MedPath

PF-04995274 Advanced Drug Monograph

Published:Nov 3, 2025

Generic Name

PF-04995274

Drug Type

Small Molecule

Chemical Formula

C23H32N2O6

CAS Number

1331782-27-4

A Comprehensive Pharmacological and Clinical Profile of PF-04995274: An Investigational 5-HT4 Receptor Partial Agonist

Executive Summary

PF-04995274 is an investigational small molecule developed by Pfizer as a potent, high-affinity, and selective partial agonist of the serotonin 4 receptor (5-HT4R). The compound was initially advanced as a potential pro-cognitive agent for the treatment of Alzheimer's disease (AD), with subsequent exploration into its utility for major depressive disorder (MDD). The therapeutic rationale was compelling, grounded in the 5-HT4R's role in modulating cholinergic neurotransmission, promoting the non-amyloidogenic processing of amyloid precursor protein (APP), and facilitating synaptic plasticity in key brain regions like the hippocampus.

Preclinical studies provided robust proof-of-concept, demonstrating that PF-04995274 could reverse cognitive deficits in animal models of AD and exhibit prophylactic, antidepressant-like effects in models of stress and depression. The compound possessed favorable physicochemical properties for an orally active, brain-penetrant CNS drug, meeting key criteria for drug-likeness and showing good potential for bioavailability.

Despite this promising preclinical profile, the clinical development program encountered significant challenges that ultimately led to its discontinuation. While Phase 1 studies in healthy volunteers established a generally acceptable safety and tolerability profile, a key safety concern emerged regarding a "high impact on plasma aldosterone," a liability that was actively monitored in early clinical trials. The pivotal pro-cognitive study, a scopolamine-induced deficit challenge in healthy volunteers, was terminated, and systems pharmacology modeling suggested that the compound's low intrinsic activity as a partial agonist was insufficient to overcome the cholinergic blockade, a prediction that was reportedly borne out by the trial's outcome.

Exploratory studies in MDD revealed a novel but complex mechanism of action. Unlike the selective serotonin reuptake inhibitor (SSRI) citalopram, PF-04995274 did not correct negative emotional biases via amygdala modulation. Instead, it increased activity in the medial-frontal cortex, suggesting a "top-down" cognitive control mechanism. Although preliminary evidence of clinical improvement was observed, this divergent and less-validated mechanism presented a higher-risk path forward.

The development of PF-04995274 was officially discontinued for both AD and depression. Its trajectory serves as an instructive case study in CNS drug development, highlighting the profound translational gap between promising preclinical models and the complexities of human pathophysiology. The program underscores the critical importance of intrinsic agonist activity for clinical efficacy and reveals potential safety liabilities, such as hormonal dysregulation, that must be carefully managed. The scientific insights gained, particularly regarding the distinct neural mechanisms of 5-HT4R agonism in depression, continue to inform the ongoing exploration of this receptor as a valid therapeutic target for CNS disorders.

Introduction: The Therapeutic Rationale for 5-HT4 Receptor Agonism in CNS Disorders

The development of PF-04995274 was predicated on a strong scientific foundation targeting the serotonin 4 receptor (5-HT4R), a G-protein coupled receptor with a strategic distribution and functional role in brain circuits implicated in both cognition and mood regulation. Its exploration for Alzheimer's disease and major depressive disorder was a direct response to the significant unmet medical needs and the limitations of existing therapeutic paradigms in these complex neurological and psychiatric conditions.

The Role of the 5-HT4 Receptor in Cognition and Mood

The 5-HT4 receptor is a post-synaptic, Gs-protein coupled receptor expressed throughout the central and enteric nervous systems.[1] In the brain, its expression is particularly dense in regions critical for learning, memory, and emotional processing, including the hippocampus (especially the CA1 region), cortex, basal ganglia (caudate, putamen, nucleus accumbens), and limbic areas like the amygdala.[2] This anatomical distribution provides a strong basis for its involvement in higher-order cognitive and affective functions.

Activation of the 5-HT4R by an agonist initiates a well-defined intracellular signaling cascade. The receptor's coupling to the Gs alpha subunit stimulates adenylate cyclase, leading to the synthesis of cyclic adenosine monophosphate (cAMP).[5] This increase in cAMP, a crucial second messenger, subsequently activates Protein Kinase A (PKA), which in turn phosphorylates the cAMP response element-binding protein (CREB). Phosphorylated CREB acts as a transcription factor, promoting the expression of genes involved in neurogenesis, synaptic plasticity, and cell survival.[4]

This molecular pathway translates into two key physiological effects relevant to CNS disorders:

  1. Pro-Cognitive and Cholinergic Enhancement: 5-HT4R agonists have been shown preclinically to enhance the release of the neurotransmitter acetylcholine (ACh).[1] Given that cognitive impairments in AD are partly due to a profound loss of cholinergic neurons and reduced ACh levels, direct modulation of this system represents a primary therapeutic strategy. By augmenting cholinergic function, 5-HT4R agonists have the potential to provide symptomatic cognitive improvement.[7]
  2. Disease-Modifying Potential in Alzheimer's Disease: Beyond symptomatic relief, 5-HT4R activation offers a potential disease-modifying mechanism. The receptor has been shown to stimulate the non-amyloidogenic α-secretase pathway for processing the Amyloid Precursor Protein (APP). This shifts APP cleavage away from the β-secretase pathway, which produces the toxic amyloid-beta (Aβ) peptide, and toward the production of the soluble, neuroprotective sAPPα fragment.[6] This dual action—enhancing cognition while potentially reducing the Aβ burden—made the 5-HT4R an exceptionally attractive target for AD drug development.[6]

