Bretaris Genuair, Duaklir, Duaklir Genuair, Eklira Genuair, Tudorza, Tudorza Genuair, Brimica Genuair
Small Molecule
C26H30NO4S2
727649-81-2
Chronic Obstructive Pulmonary Disease (COPD)
Aclidinium bromide is a small molecule pharmaceutical agent classified as a long-acting muscarinic antagonist (LAMA), a subclass of anticholinergic bronchodilators.[1] Its chemical identity is precisely defined by a comprehensive set of internationally recognized identifiers, which are essential for its unambiguous reference in clinical, regulatory, and research contexts.
The International Nonproprietary Name (INN) for the active substance is Aclidinium Bromide.[2] Chemically, it is a quaternary ammonium ion and a carboxylic ester, structurally characterized as a member of thiophenes and an aromatic ether, functionally related to 3-quinuclidinol.[1] For database and informatics purposes, it is assigned DrugBank ID DB08897 [1], ChEBI ID CHEBI:65346 [1], and ChEMBL ID CHEMBL1194325.[1]
A point of potential confusion is the existence of two primary CAS Numbers associated with the substance. The CAS Number 727649-81-2 refers to the aclidinium cation itself, while 320345-99-1 is often used for the bromide salt form in which the drug is formulated and administered.[1] Its IUPAC name is formally stated as-1-azoniabicyclo[2.2.2]octan-8-yl] 2-hydroxy-2,2-di(thiophen-2-yl)acetate or, more completely for the salt,octan-3-yl] 2-hydroxy-2,2-dithiophen-2-ylacetate;bromide.[3]
Aclidinium bromide is marketed globally under several brand names. In the United States, it is known as Tudorza Pressair.[2] In the United Kingdom and other European Union member states, it is available as
Eklira Genuair.[2] The name
Bretaris Genuair is used in the majority of EU states where it is licensed to Menarini.[2] In Canada, it is marketed as
Tudorza Genuair.[2] Furthermore, it is available in a fixed-dose combination with the long-acting beta-agonist (LABA) formoterol, marketed in the European Union under the brand names
Duaklir Genuair and Brimica Genuair.[2] Other key identifiers include the FDA UNII code K17VY42F6C and the RxNorm Concept Unique Identifier (RXCUI) 1303098.[1]
The molecular formula for aclidinium bromide is C26H30NO4S2⋅Br, with a corresponding molecular weight of approximately 564.55 g/mol.[5] The molecular formula for the active cation alone is
C26H30NO4S2+.[9] Its precise chemical structure is captured by standard chemical informatics identifiers, including the InChI string
InChI=1S/C26H30NO4S2.BrH/c28-25(26(29,23-9-4-17-32-23)24-10-5-18-33-24)31-22-19-27(14-11-20(22)12-15-27)13-6-16-30-21-7-2-1-3-8-21;/h1-5,7-10,17-18,20,22,29H,6,11-16,19H2;1H/q+1;/p-1/t20?,22-,27?;/m0./s1 and the InChIKey XLAKJQPTOJHYDR-QTQXQZBYSA-M.[2]
A critical aspect of aclidinium's structure is its stereochemistry. The molecule contains an asymmetric carbon atom and is developed and utilized exclusively as the pure R-enantiomer.[2] This stereochemical specificity is fundamental to its interaction with muscarinic receptors and, consequently, its pharmacological activity and therapeutic efficacy.
