What are the new molecules for M4 receptor agonists?

11 March 2025
Introduction to M4 Receptors
M4 muscarinic acetylcholine receptors (mAChRs) are members of the family of G protein‐coupled receptors (GPCRs) that respond to the neurotransmitter acetylcholine. Their pharmacology has attracted increasing attention due to their involvement in complex brain functions and psychiatric disorders, such as schizophrenia. An improved understanding of their structure and function has allowed researchers to develop more selective molecules aimed at modulating M4 receptor activity while avoiding off‐target effects associated with other subtypes like M2 and M3. This review‐style answer is structured to introduce basic receptor characteristics, then detail the discovery of new molecules with selective agonism at M4, discuss their therapeutic applications, and finally evaluate the challenges and future directions of this research area.

Structure and Function
M4 receptors belong to the muscarinic receptor subfamily that generally couples to the Gi/o protein, leading to inhibition of adenylyl cyclase activity and modulation of ion channels. Structurally, M4 receptors share a high sequence homology with other mAChR subtypes and feature the characteristic seven transmembrane domains of GPCRs. Unlike M1, M3, and M5 subtypes that predominantly signal through Gq/11 proteins, M4 receptors are located largely presynaptically and are crucial in modulating neurotransmitter release via the inhibition of calcium channels or activation of potassium channels. The similarity of the orthosteric binding pocket in muscarinic receptors poses a significant challenge for achieving subtype selectivity; however, the development of allosteric modulation strategies and the identification of novel chemical scaffolds have enabled researchers to design molecules with a higher selectivity for M4 receptors.

Role in the Central Nervous System
Within the central nervous system (CNS), M4 receptors are predominantly found in regions such as the striatum, hippocampus, and neocortex. Their location enables them to modulate dopaminergic neurotransmission—a key pathway implicated in neuropsychiatric disorders. For example, M4 receptors have been shown to regulate dopamine release in the basal ganglia and the ventral tegmental area, thereby affecting motor control and reward systems. In preclinical models, reduced M4 receptor function has been associated with hyperdopaminergic phenotypes and psychosis‐like behaviors, indicating that selective activation of M4 receptors holds promise for antipsychotic effects and potential cognitive improvement in conditions like schizophrenia. Moreover, selective activation of M4 receptors is believed to mitigate the side effects observed when nonselective muscarinic agonists are used, as activation of M2 and M3 receptors can lead to cardiovascular and gastrointestinal adverse effects.

Discovery of New M4 Receptor Agonists
Advancements in receptor structural biology and computational modeling have provided unprecedented insights into the binding modes of ligands at M4 receptors. This has led to the discovery and optimization of new chemical entities that show potent and selective M4 receptor agonistic activity. The approaches range from the use of allosteric modulation strategies to the careful design of orthosteric agonists with novel scaffolds that preferentially stabilize conformations of the M4 receptor.

Current Research and Development
Recent research has been primarily focused on developing molecules that overcome the intrinsic challenges linked to achieving selectivity at the M4 receptor. Since the orthosteric region is highly conserved among muscarinic receptors, researchers have turned their attention to exploiting structure–activity relationship (SAR) studies and innovative scaffolding techniques. Two major classes of new molecules are emerging:
1. Novel N‐Substituted Oxindoles
2. Molecules bearing Novel Carbamate Isosteres

The discovery of these molecules has been facilitated by advances in high‐resolution receptor structures and state‐of-the-art in silico drug design. For example, the use of allosteric binding site models has provided a robust framework to rationalize how subtle modifications in ligand structure can result in significant selectivity improvements. These advancements have been supported by multiple studies that describe not only the binding interactions but also the functional outcomes in preclinical models.

Leading Molecules and Their Characteristics
One leading candidate emerging from recent studies is a series of novel N‐substituted oxindole compounds that function as partial agonists selective for both M1 and M4 receptors. Among these, one representative compound, often referred to as “compound 1” in the literature, has demonstrated potent activity and excellent CNS penetration in preclinical models. These oxindoles have been shown to reverse psychosis‐like behaviors in animal models, with low propensity to cause peripheral side effects that are typically mediated by other muscarinic receptor subtypes. The significance of this discovery lies in the fact that the N‐substitution pattern on the oxindole scaffold confers both receptor selectivity and the appropriate physicochemical properties for blood–brain barrier penetration, making them promising for further clinical development.

