Indomethacin (DB00328): A Comprehensive Pharmacological and Clinical Monograph
Executive Summary
Indomethacin is a potent non-steroidal anti-inflammatory drug (NSAID) belonging to the indole-acetic acid class of compounds.[1] Since its introduction, it has been recognized for its powerful analgesic, antipyretic, and anti-inflammatory properties, establishing it as one of the most effective agents in its class for suppressing inflammation.[1] The primary mechanism of action involves the potent and non-selective inhibition of cyclooxygenase (COX) enzymes. Notably, Indomethacin demonstrates a greater inhibitory preference for COX-1 over COX-2, a characteristic that is fundamental to both its therapeutic efficacy and its significant profile of adverse effects, particularly gastrointestinal toxicity.[4]
Clinically, Indomethacin is approved for a range of conditions, including the management of moderate to severe inflammatory arthritides such as rheumatoid arthritis and ankylosing spondylitis, acute gouty arthritis, and acute painful shoulder from bursitis or tendinitis.[1] Beyond these common indications, it holds unique and critical therapeutic roles. In neonatology, the intravenous formulation is the standard of care for inducing the closure of a hemodynamically significant patent ductus arteriosus (PDA) in premature infants.[1] In neurology, it is uniquely effective for a group of disorders known as "indomethacin-responsive headaches," often serving as both a treatment and a diagnostic tool.[6]
Despite its efficacy, the clinical utility of Indomethacin is constrained by a significant risk profile. The U.S. Food and Drug Administration (FDA) has mandated Black Box Warnings for the risk of serious, and potentially fatal, cardiovascular thrombotic events and life-threatening gastrointestinal bleeding, ulceration, and perforation.[2] Its extensive list of potential adverse effects, including central nervous system and renal toxicity, often limits its application to short-term therapy or positions it as a second-line agent when safer alternatives have proven inadequate.[7]
In contemporary practice, while its role in the long-term management of chronic conditions has diminished in favor of drugs with more favorable safety profiles, Indomethacin remains an indispensable therapeutic agent for specific acute inflammatory states and niche indications where its high potency is paramount and the duration of treatment can be limited. The emerging field of pharmacogenomics, particularly concerning variants in the CYP2C9 gene, presents a future pathway to personalize therapy, potentially mitigating its inherent risks and refining its place in modern medicine.[9]
Historical Context and Chemical Profile
This section details the foundational chemical and historical information of Indomethacin, contextualizing its development within the broader history of pharmacology and providing the essential data for chemists, pharmacists, and formulation scientists.
Discovery and Development
Indomethacin was first discovered by scientists at Merck Sharp & Dohme in 1963 and received its initial approval for medical use from the U.S. FDA in 1965, having been patented in 1961.[1] Its development was part of a significant wave of research in the 1960s that produced several acetic acid derivative NSAIDs, including diclofenac and sulindac.[1]
A crucial aspect of Indomethacin's history is that it was developed and introduced into clinical practice before the fundamental mechanism of action for NSAIDs was fully elucidated. The discovery by Dr. John Vane that these drugs work by inhibiting the enzyme cyclooxygenase, thereby blocking the synthesis of prostaglandins, came after Indomethacin was already in use.[10] This historical context is not merely a footnote; it is the central explanation for the drug's overarching clinical profile. Engineered for maximum anti-inflammatory potency against a then-poorly understood biological target, the resulting molecule was a powerful but non-selective agent. The subsequent discovery of two distinct COX isoforms—the "housekeeping" COX-1 responsible for physiological functions like gastric protection, and the "inducible" COX-2 associated with inflammation—revealed that Indomethacin was a "blunt instrument." Its design, focused solely on potency, could not have aimed for the COX-2 selectivity that is the cornerstone of modern NSAID safety design. This legacy of non-selective, potent inhibition is the direct cause of its dual reputation as both a highly effective therapeutic and a highly toxic agent. The drug quickly established itself as one of the most potent inhibitors of prostaglandin synthesis available, a reputation it still holds.[3] Despite the availability of newer agents, it remains a frequently prescribed medication, with over 1 million prescriptions dispensed in the United States in 2022.[7]
Chemical Identity and Nomenclature
Indomethacin is identified and classified by numerous international standards.
- DrugBank ID: DB00328 [1]
- CAS Number: 53-86-1 [1]
- IUPAC Name: 2-{1-[(4-Chlorophenyl)carbonyl]-5-methoxy-2-methyl-1H-indol-3-yl}acetic acid [7]
- Synonyms: Indometacin, Indometacina, Indometacine, Indometacinum.[1] It is marketed under various brand names, including Indocin®, Tivorbex®, Amuno, and Artracin.[12]
- Chemical Classification: It is classified as an N-acylindole and a member of the indole-3-acetic acids, monochlorobenzenes, and aromatic ethers.[1]
- Other Identifiers: Additional identifiers include UNII XXE1CET956, EC Number 200-186-5, ChEBI ID CHEBI:49662, and ChEMBL ID CHEMBL6.[1]
Physicochemical Properties
The chemical structure and physical properties of Indomethacin are integral to its pharmacological activity and formulation.
- Chemical Structure: The molecule consists of an indole-3-acetic acid core. The indole ring is substituted at three key positions: a p-chlorobenzoyl group at position 1, a methyl group at position 2, and a methoxy group at position 5.[1]
- Molecular Formula: C19H16ClNO4 [12]
- Molecular Weight: Approximately 357.79 g/mol [4]
- Physical Appearance: It is a crystalline powder, typically pale-yellow to yellow-tan in color. It is odorless or has a slight odor and a slight, bitter taste.[1]
- Polymorphism: Indomethacin is known to exhibit polymorphism, meaning it can exist in multiple crystalline forms. At least three anhydrous forms (designated α, β, and γ) and various solvates have been identified.[1] This property is critically important for pharmaceutical manufacturing and formulation, as different polymorphs can have different melting points, dissolution rates, and stability, which in turn affect the drug's bioavailability. The two most commonly cited crystalline forms have distinct melting points around 155 °C and 162 °C.[1]
- Melting Point: Due to polymorphism, reported melting points vary, but generally fall within the range of 158 °C to 162 °C.[1]
- pKa: The acid dissociation constant (pKa) of Indomethacin is 4.5.[3] As a weak acid, this value is fundamental to its pharmacokinetic behavior.