As a pharmaceutical product, aclidinium bromide is a white, crystalline powder that exhibits poor solubility in water and ethanol.[2] It is formulated as a dry powder for inhalation, delivered via a breath-actuated, multi-dose inhaler known as the Genuair (in Europe) or Pressair (in the US) device.[2] The powder is an adhesive mixture composed of micronized aclidinium bromide and α-lactose monohydrate, which serves as a carrier excipient.[11] The inclusion of lactose is clinically significant, as it may contain trace amounts of milk proteins. This forms the basis for a key contraindication: the drug should not be used in patients with a severe hypersensitivity to milk proteins.[10] The device is designed to be stored at room temperature between 20 and 25 degrees C (68 and 77 degrees F) in its sealed protective pouch and should be discarded 45 days after removal from the pouch or when the dose counter reads zero.[13]
Property | Details |
---|---|
Drug Name | Aclidinium Bromide |
DrugBank ID | DB08897 1 |
Class | Long-Acting Muscarinic Antagonist (LAMA), Anticholinergic, Bronchodilator 1 |
CAS Numbers | 727649-81-2 (cation), 320345-99-1 (bromide salt) 1 |
Molecular Formula | C26H30NO4S2⋅Br 4 |
Molecular Weight | 564.55 g/mol 5 |
Key Brand Names | Tudorza Pressair, Eklira Genuair, Bretaris Genuair, Duaklir Genuair 2 |
Stereochemistry | Pure R-enantiomer 2 |
The therapeutic utility and safety of aclidinium bromide are rooted in its distinct pharmacological properties. A detailed examination of its mechanism of action, pharmacodynamics, and pharmacokinetics reveals a molecule designed for targeted local action in the lungs with minimal systemic impact.
Aclidinium bromide functions as a long-acting, competitive, and reversible antagonist of muscarinic acetylcholine receptors, categorizing it as a LAMA or anticholinergic agent.[1] In patients with Chronic Obstructive Pulmonary Disease (COPD), increased cholinergic tone is a key driver of bronchoconstriction. Aclidinium, delivered via inhalation, acts locally within the airways to block the binding of the neurotransmitter acetylcholine to these receptors on the surface of airway smooth muscle cells.[8] This blockade inhibits the downstream signaling pathways that lead to muscle contraction, resulting in the relaxation of the airway smooth muscle, a widening of the airways (bronchodilation), and consequently, easier breathing for the patient.[16]
While aclidinium exhibits high, subnanomolar affinity for all five human muscarinic receptor subtypes (M1-M5), its clinical profile is not defined by this broad affinity but rather by its differential dissociation kinetics from these subtypes.[4] The therapeutic effect—bronchodilation—is mediated primarily through the antagonism of M3 receptors, which are abundant in the smooth muscle of the bronchioles.[8] Conversely, potential cardiac side effects, such as tachycardia, are primarily associated with the antagonism of M2 receptors, which are prevalent in the heart.[2]
The defining characteristic of aclidinium is its "kinetic selectivity." It demonstrates a significantly longer residence time at M3 receptors compared to M2 receptors.[8] The dissociation half-life of aclidinium from the M3 receptor subtype is approximately 29.2 hours, which is more than six times longer than its dissociation half-life from the M2 subtype.[2] This molecular behavior provides a clear mechanistic basis for its therapeutic profile. The rapid dissociation from cardiac M2 receptors means that any potential for heart rate elevation is transient and diminishes quickly. In contrast, the prolonged binding to airway M3 receptors ensures a sustained, long-lasting bronchodilatory effect. This kinetic profile distinguishes aclidinium from other LAMAs; its M3 dissociation half-life is substantially longer than that of the short-acting ipratropium (0.47 hours) but shorter than that of the once-daily LAMA tiotropium (62.2 hours).[2] This intermediate duration of action at the M3 receptor underpins the clinical findings of sustained efficacy and supports the necessity of a twice-daily dosing regimen to maintain consistent 24-hour receptor blockade and symptom control.[20]
The pharmacodynamic effects of aclidinium are a direct consequence of its mechanism of action. It induces rapid, dose-dependent, and long-lasting bronchodilation.[12] Clinical studies have shown that the prevention of acetylcholine-induced bronchoconstriction lasts for more than 24 hours following inhalation.[2] In COPD patients, maximal bronchodilation, as measured by peak forced expiratory volume in 1 second (FEV1), is typically achieved within 1 to 3 hours after dosing.[12] This rapid onset of action can provide prompt symptom relief.
