C21H24N4O2S
223673-61-8
Neurogenic Detrusor Overactivity, Overactive Bladder Syndrome (OABS)
Mirabegron (DrugBank ID: DB08893) is a first-in-class, orally active, selective beta-3 adrenergic receptor (β3-AR) agonist. It represents a significant therapeutic advancement in the management of urological storage disorders, offering a distinct mechanism of action compared to the long-standing class of antimuscarinic agents. Developed by Astellas Pharma and marketed under brand names including Myrbetriq®, Betmiga®, and Betanis®, Mirabegron is indicated for the treatment of overactive bladder (OAB) in adults with symptoms of urge urinary incontinence, urgency, and urinary frequency. Its approval has been extended to include treatment of neurogenic detrusor overactivity (NDO) in pediatric patients and for use in combination with the antimuscarinic solifenacin succinate for refractory OAB in adults.[1]
The primary pharmacodynamic effect of Mirabegron is the relaxation of the detrusor smooth muscle during the bladder's storage phase. This is achieved through selective activation of β3-ARs, which are highly expressed in the bladder, leading to an increase in intracellular cyclic adenosine monophosphate (cAMP).[4] This mechanism increases the functional capacity of the bladder, thereby alleviating the core symptoms of OAB. Crucially, Mirabegron lacks significant antimuscarinic activity, which is the pharmacological basis for its principal clinical advantage: a more favorable tolerability profile than traditional anticholinergic drugs, with a markedly lower incidence of dry mouth and constipation.[1]
Clinical evidence from extensive Phase 3 trials demonstrates that Mirabegron has efficacy comparable to that of standard antimuscarinic agents like solifenacin and tolterodine but offers a superior balance of efficacy and tolerability.[7] This profile establishes it as a valuable first-line option for many patients, particularly the elderly or those with contraindications to anticholinergics, and as a definitive second-line therapy for patients who are intolerant to antimuscarinic side effects.
The safety profile of Mirabegron is well-defined but distinct from that of antimuscarinics. The primary safety concerns are dose-dependent increases in blood pressure and heart rate, necessitating periodic monitoring, especially in hypertensive patients. The drug is not recommended for use in individuals with severe uncontrolled hypertension.[5] Other notable risks include rare but serious angioedema and a potential for urinary retention in patients with bladder outlet obstruction.[10] From a pharmacokinetic perspective, Mirabegron is a moderate inhibitor of the cytochrome P450 2D6 (CYP2D6) enzyme, creating a significant potential for drug-drug interactions with CYP2D6 substrates, particularly those with a narrow therapeutic index.[1]
In summary, Mirabegron has secured a critical position in the clinical armamentarium for bladder dysfunction. Its unique mechanism, proven efficacy, and favorable tolerability have addressed a long-standing unmet need in OAB management, providing a vital alternative for patients and establishing a new paradigm for combination therapy in refractory cases.
Overactive Bladder (OAB) is a highly prevalent and often distressing symptom complex, defined by the International Continence Society as the presence of urinary urgency, which is typically accompanied by increased urinary frequency and nocturia, with or without urge urinary incontinence.[12] It is crucial to recognize that OAB is not a discrete disease entity but rather a clinical syndrome that can arise from various underlying etiologies.[12] The central pathophysiological feature of OAB is the involuntary contraction of the detrusor muscle, the smooth muscle layer of the bladder wall, during the bladder's storage or filling phase.[5] Normal micturition is regulated by a complex interplay between the sympathetic and parasympathetic nervous systems. During bladder filling, sympathetic stimulation promotes detrusor relaxation and bladder neck contraction, allowing for low-pressure urine storage. During voiding, parasympathetic stimulation, mediated by acetylcholine (ACh) release, triggers detrusor contraction.[6] In OAB, this control is disrupted, leading to inappropriate detrusor contractions that generate the hallmark sensation of urgency.[5] These contractions are primarily driven by the parasympathetic nervous system, with ACh acting on M2 and M3 muscarinic receptor subtypes on detrusor muscle cells.[6]
Neurogenic Detrusor Overactivity (NDO) is a related but distinct condition characterized by bladder dysfunction that stems from a confirmed neurological impairment, such as a spinal cord injury, multiple sclerosis, or spina bifida.[1] While the end result is also involuntary detrusor contractions, the origin is a disruption in the nerve signals between the brain, spinal cord, and bladder.
