Hostname: page-component-54dcc4c588-9xpg2 Total loading time: 0 Render date: 2025-10-01T13:12:23.249Z Has data issue: false hasContentIssue false

Biased agonism in psychopharmacology: an opportunity to improve efficacy and safety of treatments

Published online by Cambridge University Press:  12 August 2025

Gia Han Le
Affiliation:
Poul Hansen Family Centre for Depression, University Health Network, Toronto, ON, Canada Brain and Cognition Discovery Foundation, Toronto, ON, Canada Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
Sabrina Wong
Affiliation:
Poul Hansen Family Centre for Depression, University Health Network, Toronto, ON, Canada Brain and Cognition Discovery Foundation, Toronto, ON, Canada Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
Stavroula Bargiota
Affiliation:
Department of Psychiatry, Aristotle University of Thessaloniki, Thessaloniki, Greece
Swainson Jennifer
Affiliation:
Department of Psychiatry, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
Heidi K.Y. Lo
Affiliation:
Department of Psychiatry, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
Diana Orsini
Affiliation:
Poul Hansen Family Centre for Depression, University Health Network, Toronto, ON, Canada Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
Kayla Teopiz
Affiliation:
Poul Hansen Family Centre for Depression, University Health Network, Toronto, ON, Canada Brain and Cognition Discovery Foundation, Toronto, ON, Canada Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
Hernan F. Guillen-Burgos
Affiliation:
Pontificia Universidad Javeriana, Department of Psychiatry and Mental Health, Hospital Universitario San Ignacio, Bogota DC, Colombia Faculty of Medicine, Center for Clinical and Translational Research, Universidad El Bosque, Bogota DC, Colombia Universidad Simon Bolivar, Center for Clinical and Translational Research, Barranquilla, Colombia
Poh Khuen Lim
Affiliation:
Poul Hansen Family Centre for Depression, University Health Network, Toronto, ON, Canada
Roger S. McIntyre*
Affiliation:
Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada Department of Psychiatry, University of Toronto, Toronto, ON, Canada
*
Corresponding author: Roger S. McIntyre; Email: roger.mcintyre@bcdf.org
Rights & Permissions [Opens in a new window]

Abstract

G protein-coupled receptors (GPCRs) are involved in many physiological and pathophysiological processes. Conventional pharmacological models categorize the typology of pharmacologic ligands as agonists or antagonists. Biased agonism is a relatively newer pharmacodynamic characteristic that has potential to optimize therapeutic efficacy while minimizing adverse effects in psychiatric and neurological treatments. We conducted a narrative literature review of articles obtained from PubMed, Embase, and MEDLINE from inception to April 2025, focusing on pharmacologic antagonism (i.e., competitive, noncompetitive, uncompetitive) and agonism (i.e., full, partial, inverse, superagonism, biased). Primary and secondary articles defining these concepts were included, provided they addressed pharmacologic (rather than chemical) antagonism and agonism. Distinct mechanisms of antagonism and agonism were identified, each contributing nuanced receptor modulation beyond the conventional models. Notably, biased agonism facilitates targeted intracellular signaling (e.g., G protein- versus β-arrestin–mediated). Use cases demonstrate relatively greater efficacy (e.g., incretin receptor agonist, tirzepatide) and improved safety (e.g., serotonergic psychedelics, opioids). Biased agonism provides a potential avenue for future drug development, with emerging preclinical evidence suggesting potential to differentially activate intracellular pathways and thereby improve efficacy and safety profiles of psychopharmacologic agents—pending clinical validation. Future research vistas should aim to rigorously assess the long-term outcomes of biased agonism, explicitly addressing individual variability in receptor signaling and therapeutic response.

Information

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press

Introduction

G protein-coupled receptors (GPCRs) are large transmembrane proteins composed of seven transmembrane domains.Reference Katritch, Cherezov and Stevens1 GPCRs share common yet diverse signal transduction mechanisms which are implicated in physiology, pathophysiology, and pharmacology.Reference Katritch, Cherezov and Stevens1, Reference Insel, Sriram and Gorr2 The heterogeneity in signal transduction effects related to GPCRs introduces complexity as well as opportunity for pharmacological discovery and development.Reference Katritch, Cherezov and Stevens1, Reference Kurose and Kim3 It is currently estimated that over 35% of Food and Drug Administration (FDA)-approved pharmacologic agents target GPCRs, with over 150 GPCRs currently identified as being druggable.Reference Insel, Sriram and Gorr2, Reference Sriram and Insel4