Unmet Needs in Alzheimer's Disease and Major Depressive Disorder

The pursuit of PF-04995274 was driven by the significant therapeutic gaps in its target indications. In Alzheimer's disease, the standard of care has long been dominated by symptomatic treatments like acetylcholinesterase inhibitors (e.g., donepezil) and NMDA receptor antagonists (e.g., memantine), which offer modest and temporary benefits.[7] The landscape of disease-modifying therapies has been marked by decades of high-profile clinical failures, underscoring the desperate need for novel mechanisms that can alter the course of the disease.[6] PF-04995274, with its potential to both improve symptoms and modify pathology, represented a promising new approach.

In major depressive disorder, standard first-line treatments such as SSRIs are hampered by several critical limitations. A significant proportion of patients fail to achieve an adequate response, and those who do respond often face a delayed onset of therapeutic effect, typically taking several weeks.[9] Furthermore, residual cognitive symptoms, such as deficits in memory and executive function, are common in MDD and are not well-addressed by current antidepressants.[2] Preclinical studies consistently showed that 5-HT4R agonists produce antidepressant-like effects with a much more rapid onset than SSRIs, suggesting they could offer a faster-acting alternative or a valuable augmentation strategy to accelerate treatment response.[2]

Compound Identification and Physicochemical Properties

PF-04995274 is a synthetic organic small molecule belonging to the benzisoxazole class of compounds, which are characterized by a benzene ring fused to an isoxazole ring.[5] Its precise chemical identity is established through a variety of nomenclature systems and unique identifiers assigned by regulatory and scientific bodies.

Nomenclature and Structural Classification

The compound is most commonly referred to by its Pfizer development code, PF-04995274. It is also known by several synonyms, including PF 04995274, PF04995274, and "compound 2d" in early medicinal chemistry literature.[1]

Its formal chemical structure is defined by the IUPAC name: 4-oxy-1,2-benzoxazol-3-yl]oxymethyl]piperidin-1-yl]methyl]oxan-4-ol.[12] This name precisely describes the arrangement of its five-ring aromatic heteropolycyclic framework.[5]

The molecule is unambiguously cataloged in major chemical and drug databases with the following key identifiers:

  • DrugBank Accession Number: DB12675 [5]
  • CAS Number: 1331782-27-4 [12]
  • UNII (Unique Ingredient Identifier): XI179PG9LV [12]
  • ChEMBL ID: CHEMBL2152922 [12]

The molecular formula of PF-04995274 is $C_{23}H_{32}N_{2}O_{6}$, with an average molecular weight of approximately 432.5 g/mol and a monoisotopic mass of 432.226036758 Da.[1]

Physicochemical Characteristics and Drug-Likeness Assessment

The suitability of a compound for oral administration and central nervous system activity is heavily influenced by its physicochemical properties. Computational models provide key predictions about a molecule's absorption, distribution, metabolism, and excretion (ADME) profile. An analysis of these properties for PF-04995274 reveals a profile largely consistent with a viable CNS drug candidate.

Table 3.1 provides a consolidated summary of the key identifiers and computed physicochemical properties for PF-04995274.

Table 3.1: Summary of Identifiers and Physicochemical Properties of PF-04995274

PropertyValueSource(s)
Identifiers
Generic NamePF-049952745
DrugBank IDDB126755
CAS Number1331782-27-412
UNIIXI179PG9LV12
IUPAC Name4-oxy-1,2-benzoxazol-3-yl]oxymethyl]piperidin-1-yl]methyl]oxan-4-ol[12, 13]
Molecular Properties
Molecular Formula$C_{23}H_{32}N_{2}O_{6}$12
Average Weight432.517 g/mol5
Monoisotopic Weight432.226036758 Da5
Physicochemical Properties
Water Solubility0.205 mg/mL5
logP (Octanol-Water Partition)2.04 (ALOGPS), 1.22 (Chemaxon)5
logS (Aqueous Solubility)-3.35
pKa (Strongest Acidic)14.265
pKa (Strongest Basic)9.595
Physiological Charge15
Hydrogen Acceptor Count75
Hydrogen Donor Count15
Polar Surface Area (PSA)86.42 $Å^{2}$5
Rotatable Bond Count75
Number of Rings55
Drug-Likeness Rules
Rule of Five ComplianceYes5
Ghose Filter ComplianceYes5
Veber's Rule ComplianceNo (per source, but data suggests Yes)5
MDDR-like Rule ComplianceYes5

An expert analysis of these properties indicates a strong profile for oral bioavailability and CNS penetration. The compound's compliance with Lipinski's Rule of Five and the Ghose Filter is a positive prognostic indicator for good absorption and distribution characteristics.[5] The predicted bioavailability is high, and the logP values, ranging from 1.22 to 2.04, strike a favorable balance between aqueous solubility and lipid membrane permeability, a critical attribute for crossing the blood-brain barrier (BBB).[5]

A noteworthy detail arises from the assessment of Veber's Rule, which predicts good oral bioavailability for compounds with a Polar Surface Area (PSA) of $\leq 140 Å^{2}$ and $\leq 10$ rotatable bonds. The available data for PF-04995274 list a PSA of 86.42 $Å^{2}$ and a rotatable bond count of 7.[5] Both of these values fall comfortably within the required parameters. Despite this, the source data from DrugBank concludes that the compound does not comply with Veber's Rule.[5] This conclusion appears to be inconsistent with the underlying data. A more accurate interpretation is that PF-04995274 does in fact satisfy the criteria of Veber's Rule, further strengthening its predicted profile as a well-absorbed oral drug. This refined analysis suggests the molecule's fundamental properties are even more favorable than a cursory reading of the source data might imply.