For over three decades, the cornerstone of pharmacological management for OAB was the antimuscarinic (or anticholinergic) class of drugs.[15] These agents, such as oxybutynin, tolterodine, and solifenacin, exert their therapeutic effect by competitively blocking the action of acetylcholine at M2 and M3 muscarinic receptors on the detrusor muscle, thereby inhibiting involuntary bladder contractions and increasing bladder capacity.[1]
Despite their established efficacy, the clinical utility of antimuscarinics has been consistently hampered by a significant treatment-limiting side-effect profile.[1] Muscarinic receptors are not confined to the bladder; they are widely distributed throughout the body, including in the salivary glands, gastrointestinal tract, ciliary body of the eye, and central nervous system. Consequently, systemic blockade of these receptors leads to a constellation of well-known adverse effects, including dry mouth (xerostomia), constipation, blurred vision, and central nervous system effects such as somnolence, confusion, and cognitive impairment.[7] These side effects are often bothersome enough to cause poor patient tolerance, low adherence to therapy, and high rates of treatment discontinuation, frequently within the first few months of initiation.[7] This created a substantial unmet clinical need for an effective OAB treatment with a different mechanism and a more favorable tolerability profile.
The advent of Mirabegron, developed by Astellas Pharma, marked a paradigm shift in the pharmacological approach to OAB.[15] Approved by the U.S. Food and Drug Administration (FDA) in 2012, it was the first clinically available β3-adrenergic agonist for the treatment of OAB, representing the first new oral treatment class for the condition in over 30 years.[1]
The introduction of Mirabegron was not merely an incremental improvement but a fundamental change in therapeutic strategy. It shifted the pharmacological target away from the parasympathetic pathway responsible for bladder contraction to the sympathetic pathway involved in bladder relaxation. The human bladder expresses a high concentration of β3-adrenergic receptors, which, when stimulated, promote detrusor muscle relaxation.[6] By selectively targeting this pathway, Mirabegron was designed to improve the bladder's storage capacity without the systemic muscarinic blockade that plagued older therapies. The primary clinical value of Mirabegron, as borne out by extensive clinical trials, lies not in demonstrating superior efficacy over antimuscarinics, but in offering a comparable level of efficacy with a significantly improved tolerability profile. This distinct risk-benefit balance directly addresses the principal limitations of antimuscarinic therapy, providing a crucial alternative for patients who are intolerant to or have contraindications for the older class of drugs.
Mirabegron is classified chemically as a monocarboxylic acid amide and is a member of the 1,3-thiazole and ethanolamine structural families.[1] It is obtained through the formal condensation of the carboxyl group of 2-amino-1,3-thiazol-4-ylacetic acid with the anilino group of (1R)-2-{[2-(4-aminophenyl)ethyl]amino}-1-phenylethanol.[1]
Its formal International Union of Pure and Applied Chemistry (IUPAC) name is 2-(2-amino-1,3-thiazol-4-yl)-N-amino]ethyl]phenyl]acetamide.[1] The molecular formula is
C21H24N4O2S.[1] For structural informatics and database cross-referencing, its key identifiers are:
Mirabegron presents as a white to light-yellow or off-white crystalline solid or powder.[19] It has a molecular weight of 396.51 g/mol.[9] The reported melting point varies slightly across sources, with ranges cited as 138-140°C, 141.0-145.0°C, and a specific value of 143°C.[19]
The compound's solubility profile is a critical determinant of its pharmaceutical formulation. It is considered practically insoluble in water, with a measured solubility of 0.082 g/L.[21] It exhibits slight solubility in organic solvents such as methanol, ethanol, and chloroform.[20] Due to its sensitivity to air and heat, Mirabegron requires refrigerated storage (0-10°C) under an inert atmosphere to maintain stability.[20]
The intrinsic properties of Mirabegron, particularly its long elimination half-life of approximately 50 hours in adults and its poor water solubility, directly guided its development into extended-release dosage forms suitable for once-daily administration.[1] An immediate-release formulation would risk incomplete and variable absorption, while also potentially causing sharp peaks in plasma concentration (Cmax) that could exacerbate dose-dependent side effects like hypertension.[25] The extended-release technology mitigates these issues by ensuring a slow, controlled dissolution and absorption of the drug, which flattens the pharmacokinetic profile, maintains therapeutic concentrations over 24 hours, and enhances both efficacy and safety.