A cornerstone of aclidinium's pharmacodynamic profile is its cardiovascular safety. The kinetic selectivity for M3 over M2 receptors translates into a favorable cardiac safety profile at clinical doses. In a dedicated study assessing cardiac rhythm via Holter monitor in 336 COPD patients, treatment with aclidinium 400 mcg twice daily for three months produced no clinically significant effects on cardiac rhythm compared to placebo.[8] Furthermore, thorough QT studies have confirmed that aclidinium does not cause a prolongation of the QTc interval.[1] This robust cardiac safety evidence, later confirmed in a large-scale outcomes trial, is a key feature of the drug.
Systemic anticholinergic effects are minimal due to the drug's rapid hydrolysis in plasma upon absorption from the lungs.[12] This local action with limited systemic exposure contributes significantly to its overall tolerability and favorable side-effect profile.
The pharmacokinetic profile of aclidinium is characterized by rapid local absorption, minimal systemic exposure, extensive and rapid metabolism, and elimination primarily as inactive metabolites. This profile is central to its efficacy and safety.
Absorption: Following oral inhalation, aclidinium is rapidly absorbed from the lung, with peak plasma concentrations (Tmax) reached within approximately 5 minutes in healthy subjects and within 10 to 15 minutes in patients with COPD.[2] A 2023 study in healthy Chinese participants confirmed this rapid absorption, reporting a median Tmax of 0.08 hours (about 5 minutes).[22] Despite this rapid absorption from the lung, the absolute systemic bioavailability is extremely low, at less than 5%.[11] This indicates that the vast majority of the drug that enters the systemic circulation is quickly hydrolyzed, preventing significant systemic drug levels and associated side effects.
Distribution: Gamma scintigraphy studies show that the whole lung deposition of aclidinium delivered via the Genuair/Pressair inhaler averages approximately 30% of the metered dose.[2] After intravenous administration, aclidinium exhibits a large volume of distribution of approximately 300 L, suggesting extensive tissue distribution.[8] Due to the rapid hydrolysis of the parent drug in plasma, in vitro protein binding studies primarily reflect the binding of its metabolites. The carboxylic acid metabolite is 87% bound to plasma proteins (mainly albumin), while the alcohol metabolite is 15% bound.[2]
Metabolism: The metabolism of aclidinium is the key to its safety profile. It undergoes rapid and extensive hydrolysis to two primary, pharmacologically inactive metabolites: an alcohol derivative (LAS34823) and a dithienylglycolic acid derivative (LAS34850).[1] This breakdown occurs through both chemical (non-enzymatic) and enzymatic pathways. The primary enzyme responsible for the enzymatic hydrolysis in human plasma is butyrylcholinesterase.[2] The contribution of the cytochrome P450 (CYP) enzyme system to the overall metabolic clearance of aclidinium is minor.[8] In vitro studies have confirmed that at therapeutic concentrations, neither aclidinium nor its major metabolites inhibit or induce major CYP isoenzymes (including 1A2, 2C9, 2D6, 3A4/5) or the P-glycoprotein (P-gp) transporter.[2] This metabolic profile, which largely bypasses the CYP450 system, confers a very low potential for pharmacokinetic drug-drug interactions, a significant clinical advantage for the typically elderly COPD patient population often subject to polypharmacy. The rapid inactivation via hydrolysis is the principal reason for its low systemic exposure and the low incidence of systemic anticholinergic adverse events.
Elimination: Following its rapid hydrolysis, the inactive metabolites of aclidinium are eliminated from the body. After an intravenous dose of radiolabeled aclidinium, approximately 1% is excreted as unchanged drug in the urine. The majority of the dose is eliminated as metabolites, with up to 65% recovered in the urine and up to 33% in the feces.[2] The plasma half-life of the parent aclidinium molecule is extremely short, estimated at around 2.4 minutes, reflecting its rapid hydrolysis.[17] However, the
effective half-life, which reflects the duration of receptor binding and clinical effect, is estimated to be between 5 and 8 hours.[14] More recent, intensive PK sampling in a study of Chinese participants reported a bi-phasic decline with a geometric mean half-life of 13.5 hours after a single dose and 21.4 hours after multiple doses, though the terminal phase may not have been fully characterized.[22] This effective half-life dictates the twice-daily dosing schedule required to maintain therapeutic effect.
