MedPath

Osoresnontrine Advanced Drug Monograph

Published:Sep 24, 2025

Generic Name

Osoresnontrine

Drug Type

Small Molecule

Chemical Formula

C16H17N5O2

CAS Number

1189767-28-9

Report on Osoresnontrine (BI-409306): An Investigational PDE9A Inhibitor

Executive Summary

Osoresnontrine, identified by the developmental code BI-409306, is an orally bioavailable, small molecule compound developed by Boehringer Ingelheim as a potent and selective inhibitor of the enzyme phosphodiesterase 9A (PDE9A).[1] The therapeutic rationale for its development was predicated on a well-defined neurobiological hypothesis: the inhibition of PDE9A, an enzyme highly expressed in cognition-relevant brain regions, would lead to an elevation of intracellular cyclic guanosine monophosphate (cGMP) levels. This biochemical change was expected to enhance N-methyl-D-aspartate (NMDA) receptor-dependent signaling pathways, which are fundamental to synaptic plasticity, learning, and memory.[1] Consequently, Osoresnontrine was advanced into clinical trials as a potential symptomatic treatment for the cognitive impairments associated with Alzheimer's disease and schizophrenia, two conditions with significant unmet medical needs.

Preclinical investigations provided a robust foundation for this hypothesis, with rodent models demonstrating that Osoresnontrine successfully crossed the blood-brain barrier, engaged its target to increase brain cGMP levels, enhanced long-term potentiation (a cellular correlate of memory), and improved performance in cognitive tasks.[1] Crucially, this mechanism was confirmed in human subjects, where the drug produced a dose-dependent increase in cGMP in the cerebrospinal fluid (CSF), providing clear evidence of target engagement within the central nervous system.[6]

Despite this strong preclinical rationale and confirmed pharmacodynamic activity in humans, the extensive Phase II clinical development program for Osoresnontrine ultimately failed to demonstrate clinical efficacy. Multiple large, well-controlled trials in both Alzheimer's disease and schizophrenia did not meet their primary endpoints for cognitive improvement or relapse prevention.[9] While the drug was generally well-tolerated, it was associated with dose-dependent adverse events, most notably transient ocular disorders such as photophobia and chromatopsia.[13] Pharmacokinetic studies also revealed a significant challenge: the drug's metabolism is highly dependent on the CYP2C19 enzyme, leading to 4- to 5-fold higher systemic exposure in individuals with a "poor metabolizer" genotype, complicating dosing and safety assessments.[7]

Ultimately, development of Osoresnontrine was discontinued by Boehringer Ingelheim due to a consistent lack of efficacy across all tested indications.[4] The developmental trajectory of Osoresnontrine serves as a significant case study in translational neuroscience. It illustrates that a compound with a potent and selective mechanism, robust preclinical data, and confirmed human target engagement can still fail to produce a therapeutic benefit, highlighting the profound complexities of treating CNS disorders and the limitations of current preclinical models in predicting clinical success.

1.0 Drug Profile and Physicochemical Characteristics

This section delineates the fundamental chemical and physical identity of Osoresnontrine, providing the necessary context for subsequent pharmacological and clinical evaluation.

1.1 Identification and Nomenclature

Osoresnontrine is a small molecule drug that was under investigation for its therapeutic potential in central nervous system disorders.[4] Throughout its development and in the scientific literature, it has been referred to by several names and unique identifiers, which are essential for accurately tracking its research history.

  • International Nonproprietary Name (INN): Osoresnontrine [16]
  • Developmental Codes: BI-409306, SUB 166499 [1]
  • Drug Type: Small Molecule [5]

The compound is cataloged in major chemical and pharmacological databases under the following registry numbers:

  • Chemical Abstracts Service (CAS) Number: 1189767-28-9 [1]
  • DrugBank ID: DB16274 [16]
  • PubChem Compound ID (CID): 135908617 [16]
  • Unique Ingredient Identifier (UNII): O9OC34WOAY [16]

1.2 Molecular Structure and Chemical Properties

Osoresnontrine is a complex heterocyclic compound characterized by a fused bicyclic pyrazolopyrimidinone core structure, which is a common scaffold in pharmaceutically active molecules.[19]

  • Molecular Formula: C16​H17​N5​O2​ [1]
  • Molecular Weight: 311.34 g/mol (also reported as 311.35 g/mol) [1]
  • International Union of Pure and Applied Chemistry (IUPAC) Name: 1-(oxan-4-yl)-6-(pyridin-2-ylmethyl)-5H-pyrazolo[3,4-d]pyrimidin-4-one. An alternative, equivalent systematic name is 6-(pyridin-2-ylmethyl)-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one.[1]
  • Chemical Structure Representations:
  • SMILES (Simplified Molecular Input Line Entry System): O=c1nc(Cc2ccccn2)[nH]c2n(ncc12)C1CCOCC1.[23] Another valid representation is C1COCCC1N2C3=C(C=N2)C(=O)NC(=N3)CC4=CC=CC=N4.[16]
  • InChIKey (International Chemical Identifier Key): BZTIJCSHNVZMES-UHFFFAOYSA-N.[1]

1.3 Physical and Formulation Properties

The physical properties of Osoresnontrine influence its handling, formulation, and behavior in biological systems.