Pharmacological ligands (i.e., endogenous or synthetic ligands) bind to the GPCR extracellular domains, consequently triggering intracellular cascades which have implications for the treatment of disease processes.Reference Katritch, Cherezov and Stevens1, Reference Rosenbaum, Rasmussen and Kobilka5, Reference Kenakin6 Notwithstanding the complexity of GPCR signaling, conventional pharmacological models have typically reduced the typology of pharmacologic ligands as either an agonist (i.e., a ligand that bind and activates a receptor to promote a conformational change that increases receptor-mediated signaling) or antagonists (i.e., a ligand that binds to without activating the receptor thus blocking/diminishing the effect of an agonist).Reference Kurose and Kim3, Reference Kenakin6Reference Sum, Murphy, Li, Markossian, Grossman and Arkin8 While the agonist–antagonist paradigm is well characterized, other activities at GPCRs and subsequent effects on GPCR signaling cannot be parsimoniously reduced to agonism/antagonism.Reference Kenakin6

Emerging preclinical and/or preliminary evidence suggests that certain pharmacologic ligands, conventionally classified as agonists, can selectively (i.e., in a biased manner) activate a specific GPCR signaling pathway as opposed to contemporaneously activating multiple signaling cascades.Reference Berg and Clarke7, Reference Michel and Charlton9Reference Chisamore, Kaczmarek and Le12 Notwithstanding, clinical validation of these findings remains necessary. Consequently, pharmacologic ligands exerting biased agonism have the potential to enhance beneficial therapeutic outcomes (e.g., greater weight loss with incretin receptor agonists and neuroplastic changes) while avoiding the activation of pathways that mediate adverse drug reactions (e.g., psychedelic experiences, nausea).Reference Katritch, Cherezov and Stevens1, Reference Kenakin6, Reference Chisamore, Kaczmarek and Le12

The overarching aim herein is to not only provide a critical evaluation but also a rationale for exploring whether biased agonism could serve as a theoretical framework to guide future pharmacological discoveries and developments, with potentially improved efficacy and/or safety, for the treatment of psychiatric conditions. It is of note that while compelling preclinical data exists, caution is warranted in extrapolating these findings to clinical populations until robust human evidence becomes available.

Methods

We conducted a narrative review of articles published from inception to April, 2025. A search was conducted on literature databases including PubMed, Embase, and MEDLINE databases. The following search string was utilized for the search of relevant articles in the foregoing databases: (“agonist” OR “agonism” OR “full agonism” OR “partial agonism” OR “inverse partial agonist” OR “superagonism” OR “biased agonism” OR “antagonism” OR “antagonism” OR “competitive antagonism” OR “noncompetitive antagonism” OR “uncompetitive antagonism” OR “partial antagonism” OR “functional antagonism”). Furthermore, efficacy and safety examples of biased agonists and their effect on the discovery and development of pharmacologic agents were also searched.

The following eligibility criteria were employed during the screening process conducted by two independent reviewers (G.H.L. and S.W.). Primary research articles including human, animal, and in vitro studies were included. Secondary articles including, but not limited to, systematic reviews and meta-analyses were only included to define and characterize the different types of agonism and antagonism. Conclusions and findings from the foregoing secondary articles were not included when outlining preliminary evidence in support of a pharmacological agents’ efficacy or tolerability. Only articles referring to nonphysical/chemical antagonists were included. Furthermore, articles focused solely on chemical structure, unrelated to intracellular signaling or therapeutic implications, were excluded.

A purposive selection strategy was used to highlight representative agents, for each pharmacological ligand typology, that highlight mechanistic diversity and translational relevance in psychopharmacology. Article selection was informed by citation frequency, mechanistic clarity and clinical relevance. Due to the inherent flexibility of a narrative approach, no formal quality assessment or data synthesis was undertaken. Notwithstanding, to maintain rigor, we included all evidence when discussing efficacy, safety and tolerability of the representative agents included.

Pharmacologic antagonism and agonism: definitions

Antagonism

Pharmacologic antagonism is defined as a process wherein a pharmacologic agent binds to and inhibits the actions of a native agonist via interactions at a common receptor.Reference Kurose and Kim3, Reference Kenakin13, Reference Bardal, Waechter and Martin14 There are three main principle mechanisms wherein pharmacologic agents antagonize native ligand activity at the receptor: competitive, noncompetitive, and uncompetitive antagonism.

