death, and liver transplantation have been reported [246].

Shui-zhi-zi or sansisi — These products contain Gardenia jasminoides and are used to treat fever, liver disease, or cancer. This product contains geniposide, which has been shown to have dose dependent hepatotoxicity [247].

Polygonum multiflorum — This product has been used to improve fertility and to enhance hair color. Reports of hepatoxicity have been reported and are frequently associated with the HLA-B*35:01 allele [231,248].

WEIGHT-LOSS PRODUCTS — Numerous proprietary preparations have been associated with hepatotoxicity, including Hydroxycut [249-254], Herbalife [44,255-260], Enzyte [261], N.O.-XPLODE [212], OxyELITE [262], SlimQuick [203], and Exolise [203,206].

Herbalife products — These products are sold worldwide in the form of drinks, tablets, capsules, and energy bars. There are multiple reports of hepatotoxicity with these products, ranging from mild to severe enough to require liver transplantation [44,255,257-259,263-265]. The pattern of liver injury is hepatocellular in the majority of cases; however, there are also reports of both mixed and cholestatic injury. The mechanism of toxicity remains unknown.

Hydroxycut — Hydroxycut is a weight-loss product that contains multiple herbal products. The original formulation contained ephedra and was withdrawn from the market in 2004.The product was reformulated, and 23 reported cases of liver injury were reported. In 2009, the US Food and Drug Administration (FDA) issued a warning regarding the product’s potential hepatotoxicity and recommended that consumers discontinue use of the products carrying the brand name Hydroxycut [253,263,266,267]. The manufacturer voluntarily recalled all of its products and again reformulated the herbal mix [263]. Reformulations have included different alkaloid products, including Camellia sinensis (117 mg [epigallocatchein-3-gallate [EGCG]), yohimbe extract, garcinia cambogia, and green tea extract [7]. Hepatotoxicity generally presents within the first three months of taking the product. Patients generally have hepatocellular injury with elevated liver enzymes; however, both cholestatic injury and mixed acute hepatitis with jaundice have been reported. Although infrequent, there are still reports of liver injury in the setting of Hydroxycut.

Supplements containing Garcinia cambogia — Extracts of garcinia contain xanthones, benzophenones, amino and organic acids, including hydroxycitric acid [138]. Dietary supplements containing Garcinia cambogia are widely marketed for weight loss. As their use has gained popularity in the United States, cases of acute hepatitis and acute liver failure requiring liver transplantation have been reported [268-270]. It has been postulated that the active ingredient, hydroxycitric acid, is the cause of liver injury. This ingredient was banned by the FDA because of hepatotoxicity. Additionally, the FDA has recommended that specific products containing Garcinia cambogia be avoided as they are often contaminated with sibutramine [271]. In a case report, sibutramine was associated with hepatotoxicity, and it was removed from the US market for cardiovascular toxicity [272]. (See "Obesity in adults: Drug therapy", section on 'Therapies not recommended'.)

OxyELITE Pro — The original product contained 1,3-dimethylamylamine (DMAA), which had been noted to have caused acute liver injury [273]. Following a report of seven patients who developed severe hepatitis or acute liver failure while taking the product OxyELITE Pro [274], the initial formulation was removed from the market in 2013. The formulation was changed by the manufacturer to remove DMAA and add aegeline [275]. Aegeline is extracted from the fruit Aegle marmelos (bael) and has been used in Ayurvedic medicine. Its main alkaloid is N-[2-hydroxy-2(4-methoxyphenyl) ethyl]-3-phenyl-2-propenamide [276]. Following its release, further cases of severe liver injury were reported [234,262,274]. A total of 55 cases of liver injury were reported with the new formulation [6,62,275]. The majority of those affected were young (mean age 33.9 years) and male (60 percent). Many patients required hospitalization and liver transplantation, and deaths were reported [262]. The FDA banned the product in October 2013 and the manufacturer issued a recall of all products in November 2013.

OTHER HEPATOTOXIC HERBS — There are many case reports of hepatotoxicity with other herbal preparations. These include Aloe (A. vera; A. barbadensis, A. arborescens), Bajiaolian, Borage (Borago officinalis), Broom Corn (Sorghum vulgare), Camphor oil, Centella asiatica (gotu kola), Margosa oil (Azadirachza indica), Cocaine, Coltsfoot, Copaltra (Coutarea latiflora and Centaurium erythraea), Kombucha tea, LipoKinetix (a capsule used for weight loss that contains multiple ingredients including usnic acid from the lichen Usnea barbata), Noni juice (Morinda citrifolia), Cascara sagrada, Sassafras (Sassafras albidum), and Senna (Cassia angustifolia) [20,34,103,154,158,163,164,172,206,246,256,277-287].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Drug-induced liver injury".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Drug-induced hepatitis (The Basics)")

SUMMARY AND RECOMMENDATIONS

Background – Herbal remedies may be associated with hepatotoxicity, making it important to discuss the use of herbal and dietary supplements (HDS) and complementary and alternative medical (CAM) therapies with patients to prevent potential complications, such as herb-drug interactions. (See "Overview of herbal medicine and dietary supplements".)

The use of herbal remedies should be considered as a possible etiology in any setting of clinical manifestations of liver injury.

Clinical features and diagnosis – Proving that an herbal or dietary supplement causes liver injury relies on both chronologic and clinical criteria. Features suggesting drug toxicity include lack of illness prior to ingesting the drug, clinical illness or biochemical abnormalities developing after beginning the drug, and improvement after the product is withdrawn. (See 'Diagnosis' above.)

Obtaining a sample of the implicated HDS or CAM therapies for testing is possible, but expertise is not widely available. Costs are generally prohibitive, and most research laboratories are capable of testing only for certain compounds.

Management – The mainstay of therapy for herbal hepatotoxicity is withdrawal of the offending toxin and supportive care. (See 'Treatment' above.)

Database of hepatoxic drugs and supplements – A searchable database of hepatotoxic drugs, herbal medications, and dietary supplements has been developed by the National Institutes of Health.

