Small Molecule
C22H23N3O3S
925448-93-7
Cerlapirdine (DrugBank ID: DB12229), also known by its developmental codes SAM-531, PF-05212365, and WAY-262,531, is an investigational small molecule that was developed by Wyeth and later Pfizer for the treatment of cognitive disorders, primarily targeting Alzheimer's disease (AD) and schizophrenia.[1] As a selective and potent antagonist of the serotonin 6 (5-HT6) receptor, Cerlapirdine was designed based on the prevailing therapeutic hypothesis that blocking this central nervous system (CNS)-specific receptor could enhance cholinergic and glutamatergic neurotransmission, thereby producing a pro-cognitive effect.[2] The compound progressed through a comprehensive Phase I program and into Phase II clinical trials to evaluate its efficacy and safety in patients with mild-to-moderate AD.[2]
Despite initial reports suggesting the Phase II trial "demonstrated a trend toward efficacy" and a favorable tolerability profile within the study, the overall data package was evidently insufficient to support advancement into pivotal Phase III studies.[1] Subsequent analysis and strategic company decisions reveal that the drug ultimately "failed to show clinical efficacy at any dose".[7] The development of Cerlapirdine was officially discontinued around 2011, marking the end of its clinical investigation.[1]
The discontinuation of Cerlapirdine can be attributed to a confluence of factors. First, the ambiguous and ultimately inadequate efficacy signal failed to justify the substantial investment required for Phase III development. Second, a significant liability existed in its pharmacokinetic and pharmacodynamic profile; its metabolism via the CYP3A4 enzyme and its inherent CNS activity created a high potential for clinically significant drug-drug interactions (DDIs), particularly additive CNS depression, a critical concern for the typically poly-medicated elderly AD population.[10] Finally, the strategic context of the 2009 Pfizer-Wyeth merger likely prompted a rigorous portfolio review, leading to the deprioritization of a high-risk, borderline asset like Cerlapirdine.[2] The story of Cerlapirdine is not an isolated incident but rather a prescient case study, foreshadowing the subsequent late-stage clinical failures of other prominent 5-HT6 receptor antagonists, including Idalopirdine and Latrepirdine, and calling into question the translational validity of the entire therapeutic hypothesis for Alzheimer's disease.[7]
A comprehensive understanding of an investigational drug begins with a precise characterization of its identity and physicochemical properties. These foundational data are crucial for interpreting its pharmacological behavior, pharmacokinetic profile, and formulation development.
Cerlapirdine has been identified and tracked through its development using a variety of names and registry numbers. Establishing this nomenclature is essential for a thorough review of the scientific and clinical literature, particularly as the formal International Nonproprietary Name (INN) is noted to be "PubMed-negative," requiring searches using its developmental codes.[14]
Cerlapirdine is a synthetic organic small molecule belonging to the indazole class. Its molecular architecture was optimized to achieve potent and selective antagonism at the 5-HT6 receptor.
Predicted physicochemical data for the hydrochloride salt form indicate properties consistent with a viable oral drug candidate.[18] It demonstrates compliance with Lipinski's Rule of Five and the Ghose Filter, which are established medicinal chemistry guidelines used to predict oral bioavailability and "drug-likeness." This adherence suggests that the molecule was well-optimized from a chemical perspective, possessing favorable characteristics regarding molecular weight, lipophilicity, and hydrogen bonding potential. The implication of this successful chemical design is that the ultimate failure of Cerlapirdine's development program was not rooted in fundamental physicochemical liabilities. Instead, the focus of inquiry must shift to the compound's biological activity, clinical efficacy, and safety profile to understand the reasons for its discontinuation.