  • Appearance: The compound is a solid powder, described as white to off-white in appearance.[1]
  • Solubility: Osoresnontrine exhibits moderate polar characteristics. Its solubility is well-characterized in dimethyl sulfoxide (DMSO), a common solvent for laboratory research, with reported concentrations of 22 mg/mL (equivalent to 70.66 mM) and 25 mg/mL (80.30 mM).[19] Achieving dissolution can sometimes require sonication or warming, suggesting that preparing highly concentrated aqueous solutions may be challenging.[23]
  • Stability and Storage: As a solid powder, Osoresnontrine is stable for up to three years when stored at -20 °C. When dissolved in a solvent, it maintains stability for one year at -80 °C. For short-term use, storage in a dry, dark environment at 0-4 °C is recommended.[1]
  • Predicted Physicochemical Properties: Computational models predict properties that are consistent with a viable oral drug candidate. Its predicted water solubility is low at 0.481 mg/mL, and its predicted octanol-water partition coefficient (logP) is 0.88, indicating moderate lipophilicity suitable for crossing the blood-brain barrier.[5] The molecule adheres to Lipinski's Rule of Five, a set of guidelines used to evaluate the drug-likeness of a chemical compound and its likelihood of being an orally active drug in humans.[5]
Table 1: Key Identifiers and Physicochemical Properties of Osoresnontrine
PropertyValueSource(s)
International Nonproprietary Name (INN)Osoresnontrine16
Developmental CodesBI-409306, SUB 1664991
CAS Number1189767-28-916
DrugBank IDDB1627416
Molecular FormulaC16​H17​N5​O2​16
Molecular Weight311.34 g/mol16
IUPAC Name1-(oxan-4-yl)-6-(pyridin-2-ylmethyl)-5H-pyrazolo[3,4-d]pyrimidin-4-one1
Solubility (DMSO)22-25 mg/mL (70.66-80.30 mM)23
Water Solubility (Predicted)0.481 mg/mL5
logP (Predicted)0.25 - 0.885
Rule of Five ComplianceYes5

2.0 Pharmacology and Mechanism of Action

This section details the molecular mechanism by which Osoresnontrine exerts its effects and the scientific rationale that positioned it as a candidate for treating cognitive disorders.

2.1 The Role of Phosphodiesterase 9A in Neuronal Signaling

Phosphodiesterases (PDEs) are a large superfamily of enzymes that regulate cellular function by catalyzing the hydrolysis of the second messengers cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP).[25] This action terminates their signaling cascades, making PDEs critical control points in cellular physiology.

The PDE9 family, and specifically the PDE9A isoform, is distinguished by its exceptionally high affinity and selectivity for cGMP.[5] It functions as a key regulator of intracellular cGMP concentrations, particularly in signaling pathways linked to the neurotransmitters nitric oxide and glutamate.[1] Within the central nervous system, PDE9A is highly expressed in brain regions integral to learning and memory, including the neocortex and hippocampus.[7] The enzyme's activity is thought to modulate glutamatergic neurotransmission, particularly through the NMDA receptor pathway. NMDA receptor activation is a fundamental process for inducing synaptic plasticity, the cellular mechanism believed to underlie memory formation, which is often measured experimentally as long-term potentiation (LTP).[6] The therapeutic hypothesis for PDE9A inhibitors is that by preventing the breakdown of cGMP, they can amplify and prolong the signaling cascades downstream of NMDA receptor activation, thereby strengthening synaptic connections and improving cognitive function.[1]

2.2 In Vitro Inhibitory Profile and Selectivity of Osoresnontrine

In vitro enzymatic assays are crucial for defining a drug's potency and selectivity for its intended target. Osoresnontrine was characterized as a potent and highly selective inhibitor of the PDE9A enzyme.

  • Potency: Multiple independent sources consistently report that Osoresnontrine inhibits human PDE9A with a half-maximal inhibitory concentration (IC50​) in the range of 52 nM to 65 nM.[1] This high potency indicates that the drug can effectively block the enzyme's activity at low nanomolar concentrations, which is a desirable characteristic for a therapeutic agent.
  • Selectivity: A critical feature of a well-designed drug is its ability to interact specifically with its intended target while avoiding other related proteins, thereby minimizing the potential for off-target side effects. Osoresnontrine demonstrates a strong selectivity profile. Its inhibitory activity against other PDE isoforms is substantially weaker. For instance, its IC50​ values for PDE1C and PDE1A are approximately 1.0 µM and 1.4 µM, respectively, making it about 19 to 27 times less potent against these enzymes compared to PDE9A. For other major PDE isoforms, including PDE2A, 3A, 4B, 5A, and 10A, no significant inhibition was observed at concentrations up to 10 µM.[1] This high degree of selectivity for PDE9A was a key pharmacological attribute that supported its advancement into clinical development.
Table 2: In Vitro Inhibitory Profile of Osoresnontrine Against PDE Isoforms
PDE IsoformIC50​ (nM)Selectivity Ratio (vs. PDE9A)
PDE9A521x (Reference)
PDE1C1,000~19x
PDE1A1,400~27x
PDE2A, 3A, 4B, 5A, 6AB, 7A, 10A>10,000>192x
Data synthesized from sources.1

2.3 Rationale for Targeting Cognitive Dysfunction in Alzheimer's Disease and Schizophrenia

The decision to investigate Osoresnontrine in both Alzheimer's disease and schizophrenia stemmed from the recognition that cognitive impairment is a core, debilitating feature of both disorders and represents a major unmet medical need.[2] Both conditions are associated with disruptions in glutamatergic neurotransmission, the very system that PDE9A inhibition was hypothesized to modulate.[12] The therapeutic strategy was therefore to provide a symptomatic improvement in cognition by targeting this shared pathophysiological element. This approach was considered novel because it diverged from the prevailing therapeutic strategies in each field. In Alzheimer's research, it offered an alternative to the dominant focus on clearing amyloid-beta plaques and tau tangles.[11] In schizophrenia, it moved beyond the traditional focus on blocking dopamine D2 receptors to address the negative and cognitive symptoms that are poorly managed by existing antipsychotics.[12]

The development of Osoresnontrine represents a classic example of rational drug design, where a potent and selective molecule was created to test a clear and elegant biological hypothesis. The very clarity of this mechanism, however, ultimately contributed to the definitive nature of its clinical failure. Had the mechanism been less well-defined, ambiguity might have remained. Instead, the program's ability to later confirm target engagement in humans while still observing no clinical effect created a powerful, albeit negative, conclusion: the hypothesis, however elegant, was likely incorrect in the context of these complex human diseases. This demonstrates that while a well-defined mechanism is a necessary starting point for drug development, it is by no means a guarantee of clinical success, particularly in CNS disorders where the link between molecular targets and complex behavioral outcomes is often tenuous.