Nonclinical Pharmacology and Mechanism of Action

The pharmacological profile of PF-04995274 is defined by its specific and potent interaction with the 5-HT4 receptor. Its mechanism as a partial agonist is a central feature that dictates both its therapeutic potential and, as later analysis would suggest, its clinical limitations.

Primary Target Engagement: A High-Affinity 5-HT4R Partial Agonist

PF-04995274 is a selective, high-affinity partial agonist of the 5-HT4 receptor.[1] As an agonist, it binds to and activates the receptor, mimicking the effect of the endogenous neurotransmitter, serotonin.[5] The designation "partial agonist" is critical; it signifies that the compound activates the receptor but produces a submaximal response compared to a full agonist, regardless of concentration.[1] This property can be advantageous, as it may reduce the risk of receptor overstimulation and associated side effects, theoretically creating a wider therapeutic window.[1]

In Vitro Potency and Receptor Subtype Selectivity

The potency of PF-04995274 has been quantified through in vitro assays that measure both its binding affinity (Ki) and its functional potency (EC50) across various 5-HT4R subtypes in both human and rat cell lines. These studies confirm that the compound interacts with its target at nanomolar concentrations, classifying it as a highly potent molecule.

Table 4.1 summarizes the key in vitro pharmacological data for PF-04995274, demonstrating its high affinity and functional potency across multiple receptor isoforms.

Table 4.1: In Vitro Receptor Binding Affinity ($K_{i}$) and Functional Potency ($EC_{50}$) of PF-04995274

ParameterReceptor SubtypeSpeciesValue (nM)Source(s)
Binding Affinity ($K_{i}$)5-HT4AHuman0.3617
5-HT4BHuman0.4617
5-HT4DHuman0.1517
5-HT4EHuman0.3217
5-HT4SRat0.30[18, 19]
Functional Potency ($EC_{50}$)5-HT4AHuman0.47[1, 17]
5-HT4BHuman0.36[1, 17]
5-HT4DHuman0.37[1, 17]
5-HT4EHuman0.26[1, 17]
5-HT4SRat0.5918
5-HT4LRat0.6518
5-HT4ERat0.62[20]

The data show consistent, high-potency engagement with all tested human 5-HT4R subtypes, with $K_{i}$ and $EC_{50}$ values in the sub-nanomolar range. The potency is similarly high at rat receptor subtypes, providing a strong basis for the translation of findings from preclinical rodent models.[18]

Downstream Signaling and Functional Consequences

As a Gs-protein coupled receptor, the 5-HT4R's primary signaling mechanism involves the activation of adenylate cyclase and the subsequent production of intracellular cAMP.[5] This mechanism was experimentally confirmed for PF-04995274 in cellular assays. The compound was shown to dose-dependently increase cAMP levels in HEK293 and CHO cells engineered to express human 5-HT4R subtypes, providing direct evidence of its functional agonist activity at the molecular level.[17] This cAMP-mediated signaling is believed to underpin the receptor's role in promoting synaptic plasticity and its pro-cognitive effects.[3]

The partial agonism of PF-04995274, while theoretically a safety advantage, proved to be a critical factor in its clinical performance. The development program for AD included a human scopolamine challenge trial (NCT01345864), a standard paradigm to test pro-cognitive drugs.[3] Preclinical studies in rats had shown that PF-04995274 could reverse scopolamine-induced deficits, providing the rationale for this clinical test.[3] However, a sophisticated quantitative systems pharmacology (QSP) model was used to prospectively simulate the outcome of this human trial.[3] The model's output was stark: it projected that while a 5-HT4R agonist with high intrinsic activity (a full agonist) would likely show a beneficial effect, an agonist with low intrinsic activity—such as the partial agonist PF-04995274—would likely have no effect or could even worsen the cognitive impairment.[3] The subsequent outcome of the human trial reportedly supported the model's prediction, indicating a clinical failure.[3]

This sequence of events reveals a crucial translational insight. The preclinical animal models, while positive, were not sufficiently sensitive to distinguish the nuanced difference in functional effect between a partial and a full agonist in a manner that was predictive of the human response. The QSP model, by integrating human pharmacology data, correctly identified the compound's limited intrinsic activity as its Achilles' heel. This suggests that for overcoming a significant neurochemical challenge like a scopolamine-induced cholinergic blockade, the degree of maximal receptor activation (efficacy) is a more critical determinant of clinical success than simple receptor binding (affinity) or potency ($EC_{50}$). The failure of PF-04995274 in this context is a direct consequence of its fundamental pharmacology as a partial agonist.

Preclinical Efficacy and Proof-of-Concept

Before advancing to human trials, PF-04995274 was evaluated in a series of nonclinical animal models designed to provide in vivo evidence of its intended therapeutic effects. These studies successfully generated proof-of-concept data supporting its development for both Alzheimer's disease and depression.