Competitive antagonism

Competitive antagonists are pharmacologic agents that compete with a native ligand without activation of the receptor. Consequently, subsequent GPCR-mediated activation of signal transduction cascades is reduced and/or abrogated.Reference Williams15, Reference Zhu16 Competitive antagonists are categorized as either competitive reversible or competitive irreversible antagonists.

Competitive reversible antagonists compete with a native ligand to bind to its canonical receptor. The occupancy of the receptor is a function of both the pharmacologic agent’s concentration and its affinity to the receptor.Reference Stephenson17Reference Kenakin19 There are a finite number of receptor sites, which implies that as the concentration of the pharmacologic antagonist increases, there is a greater formation of antagonist–receptor complexes and a reduction in agonist–receptor complexes.Reference Bardal, Waechter and Martin14, Reference Ferner and Aronson18, Reference Kenakin19 In addition, a higher number of antagonist–receptor complexes would be expected as a function of higher receptor affinity by the pharmacologic antagonist. However, if the agonist concentration is sufficiently increased, the agonist can outcompete the antagonist for receptor binding to elicit maximal response (Emax).Reference Bardal, Waechter and Martin14

For example, olanzapine is an atypical antipsychotic that is a competitive, reversible antagonist at various receptor sites including, but not limited to, dopamine (D2) and serotonin 5-HT2A receptors.Reference Nadeem, Riaz and Hosawi20 By binding noncovalently to the foregoing receptors, olanzapine prevents endogenous ligands (e.g., dopamine and serotonin) from binding, which reduces excessive dopaminergic and serotonergic signaling, contributing to its therapeutic effects in schizophrenia and bipolar depression.Reference Thomas and Olanzapine21

Competitive irreversible antagonism refers to a scenario wherein the pharmacologic antagonist competes with the native agonist for the target receptor; however, the robustness of the intermolecular interaction effectively confers antagonism insofar as the covalent bonds formed between the agent and the ligand are irreversible.Reference Bardal, Waechter and Martin14, Reference Lista and Sirimaturos22 Consequently, the effects of competitive irreversible antagonists will remain constant irrespective of endogenous agonist levels.Reference Bardal, Waechter and Martin14

Noncompetitive antagonism

Noncompetitive antagonism refers to the process wherein the pharmacologic antagonist does not directly compete with the native agonist for the identical binding site; however, it will impair the ability of an agonist to bind or to activate the receptor through steric and/or allosteric mechanisms.Reference Bardal, Waechter and Martin14, Reference Arias, Bhumireddy and Bouzat23 Steric (orthosteric) noncompetitive antagonism typically involves the antagonist binding to the same site as the agonist, consequently blocking activation via an irreversible or covalent interaction.Reference Bardal, Waechter and Martin14 Specifically, the antagonist remains bound to the receptor thus removing the receptor from the pool of receptors available for activation by an agonist. Allosteric noncompetitive antagonism occurs when the antagonist binds to a different (allosteric) site on the receptor, from the orthosteric site, consequently changing the receptor’s conformation to prevent activation by the agonist.Reference Arias, Bhumireddy and Bouzat23, Reference Delaune and Alsayouri24

Ketamine is an allosteric noncompetitive N-methyl-d-aspartate receptor (NMDAR) antagonist.Reference Jelen, Young and Stone25, Reference Zorumski, Izumi and Ketamine26 Blockade of NMDARs on γ-aminobutyric acid (GABA)-ergic inhibitory interneurons by ketamine leads to disinhibition of pyramidal cells, resulting in a glutamate surge.Reference Jelen, Young and Stone25, Reference Krystal, Abdallah, Sanacora, Charney and Duman27 Although ketamine does not occupy the glutamate-binding site and therefore does not prevent glutamate from binding to the orthosteric site on NMDARs, it binds within the receptor’s ion channel pore. This interaction prevents ion flow and impedes the activation of GABAergic interneurons.Reference Bardal, Waechter and Martin14, Reference Jelen, Young and Stone25, Reference Mion and Villevieille28 Glutamate binds to and activates postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), which is believed to play a key role in ketamine’s antidepressant effects.Reference Jelen, Young and Stone25, Reference Autry, Adachi and Nosyreva29, Reference Yang, Shirayama and Jc30