  1. Tindle HA, Davis RB, Phillips RS, Eisenberg DM. Trends in use of complementary and alternative medicine by US adults: 1997-2002. Altern Ther Health Med 2005; 11:42.
  2. Smith T, May G, Eckl V, Reynolds CM. US Sales of Herbal Supplements Increase by 8.6% in 2019. http://cms.herbalgram.org/herbalgram/issue127/files/hg127-hmr.pdf (Accessed on July 29, 2023).
  3. Complementary and Alternative Medicine in the United States. Institute of Medicine Committee on the Use of Complementary and Alternative Medicine by the American Public; National Academies Press (US), Washington (DC), 2005.
  4. Bailey RL, Gahche JJ, Lentino CV, et al. Dietary supplement use in the United States, 2003-2006. J Nutr 2011; 141:261.
  5. Nahin RL, Barnes PM, Stussman BJ. Expenditures on Complementary Health Approaches: United States, 2012. Natl Health Stat Report 2016; :1.
  6. Navarro VJ, Khan I, Björnsson E, et al. Liver injury from herbal and dietary supplements. Hepatology 2017; 65:363.
  7. Gurley BJ, McGill MR, Koturbash I. Hepatotoxicity due to herbal dietary supplements: Past, present and the future. Food Chem Toxicol 2022; 169:113445.
  8. Posadzki P, Watson LK, Alotaibi A, Ernst E. Prevalence of use of complementary and alternative medicine (CAM) by patients/consumers in the UK: systematic review of surveys. Clin Med (Lond) 2013; 13:126.
  9. Garcia-Alvarez A, Egan B, de Klein S, et al. Usage of plant food supplements across six European countries: findings from the PlantLIBRA consumer survey. PLoS One 2014; 9:e92265.
  10. Kennedy ET, Luo H, Houser RF. Dietary supplement use pattern of U.S. adult population in the 2007-2008 National Health and Nutrition Examination Survey (NHANES). Ecol Food Nutr 2013; 52:76.
  11. Dickinson A, Blatman J, El-Dash N, Franco JC. Consumer usage and reasons for using dietary supplements: report of a series of surveys. J Am Coll Nutr 2014; 33:176.
  12. Steenkamp V. Traditional herbal remedies used by South African women for gynaecological complaints. J Ethnopharmacol 2003; 86:97.
  13. Bunchorntavakul C, Reddy KR. Review article: herbal and dietary supplement hepatotoxicity. Aliment Pharmacol Ther 2013; 37:3.
  14. A Lindstrom, C Ooyen, ME Lynch, et al. Sales of herbal dietary supplements increase by 7.9% in 2013, marking a decade of rising sales. HerbalGram 2014; 103:52.
  15. Bailey RL, Gahche JJ, Miller PE, et al. Why US adults use dietary supplements. JAMA Intern Med 2013; 173:355.
  16. Ni H, Simile C, Hardy AM. Utilization of complementary and alternative medicine by United States adults: results from the 1999 national health interview survey. Med Care 2002; 40:353.
  17. Graham RE, Ahn AC, Davis RB, et al. Use of complementary and alternative medical therapies among racial and ethnic minority adults: results from the 2002 National Health Interview Survey. J Natl Med Assoc 2005; 97:535.
  18. Schuppan D, Jia JD, Brinkhaus B, Hahn EG. Herbal products for liver diseases: a therapeutic challenge for the new millennium. Hepatology 1999; 30:1099.
  19. Swerdlow JL.. Nature's Medicine: Plants That Heal., National Geographic, Washington, DC 2000.
  20. Larrey D, Pageaux GP. Hepatotoxicity of herbal remedies and mushrooms. Semin Liver Dis 1995; 15:183.
  21. Eisenberg DM, Kessler RC, Van Rompay MI, et al. Perceptions about complementary therapies relative to conventional therapies among adults who use both: results from a national survey. Ann Intern Med 2001; 135:344.
  22. Stirnimann G, Kessebohm K, Lauterburg B. Liver injury caused by drugs: an update. Swiss Med Wkly 2010; 140:w13080.
  23. LiverTox. Clinical and Research Information on Drug-Induced Liver Injury. https://www.ncbi.nlm.nih.gov/books/NBK547852/ (Accessed on November 14, 2023).
  24. Dietary Supplement Label Database (DSLD). NIH Office of Dietary Supplements. https://dsld.od.nih.gov (Accessed on July 29, 2023).
  25. Goldman P. Herbal medicines today and the roots of modern pharmacology. Ann Intern Med 2001; 135:594.
  26. Dietary Supplement Health and Education Act of 1994, Public Law 103-417. Office of Dietary Supplements, National Institutes of Health. Available at: https://ods.od.nih.gov/About/DSHEA_Wording.aspx#sec3 (Accessed on July 29, 2023).
  27. U.S. Food & Drug Administration. Dietary Supplement Ingredient Directory. https://www.fda.gov/food/dietary-supplements/dietary-supplement-ingredient-directory (Accessed on July 29, 2023).
  28. United States Pharmacopeia (USP). http://www.usp.org/ (Accessed on July 29, 2023).
  29. Navarro VJ, Lucena MI. Hepatotoxicity induced by herbal and dietary supplements. Semin Liver Dis 2014; 34:172.
  30. U.S. Food & Drug Administration. Dietary Supplements. http://www.fda.gov/food/dietary-supplements (Accessed on July 29, 2023).
  31. U.S. Food and Drug Administration. Label Claims for Food & Dietary Supplements. https://www.fda/gov./food/food-labeling-nutrition/label-claims-food-dietary-supplements (Accessed on July 29, 2023).
  32. U.S. Food and Drug Administration. Guidance & Regulation (Food and Dietary Supplements). https://www.fda.gov/food/guidance-regulation-food-and-dietary-supplements (Accessed on July 29, 2023).
  33. Tomlinson B, Chan TY, Chan JC, et al. Toxicity of complementary therapies: an eastern perspective. J Clin Pharmacol 2000; 40:451.
  34. Cole MR, Fetrow CW. Adulteration of dietary supplements. Am J Health Syst Pharm 2003; 60:1576.
  35. Ernst E. Toxic heavy metals and undeclared drugs in Asian herbal medicines. Trends Pharmacol Sci 2002; 23:136.
  36. Chan TY, Chan JC, Tomlinson B, Critchley JA. Chinese herbal medicines revisited: a Hong Kong perspective. Lancet 1993; 342:1532.
  37. Ko RJ. Adulterants in Asian patent medicines. N Engl J Med 1998; 339:847.
  38. Chan TY, Critchley JA. Usage and adverse effects of Chinese herbal medicines. Hum Exp Toxicol 1996; 15:5.
  39. Huang WF, Wen KC, Hsiao ML. Adulteration by synthetic therapeutic substances of traditional Chinese medicines in Taiwan. J Clin Pharmacol 1997; 37:344.
  40. Ernst E. Adulteration of Chinese herbal medicines with synthetic drugs: a systematic review. J Intern Med 2002; 252:107.
  41. Espinoza EO, Mann MJ, Bleasdell B. Arsenic and mercury in traditional Chinese herbal balls. N Engl J Med 1995; 333:803.
  42. Gertner E, Marshall PS, Filandrinos D, et al. Complications resulting from the use of Chinese herbal medications containing undeclared prescription drugs. Arthritis Rheum 1995; 38:614.