- Solubility: It is practically insoluble in water, with a reported solubility of 0.937 mg/L at 25 °C.[1] It is sparingly soluble in alcohol but more soluble in organic solvents such as acetone, ether, and chloroform.[4] This low aqueous solubility presents a challenge for formulation, leading to the development of products like Tivorbex®, which uses micronized particles to enhance the dissolution rate and absorption.[20]
- Stability: Indomethacin is stable in neutral or slightly acidic media but is susceptible to decomposition in strong alkaline solutions. It is also light-sensitive and should be stored protected from light.[3]
The physicochemical properties of Indomethacin create a "perfect storm" for gastric toxicity through a mechanism known as ion trapping. First, as a weak acid with a pKa of 4.5, Indomethacin exists primarily in its un-ionized, lipid-soluble form in the highly acidic environment of the stomach (pH 1-2).[3] This lipophilic form allows it to readily diffuse across the lipid membranes of the gastric mucosal cells into the much more alkaline intracellular environment (pH ~7.4).[7] Once inside the cell, where the pH is significantly higher than its pKa, the drug rapidly dissociates into its ionized, water-soluble (anionic) form. This charged molecule cannot easily diffuse back across the cell membrane, causing it to become effectively "trapped" at high concentrations within the very cells that line the stomach.[7] This high intracellular accumulation, combined with the simultaneous local inhibition of cytoprotective prostaglandins due to COX-1 inhibition, leads to direct cellular damage and provides a clear molecular-to-clinical explanation for the high incidence of peptic ulcers and gastrointestinal bleeding associated with the drug.[5]
Synthesis and Formulation
The synthesis of Indomethacin has evolved over time. The "traditional" synthesis is based on the Fischer indole synthesis, a classic method for constructing the indole ring system. In this route, the indole core is formed first from precursors like 4-methoxyphenyl hydrazine and methyl levulinate, followed by N-acylation with 4-chlorobenzoyl chloride and, finally, hydrolysis of the methyl ester to yield the active carboxylic acid.[21] Recognizing the need for greater efficiency, scientists at Merck developed an elegant, improved two-step synthesis that reverses the order of operations, coupling the chlorobenzamide group onto the hydrazine precursor before the indole ring formation step.[21] More recently, advanced organometallic variations using organozinc reagents have been reported, offering milder reaction conditions and broadening the scope for creating functionalized analogs.[22]
To overcome its poor water solubility, Indomethacin is formulated into a variety of dosage forms using numerous pharmaceutical excipients. These include binders and fillers like microcrystalline cellulose, disintegrants such as carboxymethyl cellulose sodium, and various polymers (e.g., hypromellose, ethyl cellulose) to control drug release in extended-release formulations.[23]
Table 1: Chemical and Physical Properties of Indomethacin
Property | Value | Source(s) |
---|
IUPAC Name | 2-{1-[(4-Chlorophenyl)carbonyl]-5-methoxy-2-methyl-1H-indol-3-yl}acetic acid | 7 |
CAS Number | 53-86-1 | 1 |
Molecular Formula | C19H16ClNO4 | 12 |
Molecular Weight | 357.79 g/mol | 4 |
Physical Appearance | Pale-yellow to yellow-tan crystalline powder | 1 |
Polymorphism | Exhibits polymorphism (α, β, γ forms and solvates) | 1 |
Melting Point Range | 158–162 °C (varies with polymorph) | 1 |
pKa | 4.5 | 3 |
Water Solubility | 0.937 mg/L (at 25 °C); practically insoluble | 1 |
Solubility in Organic Solvents | Soluble in ethanol, ether, acetone, chloroform | 4 |
Stability Notes | Decomposes in strong alkali; light-sensitive | 3 |
Comprehensive Pharmacology
This section provides an in-depth analysis of how Indomethacin interacts with the body, from its molecular targets to its systemic effects and metabolic fate. It dissects the complex interplay of factors that define its potent therapeutic action and significant toxicity.
Mechanism of Action
The pharmacological effects of Indomethacin stem from its interaction with multiple biological targets, with its primary mechanism being well-established.
Primary Mechanism: Non-selective COX Inhibition
Indomethacin is a potent, non-specific, and reversible inhibitor of the cyclooxygenase (COX) enzymes, COX-1 and COX-2, which are also known as prostaglandin G/H synthase.1 These enzymes catalyze the rate-limiting step in the biosynthesis of prostaglandins from their precursor, arachidonic acid. By binding to the active site of the COX enzymes, Indomethacin prevents this conversion, thereby reducing the levels of prostaglandins, which are critical mediators of inflammation, pain (by sensitizing afferent nerves), and fever.3
A defining characteristic of Indomethacin is its greater inhibitory potency against COX-1 compared to COX-2. While the exact inhibitory concentrations (IC50) vary depending on the assay system used, the preference for COX-1 is a consistent finding. For example, studies have reported IC50 values of 230 nM for human COX-1 versus 630 nM for human COX-2, and in another assay, 1.67 µM for human COX-1 versus 24.6 µM for human COX-2.[4] This preferential inhibition of the constitutively expressed "housekeeping" COX-1 enzyme—which is responsible for vital physiological functions such as maintaining the protective mucosal lining of the gastrointestinal tract and supporting platelet aggregation—is the principal mechanistic explanation for Indomethacin's high incidence of gastric adverse effects relative to more COX-2 selective NSAIDs.[6]
Secondary and Novel Mechanisms
Beyond its primary action on COX enzymes, Indomethacin exhibits a more complex, multi-target profile that contributes to its overall effects:
- Phospholipase A2 (PLA2) Inhibition: Uniquely among many NSAIDs, Indomethacin has also been shown to inhibit phospholipase A2. This enzyme is responsible for releasing arachidonic acid from the phospholipid cell membrane, making it available as a substrate for COX. By acting at this upstream point in the inflammatory cascade, Indomethacin can further limit the production of pro-inflammatory mediators.[6]
- Peroxisome Proliferator-Activated Receptor (PPAR) Activation: Indomethacin is a known activator of PPARα and PPARγ, with a reported half-maximal effective concentration (EC50) of 40 nM.[4] PPARs are nuclear receptors that regulate gene expression related to inflammation and metabolism, and their activation may contribute to the anti-inflammatory effects of Indomethacin.