Uncompetitive antagonism

Similar to noncompetitive antagonism, uncompetitive antagonism also involves an agonist that binds to an allosteric site on the receptor, separate from an agonist’s binding site; however, antagonist–receptor binding only occurs postreceptor activation, during which the receptor pore is open.Reference Smith, Rao and Velázquez-Sánchez31, Reference Traynelis, Wollmuth and McBain32

Memantine, FDA-approved in the treatment of moderate-to-severe Alzheimer’s disease, is an uncompetitive antagonist of NMDARs.Reference Chen and Lipton33, Reference Lipton34 Rather than binding at the orthosteric site, memantine preferentially binds within the open, activated ion channel and blocks excessive calcium influx.Reference Lipton34, Reference Xia, Chen, Zhang and Lipton35 Memantine’s blockade of calcium influx reduces excitotoxicity in Alzheimer’s disease while still allowing for physiological neurotransmission to occur.Reference Folch, Busquets and Ettcheto36, Reference Puranik and Song37 Separately, dextromethorphan is also an uncompetitive antagonist and modulates glutamate signaling of NMDARs.Reference McCarthy, Bunn, Santalucia, Wilmouth, Muzyk and Smith38 The binding of dextromethorphan to activated NMDARs results in the inhibition of excessive excitatory neurotransmission, and ultimately reduced excitotoxicity and disrupted synaptic plasticity, which may contribute to depressive symptoms in depressive disorders.Reference McCarthy, Bunn, Santalucia, Wilmouth, Muzyk and Smith38

Agonism

In select disease states, a physiological system may be insufficiently active, providing the basis for pharmacological agents to increase activity of the system.Reference Kurose and Kim3 The foregoing pharmacological agents exhibit receptor agonism wherein binding to the receptor results in receptor activation.Reference Tate39 There are different types of receptor agonism, including full, partial, inverse, and superagonism. Another type of receptor agonism is biased agonism, which will be discussed in a separate section.

Full agonism

Full agonism refers to a substance or agent that mimics the effects of an endogenous ligand.Reference Berg and Clarke7 In this case, the agent binds to the orthosteric binding site and activates the physiological system to the same degree as the endogenous ligand, a maximal response.Reference Watts, Townsend and Neubig40 Consequently, pharmacological agents that bind to a receptor and activate it to produce a biological response, mimicking the maximal response induced by an endogenous ligand (e.g., neurotransmitter), are widely used in pain management and have high addiction potential.Reference Edinoff, Kaplan and Khan41

Morphine is an example of a full agonist which binds to and activates μ-opioid receptors (MORs) to induce profound analgesia—a property that, while therapeutically valuable, also underlies its significant side-effect profile.Reference Dhaliwal and Gupta42Reference Ricarte, Dalton and Giraldo44 Separately, methadone (MTD) is also a full MOR agonist. Racemic methadone ((R,S)-MTD) consists of two enantiomers, (R)-MTD and (S)-MTD, wherein both exhibit full MOR agonism to produce analgesia; however, they differ in their abuse potential.Reference Levinstein, De Oliveira and Casajuana-Martin45 Recent evidence indicates that compared to (R)-MTD, (S)-MTD does not robustly stimulate the dopaminergic reward pathway in the ventral tegmental area (VTA); therefore, exhibiting lower reinforcing efficacy in rats.Reference Levinstein, De Oliveira and Casajuana-Martin45 In contrast, (R)-MTD exhibits greater efficacy on dopaminergic signaling activation and was associated with reliable self-administration in rats.Reference Levinstein, De Oliveira and Casajuana-Martin45 The findings indicate that the abuse liability of (R,S)-MTD is mediated by (R)-MTD instead of (S)-MTD.Reference Levinstein, De Oliveira and Casajuana-Martin45 The foregoing phenomenon highlights that while full MOR agonism is often associated with elevated abuse liability, differences in agonist–receptor interactions at specific brain regions may modulate the risk profile of different full agonists.

