Small Molecule
C19H12O8
13739-02-1
Osteoarthritis in the Hip Joint, Osteoarthritis of the Knee
Diacerein is a small molecule drug of the anthraquinone class, developed as a symptomatic slow-acting drug for osteoarthritis (SYSADOA).[1] Its therapeutic activity is mediated by its active metabolite, rhein, which exerts a unique mechanism of action centered on the inhibition of the pro-inflammatory interleukin-1β (IL-1β) pathway. This mode of action distinguishes it from traditional non-steroidal anti-inflammatory drugs (NSAIDs), as it does not involve the inhibition of prostaglandin synthesis.[3] In its approved indication for osteoarthritis of the hip and knee, clinical evidence demonstrates a modest but statistically significant effect on pain reduction, characterized by a slow onset of action and a notable "carryover effect" where benefits persist after treatment cessation.[1]
However, the clinical utility of oral Diacerein is significantly limited by a challenging safety and tolerability profile. The most prominent adverse event is frequent, often severe, diarrhea, a direct consequence of its anthraquinone structure and pharmacokinetic properties.[1] Furthermore, post-marketing surveillance has revealed a risk of rare but serious hepatotoxicity.[6] These safety concerns have led to significant regulatory restrictions, including recommendations from the European Medicines Agency (EMA) against its use in patients over 65 and in those with liver disease.[3] The drug is not approved by the U.S. Food and Drug Administration (FDA) for osteoarthritis.[7]
In a compelling example of drug repurposing, Diacerein is being successfully re-evaluated as a topical therapy for the rare genetic disorder Epidermolysis Bullosa (EB). In this context, its potent local anti-inflammatory effects can be leveraged while circumventing the systemic toxicities associated with oral administration.[8] This investigational use has earned it an Orphan Drug designation from the FDA, suggesting that the future of Diacerein may lie not in its original indication but in a targeted application for a high-unmet-need rare disease.[10]
The compound is universally identified by its International Nonproprietary Name (INN), Diacerein.[11] Its chemical identity is precisely defined by several systematic names according to IUPAC nomenclature, including 4,5-diacetyloxy-9,10-dioxoanthracene-2-carboxylic acid and 4,5-bis(acetyloxy)-9,10-dihydro-9,10-dioxo-2-anthracenecarboxylic acid.[11]
Throughout its development and in scientific literature, Diacerein has been referred to by various synonyms and codes, such as Diacetylrhein, Diacerhein, DAR, AC-201, and SF-277.[13] Commercially, it is marketed under brand names including Artrodar and Fisiodar.[7] For unambiguous identification across global databases, it is assigned a unique set of identifiers, which are consolidated in Table 2.1.
Diacerein is a synthetic organic compound belonging to the anthraquinone chemical class, a family of compounds based on a tricyclic aromatic quinone structure.[2] This chemical backbone is fundamental to its biological activity and is also implicated in its characteristic adverse effect profile, particularly its laxative effects.[1]
The molecular formula of Diacerein is C19H12O8, corresponding to an average molecular weight of approximately 368.3 g/mol.[3] In its pure form, it presents as a yellow or light yellow crystalline solid or powder.[16] Its solubility is a key physicochemical characteristic influencing its formulation and pharmacokinetics; it is practically insoluble in water, with reported values around 0.01 mg/mL, but is sparingly soluble in organic solvents such as dimethyl sulfoxide (DMSO) and methanol.[3]
Table 2.1: Key Identifiers and Physicochemical Properties of Diacerein
Property / Identifier | Value | Source(s) |
---|---|---|
Generic Name | Diacerein | 14 |
DrugBank ID | DB11994 | 11 |
CAS Number | 13739-02-1 | 13 |
Systematic (IUPAC) Name | 4,5-diacetyloxy-9,10-dioxoanthracene-2-carboxylic acid | 12 |
Molecular Formula | C19H12O8 | 14 |
Average Molecular Weight | 368.297 g/mol | 3 |
Physical Appearance | Yellow powder or crystalline solid | 16 |
ATC Code | M01AX21 | 3 |
PubChem CID | 26248 | 3 |
FDA UNII | 4HU6J11EL5 | 3 |
Water Solubility | Practically insoluble (0.01 mg/mL at 20 °C) | 3 |
Diacerein functions as a prodrug, meaning it is pharmacologically inactive until it is metabolized in the body. Following oral administration, it undergoes complete and rapid deacetylation in the gut wall and liver, yielding its sole active metabolite, rhein.[1] All subsequent anti-inflammatory and chondroprotective effects of the drug are attributable to the actions of rhein. This metabolic activation is a critical and defining feature of its pharmacological profile.