- Cannabinoid Receptor Modulation: More recently, Indomethacin has been identified as a positive allosteric modulator (PAM) of the cannabinoid 1 (CB1) receptor. As a PAM, it does not directly activate the receptor but enhances the binding and signaling of endogenous cannabinoids, such as anandamide. This mechanism may contribute to its analgesic effects through an alternative pathway, potentially with a lower risk of the psychoactive side effects associated with direct CB1 agonists.[7]
- Other Cellular Effects: Research has indicated that Indomethacin can influence other inflammatory processes. It has been shown to inhibit the migration of polymorphonuclear leukocytes to sites of inflammation, stabilize lysosomal membranes to prevent the release of tissue-damaging enzymes, and reduce the cellular production of reactive oxygen species (ROS), thereby mitigating oxidative stress.[5] Additionally, it can induce the interferon-induced, double-stranded RNA-activated protein kinase (PKR), a mechanism which may underlie reports of its antiviral and anticancer properties.[6]
Pharmacodynamics
The inhibition of prostaglandin synthesis by Indomethacin translates into several key pharmacodynamic effects that define its clinical use.
- Core Effects: The reduction of prostaglandin levels in peripheral tissues and the central nervous system results in Indomethacin's three cardinal therapeutic properties: potent anti-inflammatory, analgesic, and antipyretic actions.[1] Its anti-inflammatory effect is particularly noteworthy, with some studies suggesting it is more potent than phenylbutazone and comparable to corticosteroids in certain experimental models.[11]
- Cerebral Hemodynamics: A unique and clinically significant pharmacodynamic property of Indomethacin is its ability to act as a potent cerebral vasoconstrictor. It has been shown to decrease cerebral blood flow and inhibit cerebrovascular reactivity to carbon dioxide more consistently than other NSAIDs.[3] While this effect appears to be transient with oral administration, it is the likely basis for its efficacy in treating conditions of raised intracranial pressure and its specific, often diagnostic, effectiveness in the "indomethacin-responsive" headache syndromes.[7]
- Closure of Patent Ductus Arteriosus (PDA): In the fetus and premature neonate, the patency of the ductus arteriosus—a blood vessel connecting the pulmonary artery to the aorta—is maintained by the vasodilatory effects of circulating prostaglandins, particularly PGE2. After birth, this vessel normally closes. In premature infants where it fails to close, Indomethacin is used as a life-saving intervention. By potently inhibiting the synthesis of PGE2, it removes the primary stimulus keeping the ductus open, allowing it to constrict and functionally close.[2]
- Renal Effects: Prostaglandins play a crucial role in maintaining renal hemodynamics, particularly by promoting vasodilation to ensure adequate blood flow. By inhibiting renal prostaglandin synthesis, Indomethacin can lead to adverse effects such as sodium and fluid retention, peripheral edema, hyperkalemia (high potassium levels), and the onset or worsening of hypertension.[7] This same mechanism is responsible for blunting the therapeutic efficacy of many antihypertensive medications, including ACE inhibitors, ARBs, and diuretics.[28]
Pharmacokinetics: ADME Profile
The disposition of Indomethacin in the body is characterized by rapid absorption, extensive protein binding, and complex elimination patterns.
- Absorption: Following oral administration, Indomethacin is readily and rapidly absorbed from the gastrointestinal tract, with a bioavailability approaching 100%.[2] Peak plasma concentrations (Tmax) are typically achieved within 1 to 2 hours in a fasting state.[2] The presence of food slows the rate of absorption, delaying the time to peak concentration, but does not significantly reduce the overall amount of drug absorbed (the area under the curve, or AUC).[30] When administered rectally as a suppository, absorption can be faster but is often incomplete and more variable, with bioavailability estimated at 80–90%.[11]
- Distribution: As a highly lipophilic molecule, Indomethacin distributes widely into body tissues. It achieves high concentrations in synovial fluid, which is relevant to its use in arthritis, and is known to cross both the blood-brain barrier and the placenta.[2] In the bloodstream, it is extensively bound (90–99%) to plasma proteins, primarily albumin.[6] Its apparent volume of distribution has been reported in the range of 0.34 to 1.57 L/kg.[6]
- Metabolism: Indomethacin undergoes extensive metabolism in the liver. The primary metabolic pathways are O-demethylation (of the methoxy group) and N-deacylation (removal of the chlorobenzoyl group) to form several major metabolites, such as O-desmethyl-indomethacin and N-deschlorobenzoyl-indomethacin.[2] These metabolites are largely pharmacologically inactive and are subsequently conjugated with glucuronic acid for excretion. A critical and defining feature of Indomethacin's pharmacokinetics is its significant and highly variable enterohepatic circulation. In this process, the glucuronide conjugates are excreted into the bile, travel to the intestine where they are hydrolyzed by gut bacteria back into the active parent drug, and are then reabsorbed into the circulation.[2]
- Excretion: The elimination of Indomethacin from the body is biphasic and occurs through multiple routes. Approximately 60% of an administered dose is excreted in the urine, both as the parent drug and its metabolites, via renal tubular secretion. The remaining 33% is eliminated in the feces after biliary secretion.[2]
The erratic and extensive nature of its enterohepatic circulation is a critical, often underappreciated, pharmacokinetic feature that directly contributes to Indomethacin's challenging clinical profile. This recycling process is highly variable among individuals, with estimates of reabsorption ranging from 27% to as high as 115% of the dose.[6] This variability is the primary reason for the exceptionally wide range reported for its elimination half-life, from as short as 1.5 hours to as long as 16 hours, with an average of around 4.5 to 7 hours.[2] A long and unpredictable half-life means that the drug can accumulate unexpectedly in certain individuals, even with standard dosing regimens. This accumulation increases the duration and intensity of systemic drug exposure, particularly the inhibition of COX-1 in the gastrointestinal tract, which directly elevates the risk of ulceration and bleeding. This pharmacokinetic quirk is therefore not just a technical detail; it is a direct mechanistic link to increased toxicity and a key reason why the drug can be difficult to manage safely. In premature infants, clearance is significantly reduced, resulting in a much longer half-life (mean of ~20 hours), which necessitates careful, age-adjusted dosing regimens.[6]
Pharmacogenomics
The metabolism of Indomethacin is significantly influenced by genetic factors, primarily related to the cytochrome P450 enzyme system.
- CYP2C9 Metabolism: The enzyme CYP2C9 is a major pathway involved in the metabolic clearance of Indomethacin.[26]
- Genetic Polymorphisms: The gene encoding this enzyme, CYP2C9, is known to have several common polymorphisms that result in reduced enzyme function. The most studied of these are the CYP2C9*2 and CYP2C9*3 alleles.
