What are the preclinical assets being developed for PDE10A?

11 March 2025
Introduction to PDE10A
Phosphodiesterase 10A (PDE10A) is a dual-substrate enzyme that hydrolyzes both cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), though it shows a substrate preference for cAMP because of its unique internal hydrogen bonding network. PDE10A is predominantly expressed in the striatum, especially in the medium spiny neurons that form the core circuitry of the basal ganglia. As such, it plays a critical role in modulating dopaminergic neurotransmission and influencing motor control, cognitive processes, and signal transduction in neural tissues. Early studies elucidated that inhibition of PDE10A results in an increase in intracellular cAMP and cGMP levels, leading to enhanced activation of protein kinase A (PKA) and downstream signaling cascades. This central role in cyclic nucleotide modulation underscores the enzyme’s significance as a therapeutic target in several central nervous system (CNS) disorders.

Biological Role and Significance
At the molecular level, PDE10A acts as a critical regulator of intracellular signaling by degrading the second messengers cAMP and cGMP. This regulatory function not only influences neuronal excitability and synaptic plasticity but also modulates the activity of other receptors linked to dopamine signaling. The enzyme’s restricted distribution in the brain, mostly within the striatum, ensures that its physiological impact is primarily confined to modulating processes such as motor control, reward, and cognition. Given this high level of selectivity, PDE10A occupies a unique niche among phosphodiesterases that makes it an attractive candidate for selectively modulating dopaminergic and glutamatergic transmission without affecting peripheral systems.

PDE10A in Disease Context
Dysregulation of PDE10A has been associated with a range of neurological and psychiatric disorders. In conditions such as schizophrenia, Huntington's disease, and Parkinson's disease, abnormal PDE10A activity can disrupt the delicate balance of neuronal signaling in the basal ganglia, leading to both motor and cognitive deficits. Preclinical data have highlighted that inhibition of PDE10A can restore or modulate dysfunctional signaling pathways, making it a promising target for therapeutic intervention in disorders that exhibit an imbalance in cAMP/cGMP homeostasis. Furthermore, some studies have also explored the enzyme’s role in non-neurological conditions, such as certain forms of cancer, where altered cyclic nucleotide signaling may contribute to tumorigenesis. This dual role has spurred significant interest in developing selective and potent inhibitors that can modulate PDE10A activity for various therapeutic applications, emphasizing the enzyme's relevance in both neurological and systemic disease states.

Current Preclinical Assets
Over the past years, a diverse array of chemical entities targeting PDE10A has been developed preclinically. Improvements in medicinal chemistry, structure-based drug design, and in silico modeling have allowed researchers to identify novel scaffolds, optimize binding affinities, and assess selectivity profiles that are critical for successful drug candidates. These preclinical assets vary from small-molecule inhibitors with high enzymatic potency (often measured in the subnanomolar to nanomolar range) to radiolabeled compounds used for positron emission tomography (PET) imaging. By combining chemical synthesis with advanced biological assays, several compounds have emerged as promising preclinical candidates with potential for clinical translation.

Overview of PDE10A Inhibitors
Preclinical research on PDE10A inhibitors has revealed a rich chemical diversity. Many compounds exhibit structural classes that include pyrazolopyrimidines, cinnoline analogues, benzimidazole derivatives, and maleimide-based scaffolds. These classes have been developed using approaches that range from high throughput screening (HTS) and fragment-based lead discovery (FBLD) to rational structure-based drug design. For instance, early discovery efforts identified compounds like MP-10, which have set the standard for potency and selectivity in PDE10A inhibition by demonstrating subnanomolar enzyme inhibition and favorable brain penetration properties. Moreover, scaffolds based on cinnoline and benzimidazole frameworks have also emerged, showing potent inhibitory activity in vitro with IC₅₀ values ranging from 1.5 to 18.6 nM and extremely high selectivity over other phosphodiesterase isoforms. These chemical series not only provide potent pharmacological tools for probing PDE10A biology but also represent potential leads for further clinical development.

