C7H17N2O2
674-38-4
Acute post-operative Urinary retention, Neurogenic atony of the urinary bladder, Non-obstructive postpartum urinary retention
Bethanechol is a synthetic choline ester classified as a direct-acting parasympathomimetic agent. It functions as a selective muscarinic receptor agonist, mimicking the effects of the endogenous neurotransmitter acetylcholine on target organs. Synthesized in 1935, bethanechol's therapeutic utility is predicated on two key molecular features: its high affinity for muscarinic receptors with negligible activity at nicotinic receptors, and its structural resistance to hydrolysis by cholinesterase enzymes.[1] This resistance confers a significantly longer duration of action compared to acetylcholine, making it a viable therapeutic agent.
The primary mechanism of action involves the stimulation of postganglionic muscarinic receptors, particularly the M3 subtype located on the smooth muscle of the urinary bladder and gastrointestinal tract.[3] This action increases the tone of the detrusor muscle, promoting bladder contraction and micturition, and enhances gastrointestinal motility. Consequently, bethanechol holds FDA approval for the treatment of acute postoperative and postpartum non-obstructive urinary retention, as well as for neurogenic atony of the bladder with retention.[5] Its pro-motility effects also support its off-label use in conditions such as gastroesophageal reflux disease (GERD).[8]
The pharmacokinetic profile of bethanechol is characterized by a rapid onset of action following oral or subcutaneous administration. A critical feature of its structure, a permanently charged quaternary ammonium group, prevents it from crossing the blood-brain barrier, thereby confining its activity to the peripheral nervous system and eliminating central nervous system side effects.[1] Notably, there is a significant gap in the scientific literature regarding its metabolism and excretion, with authoritative sources indicating these pathways are not well understood.[3]
The safety profile of bethanechol is a direct extension of its cholinergic pharmacology. Adverse effects, such as salivation, abdominal cramping, hypotension, and bronchoconstriction, are predictable consequences of generalized muscarinic stimulation.[10] Its use is contraindicated in patients with conditions that would be dangerously exacerbated by parasympathetic activation, including mechanical bladder or GI obstruction, asthma, peptic ulcer disease, and significant cardiovascular disease.[2] While bethanechol has been a cornerstone in managing specific urological disorders for decades, emerging research into its potential applications in neonatal respiratory conditions and as an adjuvant in oncology suggests its therapeutic relevance continues to evolve.[12]
The precise identification and characterization of a pharmaceutical agent's chemical and physical properties are fundamental to understanding its behavior, formulation, and biological activity. Bethanechol is a well-defined small molecule with a consistent profile across numerous chemical and drug databases.
Bethanechol is the established generic name for the active pharmaceutical ingredient.[4] It is most commonly formulated and administered as a salt, Bethanechol Chloride.[1] The drug has been marketed under various brand names, including Urecholine, Duvoid, Myotonachol, and Urocarb, although some brand name formulations are no longer commercially available in certain regions.[2]
Its chemical structure is systematically described by the International Union of Pure and Applied Chemistry (IUPAC) as 2-carbamoyloxypropyl(trimethyl)azanium or 2-(Carbamoyloxy)-N,N,N-trimethylpropan-1-aminium.[1] The name bethanechol itself is a reference to its structure as the urethane of beta-methylcholine.[2] A variety of synonyms are used in scientific literature and databases, including Carbamyl-β-methylcholine, (2-hydroxypropyl)trimethylammonium carbamate, and liberan.[2]
The compound is unambiguously identified by a set of standardized codes. The CAS (Chemical Abstracts Service) Registry Number for the active bethanechol moiety is 674-38-4, while the number for the more commonly used chloride salt is 590-63-6.[1] This distinction is crucial for precision in chemical synthesis and pharmaceutical formulation. Its unique DrugBank Accession Number is DB01019.[1]
The molecular formula for the active cationic moiety is $C_{7}H_{17}N_{2}O_{2}^{+}$.[1] This corresponds to a monoisotopic mass of 161.129 g/mol and an average molecular weight of approximately 161.22 g/mol.[2] The formula for bethanechol chloride is $C_{7}H_{17}ClN_{2}O_{2}$, with a molecular weight of 196.68 g/mol.[15]
The molecular structure of bethanechol is represented by the Simplified Molecular Input Line Entry System (SMILES) string CC(C[N+](C)(C)C)OC(=O)N.[1] Its corresponding International Chemical Identifier Key (InChIKey) is NZUPCNDJBJXXRF-UHFFFAOYSA-O, a hashed representation that serves as a digital fingerprint for the molecule.[1]
The structure contains a permanently charged quaternary ammonium ion, [N+](C)(C)C. This structural feature is the primary determinant of the drug's overarching pharmacological profile. The positive charge at physiological pH renders the molecule highly polar, which critically governs its distribution within the body. Specifically, this charge prevents the molecule from passively diffusing across lipid-rich biological membranes, most notably the blood-brain barrier.[1] This confinement to the periphery is directly responsible for its lack of central nervous system (CNS) effects, representing a key structure-activity relationship that defines its clinical utility and safety profile.
