What are the therapeutic candidates targeting PDE10A?

11 March 2025
Introduction to PDE10A
Phosphodiesterase 10A (PDE10A) is a dual-substrate enzyme that plays a critical role in the regulation of intracellular cyclic nucleotides, namely cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Predominantly expressed in the medium spiny neurons (MSNs) of the striatum, PDE10A is a key modulator of the intracellular signaling cascades downstream of dopaminergic and glutamatergic receptors. As these neurons receive converging signals from dopaminergic inputs and cortical glutamatergic projections, PDE10A is uniquely positioned to act as a “gatekeeper” in modulating neuronal excitability and synaptic plasticity. Its selective and high expression in the striatum has made it an attractive target for therapeutic intervention in various neuropsychiatric and neurodegenerative disorders.

Biological Role and Significance
At the cellular level, PDE10A is responsible for hydrolyzing cAMP and cGMP, thereby terminating the signaling actions of these cyclic nucleotides. This regulation of signaling is fundamental to the modulation of movement, cognitive function, and overall neuronal health. In the striatum, the enzyme influences the balance between the direct and indirect pathways, which are mediated by dopamine D1 and D2 receptors, respectively. Inhibition of PDE10A can increase levels of cyclic nucleotides in MSNs, enhancing signaling through these pathways and potentially altering neuronal firing patterns with profound implications for motor control and behavior. This central role in neurotransmission has spurred significant research interest in the enzyme’s pharmacology, resulting in a detailed understanding of its structure, binding domain, and the mechanistic underpinnings of its function.

Relevance in Disease Pathology
The expression pattern and signaling function of PDE10A render it particularly relevant in the context of neuropsychiatric disorders such as schizophrenia and neurodegenerative conditions like Huntington’s disease. Abnormalities in PDE10A expression or activity have been associated with dysregulated dopamine signaling and altered striatal function, contributing to the pathophysiology of these disorders. For instance, in schizophrenia, disruptions in dopaminergic circuits and cortical-striatal interactions are thought to underlie the symptoms observed, and modulation of PDE10A has been shown in preclinical studies to produce antipsychotic-like effects. Additionally, in Huntington’s disease, where the degeneration of medium spiny neurons is a hallmark feature, PDE10A inhibition has been linked to neuroprotective outcomes, improvement in motor deficits, and even modulation of cortical pathology. The emerging relationship between PDE10A dysfunction and disease progression not only provides a scientific rationale for targeting this enzyme but also establishes a promising therapeutic window for PDE10A inhibitors across a range of CNS disorders.

Therapeutic Candidates Targeting PDE10A
The focus on PDE10A as a therapeutic target has resulted in the identification and development of several candidate compounds, ranging from early discovery molecules to advanced clinical-stage agents. These therapeutic candidates are primarily small-molecule inhibitors designed to modulate intracellular cyclic nucleotide levels by impeding PDE10A activity. With the goal of restoring the balance of cAMP and cGMP signaling in the brain, these agents have been evaluated for their potential antipsychotic, pro-cognitive, and neuroprotective effects, as well as for their capability to address motor dysfunction in diseases like Huntington’s.

Overview of Current Candidates
The landscape of PDE10A inhibitors includes a diverse array of compounds developed by both major pharmaceutical companies and smaller biotech entities. One of the earliest and most well-characterized candidates is TAK-063. This compound has been extensively studied in both preclinical and clinical settings. TAK-063 is known for its high potency and selectivity for PDE10A, and its pharmacodynamic profile suggests a robust modulation of striatal signaling. In addition, MK-8189 is another notable candidate that has been identified through rigorous fragment-based lead discovery and structure-guided optimization processes. Preclinical studies on MK-8189 have demonstrated that it is highly potent, exhibits favorable pharmacokinetics, and shows promise for improved symptomatic relief in schizophrenia by acting on the striatal network.

Further candidates include Gemlapodect, a compound under investigation for its potential to modulate intracellular signaling cascades via PDE10A inhibition. Gemlapodect has garnered attention due to its unique chemical structure and mode of action that contributes to modulating both cAMP and cGMP levels within the striatum. Other candidates emerging from discovery pipelines have been designed to achieve high selectivity over other phosphodiesterase (PDE) families to minimize off-target effects and adverse events. While some compounds such as MK-8189 and TAK-063 have reached advanced clinical phases, other candidates remain in earlier stages of development, with a focus on optimizing brain penetration, metabolic stability, and overall pharmacokinetic properties. Diagnostics agents such as 18F-MNI-654, while not therapeutic per se, are used to quantitatively assess PDE10A binding in vivo and support the therapeutic monitoring of PDE10A inhibitors in clinical trials.