In parallel, radioligands for PET imaging have been developed as a means to quantify target engagement and pharmacokinetics of PDE10A inhibitors in vivo. Radiolabeled compounds such as [18F]JNJ-42259152 allow for noninvasive assessment of drug distribution and occupancy in the brain, which is crucial for bridging preclinical findings to human studies. Such imaging agents are essential for understanding the regional distribution of PDE10A and for evaluating the pharmacodynamic response following inhibitor administration.

Key Compounds in Development
Several key compounds in the preclinical pipeline exhibit promising characteristics in terms of potency, selectivity, and pharmacokinetics. Notable examples include:

• MP-10: One of the earliest and most extensively studied PDE10A inhibitors, MP-10 has demonstrated potent inhibition with IC₅₀ values in the subnanomolar range. It has been used as a benchmark compound in several preclinical evaluations, showing excellent brain penetration and target occupancy in animal models. Research using MP-10 has contributed immensely to establishing structure-activity relationships (SAR) that inform subsequent designs.

• TAK-063: This compound is another preclinical asset that has shown exceptional promise. TAK-063 was designed to achieve high inhibitory potency and extraordinary selectivity for PDE10A—with reported IC₅₀ values around 0.30 nM—and has been assessed using in vitro autoradiography to confirm its selective binding in key brain regions (such as the striatum and nucleus accumbens). Moreover, TAK-063 demonstrates favorable pharmacokinetic profiles, including effective brain penetration and predictable target occupancy, making it a strong candidate for further development in CNS disorders.

• Cinnoline and Benzimidazole Analogues: A series of compounds based on cinnoline and benzimidazole scaffolds have been synthesized, with several showing promising in vitro inhibitory activity against PDE10A. Among these, compounds such as those labeled 26a, 26b, and 33c demonstrate nanomolar potency and high selectivity over other PDE isoforms, thus representing a promising series for further preclinical evaluation. These compounds have been characterized through X-ray crystallography studies to elucidate their binding modes, thereby supporting rational design strategies for optimization.

• Novel Maleimide Derivatives: New chemical entities based on maleimide cores have been identified as allosteric inhibitors of PDE10A. These derivatives are specifically designed to target the GAF domain of PDE10A, representing an alternative mechanism of inhibition that may circumvent some of the limitations associated with active-site inhibitors. Docking studies have shed light on stabilizing interactions—such as hydrogen bonding with key amino acids—and these compounds are being evaluated further for their inhibitory potential and drug-like properties.

• Compound 39 from the 6,7-Dimethoxy-4-(pyridin-3-yl)cinnoline Series: This compound has emerged as a potent inhibitor with single-digit nanomolar activity, and its development addresses challenges related to in vivo clearance by incorporating structural modifications that enhance metabolic stability. The optimized compound shows potential for further translational studies in rodent behavioral models and may lead to future clinical candidates if safety and efficacy profiles are confirmed.

• Multitarget Ligands: Beyond single-target PDE10A inhibitors, research is also exploring multi-target compounds that simultaneously modulate PDE10A and other key neuronal receptors such as A1 and A2A receptors. This approach, representing an innovative paradigm in drug design, aims to produce synergistic effects that may be beneficial for complex neurodegenerative and psychiatric disorders. Preliminary studies using computational modeling and subsequent synthesis have validated a series of 2-aminopyridine-3-carbonitrile derivatives as promising multitarget agents, showing inhibition of PDE10A with simultaneous activity at adenosine receptors.

Together, these compounds form a robust preclinical asset portfolio that targets PDE10A from multiple angles—from traditional competitive inhibition at the enzyme’s catalytic site to allosteric modulation at regulatory domains, and from monotherapy agents to multitarget ligands intended to harness synergistic clinical benefits.

Mechanisms of Action
Understanding the mechanisms by which PDE10A preclinical assets exert their effects is foundational to their optimization and eventual clinical application. These assets are designed to modulate key molecular pathways that hinge upon cyclic nucleotide signaling in neuronal tissues.