Two primary formulations are commercially available:
Table 1: Chemical and Physical Identifiers of Mirabegron | ||
---|---|---|
Property | Value | Source(s) |
CAS Number | 223673-61-8 | 1 |
DrugBank ID | DB08893 | 1 |
Molecular Formula | C21H24N4O2S | 1 |
Molecular Weight | 396.51 g/mol | 9 |
IUPAC Name | 2-(2-amino-1,3-thiazol-4-yl)-N-amino]ethyl]phenyl]acetamide | 1 |
Appearance | White to light yellow/off-white solid or powder | 19 |
Melting Point | 138-145 °C (range from various sources) | 19 |
Solubility in Water | Insoluble (0.082 g/L) | 21 |
Solubility (Organic) | Slightly soluble in methanol, ethanol, chloroform | 20 |
InChIKey | PBAPPPCECJKMCM-IBGZPJMESA-N | 1 |
The therapeutic action of Mirabegron is rooted in its function as a potent and selective agonist of the human β3-adrenergic receptor (β3-AR).[4] The human urinary bladder is an ideal target for such an agent, as quantitative PCR studies have revealed that the β3-AR subtype constitutes more than 95% of the total β-adrenergic receptor mRNA present in the detrusor muscle.[6] This high density and specificity provide a strong rationale for targeted therapy.
Mirabegron exhibits remarkable selectivity for the β3-AR. In vitro biochemical assays demonstrate its potent agonistic activity at the human β3-AR, with a half-maximal effective concentration (EC50) of approximately 22.4 nM.[20] In stark contrast, its activity at β1-AR and β2-ARs is negligible at therapeutic concentrations, with
EC50 values exceeding 10,000 nM.[20] Clinically, evidence of β1-AR stimulation, such as a significant increase in heart rate, is generally observed only at supratherapeutic doses of 200 mg or higher, well above the recommended maximum daily dose of 50 mg.[5] This high degree of selectivity is a cornerstone of the drug's design, enabling a therapeutic window where effects on the bladder are maximized while minimizing off-target cardiovascular effects commonly associated with non-selective beta-agonists.
The β3-AR is a G-protein coupled receptor (GPCR), specifically linked to the stimulatory G-protein, Gs.[4] Upon binding of Mirabegron to the β3-AR, the Gs protein is activated. This activation, in turn, stimulates the membrane-bound enzyme adenylyl cyclase.[4] The primary function of adenylyl cyclase is to catalyze the conversion of adenosine triphosphate (ATP) into the second messenger molecule, cyclic adenosine monophosphate (cAMP).[4] The resulting elevation in intracellular cAMP concentration is the key biochemical event that initiates the downstream signaling cascade responsible for smooth muscle relaxation.[6] While the cAMP pathway is the principal accepted mechanism, some evidence also suggests that β-AR stimulation may contribute to relaxation by activating large-conductance calcium-activated potassium (
BKCa) channels, which would lead to hyperpolarization of the muscle cell membrane and reduced contractility.[18]
The ultimate physiological consequence of β3-AR activation by Mirabegron is the relaxation of the detrusor smooth muscle.[1] This effect is most relevant during the storage phase of the micturition (fill-void) cycle.[5] By relaxing the bladder wall, Mirabegron increases the functional capacity of the bladder, allowing it to store a larger volume of urine at low pressure.[4] This increased storage capacity directly addresses the core symptoms of OAB by reducing the frequency of urination, lessening the sensation of urgency, and decreasing the number of urge incontinence episodes.[4]
Furthermore, research suggests that Mirabegron may also exert a beneficial effect by suppressing non-voiding contractions, or "micromotions," of the bladder.[4] These subtle, spontaneous contractions during the filling phase are thought to be a significant source of afferent nerve signaling that contributes to the sensation of urgency, and their inhibition may be an additional mechanism by which Mirabegron provides symptomatic relief.[18]
A defining pharmacodynamic characteristic of Mirabegron is its lack of significant affinity for and activity at muscarinic receptors.[1] This is the fundamental distinction between Mirabegron and the entire class of anticholinergic agents. By avoiding the muscarinic pathway, Mirabegron circumvents the mechanism responsible for the hallmark side effects of anticholinergic drugs. This selectivity is the direct pharmacological basis for its significantly more favorable tolerability profile, particularly the much lower incidence of dry mouth, constipation, blurred vision, and cognitive side effects that often limit the use of antimuscarinics.[7]
The pharmacodynamic profile of Mirabegron extends beyond the urinary system. β3-ARs are also highly expressed in brown adipose tissue (BAT), the primary organ for nonshivering thermogenesis in mammals.[9] Studies in both animals and humans have demonstrated that Mirabegron is an effective activator of BAT.[31] Activation of β3-ARs in brown adipocytes stimulates the expression of uncoupling protein 1 (UCP1), which uncouples mitochondrial respiration from ATP synthesis, dissipating energy as heat.[31] This process consumes glucose and lipids, leading to increased energy expenditure.