  43. Kang-Yum E, Oransky SH. Chinese patent medicine as a potential source of mercury poisoning. Vet Hum Toxicol 1992; 34:235.
  44. Stickel F, Droz S, Patsenker E, et al. Severe hepatotoxicity following ingestion of Herbalife nutritional supplements contaminated with Bacillus subtilis. J Hepatol 2009; 50:111.
  45. Kennedy J. Herb and supplement use in the US adult population. Clin Ther 2005; 27:1847.
  46. Saper RB, Phillips RS, Sehgal A, et al. Lead, mercury, and arsenic in US- and Indian-manufactured Ayurvedic medicines sold via the Internet. JAMA 2008; 300:915.
  47. Kales SN, Christophi CA, Saper RB. Hematopoietic toxicity from lead-containing Ayurvedic medications. Med Sci Monit 2007; 13:CR295.
  48. Nissan R, Poperno A, Stein GY, et al. A Case of Hepatotoxicity Induced by Adulterated "Tiger King", a Chinese Herbal Medicine Containing Sildenafil. Curr Drug Saf 2016; 11:184.
  49. Medina-Caliz I, Gonzalez-Jimenez A, Bessone F, et al. Variations in drug-induced liver injury (DILI) between different prospective DILI registries. Clin Ther 2013; 35:S24.
  50. Miller GM, Stripp R. A study of western pharmaceuticals contained within samples of Chinese herbal/patent medicines collected from New York City's Chinatown. Leg Med (Tokyo) 2007; 9:258.
  51. Fleshner N, Harvey M, Adomat H, et al. Evidence for contamination of herbal erectile dysfunction products with phosphodiesterase type 5 inhibitors. J Urol 2005; 174:636.
  52. Seeff LB, Lindsay KL, Bacon BR, et al. Complementary and alternative medicine in chronic liver disease. Hepatology 2001; 34:595.
  53. Harkey MR, Henderson GL, Gershwin ME, et al. Variability in commercial ginseng products: an analysis of 25 preparations. Am J Clin Nutr 2001; 73:1101.
  54. Ye F, Wang H, Jiang S, et al. Quality evaluation of commercial extracts of Scutellaria baicalensis. Nutr Cancer 2004; 49:217.
  55. Koff RS. Herbal hepatotoxicity. Revisiting a dangerous alternative. JAMA 1995; 273:502.
  56. European Food Safety Authority. Food supplements. https://eur-lex.europa.eu/legal-content/EN/TXT/PDF/?ur4i=CELEX:02002L0046-20220930 (Accessed on July 29, 2023).
  57. Woo SM, Davis WD, Aggarwal S, et al. Herbal and dietary supplement induced liver injury: Highlights from the recent literature. World J Hepatol 2021; 13:1019.
  58. World Health Organization traditional medicine strategy: 2014-2023. https://www.who.int/publications/i/item/9789241506096 (Accessed on July 29, 2023).
  59. Navarro VJ, Barnhart H, Bonkovsky HL, et al. Liver injury from herbals and dietary supplements in the U.S. Drug-Induced Liver Injury Network. Hepatology 2014; 60:1399.
  60. Chalasani N, Fontana RJ, Bonkovsky HL, et al. Causes, clinical features, and outcomes from a prospective study of drug-induced liver injury in the United States. Gastroenterology 2008; 135:1924.
  61. Roytman MM, Poerzgen P, Navarro V. Botanicals and Hepatotoxicity. Clin Pharmacol Ther 2018; 104:458.
  62. de Boer YS, Sherker AH. Herbal and Dietary Supplement-Induced Liver Injury. Clin Liver Dis 2017; 21:135.
  63. Bessone F, Hernandez N, Sanchez A, et al. The Spanish-Latin American DILI Network: Preliminary results from a collaborative strategic initiative. J Hepatol 2013; 58:S212.
  64. Suk KT, Kim DJ, Kim CH, et al. A prospective nationwide study of drug-induced liver injury in Korea. Am J Gastroenterol 2012; 107:1380.
  65. Reuben A, Koch DG, Lee WM, Acute Liver Failure Study Group. Drug-induced acute liver failure: results of a U.S. multicenter, prospective study. Hepatology 2010; 52:2065.
  66. Wai CT, Tan BH, Chan CL, et al. Drug-induced liver injury at an Asian center: a prospective study. Liver Int 2007; 27:465.
  67. Robles-Diaz M, Gonzalez-Jimenez A, Medina-Caliz I, et al. Distinct phenotype of hepatotoxicity associated with illicit use of anabolic androgenic steroids. Aliment Pharmacol Ther 2015; 41:116.
  68. Björnsson ES, Bergmann OM, Björnsson HK, et al. Incidence, presentation, and outcomes in patients with drug-induced liver injury in the general population of Iceland. Gastroenterology 2013; 144:1419.
  69. Douros A, Bronder E, Andersohn F, et al. Herb-Induced Liver Injury in the Berlin Case-Control Surveillance Study. Int J Mol Sci 2016; 17.
  70. Devarbhavi H, Dierkhising R, Kremers WK, et al. Single-center experience with drug-induced liver injury from India: causes, outcome, prognosis, and predictors of mortality. Am J Gastroenterol 2010; 105:2396.
  71. Dağ MS, Aydınlı M, Oztürk ZA, et al. Drug- and herb-induced liver injury: a case series from a single center. Turk J Gastroenterol 2014; 25:41.
  72. Goldberg DS, Forde KA, Carbonari DM, et al. Population-representative incidence of drug-induced acute liver failure based on an analysis of an integrated health care system. Gastroenterology 2015; 148:1353.
  73. Zhao P, Wang C, Liu W, Wang F. Acute liver failure associated with traditional Chinese medicine: report of 30 cases from seven tertiary hospitals in China*. Crit Care Med 2014; 42:e296.
  74. Navarro VJ. Herbal and dietary supplement hepatotoxicity. Semin Liver Dis 2009; 29:373.
  75. Fontana RJ, Liou I, Reuben A, et al. AASLD practice guidance on drug, herbal, and dietary supplement-induced liver injury. Hepatology 2023; 77:1036.
  76. Liu SH, Chuang WC, Lam W, et al. Safety surveillance of traditional Chinese medicine: current and future. Drug Saf 2015; 38:117.
  77. Durazzo A, Sorkin BC, Lucarini M, et al. Analytical Challenges and Metrological Approaches to Ensuring Dietary Supplement Quality: International Perspectives. Front Pharmacol 2021; 12:714434.
  78. FDA Finds Problems at 52% of Supplement Manufacturing Sites in U.S. and 42% Abroad. https://www.consumerlab.com/recalls/14344/fda-finds-problems-at-52-of-supplement-manufacturing-sites-in-us-and-42-abroad (Accessed on July 29, 2023).
  79. Navarro V, Avula B, Khan I, et al. The Contents of Herbal and Dietary Supplements Implicated in Liver Injury in the United States Are Frequently Mislabeled. Hepatol Commun 2019; 3:792.
  80. Larrey D. Epidemiology and individual susceptibility to adverse drug reactions affecting the liver. Semin Liver Dis 2002; 22:145.
  81. Hayashi PH. Causality assessment in drug-induced liver injury. Semin Liver Dis 2009; 29:348.
  82. Ballotin VR, Bigarella LG, Brandão ABM, et al. Herb-induced liver injury: Systematic review and meta-analysis. World J Clin Cases 2021; 9:5490.