Partial agonism

Similar to full agonism, partial agonism also refers to a pharmacological agent that binds to the orthosteric site on the receptor. Partial agonists activate the receptor to increase the activity of the system, but only with partial efficacy compared to a full agonist or the endogenous ligand that elicits a maximal response.Reference Sandilands and Bateman46 This approach can be advantageous when a specific physiological outcome needs to be controlled, as seen with certain antipsychotics (e.g., aripiprazole, brexpiprazole, cariprazine) or pain medication (e.g., buprenorphine).Reference Mohr, Masopust and Kopeček47Reference Ragguett and McIntyre51

For example, the partial agonism of aripiprazole at dopamine and serotonin receptors allows for the balancing of neurotransmitter activity in both hyper and hypodopaminergic states.Reference Tuplin and Holahan52Reference Sciascio and Riva54 The dual action of aripiprazole addresses both positive and negative symptoms in schizophrenia as well as both depressive and manic poles of bipolar disorder and may lead to fewer side effects than are common with traditional antipsychotics.Reference McIntyre, Soczynska, Woldeyohannes, Miranda and Konarski53, Reference Lieberman55, Reference de Bartolomeis, Tomasetti and Iasevoli56

Inverse agonism

The observation that receptors may be activated in the absence of a native ligand led to the discovery of pharmacologic agents that can reduce constitutive receptor activity. Costa and Herz (1989) conducted a study of wild type, endogenously expressed delta opioid receptors in NG108-15 neuroblastoma cell membranes, and found that several ligands, previously thought to be antagonists, decreased GTPase activity stimulated by these receptors.Reference Costa and Herz57 Since their effects opposed those of agonists, these ligands were classified as inverse agonists.

While agonists are characterized by intrinsic efficacy, or the ability to enhance receptor activity, inverse agonists show negative intrinsic activity. Similar to how the intrinsic efficacy of agonists varies depending on their structure, leading to distinctions between strong and weaker (partial) agonists, the negative intrinsic efficacy of inverse agonists can also be characterized as strong or weak (partial) inverse agonists.Reference Berg and Clarke7

A range of antipsychotic medications exert their therapeutic effects through antagonism at dopamine D2 and serotonin 5-HT2A receptors. The role of the 5-HT2A receptor in the pathophysiology of psychosis has been underscored by the psychotomimetic effects of serotonergic hallucinogens such as LSD or psylocybin, that act as agonists at the 5-HT2A receptor.Reference López-Giménez and González-Maeso58, Reference Vollenweider, Vollenweider-Scherpenhuyzen, Bäbler, Vogel and Hell59 This observation provided the basis for the hypothesis that 5-HT2A antagonism could be a viable target for antipsychotic development.Reference Zhang and Stackman60 However, clinical trials involving selective 5-HT2A antagonists—notably volinanserin—failed to demonstrate sufficient efficacy in schizophrenia populations, leading to the discontinuation of such compounds in late-stage development.Reference Casey, Cui, Booth and Canal61

More recently, the 5-HT2A inverse agonist pimavanserin (Nuplazid) was FDA approved in the treatment of Parkinson’s disease (PD) psychosis.Reference Cummings, Isaacson and Mills62 Pimavanserin acts as an inverse agonist and antagonist at serotonin 5-HT2A receptors (Ki 0.087 nM) and 5-HT2C receptors (Ki 0.44 nM). Primavanserin exhibits low binding to sigma 1 receptors (Ki 120 nM) and negligible affinity (Ki > 300 nM) for 5-HT2B, dopaminergic (i.e., D2), muscarinic, histaminergic, adrenergic receptors, and calcium channels.Reference Muneta-Arrate, Diez-Alarcia, Horrillo and Meana63 Unlike typical antipsychotics, pimavanserin does not interfere with dopaminergic signaling pathways, which poses an advantage for individuals vulnerable to motor side effects.Reference Muneta-Arrate, Diez-Alarcia, Horrillo and Meana63, Reference Meltzer, Cao, Schad, King, Stoll and Standley64 Notwithstanding, by acting as an inverse agonist at 5-HT2ARs, primavanserin reduces phosphoinositide signaling thus downregulating 5-HT2A-driven excitatory signaling to dampen psychotic symptoms (e.g., hallucinations and delusions) associated with PD psychosis.Reference Rissardo, Durante, Sharon and Fornari Caprara65

Superagonism

Superagonism refers to the phenomenon wherein a ligand not only activates a receptor but can also induce a greater maximal effect than endogenous ligands or full agonists.Reference Schrage, De Min, Hochheiser, Kostenis and Mohr66, Reference Brown67 This occurs when the ligand’s intrinsic efficacy is greater than that of endogenous neurotransmitters or hormones; therefore, superagonists are able to activate the receptor to a functional level that surpasses what occurs normally under physiological conditions.Reference Schrage, De Min, Hochheiser, Kostenis and Mohr66, Reference Smith, Bennett and Milligan68