Table 1: Chemical and Physical Properties of Cerlapirdine and its Hydrochloride Salt
| Property | Value (Cerlapirdine Free Base) | Value (Cerlapirdine Hydrochloride) | Source(s) |
|---|---|---|---|
| Generic Name | Cerlapirdine | Cerlapirdine Hydrochloride | [10, 18] |
| CAS Number | 925448-93-7 | 925447-04-7 | [1, 3] |
| DrugBank ID | DB12229 | DBSALT002160 | [10, 18] |
| Chemical Formula | $C_{22}H_{23}N_{3}O_{3}S$ | $C_{22}H_{24}ClN_{3}O_{3}S$ | [10, 18] |
| Average Weight | 409.5 g·mol⁻¹ | 445.96 g·mol⁻¹ | [10, 18] |
| Monoisotopic Mass | 409.146012787 Da | 445.1226905 Da | [10, 18] |
| IUPAC Name | N,N-dimethyl-3-[(3-naphthalen-1-ylsulfonyl-2H-indazol-5-yl)oxy]propan-1-amine | dimethyl(3-{[3-(naphthalene-1-sulfonyl)-1H-indazol-5-yl]oxy}propyl)amine hydrochloride | [1, 18] |
| logP (Predicted) | 3.4 - 3.49 | 3.4 - 3.49 | 18 |
| Water Solubility (Predicted) | Not specified | 0.00476 mg/mL | 18 |
| pKa (Strongest Basic) | Not specified | 7.83 | 18 |
| Rule of Five (Predicted) | Not specified | Yes | 18 |
| Ghose Filter (Predicted) | Not specified | Yes | 18 |
The serotonin 6 (5-HT6) receptor is a G protein-coupled receptor (GPCR) that is coupled to the stimulatory G-protein, Gs, leading to the activation of adenylyl cyclase and an increase in intracellular cyclic AMP (cAMP).[20] A distinguishing feature of the 5-HT6 receptor is its highly restricted expression profile, being found almost exclusively within the CNS.[5] Its distribution is particularly dense in brain regions integral to learning, memory, and executive function, including the cerebral cortex, hippocampus, striatum, and nucleus accumbens.[5]
This specific localization led to significant interest in the 5-HT6 receptor as a therapeutic target for cognitive disorders. The prevailing neurobiological hypothesis posits that 5-HT6 receptors are predominantly located on GABAergic interneurons.[5] By acting as an antagonist at these receptors, a drug like Cerlapirdine is theorized to inhibit the inhibitory tone exerted by these GABAergic neurons on downstream effector neurons. This disinhibition is believed to result in an increased release of key pro-cognitive neurotransmitters, most notably acetylcholine (ACh) and glutamate, in the cortex and hippocampus.[13] Given that Alzheimer's disease is characterized by a profound deficit in the cholinergic system, this mechanism provided a compelling and rational basis for investigating 5-HT6 antagonists as a symptomatic treatment for the disease.[20]
Cerlapirdine was specifically designed and characterized as a selective, potent, and full antagonist of the 5-HT6 receptor.[2] Its classification in drug development databases as an "Antidementia" agent is a direct reflection of this intended mechanism and therapeutic goal.[2] By blocking the 5-HT6 receptor, Cerlapirdine was expected to leverage the neurochemical cascade described above to enhance cognitive function in patients suffering from the neurochemical imbalances associated with AD and schizophrenia, the two primary indications for which it was investigated.[1] Preclinical studies with 5-HT6 antagonists, including compounds structurally related to Cerlapirdine, consistently demonstrated pro-cognitive effects in various animal models of memory impairment, providing strong foundational support for advancing these compounds into human clinical trials.[21]
However, the history of the 5-HT6 antagonist class is a stark illustration of the challenges of translating preclinical findings into clinical reality. While the therapeutic hypothesis was robust and compelling in animal models, it has repeatedly failed to deliver meaningful benefits in human patients with AD. Cerlapirdine's own discontinuation after Phase II was an early sign of this translational disconnect.[1] This was followed by the high-profile Phase III failures of other 5-HT6 antagonists, such as Idalopirdine and Latrepirdine, which also showed promising early-stage data only to fail in large-scale pivotal trials.[12] This pattern of class-wide failure strongly suggests that the neurobiological effects observed in rodent models of cognitive deficit do not adequately represent or translate to the complex, multifactorial pathophysiology of human Alzheimer's disease. The modulation of neurotransmitter levels, while effective in simpler models, appears insufficient to overcome the profound synaptic and neuronal loss that characterizes the human disease.