The principal mechanism of action of Diacerein, via rhein, is the potent and multi-level inhibition of the pro-inflammatory cytokine Interleukin-1β (IL-1β). IL-1β is a pivotal mediator in the pathogenesis of osteoarthritis, driving inflammation, cartilage degradation, and pain.[1] The inhibitory action is comprehensive and occurs through several distinct mechanisms:
This multifaceted blockade of the IL-1β system provides a robust mechanistic foundation for its anti-inflammatory effects and its potential to modify the underlying disease process in osteoarthritis.[13]
Beyond its anti-inflammatory properties, Diacerein exhibits characteristics of a Disease-Modifying Osteoarthritis Drug (DMOAD) through direct effects on cartilage homeostasis.[1] These effects are twofold:
Furthermore, studies have shown that Diacerein can exert protective effects against the pathological remodeling of the subchondral bone, an important component of the osteoarthritic joint unit.[1]
Diacerein is classified as a Symptomatic Slow-Acting Drug in Osteoarthritis (SYSADOA), also referred to as a Slow-Acting Drug for Osteoarthritis (SADOA).[1] This classification reflects two key clinical characteristics: a delayed onset of therapeutic effect, which may take several weeks to become apparent, and a persistent "carryover" effect, where clinical benefits are maintained for a period after the drug is discontinued.[1]
The most critical distinction lies in its mechanism compared to that of NSAIDs. Unlike NSAIDs, Diacerein does not inhibit the cyclooxygenase (COX) enzymes and therefore does not interfere with prostaglandin synthesis.[3] This fundamental difference explains its divergent safety profile, notably the significantly lower risk of prostaglandin-mediated adverse effects such as gastrointestinal ulceration, renal impairment, and cardiovascular events that are major concerns with long-term NSAID use.[20] While NSAIDs provide rapid symptomatic relief by targeting inflammation and pain pathways, Diacerein's mechanism targets the upstream cytokine signaling that drives the core catabolic processes of osteoarthritis, providing a theoretical basis for its potential to modify the disease course rather than merely alleviating symptoms.
The pharmacokinetic profile of Diacerein is defined by its behavior as a prodrug, with systemic measurements focused on its active metabolite, rhein.
Following oral administration, Diacerein has a moderate bioavailability, estimated to be between 50% and 60%.[13] It undergoes complete presystemic metabolism via double deacetylation in the gastrointestinal tract and liver, meaning only the active metabolite rhein is detected in systemic circulation.[13]
The co-administration of food has a significant impact on absorption kinetics. A high-fat meal delays the rate of absorption, resulting in a lower peak plasma concentration (Cmax) and a longer time to reach that peak (Tmax).[26] However, the overall extent of absorption, as measured by the area under the curve (AUC), remains unaffected by food.[26] This pharmacokinetic property provides the rationale for the clinical recommendation to administer Diacerein with food, as slowing the absorption rate may improve gastrointestinal tolerability.[6]
Rhein is extensively bound to plasma proteins, with approximately 99% bound primarily to albumin.[3] This high degree of protein binding can influence its distribution into tissues and its potential for drug-drug interactions. The volume of distribution for rhein is reported to be in the range of 15 to 60 liters.[13]
The metabolic pathway of Diacerein is straightforward. The first and most critical step is the complete conversion of the prodrug to rhein.[3] Subsequently, rhein undergoes Phase II conjugation reactions in the liver to form more water-soluble metabolites, rhein glucuronide and rhein sulfate, which facilitates their elimination from the body.[3]
The primary route of elimination for rhein and its conjugates is through the kidneys.[3] Animal studies in rats suggest that approximately 37% of a dose is excreted in the urine and 53% in the feces.[13] The elimination half-life (
t1/2) of rhein in plasma is approximately 4 to 10 hours.[3]
The pharmacokinetic profile provides a direct explanation for both the therapeutic action and the primary adverse effect of Diacerein. While the prodrug design ensures systemic delivery of the active anti-inflammatory metabolite rhein, the incomplete absorption of the drug from the upper gastrointestinal tract is a critical factor. The unabsorbed portion of the drug and its metabolite, which belong to the laxative-acting anthraquinone class, reaches the colon. There, it exerts a local irritant effect, leading to the characteristic and dose-limiting side effect of diarrhea.[1] This clear link between pharmacokinetics and adverse events has directly motivated research into novel formulations, such as gastroretentive systems, designed to enhance absorption in the upper GI tract and thereby minimize the amount of active substance reaching the colon.[29]