Pharmacokinetics in Special Populations: The pharmacokinetic properties of aclidinium are not significantly altered in special populations. No dosage adjustments are required for elderly patients or for patients with renal or hepatic impairment.[8] The low systemic exposure and the primary metabolic pathway of hydrolysis (rather than renal or hepatic clearance) mean that organ dysfunction is unlikely to impact the drug's safety or efficacy. Furthermore, a 2023 study demonstrated that pharmacokinetic parameters in healthy Chinese participants were consistent with those observed in Caucasian populations, suggesting no dose adjustment is needed based on this ethnicity.[22]
Parameter | Value / Description | Source(s) |
---|---|---|
Absorption | ||
Tmax (Inhaled) | ~5 minutes (healthy subjects), ~10-15 minutes (COPD patients) | 2 |
Systemic Bioavailability | <5% | 11 |
Lung Deposition | ~30% of metered dose | 2 |
Distribution | ||
Volume of Distribution (IV) | ~300 L | 8 |
Protein Binding | Primarily metabolites: 87% (acid metabolite), 15% (alcohol metabolite) | 2 |
Metabolism | ||
Primary Pathway | Rapid and extensive hydrolysis (enzymatic and non-enzymatic) | 11 |
Key Enzyme | Butyrylcholinesterase (in plasma) | 2 |
Metabolites | Pharmacologically inactive alcohol and carboxylic acid derivatives | 1 |
CYP450 Interaction | Minimal; low potential for drug-drug interactions | 2 |
Elimination | ||
Excretion Routes | Metabolites: up to 65% in urine, up to 33% in feces | 8 |
Plasma Half-life (Parent) | ~2.4 minutes | 17 |
Effective Half-life | 5-8 hours | 14 |
The clinical development program for aclidinium bromide has robustly established its efficacy as a maintenance therapy for COPD. The evidence from pivotal trials, long-term studies, and investigations into combination therapy demonstrates its ability to improve lung function, reduce symptoms, and enhance patient quality of life.
Aclidinium bromide is formally indicated for the long-term, maintenance treatment of bronchospasm associated with chronic obstructive pulmonary disease (COPD), which includes chronic bronchitis and emphysema.[1] It is classified as a "controller" inhaler, designed for regular, scheduled use to manage the disease, rather than as a "rescue" therapy for the immediate relief of acute bronchospasm or a sudden COPD flare-up.[10] The primary therapeutic goals of aclidinium therapy align with the overall management strategy for COPD: to relieve and reduce the burden of symptoms such as dyspnea (shortness of breath), to improve health status and exercise tolerance, and critically, to decrease the frequency and severity of disease exacerbations.[8]
The regulatory approvals of aclidinium were supported by a comprehensive clinical trial program, with the ACCORD COPD I and ATTAIN studies serving as key pillars of evidence for its efficacy and safety. These trials established the 400 mcg twice-daily (BID) dose as the optimal regimen.
ACCORD COPD I (AClidinium in Chronic Obstructive Respiratory Disease; NCT00891462): This was a 12-week, multicenter, randomized, double-blind, placebo-controlled Phase III study designed to evaluate the efficacy and safety of aclidinium in patients with moderate to severe COPD.[28] The study compared two doses of aclidinium, 200 µg BID and 400 µg BID, against a placebo group.[29] The trial successfully met its primary endpoint. At week 12, the change from baseline in morning pre-dose (trough) FEV1 was statistically significant for both doses compared to placebo. The improvement was 86 mL for the 200 µg dose and a more robust 124 mL for the 400 µg dose, with the latter representing a clinically meaningful improvement in bronchodilation.[21] In terms of safety, aclidinium was well tolerated, with adverse event rates similar to placebo. Notably, the incidence of COPD exacerbations, the most frequent adverse event, was lower in both aclidinium arms (9.2% for 200 µg, 7.4% for 400 µg) compared to the placebo arm (12.4%).[29]
ATTAIN (Aclidinium To Treat Airway obstruction In COPD patieNts; NCT01001494): This was a larger, longer-duration Phase III trial that further solidified the evidence for aclidinium. The 24-week, randomized, double-blind, placebo-controlled study enrolled 828 patients with moderate to severe COPD.[31] Similar to ACCORD I, it compared aclidinium 200 µg BID and 400 µg BID against placebo.[32] The study also met its primary endpoint, demonstrating statistically significant improvements in trough FEV1 at week 24. The improvements versus placebo were 99 mL for the 200 µg dose and 128 mL for the 400 µg dose (p<0.0001 for both).[32] The improvement in peak FEV1 was also substantial and significant, at 185 mL and 209 mL for the 200 µg and 400 µg doses, respectively.[32]
Crucially, the ATTAIN study also demonstrated significant benefits in patient-reported outcomes (PROs). For the approved 400 µg dose, aclidinium produced a statistically significant and clinically meaningful improvement in disease-specific health status, as measured by the St. George's Respiratory Questionnaire (SGRQ), with a mean improvement of -4.6 units over placebo.[32] It also led to a clinically meaningful reduction in breathlessness, with a 1.0 unit improvement in the Transitional Dyspnea Index (TDI) focal score over placebo.[32] These PRO results are vital, as they show that the observed improvements in lung function translate into tangible benefits that patients can feel in their daily lives.