The disposition of Cerlapirdine in the human body was characterized in a human absorption, distribution, metabolism, and excretion (ADME) study. This study utilized a single oral 5 mg dose of radiolabeled $[(^{14})C]$cerlapirdine and employed accelerator mass spectrometry (AMS) for high-sensitivity detection, enabling a comprehensive profile to be built from a very low radioactive dose.[11]
Following oral administration, Cerlapirdine demonstrated good absorption. The ADME study estimated the extent of absorption to be at least 70% of the administered dose.[11] This indicates efficient passage of the drug from the gastrointestinal tract into the systemic circulation, a prerequisite for an effective oral medication.
Once in the bloodstream, unchanged Cerlapirdine was the predominant drug-related species. Metabolite profiling of pooled plasma samples revealed that the parent drug accounted for 51% of the total radiolabeled ($^{14}$C) exposure.[11] This finding is significant as it suggests that Cerlapirdine itself, rather than its metabolites, is the primary active moiety responsible for exerting the pharmacological effect at the 5-HT6 receptor in the CNS.
Cerlapirdine is subject to hepatic metabolism, primarily through oxidative pathways mediated by the cytochrome P450 (CYP) enzyme system.
The principal metabolic transformation is N-demethylation, which produces the metabolite M1, or desmethylcerlapirdine. This metabolite was detected in plasma, but at levels significantly lower than the parent drug, representing only 9% of the total circulating drug-related exposure.[11] Another metabolite, cerlapirdine-N-oxide (M3), was identified in urine.[11]
Crucially, in vitro reaction phenotyping studies identified the specific CYP isozymes responsible for the formation of M1. These studies showed that the demethylation of Cerlapirdine is mediated primarily by CYP2C8 and CYP3A4.[11] The involvement of CYP3A4 is of particular clinical importance. CYP3A4 is the most abundant and clinically significant drug-metabolizing enzyme in the human liver, responsible for the clearance of approximately half of all marketed drugs. Its activity is notoriously susceptible to inhibition and induction by a wide range of other medications. This pharmacokinetic property directly predicts a high potential for drug-drug interactions (DDIs). Co-administration of Cerlapirdine with a CYP3A4 inhibitor would be expected to decrease its metabolism, leading to elevated plasma concentrations and a corresponding exaggeration of its pharmacodynamic effects, such as CNS depression. The developers were clearly aware of this liability, as evidenced by their decision to conduct a dedicated Phase I clinical trial (NCT00745576) to formally investigate the interaction between Cerlapirdine and verapamil, a well-known inhibitor of CYP3A4.[27] This metabolic profile thus provides a clear mechanistic basis for the extensive DDI warnings associated with the drug.
The human ADME study demonstrated that the elimination of Cerlapirdine and its metabolites was nearly complete following a single oral dose. The primary route of excretion was through the feces, with urinary excretion playing a comparatively minor role.[11] Analysis of the excreta revealed that in feces, unchanged Cerlapirdine was the major component, followed by its primary metabolite, desmethylcerlapirdine. In urine, three main drug-related components were detected: cerlapirdine-N-oxide (M3), unchanged Cerlapirdine, and desmethylcerlapirdine.[11] This pattern indicates that a significant portion of the absorbed drug is eliminated via biliary excretion into the feces, either as the parent compound or after metabolism.
The clinical development of Cerlapirdine was initiated by Wyeth and subsequently managed by Pfizer following its acquisition of Wyeth in 2009.[1] The program was strategically designed to evaluate the drug's potential as a symptomatic treatment for cognitive deficits in two major neuropsychiatric disorders: Alzheimer's disease and schizophrenia.[1] The development pathway progressed logically from initial safety and tolerability studies in healthy volunteers to a proof-of-concept efficacy trial in the target patient population.
A comprehensive series of Phase I trials was conducted to establish the fundamental safety, tolerability, pharmacokinetic (PK), and pharmacodynamic (PD) properties of Cerlapirdine. These studies were performed in diverse populations to support a potential global development plan.
