Small Molecule
C22H18O11
989-51-5
Epigallocatechin gallate (EGCG) stands as the most abundant and biologically active catechin within green tea (Camellia sinensis), a natural product that has garnered immense scientific and public interest for its potential health benefits. Preclinical research has extensively documented its potent antioxidant, anti-inflammatory, antineoplastic, and neuroprotective properties, positioning it as a highly promising therapeutic agent for a wide spectrum of chronic and infectious diseases. However, the trajectory from promising preclinical compound to effective clinical therapeutic is fraught with significant challenges, and EGCG serves as a paradigmatic example of this difficult transition. The central obstacle that defines and complicates its clinical development is its exceptionally poor oral bioavailability. This pharmacokinetic limitation necessitates the administration of high oral doses to achieve potentially therapeutic systemic concentrations. This requirement, in turn, creates a profound efficacy-safety dilemma, as these high doses—particularly those at or above 800 mg per day—are directly associated with a statistically significant risk of dose-dependent hepatotoxicity.
This report provides a comprehensive, multi-disciplinary monograph on EGCG, synthesizing a vast body of evidence to construct a nuanced understanding of its molecular characteristics, pharmacological mechanisms, pharmacokinetic profile, clinical trial outcomes, and safety considerations. The analysis reveals that the very chemical features responsible for EGCG's bioactivity—its numerous phenolic hydroxyl groups—are also the root cause of its poor stability and low membrane permeability. Its mechanism of action is pleiotropic, functioning less like a targeted therapeutic and more like a "molecular shotgun" that modulates a multitude of enzymes and signaling pathways, including NF-κB, PI3K/Akt/mTOR, and STAT3.
The clinical evidence for EGCG is mixed and often contradictory. While it has shown promise for improving functional and symptomatic endpoints in conditions like multiple sclerosis and has demonstrated notable cognitive benefits in the genetically distinct Down syndrome population, it has largely failed to deliver on its promise as a broad disease-modifying agent in large-scale trials for cancer prevention or neurodegenerative disease progression. This discrepancy is likely attributable to the failure of many clinical trial designs to adequately address its challenging pharmacokinetics.
The significant risk of liver injury at high doses has prompted cautious and varied regulatory responses from global health agencies. The European Food Safety Authority (EFSA) has established a restrictive upper limit of less than 800 mg/day for EGCG in food supplements, while Australia's Therapeutic Goods Administration (TGA) has implemented a schedule exemption for products containing 300 mg or less. In the United States, the Food and Drug Administration (FDA) maintains a complex position, recognizing EGCG as Generally Recognized as Safe (GRAS) in some contexts while also granting it Orphan Drug Designation for a specific inflammatory condition. Furthermore, EGCG's capacity to inhibit key drug-metabolizing enzymes and transporters creates a significant, and often underappreciated, risk of clinically relevant drug-drug interactions.
Ultimately, EGCG remains a molecule of profound scientific interest. Its future as a therapeutic agent likely depends not on the use of the native compound in high-dose oral formulations, but on the development of advanced delivery systems, such as nanoparticle formulations or prodrug strategies, designed to overcome its fundamental pharmacokinetic and safety hurdles.
A thorough understanding of the molecular and physicochemical properties of Epigallocatechin gallate (EGCG) is fundamental to interpreting its biological activities, pharmacokinetic behavior, and challenges in clinical application. This section provides a detailed characterization of EGCG, covering its nomenclature, chemical structure, physical properties, and origins.
The compound is primarily identified by its generic name, Epigallocatechin gallate, and is cataloged in major drug and chemical databases under specific identifiers, including DrugBank ID DB12116 and Chemical Abstracts Service (CAS) Number 989-51-5.[1] An older, deprecated CAS number, 863-65-0, is also associated with the molecule.[2]
Its precise chemical structure and stereochemistry are defined by its International Union of Pure and Applied Chemistry (IUPAC) name: 3,4,5-trihydroxybenzoate.[2] This systematic name specifies the
cis configuration of the substituents at the C2 and C3 positions of the heterocyclic C-ring, a key feature distinguishing it from its trans isomer, gallocatechin gallate.