- Clinical Consequences: The clinical impact of these genetic variants is twofold. In neonates being treated for PDA, carriage of these reduced-function alleles has been associated with treatment failure, suggesting that standard doses may be insufficient to achieve the necessary therapeutic concentrations in individuals who are "poor metabolizers".[9] Conversely, in other populations, these same polymorphisms are associated with an increased risk of NSAID-related gastrointestinal bleeding.[9] This is because reduced metabolism leads to higher-than-expected plasma concentrations and prolonged exposure to the drug, increasing the risk of toxicity. In one report, a patient homozygous for the
CYP2C9*3 variant experienced major bleeding when treated with a combination of Indomethacin and an anticoagulant.[9]
This pharmacogenomic data transforms the perception of Indomethacin from a "one-size-fits-all" drug into a candidate for precision medicine. The evidence suggests that a significant proportion of adverse events and treatment failures are not random occurrences but are genetically predictable. This raises the possibility that pre-emptive CYP2C9 genotyping could one day become a standard of care before initiating Indomethacin therapy, particularly in high-risk scenarios such as in neonates or in elderly patients receiving anticoagulants. Such testing would allow for individualized dose adjustments or the selection of an alternative drug, fundamentally improving the benefit-risk assessment and moving this legacy drug toward a more modern, personalized therapeutic paradigm.
Table 2: Summary of Pharmacokinetic Parameters
Parameter | Value/Range | Clinical Significance/Note | Source(s) |
---|
Bioavailability (Oral) | ~100% | Excellent and reliable absorption from oral forms. | 2 |
Bioavailability (Rectal) | 80–90% | Absorption is faster but less complete and more variable than oral. | 11 |
Tmax (Fasted) | 1–2 hours | Rapid onset of action. | 2 |
Tmax (Fed) | Delayed | Food slows absorption rate but not total amount absorbed. | 30 |
Plasma Protein Binding | 90–99% (Albumin) | High binding limits free drug concentration but creates potential for displacement interactions. | 6 |
Volume of Distribution | 0.34–1.57 L/kg | Distributes widely into tissues, including synovial fluid and CNS. | 6 |
Major Metabolic Pathways | O-demethylation, N-deacylation, Glucuronidation | Extensive hepatic metabolism to inactive metabolites. | 2 |
Half-life (Adult) | 1.5–16 hours (avg. 4.5–7 h) | Highly variable due to enterohepatic circulation. | 2 |
Half-life (Neonate) | ~20 hours | Significantly prolonged, requiring dose adjustments. | 6 |
Excretion Routes | ~60% Renal, ~33% Fecal | Dual elimination pathways. | 2 |
Impact of Enterohepatic Circulation | Extensive and erratic | Primary cause of high inter-individual variability in half-life; complicates predictable dosing and increases risk of accumulation and toxicity. | 2 |
Clinical Applications and Administration
This section bridges pharmacology with clinical practice, detailing the approved and off-label uses of Indomethacin, and providing comprehensive guidance on its administration across different patient populations and disease states.
Approved and Off-Label Indications
Indomethacin is utilized for a wide spectrum of inflammatory and pain-related conditions.
FDA-Approved Indications:
- Musculoskeletal and Joint Disorders: It is indicated for the management of moderate to severe rheumatoid arthritis (including acute flares of chronic disease), moderate to severe osteoarthritis, and moderate to severe ankylosing spondylitis.[1]
- Acute Inflammatory Conditions: It is approved for the treatment of acute gouty arthritis and acute painful shoulder resulting from bursitis and/or tendinitis.[1]
- Neonatal Cardiology: The intravenous formulation is specifically approved for the closure of a hemodynamically significant patent ductus arteriosus (PDA) in premature infants when standard medical management has failed.[1]
- Acute Pain: The lower-dose formulation, Tivorbex®, is specifically indicated for the relief of mild-to-moderate acute pain in adults.[12]
Common Off-Label and Investigational Uses:
- Headache Syndromes: Indomethacin has a unique and profound efficacy in a specific group of primary headache disorders, often referred to as "indomethacin-responsive headaches." These include paroxysmal hemicrania and hemicrania continua, for which a positive response is considered diagnostic. It is also used off-label for cluster headaches.[6]
- Tocolysis: Due to its ability to inhibit prostaglandin-mediated uterine contractions, it has been used off-label as a tocolytic agent to suppress preterm labor.[2]
- Renal Disorders: It is used off-label in the management of certain rare salt-wasting kidney diseases, such as Bartter syndrome and Gitelman syndrome, and to reduce polyuria in nephrogenic diabetes insipidus.[12]
- Ophthalmology: While not a systemic use, ophthalmic preparations of Indomethacin have been studied and used for the management of postoperative ocular inflammation and pain, for instance, following cataract surgery.[1]
Dosage Forms and Strengths
Indomethacin is available in multiple formulations to suit different clinical needs, although not all forms are interchangeable or approved for all indications. The diverse range of indications for Indomethacin, from chronic arthritis to neonatal PDA to specific headaches, highlights its powerful, non-specific mechanism. However, this breadth also creates significant potential for clinical error. The pathophysiology, dosing regimens, and appropriate formulations vary drastically between these unrelated conditions. For example, the extended-release capsules appropriate for chronic arthritis are contraindicated for the high-dose, short-burst therapy required for acute gout.[35] This places a high burden on clinicians to be acutely aware of the indication-specific nuances of dosing, formulation choice, and duration of therapy to prevent medication errors and avoidable toxicity. The drug's versatility is, in practice, a source of its complexity.