Table 4.1: Summary of Pharmacokinetic Parameters for Diacerein/Rhein
Parameter | Value | Source(s) |
---|---|---|
Bioavailability | 50-60% | 13 |
Tmax (fasted) | ~2.6 hours | 26 |
Tmax (fed) | ~3.8 hours | 26 |
Cmax (fasted, 50 mg dose) | ~4471 ng/mL | 26 |
Cmax (fed, 50 mg dose) | ~3225 ng/mL | 26 |
Elimination Half-life (t1/2) | 4-10 hours | 3 |
Plasma Protein Binding | ~99% | 3 |
Route of Elimination | Primarily Renal | 3 |
Diacerein is approved in several countries for the symptomatic treatment of osteoarthritis (OA) affecting the hip or knee.[6] Its therapeutic effect is characterized as delayed, and it is noted to be less effective in patients with rapidly progressive forms of hip OA.[6]
A substantial body of clinical evidence, including multiple randomized controlled trials and meta-analyses, has established a consistent, albeit modest, efficacy profile. A 2014 Cochrane systematic review concluded that Diacerein has a "small beneficial effect on pain" when compared to placebo.[3] This effect was quantified as an average 9-point greater reduction in pain on a 100-point visual analogue scale (VAS) over treatment periods of 3 to 36 months.[24] In contrast, its effect on improving physical function has generally not reached statistical significance in these analyses.[24] There is also low-quality evidence suggesting that Diacerein may slightly slow the rate of joint space narrowing (a marker of structural progression) in the hip, though a similar effect has not been demonstrated in the knee.[24] When compared directly with NSAIDs, Diacerein demonstrates comparable efficacy for pain relief during the active treatment phase.[5]
A distinguishing clinical feature of Diacerein and other SYSADOAs is the presence of a "carryover effect." This refers to the sustained therapeutic benefit, particularly pain relief, that persists for several weeks to months after the cessation of treatment.[1] This phenomenon stands in stark contrast to the rapid waning of effects seen upon discontinuation of NSAIDs.[33] The carryover effect lends clinical support to the hypothesis that Diacerein's mechanism involves a deeper, disease-modifying action on cartilage and synovial tissue, rather than purely symptomatic modulation.
A significant and promising new direction for Diacerein is its repurposing as a topical therapy for Epidermolysis Bullosa (EB), a group of rare and severe genetic blistering skin disorders.[8] Specifically, a 1% ointment formulation (coded as AC-203 or CCP-020) is under active investigation for Epidermolysis Bullosa Simplex (EBS).[35]
The therapeutic rationale in EB stems from Diacerein's ability to inhibit IL-1β. In EBS, mutations in keratin genes (KRT5/KRT14) lead to a pathogenic inflammatory feedback loop involving IL-1β and keratin 14 overexpression, which destabilizes the epidermis.[34] By disrupting this cycle locally, topical Diacerein has been shown to reduce blister formation.[34] Clinical trials have yielded promising results; a phase 2/3 crossover study demonstrated a statistically significant reduction in blister counts in patients treated with 1% Diacerein cream compared to placebo.[37]
This potential has been recognized by regulatory agencies. The U.S. FDA granted Diacerein Orphan Drug designation in November 2018 and Fast Track designation for the treatment of EB, highlighting the high unmet medical need.[8] Ongoing international phase 2/3 trials are further evaluating its long-term efficacy and safety.[35] There is also a case report suggesting potential efficacy in recessive dystrophic EB (RDEB), a different subtype of the disease.[39]
This clinical development path highlights a critical therapeutic pivot. The oral formulation for osteoarthritis is constrained by a narrow therapeutic window due to modest efficacy and significant systemic side effects. In contrast, the topical formulation for EB offers a solution to this risk-benefit challenge. By delivering the drug directly to the skin, it maximizes local anti-inflammatory efficacy at the site of pathology while minimizing systemic absorption, thereby avoiding the dose-limiting gastrointestinal and hepatic toxicities associated with oral use.[8] This change in administration route effectively unlocks the drug's potent mechanism for a new indication where its benefits can be realized with a much more favorable safety profile.
Diacerein has also been investigated for its potential role in metabolic diseases. It has been studied for the treatment of Type 2 Diabetes Mellitus and Metabolic Dysfunction-associated Steatohepatitis (MASH).[13] A completed Phase 3 clinical trial (NCT02242149) was conducted to evaluate its efficacy on glycemic control and liver fat content in subjects with Type 2 Diabetes, suggesting interest in its potential systemic metabolic effects.[7]
The use of oral Diacerein is defined as much by its safety profile as by its efficacy. The risks associated with its use have led to significant restrictions and require careful patient selection and monitoring.