The cornerstone of the Cerlapirdine program was the Phase II trial designed to obtain the first evidence of efficacy in patients. The primary study appears to be NCT00481520, a randomized, double-blind, placebo-controlled trial evaluating multiple ascending fixed doses of Cerlapirdine in subjects with mild-to-moderate Alzheimer's disease over a 28-day period.[29] Another Phase II trial, NCT00895895, was also conducted in AD patients.[2]
The reported outcomes of these trials present a narrative that requires careful interpretation. On one hand, several sources state that the Phase II trial "demonstrated a trend toward efficacy" and was associated with a "good side effect profile and no incidence of serious adverse events".[1] This phrasing is common in pharmaceutical communications for a trial that has failed to meet its prespecified primary endpoint with statistical significance, but may have shown some numerical advantage over placebo or a positive signal on a secondary or exploratory endpoint. It is a way to maintain a degree of optimism about a program or target while acknowledging a technical failure.
Conversely, a more definitive assessment comes from a review of the development history of the 5-HT6 antagonist class. One publication explicitly states that a follow-up compound, SAM-760, was developed by Pfizer precisely because Cerlapirdine (SAM-531) "failed to show clinical efficacy at any dose in phase II trials".[7] This statement reflects the internal, strategic conclusion that drives business decisions. While the external communication may have highlighted a "trend," the internal assessment was one of failure.
The most definitive evidence of a drug's perceived performance is not the language used in abstracts or press releases, but the actions taken by the developing company. Pfizer's decision to halt the development of Cerlapirdine while simultaneously investing in a next-generation 5-HT6 antagonist (SAM-760) is a clear and unambiguous signal. It indicates that while the company may have retained some belief in the therapeutic potential of the target (the 5-HT6 receptor), they concluded that Cerlapirdine itself was not a viable candidate. Therefore, the conflicting reports are best reconciled by concluding that the Phase II trial failed to generate data sufficiently compelling to warrant the high cost and risk of a Phase III program, leading to its termination.
Table 2: Summary of Key Clinical Trials for Cerlapirdine (SAM-531)
| NCT Number | Phase | Study Title / Purpose | Target Population | Status / Outcome | Source(s) |
|---|---|---|---|---|---|
| NCT00479700 | 1 | Safety, Tolerability, PK, and PD of SAM-531 | Healthy Subjects | Completed | 27 |
| NCT00479349 | 1 | Safety, PK, and PD of SAM-531 | Healthy Subjects | Completed | 27 |
| NCT00479297 | 1 | Safety, Tolerability, PK, and PD of SAM-531 | Healthy Japanese Males | Completed | 27 |
| NCT00480818 | 1 | Safety, PK, and PD of SAM-531 | Healthy Young and Elderly Subjects | Completed | 27 |
| NCT00726115 | 1 | Safety, Tolerability, and PK of Single and Multiple Doses of SAM-531 | Not specified | Completed | 27 |
| NCT00745576 | 1 | Potential Interaction Between Verapamil and SAM-531 | Not specified | Completed | 27 |
| NCT00481520 | 2 | Safety, Tolerability, PK, and PD of Multiple Ascending Doses of SAM-531 | Mild to Moderate Alzheimer's Disease | Terminated/Completed (Development Discontinued) | [22, 30] |
| NCT00895895 | 2 | Study in Alzheimer's Disease | Alzheimer's Disease | Completed (Enrollment) / Development Discontinued | 2 |
While the Phase II trial reported a generally good side effect profile in a controlled setting, a broader analysis of Cerlapirdine's pharmacology reveals a significant and pervasive safety concern: the potential for drug-drug interactions leading to additive Central Nervous System (CNS) depression.[1] The DrugBank database provides an extensive list of potential pharmacodynamic interactions where the concomitant use of Cerlapirdine with other CNS-active agents could increase the risk or severity of adverse effects such as sedation, somnolence, dizziness, and cognitive impairment.[10]
The list of interacting drug classes is exceptionally broad and includes many medications commonly prescribed to the elderly. Key examples include:
The clinical implications of this extensive DDI profile are profound and severely compromise the potential risk-benefit profile of Cerlapirdine. The target population for an Alzheimer's disease therapeutic is, by definition, elderly. This demographic has a high prevalence of comorbidities such as anxiety, depression, insomnia, chronic pain, and behavioral disturbances, which are frequently managed with medications from the very classes listed above. Polypharmacy is the norm, not the exception, in this patient group.