Pro-Cognitive Effects in Animal Models

The primary rationale for developing PF-04995274 for AD was its potential to enhance cognitive function. This was tested in the scopolamine-induced deficit model in rats, a widely used preclinical paradigm that mimics the cholinergic deficits seen in AD.[3] In the Morris water maze, a test of spatial learning and memory, scopolamine administration typically impairs performance, causing animals to take longer and travel a greater distance to find a hidden platform. Treatment with PF-04995274 at a dose of 0.032 mg/kg was shown to significantly attenuate this deficit, decreasing the scopolamine-induced increase in distance traveled.[17] This result provided the key preclinical evidence that PF-04995274 could act as a pro-cognitive agent in vivo, justifying its progression into clinical trials for AD.[3]

Prophylactic Efficacy in Models of Stress and Depression

The therapeutic hypothesis was later expanded to include depression, based on the role of the 5-HT4 receptor in mood regulation and stress resilience. Preclinical studies in mice were conducted to assess the compound's potential antidepressant-like and anxiolytic effects.[1] These experiments utilized a prophylactic design, where the drug was administered prior to a stressor to evaluate its ability to enhance stress resilience.

In a contextual fear conditioning paradigm, which models aspects of trauma- and stressor-related disorders, a single injection of PF-04995274 (10 mg/kg) administered five days before the conditioning event significantly attenuated the expression of learned fear.[1] Furthermore, in the forced swim test, a common screening model for antidepressant efficacy, pretreatment with PF-04995274 was found to decrease stress-induced depressive-like behavior.[1] Together, these findings indicated that PF-04995274 not only had antidepressant-like properties but could also act prophylactically to buffer the negative behavioral consequences of stress, providing a strong rationale for its clinical investigation in MDD.[22]

Pharmacokinetic and Safety Profile

A thorough understanding of a drug's pharmacokinetic properties—its absorption, distribution, metabolism, and excretion (ADME)—and its safety profile is paramount for successful clinical development. For PF-04995274, the available data indicate a profile suitable for a CNS therapeutic, though a specific safety concern regarding aldosterone was identified early in development.

Absorption, Distribution, Metabolism, and Excretion (ADME) Profile

PF-04995274 was designed and confirmed to be an orally active and brain-penetrant compound, two essential characteristics for a drug intended to treat CNS disorders like AD and depression.[1] Its physicochemical properties, including a predicted high bioavailability and an optimal logP value, are consistent with these features.[5]

Human pharmacokinetic data were collected in several Phase 1 trials. These studies were designed to measure key PK parameters, including maximum plasma concentration ($C_{max}$), time to maximum concentration ($T_{max}$), area under the concentration-time curve (AUC), and elimination half-life ($t_{1/2}$).[26] Crucially, these parameters were assessed not only in plasma but also in cerebrospinal fluid (CSF), providing direct evidence of the drug's ability to cross the blood-brain barrier and achieve exposure at its site of action in the CNS.[27] One publication noted that the compound has a long half-life in humans, which would be favorable for once-daily dosing regimens.[29]

Metabolism studies identified at least one significant metabolite, designated PF-05082547. The importance of this metabolite is underscored by the fact that its concentration was also systematically measured in both plasma and CSF alongside the parent drug in clinical trials, indicating it may contribute to the overall pharmacological or safety profile of the compound.[26] While comprehensive human ADME studies detailing all metabolic pathways and routes of excretion are not publicly available, the consistent tracking of this metabolite points to its relevance in the drug's disposition.

Preclinical and Clinical Safety and Tolerability

The overall safety profile of PF-04995274 was found to be generally acceptable throughout its development. Preclinical toxicology studies did not reveal any major safety concerns that would have prevented its advancement into human trials.[7]

In Phase 1 clinical studies involving single and multiple ascending doses in healthy young and elderly volunteers, PF-04995274 was reported to be well tolerated.[29] This favorable safety profile was largely maintained in studies involving patients with depression. The RESTAND study (NCT03516604), which administered 15 mg daily for 7 days, reported no major adverse events.[9] The most common side effect was fatigue, which was reported significantly more often in the PF-04995274 group compared to placebo or citalopram; however, all instances were described as mild-to-moderate in severity.[10] One participant in the drug group withdrew due to an adverse event, which resolved within 24 hours of discontinuing the medication.[10] According to the Globally Harmonized System (GHS) of Classification and Labelling of Chemicals, the compound is classified as a potential skin, eye, and respiratory irritant, which is a standard handling precaution for the chemical substance rather than an indicator of systemic toxicity.[12]

Analysis of a Key Safety Concern: Impact on Plasma Aldosterone

Despite the generally benign safety profile, a specific and noteworthy safety signal was identified during the drug's development. An abstract summarizing early clinical findings stated that while PF-04995274 was well tolerated, "a high impact on plasma aldosterone is an issue of concern".[29] Aldosterone is a mineralocorticoid hormone that plays a central role in regulating blood pressure and electrolyte balance. Significant drug-induced elevation of aldosterone could pose risks, including hypertension, hypokalemia, and other cardiovascular complications, which would be a major liability for a drug intended for chronic administration.