Table 3: Available Dosage Forms and Strengths
Formulation | Strength(s) | Common Brand Name(s) |
---|
Immediate-Release Capsule | 20 mg, 25 mg, 40 mg, 50 mg | Indocin®, Tivorbex® |
Extended-Release (SR) Capsule | 75 mg | Indocin SR® |
Oral Suspension | 25 mg/5 mL | Indocin® |
Rectal Suppository | 50 mg | Indocin® |
Powder for IV Injection | 1 mg per vial | Indocin IV® |
Sources: [2]
Dosing and Administration Guidelines (Adults)
The guiding principle for Indomethacin therapy is to use the lowest effective dose for the shortest duration consistent with patient treatment goals to minimize the risk of adverse events.[2] To reduce the incidence of gastrointestinal upset, it is generally recommended that oral forms be taken with food.[37]
Table 4: Dosing Guidelines for Adult Indications
Indication | Formulation(s) | Starting Dose | Titration/Maintenance | Maximum Daily Dose | Key Clinical Notes |
---|
Rheumatoid Arthritis, Osteoarthritis, Ankylosing Spondylitis | IR Capsules, Suspension, Suppositories | 25 mg 2-3 times daily | Increase weekly by 25-50 mg as needed. | 200 mg | A larger dose (up to 100 mg) at bedtime can help with night pain/morning stiffness. |
| ER Capsules | 75 mg once daily | May be increased to 75 mg twice daily. | 150 mg | Provides more consistent plasma levels for chronic conditions. |
Acute Gouty Arthritis | IR Capsules, Suspension, Suppositories | 50 mg 3 times daily | Continue until pain is tolerable, then rapidly reduce dose and discontinue. | N/A | ER capsules are not indicated for acute gout. Relief is often rapid (2-4 hours). |
Acute Painful Shoulder (Bursitis/Tendinitis) | All formulations | 75-150 mg daily in 3-4 divided doses | Treat for 7-14 days. Discontinue after inflammation subsides. | 150 mg | Intended for short-term use only. |
Sources: [2]
Special Dosing Considerations: Pediatrics and Neonatology
The use of Indomethacin in children and neonates is reserved for specific indications and requires precise, weight-based dosing and careful monitoring.
Patent Ductus Arteriosus (PDA) Closure (IV Administration)
The intravenous administration of Indomethacin to close a PDA is a critical, high-stakes intervention in premature infants. The standard regimen consists of a course of three IV doses, infused over 20-30 minutes and administered at 12- to 24-hour intervals.33 Dosing is stratified by the infant's postnatal age at the time of the first dose, as renal clearance of the drug changes rapidly in the first week of life. A second course of 1 to 3 doses may be considered if the ductus re-opens 48 hours or more after the completion of the first course.33
Table 5: Intravenous Dosing Guidelines for Patent Ductus Arteriosus in Neonates
Postnatal Age at First Dose | Dose 1 (mg/kg) | Dose 2 (mg/kg) | Dose 3 (mg/kg) | Dosing Interval | Critical Monitoring Parameters |
---|
< 48 hours | 0.2 | 0.1 | 0.1 | 12-24 hours | Withhold dose if urine output <0.6 mL/kg/hr. Monitor renal function, electrolytes, and platelet count. |
2–7 days | 0.2 | 0.2 | 0.2 | 12-24 hours | Withhold dose if urine output <0.6 mL/kg/hr. Monitor renal function, electrolytes, and platelet count. |
> 7 days | 0.2 | 0.25 | 0.25 | 12-24 hours | Withhold dose if urine output <0.6 mL/kg/hr. Monitor renal function, electrolytes, and platelet count. |
Sources: [33]
Juvenile Rheumatoid Arthritis (Oral Administration)
For children aged 2 to 14 years, the recommended starting dose is 1 to 2 mg/kg/day, administered orally in 2 to 4 divided doses. The dosage can be cautiously increased as needed to a maximum of 4 mg/kg/day or 150-200 mg/day, whichever is less.36
Safety Profile, Risks, and Management
This section provides a comprehensive and critical evaluation of Indomethacin's safety profile, detailing the full spectrum of potential harms from common side effects to life-threatening toxicities, and outlining contraindications and precautions for its use. The safety profile is a direct, system-by-system manifestation of its potent, non-selective inhibition of prostaglandins, which are ubiquitous and vital signaling molecules throughout the body. Nearly every major therapeutic benefit and adverse event can be traced back to this single, powerful mechanism. For instance, the inhibition of prostaglandins critical for GI mucosal defense leads to ulcers; inhibition of those maintaining renal blood flow leads to kidney injury; and inhibition of those keeping the ductus arteriosus patent leads to its therapeutic closure in neonates but is a contraindication in late pregnancy.[2]
Black Box Warnings (FDA Mandated)
Indomethacin carries the highest level of warning from the FDA for two major classes of risk.
Cardiovascular Thrombotic Events
- Like other NSAIDs, Indomethacin is associated with an increased risk of serious and potentially fatal cardiovascular thrombotic events, including myocardial infarction (MI) and stroke. This risk can occur early in treatment and may increase with the duration of use. The risk is more pronounced in patients with pre-existing cardiovascular disease or risk factors for it.[2]
- Contraindication: Due to a demonstrated increase in the incidence of MI and stroke, the use of Indomethacin is contraindicated for the treatment of peri-operative pain in the setting of coronary artery bypass graft (CABG) surgery.[2]
- Post-MI Patients: Treatment with NSAIDs in patients who have had a recent MI has been shown to increase the risk of reinfarction, cardiovascular-related death, and all-cause mortality, beginning within the first week of treatment. Therefore, the use of Indomethacin in this population should be avoided unless the benefits are expected to clearly outweigh these substantial risks.[41]
Gastrointestinal Bleeding, Ulceration, and Perforation
- Indomethacin causes an increased risk of serious gastrointestinal (GI) adverse events, including inflammation, bleeding, ulceration, and perforation of the esophagus, stomach, small intestine, or large intestine. These events can be fatal.[2]
- These serious complications can occur at any point during therapy, with or without preceding warning symptoms. It is estimated that only one in five patients who develop a serious upper GI adverse event on NSAID therapy is symptomatic beforehand.[41]
- The risk is significantly elevated in certain populations, including patients with a prior history of peptic ulcer disease (PUD) or GI bleeding (a greater than 10-fold increased risk), those on longer-term therapy, the elderly, individuals in poor general health, and those with concomitant use of oral corticosteroids, anticoagulants (e.g., warfarin), or selective serotonin reuptake inhibitors (SSRIs).[27]
Adverse Effects
The incidence of adverse reactions with Indomethacin is high, which often leads to its consideration as a second-choice agent when safer alternatives are available.[7]
Table 6: Common and Severe Adverse Reactions by System Organ Class
System Organ Class | Common (>1%) and/or Clinically Significant Adverse Reactions | Severe and/or Rare (<1%) Adverse Reactions |
---|
Gastrointestinal | Nausea, vomiting, dyspepsia (heartburn, indigestion), abdominal pain, diarrhea, constipation, anorexia 8 | Peptic ulcer, GI bleeding, perforation, intestinal ulceration with stenosis and obstruction, toxic hepatitis, jaundice (some fatal), ulcerative colitis, pancreatitis 2 |
Central Nervous System | Headache (11.7%), dizziness, vertigo, fatigue, depression, somnolence 8 | Confusion, psychosis, syncope, seizures, coma, peripheral neuropathy, aggravation of epilepsy and Parkinsonism 2 |
Cardiovascular | Hypertension, edema, fluid retention 3 | Myocardial infarction, stroke, congestive heart failure, arrhythmia, palpitations 8 |
Renal | Fluid and sodium retention, increased serum creatinine | Acute renal failure, interstitial nephritis, nephrotic syndrome, hyperkalemia, papillary necrosis 2 |
Hepatic | Elevated liver enzymes (transaminases) 2 | Severe hepatitis (cholestatic, hepatocellular, or mixed), jaundice, liver failure (some fatal) 2 |
Hematologic | Inhibition of platelet aggregation | Leukopenia, agranulocytosis, aplastic anemia, hemolytic anemia, thrombocytopenia, disseminated intravascular coagulation 2 |
Dermatologic/ Hypersensitivity | Rash, pruritus 8 | Stevens-Johnson Syndrome (SJS), Toxic Epidermal Necrolysis (TEN), Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS), exfoliative dermatitis, angioedema, anaphylaxis 8 |
Contraindications and Precautions
Given its risk profile, the use of Indomethacin is contraindicated in several situations and requires significant caution in others.