This discovery has profound implications, suggesting that Mirabegron is not merely a "bladder drug" but a systemic β3-AR agonist with potential pleiotropic metabolic effects. This has spurred significant research interest into the potential repurposing of Mirabegron or the development of new β3-AR agonists for the treatment of metabolic disorders such as obesity and type 2 diabetes.[9] For instance, preclinical studies in mice have shown that combining Mirabegron with metformin resulted in greater weight loss than either agent alone, highlighting a potential synergistic effect.[9] This opens up an entirely new avenue of investigation, raising questions about whether long-term Mirabegron use for OAB might confer unintended but potentially beneficial metabolic effects in patients, a third-order question that warrants clinical exploration.
The pharmacokinetic profile of Mirabegron is characterized by oral absorption from an extended-release formulation, extensive tissue distribution, complex metabolism involving multiple enzymatic pathways, and elimination through both renal and fecal routes. This profile has significant clinical implications for dosing, drug interactions, and use in special populations.
Following oral administration of the extended-release tablet, Mirabegron reaches maximum plasma concentrations (Tmax) in approximately 3.5 hours.[12] Its absolute bioavailability is dose-dependent, increasing from 29% at the 25 mg dose to 35% at the 50 mg dose, suggesting that absorption processes become more efficient at the higher therapeutic dose.[25]
The pharmacokinetics of Mirabegron are non-linear at doses above the therapeutic range. The area under the concentration-time curve (AUC) and maximum concentration (Cmax) increase in a manner that is more than dose-proportional.[25] For example, doubling the dose from 50 mg to 100 mg results in a 2.6-fold increase in AUC and a 2.9-fold increase in
Cmax.[25] This non-linearity underscores the importance of adhering to recommended dosing and not exceeding the 50 mg maximum daily dose.
In adults, the tablet can be administered with or without food, as food does not significantly impact its overall exposure.[14] However, for pediatric patients receiving the granule suspension, administration
with food is recommended to lower the risk of side effects such as tachycardia.[13] Steady-state plasma concentrations are typically achieved within 7 days of initiating once-daily dosing.[25]
Mirabegron undergoes extensive distribution into body tissues, which is reflected by its large apparent steady-state volume of distribution (Vd) of approximately 1670 L following intravenous administration.[20] This value, far exceeding total body water, indicates significant partitioning of the drug from the plasma into peripheral tissues. In human plasma, Mirabegron is moderately bound to proteins (approximately 71%), primarily to albumin and alpha-1 acid glycoprotein.[1]
Mirabegron is subject to extensive metabolism via several parallel pathways, including amide hydrolysis, oxidative metabolism, and direct glucuronidation.[20] Despite this extensive biotransformation, the unchanged parent drug remains the major circulating component in plasma following oral administration.[25]
The oxidative metabolism of Mirabegron is mediated by two key cytochrome P450 isoenzymes: CYP3A4 and CYP2D6.[1] The conjugation reactions (glucuronidation) are carried out by UDP-glucuronosyltransferase (UGT) enzymes, specifically UGT2B7, UGT1A3, and UGT1A8.[25] Other enzymes, including butylcholinesterase and alcohol dehydrogenase, are also thought to contribute to its metabolism.[25]
This complex metabolic profile creates a high potential for drug-drug interactions. A critical pharmacokinetic property of Mirabegron is that it acts as a moderate inhibitor of CYP2D6.[1] This dual role—being both a substrate for multiple enzymes and an inhibitor of a key enzyme—is a central consideration in its clinical use. Its inhibitory action on CYP2D6 means it can significantly increase the plasma concentrations of co-administered drugs that are primarily metabolized by this enzyme, a point of major clinical significance detailed in Section 9.0.
In individuals who are genetically deficient in CYP2D6 activity (CYP2D6 poor metabolizers), the mean Cmax and AUC of Mirabegron are approximately 16-17% higher compared to extensive metabolizers. This modest increase is not considered clinically significant enough to warrant routine genetic testing or a priori dose adjustments.[38]
Mirabegron and its metabolites are eliminated from the body through both the kidneys and the liver. Following administration of a radiolabeled dose, approximately 55% of the radioactivity is recovered in the urine and 34% is recovered in the feces.[9] Renal elimination of the parent drug accounts for about 25% of the total dose and occurs through a combination of glomerular filtration and active tubular secretion.[20] The active secretion is mediated by organic cation transporters (OCT).