3.0 Preclinical Efficacy and Proof-of-Concept

Before advancing to human trials, investigational drugs must demonstrate evidence of biological activity and therapeutic potential in non-human models. The preclinical data package for Osoresnontrine was robust and provided a compelling, logical basis for its clinical development program.

3.1 In Vivo Target Engagement: Modulation of Brain cGMP Levels

A critical first step in preclinical in vivo assessment is to confirm that the drug can reach its target in the brain and produce the intended biochemical effect. Studies in rodents provided direct evidence of Osoresnontrine's ability to engage PDE9A in the central nervous system.

  • Brain Penetrance and Target Modulation: Following oral administration in rats, Osoresnontrine produced a dose-dependent increase in cGMP levels in both the prefrontal cortex and the cerebrospinal fluid (CSF).[1] This finding was crucial, as it confirmed that the compound was not only capable of crossing the blood-brain barrier but was also actively inhibiting its target enzyme in relevant brain regions.
  • Reversal of Pharmacological Deficit: To further validate its mechanism, Osoresnontrine was tested in a pharmacological challenge model. The NMDA receptor antagonist MK-801 is known to disrupt glutamatergic signaling and reduce cGMP levels. Co-administration of Osoresnontrine in mice successfully attenuated this MK-801-induced reduction in striatal cGMP, demonstrating that the drug could restore cGMP signaling even under conditions of pathway disruption.[1]

3.2 Effects on Synaptic Plasticity and Long-Term Potentiation (LTP)

With target engagement confirmed, the next step was to determine if this biochemical effect translated to a functional change at the cellular level. The primary hypothesis was that increased cGMP would enhance synaptic plasticity, a key cellular process underlying memory formation.

  • Enhancement of LTP: Studies using ex vivo rat hippocampal slices, a standard model for studying synaptic plasticity, showed that Osoresnontrine significantly enhanced LTP.[6] This effect was observed in response to both weak and strong electrical stimulation protocols, suggesting that the drug could promote both the initial phase (early-LTP) and the more durable, protein synthesis-dependent phase (late-LTP) of synaptic strengthening.[6] The ability to enhance late-LTP was particularly promising, as it is more closely associated with the consolidation of long-term memories.

3.3 Efficacy in Rodent Models of Cognition and Memory

The final and most important piece of the preclinical puzzle was to demonstrate that these cellular effects could translate into improved cognitive performance in behavioral tasks.

  • Working Memory Improvement: In a T-maze spontaneous alternation task, a test of spatial working memory, Osoresnontrine reversed the cognitive deficits induced by MK-801 in mice.[1] This provided evidence for its potential to treat deficits in short-term or "online" memory processing.
  • Long-Term Memory Enhancement: In a novel object recognition task, which assesses long-term episodic memory, mice treated with Osoresnontrine showed improved memory performance.[6] This result supported the hypothesis that the drug could enhance the consolidation and retrieval of long-term memories.

Collectively, these preclinical findings constructed a strong and internally consistent "translational bridge" from the molecular target to behavioral outcomes. The evidence followed a clear, logical progression: the drug potently and selectively inhibited PDE9A in vitro; it crossed the blood-brain barrier to increase cGMP levels in vivo; this biochemical change enhanced the cellular mechanisms of synaptic plasticity; and this enhancement of plasticity translated into measurable improvements in cognitive function in animal models.[2] This seemingly robust chain of evidence provided a compelling justification for advancing Osoresnontrine into human clinical trials. However, the subsequent failure of the drug in those trials serves as a stark reminder that even the most well-constructed preclinical translational bridge can collapse when faced with the complexity of human disease, calling into question the predictive validity of these specific animal models for multifactorial conditions like Alzheimer's disease and schizophrenia.

4.0 Human Pharmacokinetics, Pharmacodynamics, and Metabolism (ADME)

The transition from preclinical models to human subjects is a critical phase in drug development. This section details the pharmacokinetic (what the body does to the drug) and pharmacodynamic (what the drug does to the body) profile of Osoresnontrine in humans, as determined through a series of Phase I clinical trials.

4.1 Absorption, Distribution, and Elimination Profile

Phase I studies in healthy volunteers established the fundamental ADME properties of Osoresnontrine.

  • Absorption: Following oral administration, Osoresnontrine is absorbed very rapidly. Peak plasma concentrations (Cmax​) are typically reached in under one hour, with studies reporting a time to maximum concentration (Tmax​) of 30-45 minutes.[7]
  • Elimination: The drug is also eliminated rapidly from the body. The terminal elimination half-life (t1/2​) is short, generally ranging from 1 to 2.7 hours.[7] This rapid clearance means the drug does not persist in the system for long periods.
  • Accumulation: The short half-life results in minimal drug accumulation with once-daily dosing. Pharmacokinetic modeling and empirical data show that steady-state concentrations are achieved quickly, typically by the second or third day of continuous administration.[14]
  • Dose Proportionality: Across the tested dose ranges, systemic exposure, as measured by both Cmax​ and the area under the concentration-time curve (AUC), increased in a dose-dependent manner. This increase was characterized as slightly more than dose-proportional, meaning a doubling of the dose resulted in slightly more than a doubling of the plasma concentration.[7]

4.2 The Critical Role of CYP2C19 Metabolism and Genetic Polymorphisms

A pivotal finding from the early clinical studies was the identification of the cytochrome P450 2C19 (CYP2C19) enzyme as the primary pathway for Osoresnontrine's metabolism.[7] The gene for this enzyme is known to have common genetic variations (polymorphisms) that result in different metabolizer phenotypes within the population.