Absolute Contraindications:
- Patients with a known history of hypersensitivity to Indomethacin, aspirin, or other NSAIDs, particularly those who have experienced asthma, urticaria, or other allergic-type reactions.[2]
- For the treatment of peri-operative pain in the setting of CABG surgery.[2]
- Patients with active peptic ulceration or a history of recent GI bleeding.[2]
- Indomethacin suppositories are contraindicated in patients with a history of proctitis or recent rectal bleeding.[11]
- In neonates, the IV formulation is contraindicated in the presence of proven or suspected necrotizing enterocolitis, active bleeding (especially intracranial or GI), significant renal impairment, thrombocytopenia, coagulation defects, or congenital heart disease in which patency of the ductus arteriosus is necessary for pulmonary or systemic blood flow.[2]
Precautions:
- Therapy should be initiated and maintained at the lowest effective dose for the shortest possible duration.
- Extreme caution is warranted in patients with a history of PUD, cardiovascular disease, renal or hepatic impairment, congestive heart failure, hypertension, or coagulation disorders.[2]
- Patients with pre-existing asthma should be monitored closely, as NSAIDs can precipitate acute bronchospasm.[2]
- Regular monitoring for signs and symptoms of GI bleeding, as well as periodic monitoring of renal function (serum creatinine), liver function tests, and blood pressure is recommended during long-term therapy.[7]
Use in Specific Populations
- Elderly: Indomethacin is identified as a potentially inappropriate medication for use in older adults by the American Geriatrics Society (AGS) Beers Criteria. The risks of GI bleeding, peptic ulcer disease, renal failure, and adverse CNS effects are significantly increased in this population. If its use is unavoidable, it must be at the lowest possible dose for the shortest duration.[2]
- Pregnancy: Use of NSAIDs, including Indomethacin, is contraindicated starting at 30 weeks of gestation. Its use can cause premature closure of the fetal ductus arteriosus, leading to persistent pulmonary hypertension of the newborn. Use during the first and second trimesters should be avoided unless the potential benefit justifies the potential risk to the fetus.[2]
- Lactation: Indomethacin is excreted into breast milk in low concentrations. While not an absolute contraindication, alternative NSAIDs with more established safety profiles during lactation, such as ibuprofen, are generally preferred, especially when nursing a newborn or preterm infant.[2]
- Renal Impairment: Indomethacin should be avoided in patients with advanced renal disease (e.g., GFR < 30 mL/min/1.73 m²). It should be used with caution in patients with mild-to-moderate renal impairment, with close monitoring of renal function.[2]
Drug-Drug and Other Interactions
Indomethacin participates in numerous clinically significant drug interactions, which can increase the risk of toxicity or reduce therapeutic efficacy. Prescribing Indomethacin requires a thorough medication reconciliation and a deep understanding of its "triple threat" interaction profile: it synergistically increases bleeding risk, it antagonizes common treatments for cardiovascular and renal disease, and it alters the pharmacokinetics of several narrow-therapeutic-index drugs.
Pharmacodynamic Interactions (Additive or Antagonistic Effects)
- Anticoagulants and Antiplatelet Agents: The concomitant use of Indomethacin with anticoagulants (e.g., warfarin, apixaban, dabigatran), other antiplatelet drugs (e.g., aspirin, clopidogrel), or even certain herbal supplements with antiplatelet effects (e.g., garlic, ginger, ginkgo biloba) dramatically increases the risk of serious bleeding. This occurs through a synergistic effect, as Indomethacin both inhibits platelet function via COX-1 and directly damages the GI mucosa.[29]
- Other NSAIDs: Co-administration with other systemic NSAIDs (e.g., ibuprofen, naproxen) or with ketorolac (which is contraindicated) provides no additional therapeutic benefit while significantly amplifying the risk of GI toxicity and other adverse effects.[29]
- Corticosteroids: The concurrent use of oral corticosteroids (e.g., prednisone, dexamethasone) with Indomethacin increases the risk of GI ulceration and bleeding.[29]
- SSRIs/SNRIs: Drugs that interfere with serotonin reuptake, such as selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs), also impair platelet aggregation. Their use with Indomethacin potentiates the risk of bleeding more than either agent alone.[29]
- Antihypertensives: Indomethacin can diminish the blood pressure-lowering effects of many classes of antihypertensive drugs, including ACE inhibitors (e.g., lisinopril), angiotensin II receptor blockers (ARBs, e.g., losartan), beta-blockers (e.g., metoprolol), and diuretics (e.g., hydrochlorothiazide, furosemide). This antagonism occurs because Indomethacin inhibits the synthesis of vasodilating renal prostaglandins. This can lead to a loss of blood pressure control and, particularly when combined with ACE inhibitors, ARBs, or diuretics, can precipitate acute kidney injury, especially in elderly, volume-depleted, or renally-impaired patients.[28]
Pharmacokinetic Interactions (Altered Drug Levels)
- Lithium: Indomethacin significantly reduces the renal clearance of lithium, which can lead to elevated, potentially toxic lithium levels. Close monitoring of serum lithium concentrations is mandatory if these drugs must be used together.[28]
- Methotrexate: Indomethacin can decrease the renal clearance of methotrexate, resulting in elevated and prolonged serum concentrations. This can lead to severe, sometimes fatal, hematologic and gastrointestinal toxicity. This combination should be approached with extreme caution, often with dose reduction and intensive monitoring of methotrexate levels, or avoided entirely.[29]