The terminal elimination half-life (t1/2) of Mirabegron is notably long, which supports the once-daily dosing regimen.
The clearance of Mirabegron is significantly affected by both renal and hepatic function, necessitating dose adjustments in patients with organ impairment.
Table 2: Summary of Key Pharmacokinetic Parameters in Adult and Pediatric Populations | ||
---|---|---|
Parameter | Adult Population | Pediatric Population (3 to <18 years) |
Formulation | Extended-Release Tablet | Extended-Release Granules for Suspension |
Absolute Bioavailability | 29% (25 mg dose) to 35% (50 mg dose) 25 | Not specifically stated, but dosing is weight-based to achieve target exposures. |
Time to Peak (Tmax) | ~3.5 hours 12 | ~4-5 hours 25 |
Elimination Half-Life (t1/2) | ~50 hours 20 | ~26-31 hours 25 |
Volume of Distribution (Vd) | ~1670 L 20 | Proportional to body weight, higher relative to adults 12 |
Plasma Protein Binding | ~71% 12 | Not specifically stated, presumed similar. |
Food Effect | No clinically significant effect 29 | Recommended to be taken with food 13 |
Mirabegron is formally indicated for the symptomatic treatment of OAB in adults, including the management of urge urinary incontinence, urgency, and urinary frequency.[1] Its efficacy was established in a series of large, multinational, randomized, placebo-controlled Phase 3 clinical trials involving over 10,000 patients.[15]
In these pivotal 12-week studies, both the 25 mg and 50 mg once-daily doses of Mirabegron demonstrated statistically significant and clinically meaningful improvements in the co-primary efficacy endpoints compared to placebo.[6] For instance, data submitted for its initial approval showed that treatment with Mirabegron 50 mg reduced the mean number of incontinence episodes by 1.49 per 24 hours (from a baseline of 2.71) and the mean number of micturitions by 1.75 per 24 hours (from a baseline of 11.70). These reductions were statistically significant versus the changes observed with placebo.[15] The therapeutic effect of Mirabegron is typically apparent within the first few weeks of treatment, with maximal efficacy often observed by 8 weeks.[10]
In March 2021, the FDA expanded the approved use of Mirabegron to include the treatment of NDO in pediatric patients aged 3 years and older.[1] This indication addresses a significant need in a vulnerable population where bladder dysfunction is secondary to neurological conditions. The approval was specifically for a new formulation, prolonged-release granules for oral suspension, to allow for accurate weight-based dosing.[13] The efficacy in this population was demonstrated in a clinical study involving 86 children and adolescents. The primary measure of effectiveness was the change in maximum cystometric capacity (MCC), which is the volume of urine the bladder can comfortably hold. After 24 weeks of treatment, patients receiving Mirabegron showed a mean increase in MCC of approximately 87 mL, indicating an improved ability of the bladder to store urine.[28]
A major evolution in the management of OAB was the FDA approval in May 2018 for the use of Mirabegron in combination with the M3-selective antimuscarinic agent, solifenacin succinate.[2] This approval provides an evidence-based option for patients who have an inadequate response to monotherapy with either agent alone.[10]
This strategy is built upon a strong pharmacological rationale of complementary mechanisms of action. Mirabegron promotes detrusor relaxation via the sympathetic β3-AR pathway, while solifenacin inhibits involuntary detrusor contractions by blocking the parasympathetic M3 muscarinic pathway.[1] This dual-pathway approach offers the potential for an additive therapeutic effect.
The efficacy of this combination was rigorously established in large-scale clinical trials, including the SYNERGY I & II and BESIDE studies.[3] These trials demonstrated that combination therapy with Mirabegron and solifenacin was statistically superior to monotherapy with either drug alone in improving key OAB symptoms, such as the mean number of incontinence episodes, micturitions, and urgency episodes per 24 hours.[3] Importantly, this enhanced efficacy was achieved without a clinically meaningful increase in the overall incidence of adverse events, establishing the combination as a valuable and well-tolerated third-line treatment strategy before consideration of more invasive procedures like onabotulinumtoxinA injections or sacral neuromodulation.