  83. García-Cortés M, Robles-Díaz M, Ortega-Alonso A, et al. Hepatotoxicity by Dietary Supplements: A Tabular Listing and Clinical Characteristics. Int J Mol Sci 2016; 17:537.
  84. Chalasani NP, Maddur H, Russo MW, et al. ACG Clinical Guideline: Diagnosis and Management of Idiosyncratic Drug-Induced Liver Injury. Am J Gastroenterol 2021; 116:878.
  85. Andrade RJ, Lucena MI, Fernández MC, et al. Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology 2005; 129:512.
  86. Björnsson E, Olsson R. Outcome and prognostic markers in severe drug-induced liver disease. Hepatology 2005; 42:481.
  87. Bonkovsky HL, Kleiner DE, Gu J, et al. Clinical presentations and outcomes of bile duct loss caused by drugs and herbal and dietary supplements. Hepatology 2017; 65:1267.
  88. Zimmerman HJ, Ishak KG. General aspects of drug-induced liver disease. Gastroenterol Clin North Am 1995; 24:739.
  89. Larrey D, Vial T, Pauwels A, et al. Hepatitis after germander (Teucrium chamaedrys) administration: another instance of herbal medicine hepatotoxicity. Ann Intern Med 1992; 117:129.
  90. Bénichou C. Criteria of drug-induced liver disorders. Report of an international consensus meeting. J Hepatol 1990; 11:272.
  91. USFDA Guidance for Industry. Drug-Induced Liver Injury: Premarketing Clinical Evaluation. https://www.fda.gov/media/116737/download (Accessed on August 05, 2023).
  92. Lucena MI, Camargo R, Andrade RJ, et al. Comparison of two clinical scales for causality assessment in hepatotoxicity. Hepatology 2001; 33:123.
  93. Kaplowitz N. Causality assessment versus guilt-by-association in drug hepatotoxicity. Hepatology 2001; 33:308.
  94. Benichou C, Danan G, Flahault A. Causality assessment of adverse reactions to drugs--II. An original model for validation of drug causality assessment methods: case reports with positive rechallenge. J Clin Epidemiol 1993; 46:1331.
  95. Maria VA, Victorino RM. Development and validation of a clinical scale for the diagnosis of drug-induced hepatitis. Hepatology 1997; 26:664.
  96. García-Cortés M, Lucena MI, Pachkoria K, et al. Evaluation of naranjo adverse drug reactions probability scale in causality assessment of drug-induced liver injury. Aliment Pharmacol Ther 2008; 27:780.
  97. Hayashi PH, Lucena MI, Fontana RJ. RECAM: A New and Improved, Computerized Causality Assessment Tool for DILI Diagnosis. Am J Gastroenterol 2022; 117:1387.
  98. Rockey DC, Seeff LB, Rochon J, et al. Causality assessment in drug-induced liver injury using a structured expert opinion process: comparison to the Roussel-Uclaf causality assessment method. Hepatology 2010; 51:2117.
  99. O'Grady JG, Alexander GJ, Hayllar KM, Williams R. Early indicators of prognosis in fulminant hepatic failure. Gastroenterology 1989; 97:439.
  100. Björnsson E. Drug-induced liver injury: Hy's rule revisited. Clin Pharmacol Ther 2006; 79:521.
  101. Reuben A. Hy's law. Hepatology 2004; 39:574.
  102. Shaw D, Leon C, Kolev S, Murray V. Traditional remedies and food supplements. A 5-year toxicological study (1991-1995). Drug Saf 1997; 17:342.
  103. Pittler MH, Ernst E. Systematic review: hepatotoxic events associated with herbal medicinal products. Aliment Pharmacol Ther 2003; 18:451.
  104. Woolf GM, Petrovic LM, Rojter SE, et al. Acute hepatitis associated with the Chinese herbal product jin bu huan. Ann Intern Med 1994; 121:729.
  105. Picciotto A, Campo N, Brizzolara R, et al. Chronic hepatitis induced by Jin Bu Huan. J Hepatol 1998; 28:165.
  106. Neff GW, Reddy KR, Durazo FA, et al. Severe hepatotoxicity associated with the use of weight loss diet supplements containing ma huang or usnic acid. J Hepatol 2004; 41:1062.
  107. Bajaj J, Knox JF, Komorowski R, Saeian K. The irony of herbal hepatitis: Ma-Huang-induced hepatotoxicity associated with compound heterozygosity for hereditary hemochromatosis. Dig Dis Sci 2003; 48:1925.
  108. Nadir A, Agrawal S, King PD, Marshall JB. Acute hepatitis associated with the use of a Chinese herbal product, ma-huang. Am J Gastroenterol 1996; 91:1436.
  109. Lekehal M, Pessayre D, Lereau JM, et al. Hepatotoxicity of the herbal medicine germander: metabolic activation of its furano diterpenoids by cytochrome P450 3A Depletes cytoskeleton-associated protein thiols and forms plasma membrane blebs in rat hepatocytes. Hepatology 1996; 24:212.
  110. Laliberté L, Villeneuve JP. Hepatitis after the use of germander, a herbal remedy. CMAJ 1996; 154:1689.
  111. Mostefa-Kara N, Pauwels A, Pines E, et al. Fatal hepatitis after herbal tea. Lancet 1992; 340:674.
  112. Castot A, Larrey D. [Hepatitis observed during a treatment with a drug or tea containing Wild Germander. Evaluation of 26 cases reported to the Regional Centers of Pharmacovigilance]. Gastroenterol Clin Biol 1992; 16:916.
  113. Castot A, Djezzar S, Deleau N, et al. [Pharmacovigilance off the beaten track: herbal surveillance or pharmacovigilance of medicinal plants]. Therapie 1997; 52:97.
  114. Mimidis KP, Papadopoulos VP, Baltatzidis G, et al. Severe acute cholestasis caused by Teucrium polium. J Gastrointestin Liver Dis 2009; 18:387.
  115. MacGregor FB, Abernethy VE, Dahabra S, et al. Hepatotoxicity of herbal remedies. BMJ 1989; 299:1156.
  116. Harvey J, Colin-Jones DG. Mistletoe hepatitis. Br Med J (Clin Res Ed) 1981; 282:186.
  117. Hullar TE, Sapers BL, Ridker PM, et al. Herbal toxicity and fatal hepatic failure. Am J Med 1999; 106:267.
  118. Sheikh NM, Philen RM, Love LA. Chaparral-associated hepatotoxicity. Arch Intern Med 1997; 157:913.
  119. Katz M, Saibil F. Herbal hepatitis: subacute hepatic necrosis secondary to chaparral leaf. J Clin Gastroenterol 1990; 12:203.
  120. Gordon DW, Rosenthal G, Hart J, et al. Chaparral ingestion. The broadening spectrum of liver injury caused by herbal medications. JAMA 1995; 273:489.
  121. Smith BC, Desmond PV. Acute hepatitis induced by ingestion of the herbal medication chaparral. Aust N Z J Med 1993; 23:526.
  122. Batchelor WB, Heathcote J, Wanless IR. Chaparral-induced hepatic injury. Am J Gastroenterol 1995; 90:831.
  123. Bach N, Thung SN, Schaffner F. Comfrey herb tea-induced hepatic veno-occlusive disease. Am J Med 1989; 87:97.
  124. Ridker PM, McDermott WV. Comfrey herb tea and hepatic veno-occlusive disease. Lancet 1989; 1:657.
  125. Weston CF, Cooper BT, Davies JD, Levine DF. Veno-occlusive disease of the liver secondary to ingestion of comfrey. Br Med J (Clin Res Ed) 1987; 295:183.