Beyond the initial pivotal trials, the long-term efficacy and safety of aclidinium have been well-established. A 52-week extension of the ACCORD COPD I study confirmed that the improvements in lung function and health status seen in the initial 12 weeks were sustained over a year of continuous treatment.[34] Similarly, long-term safety studies of up to 52 weeks, involving over 890 patients, found no new safety signals and confirmed the tolerability profile seen in shorter trials.[24]
The most definitive evidence for aclidinium's long-term profile comes from the ASCENT-COPD (Aclidinium Bromide on Long‑Term Cardiovascular Safety and COPD Exacerbations in PatieNTs with Moderate to Very Severe COPD; NCT01966344) trial. This landmark Phase IV study was specifically designed to address a critical evidence gap. Pivotal registration trials typically exclude patients with recent or unstable cardiovascular (CV) disease to obtain a clear efficacy signal, yet this patient profile is extremely common in real-world clinical practice, where up to 70% of COPD patients have CV comorbidities.[28] The ASCENT trial was therefore a crucial post-marketing commitment that prospectively evaluated aclidinium in the very population it was intended to treat but who were underrepresented in earlier studies. The large-scale (3630 patients), long-term (up to 3 years) study enrolled patients with moderate-to-very-severe COPD who also had a documented history of CV disease or significant CV risk factors.[35]
The results of ASCENT were transformative for the clinical profile of aclidinium. The study's primary safety objective was met, demonstrating conclusively that aclidinium 400 mcg BID did not increase the risk of major adverse cardiovascular events (MACE) compared to placebo in this high-risk population.[36] This finding moved the drug's CV safety profile from a "lack of negative evidence" to "positive evidence of safety." Concurrently, the study's primary efficacy objective was also met, showing that aclidinium significantly reduced the rate of moderate-to-severe COPD exacerbations and the rate of exacerbation-related hospitalizations during the first year of treatment.[25] This dual message of proven cardiovascular safety and robust exacerbation reduction in a clinically relevant, high-risk population led to a significant update to the FDA label and strongly differentiates aclidinium within the LAMA class.[36]
Modern COPD management guidelines increasingly recommend dual bronchodilation with a LAMA and a LABA for patients who remain symptomatic on monotherapy.[38] The combination of aclidinium with the LABA formoterol leverages two distinct and complementary mechanisms of action to achieve superior bronchodilation.[16] Clinical trials, such as the ACTIVATE study (NCT02424344), have evaluated the fixed-dose combination (FDC) of aclidinium/formoterol (marketed as Duaklir/Brimica).[41] These studies have consistently shown that the FDC provides statistically significant and sustained improvements in lung function that are superior to those achieved with either the individual monotherapy components or placebo.[6] This FDC provides an important therapeutic option for escalating treatment in appropriate patients.