In scientific literature and commercial products, EGCG is referred to by a variety of synonyms, including (-)-Epigallocatechin 3-gallate, EGCG, (-)-EPIGALLOCATECHIN-3-O-GALLATE, and 3-O-GALLOYL-(-)-EPIGALLOCATECHIN.[1]
Chemically, EGCG is classified as a small molecule flavonoid, a large family of polyphenolic compounds widely found in plants.[1] More specifically, it is a flavan-3-ol, characterized by a C6-C3-C6 backbone. It belongs to the direct parent class of catechin gallates, which are defined as compounds containing a gallate (3,4,5-trihydroxybenzoate) moiety ester-linked to a catechin core.[1] This structural classification is critical, as it places EGCG within a well-studied group of natural products renowned for their antioxidant and biological activities. Further hierarchical classifications include its membership in the superclass of Phenylpropanoids and polyketides and broader categories such as Benzopyrans and Chromans.[1] Its broad relevance across biomedical research is underscored by its numerous identifiers in key databases, such as PubChem Compound ID (CID) 65064, ChEBI ID CHEBI:4806, Human Metabolome Database (HMDB) ID HMDB0003153, and FDA Unique Ingredient Identifier (UNII) BQM438CTEL.[2]
The molecular formula of EGCG is C22H18O11, corresponding to an average molecular weight of approximately 458.37 to 458.38 g/mol and a monoisotopic mass of approximately 458.085 Da.[1] Its two-dimensional structure and three-dimensional conformation are unambiguously defined by standard chemical line notations. The canonical Simplified Molecular-Input Line-Entry System (SMILES) string is
C1[C@H]([C@H](OC2=CC(=CC(=C21)O)O)C3=CC(=C(C(=C3)O)O)O)OC(=O)C4=CC(=C(C(=C4)O)O)O, and its International Chemical Identifier Key (InChIKey) is WMBWREPUVVBILR-WIYYLYMNSA-N.[2]
The structure of EGCG consists of four rings: two phenolic rings (A and B rings), a dihydropyran heterocyclic ring (C ring), and a galloyl group (D ring) attached at the 3-position of the C ring.[1] A defining feature of this structure is the presence of eight phenolic hydroxyl groups distributed across the A, B, and D rings. These hydroxyl groups are the primary sites of its chemical reactivity and are directly responsible for its potent biological activities, particularly its antioxidant capacity.
Physically, pure EGCG is described as a white to light brown or off-white to light yellow solid or crystalline powder.[5] There is significant variability in the reported melting point of the compound, with values ranging from 140–142 °C [11], 213–216 °C [5], to as high as 222–224 °C.[4] Such a wide discrepancy is unusual for a single pure compound and is a strong indicator of the challenges associated with its characterization. This variability likely stems from differences in sample purity, the presence of various polymorphic crystalline forms, or degradation of the sample during analysis, which can lead to inconsistent experimental results and complicates the comparison of data across different studies. The estimated boiling point is consistently high, around 909–910 °C.[4]
The solubility and lipophilicity of EGCG are complex and, like its melting point, are reported with considerable variation. It is generally considered to have poor water solubility, with quantitative values cited as 0.0728 mg/mL and 32.77 mg/L (equivalent to 0.03277 mg/mL).[1] In contrast, other sources describe it as being soluble in water up to 100 mM (approximately 45.8 mg/mL) and readily soluble in organic solvents like ethanol and dimethyl formamide.[5] This conflicting information underscores the difficulty in working with the compound. Its lipophilicity, measured as the octanol-water partition coefficient (
logP), also varies across different predictive models, with values ranging from 0.639 to 3.58, suggesting a moderately lipophilic character.[1]
A critical characteristic of EGCG is its inherent instability. The molecule is highly susceptible to degradation, particularly in aqueous solutions at neutral or alkaline pH, and is sensitive to light, heat, and oxidation.[5] Its stability is significantly improved under acidic conditions (e.g., pH 3.5), at lower temperatures (4 °C or -20 °C), or in the presence of reducing agents such as ascorbic acid.[5] This instability is a major factor limiting its shelf-life in formulations and its bioavailability following oral administration.