Mechanisms of Action
The therapeutic action of PDE10A inhibitors revolves around the enzyme’s role in hydrolyzing cyclic nucleotides. By selectively blocking PDE10A, these compounds increase intracellular concentrations of cAMP and cGMP. This elevation in cyclic nucleotide levels leads to a cascade of downstream effects that enhance dopamine signaling in the direct pathway, while concurrently modulating the indirect pathway by influencing adenosine receptor signaling—specifically, enhancing adenosine A2A receptor-mediated signaling and suppressing dopamine D2 receptor signaling. The net effect of these molecular mechanisms is the restoration of signaling balance in striatal neurons, which is particularly beneficial in disorders where dopaminergic dysregulation plays a central role.

In addition, the inhibitory action on PDE10A supports neuroprotective mechanisms by influencing key transcriptional pathways including the cAMP response element-binding protein (CREB) signaling cascade. Increased CREB phosphorylation, as a result of PDE10A inhibition, has been correlated with elevated brain-derived neurotrophic factor (BDNF) levels, contributing further to neuroprotection and synaptic plasticity. These comprehensive mechanisms of action not only illustrate the biochemical efficacy of these compounds but also underscore their potential versatility in treating a spectrum of CNS disorders. Moreover, the structural studies using X-ray co-crystallography have shed light on the binding interactions between PDE10A and its inhibitors, helping inform the rational design of next-generation compounds with improved selectivity and potency.

Clinical Development and Trials
The translation of PDE10A inhibitors from the bench to clinical practice has been marked by a series of well-structured clinical trials aimed at evaluating safety, tolerability, and efficacy in human subjects. These clinical investigations are critical because they not only validate the preclinical findings but also help in determining optimal dosing regimens and therapeutic windows for treating CNS disorders.

Clinical Trial Phases and Results
Several PDE10A inhibitors have already been subjected to clinical trials, with TAK-063 and MK-8189 among the most prominent examples. TAK-063 has been advanced through Phase 1 trials, which demonstrated not only its excellent pharmacokinetic profile but also significant target engagement as verified by positron emission tomography (PET) imaging using specific radiotracers. In these early studies, TAK-063 exhibited robust striatal occupancy, clearly correlating with the intended inhibition of PDE10A activity. In Phase 1 studies, subjects receiving TAK-063 showed modulation of biomarkers related to antipsychotic activity, such as changes in blood oxygen-level-dependent (BOLD) signaling and electroencephalography (EEG) patterns, which further supported its mechanism of action.

MK-8189 has similarly progressed through early clinical phases, with results indicating a promising efficacy profile in terms of improving symptomatic measures in schizophrenia and related disorders. The clinical trial results of MK-8189 have reported the occurrence of adverse events at relatively low frequencies and a favorable safety margin, which is essential for therapies targeting the central nervous system. Moreover, PET studies conducted in the context of MK-8189 trials have provided valuable insights into the binding properties of the compound in human subjects, revealing a high degree of target engagement in key dopaminergic regions of the brain.

In addition to these two agents, Gemlapodect is another candidate that has been evaluated in preclinical and early clinical settings. While still in the earlier phases of clinical development, Gemlapodect has demonstrated promising pharmacological effects by altering the downstream signaling of PDE10A in animal models, which provides a strong rationale for its continued investigation in human trials. Although not as advanced as TAK-063 or MK-8189, the ongoing evaluation of other PDE10A inhibitors continues to underscore the clinical potential of targeting this enzyme in managing neuropsychiatric conditions.

It is also noteworthy that alongside these therapeutic candidates, diagnostic agents such as 18F-MNI-654 have been developed and are valuable for monitoring PDE10A expression in vivo during clinical trials. This dual strategy of therapeutic intervention and imaging biomarker development enhances the overall translational process, as it allows for the quantification of target engagement and helps to guide dose selection in early-phase clinical studies.

Safety and Efficacy Assessments
Safety and efficacy are of paramount importance when developing compounds for CNS disorders. In the case of PDE10A inhibitors, extensive assessments of adverse events, pharmacokinetics, and pharmacodynamics have been conducted during clinical trials. TAK-063, for example, exhibited a favorable safety profile with only a limited number of treatment-emergent adverse events (TEAEs) reported during the single-rising dose and multiple-dose studies. In trials involving subjects with stable schizophrenia, TAK-063 not only improved cognitive and behavioral parameters but also was well tolerated, exhibiting minimal extrapyramidal symptoms and negligible adverse effects on metabolic parameters.