Molecular Pathways Involved
PDE10A is a critical molecular gatekeeper in the degradation of cAMP and cGMP, both of which are essential secondary messengers in a multitude of neuronal processes. By inhibiting PDE10A, these preclinical assets effectively elevate intracellular levels of cAMP and cGMP. This elevation enhances the activation of protein kinase A (PKA) and other downstream kinases that regulate diverse cellular functions—ranging from gene transcription to neuronal plasticity and synaptic transmission. The net result is often an enhancement of dopaminergic and glutamatergic signaling through indirect modulation of receptor activity. Specific studies have demonstrated that PDE10A inhibitors can normalize aberrant signaling pathways in basal ganglia circuits, which are often dysregulated in conditions such as schizophrenia and Huntington’s disease.

Another essential aspect of the mechanism of action is the modulation of β-catenin levels. In some contexts, PDE10A inhibition has been linked to the suppression of oncogenic β-catenin-dependent transcriptional activity, an effect that may provide therapeutic benefits in non-CNS diseases such as colon tumorigenesis. Thus, these assets work through multiple molecular pathways that involve regulation of cyclic nucleotides, ultimately translating into physiological responses at the cellular and tissue levels.

Pharmacodynamics and Pharmacokinetics
The pharmacodynamic profiles of preclinical PDE10A assets are characterized by high binding affinity, robust inhibition of enzyme activity, and resultant increases in intracellular cyclic nucleotide levels. For example, TAK-063 has been shown to produce significant occupancy of PDE10A in key brain regions at relatively low doses, correlating with marked improvements in animal behavioral models relevant to schizophrenia. Such compounds are not only potent inhibitors in vitro but have been optimized for rapid and sustained effects in vivo. Preclinical pharmacokinetic assessments have focused on ensuring adequate brain penetration, stable plasma concentrations, and minimal off-target effects. Techniques such as PET imaging with radiolabeled ligands—like [18F]JNJ-42259152—provide critical data on the temporal and spatial distribution of these assets in the brain, further guiding dosage and formulation strategies.

Moreover, many preclinical assets have been engineered to overcome challenges inherent to CNS drug delivery. Structural modifications intended to increase lipophilicity while maintaining high-affinity binding have led to compounds with improved oral bioavailability and blood–brain barrier penetration. Studies employing in vivo animal models have yielded valuable insights into parameters such as clearance rates, half-life, and metabolic stability, with some compounds (e.g., Compound 39 in the cinnoline series) demonstrating enhancements in metabolic stability and reduced high in vivo clearance. This dual optimization for pharmacodynamics and pharmacokinetics is critical, as it provides a platform for translating preclinical success into clinical efficacy.

Potential Therapeutic Applications
PDE10A inhibitors are primarily being developed to address disorders of the central nervous system, particularly those that involve dysfunction within the basal ganglia circuits. Preclinical assets targeting PDE10A have the potential not only to alleviate motor and cognitive deficits but also to modulate psychiatric symptoms.

Neurological Disorders
In the realm of neurological disorders, the therapeutic potential of PDE10A inhibitors is largely tied to their ability to restore the balance of cyclic nucleotide signaling within the striatum. Disorders such as Huntington's disease and Parkinson's disease are characterized by abnormalities in basal ganglia circuitry—often manifesting as dyskinesia, rigidity, and bradykinesia. Preclinical studies using compounds like MP-10 and TAK-063 have demonstrated the capacity to modulate these circuits by increasing cAMP/cGMP levels, thereby enhancing PKA-mediated signaling and normalizing motor function in rodent models. Additionally, preclinical assets have been tested in models of dystonia and other hyperkinetic movement disorders, where selective modulation of PDE10A activity leads to improvements in motor output and basal ganglia communication.

The development of radiolabeled PDE10A inhibitors also supports the evaluation of these compounds in diseases characterized by progressive striatal degeneration. By enabling the tracking of drug occupancy and distribution, these compounds can help refine dosing regimens and provide early indicators of therapeutic efficacy in neurodegenerative conditions. Overall, preclinical assets in this space offer hope for striking a balance between efficacy and tolerability in disorders that are currently difficult to treat.