  126. Yeong ML, Swinburn B, Kennedy M, Nicholson G. Hepatic veno-occlusive disease associated with comfrey ingestion. J Gastroenterol Hepatol 1990; 5:211.
  127. Stickel F, Seitz HK. The efficacy and safety of comfrey. Public Health Nutr 2000; 3:501.
  128. Kumana CR, Ng M, Lin HJ, et al. Herbal tea induced hepatic veno-occlusive disease: quantification of toxic alkaloid exposure in adults. Gut 1985; 26:101.
  129. Smith LW, Culvenor CC. Plant sources of hepatotoxic pyrrolizidine alkaloids. J Nat Prod 1981; 44:129.
  130. Mattocks AR. Toxicity of pyrrolizidine alkaloids. Nature 1968; 217:723.
  131. Gordon WP, Forte AJ, McMurtry RJ, et al. Hepatotoxicity and pulmonary toxicity of pennyroyal oil and its constituent terpenes in the mouse. Toxicol Appl Pharmacol 1982; 65:413.
  132. Centers for Disease Control and Prevention (CDC). Hepatic toxicity possibly associated with kava-containing products--United States, Germany, and Switzerland, 1999-2002. MMWR Morb Mortal Wkly Rep 2002; 51:1065.
  133. Ernst E. Safety concerns about kava. Lancet 2002; 359:1865.
  134. Wooltorton E. Herbal kava: reports of liver toxicity. CMAJ 2002; 166:777.
  135. Campo JV, McNabb J, Perel JM, et al. Kava-induced fulminant hepatic failure. J Am Acad Child Adolesc Psychiatry 2002; 41:631.
  136. Gow PJ, Connelly NJ, Hill RL, et al. Fatal fulminant hepatic failure induced by a natural therapy containing kava. Med J Aust 2003; 178:442.
  137. Yoshida EM, McLean CA, Cheng ES, et al. Chinese herbal medicine, fulminant hepatitis, and liver transplantation. Am J Gastroenterol 1996; 91:2647.
  138. LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. Available at: http://www.ncbi.nlm.nih.gov/books/NBK547852/ (Accessed on July 29, 2023).
  139. Roeder E. Medicinal plants in China containing pyrrolizidine alkaloids. Pharmazie 2000; 55:711.
  140. Li N, Xia Q, Ruan J, et al. Hepatotoxicity and tumorigenicity induced by metabolic activation of pyrrolizidine alkaloids in herbs. Curr Drug Metab 2011; 12:823.
  141. Guzman G, Kallwitz ER, Wojewoda C, et al. Liver Injury with Features Mimicking Autoimmune Hepatitis following the Use of Black Cohosh. Case Rep Med 2009; 2009:918156.
  142. Pearson W. Pyrrolizidine alkaloids in higher plants: hepatic veno-occlusive disease associated with chronic consumption. J Neutraceuticals 2000; 3:87.
  143. Benninger J, Schneider HT, Schuppan D, et al. Acute hepatitis induced by greater celandine (Chelidonium majus). Gastroenterology 1999; 117:1234.
  144. Xia Q, Zhao Y, Lin G, et al. Pyrrolizidine Alkaloid-Protein Adducts: Potential Non-invasive Biomarkers of Pyrrolizidine Alkaloid-Induced Liver Toxicity and Exposure. Chem Res Toxicol 2016; 29:1282.
  145. Steenkamp V, Stewart MJ, Zuckerman M. Clinical and analytical aspects of pyrrolizidine poisoning caused by South African traditional medicines. Ther Drug Monit 2000; 22:302.
  146. Tandon HD, Tandon BN, Ramalingaswami V. Epidemic of toxic hepatitis in India of possible mycotoxic origin. Arch Pathol Lab Med 1978; 102:372.
  147. FDA Advises Dietary Supplement Manufacturers to Remove Comfrey Products From the Market. http://wayback.archive-it.org/7993/20171114115012/https://www.fda.gov/Food/RecallsOutbreaksEmergencies/SafetyAlertsAdvisories/ucm111219.htm (Accessed on August 05, 2023).
  148. Conradie J, Stewart MJ, Steenkamp V. GC/MS identification of toxic pyrrolizidine alkaloids in traditional remedies given to two sets of twins. Ann Clin Biochem 2005; 42:141.
  149. Gao H, Li N, Wang JY, et al. Definitive diagnosis of hepatic sinusoidal obstruction syndrome induced by pyrrolizidine alkaloids. J Dig Dis 2012; 13:33.
  150. Geburek I, Rutz L, Gao L, et al. Metabolic Pattern of Hepatotoxic Pyrrolizidine Alkaloids in Liver Cells. Chem Res Toxicol 2021; 34:1101.
  151. Neuman MG, Jia AY, Steenkamp V. Senecio latifolius induces in vitro hepatocytotoxicity in a human cell line. Can J Physiol Pharmacol 2007; 85:1063.
  152. Ji LL, Zhang M, Sheng YC, Wang ZT. Pyrrolizidine alkaloid clivorine induces apoptosis in human normal liver L-02 cells and reduces the expression of p53 protein. Toxicol In Vitro 2005; 19:41.
  153. Valla D, Benhamou JP. Drug-induced vascular and sinusoidal lesions of the liver. Baillieres Clin Gastroenterol 1988; 2:481.
  154. Schiano TD. Liver injury from herbs and other botanicals. Clin Liver Dis 1998; 2:607.
  155. Ridker PM, Ohkuma S, McDermott WV, et al. Hepatic venocclusive disease associated with the consumption of pyrrolizidine-containing dietary supplements. Gastroenterology 1985; 88:1050.
  156. Neuman MG, Cohen L, Opris M, et al. Hepatotoxicity of Pyrrolizidine Alkaloids. J Pharm Pharm Sci 2015; 18:825.
  157. E Roeder, H Wiedenfeld. Plants containing pyrrolizidine alkaloids used in the traditional Indian medicine-including Ayurveda. Pharmazie 2013; 68:83.
  158. Jorge OA, Jorge AD. Hepatotoxicity associated with the ingestion of Centella asiatica. Rev Esp Enferm Dig 2005; 97:115.
  159. Teschke R, Bahre R. Severe hepatotoxicity by Indian Ayurvedic herbal products: a structured causality assessment. Ann Hepatol 2009; 8:258.
  160. Ernst E. Heavy metals in traditional Indian remedies. Eur J Clin Pharmacol 2002; 57:891.
  161. Björnsson HK, Björnsson ES, Avula B, et al. Ashwagandha-induced liver injury: A case series from Iceland and the US Drug-Induced Liver Injury Network. Liver Int 2020; 40:825.
  162. Tóth M, Benedek AE, Longerich T, Seitz HK. Ashwagandha-induced acute liver injury: A case report. Clin Case Rep 2023; 11:e7078.
  163. Bateman J, Chapman RD, Simpson D. Possible toxicity of herbal remedies. Scott Med J 1998; 43:7.
  164. Farrell GC. Drug-induced hepatic injury. J Gastroenterol Hepatol 1997; 12:S242.
  165. Fau D, Lekehal M, Farrell G, et al. Diterpenoids from germander, an herbal medicine, induce apoptosis in isolated rat hepatocytes. Gastroenterology 1997; 113:1334.
  166. Mattéi A, Rucay P, Samuel D, et al. Liver transplantation for severe acute liver failure after herbal medicine (Teucrium polium) administration. J Hepatol 1995; 22:597.
  167. Savvidou S, Goulis J, Giavazis I, et al. Herb-induced hepatitis by Teucrium polium L.: report of two cases and review of the literature. Eur J Gastroenterol Hepatol 2007; 19:507.
  168. Dourakis SP, Papanikolaou IS, Tzemanakis EN, Hadziyannis SJ. Acute hepatitis associated with herb (Teucrium capitatum L.) administration. Eur J Gastroenterol Hepatol 2002; 14:693.