Similarly, MK-8189 also has a strong safety profile based on early clinical data. The incidence of adverse events observed in these trials was relatively low, and the compounds maintained a broad therapeutic window. These safety assessments are further corroborated by preclinical toxicity studies that evaluated the effects of PDE10A inhibitors in animal models, where significant improvements in motor and cognitive functions were observed without pronounced off-target effects. The detailed analysis of adverse event rates, along with quantitative assessments such as the number of participants experiencing adverse events in both placebo-treated and compound-treated groups, has assisted in refining dosing regimens and optimizing the risk–benefit balance of these therapies.

The efficacy measures in clinical trials have been multifaceted and include both behavioral and neuroimaging endpoints. Functional magnetic resonance imaging (fMRI) and PET imaging studies have been instrumental in demonstrating target engagement and the consequent modulation of neural circuits involved in schizophrenia and Huntington’s disease. The studies indicate that the inhibition of PDE10A leads to improved connectivity in the striatum and prefrontal cortical regions, which is directly associated with symptomatic improvements in cognitive and motor functions. Moreover, assessments of electrophysiological parameters such as EEG have provided evidence that PDE10A inhibitors alter neural oscillatory patterns in a manner that is consistent with antipsychotic activity. These converging lines of evidence from various modalities underscore the clinical promise of PDE10A inhibitors and reinforce their role as a novel therapeutic strategy for central nervous system disorders.

Challenges and Future Directions
Despite considerable promise, the clinical development of PDE10A inhibitors is accompanied by several challenges that must be addressed to fully realize their therapeutic potential. Researchers and pharmaceutical developers have identified key issues ranging from issues of selectivity and brain penetration to the complex interplay of signaling networks in the striatal circuitry. Addressing these challenges is critical for the continued advancement of PDE10A-targeted therapeutics and for optimizing their use as monotherapy or in combination with other agents.

Current Development Challenges
One of the most significant challenges in the development of PDE10A inhibitors is achieving and maintaining a high level of selectivity over other phosphodiesterase subtypes. Given the structural similarities among phosphodiesterases, off-target inhibition can lead to unintended side effects, particularly when compounds inadvertently affect PDE families that regulate cardiovascular or inflammatory processes. Recent studies have focused on structure-based drug design to refine the binding pocket interactions of inhibitors, thereby enhancing selectivity while minimizing off-target effects. However, achieving a balance between potency, selectivity, and drug-like properties remains an ongoing challenge in the field.

Brain penetration represents another critical hurdle. CNS active molecules require the ability to cross the blood–brain barrier (BBB), and many PDE10A inhibitors have been optimized for this purpose. While compounds such as TAK-063 and MK-8189 have shown favorable brain penetration in preclinical and early clinical studies, the quest for improved pharmacokinetic profiles continues. Factors such as high clearance rates, rapid metabolism, and variable CNS exposure complicate dose optimization and may limit therapeutic efficacy if not properly addressed.

Furthermore, the complex neuropharmacology of the striatal network adds another layer of difficulty. PDE10A interacts with multiple signaling pathways in medium spiny neurons, and modulation of these pathways may have differential effects in the direct versus indirect pathways. This complexity necessitates careful titration of inhibitor dose to achieve the desired symptomatic outcome without triggering adverse dopaminergic imbalances. Such challenges are compounded by natural variations in patient populations, which may influence both the pharmacokinetics and pharmacodynamics of these agents.

Clinical trials themselves have encountered challenges in terms of demonstrating robust efficacy in heterogeneous patient populations. Early-phase trials often rely on biomarker endpoints and neuroimaging readouts, while later-phase studies must contend with the clinical heterogeneity inherent in disorders like schizophrenia. In some instances, despite promising preclinical data, the translation to clinical efficacy has been modest due to factors such as patient selection, study design limitations, and the complexity of CNS disorders.

Future Research and Development Opportunities
Looking forward, several promising avenues exist that could enhance the development of PDE10A inhibitors. Advances in medicinal chemistry and structure-based drug design continue to provide opportunities to improve selectivity, potency, and metabolic stability. High-resolution crystallographic studies, combined with computational modeling and fragment-based lead optimization, are likely to yield next-generation inhibitors with superior pharmacological profiles. These efforts can further tighten the structure–activity relationships (SAR) and guide modifications that enhance BBB penetration and prolong brain exposure.