From a drug development perspective, EGCG's structure presents a significant challenge. It violates Lipinski's Rule of Five, a set of guidelines used to predict the druglikeness of a molecule, specifically its potential for oral absorption.[1] The violations stem from its high number of hydrogen bond donors (8) and acceptors (10–11), as well as its large polar surface area of 197.37
A˚2.[1] These properties predict poor membrane permeability and, consequently, low oral absorption. The structural features of EGCG thus represent a fundamental paradox: the numerous hydroxyl groups that endow it with potent antioxidant and metal-chelating activities are the very same features that render it unstable, poorly permeable, and susceptible to extensive metabolism. This inherent chemical conflict between biological potency and pharmacokinetic liability is a central theme that dictates its therapeutic potential.
| Property | Value / Range | Source(s) |
|---|---|---|
| Identifiers | ||
| DrugBank ID | DB12116 | 1 |
| CAS Number | 989-51-5 | 2 |
| IUPAC Name | 3,4,5-trihydroxybenzoate | 2 |
| InChIKey | WMBWREPUVVBILR-WIYYLYMNSA-N | 2 |
| Molecular Properties | ||
| Molecular Formula | C22H18O11 | 1 |
| Average Molecular Weight | 458.37–458.38 g/mol | 1 |
| Monoisotopic Mass | 458.084911418 Da | 1 |
| Physical Properties | ||
| Appearance | White to light brown/yellow solid powder | 5 |
| Melting Point | 140–142 °C | 11 |
| 213–216 °C | 5 | |
| 222–224 °C | 4 | |
| Boiling Point (est.) | 909.00–910.00 °C | 4 |
| Water Solubility | 0.0728 mg/mL (ALOGPS) | 1 |
| 32.77 mg/L @ 25 °C (est.) | 4 | |
| Soluble to 100 mM | 8 | |
| Druglikeness Parameters | ||
| logP (o/w) | 0.639 to 3.58 | 1 |
| pKa (Strongest Acidic) | 7.75–7.99 | 1 |
| Hydrogen Bond Donors | 8 | 1 |
| Hydrogen Bond Acceptors | 10–11 | 1 |
| Polar Surface Area | 197.37 A˚2 | 1 |
| Lipinski's Rule of Five | No (Violated) | 1 |
EGCG is predominantly sourced from the leaves of the tea plant, Camellia sinensis, where it is the most abundant and biologically active of the catechins.[6] It can comprise 50–80% of the total catechin content and up to 30% of the dry weight of green tea leaves.[6] The concentration of EGCG varies significantly depending on the type of tea, which is determined by the processing method. Green tea, which is produced from unoxidized leaves, contains the highest concentration, with reported values around 7380 mg per 100 g of dried leaves. White tea also retains a high concentration (4245 mg/100 g). In contrast, black tea, which undergoes extensive enzymatic oxidation, has a significantly lower EGCG content (936 mg/100 g) because the catechins are converted into more complex polyphenols like theaflavins and thearubigins.[14] Trace amounts of EGCG can also be found in other plant-based foods, including plums, apple skin, hazelnuts, pecans, and carob powder.[9]
The extraction of EGCG from tea leaves is a well-established process. Traditional methods often employ solvent extraction using hot water (as in brewing tea) or organic solvents such as ethanol, ethyl acetate, or acetone.[16] More advanced and selective methods have been developed to improve yield and purity. Supercritical Fluid Extraction (SFE) using carbon dioxide (
CO2) modified with a co-solvent or "entrainer" like ethanol is a prominent example. This technique offers high selectivity and minimizes the use of harsh organic solvents.[14] The purification of the crude extract is a critical step to obtain high-purity EGCG for research or supplementation. This typically involves a decaffeination step, often using a solvent like chloroform, followed by liquid-liquid partitioning and chromatographic techniques to isolate EGCG from other catechins and plant components, achieving purities of over 98%.[8]
While natural extraction is the primary source of EGCG, total chemical synthesis has also been successfully achieved. An enantioselective synthesis route has been developed that involves the stereospecific cyclization of a product from Sharpless asymmetric dihydroxylation, effectively building the complex molecule from three separate aromatic fragments.[17] This synthetic capability is crucial not only for confirming the structure of the natural product but also for producing analogues with modified structures for structure-activity relationship (SAR) studies. Furthermore, chemical modification strategies are actively being explored to create EGCG derivatives with improved physicochemical properties. Examples include the synthesis of EGCG palmitate or various EGCG glycosides, which are designed to enhance stability, water solubility, and potentially bioavailability, thereby addressing some of the inherent limitations of the parent molecule.[18] These synthetic endeavors represent a key research direction aimed at unlocking the therapeutic potential of the EGCG scaffold.