Psychiatric Disorders
Schizophrenia represents one of the most promising areas for the application of PDE10A inhibitors. Abnormalities in dopaminergic signaling and dysregulated intracellular cyclic nucleotide levels have been implicated in the pathophysiology of schizophrenia. Preclinical studies indicate that compounds such as TAK-063 and MP-10 can reduce hyperlocomotion and ameliorate behavioral deficits in rodent models mimicking schizophrenia symptomatology. In these models, PDE10A inhibition helps to normalize dopamine receptor-mediated signaling and improve the performance of tasks related to cognitive and negative symptoms.

Furthermore, given the excellent selectivity and controlled pharmacokinetics of the preclinical assets, they may offer a therapeutic profile that minimizes side effects typical of conventional antipsychotic drugs (such as extrapyramidal symptoms or metabolic disturbances). The combination of multitarget ligand strategies—where PDE10A inhibition is paired with modulation of adenosine receptors—suggests that future compounds might provide broad-spectrum benefits, potentially addressing both positive and negative symptoms of schizophrenia while also improving cognitive deficits.

Challenges and Future Directions
Despite the encouraging preclinical data and the diverse portfolio of PDE10A inhibitors under development, several challenges remain that must be addressed before these compounds can transition successfully into clinical applications. Research continues to uncover gaps in our understanding of the structure–activity relationships, metabolic pathways, and off-target effects associated with these compounds.

Current Research Gaps
One of the primary challenges is translating the potent in vitro inhibition of PDE10A into consistent in vivo efficacy with minimal side effects. Although many compounds exhibit subnanomolar IC₅₀ values and excellent selectivity in preclinical assays, issues such as metabolic instability or poor brain penetration have restricted the progression of some assets beyond the preclinical stage. Furthermore, the diversity in binding modes across different chemical series sometimes leads to unpredictable off-target interactions or adverse pharmacokinetic outcomes that need to be carefully characterized. For instance, while compounds like TAK-063 show remarkable selectivity, others deployed from the maleimide series or multipharmacophore approaches may encounter variability in allosteric binding modes that complicate drug optimization and predictability.

Additionally, the use of PET imaging agents—although a powerful tool—requires further refinement. Standardizing kinetic models and ensuring reproducibility between whole-cell preparations and membrane studies remain areas for improvement. Such discrepancies can affect the accurate measurement of target engagement and drug occupancy, making it harder to draw definitive conclusions about dosing and efficacy. Moreover, the inherent plasticity of the basal ganglia circuitry and the potential for compensatory mechanisms in chronic drug administration are areas that require further investigation to understand long-term treatment outcomes.

Another research gap lies in the need for better elucidation of the downstream effects of PDE10A inhibition. While many compounds have been shown to elevate cyclic nucleotide levels, the precise molecular and transcriptional cascades that are activated—and their eventual impact on neuronal survival, plasticity, and behavior—still need further exploration. Detailed systems-level analyses, including genomic and proteomic studies, could shed light on the long-term implications of PDE10A inhibition in diverse neural and non-neural tissues.

Future Research Opportunities
Future research into PDE10A inhibitors should prioritize the integration of advanced in silico modeling, high-throughput screening, and structure-based drug design to refine and diversify chemical scaffolds. Current efforts can be expanded by employing techniques such as molecular dynamics simulations and free-energy perturbation calculations to predict subtler aspects of binding affinity, metabolism, and off-target activity. Novel scaffolds—such as advanced maleimide derivatives that target allosteric sites—as well as multitarget ligands that combine PDE10A inhibition with modulation of A1 and A2A receptors, will likely represent the next generation of candidates for neuropsychiatric indications.

In addition, further optimization of pharmacokinetic properties is essential. Improving oral bioavailability and ensuring sufficient penetration of the blood–brain barrier are recurring themes in preclinical asset development. Methods such as prodrug approaches, incorporation of specific lipid moieties, and nanotechnology-based drug delivery systems may provide innovative solutions to these challenges. Simultaneously, employing rigorous in vivo evaluations to monitor parameters like clearance, half-life, and metabolic stability will help in selecting the most promising candidates for clinical trials.