Bethanechol chloride is a white, hygroscopic crystalline solid that emits a slight amine-like odor.[16] It is freely soluble in water, with one gram dissolving in approximately 0.6 mL of water, and is also soluble in alcohol.[16] The melting point of the chloride salt is in the range of 217–221 °C.[1] Aqueous solutions are relatively stable and can be sterilized by autoclaving.[16]
The molecule's properties align well with established criteria for orally bioavailable drugs, known as Lipinski's Rule-of-Five. It has 0 violations of these rules, with a molecular weight under 500, a calculated logarithm of the octanol-water partition coefficient (XLogP) of -0.52, 3 hydrogen bond acceptors, and 1 hydrogen bond donor, suggesting favorable characteristics for absorption after oral administration.[19]
The comprehensive chemical and physical data for bethanechol are summarized in Table 2.1. The high degree of concordance for key identifiers such as the CAS number and InChIKey across multiple independent and authoritative databases (e.g., DrugBank, PubChem, FDA) provides a strong validation of the compound's structural identity and serves as a reliable foundation for all subsequent pharmacological and clinical analysis.[1]
Table 2.1: Comprehensive Identifiers and Physicochemical Properties of Bethanechol
| Property | Value | Source(s) |
|---|---|---|
| Nomenclature | ||
| Generic Name | Bethanechol | 4 |
| Common Salt | Bethanechol Chloride | 1 |
| IUPAC Name | 2-carbamoyloxypropyl(trimethyl)azanium | 1 |
| Brand Names | Urecholine, Duvoid, Myotonachol | 2 |
| Identifiers | ||
| DrugBank ID | DB01019 | 2 |
| CAS Number (Moiety) | 674-38-4 | 1 |
| CAS Number (Chloride Salt) | 590-63-6 | 1 |
| UNII | 004F72P8F4 | 1 |
| Chemical Formula & Weight | ||
| Molecular Formula (Moiety) | $C_{7}H_{17}N_{2}O_{2}^{+}$ | 1 |
| Molecular Weight (Moiety) | 161.22 g/mol | 19 |
| Molecular Formula (Chloride) | $C_{7}H_{17}ClN_{2}O_{2}$ | 22 |
| Molecular Weight (Chloride) | 196.68 g/mol | 15 |
| Structural Information | ||
| SMILES | CC(C[N+](C)(C)C)OC(=O)N | 1 |
| InChIKey | NZUPCNDJBJXXRF-UHFFFAOYSA-O | 1 |
| Physicochemical Properties | ||
| Physical Description | White, hygroscopic crystalline solid | 1 |
| Melting Point (Chloride) | 217–221 °C | 1 |
| Water Solubility | Freely soluble | 16 |
| Topological Polar Surface Area | 52.32 Ų | 19 |
| Lipinski's Rule Violations | 0 | 19 |
The therapeutic effects and adverse reaction profile of bethanechol are direct consequences of its interaction with the autonomic nervous system. A thorough understanding of its pharmacology is essential for its safe and effective clinical use.
