C6H8ClN7O
2609-46-3
Ascites, Calcium Nephrolithiasis, Congestive Heart Failure (CHF), Hypertension, Hypokalemia, Metabolic Alkalosis, Polyuria
Amiloride is a small molecule drug classified as a potassium-sparing diuretic, belonging to the pyrazine-carbonyl-guanidine chemical class.[1] It is identified by DrugBank Accession Number DB00594 and CAS Number 2609-46-3.[1] Developed in 1967 and first approved by the U.S. Food and Drug Administration (FDA) in 1981, amiloride is recognized on the World Health Organization's List of Essential Medicines, underscoring its global importance.[5]
The primary therapeutic indications for amiloride are the management of hypertension and edematous states, such as those associated with congestive heart failure and hepatic cirrhosis with ascites.[9] However, its clinical utility in these prevalent conditions is nuanced. Amiloride possesses only weak intrinsic natriuretic and antihypertensive properties and is therefore rarely employed as a monotherapy.[4] Its principal role in mainstream cardiovascular medicine is as an adjunctive agent, co-administered or co-formulated with more potent kaliuretic diuretics, such as thiazides (e.g., hydrochlorothiazide) or loop diuretics (e.g., furosemide). In this capacity, its primary function is to counteract the significant adverse effect of these diuretics: the excessive urinary loss of potassium (hypokalemia).[5]
The pharmacological basis for this potassium-sparing effect is the direct, reversible blockade of the epithelial sodium channel (ENaC) located on the apical membrane of principal cells in the late distal convoluted tubule and collecting duct of the nephron.[1] This mechanism is distinct from that of other potassium-sparing diuretics like spironolactone, as it is independent of aldosterone levels.[3] By inhibiting sodium reabsorption at this site, amiloride reduces the electrical driving force for potassium secretion into the tubular lumen, thereby conserving body potassium stores.
This highly specific mechanism of action also underpins amiloride's critical role in several off-label applications where ENaC dysregulation is the central pathophysiological defect. It is the treatment of choice for Liddle Syndrome, a rare genetic form of hypertension caused by a gain-of-function mutation in ENaC. It is also a preferred therapy for managing lithium-induced nephrogenic diabetes insipidus, where it prevents the toxic entry of lithium into renal cells via the ENaC.[2] The most significant risk associated with amiloride therapy is hyperkalemia (elevated serum potassium), which is a direct extension of its therapeutic action and can be life-threatening, particularly in patients with renal impairment or diabetes, or those taking other potassium-elevating medications.[12]
A comprehensive understanding of amiloride's place in modern therapeutics requires an appreciation of its fundamentally dualistic clinical identity. In the context of widespread cardiovascular diseases like hypertension and heart failure, it functions as a secondary, supportive agent. Its primary purpose is not to be the main driver of diuresis or blood pressure reduction, but rather to serve as a "potassium-sparing" safeguard that mitigates the well-known metabolic side effects of more powerful primary diuretic therapies.[1] Its own diuretic effect is described as modest, and clinical guidelines have historically positioned it as a secondary-line agent.[7]
In stark contrast, when applied to specific genetic and iatrogenic channelopathies, amiloride transforms from a supportive agent into a highly targeted, first-line precision medicine. For conditions like Liddle Syndrome, where the underlying pathology is an overactive ENaC, amiloride is not merely helpful; it is the definitive treatment that directly corrects the molecular defect.[8] Similarly, in lithium-induced nephrogenic diabetes insipidus, its ability to block the ENaC serves as a targeted intervention to prevent the entry of the toxic agent into the renal tubular cells, thereby addressing the root cause of the condition.[18] Therefore, the very property that makes it a "weak" diuretic in a general sense—its limited site of action in the nephron—is precisely what makes it a potent and specific therapeutic in diseases where that site is the nexus of the pathology. This duality, from a general adjunctive drug to a specific disease-modifying agent, is the central theme that defines its unique and varied role in medicine.
Amiloride is a small molecule drug with a well-defined chemical structure and numerous identifiers used across scientific and regulatory databases. Its systematic IUPAC (International Union of Pure and Applied Chemistry) name is 3,5-diamino-6-chloro-N-(diaminomethylidene)pyrazine-2-carboxamide.[1] It is also referred to by the synonym N-Amidino-3,5-diamino-6-chloropyrazinecarboxamide.[24]
In clinical and research settings, it is known by various synonyms, including Amilorida, Amiloridum, Amipramidin, Amipramizid, and Guanamprazin.[24] A key research code used during its development was MK-870.[1] As a single-agent therapy, it is most widely known by the brand name Midamor.[9] However, it is more frequently encountered in combination products, particularly with hydrochlorothiazide, under brand names such as Moduretic and Moduretic 5-50.[9]
The fundamental chemical and physical properties of amiloride are critical to understanding its absorption, distribution, mechanism of action, and formulation. These properties are consolidated in Table 1.