ACCORD COPD I | ATTAIN | |
---|---|---|
NCT Number | NCT00891462 28 | NCT01001494 31 |
Duration | 12 weeks 29 | 24 weeks 32 |
Patient N (Aclidinium 400 mcg vs. Placebo) | 185 vs. 180 29 | 278 vs. 272 32 |
Primary Endpoint | Change in trough FEV1 at Week 12 29 | Change in trough FEV1 at Week 24 32 |
Result (Change in Trough FEV1) | +124 mL vs. placebo (p<0.0001) 21 | +128 mL vs. placebo (p<0.0001) 32 |
Key Secondary Outcome (Change in Peak FEV1) | +192 mL vs. placebo (at Week 12) 21 | +209 mL vs. placebo (at Week 24) 32 |
Key PRO Result | Statistically significant improvements in health status and symptoms 42 | SGRQ: -4.6 units vs. placebo (p<0.0001)TDI: +1.0 units vs. placebo (p<0.001) 32 |
While pivotal trials establish efficacy against placebo, a drug's true clinical value is often determined by its performance relative to other available treatments. A substantial body of evidence from network meta-analyses (NMAs) allows for a nuanced comparison of aclidinium against other leading LAMAs, providing critical information for clinical decision-making and formulary placement.
The therapeutic class of LAMAs for COPD includes several key agents, most notably tiotropium (the first widely available once-daily LAMA), glycopyrronium, and umeclidinium, in addition to aclidinium.[40] Numerous NMAs have been conducted to indirectly compare the relative efficacy of these agents, as large-scale, head-to-head trials between all competitors are not always feasible.[8] These analyses synthesize data from dozens of randomized controlled trials to estimate the comparative effectiveness on key outcomes.
A surface-level interpretation of the NMA data often concludes that all LAMAs demonstrate "comparable efficacy." However, a more detailed analysis reveals important nuances across different clinical endpoints that can help guide therapeutic choice.
Lung Function (FEV1): Across multiple NMAs, aclidinium 400 µg BID has been shown to produce improvements in trough FEV1 that are comparable to those of tiotropium (both 5 µg and 18 µg once-daily formulations) and glycopyrronium 50 µg OD.[44] For example, one NMA found the difference in FEV1 change from baseline between aclidinium and tiotropium 18 µg was a negligible 0.02 L, and between aclidinium and glycopyrronium was 0.00 L.[44] Some analyses have suggested that umeclidinium may offer a small numerical advantage in trough FEV1 improvement over other LAMAs at 12 weeks, though this difference was not always statistically significant and its clinical relevance is debatable.[43] Overall, on the primary measure of bronchodilation, aclidinium is considered to be in the same efficacy tier as its main competitors.
Health Status (SGRQ): Patient-reported outcomes, such as health status, are arguably more important than spirometry for assessing treatment benefit. In this domain, aclidinium performs strongly. One NMA found that aclidinium resulted in a greater improvement in SGRQ total score at 24 weeks compared to tiotropium 5 µg (a difference of -2.44 units), while being comparable to tiotropium 18 µg and glycopyrronium.[44] Another analysis noted that aclidinium and umeclidinium were the two LAMAs whose mean improvement versus placebo consistently met or exceeded the minimal clinically important difference (MCID) of 4 units on the SGRQ scale.[43] This suggests that aclidinium's benefits may be particularly apparent in how patients feel and function in their daily lives.
Dyspnea (TDI): For the outcome of breathlessness, as measured by the Transition Dyspnea Index (TDI), improvements were found to be generally comparable across all major LAMAs, including aclidinium, tiotropium, and glycopyrronium.[44]
Exacerbation Prevention: Preventing exacerbations is a primary goal of COPD management, as they drive disease progression, hospitalizations, and mortality. A large NMA including 27 studies and over 48,000 patients found that while all LAMAs were effective versus placebo in reducing moderate-to-severe exacerbations, a critical finding emerged from a subgroup analysis of trials lasting six months or longer. In this longer-term analysis, aclidinium was associated with the greatest probability of being the most effective therapy for preventing severe exacerbations—those typically requiring hospitalization.[47] This finding, which predated and aligns with the positive exacerbation results from the ASCENT-COPD trial, represents a significant potential differentiator for aclidinium, particularly in patients at high risk for severe events.