The therapeutic potential of Mirabegron may extend beyond its currently approved indications. There is ongoing research into its utility for other urological conditions. Notably, a clinical trial (NCT05617664) is actively recruiting participants to evaluate the efficacy of Mirabegron 25 mg for the treatment of primary nocturnal enuresis, commonly known as bedwetting, in adults.[45] The rationale for this investigation likely stems from Mirabegron's established ability to increase bladder capacity, which could be beneficial in preventing nocturnal voiding.
A comprehensive assessment of Mirabegron's place in therapy requires a detailed analysis of its performance relative to placebo, the established class of anticholinergic agents, and newer drugs within its own class. The collective evidence paints a clear picture of a drug defined more by its favorable tolerability profile than by superior efficacy.
The efficacy of Mirabegron is unequivocally established against placebo. The initial regulatory approvals in the US and Europe were based on a robust program of three large, 12-week, randomized, double-blind Phase 3 studies (e.g., SCORPIO, ARIES, CAPRICORN).[15] Across these trials, both the 25 mg and 50 mg once-daily doses of Mirabegron consistently demonstrated statistically significant superiority over placebo in reducing the co-primary endpoints of mean change from baseline in incontinence episodes and micturition frequency per 24 hours.[15]
Direct comparisons with the former standard of care, anticholinergic drugs, are crucial for positioning Mirabegron in clinical practice.
The introduction of a second β3-agonist, Vibegron (Gemtesa®), has prompted direct comparisons to further refine the role of this drug class.
The choice between Mirabegron and an anticholinergic is therefore a classic clinical trade-off. It requires a personalized assessment, weighing the adrenergic risks of Mirabegron (hypertension) against the anticholinergic risks of the alternative (dry mouth, constipation, cognitive effects). Mirabegron's role is not to replace anticholinergics universally, but to provide a mechanistically distinct alternative that allows for more individualized and better-tolerated management of OAB.
Table 3: Summary of Pivotal Phase III Clinical Trials vs. Placebo and Anticholinergics | |||||
---|---|---|---|---|---|
Trial (Identifier) | Comparator(s) | Patient Population | Primary Endpoints | Key Efficacy Results (Change from Baseline) | Key Tolerability Finding (Incidence of Dry Mouth) |
SCORPIO (178-CL-046) 15 | Placebo, Tolterodine ER 4 mg | Adults with OAB (US) | Change in micturitions/24h; Change in incontinence episodes/24h | Mira 50mg: -1.8 micturitions; -1.5 episodes. Statistically significant vs. placebo. | Mira 50mg: 2.8% vs. Placebo: 2.3% vs. Tolterodine: 10.1% |
ARIES (178-CL-047) 15 | Placebo | Adults with OAB (US) | Change in micturitions/24h; Change in incontinence episodes/24h | Mira 50mg: -1.9 micturitions; -1.5 episodes. Statistically significant vs. placebo. | Mira 50mg: 3.4% vs. Placebo: 2.0% |
CAPRICORN (178-CL-049) 16 | Placebo | Adults with OAB (Europe/Australia) | Change in micturitions/24h; Change in incontinence episodes/24h | Mira 50mg: -1.9 micturitions; -1.6 episodes. Statistically significant vs. placebo. | Mira 50mg: 3.0% vs. Placebo: 2.1% |
BESIDE (NCT01908829) 3 | Solifenacin 5 mg monotherapy | Adults with OAB inadequately treated with solifenacin | Change in micturitions/24h; Change in incontinence episodes/24h | Combination therapy (Mira 50mg + Soli 5mg) was statistically superior to solifenacin 5mg monotherapy on all primary endpoints. | Combination: 9.3% vs. Soli 5mg: 6.5% |
The safety profile of Mirabegron is well-characterized through extensive clinical trials and over a decade of post-marketing surveillance. Its adverse event profile is fundamentally different from that of antimuscarinic agents, trading cholinergic side effects for adrenergic ones.
Data pooled from Phase 3 clinical trials provide a clear picture of the most common adverse reactions associated with Mirabegron.[9]
The official prescribing information for Mirabegron includes several important warnings and precautions that guide its safe use.
Post-marketing surveillance provides valuable insights into the real-world safety profile of a drug. An analysis of the FDA Adverse Event Reporting System (FAERS) database from 2012 to 2023, encompassing over 18,000 reports associated with Mirabegron, confirmed the known common adverse events identified in clinical trials, such as hypertension, urinary retention, atrial fibrillation, and tachycardia.[49]
This pharmacovigilance study also identified several new, unexpected, and serious potential safety signals, including arrhythmia, palpitations, dementia, transient ischemic attack (TIA), Parkinson's disease, and ANCA-positive vasculitis.[50] It is critical to note that FAERS data can show an association but cannot establish causality. These signals require further investigation through rigorous epidemiological studies to determine if a true causal link exists. The potential signal for neurological events like dementia is particularly noteworthy, as a presumed lack of CNS effects is a key theoretical advantage over anticholinergics. The FAERS analysis also found that while the majority of adverse events (56%) had an onset within the first 30 days of treatment, reports of adverse events occurring even after one year of use were possible.[50]
Extensive non-clinical toxicology studies in animals were conducted to support the safety of Mirabegron.