Table 1: Chemical and Physical Properties of Amiloride
Property | Value | Source(s) |
---|---|---|
Drug Type | Small Molecule | User Query |
Chemical Formula | C6H8ClN7O | 9 |
Molecular Weight | 229.63 g/mol | 24 |
CAS Number (Free Base) | 2609-46-3 | 1 |
Related CAS Numbers | 17440-83-4 (hydrochloride); 2016-88-8 (anhydrous hydrochloride) | 1 |
DrugBank ID | DB00594 | 1 |
ATC Code | C03DB01 | 6 |
IUPAC Name | 3,5-diamino-6-chloro-N-(diaminomethylidene)pyrazine-2-carboxamide | 1 |
InChIKey | XSDQTOBWRPYKKA-UHFFFAOYSA-N | 1 |
SMILES | C1(=C(N=C(C(=N1)Cl)N)N)C(=O)N=C(N)N | 1 |
Physical Description | Solid; Off-white to light yellow powder | 1 |
Melting Point | 240.5–241.5 °C | 1 |
Water Solubility | 659 mg/L at 25 °C; described as "slightly soluble" | 1 |
LogP (Octanol-Water Partition Coefficient) | -0.3 to -1.08 | 1 |
pKa | 8.7 | 1 |
The physicochemical characteristics of amiloride are not merely descriptive data points; they are intrinsically linked to its pharmacokinetic profile and mechanism of action. The drug's low lipophilicity, as evidenced by a consistently negative LogP value, indicates that it is a hydrophilic molecule.[1] This preference for aqueous environments over lipid membranes is a key factor contributing to its incomplete and variable oral absorption.[7]
Furthermore, amiloride is a moderately strong base with a pKa of 8.7.[1] According to the Henderson-Hasselbalch equation, at the physiological pH of blood and extracellular fluid (~7.4), a base with this pKa will exist predominantly in its protonated, positively charged (ionized) form. This ionization of the guanidinium group is fundamental to its pharmacological activity.[1] The resulting positive charge facilitates a critical electrostatic interaction with negatively charged amino acid residues within the pore of the ENaC. This interaction allows amiloride to act as a physical "plug," effectively blocking the channel and preventing sodium transport. Thus, the chemical properties of the molecule—its basicity and the resulting charge at physiological pH—are directly responsible for its therapeutic effect. The combination of being hydrophilic and ionized also explains its limited ability to cross biological membranes, leading to its incomplete absorption and its distribution characteristics.
The therapeutic and adverse effects of amiloride are almost exclusively attributable to its interaction with a single primary target: the epithelial sodium channel (ENaC).
Amiloride functions as a direct, competitive, and reversible blocker of the ENaC.[1] It exerts its effect from the luminal (urine) side of the kidney tubules, specifically targeting the principal cells of the late distal convoluted tubule (DCT) and the cortical collecting duct.[1] By physically obstructing the channel's outer pore, amiloride inhibits the reabsorption of sodium ions (
Na+) from the tubular fluid back into the circulation.[1] This blockade is highly potent, with a half-maximal inhibitory concentration (
IC50) in the sub-micromolar range of 0.1 to 0.5 µmol/L, a concentration readily achieved with standard clinical dosing.[8]
The reabsorption of positively charged sodium ions through ENaC is the primary mechanism that generates a negative electrical potential in the tubular lumen relative to the inside of the principal cell. This lumen-negative transepithelial potential difference is the principal driving force for the secretion of other positive ions, namely potassium (K+) and hydrogen (H+), from the cell into the urine for excretion.[1]
By blocking Na+ influx, amiloride effectively dissipates this electrical gradient. The reduction in the lumen-negative potential directly diminishes the electrochemical driving force for K+ and H+ secretion. The ultimate result is a "potassium-sparing" or "antikaliuretic" effect, where the excretion of potassium is significantly reduced.[1] This mechanism also leads to a reduction in the excretion of other cations, including calcium (
Ca2+) and magnesium (Mg2+).[2] The overall diuretic effect—the increased excretion of sodium and water—is considered mild because the distal nephron is responsible for reabsorbing only a small fraction (~5%) of the total filtered sodium load.[31]
At therapeutic concentrations, amiloride is highly selective for ENaC. However, at much higher, supra-pharmacological concentrations, it can exhibit off-target effects by inhibiting other ion transporters. These include the Na+/H+ exchanger (NHE) and the Na+/Ca2+ exchanger (NCX). Amiloride's potency against these transporters is significantly lower than for ENaC. The IC50 for NHE inhibition ranges from approximately 3 µmol/L to as high as 1 mmol/L, while the IC50 for NCX inhibition is around 1 mmol/L.[15] This wide separation in potency means that at standard clinical doses that produce plasma concentrations of ≤1 µmol/L, amiloride functions as a relatively pure ENaC inhibitor. This distinction is important for interpreting research suggesting that low-dose amiloride may have cardiovascular effects independent of its renal actions, potentially through blockade of ENaC expressed in non-renal tissues like blood vessels or the central nervous system.[15]
The clinical use of amiloride is guided by its pharmacokinetic profile, which describes its movement into, through, and out of the body. Key parameters are summarized in Table 2.