Notwithstanding the efficacy of superagonists, it is also associated with greater risk of side effects or potential receptor desensitization as a result of overstimulation of targeted pathways.Reference Miess, Gondin and Yousuf69, Reference Lowe, Sanderson and Cooke70 For example, isotonitazene is a synthetic opioid that is a superagonist of μ-opioid receptors (MORs). Compared to other opioids such as morphine, hydromorphone and fentanyl, isotonitazene demonstrates greater MOR signaling efficacy; therefore, it exhibits greater potency and overall efficacy for reducing pain (isotonitazene > fentanyl; F(1,26) = 8.25, p = 0.008).Reference Malcolm, Palkovic and Sprague71, Reference Vandeputte, Van Uytfanghe, Layle, St Germaine, Iula and Stove72 It is of note that preclinical findings from extant literature also indicate that isotonitazene’s superagonism is associated with greater and prolonged respiratory depression than fentanyl.Reference Malcolm, Palkovic and Sprague71 The foregoing example underscores the importance of balancing therapeutic potency and safety considerations, which highlights a broader principle relevant to biased agonism wherein targeted efficacy must be carefully weighed against potential adverse outcomes and their severities.

Biased agonism

Contemporary mathematical and pharmacological studies indicate that GPCRs are highly dynamic with the ability of adopting multiple structural conformations and signaling states.Reference Hilger73, Reference Calebiro, Koszegi, Lanoiselée, Milijus and O’Brien74 Depending on the ligand that binds to the GPCR, both G protein-mediated and β-arrestin pathways may be activated or one pathway may be preferentially stimulated over the other (Figure 1).Reference Eiger, Pham, Gardner, Hicks and Rajagopal75, Reference Lamichhane, Liu and White76 Preferential activation of a particular pathway is known as biased agonism, also referred to as functional selectivity.Reference Michel and Charlton9 Biased and selective agonism are distinct, wherein the latter refers to receptor selectivity rather than preferential pathway activation after a ligand binds.Reference Sum, Murphy, Li, Markossian, Grossman and Arkin8 This phenomenon allows for a more targeted modulation of distinct intracellular responses and treatment of clinical symptoms.Reference Michel and Charlton9, Reference Ehlert77Reference Chang and Bruchas79 Biased agonism is a well-established principle in GPCR research and various GPCR families have been studied including, serotonergic, opioid, adrenergic, cannabinoid, muscarinic, and metabotropic glutamate receptors.Reference Chisamore, Kaczmarek and Le12, Reference Yang, Zhou and Laborska80Reference Lamberts and Traynor82

Figure 1. G-protein-coupled receptor-biased agonism. Left: Balanced agonist that activates both the G-protein- and β-arrestin-mediated pathways. Middle: G-protein-biased agonist that selectively activates the G-protein intracellular pathway. Right: βarrestin-biased agonist that selectively activates the β-arrestin intracellular pathway. Created in BioRender. Le, G. (2025). https://BioRender.com/3tmy5ld.

Extant literature indicates that ligands exhibiting biased agonism may be a novel avenue to achieve targeted symptom relief while minimizing additional side effects. For example, current data indicate that functionally selective ligands at opioid receptors are able to achieve pain relief without the normally associated abuse liability, dysphorigenic properties, or psychomimetic components of ligands that traditionally bind at these sites.Reference Bruchas and Chavkin83, Reference Bruchas, Schindler and Shankar84 In addition, recent pharmacological advances indicate that engaging D2R arrestin-biased signaling via GSK3β or 5HT2A arrestin-biased Src/Akt signaling may enable more targeted treatments for schizophrenia, psychosis, and other mood disorders.Reference Schmid and Bohn85Reference Urs, Bido and Peterson87 Preclinical evidence suggests that biased agonism may present a novel strategy for potentially enhancing both therapeutic efficacy and safety to achieve better patient tolerability. However, robust clinical studies are essential to confirm these hypothesized advantages in patient populations.

Opportunity for enhancing efficacy with biased agonism

Preclinical studies suggest that regulating downstream effectors such as GSK3β may contribute to achieving antipsychotic effects without impairing motor function.Reference Schmid and Bohn85Reference Urs, Bido and Peterson87 In psychiatry, biased ligands targeting dopamine D2 receptors—selectively activating arrestin-dependent pathways—have been explored to decrease extrapyramidal symptoms primarily induced by conventional antipsychotics. While promising in preclinical settings, rigorous clinical validation remains to be necessary to determine real-world applicability.