Enhanced imaging tools and biomarkers also represent a major opportunity for advancing PDE10A research. The development of more refined radioligands not only aids in early target engagement assessments but also provides a platform for personalized medicine strategies. By correlating imaging data with genetic and clinical metrics, researchers can identify patient subgroups that are most likely to benefit from PDE10A inhibitors. Such approaches could mitigate the high placebo effects and variability observed in earlier clinical trials, as seen in other classes of CNS drugs.

Moreover, there is a growing opportunity to study the long-term effects of PDE10A inhibition on neurodevelopment and neuroprotection. Preclinical studies should be extended to understand how chronic inhibition of PDE10A might impact neuronal circuitry, synaptic plasticity, and compensatory responses. Collaborative efforts between pharmacologists, neuroscientists, and clinicians will be imperative to develop longitudinal studies and validate the safety profile of these assets, thereby laying a robust groundwork for future clinical trials.

Finally, an unmet area in current research is the exploration of combination therapies. Given that PDE10A inhibitors modulate key pathways implicated in both motor and psychiatric conditions, combining these compounds with other agents—such as immune checkpoint modulators, neuroprotective agents, or even conventional antipsychotics—could yield synergistic benefits that exceed the efficacy of monotherapy. Such strategies may not only address individual symptoms more effectively but also help in minimizing side effects by allowing for lower effective doses.

Conclusion
In summary, the preclinical asset landscape for PDE10A inhibitors is both broad and dynamic. The continuous refinement of chemical scaffolds—from the well-established agents like MP-10 and TAK-063 to novel cinnoline, benzimidazole, and maleimide derivatives—reflects a deepening understanding of the enzyme’s biology and its role in brain function. Initially identified as critical regulators of cyclic nucleotide signaling in striatal medium spiny neurons, PDE10A continues to attract significant attention, mainly in the context of neurological disorders such as Huntington’s and Parkinson’s diseases as well as psychiatric conditions like schizophrenia. Preclinical assets demonstrate remarkable potency, often achieving IC₅₀ values in the subnanomolar range, and are being optimized for selectivity and favorable pharmacokinetic profiles that ensure adequate blood–brain barrier penetration and sustained drug exposure.

The mechanisms of action underlying these assets are multifaceted, involving the elevation of intracellular cAMP and cGMP levels and the stimulation of downstream signaling cascades that modulate neurotransmission, motor control, and even transcription factor activity (e.g., β-catenin suppression). This intricate interplay of molecular pathways supports the therapeutic rationale behind PDE10A inhibition, especially given its localized expression in regions that are crucial for dopaminergic signaling. Radiolabeled PET imaging agents add an extra dimension by offering noninvasive methods to track drug occupancy and distribution, thereby facilitating the translation of preclinical results into clinical contexts.

Despite these promising advances, several challenges remain. Key research gaps include the variability in binding modes across different scaffolds, issues with metabolic stability, and the need for precise biomarkers to evaluate long-term pharmacodynamic effects. Future directions should focus on enhancing structure–activity relationships using advanced computational techniques, improving drug delivery systems, and exploring synergistic combination therapies to maximize therapeutic potential while minimizing adverse effects. Additionally, refining imaging methodologies and integrating multi-target approaches can further advance the field, offering hope for more effective treatment options for both neurological and psychiatric disorders.

Ultimately, the preclinical assets being developed for PDE10A represent a robust and promising foundation for the next generation of CNS therapeutics. With continued innovation in medicinal chemistry and a comprehensive understanding of the molecular mechanisms involved, these assets have the potential to address unmet clinical needs in disorders characterized by aberrant basal ganglia signaling. The strategic combination of potent enzyme inhibitors, optimized pharmacokinetics, and advanced imaging techniques paves the way for future clinical successes that may transform the therapeutic landscape for diseases such as schizophrenia, Huntington’s disease, Parkinson’s disease, and beyond. The integration of these diverse perspectives—from molecular biology to pharmacodynamics, from rigorous preclinical testing to advanced imaging—will be essential to overcoming current barriers and fully realizing the therapeutic promise of PDE10A inhibition.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.