The extensive preclinical investigation of EGCG has revealed a remarkably broad and complex pharmacological profile. Its biological effects are pleiotropic, stemming from its ability to interact with a multitude of molecular targets and modulate a wide array of cellular signaling pathways. This section details the key mechanisms of action that underpin its therapeutic potential, from its dual role as an antioxidant and pro-oxidant to its specific interactions with enzymes and signaling cascades.
The most widely recognized property of EGCG is its potent antioxidant activity.[2] This activity is primarily attributed to its chemical structure, which features eight phenolic hydroxyl groups. These groups act as excellent hydrogen or electron donors, allowing EGCG to effectively scavenge a wide range of reactive oxygen species (ROS) and other free radicals, thereby neutralizing their damaging effects on cellular components like DNA, proteins, and lipids.[10] The mechanism involves the oxidation of these phenolic groups, leading to the formation of more stable semiquinone and quinone products.[10] In addition to direct radical scavenging, EGCG also functions as an antioxidant by chelating transition metal ions, such as iron (
Fe3+) and copper (Cu2+), which can otherwise catalyze the formation of highly reactive hydroxyl radicals via Fenton-type reactions.[23]
Paradoxically, under certain conditions, EGCG can exhibit pro-oxidant activity. This functional switch is not a contradiction but rather a dose- and context-dependent phenomenon. At higher concentrations, or in the presence of metal ions like Fe3+, EGCG can auto-oxidize and generate ROS, including hydrogen peroxide (H2O2) and hydroxyl radicals.[24] This pro-oxidant behavior is believed to be a crucial component of its anticancer mechanism. While low, physiological concentrations of EGCG act as a cytoprotective antioxidant, the high, pharmacological concentrations used in many preclinical cancer studies induce significant oxidative stress within tumor cells. This elevated stress can overwhelm the cancer cell's antioxidant defenses, leading to damage of cellular machinery and ultimately triggering programmed cell death, or apoptosis.[24] This dose-dependent functional shift is a critical concept for understanding both the therapeutic potential and the potential toxicity of EGCG.
Beyond its redox activities, EGCG demonstrates significant anti-inflammatory effects. It modulates several key signaling pathways involved in the inflammatory response. Notably, it has been shown to inhibit the activation of nuclear factor-kappa B (NF-κB), a master regulator of inflammatory gene expression.[8] By blocking NF-κB, EGCG can suppress the production of numerous pro-inflammatory cytokines and enzymes, such as tumor necrosis factor-alpha (TNF-α) and cyclooxygenase-2 (COX-2).[8] It also inhibits the mitogen-activated protein kinase (MAPK) signaling cascade, another critical pathway in inflammation and cellular stress responses.[8]
The pleiotropic effects of EGCG are a direct result of its ability to interact with a vast and diverse range of molecular targets. It does not function as a highly specific ligand for a single receptor but rather as a promiscuous binder that can directly interact with numerous proteins, including cell surface receptors, kinases, and transcription factors, thereby altering their conformation and function.[24] This broad binding profile explains both its wide range of potential therapeutic applications and the significant challenges in developing it as a targeted drug. It acts less like a precision "magic bullet" and more like a "molecular shotgun," simultaneously impacting multiple cellular processes. This lack of specificity makes it difficult to pinpoint a primary mechanism of action for a given disease, complicates dose-response studies, and increases the potential for off-target effects and drug interactions.
A substantial number of enzymes have been identified as direct targets of EGCG inhibition. These targets span multiple disease areas:
Through its direct interactions with key proteins, EGCG modulates several of the most critical intracellular signaling cascades that govern cell fate, including proliferation, survival, and death.