Biased agonism has also shown potential in metabolic and neuroendocrine contexts. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are widely prescribed for treating type 2 diabetes mellitus (T2DM) and obesity.Reference Kurtzhals, Flindt Kreiner and Singh Bindra88 Preliminary evidence from secondary analyses and observational studies suggests that GLP-1 RAs may exert indirect benefits for mood, cognitive, and substance-related clinical outcomes, largely through improvements in metabolic parameters (e.g., weight management and glycemic modulation).Reference Badulescu, Tabassum and Le89Reference Cooper, Ramachandra and Ceban93 However, clinical trials in psychiatric populations directly measuring the foregoing clinical symptom outcomes at endpoints independent of metabolic changes are necessary to confirm these potential effects. Notwithstanding, there remain concerns regarding suicidality among persons treated with GLP-1 RAs; however, evidence of causality has not been established.Reference McIntyre94 Evidence from extant literature indicates that metabolic disturbances, including obesity and untreated/poorly maintained diabetes, may predispose individuals to and exacerbate psychiatric symptoms (e.g., depression, anxiety), which emphasizes the importance of careful monitoring.Reference Segal and Gunturu95, Reference Busili, Kumar, Kudrna and Busaily96 Recent evidence in older adults and animal models indicates that GLP-1 RAs exert antidepressant effects independent of glycemic control.Reference Chen, Zhao, Wang, Guo and Pan97, Reference Camkurt, Lavagnino, Zhang and Teixeira98 Supporting the foregoing findings, Gunturu et al. (2024) similarly highlighted the promise of GLP-1 RAs for psychiatric treatment, specifically in improving mood and cognitive functioning.Reference Gunturu99 It is hypothesized that the therapeutic effects that GLP-1s may exert across dimensions of psychopathology are a consequence of their ability to target neurobiological systems relevant to neuroplasticity and neuroprotection.Reference McIntyre, Rasgon and Goldberg100 Notwithstanding the preliminary findings in support of GLP-1 receptor agonists’ potential in psychiatric contexts, evidence of antidepressant efficacy and precognitive effects independent of metabolic improvements is currently limited, highlighting the need for focused clinical studies.

Building upon this therapeutic foundation, tirzepatide, a dual glucose-dependent insulinotropic polypeptide (GIP) and GLP-1 receptor agonist exhibiting biased signaling, has demonstrated superior efficacy in weight reduction and glycemic control compared to standard GLP-1 agonists.Reference Coskun, Sloop and Loghin101Reference Chuang, Chen, Wang, Jiang and Wu103 Extant literature reports that while tirzepatide’s primary indication is for the treatment of T2DM, its biased activation of distinct intracellular pathways may also benefit psychiatric populations with metabolic comorbidities.Reference Camkurt, Lavagnino, Zhang and Teixeira98, Reference Anthamatten and Henry-Okafor104, Reference Tobaiqy and Elkout105 The precise mechanisms underlying tirzepatide’s potential effects on mood and cognitive functioning remain currently unclear and require focused clinical investigation. Notwithstanding, current hypotheses propose that biased agonism at the GLP-1Rs, favoring cyclic adenosine monophosphate (cAMP) over β-arrestin signaling, may be a key mechanism underlying its superior efficacy in metabolic improvements (Figure 2).Reference Willard, Douros and Gabe106Reference Baggio and Drucker108 By improving metabolic health, which has been reported to be associated with mood and cognitive disturbances in extant literature, tirzepatide’s biased agonism may represent a promising approach, with potentially reduced risk of adverse events (e.g., nausea and vomiting), for individuals with metabolic disorders.Reference Tobaiqy and Elkout105, Reference Rajan and Menon109 However, any clinical psychiatric benefits (e.g., improvements in mood or cognitive functioning) have yet to be explicitly demonstrated to be independent of metabolic changes. Therefore, further research is required to elucidate tirzepatide’s mood and cognitive effects in persons with comorbid T2DM or obesity and psychiatric disorder. While evidence for tirzepatide’s full impact on the foregoing parameters are still emerging, it is posited that tirzepatide’s biased agonism may underscore its superior efficacy in improving therapeutic indices across metabolic and psychiatric conditions compared to other GLP-1 RAs.