The collective evidence suggests that the choice between LAMAs is a multi-factorial decision that goes beyond a simple comparison of average FEV1 improvement. While all are effective bronchodilators, aclidinium's profile, characterized by strong performance on patient-reported outcomes like health status and a potential advantage in preventing the most severe exacerbations, makes it a compelling option. The twice-daily dosing regimen, while potentially posing a greater challenge for adherence than once-daily options, may offer the benefit of more consistent 24-hour symptom control, especially for patients troubled by nocturnal or early morning symptoms, a benefit supported by early phase II data.[20]
Endpoint | Aclidinium vs. Tiotropium | Aclidinium vs. Glycopyrronium | Aclidinium vs. Umeclidinium |
---|---|---|---|
Trough FEV1 | Comparable efficacy to both 5 µg and 18 µg OD doses.44 One NMA showed a non-significant difference of -12.8 mL vs. tiotropium 18 µg at 12 weeks.43 | Comparable efficacy. Difference in change from baseline at 24 weeks was 0.00 L.44 | Umeclidinium showed a non-significant numerical advantage in some analyses. Difference vs. umeclidinium was -35.4 mL at 12 weeks.43 |
Health Status (SGRQ Score) | Favored over tiotropium 5 µg OD (difference of -2.44 units). Comparable to tiotropium 18 µg OD.44 | Comparable efficacy.44 | Comparable efficacy. Both agents met MCID vs. placebo.43 |
Dyspnea (TDI Score) | Comparable efficacy.44 | Comparable efficacy.44 | Comparable efficacy. Both agents met MCID vs. placebo.43 |
Severe Exacerbation Risk | Not directly compared in NMA for this outcome. | Aclidinium associated with the highest probability of being the best therapy in preventing severe exacerbations in studies ≥6 months. Glycopyrronium was associated with the least effective strategy.47 | Not directly compared in NMA for this outcome. |
The safety and tolerability of a maintenance medication for a chronic condition are paramount, particularly in the context of COPD, which predominantly affects an older population with multiple comorbidities. Aclidinium bromide has demonstrated a favorable safety profile across its extensive clinical development program.
In placebo-controlled clinical trials of 3 to 6 months duration, aclidinium was generally well tolerated.[20] The most commonly reported adverse events that occurred with an incidence of 1% or greater and at a higher rate than placebo were headache (6.6% vs. 5.0% for placebo), nasopharyngitis (5.5% vs. 3.9%), and cough (3.0% vs. 2.2%).[2] Other less frequent events included diarrhea, sinusitis, and rhinitis.[2]
A key feature of aclidinium's tolerability is the low incidence of systemic anticholinergic side effects. This is a direct result of its pharmacokinetic profile, specifically its rapid hydrolysis in plasma to inactive metabolites, which limits systemic drug exposure. Consequently, the incidence of events like dry mouth, tachycardia, blurred vision, and urinary retention was low and comparable to that seen with placebo in clinical trials.[2] In a 52-week extension study, dry mouth was reported in only a single patient receiving the 400 µg dose.[34]
Long-term safety has been confirmed in trials lasting up to 3 years. These studies, including the landmark ASCENT-COPD trial, reported no new safety signals and confirmed the adverse event profile observed in the shorter registration studies.[15]
The prescribing information for aclidinium includes several contraindications, warnings, and precautions, many of which are class effects for inhaled anticholinergic agents.
Contraindications: Aclidinium is contraindicated in patients with a history of severe hypersensitivity to milk proteins, due to the lactose monohydrate excipient in the formulation.[14] It is also contraindicated in patients with a known hypersensitivity to aclidinium bromide, atropine, or its derivatives (including ipratropium and tiotropium).[10]
Warnings and Precautions:
Cardiovascular Safety: The cardiovascular safety of aclidinium is a well-documented and distinguishing feature. Preclinical and early clinical studies indicated a low propensity for cardiac effects.[1] This was prospectively and definitively confirmed in the ASCENT-COPD trial, which demonstrated no increased risk of major adverse cardiovascular events (MACE) in a large population of COPD patients with high underlying cardiovascular risk.[35] This robust evidence provides significant reassurance for prescribing aclidinium to the typical, co-morbid COPD patient.