In parallel, another series of compounds has been developed based on novel carbamate isosteres. These molecules are specifically designed to enhance M4 receptor selectivity by altering the electrostatic potential isosurface that is critical for receptor activation. The introduction of carbamate isosteres into the pharmacophore results in a subtle reorientation of molecular interactions within the receptor binding pocket. This modification is believed to promote a ligand–receptor interaction mode that is optimal for M4 receptor activation while reducing the likelihood of cross‐activation of M2/M3 receptors. Detailed SAR studies have demonstrated that small molecular changes, such as variations in the substituents on the carbamate moiety, can significantly alter potency, efficacy, and pharmacokinetic profiles of these compounds.

Another molecule that has attracted attention in the broader context of M1/M4 receptor pharmacology is xanomeline. Although xanomeline itself is not entirely new, it has undergone several modifications—such as pairing with trospium—to enhance tolerability by mitigating peripheral side effects. However, xanomeline represents a prototype for a class of molecules targeting M1/M4 receptors in which further medicinal chemistry efforts are being applied to produce even more selective M4 agonists. The ongoing development of such dual agonists illustrates the translational potential of M4 modulators in psychiatric indications.

Overall, the emerging molecules can be summarized with the following key points:
• N‐Substituted Oxindoles: These molecules offer selective partial agonism at M1 and M4 receptors and have demonstrated efficacy in reversing psychosis‐like behaviors in animal models with good CNS penetration. Their design heavily relies on modifications of the oxindole core to optimize receptor interactions.
• Carbamate Isosteres: This class of compounds has been specifically engineered to enhance M4 receptor selectivity through the introduction of novel isosteres. These molecules not only exhibit potent receptor activation but also possess favorable metabolic stability and pharmacokinetics, making them exciting candidates for further development.
• Prototype Molecules like Xanomeline: Though older, xanomeline has informed much of the current research strategy towards dual M1/M4 receptor activation, inspiring new chemotypes that aim to achieve improved selectivity and reduced side effects.

Therapeutic Applications of M4 Agonists
The advances in selective M4 receptor agonist molecules are driven largely by their promising therapeutic potential in various CNS disorders. Given the critical role of the M4 receptor in modulating dopaminergic signaling and neural network excitability, these new molecules have significant implications for treating neuropsychiatric conditions such as schizophrenia and related cognitive disorders.

Potential Medical Uses
M4 receptor agonists are being developed with the expectation that they may offer a novel mechanism for treating psychosis and cognitive deficits in disorders like schizophrenia. Preclinical evidence supports the hypothesis that M4 receptor activation can normalize dopaminergic hyperactivity, a pathological hallmark observed in schizophrenic patients. In animal models, selective M4 receptor activation has demonstrated antipsychotic‐like efficacy by reducing abnormal locomotor activity and psychostimulant‐induced behaviors. In addition, given that the M4 receptor modulates the signaling of dopamine in regions implicated in cognitive functions such as the striatum and hippocampus, M4 agonists could potentially improve cognitive performance, which is an unmet need in current antipsychotic therapies.

Beyond schizophrenia, there is also interest in M4 receptor agonists for other CNS disorders where cholinergic dysfunction plays a key role. This includes Alzheimer’s disease and Parkinson’s disease, where cognitive impairment and dysregulated motor control, respectively, are significant clinical issues. The selective activation of M4 receptors—without the off-target activation of M2/M3 receptors—could result in antipsychotic effects with a lower incidence of adverse effects related to gastrointestinal and cardiovascular systems.

In some cases, dual-acting M1/M4 receptor agonists are being evaluated for their potential to address both psychotic and cognitive symptoms concurrently. As the muscarinic system influences numerous aspects of brain function, a carefully balanced activation of these receptors can modulate neurotransmitter pathways, resulting in a therapeutic outcome that surpasses what is currently achievable with conventional antipsychotic drugs.

Clinical Trials and Studies
Recent clinical studies have begun to translate these preclinical successes into the human setting. One notable example is the clinical development of compounds like emraclidine, which is considered to be an M1/M4 receptor agonist. Emraclidine has progressed into early phase clinical trials and has shown promising results in terms of efficacy and a tolerable safety profile. Preliminary data indicate reductions in positive and negative symptoms of schizophrenia, along with improvements in cognitive measures, confirming the relevance of M4 receptor modulation in a clinical setting.