| Target Category | Specific Target / Pathway | Observed Action | Associated Disease Area | Source(s) |
|---|---|---|---|---|
| Enzymes | Fatty Acid Synthase (FASN) | Inhibitor | Cancer | 1 |
| Matrix Metalloproteinase-2 (MMP-2) | Inhibitor | Cancer, Metastasis | 1 | |
| DNA Methyltransferase 1 (DNMT1) | Inhibitor | Cancer (Epigenetics) | 1 | |
| Neutrophil Elastase | Inhibitor | Inflammation | 1 | |
| Hsp90 | Inhibitor | Cancer | 2 | |
| Telomerase | Inhibitor | Cancer | 7 | |
| α-Amylase, α-Glucosidase | Inhibitor | Diabetes, Metabolism | 31 | |
| Transcription Factors | Signal Transducer and Activator of Transcription 3 (STAT3) | Inhibitor | Cancer, Inflammation | 1 |
| Nuclear Factor-kappa B (NF-κB) | Inhibitor | Inflammation, Cancer | 8 | |
| Signaling Pathways | PI3K/Akt/mTOR Pathway | Suppressor | Cancer, Autophagy | 26 |
| MAPK/ERK Pathway | Suppressor | Cancer, Inflammation | 8 | |
| Apoptosis Regulators | B-cell lymphoma 2 (Bcl-2) | Inhibitor | Cancer | 7 |
| Caspases | Activator | Cancer | 26 | |
| Viral Proteins | HIV-1 Gag-Pol polyprotein | Inhibitor | HIV/AIDS | 1 |
| SARS-CoV-2 Mpro, PLpro, S protein | Interacts with / Inhibitor | COVID-19 | 3 | |
The clinical utility of any orally administered therapeutic agent is fundamentally dependent on its pharmacokinetic profile—its absorption, distribution, metabolism, and excretion (ADME). For EGCG, this profile is characterized by significant challenges, most notably its extremely low oral bioavailability. This section provides a critical examination of the ADME properties of EGCG in humans, the factors that influence its systemic exposure, and the strategies being developed to overcome these limitations.
Absorption: Following oral administration, EGCG exhibits very poor and highly variable absorption from the gastrointestinal tract. Human bioavailability studies have consistently demonstrated that only a very small fraction of the ingested dose reaches systemic circulation, with estimates ranging from less than 0.1% to approximately 1.6%.[11] The absorption process is rapid, with peak plasma concentrations (
Tmax) typically observed between 1.3 and 2.2 hours after ingestion in fasted individuals.[12] The primary site of absorption is believed to be the small intestine, where it likely crosses the intestinal epithelium via passive diffusion.[10]
Distribution: Despite its low absorption, the small fraction of EGCG that enters the bloodstream is rapidly distributed throughout the body. Studies in animals have detected EGCG and its metabolites in various tissues, although the levels found represent a minute percentage (0.0003–0.45%) of the total ingested dose.[21]
Metabolism: EGCG undergoes extensive metabolism, which is a primary contributor to its low bioavailability. This metabolic transformation begins immediately upon ingestion and occurs through two major routes:
Excretion: The primary route of elimination for EGCG and its metabolites is through biliary excretion into the feces.[10] Very little of the parent compound (less than 1%) is excreted unchanged in the urine, further highlighting its extensive metabolism and biliary clearance.[10] The biliary excretion pathway also allows for enterohepatic recirculation, where metabolites secreted into the intestine with bile can be reabsorbed, potentially prolonging their presence in the body.[21] The elimination half-life (
t1/2) of parent EGCG from the plasma is relatively short, typically ranging from 1.9 to 5 hours.[11]
The pharmacokinetic profile of EGCG creates a profound "efficacy-safety dilemma" that stands as the single greatest obstacle to its clinical development. The potent biological effects observed in in vitro experiments are typically seen at micromolar concentrations. To achieve these concentrations in human plasma through oral administration, very large doses, often exceeding 1 gram, are required due to the sub-1% bioavailability.[21] However, as detailed in Section 5, oral doses at or above 800 mg/day are precisely the levels at which a statistically significant risk of hepatotoxicity emerges.[34] Thus, the dose required for potential efficacy dangerously overlaps with the dose known to cause toxicity. This narrow therapeutic window, dictated by poor bioavailability, severely limits the practical application of oral EGCG as a systemic therapeutic.
The bioavailability of EGCG is further complicated by a number of influencing factors that contribute to high inter-individual variability:
The significant challenge of low bioavailability has spurred intensive research into novel formulation and prodrug strategies aimed at improving the systemic delivery of EGCG.