Table 2: Pharmacokinetic Parameters of Amiloride
Parameter | Value / Description | Source(s) |
---|---|---|
Absorption | Incomplete oral bioavailability of ~50%. Co-administration with food reduces the extent of absorption by ~30% but does not affect the rate. | 7 |
Onset of Action | 2 hours | 2 |
Time to Peak Plasma Concentration (Tmax) | 3 to 4 hours | 2 |
Time to Peak Diuretic Effect | 6 to 10 hours | 3 |
Duration of Action | Approximately 24 hours | 2 |
Distribution | High volume of distribution (Vd) of 350 to 380 L, indicating significant distribution into extravascular tissues. | 2 |
Protein Binding | Low plasma protein binding, <40%. | 2 |
Metabolism | Not metabolized by the liver. | 2 |
Elimination Half-life (t1/2) | 6 to 9 hours in individuals with normal renal function. | 2 |
Excretion | Excreted unchanged from the body. Approximately 50% is recovered in the urine and 40% in the feces within 72 hours. | 2 |
The pharmacokinetic and pharmacodynamic properties of amiloride have significant clinical implications that extend beyond basic dosing. The fact that amiloride is not metabolized by the liver is a notable feature, suggesting that drug accumulation would not be expected in patients with hepatic dysfunction alone.[2] This would seem to make it an ideal diuretic for patients with liver cirrhosis and ascites, a key indication. However, this creates a clinical paradox. Patients with severe liver disease are frequently at high risk for developing renal complications, most notably hepatorenal syndrome. Since amiloride is cleared entirely by the kidneys and its primary toxicity, hyperkalemia, is profoundly exacerbated by any degree of renal impairment, its use in this population is extremely hazardous.[13] Therefore, while the drug itself is not hepatotoxic, the common comorbidities of its target patient population (i.e., cirrhotic patients) often create the very contraindication (renal dysfunction) that makes its use unsafe. This necessitates extreme caution and vigilant monitoring of renal function and electrolytes, a critical nuance that belies the simplistic assumption that it is "safe in liver disease."
Amiloride is approved by regulatory agencies for the treatment of hypertension and edematous conditions, with its primary value derived from its potassium-sparing effect when used in combination with other diuretics.