Furthermore, extensive research in preclinical animal models has demonstrated that the new molecules—specifically those based on the N‐substituted oxindole and carbamate isostere scaffolds—can reverse psychostimulant-induced behavioral abnormalities. Studies have highlighted that these molecules not only engage the central M4 receptors effectively but also do so with a reduced propensity to induce extrapyramidal side effects—a critical factor for antipsychotic agents. Although these molecules are still in early stages of development, their favorable pharmacokinetic properties, such as metabolic stability and adequate brain penetration, are encouraging from a clinical perspective.

In addition to receptor activation studies, the research community has employed state-of-the-art in vitro assays—such as calcium mobilization and cAMP signaling assays— to profile the activity, potency (EC50 values), and selectivity of these compounds across the muscarinic receptor subtypes. Such assays have confirmed that modifications in the chemical structure directly influence the functional activity at M4 receptors and have provided important insights into the receptor-ligand interaction mechanics. The success of these preclinical validations has consequently paved the way for further clinical evaluations, with several molecules anticipated to enter phase I/II studies in the near future.

Challenges and Future Directions
Despite the significant progress in identifying promising molecules for M4 receptor activation, several challenges remain that need to be addressed to bring these agents from bench to bedside. Critical issues include achieving robust subtype selectivity, mitigating off-target effects, and ensuring that in vivo pharmacokinetic profiles are suitable for long-term therapeutic use.

Developmental Challenges
One of the primary challenges in developing M4 receptor agonists is the high sequence and structural homology shared among muscarinic receptor subtypes. This overlap makes it difficult to design ligands that are both potent and selective for M4 without inadvertently activating M2 or M3 receptors, which are typically associated with undesirable peripheral side effects. For instance, while xanomeline has provided proof-of-concept data for M1/M4 agonism, its clinical utility was hampered by side effects such as gastrointestinal disturbances and cardiovascular effects due to nonselective receptor activation.

Another challenge lies in the process of optimizing the pharmacokinetic Profile of new molecules. Molecules must possess sufficient metabolic stability, low potential for off-target covalent binding, and robust permeability through the blood–brain barrier. The dynamic nature of the receptor conformation during activation also complicates the optimization process. Moreover, ensuring that the newly designed molecules maintain a favorable balance between efficacy and safety is crucial; even slight deviations in molecular modifications can lead to unexpected toxicities or reduced therapeutic windows.

Additionally, the reliance on preclinical animal models, while invaluable, presents translational challenges. Differences in receptor expression, signaling pathways, and pharmacodynamic responses between animal models and humans mean that promising preclinical candidates might not always exhibit the same therapeutic profiles in clinical trials. For example, while the selective N‐substituted oxindoles and carbamate isosteres have shown great promise in rodent models, extensive safety and efficacy evaluations in humans are still required before these compounds can be considered viable therapeutic agents.

Future Research and Innovations
Looking forward, continued integration of structure-based drug design, high-throughput screening, and advanced SAR analyses will be essential in refining M4 receptor agonists. The recent success in identifying novel scaffolds such as the N‐substituted oxindoles and carbamate isosteres highlights the potential of these methodologies when combined with detailed receptor structural data derived from techniques like cryo-electron microscopy. Future research will likely focus on several fronts:

• Refinement of molecular scaffolds to further enhance selectivity and efficacy: Researchers will continue to tweak the chemical structure of these molecules to improve binding affinity to the M4 receptor while ensuring that adverse effects are minimized. Novel substituent patterns, changes in stereochemistry, and the exploration of additional isosteric replacements may provide further gains in selectivity.

• Development of allosteric modulators: In parallel with orthosteric agonists, there is a growing interest in positive allosteric modulators (PAMs) that enhance the activity of the endogenous ligand acetylcholine only when it is present. This “only when needed” mode of action could reduce the risk of receptor desensitization and side effects while providing fine-tuned control over receptor activation. Such strategies have already been demonstrated in other GPCR systems and are being actively explored for M4 receptors.

• Application of computational modeling and machine learning: With the availability of detailed structural information, computational approaches can predict optimal ligand conformations and dynamic interactions within the receptor binding pocket. Using machine learning models trained on existing SAR data may accelerate the iterative cycle of design, synthesis, and testing for new M4 agonists.

• Investigation of combination therapies: Considering that muscarinic receptor agonism (particularly through dual M1/M4 agents) shows promise for complex CNS disorders, future research might explore combination regimens that include selective M4 agonists alongside other therapeutic modalities. For example, combining M4 agonists with agents that target dopamine receptors or with cognitive enhancers might yield synergistic effects, especially in multifactorial diseases like schizophrenia.