- Digoxin: Indomethacin can increase serum digoxin concentrations, likely by reducing its renal clearance. Digoxin levels should be monitored.[29]
- Probenecid: Probenecid inhibits both the metabolism and renal tubular secretion of Indomethacin. This interaction leads to markedly increased and prolonged plasma concentrations of Indomethacin, which may enhance its therapeutic effect but also significantly increases the risk of toxicity.[31]
- CYP2C9 Substrates/Inhibitors: As a substrate and inhibitor of the CYP2C9 enzyme, Indomethacin can interact with other drugs metabolized by this pathway. It may increase the levels of other CYP2C9 substrates, such as the multiple sclerosis drug siponimod. Conversely, strong inhibitors or inducers of CYP2C9 can alter Indomethacin's own plasma levels, affecting its efficacy and safety.[29]
Table 7: Clinically Significant Drug Interactions
Interacting Drug/Class | Mechanism of Interaction | Clinical Effect/Consequence | Management Recommendation |
---|
Anticoagulants (e.g., Warfarin, Apixaban) | Pharmacodynamic (PD) | Synergistic effect on hemostasis, dramatically increased risk of serious bleeding. | Avoid combination if possible. If necessary, monitor closely for signs of bleeding. |
Antiplatelet Agents (e.g., Aspirin, Clopidogrel) | PD | Additive antiplatelet effects, increased risk of GI and other bleeding. | Avoid routine co-administration, especially with high-dose aspirin. |
Other NSAIDs (e.g., Ibuprofen, Ketorolac) | PD | Increased risk of GI toxicity and other adverse effects with no added benefit. | Avoid concomitant use. Ketorolac is contraindicated. |
Corticosteroids (e.g., Prednisone) | PD | Increased risk of GI ulceration and bleeding. | Use with caution and monitor for GI toxicity. |
SSRIs/SNRIs (e.g., Fluoxetine, Venlafaxine) | PD | Potentiated risk of bleeding due to impaired platelet serotonin uptake. | Use with caution; educate patient on bleeding risk. |
ACE Inhibitors/ARBs (e.g., Lisinopril, Losartan) | PD | Antagonism of antihypertensive effect; increased risk of acute kidney injury. | Monitor blood pressure and renal function. Avoid in high-risk patients. |
Diuretics (e.g., Furosemide, HCTZ) | PD | Reduced natriuretic and antihypertensive effects; increased risk of renal toxicity. | Monitor blood pressure, electrolytes, and renal function. |
Lithium | Pharmacokinetic (PK) | Decreased renal clearance of lithium, leading to increased levels and risk of toxicity. | Monitor lithium levels closely and adjust dose. |
Methotrexate | PK | Decreased renal clearance of methotrexate, leading to potentially fatal toxicity. | Avoid high-dose methotrexate. Use with extreme caution and intensive monitoring at lower doses. |
Digoxin | PK | Increased serum digoxin levels due to reduced renal clearance. | Monitor digoxin levels. |
Probenecid | PK | Increased plasma concentrations and half-life of Indomethacin. | May require Indomethacin dose reduction. |
Sources: [6]
Concluding Analysis and Place in Therapy
Indomethacin occupies a complex and enduring position in the modern pharmacopeia, defined by a fundamental duality: it is at once one of the most potent anti-inflammatory agents available and one of the most fraught with risk. Its clinical profile is a direct reflection of its history as a drug designed for maximum potency in an era before the discovery of targeted enzyme isoforms. The result is a powerful but non-selective molecule that serves as a classic pharmacological lesson—with great potency comes great responsibility.
In contemporary clinical practice, the role of Indomethacin has been significantly refined. For the long-term management of chronic inflammatory conditions like rheumatoid and osteoarthritis, it has largely been relegated to a second-line agent. The development of safer alternatives, including more COX-2 selective NSAIDs, disease-modifying antirheumatic drugs (DMARDs), and biologic agents, means that Indomethacin is typically reserved for patients who have failed or are unable to tolerate other therapies.[7]
However, Indomethacin remains a first-line or indispensable agent for a select group of acute and niche conditions where its potent, rapid action is a distinct advantage and the limited duration of therapy helps to mitigate its risk profile. These key areas include:
- Acute Gouty Arthritis: Its ability to provide rapid and definitive relief from the severe pain and inflammation of a gout flare is highly valued, making it a standard choice for short-term treatment.[7]
- Closure of Patent Ductus Arteriosus: In neonatal medicine, it is the established, life-saving medical therapy for closing a hemodynamically significant PDA in premature infants, an indication for which few alternatives exist.[1]
- Indomethacin-Responsive Headaches: It has a unique, almost pathognomonic, efficacy for certain rare headache syndromes like paroxysmal hemicrania and hemicrania continua, where it often succeeds after all other NSAIDs have failed.[6]
Looking forward, the most promising avenue for refining the use of Indomethacin lies in the application of pharmacogenomics. There is strong evidence linking genetic variants in the CYP2C9 enzyme to both treatment outcomes and toxicity.[9] The ability to pre-emptively identify patients who are poor metabolizers (at higher risk of toxicity) or whose genotype might predict treatment failure (as in PDA) could allow for a more personalized and safer approach to its use. This strategy could help to reclaim a more prominent role for this powerful drug by prospectively identifying the patients most likely to benefit and least likely to be harmed.
In conclusion, Indomethacin is a legacy drug that has withstood the test of time, not because it is the safest option, but because in certain clinical scenarios, it remains one of the most effective. Its continued use demands a profound respect for its pharmacology, meticulous patient selection, a short duration of therapy whenever possible, and vigilant monitoring for its well-documented toxicities.