Amiloride is commercially available as a 5 mg oral tablet.[19] The typical initial dosage for adults is 5 mg administered once daily. This dose can be titrated upwards to a maximum of 10 mg per day if the desired potassium-sparing or diuretic effect is not achieved.[3] While higher doses up to 20 mg daily have been investigated in clinical trials, they are associated with a greater risk of adverse effects, particularly hyperkalemia, and are not commonly used in routine practice.[11] To minimize the potential for gastrointestinal side effects such as nausea, it is recommended that amiloride be taken with food.[7]
Amiloride was developed by researchers at Merck in 1967.[5] It received its initial approval from the U.S. FDA on October 5, 1981, under the brand name Midamor for use as a single agent.[6] Subsequent approvals for generic versions and combination products have occurred over the following decades; for example, a supplemental new drug application for Midamor was approved on March 21, 2002.[38] Its established efficacy and safety profile have earned it a place on the World Health Organization's List of Essential Medicines.[7] In the United States, United Kingdom, and Australia, amiloride is available by prescription only (Rx-only).[7]
The clinical positioning of amiloride has evolved since its introduction. Initially conceived and studied as a standalone diuretic, its relatively weak efficacy in this role led to its relegation primarily to an adjunctive agent for potassium conservation.[12] The 2017 American College of Cardiology/American Heart Association (ACC/AHA) hypertension guidelines, for instance, classify it as a "secondary" oral antihypertensive with minimal efficacy when used alone.[7]
However, this traditional view is being challenged by emerging evidence. Its established, first-line efficacy in treating ENaC-driven hypertension in Liddle Syndrome provides a clear precedent for its use as a targeted primary therapy. More recently, a 2024 clinical trial (NCT04331691) demonstrated that amiloride was non-inferior to the mineralocorticoid receptor antagonist spironolactone for treating resistant hypertension—a condition where spironolactone is a guideline-recommended add-on therapy.[11] This finding is significant because it suggests that for a subset of patients with difficult-to-treat hypertension, possibly those with underlying high ENaC activity, amiloride could be a highly effective agent in its own right, not merely a supportive one. This new evidence may prompt a re-evaluation of its potential and a shift away from its purely adjunctive classification toward a more mechanistically informed, targeted application in specific hypertensive populations.
The safety profile of amiloride is overwhelmingly dominated by the predictable consequences of its primary pharmacological action on renal electrolyte handling.
The most important and potentially fatal adverse effect of amiloride is hyperkalemia.[19] This risk is a direct extension of its therapeutic mechanism.
The use of amiloride is strictly prohibited in certain patient populations where the risk of severe hyperkalemia is unacceptably high.
Given the risks, vigilant monitoring and cautious use in specific populations are paramount.
The safety profile of amiloride is a clear illustration of on-target toxicity. Unlike drugs with unpredictable, idiosyncratic adverse effects, the primary danger of amiloride—hyperkalemia—is a direct and logical extension of its therapeutic mechanism. This predictability makes risk management a straightforward exercise centered on two core principles: meticulous patient selection to avoid individuals with impaired potassium excretion capabilities, and diligent laboratory monitoring to detect electrolyte shifts before they become clinically significant. A clinician who understands the drug's mechanism can anticipate nearly all of its major risks and knows precisely which parameters (serum potassium and renal function) to monitor.
The potential for drug-drug and drug-disease interactions with amiloride is significant and centers on its effects on potassium homeostasis and renal function.
Amiloride is known to interact with numerous medications, with over 300 potential interactions identified.[46] The most clinically relevant of these involve drugs that also affect serum potassium levels or renal function. These are summarized in Table 3.
Table 3: Clinically Significant Drug Interactions with Amiloride
Interacting Drug or Class | Nature of Interaction | Clinical Recommendation and Management | Source(s) |
---|---|---|---|
Potassium-Sparing Diuretics (e.g., Spironolactone, Triamterene, Eplerenone) | Additive pharmacodynamic effect, leading to a severe and unacceptable risk of life-threatening hyperkalemia. | Contraindicated. Combination use should be avoided. | 12 |
Potassium Supplements & Salt Substitutes (containing KCl) | Direct addition of potassium load to a system where renal potassium excretion is already inhibited, leading to a high risk of severe hyperkalemia. | Contraindicated. Avoid concomitant use unless treating severe, refractory hypokalemia, which requires intensive monitoring. | 12 |
ACE Inhibitors (e.g., Lisinopril, Ramipril) & Angiotensin II Receptor Blockers (ARBs) (e.g., Losartan, Valsartan) | These agents decrease aldosterone production, which reduces potassium excretion. This effect is additive with amiloride's direct ENaC blockade, increasing the risk of hyperkalemia. | Use with caution. Monitor serum potassium levels frequently, especially upon initiation or dose titration of either agent. | 5 |
Calcineurin Inhibitors (Cyclosporine, Tacrolimus) | These immunosuppressants can impair renal function and independently increase serum potassium. The combination with amiloride significantly elevates the risk of hyperkalemia. | Use with caution. Requires frequent monitoring of serum potassium and renal function. | 12 |
Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) (e.g., Ibuprofen, Indomethacin, Naproxen) | NSAIDs can reduce the diuretic and antihypertensive effects of amiloride by inhibiting renal prostaglandin synthesis. They can also impair renal function and may independently increase serum potassium. | Monitor blood pressure, signs of fluid retention, renal function, and serum potassium. The desired diuretic effect may be diminished. | 2 |
Lithium | Amiloride can reduce the renal clearance of lithium, leading to increased serum lithium concentrations and a heightened risk of lithium toxicity. | Concomitant use is generally contraindicated for routine indications. If necessary (e.g., for lithium-induced NDI), it requires specialist management with intensive monitoring of serum lithium levels and dose adjustments. | 12 |
Digoxin | The interaction is not well-defined, but there is a potential for amiloride to alter the clinical response to digoxin. Electrolyte imbalances (hyperkalemia) can also affect cardiac sensitivity to digoxin. | Observe the patient carefully for signs of altered digoxin efficacy or toxicity. Maintain normal electrolyte levels. | 12 |
The conditions that predispose patients to adverse outcomes with amiloride are largely synonymous with its contraindications and represent critical drug-disease interactions.