This was not an incidental finding but a pre-identified risk that was proactively investigated by the sponsor, Pfizer. The clinical trial protocol for NCT01169714, a multiple-dose study in healthy adult and elderly subjects, explicitly lists "Aldosterone concentration in healthy adult subjects" as a key pharmacodynamic endpoint to be measured over the 14-day dosing period.[26] The inclusion of a specific biomarker for a potential off-target hormonal effect as a formal endpoint in a Phase 1 study is highly indicative. It demonstrates that the development team had a preclinical signal or a strong mechanistic hypothesis to suspect this interaction and designed the study to quantify the risk in humans. While the specific results of these aldosterone measurements are not available in the provided documentation, the existence of this concern adds a significant layer of complexity to the drug's safety assessment and may have contributed to the overall risk-benefit calculation that led to its eventual discontinuation.

Clinical Development Program and Trial Analysis

The clinical development of PF-04995274 was a multi-faceted program that initially focused on Alzheimer's disease before pivoting to explore its potential in major depressive disorder. The program, which did not advance beyond Phase 1, comprised a series of studies designed to evaluate the drug's safety, pharmacokinetics, target engagement, and preliminary efficacy in both healthy volunteers and patient populations.

Overview of the Clinical Strategy and Investigational Indications

The initial and primary therapeutic target for PF-04995274 was Alzheimer's disease. The clinical strategy was twofold: to assess its potential for symptomatic cognitive improvement, leveraging its pro-cholinergic effects, and to investigate its disease-modifying potential by measuring its impact on central biomarkers of APP processing, namely sAPPα and Aβ fragments in the CSF.[1]

Following the generation of supportive preclinical data in animal models of stress, the program was expanded to investigate Major Depressive Disorder. This exploration was also multifaceted, targeting both unmedicated patients to assess its primary antidepressant effects and treatment-resistant patients to evaluate its utility as an adjunctive therapy.[35] Across all indications, the compound remained in the investigational, Phase 1 stage of development.[12]

Detailed Review of Phase 1 Studies

A comprehensive overview of the known clinical trials for PF-04995274 is essential to understand the scope and progression of its human testing. Table 7.1 summarizes the key details of these studies.

Table 7.1: Summary of Key Clinical Trials for PF-04995274

NCT IdentifierTitle / PurposePhaseStatusPopulationKey InterventionsKey Outcome MeasuresSource(s)
NCT01091272First-in-human, single ascending dose (SAD) study1CompletedHealthy AdultsSingle oral doses (0.15 mg to 210 mg) vs. PlaceboSafety, Tolerability, PK (plasma)[28]
NCT01169714Multiple ascending dose (MAD) study1CompletedHealthy Adults & ElderlyMultiple oral doses (0.1 mg to 15 mg daily for 14 days) vs. PlaceboSafety, Tolerability, PK (plasma), PD (Aldosterone)[26, 30, 31]
NCT01193062AD biomarker study1CompletedHealthy AdultsSingle oral doses (0.1 mg to 40 mg) vs. PlaceboSafety, PK (plasma & CSF), PD (CSF sAPPα & Aβ)5
NCT01345864Scopolamine challenge study for AD1TerminatedHealthy AdultsPF-04995274 +/- Donepezil vs. Placebo after scopolamineEffects on scopolamine-induced cognitive deficits[21, 32]
NCT03516604RESTAND Study1CompletedUnmedicated MDD Patients15 mg PF-04995274 vs. 20 mg Citalopram vs. Placebo for 7 daysEmotional processing (behavioral & fMRI), Depression symptoms[9, 32, 35]
NCT03515733RESTART Study1CompletedTreatment-Resistant MDD Patients15 mg PF-04995274 vs. Placebo (adjunctive) for 7-9 daysEmotional & non-emotional cognition, Depression symptoms[32, 35, 36, 39]

Synthesis and Interpretation of Clinical Outcomes

A synthesis of the findings from these trials reveals a complex picture of a compound with acceptable safety but challenging efficacy and pharmacodynamic profiles.

Safety and Pharmacokinetics: The foundational Phase 1 studies (NCT01091272, NCT01169714) successfully established the safety and tolerability of single and multiple doses of PF-04995274 in both young and elderly healthy volunteers. These studies also provided the essential pharmacokinetic characterization in plasma, confirming its suitability for further development.[26]

Alzheimer's Disease Program: The AD program hinged on two key studies. The biomarker study (NCT01193062) was designed to provide the first human evidence of central target engagement by measuring the drug's effect on sAPPα and Aβ in the CSF.[5] While the results of this study have not been publicly detailed, the fate of the AD program was likely sealed by the outcome of the scopolamine challenge study (NCT01345864). This trial was terminated prematurely, and as discussed previously, QSP modeling strongly suggests it failed to demonstrate the expected pro-cognitive effect.[3] This failure to translate a positive preclinical signal in a core human pharmacology model represented a major setback and likely undermined confidence in the AD indication.