The primary therapeutic goal of Cerlapirdine was to improve cognition and function. However, the most predictable adverse outcome in a real-world setting would be the exacerbation of CNS depression. The resulting sedation, dizziness, confusion, and somnolence are not merely uncomfortable side effects; they are directly antithetical to the therapeutic goal. An increase in these symptoms would lead to a decline in daily functioning, worsen cognitive measures, and significantly increase the risk of falls, which are a major cause of morbidity and mortality in the elderly. Therefore, even if Cerlapirdine possessed a modest intrinsic pro-cognitive effect, this benefit would likely be negated or entirely outweighed by the functional impairment caused by its unavoidable interactions in a typical, poly-medicated AD patient. This creates a fundamentally unfavorable risk-benefit equation, representing a critical, and likely insurmountable, safety hurdle for the drug's development.
The clinical development of Cerlapirdine was formally discontinued, with no further development activity reported since 2011.[1] The cessation of the program can be understood as the result of a "triad of failure," a convergence of insufficient efficacy, a challenging safety profile, and unfavorable strategic business factors.
The story of Cerlapirdine's failure was not an isolated event but rather a harbinger of a class-wide disappointment. The therapeutic hypothesis that 5-HT6 receptor antagonism could provide meaningful cognitive benefits in AD was tested by several other companies with different chemical entities, all of which ultimately failed in late-stage clinical trials. This pattern of repeated failure points not to the flaws of a single molecule, but to a fundamental problem with the underlying therapeutic strategy in the context of human AD.
Table 3: Comparative Profile of 5-HT6 Receptor Antagonists in Alzheimer's Disease
| Drug Name | Developer(s) | Mechanism of Action | Highest Phase Reached (AD) | Key Trial Outcome (AD) | Status |
|---|---|---|---|---|---|
| Cerlapirdine | Wyeth / Pfizer | 5-HT6 Receptor Antagonist | Phase 2 | Failed to show sufficient clinical efficacy | Discontinued |
| Idalopirdine | Lundbeck | 5-HT6 Receptor Antagonist | Phase 3 | Failed to meet primary endpoints in three pivotal trials | Discontinued |
| Latrepirdine | Medivation / Pfizer | Mixed (incl. 5-HT6 Antagonism) | Phase 3 | Failed to meet primary endpoints in multiple pivotal trials | Discontinued |
The development and subsequent discontinuation of Cerlapirdine, viewed within the broader context of the failures of Idalopirdine and Latrepirdine, offers critical lessons for CNS drug development. It serves as a powerful cautionary tale about the profound challenges of translating a compelling preclinical therapeutic hypothesis into a clinically effective medicine for Alzheimer's disease. The consistent failure of this class suggests that simply modulating neurotransmitter systems is likely insufficient to overcome the complex and overwhelming pathology of synaptic loss and neurodegeneration that drives cognitive decline in AD.
Furthermore, the case of Cerlapirdine underscores the vital importance of considering the real-world clinical context from the earliest stages of development. A drug's interaction profile and its suitability for a poly-medicated target population are not secondary considerations but are fundamental to its viability. The unfavorable DDI profile of Cerlapirdine created a risk-benefit imbalance that was likely as instrumental in its termination as its lukewarm efficacy.
While the 5-HT6 receptor remains a validated and intriguing CNS target for research, its pursuit as a primary mechanism for cognitive enhancement in Alzheimer's disease has been largely abandoned. The collective failures of this class have forced the field to look beyond simple neurotransmitter modulation and focus on more fundamental disease-modifying pathways. The story of Cerlapirdine is a valuable, if disappointing, chapter in the long and difficult search for effective treatments for Alzheimer's disease.
Published at: November 1, 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.