Table 4: Comparative Incidence of Key Adverse Events (Mirabegron vs. Anticholinergics) | ||||
---|---|---|---|---|
Adverse Event | Placebo | Mirabegron 25 mg | Mirabegron 50 mg | Solifenacin 5 mg |
Hypertension | 7.6% | 11.3% | 7.5% | (Not typically elevated) |
Urinary Tract Infection | 1.8% | 4.2% | 2.9% | 3.6% |
Headache | 3.0% | 2.1% | 3.2% | (Variable, ~2-4%) |
Dry Mouth | 2.2% - 2.3% | 2.8% - 3.8% | 2.8% - 3.4% | ~23% - 28% |
Constipation | 1.2% | 1.2% | 2.8% | ~13% - 15% |
Data compiled and synthesized from multiple sources, including prescribing information and clinical trial reports, to represent typical incidences. Solifenacin data is representative of the anticholinergic class. 7 |
This table visually quantifies the fundamental risk-benefit trade-off in OAB pharmacotherapy. A clinician and patient choosing Mirabegron are accepting a higher risk of increased blood pressure in exchange for a substantially lower risk of the bothersome anticholinergic effects of dry mouth and constipation.
The complex metabolism and transport of Mirabegron create a significant potential for drug-drug interactions (DDIs). A thorough understanding of these interactions is essential for its safe clinical use, particularly in patients with polypharmacy. The most critical interaction involves its inhibition of the CYP2D6 enzyme.
Mirabegron is a moderate inhibitor of the cytochrome P450 2D6 (CYP2D6) isoenzyme.[1] This is its most clinically significant pharmacokinetic property. By inhibiting CYP2D6, Mirabegron can decrease the metabolism and thereby increase the plasma concentrations and potential for toxicity of co-administered drugs that are substrates of this enzyme.
This interaction is of particular concern for CYP2D6 substrates that have a narrow therapeutic index, where a modest increase in exposure can lead to serious adverse events. Therefore, appropriate monitoring and potential dose adjustment of the co-administered drug are necessary when used with Mirabegron. Drugs in this category include:
The interaction also affects more commonly used drugs, such as the beta-blocker metoprolol, where co-administration with Mirabegron leads to increased exposure.[11] This DDI profile means that a clinician must perform a careful medication review before prescribing Mirabegron. In a patient taking a narrow therapeutic index CYP2D6 substrate, an alternative OAB agent without this interaction, such as an anticholinergic or vibegron, may be a safer choice.
Mirabegron itself is a substrate of CYP3A4, another major drug-metabolizing enzyme.[25]
Table 6: Guide to Clinically Significant Drug-Drug Interactions with Mirabegron | |||
---|---|---|---|
Interacting Drug/Class | Mechanism of Interaction | Clinical Effect | Recommended Management |
CYP2D6 Substrates (especially with narrow therapeutic index, e.g., flecainide, propafenone, thioridazine, desipramine) | Mirabegron is a moderate CYP2D6 inhibitor. 10 | Increased plasma concentrations and potential toxicity of the co-administered drug. | Use with caution. Appropriate monitoring (e.g., ECG, drug levels) and potential dose adjustment of the CYP2D6 substrate may be necessary. Consider alternative OAB therapy. 10 |
Strong CYP3A4 Inhibitors (e.g., ketoconazole, itraconazole, ritonavir, clarithromycin) | Inhibition of CYP3A4-mediated metabolism of Mirabegron. 36 | Increased plasma concentrations and potential toxicity of Mirabegron. | Reduce Mirabegron dose to a maximum of 25 mg once daily. Not recommended in patients with severe renal or moderate/severe hepatic impairment. 36 |
Digoxin | Mirabegron inhibits P-glycoprotein (P-gp), a transporter responsible for digoxin efflux. 11 | Increased plasma concentrations and potential toxicity of digoxin. | When initiating, prescribe the lowest dose of digoxin. Monitor serum digoxin concentrations and titrate digoxin dose based on clinical effect. 11 |
Antimuscarinic Agents (e.g., solifenacin, tolterodine) | Additive pharmacodynamic effects. | Increased risk of urinary retention, especially in patients with Bladder Outlet Obstruction (BOO). 10 | Administer with caution. Monitor for signs and symptoms of urinary retention. 10 |
CYP3A4 Inducers (e.g., rifampin) | Induction of CYP3A4-mediated metabolism of Mirabegron. 36 | Decreased plasma concentrations of Mirabegron, potentially reducing efficacy. | No dose adjustment for Mirabegron is generally required. 11 |
The dosing of Mirabegron is tailored to the specific indication and patient population.