Depression Program (RESTAND & RESTART): The later-stage exploration in depression, particularly the RESTAND study (NCT03516604) in unmedicated patients, produced the most scientifically intriguing results. The study found that after one week of treatment, PF-04995274, like the active comparator citalopram, produced a statistically significant reduction in observer-rated depression scores compared to placebo. PF-04995274 also reduced self-reported depression, anxiety, and negative affect.[9] However, the underlying neural mechanisms were starkly different. Citalopram produced the expected SSRI effect: it reduced the recognition of negative facial expressions and attenuated the response of the amygdala to emotional faces.[9] In contrast, PF-04995274 had no effect on this negative emotional bias and did not modulate amygdala activity. Instead, it was associated with a distinct increase in activation across the medial-frontal cortex during the emotional processing task.[9]

This divergence in neural effects points to fundamentally different mechanisms of antidepressant action. The effect of citalopram can be characterized as a "bottom-up" mechanism, where the drug blunts the initial, automatic processing of negative emotional information in core limbic structures like the amygdala. The effect of PF-04995274, however, suggests a "top-down" mechanism. The medial-frontal cortex is a key node in the brain's cognitive control and emotion regulation networks. The increased activation in this region suggests that PF-04995274 may not be reducing the initial negative emotional signal but rather enhancing the brain's cognitive capacity to regulate and control the response to that signal. While this presents a fascinating and novel pro-cognitive approach to treating depression, it also represents a departure from the well-understood and validated mechanism of SSRIs. This novel, unproven mechanism, combined with the earlier efficacy failures in the AD program, would have represented a significantly higher-risk development path for Pfizer.

Developmental Trajectory and Expert Perspective

The journey of PF-04995274 from a promising preclinical candidate to a discontinued asset provides a valuable lens through which to view the challenges of CNS drug development. Its history within the Pfizer pipeline, the reasons for its discontinuation, and the lessons learned offer critical insights for the future of 5-HT4R agonists and the broader field of neuroscience therapeutics.

History within the Pfizer Pipeline and Eventual Discontinuation

PF-04995274 was an active program in Pfizer's CNS pipeline by at least late 2010. The company's public pipeline report from September 27, 2010, lists PF-04995274 in Phase 1 for Alzheimer's Disease.[40] It remained in Phase 1 for AD in subsequent pipeline updates through February and May of 2011.[41]

A pivotal moment in the drug's trajectory occurred on October 12, 2011, with the termination of the scopolamine challenge study (NCT01345864).[21] The official reason cited was "changes in sponsor's organizational strategy," a common corporate euphemism that often masks portfolio reprioritization based on emerging data, changing market dynamics, or a reassessment of a program's probability of success.[21] Given the likely failure of this key pro-cognitive trial, this termination was a strong signal of waning confidence in the AD indication.

Ultimately, the development of PF-04995274 was discontinued for both Alzheimer's disease and depressive disorders.[43] The later clinical studies in depression, such as RESTAND and RESTART, which were initiated after 2018, appear to have been conducted as investigator-initiated trials or academic-industry collaborations, with Pfizer providing the drug and expertise.[37] This suggests that while Pfizer ceased its internal development efforts, the compound was made available to external researchers to further probe its unique mechanism of action, a common practice for scientifically interesting but commercially non-viable assets.

The Dichotomy of Preclinical Promise and Clinical Reality

The story of PF-04995274 is a classic example of the translational gap that so often plagues CNS drug development. The compound's failure can be attributed to a confluence of factors where promising preclinical data did not accurately predict human outcomes:

  1. The Limits of Animal Models: The positive results in the rat scopolamine model and mouse depression models provided a strong, but ultimately misleading, foundation. These models were not sensitive enough to capture the critical nuance of the drug's partial agonism, which proved to be a fatal flaw in the human scopolamine challenge. Similarly, behavioral tests like the forced swim test are crude surrogates for the complex psychopathology of human depression and could not predict the divergent neural mechanism observed with fMRI.
  2. Insufficient Target Engagement (Efficacy): The core issue for the AD indication was not a lack of potency or affinity, but insufficient intrinsic activity. The failure to overcome the scopolamine challenge strongly suggests that the level of 5-HT4R activation provided by this partial agonist was simply not enough to produce a clinically meaningful pro-cognitive effect in humans.
  3. A Complicating Safety Signal: The identified "issue of concern" regarding elevated plasma aldosterone introduced a significant safety liability.[29] While the full extent of this risk is not publicly known, any potential for long-term cardiovascular or metabolic disruption would have substantially narrowed the therapeutic index and complicated the risk-benefit assessment, particularly for a drug intended for chronic use in elderly or depressed populations.
  4. A High-Risk Mechanism in Depression: The novel "top-down" cognitive control mechanism observed in the RESTAND study, while scientifically compelling, represented a high-risk, unvalidated path to an antidepressant approval compared to the well-trodden ground of SSRIs. For a large pharmaceutical company managing a portfolio, pivoting to such a high-risk mechanism after the failure of the primary indication would be a difficult strategic decision.

Future Outlook for 5-HT4R Agonists in CNS Therapeutics

Despite the discontinuation of PF-04995274, the 5-HT4 receptor remains a viable and compelling therapeutic target for CNS disorders.[2] The development program for this compound, though unsuccessful, has provided invaluable lessons that can guide future efforts.

The primary takeaway is that future 5-HT4R agonists intended to produce robust pro-cognitive or antidepressant effects should likely possess higher intrinsic activity than PF-04995274.[3] The need for a more substantial degree of receptor activation appears critical for translating preclinical efficacy into clinical benefit.

Furthermore, the unique neural signature identified in the RESTAND study suggests a promising niche for 5-HT4R agonists in treating the cognitive symptoms of depression, an area of profound unmet need where current treatments fall short.[38] A drug that can enhance cognitive control could be a valuable new tool in the psychiatric armamentarium. Finally, the exploration of 5-HT4R agonists as prophylactic agents to enhance stress resilience, as detailed in recent patent applications, represents an innovative therapeutic paradigm that warrants continued investigation.[25] The story of PF-04995274 is therefore not merely a chronicle of failure, but a crucial scientific chapter that refines our understanding of the 5-HT4R system and provides a clearer, albeit more challenging, roadmap for the development of the next generation of therapeutics for this important CNS target.