  169. Poon WT, Chau TL, Lai CK, et al. Hepatitis induced by Teucrium viscidum. Clin Toxicol (Phila) 2008; 46:819.
  170. Vahlensieck U, Hahn R, Winterhoff H, et al. The effect of Chelidonium majus herb extract on choleresis in the isolated perfused rat liver. Planta Med 1995; 61:267.
  171. Moro PA, Cassetti F, Giugliano G, et al. Hepatitis from Greater celandine (Chelidonium majus L.): review of literature and report of a new case. J Ethnopharmacol 2009; 124:328.
  172. Stickel F, Pöschl G, Seitz HK, et al. Acute hepatitis induced by Greater Celandine (Chelidonium majus). Scand J Gastroenterol 2003; 38:565.
  173. Crijns AP, de Smet PA, van den Heuvel M, et al. [Acute hepatitis after use of a herbal preparation with greater celandine (Chelidonium majus)]. Ned Tijdschr Geneeskd 2002; 146:124.
  174. De Smet PA. Safety concerns about kava not unique. Lancet 2002; 360:1336.
  175. Greving I, Meister V, Monnerjahn C, et al. Chelidonium majus: a rare reason for severe hepatotoxic reaction. Pharmacoepidemiol Drug Saf 1998; 7 Suppl 1:S66.
  176. Rifai K, Flemming P, Manns MP, Trautwein C. [Severe drug hepatitis caused by Chelidonium]. Internist (Berl) 2006; 47:749.
  177. Teschke R, Glass X, Schulze J, Eickhoff A. Suspected Greater Celandine hepatotoxicity: liver-specific causality evaluation of published case reports from Europe. Eur J Gastroenterol Hepatol 2012; 24:270.
  178. Teschke R, Frenzel C, Glass X, et al. Greater Celandine hepatotoxicity: a clinical review. Ann Hepatol 2012; 11:838.
  179. Teschke R, Glass X, Schulze J. Herbal hepatotoxicity by Greater Celandine (Chelidonium majus): causality assessment of 22 spontaneous reports. Regul Toxicol Pharmacol 2011; 61:282.
  180. Kassler WJ, Blanc P, Greenblatt R. The use of medicinal herbs by human immunodeficiency virus-infected patients. Arch Intern Med 1991; 151:2281.
  181. Kauma H, Koskela R, Mäkisalo H, et al. Toxic acute hepatitis and hepatic fibrosis after consumption of chaparral tablets. Scand J Gastroenterol 2004; 39:1168.
  182. Zaim N, Guemouri L, Lamnaouer D, Benjouad A. [Study of four cases of poisoning by Atractylis gummifera L. in Morocco]. Therapie 2008; 63:49.
  183. Ahid S, El Cadi MA, Meddah B, Cherrah Y. [Atractylis gummifera: from poisoning to the analytic methods]. Ann Biol Clin (Paris) 2012; 70:263.
  184. Daniele C, Dahamna S, Firuzi O, et al. Atractylis gummifera L. poisoning: an ethnopharmacological review. J Ethnopharmacol 2005; 97:175.
  185. Hamouda C, Hédhili A, Ben Salah N, et al. A review of acute poisoning from Atractylis gummifera L. Vet Hum Toxicol 2004; 46:144.
  186. Popat A, Shear NH, Malkiewicz I, et al. The toxicity of Callilepis laureola, a South African traditional herbal medicine. Clin Biochem 2001; 34:229.
  187. Stewart MJ, Steenkamp V, van der Merwe S, et al. The cytotoxic effects of a traditional Zulu remedy, impila (Callilepis laureola). Hum Exp Toxicol 2002; 21:643.
  188. Anderson IB, Mullen WH, Meeker JE, et al. Pennyroyal toxicity: measurement of toxic metabolite levels in two cases and review of the literature. Ann Intern Med 1996; 124:726.
  189. Bakerink JA, Gospe SM Jr, Dimand RJ, Eldridge MW. Multiple organ failure after ingestion of pennyroyal oil from herbal tea in two infants. Pediatrics 1996; 98:944.
  190. Thomassen D, Slattery JT, Nelson SD. Menthofuran-dependent and independent aspects of pulegone hepatotoxicity: roles of glutathione. J Pharmacol Exp Ther 1990; 253:567.
  191. Stickel F, Baumüller HM, Seitz K, et al. Hepatitis induced by Kava (Piper methysticum rhizoma). J Hepatol 2003; 39:62.
  192. Teschke R. Kava hepatotoxicity--a clinical review. Ann Hepatol 2010; 9:251.
  193. Teschke R, Schwarzenboeck A, Hennermann KH. Kava hepatotoxicity: a clinical survey and critical analysis of 26 suspected cases. Eur J Gastroenterol Hepatol 2008; 20:1182.
  194. Teschke R. Kava hepatotoxicity: pathogenetic aspects and prospective considerations. Liver Int 2010; 30:1270.
  195. Teschke R, Qiu SX, Lebot V. Herbal hepatotoxicity by kava: update on pipermethystine, flavokavain B, and mould hepatotoxins as primarily assumed culprits. Dig Liver Dis 2011; 43:676.
  196. Sarris J, Kavanagh DJ, Byrne G, et al. The Kava Anxiety Depression Spectrum Study (KADSS): a randomized, placebo-controlled crossover trial using an aqueous extract of Piper methysticum. Psychopharmacology (Berl) 2009; 205:399.
  197. Teschke R, Genthner A, Wolff A. Kava hepatotoxicity: comparison of aqueous, ethanolic, acetonic kava extracts and kava-herbs mixtures. J Ethnopharmacol 2009; 123:378.
  198. Javaid A, Bonkovsky HL. Hepatotoxicity due to extracts of Chinese green tea (Camellia sinensis): a growing concern. J Hepatol 2006; 45:334.
  199. Bonkovsky HL. Hepatotoxicity associated with supplements containing Chinese green tea (Camellia sinensis). Ann Intern Med 2006; 144:68.
  200. Molinari M, Watt KD, Kruszyna T, et al. Acute liver failure induced by green tea extracts: case report and review of the literature. Liver Transpl 2006; 12:1892.
  201. Oketch-Rabah HA, Roe AL, Rider CV, et al. United States Pharmacopeia (USP) comprehensive review of the hepatotoxicity of green tea extracts. Toxicol Rep 2020; 7:386.
  202. Galati G, Lin A, Sultan AM, O'Brien PJ. Cellular and in vivo hepatotoxicity caused by green tea phenolic acids and catechins. Free Radic Biol Med 2006; 40:570.
  203. Weinstein DH, Twaddell WS, Raufman JP, et al. SlimQuick™ - associated hepatotoxicity in a woman with alpha-1 antitrypsin heterozygosity. World J Hepatol 2012; 4:154.
  204. Hoofnagle JH, Bonkovsky HL, Phillips EJ, et al. HLA-B*35:01 and Green Tea-Induced Liver Injury. Hepatology 2021; 73:2484.
  205. Isomura T, Suzuki S, Origasa H, et al. Liver-related safety assessment of green tea extracts in humans: a systematic review of randomized controlled trials. Eur J Clin Nutr 2016; 70:1221.
  206. Mazzanti G, Menniti-Ippolito F, Moro PA, et al. Hepatotoxicity from green tea: a review of the literature and two unpublished cases. Eur J Clin Pharmacol 2009; 65:331.
  207. Ishak KG. Hepatic lesions caused by anabolic and contraceptive steroids. Semin Liver Dis 1981; 1:116.
  208. Hepatic effects of 17 alpha-alkylated anaboli-androgenic steroids. HIV Hotline 1998; 8:2.
  209. Mohideen H, Hussain H, Dahiya DS, Wehbe H. Selective Androgen Receptor Modulators: An Emerging Liver Toxin. J Clin Transl Hepatol 2023; 11:188.