  • Pharmacogenetic Variability: Clinical trials specifically enrolled and compared individuals genotyped as "Extensive Metabolizers" (EMs), who have normal CYP2C19 function, and "Poor Metabolizers" (PMs), who have significantly reduced or absent enzyme function.[7]
  • Impact on Exposure: This genetic difference had a profound impact on the pharmacokinetics of Osoresnontrine. At the same oral dose, individuals who were PMs exhibited dramatically higher systemic exposure to the drug compared to EMs. Specifically, Cmax​ was found to be 2.2 to 2.3-fold higher, and the total drug exposure (AUC) was 4.1 to 5.0-fold higher in PMs.[7] This large variability in exposure based on a common genetic polymorphism presents a significant clinical challenge for consistent and safe dosing.
Table 3: Summary of Human Pharmacokinetic Parameters for Osoresnontrine by CYP2C19 Genotype
ParameterValue in Extensive Metabolizers (EMs)Value in Poor Metabolizers (PMs)Fold-Difference (PM vs. EM)
Tmax​ (Time to Peak Concentration)<1 hour<1 hourNo significant difference
t1/2​ (Elimination Half-Life)0.99 - 2.71 hoursWithin a similar range as high doses in EMs~1-1.5x
Cmax​ (Peak Concentration)Dose-dependent2.2 - 2.3x higher than EMs at the same dose~2.3x
AUC (Total Exposure)Dose-dependent4.1 - 5.0x higher than EMs at the same dose~4.5x
Data synthesized from the first-in-human study NCT01343706 as reported in sources.7

4.3 Pharmacodynamic Effects: cGMP Modulation in Human CSF

A key objective of the early clinical program was to confirm that the mechanism of action observed in animals was also present in humans. The clinical trial NCT01493570 was a dedicated "proof of mechanism" study designed specifically for this purpose.[8] The trial successfully demonstrated that single oral doses of Osoresnontrine led to a dose-dependent and statistically significant increase in cGMP levels in the cerebrospinal fluid of healthy male volunteers.[6] This result was a landmark achievement for the development program. It provided unequivocal evidence that Osoresnontrine crossed the human blood-brain barrier and engaged its target, PDE9A, to produce the intended biochemical effect in the central nervous system.

This confirmation of target engagement, however, became a double-edged sword in the context of the program's subsequent failures. The positive pharmacodynamic data effectively eliminated the possibility that the drug failed due to poor brain penetration or an inability to inhibit its target in humans. Instead, it strongly implied that the target itself, when modulated, did not produce the desired therapeutic effect. This shifted the focus of the failure from the drug molecule to the fundamental validity of the therapeutic hypothesis, making the negative efficacy results far more definitive.

4.4 Drug-Drug Interaction Profile

Given the drug's reliance on a specific metabolic pathway, understanding its potential for interactions with other medications was essential. Several dedicated studies were conducted:

  • CYP Interactions: Trial NCT02248259 was designed to investigate interactions with itraconazole, a potent inhibitor of the CYP3A4 enzyme, in both CYP2C19 EMs and PMs, to fully characterize its metabolic profile.[34]
  • Interactions with Concomitant Medications: To assess its use in the target patient populations, interactions with standard-of-care medications were studied. Trials NCT02635750 and NCT03151499 evaluated the pharmacokinetic interplay between Osoresnontrine and donepezil, a widely used treatment for Alzheimer's disease.[35] Another study, NCT03193307, assessed potential interactions with oral contraceptives.[38]

5.0 Clinical Development in Alzheimer's Disease

Following the promising preclinical data and confirmation of human target engagement, Boehringer Ingelheim initiated a Phase II clinical program to evaluate the efficacy and safety of Osoresnontrine for treating cognitive impairment in patients with Alzheimer's disease.

5.1 Overview of the Phase II Clinical Program

The strategy for Alzheimer's disease involved two large, parallel, multi-center, randomized, double-blind, placebo-controlled studies. The primary objective of these trials was to demonstrate that Osoresnontrine could produce a statistically significant improvement in cognitive function compared to placebo over a 12-week treatment period.[2] These trials represented the first major test of the PDE9A inhibition hypothesis in a patient population.

Table 4: Overview of Key Phase I & II Clinical Trials for Osoresnontrine
Trial ID (NCT)PhaseStatusIndication(s)Patient PopulationPrimary Endpoint(s)Key Outcome
NCT013437061CompletedHealthy VolunteersHealthy Male EMs & PMsSafety, Tolerability, PKGenerally safe; PK highly dependent on CYP2C19 genotype
NCT014935701CompletedHealthy VolunteersHealthy Male VolunteersPK/PD (cGMP in CSF)Confirmed dose-dependent increase of cGMP in human CSF
NCT022406932CompletedAlzheimer's DiseasePatients with Prodromal ADChange in NTB z-scoreFailed to show superiority over placebo
NCT023379072CompletedAlzheimer's DiseasePatients with Mild ADChange in NTB z-scoreFailed to show superiority over placebo
NCT022817732CompletedSchizophreniaPatients with Cognitive ImpairmentChange in MCCB composite scoreFailed to meet primary endpoint; no cognitive improvement
NCT033512442TerminatedSchizophreniaPatients on AntipsychoticsPrevention of RelapseTerminated (COVID-19); did not meet primary endpoint
NCT032300972TerminatedPsychotic DisordersAttenuated Psychosis SyndromeEfficacy and TolerabilityTerminated
Table synthesizes data from multiple sources including.2

5.2 Analysis of Key Efficacy Trials

The core of the Alzheimer's program consisted of two sister studies:

  • NCT02337907: This trial enrolled patients with cognitive impairment due to mild Alzheimer's disease. It was designed as a parallel-group study comparing different doses of Osoresnontrine to both placebo and an active comparator, donepezil.[35] The primary outcome measure was the change from baseline in the total z-score of a comprehensive Neuropsychological Test Battery (NTB) after 12 weeks of treatment.[36] Patients were permitted to be on a stable dose of an acetylcholinesterase inhibitor, reflecting a real-world treatment setting.[40]
  • NCT02240693: This was a similar "proof of concept" study that enrolled patients with a confirmed diagnosis of prodromal Alzheimer's disease, an earlier stage of the condition.[12] It compared four different doses of Osoresnontrine against placebo, also using the 12-week change in NTB score as the primary efficacy endpoint.[42]

5.3 Efficacy Outcomes and Failure to Meet Primary Endpoints

The results from these trials were definitive and disappointing. Topline data from a pooled analysis of 457 patients across both NCT02240693 and NCT02337907 were released, showing that Osoresnontrine failed to demonstrate any statistically significant benefit on cognitive function compared to placebo.[11] The primary endpoint of improving the NTB total z-score was not met at any of the tested doses.[11]

5.4 Strategic Decision to Halt Development for Alzheimer's Disease

Following the unequivocal negative results from the Phase II program, Boehringer Ingelheim announced its decision to terminate the development of Osoresnontrine for the treatment of Alzheimer's disease.[4] This outcome added Osoresnontrine to a long list of investigational Alzheimer's drugs that have failed in mid- to late-stage clinical trials, further highlighting the immense difficulty of developing effective treatments for this neurodegenerative disorder. The focus of the asset was subsequently shifted entirely to its potential in psychiatric indications.[12]

6.0 Clinical Development in Schizophrenia and Psychotic Disorders

Concurrent with and following the Alzheimer's program, Osoresnontrine was extensively investigated in a series of Phase II trials for schizophrenia and related psychotic disorders. This program was ambitious, targeting three distinct therapeutic goals: symptomatic improvement of cognitive impairment, prevention of relapse, and early intervention in individuals at high risk of developing psychosis.

6.1 Investigation for Cognitive Impairment (Trial NCT02281773)

Cognitive Impairment Associated with Schizophrenia (CIAS) is a core feature of the illness and a primary driver of functional disability. This trial was the largest and most definitive study of Osoresnontrine in this indication.

  • Design: NCT02281773 was a large, multi-center, double-blind, parallel-group Phase II trial that randomized 518 patients with stable schizophrenia to receive one of four once-daily doses of Osoresnontrine (10, 25, 50, or 100 mg) or placebo for 12 weeks, in addition to their ongoing antipsychotic medication.[5] The study employed a sophisticated "learn-and-confirm" adaptive trial design. An initial "learn" stage was intended to identify the most responsive cognitive domains on one test battery (CANTAB) to select the primary endpoint for the final "confirm" stage analysis.[15]
  • Endpoints: In the "learn" stage, no single cognitive domain showed a sufficient signal of efficacy. Therefore, as per the protocol, the primary endpoint for the final analysis defaulted to the change from baseline in the composite score of the MATRICS Consensus Cognitive Battery (MCCB), a standard and comprehensive measure of cognition in schizophrenia. A key secondary endpoint was the change in the Schizophrenia Cognition Rating Scale (SCoRS) total score, which measures cognition-related functional capacity.[15]
  • Results: The trial failed to meet its primary and key secondary endpoints. There was no statistically significant difference in the change from baseline on the MCCB composite score between any of the four Osoresnontrine dose groups and the placebo group. Similarly, no significant improvement was observed on the SCoRS total score.[9] The results were unequivocally negative, indicating that 12 weeks of treatment with Osoresnontrine did not improve cognitive function in patients with schizophrenia.

6.2 Investigation for Relapse Prevention (Trial NCT03351244)

This study tested a different therapeutic hypothesis: whether adjunctive treatment with Osoresnontrine could help maintain stability and prevent the recurrence of psychotic symptoms.

  • Design: NCT03351244 was a Phase II, randomized, double-blind, placebo-controlled study designed to evaluate the efficacy of Osoresnontrine as an add-on to standard antipsychotic treatment for the prevention of relapse over a 28-week period.[37]
  • Status and Outcome: The trial was officially terminated prematurely, with the sponsor citing disruption due to the COVID-19 pandemic as the reason.[3] However, an analysis of the available data indicated that the study had not met its primary endpoint for relapse prevention.[4] A post-hoc analysis suggested a potential reduction in relapse risk specifically among patients who were highly adherent to the medication, but this exploratory finding from a terminated trial is not sufficient to establish efficacy.[4]

6.3 Investigation in Attenuated Psychosis Syndrome (Trial NCT03230097)

This trial represented an early-intervention strategy, testing whether Osoresnontrine could alter the course of illness in individuals identified as being at clinically high risk for developing a first episode of psychosis.

  • Design: NCT03230097 was a Phase II trial evaluating the efficacy, safety, and tolerability of Osoresnontrine in individuals with Attenuated Psychosis Syndrome (APS).[2] The goal was to determine if treatment could prevent or delay conversion to a full-blown psychotic disorder.
  • Status: Similar to the relapse prevention study, this trial was also terminated before completion.[37]

The consistent failure of Osoresnontrine across this broad and well-designed clinical program in schizophrenia and related disorders was definitive. The drug was tested for improving chronic cognitive deficits, preventing acute relapse, and intervening in an at-risk population. Its inability to show a clear efficacy signal in any of these distinct therapeutic paradigms provided a comprehensive and powerful refutation of its clinical utility in these conditions. This pattern of failure across multiple indications and endpoints strongly suggests that the therapeutic mechanism of PDE9A inhibition, while biochemically active in the human brain, is clinically inert for treating the complex neuropsychiatric symptoms under investigation.

7.0 Comprehensive Safety and Tolerability Assessment

A thorough evaluation of a drug's safety and tolerability profile is as important as assessing its efficacy. Data from the entire Osoresnontrine clinical program, from Phase I studies in healthy volunteers to Phase II studies in patient populations, provides a comprehensive picture of its safety profile.