Works cited

  1. PF-04995274 | CAS#1331782-27-4 | 5-HT4 Receptor Agonist - MedKoo Biosciences, accessed November 3, 2025, https://www.medkoo.com/products/10639
  2. Translating the promise of 5HT4 receptor agonists for the treatment of depression - PMC, accessed November 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8188527/
  3. Systems pharmacology modeling in neuroscience: Prediction and outcome of PF-04995274, a 5-HT4 partial agonist, in a clinical scopolamine impairment trial - Scirp.org., accessed November 3, 2025, https://www.scirp.org/journal/paperinformation?paperid=37276
  4. 5-HT4 receptor agonists treatment reduces tau pathology and behavioral deficit in the PS19 mouse model of tauopathy - Frontiers, accessed November 3, 2025, https://www.frontiersin.org/journals/cellular-neuroscience/articles/10.3389/fncel.2024.1338502/full
  5. PF-04995274: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed November 3, 2025, https://go.drugbank.com/drugs/DB12675
  6. 5-HT4 Receptor Agonists for the Treatment of Alzheimer's Dsease - Scirp.org., accessed November 3, 2025, https://www.scirp.org/journal/paperinformation?paperid=5519
  7. PF 04995274, a 5-HT4Partial Agonist | New Drug Approvals, accessed November 3, 2025, https://newdrugapprovals.org/2016/02/03/pf-04995274-a-5-ht4partial-agonist/
  8. Early administration of RS 67333, a specific 5-HT4 receptor agonist, prevents amyloidogenesis and behavioral deficits in the 5XFAD mouse model of Alzheimer's disease - Frontiers, accessed November 3, 2025, https://www.frontiersin.org/journals/aging-neuroscience/articles/10.3389/fnagi.2013.00096/full
  9. Early effects of a novel 5-HT4 agonist (PF-04995274) and the SSRI citalopram on emotional cognition in unmedicated depression: the RESTAND study | medRxiv, accessed November 3, 2025, https://www.medrxiv.org/content/10.1101/2025.08.28.25334462v1
  10. (PDF) Early effects of a novel 5-HT4 agonist (PF-04995274) and the ..., accessed November 3, 2025, https://www.researchgate.net/publication/395040639_Early_effects_of_a_novel_5-HT4_agonist_PF-04995274_and_the_SSRI_citalopram_on_emotional_cognition_in_unmedicated_depression_the_RESTAND_study
  11. The Effect of the 5-HT4 Agonist, Prucalopride, on a Functional Magnetic Resonance Imaging Faces Task in the Healthy Human Brain - Frontiers, accessed November 3, 2025, https://www.frontiersin.org/journals/psychiatry/articles/10.3389/fpsyt.2022.859123/full
  12. Pf-04995274 | C23H32N2O6 | CID 53354764 - PubChem, accessed November 3, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/pf-04995274
  13. PF-04995274 | Ligand page - IUPHAR/BPS Guide to PHARMACOLOGY, accessed November 3, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=9059
  14. PF-04995274 [CAS: 1331782-27-4] glixxlabs - Glixx Laboratories, accessed November 3, 2025, http://glixxlabs.com/chemical-products/bioactive-screen-leads-p6/GLXC-12295
  15. PF-04995274 - precisionFDA, accessed November 3, 2025, https://precision.fda.gov/ginas/app/ui/substances/fe9315f8-7964-4baf-9763-e911084314a5
  16. GLASS2.0: GPCR-ligand Association Database - The Yang Zhang Lab, accessed November 3, 2025, https://zhanggroup.org/GLASS/index.html?page=ligand.html%3Fid%3D30569143%26search%3D
  17. PF-04995274 (CAS Number: 1331782-27-4) | Cayman Chemical, accessed November 3, 2025, https://www.caymanchem.com/product/28776/pf-04995274
  18. PF-04995274 | 5-HT Receptor - TargetMol, accessed November 3, 2025, https://www.targetmol.com/compound/pf-04995274
  19. PF-04995274 | 5-HT4 Receptor Agonist | MedChemExpress, accessed November 3, 2025, https://www.medchemexpress.com/pf-04995274.html
  20. PF-04995274 | 5-HT Receptor agonist | Mechanism | Concentration - Selleck Chemicals, accessed November 3, 2025, https://www.selleckchem.com/products/pf-04995274.html
  21. Assessment Of The Effects Of Single Doses Of An Investigational Drug, Given Alone Or With Donepezil, On Scopolamine-Induced Changes In Memory And Learning In Healthy Adults | PatLynk, accessed November 3, 2025, https://www.patlynk.com/es/trial/NCT01345864
  22. Prophylactic efficacy of 5-HT4R agonists against stress - PMC - PubMed Central, accessed November 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6969048/
  23. Prophylactic efficacy of 5-HT4R agonists against stress - PubMed - NIH, accessed November 3, 2025, https://pubmed.ncbi.nlm.nih.gov/31600767/
  24. Prophylactic efficacy of 5-HT4R agonists against stress - bioRxiv, accessed November 3, 2025, https://www.biorxiv.org/content/10.1101/712786v1.full.pdf