Works cited
- Indomethacin | C19H16ClNO4 | CID 3715 - PubChem, accessed August 2, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Indomethacin
- Indomethacin - StatPearls - NCBI Bookshelf, accessed August 2, 2025, https://www.ncbi.nlm.nih.gov/books/NBK555936/
- INDOCIN® (INDOMETHACIN) ORAL SUSPENSION Cardiovascular Risk - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/018332s032lbl.pdf?ref=i4ht29
- Indomethacin, Non-selective COX inhibitor (CAS 53-86-1) (ab120719) | Abcam, accessed August 2, 2025, https://www.abcam.com/en-us/products/biochemicals/indomethacin-non-selective-cox-inhibitor-ab120719
- What is the mechanism of Indomethacin? - Patsnap Synapse, accessed August 2, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-indomethacin
- Indomethacin: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 2, 2025, https://go.drugbank.com/drugs/DB00328
- Indometacin - Wikipedia, accessed August 2, 2025, https://en.wikipedia.org/wiki/Indometacin
- INDOCIN® (INDOMETHACIN) ORAL ... - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/018332s032lbl.pdf
- indomethacin - PharmGKB, accessed August 2, 2025, https://www.pharmgkb.org/chemical/PA449982/literature
- Be Informed About Non-Steroidal Anti-Inflammatory Drug Combinations | ARAPC, accessed August 2, 2025, https://arapc.com/nsaid-story/
- INDOCIN® - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2007/016059s097,017814s040,018332s030lbl.pdf
- Indomethacin - brand name list from Drugs.com, accessed August 2, 2025, https://www.drugs.com/ingredient/indomethacin.html
- CAS 53-86-1: Indomethacin | CymitQuimica, accessed August 2, 2025, https://cymitquimica.com/cas/53-86-1/
- indomethacin | Ligand page - IUPHAR/BPS Guide to PHARMACOLOGY, accessed August 2, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=1909
- 3D structure for Indomethacin (DB00328) | DrugBank Online, accessed August 2, 2025, https://go.drugbank.com/structures/small_molecule_drugs/DB00328
- Indomethacin 53-86-1 | TCI AMERICA, accessed August 2, 2025, https://www.tcichemicals.com/CA/en/p/I0655
- Indomethacin | CAS 53-86-1 | SCBT - Santa Cruz Biotechnology, accessed August 2, 2025, https://www.scbt.com/p/indomethacin-53-86-1
- Structural and Dynamical Properties of Indomethacin Molecules Embedded within the Mesopores of SBA-15: A Solid-State NMR View - ACS Publications, accessed August 2, 2025, https://pubs.acs.org/doi/10.1021/jp2087016
- Indometacin | 53-86-1 - ChemicalBook, accessed August 2, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB1750267.htm
- 204768Orig1s000 - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/204768Orig1s000ClinPharmR.pdf
- Organic synthesis of indomethacin - The Science Snail, accessed August 2, 2025, https://www.sciencesnail.com/science/organic-synthesis-of-indomethacin
- Synthesis of Indomethacin and Iprindole - Thieme Connect, accessed August 2, 2025, https://www.thieme-connect.com/products/ejournals/pdf/10.1055/s-0030-1259403.pdf
- Indomethacin | Drug Information, Uses, Side Effects, Chemistry - PharmaCompass.com, accessed August 2, 2025, https://www.pharmacompass.com/chemistry-chemical-name/indomethacin
- Indomethacin Capsules, USP 25 mg - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/018829s022lbl.pdf
- Indomethacin (CAS Number: 53-86-1) | Cayman Chemical, accessed August 2, 2025, https://www.caymanchem.com/product/70270/indomethacin
- Indomethacin Gene Set - Ma'ayan Laboratory, Computational Systems Biology, accessed August 2, 2025, https://maayanlab.cloud/Harmonizome/gene_set/Indomethacin/DrugBank+Drug+Targets
- Indomethacin (Indocin, Indocin SR): Uses, Side Effects, Interactions, Pictures, Warnings & Dosing - WebMD, accessed August 2, 2025, https://www.webmd.com/drugs/2/drug-8880-5186/indomethacin-oral/indomethacin-oral/details
- Indomethacin - Mechanism, Indication, Contraindications, Dosing, Adverse Effect, Interaction, Renal Dose, Hepatic Dose | Drug Index | Pediatric Oncall, accessed August 2, 2025, https://www.pediatriconcall.com/drugs/indomethacin/667
- Indocin (indomethacin) dosing, indications, interactions, adverse effects, and more - Medscape Reference, accessed August 2, 2025, https://reference.medscape.com/drug/indocin-indomethacin-343290
- Pharmacology, Pharmacokinetics and Future Prospective of Indomethacin, accessed August 2, 2025, https://sifisheriessciences.com/index.php/journal/article/download/3246/2367/4378
- Clinical Pharmacokinetics of indomethacin - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/7249487/
- indomethacin capsules - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/018851s028lbl.pdf
- Indomethacin for Injection - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/022536lbl.pdf
- Premature Births Unknown Status Phase 2 Trials for Indomethacin (DB00328) - DrugBank, accessed August 2, 2025, https://go.drugbank.com/indications/DBCOND0043809/clinical_trials/DB00328?phase=2&status=unknown_status
- Indomethacin Uses, Side Effects & Warnings - Drugs.com, accessed August 2, 2025, https://www.drugs.com/mtm/indomethacin.html
- Indomethacin dosage, forms, and strengths - SingleCare, accessed August 2, 2025, https://www.singlecare.com/prescription/indomethacin/dosage
- How to Determine Dosage for Indomethacin - GoodRx, accessed August 2, 2025, https://www.goodrx.com/indomethacin/dosage
- Indomethacin (oral route) - Side effects & dosage - Mayo Clinic, accessed August 2, 2025, https://www.mayoclinic.org/drugs-supplements/indomethacin-oral-route/description/drg-20069700
- Indomethacin for Injection, USP Dosage and Administration | Pfizer Medical - US, accessed August 2, 2025, https://www.pfizermedical.com/indomethacin/dosage-admin
- Table: Indomethacin Dosing Guidelines*-Merck Manual Professional Edition, accessed August 2, 2025, https://www.merckmanuals.com/professional/multimedia/table/indomethacin-dosing-guidelines
- Indocin (Indomethacin): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed August 2, 2025, https://www.rxlist.com/indocin-drug.htm
- Indomethacin (intravenous route) - Side effects & uses - Mayo Clinic, accessed August 2, 2025, https://www.mayoclinic.org/drugs-supplements/indomethacin-intravenous-route/description/drg-20069693
- Indomethacin | Davis's Drug Guide for Rehabilitation Professionals, accessed August 2, 2025, https://fadavispt.mhmedical.com/content.aspx?bookid=1873§ionid=139014420