Bethanechol is classified as a direct-acting parasympathomimetic (or cholinergic) agent.[6] It is a synthetic choline carbamate, structurally related to the endogenous neurotransmitter acetylcholine.[2] Its primary pharmacological identity is that of a muscarinic receptor agonist, meaning it binds to and activates muscarinic acetylcholine receptors, thereby mimicking the "rest and digest" functions of the parasympathetic nervous system.[1]
Bethanechol exerts its effects by directly stimulating postganglionic muscarinic receptors located on the surface of effector cells.[3] This action initiates a cascade of intracellular signaling events that produce a physiological response characteristic of parasympathetic nerve stimulation.
There are five known subtypes of muscarinic receptors ($M_{1}$ through $M_{5}$), and bethanechol functions as a non-selective agonist at all of them.[3] However, the clinical effects of the drug are primarily dictated by its actions on specific subtypes in key tissues:
A crucial aspect of bethanechol's molecular profile is its high selectivity for muscarinic receptors over nicotinic receptors.[1] This selectivity is clinically important because it avoids the effects associated with nicotinic receptor activation, such as skeletal muscle tremors, fasciculations, and stimulation of autonomic ganglia.
The therapeutic viability of bethanechol stems from a key innovation in its chemical design compared to the natural ligand, acetylcholine. Acetylcholine is an ineffective systemic drug because it is almost instantaneously hydrolyzed and inactivated by the enzyme acetylcholinesterase.[25] Bethanechol, by contrast, possesses a carbamate ester group instead of an acetate ester, which makes it resistant to degradation by cholinesterase.[1] This chemical modification was a deliberate design choice to overcome the profound pharmacokinetic limitations of the endogenous neurotransmitter. The result is a molecule with a significantly more prolonged duration of action, allowing it to exert sustained therapeutic effects following administration.[2]
As a global muscarinic agonist, bethanechol produces a wide range of physiological effects throughout the body. The clinical utility of the drug is fundamentally a matter of balancing its desired therapeutic effects, which are primarily mediated by $M_{3}$ receptors, against its dose-limiting side effects, which arise from the stimulation of muscarinic receptors ($M_{1}$, $M_{2}$, and $M_{3}$) in other organ systems. This balance defines its therapeutic window and necessitates careful, individualized dosing.
The pharmacokinetic profile of bethanechol describes its journey through the body and is critical for determining appropriate dosing regimens and administration routes. While some aspects of its ADME are well-documented, significant knowledge gaps persist, particularly concerning its metabolic fate.
Bethanechol can be administered orally or via subcutaneous injection, with each route having a distinct pharmacokinetic profile that dictates its clinical use.
The stark differences in the pharmacokinetic profiles between the two routes are summarized in Table 4.1, providing a clear basis for clinical decision-making.
Table 4.1: Comparative Pharmacokinetic Profile of Bethanechol by Route of Administration
| Pharmacokinetic Parameter | Oral Route | Subcutaneous Route |
|---|---|---|
| Onset of Action | 30–90 minutes | 5–15 minutes |
| Time to Peak Effect | 60–90 minutes | 15–30 minutes |
| Duration of Action | ~1 hour (up to 6 hours with large doses) | ~2 hours |
| Data compiled from sources.3 |
The distribution of bethanechol is severely restricted by its molecular structure. As previously noted, the presence of a permanently charged quaternary amine makes the molecule highly hydrophilic and prevents it from crossing the lipid-rich blood-brain barrier.[1] This is a defining characteristic of the drug, effectively confining its pharmacological actions to the peripheral nervous system and preventing any direct effects on the central nervous system.[3]
A critical review of the available literature reveals a significant and persistent knowledge gap regarding the metabolism and excretion of bethanechol. Multiple authoritative sources, including the National Center for Biotechnology Information's StatPearls resource, explicitly state that the metabolic breakdown pathways and elimination mechanisms of bethanechol have not been extensively studied or fully understood.[3] This is a remarkable finding for a drug that has been in clinical use for over 80 years.
This uncertainty is contrasted by a statement from one source suggesting the drug "undergoes limited metabolism and is largely excreted unchanged in the urine".[33] However, this claim is not substantiated by primary pharmacokinetic studies within the provided materials and stands in direct opposition to the more cautious and prevalent view that these processes remain uncharacterized. An expert assessment must prioritize the consensus from clinical pharmacology resources, which acknowledge the lack of definitive data.