Hepatotoxicity: Aclidinium has not been linked to instances of liver enzyme elevations or clinically apparent acute liver injury.[1] A major contributing factor to its hepatic safety is its very low systemic absorption and exposure following inhalation, minimizing the amount of drug that reaches the liver.[1]
The potential for clinically significant drug-drug interactions with aclidinium is low.
Adverse Reaction | Aclidinium 400 mcg BID (%) | Placebo (%) |
---|---|---|
Headache | 6.6 | 5.0 |
Nasopharyngitis | 5.5 | 3.9 |
Cough | 3.0 | 2.2 |
Diarrhea | 2.7 | 1.4 |
Sinusitis | 1.7 | 0.8 |
Rhinitis | 1.6 | 1.2 |
Toothache | 1.1 | 0.8 |
Fall | 1.1 | 0.5 |
Vomiting | 1.1 | 0.5 |
Data sourced from the TUDORZA PRESSAIR Prescribing Information, reflecting pooled data from 3- and 6-month placebo-controlled trials.[15]
The successful translation of aclidinium from a promising molecule to a globally recognized therapy is reflected in its regulatory approvals and its inclusion in international treatment guidelines. Practical information regarding its administration is crucial for ensuring optimal patient outcomes.
Recommended Dosage: The recommended and approved dose of aclidinium is one oral inhalation of 400 mcg, administered twice daily.[8] To ensure consistent bronchodilation over a 24-hour period, the doses should be taken at approximately the same times each day, once in the morning and once in the evening, about 12 hours apart.[18] Each actuation of the inhaler meters 400 mcg of aclidinium bromide powder. The amount delivered from the mouthpiece is specified as 375 mcg in the US and as 322 mcg of the active aclidinium moiety in the EU.[10]
Inhaler Device Use (Genuair/Pressair): The delivery device is an integral part of the therapeutic system. The Genuair/Pressair is a breath-actuated, multi-dose dry powder inhaler designed to be user-friendly and to provide feedback on correct usage. Proper administration technique is critical for efficacy:
[10]
The design of the inhaler addresses a common point of failure in respiratory therapy: incorrect inhaler technique. The multi-sensory feedback mechanisms—the visual color change of the control window and the auditory "click"—provide immediate and unambiguous confirmation to the patient that the dose has been successfully loaded and inhaled. This user-centric design can improve patient confidence, enhance proper technique, and ultimately improve adherence and real-world effectiveness. Patient satisfaction studies have supported this, finding that patients using the aclidinium device made fewer critical errors affecting drug delivery compared to those using the tiotropium HandiHaler device.[30]
Aclidinium bromide has established itself as an effective, safe, and well-tolerated LAMA for the maintenance treatment of a broad range of patients with COPD. Its clinical profile is defined by several key attributes: consistent and clinically meaningful improvements in lung function and patient-reported outcomes; a twice-daily dosing regimen that provides rapid and sustained 24-hour bronchodilation; and a user-friendly inhaler device with feedback mechanisms.
Its most prominent differentiating features are its robust cardiovascular safety, prospectively proven in the high-risk ASCENT-COPD trial, and its demonstrated ability to reduce the rate of moderate-to-severe exacerbations, including a potential advantage in preventing severe, hospital-worthy events.
Based on this profile, aclidinium is a strong therapeutic choice for many symptomatic COPD patients. It is a particularly compelling option for several specific patient phenotypes:
The primary consideration in its clinical placement is the twice-daily dosing schedule. This should be discussed with the patient, weighing the potential benefits of consistent 24-hour symptom control against the convenience of once-daily alternatives. The intuitive design of the Genuair/Pressair inhaler may help to mitigate potential adherence challenges. Aclidinium is also available as a fixed-dose combination with formoterol, providing a clear and effective pathway for treatment escalation in patients who require dual bronchodilation, in line with current global treatment strategies.
Published at: August 12, 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.