  210. Kafrouni MI, Anders RA, Verma S. Hepatotoxicity associated with dietary supplements containing anabolic steroids. Clin Gastroenterol Hepatol 2007; 5:809.
  211. Timcheh-Hariri A, Balali-Mood M, Aryan E, et al. Toxic hepatitis in a group of 20 male body-builders taking dietary supplements. Food Chem Toxicol 2012; 50:3826.
  212. Martin DJ, Partridge BJ, Shields W. Hepatotoxicity associated with the dietary supplement N.O.-XPLODE. Ann Intern Med 2013; 159:503.
  213. FDA Warns of Use of Selective Androgen Receptor Modulators (SARMs) Among Teens, Young Adults. https://www.fda.gov/consumers/consumer-updates/fda-warns-use-selective-androgen-receptor-modulators-sarms-among-teens-young-adults (Accessed on May 03, 2023).
  214. U.S. Food & Drug Administration. FDA issues warning about body-building products labeled to contain steroid and steroid-like substances. https://www.fda.gov/drugs/drug-safety-and-availability/fda-issues-warning-about-body-building-products-labeled-contain-steroid-and-steroid-substances (Accessed on August 05, 2023).
  215. Pierard S, Coche JC, Lanthier P, et al. Severe hepatitis associated with the use of black cohosh: a report of two cases and an advice for caution. Eur J Gastroenterol Hepatol 2009; 21:941.
  216. Mahady GB, Low Dog T, Barrett ML, et al. United States Pharmacopeia review of the black cohosh case reports of hepatotoxicity. Menopause 2008; 15:628.
  217. Cohen SM, O'Connor AM, Hart J, et al. Autoimmune hepatitis associated with the use of black cohosh: a case study. Menopause 2004; 11:575.
  218. Levitsky J, Alli TA, Wisecarver J, Sorrell MF. Fulminant liver failure associated with the use of black cohosh. Dig Dis Sci 2005; 50:538.
  219. Lynch CR, Folkers ME, Hutson WR. Fulminant hepatic failure associated with the use of black cohosh: a case report. Liver Transpl 2006; 12:989.
  220. ET Enbom, MD Le, L Oestrich, et al.. Mechanism of hepatotoxicity due to black cohosh: (Cimicifuga racemose): historical, immunohistorical and electron microscopy analysis of two liver biopsies with clinical correlation. Exp Mol Pathol 2014; 96:279.
  221. Sarri G, Pedder H, Dias S, et al. Vasomotor symptoms resulting from natural menopause: a systematic review and network meta-analysis of treatment effects from the National Institute for Health and Care Excellence guideline on menopause. BJOG 2017; 124:1514.
  222. Castelo-Branco C, Gambacciani M, Cano A, et al. Review & meta-analysis: isopropanolic black cohosh extract iCR for menopausal symptoms - an update on the evidence. Climacteric 2021; 24:109.
  223. https://www.fda.gov/food/alerts-advisories-safety-information/fda-alerts-consumers-about-potentially-life-threatening-health-problems-linked-limbrel (Accessed on November 17, 2020).
  224. Chalasani N, Vuppalanchi R, Navarro V, et al. Acute liver injury due to flavocoxid (Limbrel), a medical food for osteoarthritis: a case series. Ann Intern Med 2012; 156:857.
  225. Garcia-Romeu A, Cox DJ, Smith KE, et al. Kratom (Mitragyna speciosa): User demographics, use patterns, and implications for the opioid epidemic. Drug Alcohol Depend 2020; 208:107849.
  226. Post S, Spiller HA, Chounthirath T, Smith GA. Kratom exposures reported to United States poison control centers: 2011-2017. Clin Toxicol (Phila) 2019; 57:847.
  227. Ahmad J, Odin JA, Hayashi PH, et al. Liver injury associated with kratom, a popular opioid-like product: Experience from the U.S. drug induced liver injury network and a review of the literature. Drug Alcohol Depend 2021; 218:108426.
  228. Griffin OH, Webb ME. The Scheduling of Kratom and Selective Use of Data. J Psychoactive Drugs 2018; 50:114.
  229. Perharic-Walton L, Murray V. Toxicity of Chinese herbal remedies. Lancet 1992; 340:674.
  230. Graham-Brown R. Toxicity of Chinese herbal remedies. Lancet 1992; 340:673.
  231. Jung KA, Min HJ, Yoo SS, et al. Drug-Induced Liver Injury: Twenty Five Cases of Acute Hepatitis Following Ingestion of Polygonum multiflorum Thunb. Gut Liver 2011; 5:493.
  232. Teschke R. Traditional Chinese Medicine Induced Liver Injury. J Clin Transl Hepatol 2014; 2:80.
  233. Jing J, Teschke R. Traditional Chinese Medicine and Herb-induced Liver Injury: Comparison with Drug-induced Liver Injury. J Clin Transl Hepatol 2018; 6:57.
  234. Fu PP, Xia Q, Lin G, Chou MW. Pyrrolizidine alkaloids--genotoxicity, metabolism enzymes, metabolic activation, and mechanisms. Drug Metab Rev 2004; 36:1.
  235. Wu GL, Yu GY, Chen J. [Clinical analysis of hepatic veno-occlusive disease induced by Sedum aizoon]. Zhongguo Zhong Yao Za Zhi 2008; 33:2402.
  236. Lin G, Wang JY, Li N, et al. Hepatic sinusoidal obstruction syndrome associated with consumption of Gynura segetum. J Hepatol 2011; 54:666.
  237. Xiong A, Fang L, Yang X, et al. An application of target profiling analyses in the hepatotoxicity assessment of herbal medicines: comparative characteristic fingerprint and bile acid profiling of Senecio vulgaris L. and Senecio scandens Buch.-Ham. Anal Bioanal Chem 2014; 406:7715.
  238. Xu JJ, Henstock PV, Dunn MC, et al. Cellular imaging predictions of clinical drug-induced liver injury. Toxicol Sci 2008; 105:97.
  239. Horowitz RS, Feldhaus K, Dart RC, et al. The clinical spectrum of Jin Bu Huan toxicity. Arch Intern Med 1996; 156:899.
  240. Borum ML. Fulminant exacerbation of autoimmune hepatitis after the use of ma huang. Am J Gastroenterol 2001; 96:1654.
  241. Skoulidis F, Alexander GJ, Davies SE. Ma huang associated acute liver failure requiring liver transplantation. Eur J Gastroenterol Hepatol 2005; 17:581.
  242. Verma S, Thuluvath PJ. Complementary and alternative medicine in hepatology: review of the evidence of efficacy. Clin Gastroenterol Hepatol 2007; 5:408.
  243. Itoh S, Marutani K, Nishijima T, et al. Liver injuries induced by herbal medicine, syo-saiko-to (xiao-chai-hu-tang). Dig Dis Sci 1995; 40:1845.
  244. Kamiyama T, Nouchi T, Kojima S, et al. Autoimmune hepatitis triggered by administration of an herbal medicine. Am J Gastroenterol 1997; 92:703.
  245. Hsu LM, Huang YS, Tsay SH, et al. Acute hepatitis induced by Chinese hepatoprotective herb, xiao-chai-hu-tang. J Chin Med Assoc 2006; 69:86.
  246. Adachi M, Saito H, Kobayashi H, et al. Hepatic injury in 12 patients taking the herbal weight loss AIDS Chaso or Onshido. Ann Intern Med 2003; 139:488.
  247. Ding Y, Zhang T, Tao JS, et al. Potential hepatotoxicity of geniposide, the major iridoid glycoside in dried ripe fruits of Gardenia jasminoides (Zhi-zi). Nat Prod Res 2013; 27:929.