7.1 Overview of Adverse Events from Phase I Studies in Healthy Volunteers

Early-phase trials in healthy subjects are designed to establish initial safety, tolerability, and pharmacokinetic parameters.

  • General Tolerability: In these studies, Osoresnontrine was generally reported to be safe and well-tolerated. The highest tolerated single dose in one study was 350 mg.[7] Most adverse events (AEs) were of mild to moderate intensity.[14]
  • Dose-Dependent and Exposure-Related AEs: A clear trend was observed where the frequency and intensity of AEs increased with higher doses of Osoresnontrine.[14] Furthermore, consistent with their 4- to 5-fold higher systemic drug exposure, CYP2C19 Poor Metabolizers (PMs) reported a higher incidence of AEs compared to Extensive Metabolizers (EMs) at the same dose level.[7]
  • Most Common AEs: The most frequently reported categories of AEs in healthy volunteers were nervous system disorders and eye disorders.[7]

7.2 Safety Profile in Patient Populations

Safety data from the larger Phase II trials in patients with Alzheimer's disease and schizophrenia confirmed the patterns observed in healthy volunteers.

  • Schizophrenia (NCT02281773): In this large study, the incidence of AEs was clearly dose-dependent. The rate of AEs increased from 33.3% in the 10 mg group to 53.5% in the 100 mg group, compared to an incidence of 36.4% in the placebo group.[9] Despite the increased AE rate at higher doses, the overall conclusion from the trial was that Osoresnontrine was well-tolerated and possessed an acceptable safety profile.[15]
  • GHS Hazard Classification: It is important for handling and manufacturing purposes to note that the raw chemical substance is classified under the Globally Harmonized System (GHS) with the signal word "Warning" and the hazard statement H302: "Harmful if swallowed" (Acute Toxicity, Oral, Category 4).[16] This classification applies to the concentrated chemical and not necessarily to the formulated drug at therapeutic doses.
Table 5: Summary of Most Frequently Reported Adverse Events with Osoresnontrine
System Organ ClassSpecific Adverse Events
Eye DisordersPhotophobia (light sensitivity), Photopsia (perceived flashes of light), Chromatopsia (distorted color vision), Blurred vision, Abnormal sensation in eye, Eye pain
Nervous System DisordersHeadache, Dizziness
Events compiled from Phase I and II trial data reported in sources.7

7.3 Specific Adverse Events of Interest (Ocular Disorders)

The most distinctive and consistently reported AEs associated with Osoresnontrine were transient visual disturbances.

  • Prevalence and Nature: Eye disorders were the most commonly reported class of AEs in multiple Phase I trials, with incidence rates increasing with dose and systemic exposure.[13] Specific symptoms included photophobia, photopsia, chromatopsia, and blurred vision.[13]
  • Temporal Profile: These visual AEs had a characteristic temporal profile. They typically appeared shortly after drug administration (within 20-30 minutes) and were transient, resolving on their own within 1-2 hours.[7]

The consistent reporting of these specific visual phenomena is noteworthy. These symptoms are highly reminiscent of the known side effects of PDE5 inhibitors (e.g., sildenafil) and are mechanistically linked to the inhibition of PDE6, an enzyme critical for the phototransduction cascade in the retina. Although the in vitro data for Osoresnontrine showed high selectivity for PDE9A over other isoforms, the clinical presentation of these visual AEs suggests that at the plasma concentrations achieved in humans—particularly at higher doses or in CYP2C19 PMs—there may have been functionally significant off-target inhibition of PDE6 in the eye. This represents a potential disconnect between the clean in vitro selectivity profile and the integrated physiological effects in vivo. While these AEs were not the reason for the drug's discontinuation, their presence could have posed a dose-limiting toxicity and complicated further development had the drug demonstrated any signal of efficacy.

8.0 Conclusion: The Developmental Trajectory and Future Perspective

The story of Osoresnontrine (BI-409306) provides a comprehensive and instructive case study in modern CNS drug development. It encapsulates the journey of a rationally designed molecule from a promising biological hypothesis through to a definitive clinical failure, offering valuable lessons for the pharmaceutical industry and the field of neuroscience.

8.1 Synthesis of Findings: A Profile of a Preclinically Promising but Clinically Ineffective Agent

Osoresnontrine was, by many measures, an exemplary drug candidate at the outset. It was born from a clear and compelling scientific rationale: targeting PDE9A to modulate cGMP and enhance synaptic plasticity. It was a potent and selective molecule that performed exceptionally well in preclinical models, demonstrating target engagement and pro-cognitive effects in rodents. This success continued into early human trials, where the drug's mechanism of action was confirmed through the measurement of increased cGMP in cerebrospinal fluid. The program successfully built and validated a translational bridge from the laboratory bench to human pharmacodynamics.

However, this bridge did not extend to clinical efficacy. In large, well-controlled Phase II trials, Osoresnontrine failed to show any meaningful therapeutic benefit for patients with Alzheimer's disease or schizophrenia. The lack of efficacy was not subtle or confined to a single trial; it was a consistent and definitive finding across multiple indications, patient populations, and clinical endpoints.

8.2 Analysis of the Translational Failure

The failure of Osoresnontrine cannot be attributed to poor drug properties, a lack of brain penetration, or a failure to engage its molecular target. The clinical data clearly show the drug reached the CNS and produced its intended biochemical effect. Therefore, the failure lies squarely with the therapeutic hypothesis itself. The core assumption—that elevating cGMP levels via PDE9A inhibition would be sufficient to overcome or meaningfully improve the complex cognitive deficits of Alzheimer's disease and schizophrenia—was proven incorrect.

This outcome forces a critical re-evaluation of the preclinical models that supported the program's initiation. While models involving pharmacological challenges (e.g., MK-801) or memory tests in healthy rodents are valuable for establishing proof-of-concept for a mechanism, the Osoresnontrine story suggests they possess poor predictive validity for the deeply rooted and multifactorial pathologies of human neurodegenerative and neurodevelopmental disorders. The cognitive deficits in these patients are the result of decades of complex genetic, environmental, and cellular dysfunctions—a state that is not adequately replicated by short-term, chemically induced deficits in an otherwise healthy animal brain.

