Small Molecule
C16H17N5O2
1189767-28-9
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.
This section delineates the fundamental chemical and physical identity of Osoresnontrine, providing the necessary context for subsequent pharmacological and clinical evaluation.
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.
The compound is cataloged in major chemical and pharmacological databases under the following registry numbers:
Osoresnontrine is a complex heterocyclic compound characterized by a fused bicyclic pyrazolopyrimidinone core structure, which is a common scaffold in pharmaceutically active molecules.[19]
The physical properties of Osoresnontrine influence its handling, formulation, and behavior in biological systems.
Table 1: Key Identifiers and Physicochemical Properties of Osoresnontrine | ||
---|---|---|
Property | Value | Source(s) |
International Nonproprietary Name (INN) | Osoresnontrine | 16 |
Developmental Codes | BI-409306, SUB 166499 | 1 |
CAS Number | 1189767-28-9 | 16 |
DrugBank ID | DB16274 | 16 |
Molecular Formula | C16H17N5O2 | 16 |
Molecular Weight | 311.34 g/mol | 16 |
IUPAC Name | 1-(oxan-4-yl)-6-(pyridin-2-ylmethyl)-5H-pyrazolo[3,4-d]pyrimidin-4-one | 1 |
Solubility (DMSO) | 22-25 mg/mL (70.66-80.30 mM) | 23 |
Water Solubility (Predicted) | 0.481 mg/mL | 5 |
logP (Predicted) | 0.25 - 0.88 | 5 |
Rule of Five Compliance | Yes | 5 |
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.
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]
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.
Table 2: In Vitro Inhibitory Profile of Osoresnontrine Against PDE Isoforms | ||
---|---|---|
PDE Isoform | IC50 (nM) | Selectivity Ratio (vs. PDE9A) |
PDE9A | 52 | 1x (Reference) |
PDE1C | 1,000 | ~19x |
PDE1A | 1,400 | ~27x |
PDE2A, 3A, 4B, 5A, 6AB, 7A, 10A | >10,000 | >192x |
Data synthesized from sources.1 |
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.
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.
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.
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.
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.
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.
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.
Phase I studies in healthy volunteers established the fundamental ADME properties of Osoresnontrine.
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.
Table 3: Summary of Human Pharmacokinetic Parameters for Osoresnontrine by CYP2C19 Genotype | |||
---|---|---|---|
Parameter | Value in Extensive Metabolizers (EMs) | Value in Poor Metabolizers (PMs) | Fold-Difference (PM vs. EM) |
Tmax (Time to Peak Concentration) | <1 hour | <1 hour | No significant difference |
t1/2 (Elimination Half-Life) | 0.99 - 2.71 hours | Within a similar range as high doses in EMs | ~1-1.5x |
Cmax (Peak Concentration) | Dose-dependent | 2.2 - 2.3x higher than EMs at the same dose | ~2.3x |
AUC (Total Exposure) | Dose-dependent | 4.1 - 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 |
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.
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:
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.
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) | Phase | Status | Indication(s) | Patient Population | Primary Endpoint(s) | Key Outcome |
NCT01343706 | 1 | Completed | Healthy Volunteers | Healthy Male EMs & PMs | Safety, Tolerability, PK | Generally safe; PK highly dependent on CYP2C19 genotype |
NCT01493570 | 1 | Completed | Healthy Volunteers | Healthy Male Volunteers | PK/PD (cGMP in CSF) | Confirmed dose-dependent increase of cGMP in human CSF |
NCT02240693 | 2 | Completed | Alzheimer's Disease | Patients with Prodromal AD | Change in NTB z-score | Failed to show superiority over placebo |
NCT02337907 | 2 | Completed | Alzheimer's Disease | Patients with Mild AD | Change in NTB z-score | Failed to show superiority over placebo |
NCT02281773 | 2 | Completed | Schizophrenia | Patients with Cognitive Impairment | Change in MCCB composite score | Failed to meet primary endpoint; no cognitive improvement |
NCT03351244 | 2 | Terminated | Schizophrenia | Patients on Antipsychotics | Prevention of Relapse | Terminated (COVID-19); did not meet primary endpoint |
NCT03230097 | 2 | Terminated | Psychotic Disorders | Attenuated Psychosis Syndrome | Efficacy and Tolerability | Terminated |
Table synthesizes data from multiple sources including.2 |
The core of the Alzheimer's program consisted of two sister studies:
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]
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]
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.
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.
This study tested a different therapeutic hypothesis: whether adjunctive treatment with Osoresnontrine could help maintain stability and prevent the recurrence of psychotic symptoms.
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.
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.
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.
Early-phase trials in healthy subjects are designed to establish initial safety, tolerability, and pharmacokinetic parameters.
Safety data from the larger Phase II trials in patients with Alzheimer's disease and schizophrenia confirmed the patterns observed in healthy volunteers.
Table 5: Summary of Most Frequently Reported Adverse Events with Osoresnontrine | |
---|---|
System Organ Class | Specific Adverse Events |
Eye Disorders | Photophobia (light sensitivity), Photopsia (perceived flashes of light), Chromatopsia (distorted color vision), Blurred vision, Abnormal sensation in eye, Eye pain |
Nervous System Disorders | Headache, Dizziness |
Events compiled from Phase I and II trial data reported in sources.7 |
The most distinctive and consistently reported AEs associated with Osoresnontrine were transient visual disturbances.
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.
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.
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.
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.
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.
Published at: September 24, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.