Proper administration is crucial for ensuring the efficacy and safety of Mirabegron's extended-release formulations.
Mirabegron clearance is dependent on both renal and hepatic function, requiring specific dose adjustments in patients with impairment.
Effective patient management involves clear counseling and routine monitoring.
Table 5: Dosing Recommendations and Adjustments for Special Populations | |||
---|---|---|---|
Patient Population | Starting Dose | Maximum Dose | Special Considerations |
Adult OAB (Normal Function) | 25 mg once daily | 50 mg once daily | May increase dose after 4-8 weeks based on efficacy/tolerability. 10 |
Pediatric NDO (by weight) | 3-6 mL (24-48 mg) once daily | 6-10 mL (48-80 mg) once daily | Dose is weight-based using oral suspension. Must be taken with food. 13 |
Severe Renal Impairment (eGFR 15-29) | 25 mg once daily | 25 mg once daily | Dose increase is not recommended. 10 |
ESRD (eGFR <15) / Dialysis | Not Recommended | Not Recommended | Insufficient data to support safe use. 10 |
Moderate Hepatic Impairment (Child-Pugh B) | 25 mg once daily | 25 mg once daily | Dose increase is not recommended. 10 |
Severe Hepatic Impairment (Child-Pugh C) | Not Recommended | Not Recommended | Not studied in this population. 12 |
Concomitant use with Strong CYP3A4 Inhibitors | 25 mg once daily | 25 mg once daily | Further restrictions apply if patient also has renal/hepatic impairment. 36 |
The regulatory journey of Mirabegron reflects a well-executed lifecycle management strategy, beginning with a core indication and methodically expanding to include combination therapy and a special pediatric population.
The development and approval process in the United States, overseen by the FDA, followed a clear timeline:
This stepwise expansion of the drug's label—from a foundational adult monotherapy indication to combination use and finally to a specialized pediatric indication with a new formulation—demonstrates a strategic approach to maximizing the clinical and commercial value of the asset over its lifecycle.
Mirabegron underwent a parallel regulatory review process in Europe under the oversight of the European Medicines Agency (EMA).
Mirabegron is marketed globally under several distinct brand names, reflecting regional marketing strategies by Astellas Pharma.
Mirabegron has established itself as a cornerstone in the management of OAB and pediatric NDO, primarily by offering a distinct and favorable risk-benefit profile compared to the long-established antimuscarinic class. Its core clinical benefit is the provision of effective OAB symptom control, with a magnitude of effect comparable to that of standard anticholinergics, for a population of patients who are often unable to tolerate traditional therapy. The complementary mechanism of action also provides a strong rationale for its use in combination therapy for refractory cases.
This benefit must be weighed against a well-defined set of risks. The primary safety concerns are adrenergic in nature and are a direct consequence of its mechanism of action. These include dose-dependent increases in blood pressure and heart rate, which necessitate careful patient selection and monitoring. The risk of rare but serious angioedema, while low, requires immediate recognition and drug discontinuation. From a clinical pharmacology standpoint, the most significant risk is its potential for drug-drug interactions stemming from its role as a moderate inhibitor of CYP2D6, which complicates its use in the context of polypharmacy, particularly with narrow therapeutic index drugs.
Overall, for a properly selected patient—one without severe uncontrolled hypertension, significant bladder outlet obstruction, or conflicting medications—the risk-benefit profile of Mirabegron is highly favorable. It successfully addresses the most common reason for treatment failure with antimuscarinics: intolerable side effects.
The positioning of Mirabegron in clinical guidelines has evolved as experience with the drug has grown. It is now firmly established as a first-line pharmacotherapy option for OAB, alongside antimuscarinic agents. The choice between these two classes for a treatment-naïve patient should be guided by an individualized assessment of their comorbidities and risk factors.
Clinical Recommendations:
Future Research Directions:
Published at: July 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.