8.3 Implications for the Future of PDE9A Inhibitors in CNS Disorders

The comprehensive and unambiguous failure of Osoresnontrine, a leading compound in its class, casts significant doubt on the viability of PDE9A inhibition as a therapeutic strategy for cognitive enhancement in major psychiatric and neurodegenerative diseases. While the target may hold relevance for other physiological systems and is being explored for conditions like heart failure [4], its promise in mainstream neuropsychiatry has been substantially diminished. Future efforts to target this mechanism for cognition would require a fundamentally new hypothesis or a drastically different clinical approach.

Ultimately, the developmental history of Osoresnontrine serves as a critical lesson. It underscores the immense risk and profound challenges inherent in CNS drug development. It demonstrates that even a program executed with scientific rigor, based on a strong rationale, and achieving its pharmacodynamic goals can falter at the final hurdle of clinical efficacy. This case reinforces the urgent need for the development of more disease-relevant, translatable preclinical models and for biomarkers that are not only indicative of target engagement but are also predictive of a meaningful clinical response.

Works cited

  1. Osoresnontrine | CAS#1189767-28-9 | PDE9A inhibitor | MedKoo, accessed September 24, 2025, https://www.medkoo.com/products/30215
  2. Osoresnontrine - Wikipedia, accessed September 24, 2025, https://en.wikipedia.org/wiki/Osoresnontrine
  3. Osoresnontrine - Boehringer Ingelheim - AdisInsight - Springer, accessed September 24, 2025, https://adisinsight.springer.com/drugs/800034095
  4. BI-409306 - Drug Targets, Indications, Patents - Synapse, accessed September 24, 2025, https://synapse-patsnap-com.libproxy1.nus.edu.sg/drug/c3ac00dd8ef546bd816de4471500cd7b
  5. Osoresnontrine: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed September 24, 2025, https://go.drugbank.com/drugs/DB16274
  6. The selective phosphodiesterase 9 (PDE9) inhibitor PF-04447943 (6-[(3S,4S)-4-methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d - ResearchGate, accessed September 24, 2025, https://www.researchgate.net/publication/51172198_The_selective_phosphodiesterase_9_PDE9_inhibitor_PF-04447943_6-3S4S-4-methyl-1-pyrimidin-2-ylmethylpyrrolidin-3-yl-1-tetrahydro-2H-pyran-4-yl-15-dihydro-4H-pyrazolo34-dpyrimidin-4-one_enhances_synapti
  7. First‐in‐human study assessing safety, tolerability and ..., accessed September 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5061793/
  8. Study Details | NCT01493570 | Assessment of Exposure of BI 409306 in Cerebrospinal Fluid (CSF) Relative to Plasma as Well as to Evaluation of the Effect of Different Doses of BI 409306 on the cGMP (Cyclic Guanosine Monophosphate) Levels in CSF in Healthy Male Volunteers | ClinicalTrials.gov, accessed September 24, 2025, https://clinicaltrials.gov/study/NCT01493570?intr=guanosine&rank=9
  9. (PDF) Evaluation of the Efficacy, Safety, and Tolerability of BI ..., accessed September 24, 2025, https://www.researchgate.net/publication/324919905_Evaluation_of_the_Efficacy_Safety_and_Tolerability_of_BI_409306_a_Novel_Phosphodiesterase_9_Inhibitor_in_Cognitive_Impairment_in_Schizophrenia_A_Randomized_Double-Blind_Placebo-Controlled_Phase_II_Tri
  10. Overview of Novel Antipsychotic Drugs: State of the Art, New Mechanisms, and Clinical Aspects of Promising Compounds - PMC - PubMed Central, accessed September 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11763187/
  11. Boehringer ends Alzheimer's research for PDE9 blocker - BioPharma Dive, accessed September 24, 2025, https://www.biopharmadive.com/news/boehringer-ends-alzheimers-research-for-pde9-blocker/516762/
  12. Schizophrenia: BI Focuses PDE9 Inhibition Brain Research | BI US - Boehringer Ingelheim, accessed September 24, 2025, https://www.boehringer-ingelheim.com/us/media/press-releases/boehringer-ingelheim-refocuses-pde9-inhibition-brain-research-schizophrenia-following-results-phase
  13. Alzheimer's Association International Conference on Alzheimer's Disease 2015 (AAIC 2015) (July 18-23, 2015, accessed September 24, 2025, https://access.portico.org/Portico/show?viewFile=pdf&auId=pjbf7dcx858
  14. The safety, tolerability and pharmacokinetics of BI 409306, a novel and potent PDE9 inhibitor: Overview of three Phase I randomised trials in healthy volunteers - PubMed, accessed September 24, 2025, https://pubmed.ncbi.nlm.nih.gov/29567399/
  15. Evaluation of the Efficacy, Safety, and Tolerability of BI 409306, a Novel Phosphodiesterase 9 Inhibitor, in Cognitive Impairment in Schizophrenia: A Randomized, Double-Blind, Placebo-Controlled, Phase II Trial - PubMed, accessed September 24, 2025, https://pubmed.ncbi.nlm.nih.gov/29718385/
  16. Osoresnontrine | C16H17N5O2 | CID 135908617 - PubChem, accessed September 24, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Osoresnontrine
  17. BI-409306 - Ace Therapeutics, accessed September 24, 2025, https://www.acetherapeutics.com/psychiatry/bi-409306-item-1406.html
  18. Osoresnontrine API 공급업체 - 모두 찾기 GMP 제조 업체, accessed September 24, 2025, https://pharmaoffer.com/ko/api-excipient-supplier/antipsychotics/osoresnontrine