Another promising research direction involves the integration of multi-modal diagnostic tools and biomarkers in clinical trials. The use of PET and fMRI imaging together with electrophysiological assessments (such as EEG) provides a comprehensive view of target engagement and neural circuit modulation. Quantitative measures of striatal occupancy and functional connectivity may serve as surrogate endpoints that improve the predictability of clinical outcomes. The incorporation of these biomarkers into clinical trial designs will not only help in dose optimization but also in personalized medicine approaches where patient stratification becomes key to identifying responders from non-responders.

In addition, understanding the differential roles of PDE10A in the direct and indirect striatal pathways could lead to more refined therapeutic strategies. There is potential for combination therapies that leverage the unique contributions of PDE10A inhibition alongside other neuromodulatory agents. For instance, combining PDE10A inhibitors with agents targeting dopamine receptors or with modulators of glutamatergic signaling might produce synergistic effects, enhancing both efficacy and safety profiles. Such combination regimens could be especially beneficial in patients with complex neuropsychiatric profiles where multiple dysregulated pathways contribute to disease pathology.

Emerging multi-target pharmacological strategies also represent an area of opportunity where PDE10A inhibitors could be combined with other agents to address broader aspects of CNS disorders. Recent computational studies have explored the possibility of designing multi-target ligands that simultaneously engage PDE10A and adenosine receptors (such as A1R and A2AR), thereby modulating cAMP signaling in a more comprehensive manner. This polypharmacological approach may prove to be particularly effective in treating complex neurodegenerative and psychiatric disorders, where a single target intervention might be insufficient.

Furthermore, the development of diagnostics and companion biomarkers will play an integral role in the future of PDE10A inhibitor research. The use of advanced imaging agents, such as 18F-MNI-654 and other PET radiotracers, allows for precise quantification of PDE10A expression in vivo and better monitoring of the pharmacodynamic effects of therapeutic candidates. These tools will be critical not only in clinical trials but eventually in routine clinical practice, to tailor therapy, monitor treatment response, and adjust doses in a personalized manner.

Finally, there is growing interest in exploring the role of PDE10A inhibitors beyond traditional indications such as schizophrenia and Huntington’s disease. Preclinical research suggests potential applications in other neurological conditions, such as Parkinson’s disease, where dysregulation of striatal signaling plays a significant role, as well as in certain cancers where aberrant cyclic nucleotide signaling contributes to tumorigenesis. The identification of novel indications and the stratification of patients based on biomarker profiles could considerably expand the therapeutic utility of PDE10A inhibitors in the future.

Conclusion
In summary, PDE10A plays a central role in modulating the signal transduction pathways in the striatum through its regulation of cAMP and cGMP levels. Its unique expression in medium spiny neurons and its influence on both direct and indirect dopaminergic pathways make it a critical target in the treatment of neuropsychiatric and neurodegenerative disorders. A number of promising therapeutic candidates targeting PDE10A have been identified, with TAK-063 and MK-8189 leading the field. These compounds function by inhibiting PDE10A activity, resulting in increased cyclic nucleotide signaling that restores the delicate balance of neuronal activity and produces beneficial antipsychotic, neuroprotective, and pro-cognitive effects. Extensive clinical development efforts have yielded encouraging results, with early phase trials demonstrating significant target engagement, favorable safety profiles, and early efficacy signals as measured by neuroimaging and electrophysiological biomarkers.

However, despite the progress made, several challenges remain. Issues of selectivity over other phosphodiesterase subtypes, achieving optimal blood–brain barrier penetration, and managing the complex interplay of dopaminergic signaling in heterogeneous patient populations require further research and innovative solutions. Future opportunities lie in refining the chemical structures of PDE10A inhibitors, integrating advanced diagnostic biomarkers into clinical trial protocols, and exploring combination therapies and multi-target strategies that could enhance overall therapeutic efficacy.

The trajectory of PDE10A inhibitor development reflects a classic progression from basic molecular understanding, through preclinical validation, to sophisticated clinical assessments using advanced imaging and biomarker measures. As the field moves forward, continued collaboration between medicinal chemists, pharmacologists, and clinicians—supported by robust computational tools and in vivo diagnostic technologies—will be essential for overcoming current challenges and fully realizing the therapeutic potential of PDE10A-targeted interventions. Ultimately, successful development of these agents could significantly enhance treatment options for patients with schizophrenia, Huntington’s disease, and possibly other CNS disorders, thereby contributing to improved patient outcomes and advancing the broader field of neurotherapeutics.

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.