| Parameter | Value / Range | Conditions | Source(s) |
|---|---|---|---|
| Bioavailability | < 0.1% – 1.6% | Oral administration | 11 |
| Tmax (Time to Peak) | 1.3 – 2.2 hours | Single oral dose, fasted | 12 |
| t1/2 (Elimination Half-life) | 1.9 – 5 hours | Single oral dose | 11 |
| Cmax (Peak Concentration) | Highly variable, dose-dependent | 50–1600 mg single oral dose | 21 |
| > 1 µM ( 458 ng/mL) achieved with doses > 1 g | 21 | ||
| AUC (Total Exposure) | 442 – 10,368 ng·h/mL | 50–1600 mg single oral dose | 21 |
| Effect of Food | AUC reduced by 2.7–3.9 times | Taken with food vs. fasted | 12 |
| Excretion | Primarily biliary/fecal; <1% in urine (parent) | Oral administration | 11 |
Despite the formidable pharmacokinetic challenges, EGCG has been the subject of numerous clinical trials across a wide range of human diseases. The results of these investigations have been mixed, revealing a complex picture of its therapeutic potential. This section provides a critical synthesis of the available clinical evidence, evaluating its efficacy in neurodegenerative, metabolic, oncologic, and inflammatory conditions.
The neuroprotective properties of EGCG, demonstrated extensively in preclinical models through mechanisms like metal chelation, antioxidant action, and anti-inflammatory effects, have prompted clinical investigation in several neurological disorders.[23]
EGCG's potential to modulate lipid metabolism and inflammation has made it a popular subject for research in cardiovascular and metabolic disorders.
The potent anticancer activities of EGCG observed in countless in vitro and animal studies—where it acts as an antineoplastic agent, an inducer of apoptosis, and an inhibitor of angiogenesis and metastasis—have generated immense hope for its use in cancer prevention and treatment.[2] However, this strong preclinical promise has not yet translated into clear clinical benefit. To date, clinical trials conducted in human populations at increased risk of cancer have generally failed to demonstrate a significant preventive effect of EGCG supplementation.[23] It is noteworthy that some earlier pharmaceutical development programs for EGCG in cancer chemoprevention were reportedly halted for corporate reasons unrelated to efficacy or safety concerns.[28] The stark discrepancy between the preclinical and clinical results in oncology is a major theme in EGCG research and is widely believed to be a direct consequence of the pharmacokinetic challenges that prevent the achievement of sustained, effective anticancer concentrations in human tissues after oral dosing.
A critical evaluation of the overall pattern of clinical trial results reveals that many studies may have been hampered by design flaws that did not adequately account for EGCG's challenging pharmacokinetics. The failure to standardize administration relative to food intake, control for co-administration of potential enhancers, or account for genetic variability in metabolism (e.g., COMT status) could introduce significant pharmacokinetic variability among subjects. This high level of variability could easily mask a true, albeit modest, treatment effect, potentially leading to inconclusive or erroneously negative results. Therefore, many "negative" trials may not be a definitive verdict on EGCG's potential but rather a reflection of the difficulty in achieving consistent and adequate drug exposure.
| Condition | Trial Identifier / Study Type | Phase | Participants (N) | Dosage | Duration | Primary Outcome | Result Summary | Source(s) |
|---|---|---|---|---|---|---|---|---|
| Alzheimer's Disease (Early Stage) | NCT00951834 | 2 / 3 | Not specified | Not specified | Not specified | Not specified | Completed, results not detailed | 39 |
| Down Syndrome | Phase II RCT | 2 | 84 | 9 mg/day + cognitive training | 6 months | Cognitive scores | Significant improvement in visual memory, inhibitory control, adaptive behavior vs. placebo | 23 |
| Multiple Sclerosis (Relapsing-Remitting) | RCT | Not specified | 122 | 800 mg/day | 18 months | Proportion of patients without new hyperintense lesions | No significant improvement in primary outcome or other clinical/radiological parameters | 23 |
| Multiple Sclerosis (Progressive) | RCT | Not specified | 61 | Up to 1200 mg/day | 36 months | Rate of brain atrophy | No significant improvement in primary outcome or any secondary endpoints | 23 |
| Idiopathic Pulmonary Fibrosis (IPF) | NCT05195918 | 1 | Not specified | 300 mg or 600 mg/day | 12 weeks | Safety and PK | Ongoing study to assess safety as add-on therapy | 44 |
| Inflammatory Bowel Disease (IBD) | Clinical Studies | Not specified | Not specified | EGCG-enriched extract | Not specified | Clinical & endoscopic improvement | Positive results leading to FDA Orphan Drug Designation for pouchitis | 28 |
| Cancer Prevention | Clinical Trials | Not specified | Not specified | Varied | Varied | Cancer incidence | Generally no preventive benefits found in people at increased risk | 23 |
While EGCG is often perceived as a benign natural compound, its use in concentrated forms, particularly in dietary supplements, is associated with significant safety concerns. A thorough understanding of its adverse effect profile, potential for toxicity, and drug-drug interactions is essential for its safe use and for navigating the complex global regulatory landscape that has emerged in response to these risks.