This major research gap has direct clinical consequences. For instance, the absence of clear metabolic data means that no specific dosage adjustment guidelines can be provided for patients with hepatic impairment.[28] While it is noted that no dosage adjustment is needed for renal impairment, the scientific basis for this recommendation is unclear without a complete understanding of the drug's excretion pathways.[28] This uncertainty underscores a critical need for modern pharmacokinetic studies to fully characterize the disposition of bethanechol in the human body.
Bethanechol occupies a specific niche in modern pharmacotherapy, with well-established indications for conditions involving smooth muscle atony. Its use is also being explored in several novel therapeutic areas.
The U.S. Food and Drug Administration (FDA) has approved bethanechol for the treatment of specific urological conditions where there is a functional, rather than mechanical, impairment of bladder emptying.[6]
Across all approved indications, the "non-obstructive" caveat is the single most important diagnostic criterion for its use. Before initiating therapy, it is imperative to rule out any physical blockage of the urinary tract (e.g., prostatic hypertrophy, urethral stricture).[2] Applying a powerful contractile stimulant against a mechanical obstruction can lead to dangerous increases in pressure, potentially causing vesicoureteral reflux or, in extreme cases, bladder rupture.[3]
The pharmacological properties of bethanechol have led to its use and investigation in a variety of other conditions beyond its FDA-approved labels.
Dosage of bethanechol must be carefully individualized based on the condition being treated, the route of administration, and the patient's response.[25] The range of dosing regimens is summarized in Table 5.1.
Table 5.1: Dosing Regimens for Bethanechol by Indication and Population
| Indication | Patient Population | Route | Recommended Dosage | Source(s) |
|---|---|---|---|---|
| FDA-Approved | ||||
| Urinary Retention (Non-obstructive) | Adult | Oral | 10–50 mg, 3 to 4 times daily. May titrate hourly from 5-10 mg up to a 50 mg max to find effective dose. | 25 |
| Neurogenic Bladder Atony | Adult | Oral | 10–50 mg, 3 to 4 times daily. | 3 |
| Urinary Retention (Acute) | Adult | Subcutaneous | 5 mg, 3 to 4 times daily. | 31 |
| Off-Label | ||||
| GERD | Adult | Oral | 25 mg, 4 times daily. | 8 |
| Urinary Retention | Pediatric | Oral | 0.3–0.6 mg/kg/day, divided into 3 to 4 doses. | 8 |
| GERD | Pediatric | Oral | 0.3–0.6 mg/kg/day, divided into 4 doses. | 8 |
| All oral doses should be taken on an empty stomach (1 hour before or 2 hours after meals).25 Subcutaneous administration is not for intramuscular or intravenous use.35 |
The safety profile of bethanechol is well-characterized and is almost entirely predictable from its mechanism of action as a global muscarinic agonist. Adverse events are extensions of its pharmacology, and contraindications are logically derived from conditions that would be exacerbated by parasympathetic stimulation.
The use of bethanechol is absolutely contraindicated in patients with certain pre-existing conditions where cholinergic stimulation would pose a significant risk. These contraindications represent patient groups with no physiological margin for cholinergic error, where a typical side effect could become a life-threatening event.
Adverse reactions are common, generally mild to moderate, and are dose-dependent. They are more frequent and intense following subcutaneous injection compared to oral administration.[29] The profile of ADRs is a coherent set of symptoms that can be reasoned from the drug's mechanism, not a random list to be memorized.
Overdosage with bethanechol results in a cholinergic crisis. Early signs are an exaggeration of the common adverse effects and include severe abdominal discomfort, excessive salivation, flushing of the skin, sweating, nausea, and vomiting.[25]
The specific antidote for bethanechol toxicity is Atropine Sulfate, a competitive muscarinic receptor antagonist. For adults, the recommended dose is 0.6 mg, administered subcutaneously or intravenously, which can be repeated every two hours as needed based on clinical response. For children, the dose is 0.01 mg/kg (up to a maximum of 0.4 mg).[25]
The drug interaction profile for bethanechol is overwhelmingly pharmacodynamic in nature, involving direct or indirect modulation of the cholinergic system at the receptor level. This observation aligns with the lack of well-defined metabolic pathways for the drug, as a compound that is not significantly metabolized is unlikely to participate in metabolism-based (i.e., pharmacokinetic) drug interactions.[30]
Interactions are broadly categorized as either antagonistic (reducing bethanechol's effect) or synergistic (increasing its effect and/or toxicity).