• Long-term safety studies and translational research: As many of the newly discovered molecules progress into clinical stages, it will be essential to monitor long-term safety and efficacy. Particular attention will be given to any signs of receptor desensitization, tolerance development, or peripheral off-target effects. Establishing robust pharmacodynamic biomarkers in early-phase clinical trials will be crucial in ensuring that the therapeutic benefits outweigh potential risks.

• Addressing patient heterogeneity: Advances in pharmacogenomics can aid in identifying patient subgroups that might benefit most from M4 receptor agonist therapy. Since psychiatric disorders like schizophrenia manifest with considerable heterogeneity, personalized medicine approaches that tailor treatment based on genetic makeup and receptor expression patterns could optimize therapeutic outcomes.

Detailed Summary and Conclusion
In summary, new molecules for M4 receptor agonists have emerged primarily from two innovative chemical scaffolds: one based on novel N‐substituted oxindoles and the other on compounds featuring novel carbamate isosteres. These molecules are designed to overcome the longstanding challenge of achieving high selectivity for the M4 receptor given the conserved orthosteric binding sites among muscarinic receptor subtypes. The N‐substituted oxindoles act as partial agonists with confirmed efficacy in reversing psychosis-like behaviors in animal models, while the carbamate isostere derivatives have been optimized to stabilize the receptor in a conformation conducive to M4 activation. Together, these discoveries address the dual challenge of potency and selectivity while also providing favorable pharmacokinetic attributes such as good brain penetration and metabolic stability.

From a general perspective, M4 receptors play a critical role in modulating neuronal signaling, particularly by influencing dopaminergic transmission in brain regions that are dysregulated in neuropsychiatric disorders. This makes them attractive targets for novel therapeutic interventions, especially in conditions such as schizophrenia where traditional dopamine antagonists have limited efficacy and can lead to undesirable side effects. The new molecules exploit advanced drug design principles and SAR studies, which have enabled medicinal chemists to design ligands that not only bind selectively to the M4 receptor but also produce the desired functional responses with minimal off-target activity.

More specifically, the development of these new M4 agonists reflects an important evolution in our understanding of muscarinic receptor pharmacology. The identification of novel chemical motifs such as N‐substituted oxindoles and carbamate isosteres has allowed researchers to modulate receptor activity through both orthosteric and allosteric mechanisms. Preclinical studies have demonstrated that these compounds are capable of reversing hyperdopaminergic states in animal models, providing a solid rationale for their continued development. Moreover, the translational potential of these molecules is underlined by their promising early clinical data, which indicate that they might offer a new class of antipsychotic agents with improved safety profiles compared to nonselective muscarinic agonists like xanomeline.

On a general-specific level, while the initial focus remains on understanding the basic receptor structure and function in the CNS, recent efforts in molecular design are directly contributing to the pipeline of new therapeutic agents. Effective modulation of the M4 receptor requires an intricate balance: robust activation sufficient to achieve antipsychotic and cognitive enhancing effects, yet selective enough to avoid unwanted peripheral effects. Advances in using machine learning and computational modeling, along with high-throughput screening methodologies, are expected to further optimize these molecules. From this perspective, the research into N‐substituted oxindoles and carbamate isostere derivatives represents a promising convergence of academic insight and applied medicinal chemistry, with strong potential to benefit patients suffering from complex neuropsychiatric disorders.

Finally, considering future directions, ongoing research must now address the translational challenges that come with moving from preclinical successes to clinical efficacy. This involves detailed safety assessments, long-term pharmacokinetic studies, and the exploration of combination therapies that may harness the synergistic potential of modulating multiple neurotransmitter systems simultaneously. There is also the opportunity to integrate personalized medicine strategies—using pharmacogenomics to predict which patients may respond best to M4 receptor agonists—thus paving the way for more tailored and effective therapeutic regimens.

In conclusion, the development of new molecules for M4 receptor agonists—exemplified by the novel N‐substituted oxindoles and carbamate isostere compounds—represents a significant breakthrough in the field of neuropsychiatric drug discovery. These molecules not only offer unprecedented receptor selectivity and favorable pharmacokinetic profiles but also hold the promise of revolutionizing the treatment landscape for disorders such as schizophrenia and potentially other CNS diseases. With continued research and clinical evaluation, these new molecules are set to become key components in next-generation therapies, combining the power of advanced chemistry and deep biological insight to meet major unmet medical needs.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成