The most commonly reported adverse effects associated with EGCG supplementation are mild to moderate and primarily affect the gastrointestinal system. These include nausea, abdominal pain or discomfort, diarrhea, and dyspepsia (heartburn).[42] Other less frequent side effects include headache, dizziness, and muscle pain.[42]
The most critical and dose-limiting safety concern for EGCG is hepatotoxicity, or liver damage.[34] This risk has been identified through case reports and confirmed in systematic reviews of clinical trials. The primary manifestation of this toxicity is an elevation in serum levels of liver enzymes, such as alanine transaminase (ALT) and aspartate transaminase (AST), which are biomarkers of liver cell injury.[35] The evidence strongly indicates that this risk is dose-dependent. Data from interventional clinical trials show that intake of EGCG at doses equal to or above 800 mg per day, particularly when taken as a food supplement, induces a statistically significant increase in serum transaminases compared to placebo.[34] The risk appears to be exacerbated when high doses are taken in a fasted state, which enhances absorption and leads to higher peak plasma concentrations.[45]
Most reported cases of hepatotoxicity have been mild to moderate in severity and were reversible upon discontinuation of the supplement.[45] However, serious adverse events have occurred. There are documented cases of severe liver injury associated with concentrated green tea extracts, including at least one case in Australia that ultimately required a liver transplant.[45]
Regarding other organ systems, animal models have suggested the potential for damage to the heart, kidneys, and pancreas, but these effects were only observed in the context of severe gastrointestinal and liver toxicity and are not considered primary toxicities in humans.[45] Importantly, extensive toxicological reviews have found no evidence to suggest that EGCG is carcinogenic, mutagenic, or genotoxic.[45]
Certain populations may be at higher risk. The immature metabolic capacity of the liver in children may make them more vulnerable to hepatotoxicity.[48] Consequently, regulatory bodies generally advise against the use of concentrated EGCG supplements in children under 18, as well as in pregnant or lactating women, due to a lack of specific safety data in these groups.[46] Individuals with pre-existing liver disease should also exercise extreme caution and consult a healthcare provider before using EGCG supplements.[42]
EGCG has the potential to cause clinically significant drug-drug interactions by altering the pharmacokinetics of co-administered medications.[50] These interactions are not random but are mechanistically predictable, primarily stemming from EGCG's ability to inhibit key drug-metabolizing enzymes and drug transporters in the gut and liver. The most well-documented mechanisms include the inhibition of cytochrome P450 (CYP) enzymes, particularly CYP3A4 and CYP1A2, and the inhibition of drug transporters like P-glycoprotein (P-gp) and organic anion-transporting polypeptide 1A2 (OATP1A2).[50]
These interactions can lead to two opposing, and equally dangerous, clinical outcomes:
Additionally, a pharmacodynamic interaction has been noted with the immunosuppressant drug Etrasimod, where co-administration with EGCG may increase the overall risk or severity of immunosuppression.[1] The potential for these interactions represents a significant and underappreciated clinical risk, especially for patients with cardiovascular disease or cancer who may be taking multiple medications while also self-medicating with high-dose green tea supplements.
The primary contraindication for EGCG is a known hypersensitivity or allergy to EGCG or green tea products.[42] Given the risk of hepatotoxicity, strong caution is advised for individuals with pre-existing liver conditions.[42]
Acute toxicity data from animal studies provide a benchmark for its toxic potential. The median lethal dose (LD50) following oral administration in mice has been reported as 2170 mg/kg.[7] In rats, an oral dose of 2000 mg/kg was found to be lethal.[58] The dermal LD50 in rats is reported as 1860 mg/kg.[57] These values classify EGCG as moderately toxic upon acute ingestion under the Globally Harmonized System (GHS).[7]
The growing body of evidence regarding the risk of hepatotoxicity has prompted major regulatory agencies around the world to take action, resulting in a varied but generally cautious regulatory landscape for EGCG-containing supplements. These regulatory actions are a direct, evidence-based response to the efficacy-safety dilemma, effectively establishing dose caps that are intended to prevent liver injury but may, in doing so, also limit the potential for achieving therapeutic efficacy.