A significant clinical paradox emerges from the interaction profile, particularly concerning the use of amiloride and lithium. For general therapeutic purposes, the combination is listed as contraindicated due to the risk of amiloride reducing lithium's renal clearance and precipitating toxicity.[12] However, in the specific context of treating lithium-induced nephrogenic diabetes insipidus (NDI), amiloride is considered a first-line therapy.[47] This highlights that the risk-benefit calculation is highly dependent on the clinical scenario. In NDI, the primary therapeutic goal is to block the entry of lithium into the renal principal cells via ENaC, thereby preventing the downstream pathology that causes debilitating polyuria.[18] The benefit of this targeted mechanism outweighs the manageable risk of increased systemic lithium levels. This situation requires expert clinical judgment, where the known drug interaction is not a barrier to use but rather a factor to be actively managed through dose adjustments and vigilant monitoring of serum lithium levels.
The highly specific mechanism of amiloride has led to its successful application in several off-label and investigational contexts where ENaC function is central to the disease pathophysiology.
The history of amiloride's off-label use provides a compelling lesson in the evolution from broad-spectrum therapy toward precision medicine. Its successes and failures are not arbitrary but are dictated by the centrality of the ENaC channel to the disease in question. In Liddle Syndrome, the overactive ENaC is the entire disease; blocking it is curative. In lithium-induced NDI, ENaC is the gateway for the toxin; blocking the gate prevents the disease. In a subset of patients with resistant hypertension, ENaC overactivity may be a primary driver, making amiloride a targeted therapy. Conversely, in cystic fibrosis, while ENaC hyperabsorption is an important part of the pathophysiology, it is a downstream consequence of the primary CFTR defect in a complex multifactorial disease. In this context, a short-acting drug targeting a secondary mechanism proved insufficient. This pattern demonstrates that amiloride's utility is directly proportional to the degree to which ENaC is the primary, rate-limiting driver of the pathology, providing a clear framework for predicting its potential success in future applications.
Amiloride occupies a unique and multifaceted position in the therapeutic armamentarium. Its clinical profile is defined by a distinct duality. For the most common cardiovascular indications—hypertension and heart failure—it serves as a valuable adjunctive agent, a workhorse whose primary role is to safeguard against the potassium-wasting effects of more potent diuretics. In this capacity, its own weak diuretic and antihypertensive effects are secondary to its crucial role in maintaining electrolyte homeostasis.
Simultaneously, for rare but serious channelopathies like Liddle Syndrome and lithium-induced nephrogenic diabetes insipidus, amiloride is transformed into a first-line, highly specific, and mechanistically targeted therapy. In these conditions, it directly corrects the underlying molecular pathology centered on the epithelial sodium channel (ENaC). This therapeutic value is inextricably linked to its on-target effect on ENaC, which is concurrently the source of its efficacy, the cause of its principal adverse effect (hyperkalemia), and the scientific rationale for its most successful off-label applications. The appropriate and safe use of amiloride, therefore, requires a sophisticated understanding of this dual identity and a deep appreciation for the central role of potassium and renal function in its clinical effects.
Despite being a drug with over five decades of clinical use, several promising avenues for future research remain, aimed at refining its role and expanding its applications.
In conclusion, amiloride is far more than a simple diuretic. It is a model drug for understanding how a highly specific mechanism of action can be leveraged across a spectrum of diseases, from common to rare. Future research focused on personalized medicine and exploring its non-renal effects will ensure that the full therapeutic potential of this venerable drug continues to be realized.
Published at: August 13, 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.