  25. WO2020210393A1 - Prophylactic efficacy of serotonin 4 receptor ..., accessed November 3, 2025, https://patents.google.com/patent/WO2020210393A1/en
  26. NCT01169714 | A Study To Observe Safety And Blood Concentrations Of PF-04995274 During And Following the Administration of Multiple Doses Of PF-04995274 In Healthy Adult And Healthy Elderly Volunteers. | ClinicalTrials.gov, accessed November 3, 2025, https://clinicaltrials.gov/study/NCT01169714
  27. NCT01193062 | Study In Healthy Subjects To Evaluate The Changes In The Protein sAPP-Alpha In Cerebrospinal Fluid Following A Single Oral Dose Of PF-04995274 | ClinicalTrials.gov, accessed November 3, 2025, https://clinicaltrials.gov/study/NCT01193062
  28. NCT01091272 | A Study To Evaluate The Safety, Tolerability And Plasma Drug Levels Following A Single Dose Of PF-04995274 In Healthy Adult Volunteers | ClinicalTrials.gov, accessed November 3, 2025, https://clinicaltrials.gov/study/NCT01091272
  29. Pharmacokinetics, safety and tolerability of PF04995274: A 5HT4, accessed November 3, 2025, https://www.researchgate.net/publication/251687261_Pharmacokinetics_safety_and_tolerability_of_PF04995274_A_5HT4_partial_agonist_being_developed_for_the_treatment_of_Alzheimer's_disease
  30. NCT01169714 | A Study To Observe Safety And Blood Concentrations Of PF-04995274 During And Following the Administration of Multiple Doses Of PF-04995274 In Healthy Adult And Healthy Elderly Volunteers. | ClinicalTrials.gov, accessed November 3, 2025, https://www.clinicaltrials.gov/study/NCT01169714
  31. A Study To Observe Safety And Blood Concentrations Of PF-04995274 During And Following the Administration of Multiple Doses Of PF-04995274 In Healthy Adult And Healthy Elderly Volunteers. | MedPath, accessed November 3, 2025, https://trial.medpath.com/clinical-trial/01c95611c2f5a2c1/nct01169714-investigator-subject-blind-phase-1-study-pf-04995274
  32. PF-4995274 - Drug Targets, Indications, Patents - Patsnap Synapse, accessed November 3, 2025, https://synapse.patsnap.com/drug/7b3c3daa95cd42eba6fc0bc8b4941ec1
  33. Early effects of a novel 5-HT4 agonist (PF-04995274) and the SSRI citalopram on emotional cognition in unmedicated depression: the RESTAND study - eLife, accessed November 3, 2025, https://sciety-labs.elifesciences.org/articles/by?article_doi=10.1101/2025.08.28.25334462
  34. Uncategorized | New Drug Approvals | Page 128, accessed November 3, 2025, https://newdrugapprovals.org/category/uncategorized/page/128/
  35. Unipolar Depression Completed Phase 1 Trials for PF-04995274 (DB12675) - DrugBank, accessed November 3, 2025, https://go.drugbank.com/indications/DBCOND0028284/clinical_trials/DB12675?phase=1&status=completed
  36. PF-04995274 Completed Phase 1 Trials for Major depressive disorder, recurrent episode / Unipolar Depression Basic Science - DrugBank, accessed November 3, 2025, https://go.drugbank.com/drugs/DB12675/clinical_trials?conditions=DBCOND0089536%2CDBCOND0028284&phase=1&purpose=basic_science&status=completed
  37. PF-04995274 and emotional processing in treatment-resistant depression, accessed November 3, 2025, https://www.hra.nhs.uk/planning-and-improving-research/application-summaries/research-summaries/pf-04995274-and-emotional-processing-in-treatment-resistant-depression/
  38. MICA: 5-HT4 receptor activation as a novel mechanism of antidepressant action - GtR, accessed November 3, 2025, https://gtr.ukri.org/projects?ref=MR%2FP012604%2F1
  39. Pfizer Pipeline, accessed November 3, 2025, https://cdn.pfizer.com/pfizercom/research/pipeline/2010_0927/pipeline_2010_0927.pdf
  40. Pfizer Pipeline, accessed November 3, 2025, https://cdn.pfizer.com/pfizercom/research/pipeline/2011_0512/pipeline_2011_0512.pdf
  41. Pfizer Pipeline, accessed November 3, 2025, https://cdn.pfizer.com/pfizercom/research/pipeline/2011_0228/pipeline_2011_0228.pdf
  42. PF 4995274 - AdisInsight, accessed November 3, 2025, https://adisinsight.springer.com/drugs/800031903
  43. T59679 - TTD - Therapeutic Target Database, accessed November 3, 2025, https://db.idrblab.net/ttd/target/t59679
  44. Report to the Meeting of the Oxford Health NHS Foundation Trust Board of Directors APRIL 2021 Research and Development Report Fo, accessed November 3, 2025, https://www.oxfordhealth.nhs.uk/wp-content/uploads/2021/01/24_BOD_RD-Board-Report-March-2021-V13-JG-MH-approved-070421.pdf
  45. Effects on hippocampal activity following 5-HT4 receptor agonism in ..., accessed November 3, 2025, https://pubmed.ncbi.nlm.nih.gov/40950496/

Published at: November 3, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.