A summary of these key interactions is provided in Table 7.1.
Table 7.1: Clinically Significant Drug Interactions with Bethanechol
| Interacting Drug/Class | Mechanism of Interaction | Potential Clinical Outcome | Management Recommendation |
|---|---|---|---|
| Anticholinergic Agents (e.g., atropine, dicyclomine, amitriptyline) | Pharmacodynamic Antagonism (Competition at muscarinic receptors) | Decreased or abolished therapeutic effect of bethanechol. | Avoid combination if possible. Monitor for lack of efficacy. |
| Sympathomimetic Agents (e.g., pseudoephedrine, amphetamine) | Physiological Antagonism (Opposing effects of sympathetic vs. parasympathetic stimulation) | Offset of bethanechol's effects, particularly on the cardiovascular system. | Monitor clinical response (e.g., blood pressure, heart rate). |
| Cholinesterase Inhibitors (e.g., donepezil, neostigmine) | Pharmacodynamic Synergism (Increased overall cholinergic tone) | Increased risk and severity of cholinergic adverse effects (e.g., bradycardia, diarrhea, salivation). | Use with caution. Monitor closely for signs of cholinergic toxicity. |
| Other Muscarinic Agonists (e.g., pilocarpine, cevimeline) | Additive Pharmacodynamic Effects | Markedly increased risk of cholinergic toxicity. | Combination is generally not recommended. |
| Ganglion Blocking Compounds | Synergistic Hypotensive Effect | Critical fall in blood pressure. | Use with extreme caution and intensive monitoring. |
| Beta-Blockers | Additive Bradycardic Effect | Increased risk of severe bradycardia or heart block. | Monitor heart rate closely. |
| Data compiled from sources.10 |
The use of bethanechol requires special consideration in certain patient populations due to potential for altered pharmacodynamics, increased risk of adverse effects, or lack of safety data.
The safety and effectiveness of bethanechol in pediatric patients have not been formally established through rigorous clinical trials, and it is not FDA-approved for this population.[29] Despite this, it is used off-label in children and adolescents for conditions such as non-obstructive urinary retention and GERD.[8] When used in this population, dosing is typically based on body weight (mg/kg), and special care and close monitoring are required.[10]
Older adults may exhibit increased sensitivity to the effects of bethanechol. They may experience sensory responses in the bladder, such as urgency, at lower bladder volumes compared to younger adults.[3] Furthermore, the geriatric population is often at a higher risk for the adverse cardiovascular and neurological effects of the drug, including hypotension, bradycardia, dizziness, and lightheadedness. These side effects can significantly increase the risk of falls and related injuries in frail or elderly patients.[17] Therefore, cautious dosing and vigilant monitoring for adverse events are essential when prescribing bethanechol to older adults.
Bethanechol is a long-established parasympathomimetic agent whose clinical role is defined by its potent and selective muscarinic agonist activity. Its primary utility in managing non-obstructive urinary retention and neurogenic bladder atony is a direct result of its ability to stimulate detrusor muscle contraction. The drug's rational design, specifically its resistance to cholinesterase and its inability to cross the blood-brain barrier, underpins its therapeutic viability and favorable peripheral safety profile. The adverse effects and contraindications are well-understood and are predictable extensions of its core pharmacology, allowing for effective risk mitigation through careful patient selection and monitoring.
Despite its long history of use, this comprehensive review highlights critical areas for future investigation and development that could refine and potentially expand the role of bethanechol in medicine.
In conclusion, bethanechol remains a valuable and clinically relevant medication for its specific, well-defined indications. However, its full potential may yet be untapped. Addressing the fundamental gaps in its pharmacokinetic profile and rigorously exploring its novel therapeutic applications will ensure that this venerable drug can continue to contribute to patient care in the 21st century.
Published at: October 22, 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.