  248. Li C, Rao T, Chen X, et al. HLA-B*35:01 Allele Is a Potential Biomarker for Predicting Polygonum multiflorum-Induced Liver Injury in Humans. Hepatology 2019; 70:346.
  249. Stevens T, Qadri A, Zein NN. Two patients with acute liver injury associated with use of the herbal weight-loss supplement hydroxycut. Ann Intern Med 2005; 142:477.
  250. Jones FJ, Andrews AH. Acute liver injury associated with the herbal supplement hydroxycut in a soldier deployed to Iraq. Am J Gastroenterol 2007; 102:2357.
  251. Dara L, Hewett J, Lim JK. Hydroxycut hepatotoxicity: a case series and review of liver toxicity from herbal weight loss supplements. World J Gastroenterol 2008; 14:6999.
  252. Shim M, Saab S. Severe hepatotoxicity due to Hydroxycut: a case report. Dig Dis Sci 2009; 54:406.
  253. Fong TL, Klontz KC, Canas-Coto A, et al. Hepatotoxicity due to hydroxycut: a case series. Am J Gastroenterol 2010; 105:1561.
  254. https://www.fda.gov/media/76365/download (Accessed on November 17, 2020).
  255. Schoepfer AM, Engel A, Fattinger K, et al. Herbal does not mean innocuous: ten cases of severe hepatotoxicity associated with dietary supplements from Herbalife products. J Hepatol 2007; 47:521.
  256. Elinav E, Pinsker G, Safadi R, et al. Association between consumption of Herbalife nutritional supplements and acute hepatotoxicity. J Hepatol 2007; 47:514.
  257. Duque JM, Ferreiro J, Salgueiro E, Manso G. [Hepatotoxicity associated with the consumption of herbal slimming products]. Med Clin (Barc) 2007; 128:238.
  258. Chao S, Anders M, Turbay M, et al. [Toxic hepatitis by consumption Herbalife products a case report]. Acta Gastroenterol Latinoam 2008; 38:274.
  259. Manso G, López-Rivas L, Duque JM, Salgueiro E. Spanish reports of hepatotoxicity associated with Herbalife products. J Hepatol 2008; 49:289.
  260. Manso G, López-Rivas L, Salgueiro ME, et al. Continuous reporting of new cases in Spain supports the relationship between Herbalife® products and liver injury. Pharmacoepidemiol Drug Saf 2011; 20:1080.
  261. Ramanathan VS, Mitropoulos E, Shlopov B, et al. An Enzyte'ing' case of acute hepatitis. J Clin Gastroenterol 2011; 45:834.
  262. Chatham-Stephens K, Taylor E, Chang A, et al. Hepatotoxicity associated with weight loss or sports dietary supplements, including OxyELITE Pro™ - United States, 2013. Drug Test Anal 2017; 9:68.
  263. Stickel F, Kessebohm K, Weimann R, Seitz HK. Review of liver injury associated with dietary supplements. Liver Int 2011; 31:595.
  264. Pacheco A, González-Sainz J, Arocena C, et al. Community acquired pneumonia caused by Chlamydia pneumoniae strain TWAR in chronic cardiopulmonary disease in the elderly. Respiration 1991; 58:316.
  265. Bessone F, García-Cortés M, Medina-Caliz I, et al. Herbal and Dietary Supplements-Induced Liver Injury in Latin America: Experience From the LATINDILI Network. Clin Gastroenterol Hepatol 2022; 20:e548.
  266. Wang ZG, Ren J. Current status and future direction of Chinese herbal medicine. Trends Pharmacol Sci 2002; 23:347.
  267. U.S. Department of Health and Human Services. Warning on Hydroxycut Products. http://wayback.archive-it.org/7993/20170722060237/https://www.fda.gov/ForConsumers/ConsumerUpdates/ucm152152.htm (Accessed on August 05, 2023).
  268. Melendez-Rosado J, Snipelisky D, Matcha G, Stancampiano F. Acute hepatitis induced by pure Garcinia cambogia. J Clin Gastroenterol 2015; 49:449.
  269. Corey R, Werner KT, Singer A, et al. Acute liver failure associated with Garcinia cambogia use. Ann Hepatol 2016; 15:123.
  270. Lunsford KE, Bodzin AS, Reino DC, et al. Dangerous dietary supplements: Garcinia cambogia-associated hepatic failure requiring transplantation. World J Gastroenterol 2016; 22:10071.
  271. U.S. Food & Drug Administration. Public Notification: Fruta Planta Life (Garcinia Cambogia Premium) contains hidden drug ingredient. https://www.fda.gov/drugs/medication-health-fraud/public-notification-fruta-planta-life-garcinia-cambogia-premium-contains-hidden-drug-ingredient (Accessed on August 05, 2023).
  272. Chounta A, Tsiodras S, Zouridakis S, et al. Sibutramine use associated with reversible hepatotoxicity. Ann Intern Med 2005; 143:763.
  273. Foley S, Butlin E, Shields W, Lacey B. Experience with OxyELITE pro and acute liver injury in active duty service members. Dig Dis Sci 2014; 59:3117.
  274. Roytman MM, Pörzgen P, Lee CL, et al. Outbreak of severe hepatitis linked to weight-loss supplement OxyELITE Pro. Am J Gastroenterol 2014; 109:1296.
  275. Klontz KC, DeBeck HJ, LeBlanc P, et al. The Role of Adverse Event Reporting in the FDA Response to a Multistate Outbreak of Liver Disease Associated with a Dietary Supplement. Public Health Rep 2015; 130:526.
  276. Aegeline. In PubChem, National Library of Medicine. https://pubchem.ncbi.nlm.nih.gov/substance/165224873#section=Source (Accessed on October 03, 2023).
  277. Miller LG. Herbal medicinals: selected clinical considerations focusing on known or potential drug-herb interactions. Arch Intern Med 1998; 158:2200.
  278. Perron AD, Patterson JA, Yanofsky NN. Kombucha "mushroom" hepatotoxicity. Ann Emerg Med 1995; 26:660.
  279. Favreau JT, Ryu ML, Braunstein G, et al. Severe hepatotoxicity associated with the dietary supplement LipoKinetix. Ann Intern Med 2002; 136:590.
  280. Nadir A, Reddy D, Van Thiel DH. Cascara sagrada-induced intrahepatic cholestasis causing portal hypertension: case report and review of herbal hepatotoxicity. Am J Gastroenterol 2000; 95:3634.
  281. Kao WF, Hung DZ, Tsai WJ, et al. Podophyllotoxin intoxication: toxic effect of Bajiaolian in herbal therapeutics. Hum Exp Toxicol 1992; 11:480.
  282. Uc A, Bishop WP, Sanders KD. Camphor hepatotoxicity. South Med J 2000; 93:596.
  283. Mallat A, Dhumeaux D. Cocaine and the liver. J Hepatol 1991; 12:275.
  284. Wurtz AS, Vial T, Isoard B, Saillard E. Possible hepatotoxicity from Copaltra, an herbal medicine. Ann Pharmacother 2002; 36:941.
  285. Millonig G, Stadlmann S, Vogel W. Herbal hepatotoxicity: acute hepatitis caused by a Noni preparation (Morinda citrifolia). Eur J Gastroenterol Hepatol 2005; 17:445.
  286. Beuers U, Spengler U, Pape GR. Hepatitis after chronic abuse of senna. Lancet 1991; 337:372.
  287. Yang HN, Kim DJ, Kim YM, et al. Aloe-induced toxic hepatitis. J Korean Med Sci 2010; 25:492.
Topic 3618 Version 35.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