  19. Buy Osoresnontrine | 1189767-28-9 | >98% - Smolecule, accessed September 24, 2025, https://www.smolecule.com/products/s521137
  20. Certificate of Analysis - ChemScene, accessed September 24, 2025, https://file.chemscene.com/pdf/COA/COACS33546.pdf
  21. Osoresnontrine 98% | CAS: 1189767-28-9 | AChemBlock, accessed September 24, 2025, https://www.achemblock.com/osoresnontrine.html
  22. Osoresnontrine Completed Phase 1 Trials for Alzheimer's Disease (AD) / Schizophrenia Treatment | DrugBank Online, accessed September 24, 2025, https://go.drugbank.com/drugs/DB16274/clinical_trials?conditions=DBCOND0049114%2CDBCOND0018504&phase=1&purpose=treatment&status=completed
  23. Osoresnontrine | PDE - TargetMol, accessed September 24, 2025, https://www.targetmol.com/compound/osoresnontrine
  24. Osoresnontrine (BI-409306) | PDE9A Inhibitor - MedchemExpress.com, accessed September 24, 2025, https://www.medchemexpress.com/BI-409306.html
  25. Phosphodiesterase 9 inhibitor - Wikipedia, accessed September 24, 2025, https://en.wikipedia.org/wiki/Phosphodiesterase_9_inhibitor
  26. Recent developments of phosphodiesterase inhibitors: Clinical trials, emerging indications and novel molecules - PMC - PubMed Central, accessed September 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9731127/
  27. Overview of Novel Antipsychotic Drugs: State of the Art, New Mechanisms, and Clinical Aspects of Promising Compounds - MDPI, accessed September 24, 2025, https://www.mdpi.com/2227-9059/13/1/85
  28. Baseline Demographics and Characteristics (TS) - ResearchGate, accessed September 24, 2025, https://www.researchgate.net/figure/Baseline-Demographics-and-Characteristics-TS_tbl1_324919905
  29. Osoresnontrine (BI-409306) | PDE9A Inhibitor | AmBeed.com, accessed September 24, 2025, https://www.ambeed.com/products/osoresnontrine.html
  30. Discovery of Effective Inhibitors Against Phosphodiesterase 9, a Potential Therapeutic Target of Alzheimer's Disease with Antioxidant Capacities - MDPI, accessed September 24, 2025, https://www.mdpi.com/2076-3921/14/2/123
  31. en.wikipedia.org, accessed September 24, 2025, https://en.wikipedia.org/wiki/Osoresnontrine#:~:text=Osoresnontrine%20(BI%2D409306)%20is,it%20increases%20memory%20in%20rodents.
  32. A Phase IC Study Evaluating the Safety, Tolerability, Pharmacokinetics, and Cognitive Outcomes of BI 409306 in Patients with Mild-to-Moderate Schizophrenia - PubMed, accessed September 24, 2025, https://pubmed.ncbi.nlm.nih.gov/29177699/
  33. TTD | Target: T06063 - Therapeutic Target Database, accessed September 24, 2025, https://db.idrblab.net/ttd/target/t06063
  34. Itraconazole Completed Phase 1 Trials for Healthy Lifestyle, accessed September 24, 2025, https://go.drugbank.com/drugs/DB01167/clinical_trials?conditions=DBCOND0059969&phase=1&purpose=treatment&status=completed
  35. Donepezil Completed Phase 2 Trials for Alzheimer's Disease (AD) Treatment - DrugBank, accessed September 24, 2025, https://go.drugbank.com/drugs/DB00843/clinical_trials?conditions=DBCOND0049114&phase=2&purpose=treatment&status=completed
  36. Clinical trials data Insight - Clival Database, accessed September 24, 2025, https://clival.com/product-table?graphSearchValue=Donepezil%3B+Osoresnontrine&graphClickVar=&fromPage=product&coloumnForGraph=product&extraInfo=tab&searchingType=1&searchCountry=US,EU,Japan,Canada,Korea,India,Aus/NZ
  37. Osoresnontrine | MedPath, accessed September 24, 2025, https://trial.medpath.com/drug/4fe5eab901ae17d5/osoresnontrine
  38. This Study Tests in Healthy Korean Women Which Effects BI 409306 and a Birth-control Pill Have on Each Other | Clinical Research Trial Listing - CenterWatch, accessed September 24, 2025, https://www.centerwatch.com/clinical-trials/listings/NCT03193307/this-study-tests-in-healthy-korean-women-which-effects-bi-409306-and-a-birth-control-pill-have-on-each-other?activelyRecruiting=false&page=16&id=848&slug=birth-control
  39. NCT03351244 | This Study Tests Whether BI 409306 Prevents Patients With Schizophrenia From Becoming Worse. This Study Looks at How Well Patients Tolerate BI 409306 and How Effective it is Over 6 Months | ClinicalTrials.gov, accessed September 24, 2025, https://clinicaltrials.gov/study/NCT03351244
  40. Study Details | NCT02337907 | BI 409306 in Patients With Cognitive Impairment Due to Alzheimer's Disease. | ClinicalTrials.gov, accessed September 24, 2025, https://clinicaltrials.gov/study/NCT02337907
  41. NCT02337907: A reported trial by Boehringer Ingelheim, accessed September 24, 2025, https://fdaaa.trialstracker.net/trial/NCT02337907/
  42. NCT02240693 | Alzheimer Disease Proof of Concept Study With BI 409306 Versus Placebo, accessed September 24, 2025, https://clinicaltrials.gov/study/NCT02240693
  43. Osoresnontrine Completed Phase 2 Trials for Schizophrenia, accessed September 24, 2025, https://go.drugbank.com/drugs/DB16274/clinical_trials?conditions=DBCOND0018504&phase=2&purpose=treatment&status=completed
  44. Poster Abstracts - CONICET, accessed September 24, 2025, https://bicyt.conicet.gov.ar/fichas/produccion/en/7997535

Published at: September 24, 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.