| Interacting Drug / Class | Effect on Drug's Pharmacokinetics | Proposed Mechanism | Potential Clinical Outcome | Source(s) |
|---|---|---|---|---|
| Beta-Blockers | ||||
| Nadolol, Atenolol, Bisoprolol | Decreased AUC and Cmax | Inhibition of intestinal uptake transporter OATP1A2 | Therapeutic Failure (e.g., inadequate blood pressure control) | 52 |
| Statins | ||||
| Atorvastatin (Lipitor) | Decreased absorption | Not specified | Therapeutic Failure (inadequate cholesterol control) | 42 |
| Simvastatin, Rosuvastatin | Increased systemic circulation (AUC) | Inhibition of P-glycoprotein and/or CYP enzymes | Increased Risk of Toxicity (e.g., myopathy) | 54 |
| Calcium Channel Blockers | ||||
| Verapamil, Amlodipine | Increased AUC and Cmax | Inhibition of P-glycoprotein and CYP3A4 | Increased Risk of Toxicity (e.g., hypotension, bradycardia) | 50 |
| Immunosuppressants | ||||
| Etrasimod | Pharmacodynamic interaction | Not specified | Increased Risk of Immunosuppression | 1 |
| Other Cardiovascular Drugs | ||||
| Digoxin | Decreased serum levels | Not specified | Therapeutic Failure (inadequate heart rate control) | 51 |
| Oncology Drugs | ||||
| Tamoxifen, Doxorubicin | Increased serum levels | Inhibition of P-glycoprotein and/or CYP enzymes | Increased Risk of Toxicity | 51 |
This final section synthesizes the comprehensive data presented in this monograph to provide a holistic, expert-level perspective on Epigallocatechin gallate. It bridges the gap between its molecular actions and clinical outcomes, identifies critical knowledge gaps, and outlines key directions for future research to potentially unlock its therapeutic value.
Epigallocatechin gallate presents a compelling yet challenging case study in natural product drug discovery. Its molecular pharmacology is undeniably impressive; the pleiotropic mechanisms of action detailed in Section 2, including potent antioxidant, anti-inflammatory, and antineoplastic activities, provide a strong scientific rationale for its investigation in a multitude of human diseases. The ability of EGCG to act as a "molecular shotgun," modulating dozens of enzymes and critical signaling pathways like NF-κB and PI3K/Akt/mTOR, explains its broad preclinical efficacy.
However, this preclinical promise is profoundly and consistently undermined by its clinical pharmacokinetics, as detailed in Section 3. The fundamental disconnect between its potent in vitro activity and its limited in vivo efficacy stems directly from its extremely poor oral bioavailability. This pharmacokinetic barrier forces the use of high oral doses in an attempt to achieve therapeutic systemic concentrations, which in turn triggers the primary safety concern of dose-dependent hepatotoxicity. This "efficacy-safety dilemma" is the central, unifying theme of the EGCG story.
This dilemma directly explains the mixed and often disappointing results from clinical trials discussed in Section 4. The failure of EGCG to act as a broad disease-modifying agent in conditions like multiple sclerosis or as a cancer preventative is likely a direct consequence of the inability to safely achieve and sustain the high systemic concentrations that would be required to replicate its preclinical effects. Conversely, its successes in more niche areas—such as providing symptomatic relief in MS, showing cognitive benefits in the specific genetic context of Down syndrome, or exerting local anti-inflammatory effects in the gastrointestinal tract for IBD—may represent scenarios where lower, achievable systemic concentrations are sufficient, or where local, non-systemic action is the primary driver of efficacy.
The global regulatory actions detailed in Section 5 are a logical and necessary public health response to this dilemma. By establishing upper intake limits (e.g., <800 mg/day in the EU), regulators have prioritized safety, effectively capping the permissible dose at a level that is intended to prevent liver injury. While essential for consumer protection, this regulatory ceiling further complicates the path forward for clinical research aiming to demonstrate efficacy for conditions that may require higher systemic exposure.
In its native form, therefore, EGCG is best viewed not as a viable standalone oral therapeutic for most chronic systemic diseases, but rather as a highly promising lead compound and a beneficial component of a healthy diet through green tea consumption. Its future as a pharmaceutical agent hinges on overcoming its inherent limitations.
To move beyond the current impasse and realize the therapeutic potential of the EGCG scaffold, future research must be strategically directed at addressing the key knowledge gaps identified in this report.
Published at: September 23, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.