The most frequently reported adverse events are gastrointestinal in nature. Diarrhea is classified as a "very common" side effect, affecting more than 1 in 10 patients, with abdominal pain also being very common.[1] Meta-analyses have calculated the risk ratio for developing diarrhea with Diacerein versus placebo to be approximately 3.5, with some studies reporting that up to 39% of patients experience at least one episode.[1]
These effects are typically mild to moderate in severity, tend to occur within the first two to four weeks of initiating therapy, and often subside with continued treatment.[1] However, severe cases of diarrhea have been reported, which can lead to complications such as dehydration and electrolyte imbalances, a particular concern in elderly patients.[1]
A less frequent but more serious risk associated with Diacerein is hepatotoxicity. Post-marketing surveillance has identified cases of acute liver injury, including clinically significant elevations in serum hepatic enzymes and cases of hepatitis.[3] Most of these events occurred within the first few months of treatment. Although preclinical toxicology studies did not identify the liver as a target organ for toxicity, the mechanism of liver injury in humans is suspected to be idiosyncratic.[1] This risk has prompted regulatory agencies to issue strong warnings and contraindicate its use in patients with liver disease.[3]
Table 6.1: Incidence of Key Adverse Events from Clinical Trials (Diacerein vs. Placebo)
Adverse Event | Diacerein Incidence | Placebo Incidence | Absolute Risk Increase | Risk Ratio (95% CI) | Source(s) |
---|---|---|---|---|---|
Diarrhea | 36 per 100 patients | 10 per 100 patients | 26% | 3.52 (2.42 - 5.11) | 1 |
Any Adverse Event | 42 per 100 patients | 18 per 100 patients | 24% | - | 42 |
Withdrawal due to AE | 18 per 100 patients | 13 per 100 patients | 5% | 1.29 (0.83 - 2.01) | 24 |
Based on its safety profile, the use of Diacerein is subject to several important restrictions:
The safety of Diacerein is influenced by its potential for interactions with other drugs, as well as with food and alcohol. The overall risk profile can be understood as a combination of a predictable, mechanism-based gastrointestinal effect, a rare but serious idiosyncratic hepatic effect, and a broad potential for pharmacokinetic drug-drug interactions. This last category is particularly important in the typical elderly osteoarthritis patient, who is often on multiple concurrent medications.
Pharmacokinetic data suggest that Diacerein and its active metabolite rhein can inhibit various metabolic enzymes, including members of the Cytochrome P450 (CYP) 3A subfamily.[44] This creates a broad potential for drug-drug interactions by decreasing the metabolism of co-administered drugs that are substrates for these enzymes. This can lead to elevated plasma concentrations and an increased risk of toxicity for a wide range of medications. The list of potentially affected drugs is extensive and includes agents from many common therapeutic classes, such as anticoagulants (acenocoumarol, apixaban), analgesics (acetaminophen, where hepatotoxic risk may be increased), antidepressants (amitriptyline), and antidiabetic agents (acetohexamide).[14]
The regulatory status of Diacerein varies significantly across different regions, reflecting divergent assessments of its benefit-risk profile for osteoarthritis.
The following dosing recommendations apply to the approved indication of symptomatic treatment of osteoarthritis of the hip and knee. Treatment should only be initiated and supervised by physicians experienced in managing osteoarthritis.[6]
Diacerein occupies a complex and evolving position in the therapeutic landscape. As an oral agent for osteoarthritis, it is a drug defined by a compelling mechanistic rationale that has been largely overshadowed by a challenging clinical profile. Its unique IL-1β inhibitory action offers a theoretical advantage over traditional NSAIDs by targeting the underlying pathophysiology of cartilage degradation. However, this potential has not translated into superior clinical efficacy, with studies consistently showing only a modest benefit for pain relief. This limited efficacy is difficult to balance against a significant safety and tolerability burden, dominated by a high incidence of diarrhea and the rare but serious risk of idiosyncratic hepatotoxicity. Consequently, its role in osteoarthritis has been curtailed by regulatory bodies, confining it to a niche population where other first-line agents are contraindicated.
In stark contrast, the repurposing of Diacerein as a topical formulation for Epidermolysis Bullosa represents a remarkable therapeutic pivot. This strategic shift is a salient example of modern pharmaceutical development, where a deep understanding of a drug's mechanism can be applied to a new disease context. By changing the route of administration, developers have successfully uncoupled the drug's potent local anti-inflammatory effects from the systemic toxicities that limited its original application. This approach allows the targeted delivery of the active molecule to the site of pathology, maximizing benefit while minimizing risk.
In conclusion, while Diacerein's journey as a systemic treatment for the widespread condition of osteoarthritis appears to be one of limited and declining utility, its future as a targeted, topically delivered orphan drug for a severe rare disease is highly promising. This evolution underscores a fundamental principle in pharmacology: the ultimate value of a molecule is determined not just by its intrinsic mechanism, but by the precision with which that mechanism can be applied to a specific clinical problem.
Published at: September 6, 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.