Figure 2. Tirzepatide’s hypothesized mechanism of action. Once bound to the GIP/GLP-1 receptor, tirzepatide selectively activates the G-protein intracellular pathway, specifically the Gɑs-protein-mediated signaling pathway. This ligand–receptor interaction results in receptor conformational change and activation. Subsequently, the activated receptor exchanges the guanosine diphosphate (GDP) on the ɑs-subunit to guanosine triphosphate (GTP), which activates the G-protein, leading to the dissociation of the ɑ-subunit. The activated ɑs-subunit binds to and stimulates the activation of its effector protein adenylyl cyclase (AC). Activated AC catalyzes the conversion of adenosine triphosphate (ATP) to cyclic adenosine monophosphate (cAMP). cAMP will then bind to and activate protein kinase A (PKA), which (1) activates glucose metabolism to increase the release of insulin and (2) initiates insulin gene transcription for insulin synthesis.Reference Yang, Hou and Tao136 Created in BioRender. Le, G. (2025). https://BioRender.com/ce23rn8.

Opportunity for enhancing safety with biased agonism

By selectively activating intracellular pathways, biased agonism can potentially minimize off-target effects associated with adverse outcomes while enhancing therapeutic benefits.Reference Michel and Charlton9, Reference El Eid, Reynolds, Tomas and Jones110, Reference Luttrell, Maudsley and Bohn111 Oliceridine (TRV130), is a G protein-biased mu-opioid receptor agonist able to produce analgesic efficacy and at the same time reduce adverse effects associated with beta-arrestin engagement.Reference DeWire, Yamashita and Rominger112 Initial clinical trials suggest that oliceridine may maintain analgesic efficacy with reduced hypoventilation compared to morphine; however, its long-term safety and efficacy as well as in comparison with traditional opioids remain to be conclusively demonstrated in larger-scale and long-term clinical trials.Reference Viscusi, Skobieranda, Soergel, Cook, Burt and Singla113 In addition, it has also been demonstrated in rodent models that cannabinoid CB1 receptor-biased agonists are able to reduce sedation and psychomotor impairment and still maintain therapeutic efficacy.Reference Khajehali, Malone, Glass, Sexton, Christopoulos and Leach114

Separately, there is uncertainty about whether hallucinogenic experiences mediate the antidepressant effects of serotonergic psychedelics (e.g., psilocybin).Reference Chisamore, Kaczmarek and Le12, Reference McIntyre115, Reference McIntyre, Kwan and Mansur116 Preliminary evidence suggests that coadministration of 5-HT2A antagonist not only prevents psychedelic experiences with psilocybin but does not appear to interfere with antidepressant efficacy.Reference Rosenblat, Leon-Carlyle, Ali, Husain and McIntyre117 Research efforts are attempting to determine whether fully antagonizing 5-HT2A activity or biased agonism of the 5-HT2A which aims to block the hallucinogenic effects associated with the activation of G protein signaling pathway is capable of antidepressant effects in the absence of a psychedelic experience.Reference Chisamore, Kaczmarek and Le12, Reference Halman, Kong, Sarris and Perkins118, Reference Husain, Blumberger and Castle119

Biased agonism endeavors for psilocybin could similarly target 5-HT2A pathways to minimize the profound hallucinogenic “trip.” By selectively activating downstream intracellular pathways hypothesized to underlie its therapeutic benefits and minimizing the activation of signalling pathways associated with perceptual distortions, drug tolerability would improve.Reference Chisamore, Kaczmarek and Le12, Reference López-Giménez and González-Maeso58 Notwithstanding, the foregoing potential mechanisms are currently theoretical as the ability to fully separate therapeutic from hallucinogenic effects, via biased signaling, remains to be demonstrated in clinical contexts.

Preclinical evidence from recent animal studies indicates that when only the β-arrestin-2 signaling pathway was engaged, no hallucinogenic effects were observed (i.e., head twitch response).Reference Wallach, Cao and Calkins120, Reference Lewis, Bonniwell and Lanham121 In addition, 5-HT2AR biased agonism of the β-arrestin-2 signaling pathway was also associated with antidepressant effects in mice without producing psychoactive effects (Figure 3).Reference Chisamore, Kaczmarek and Le12, Reference Cao, Yu and Wang122 The foregoing findings in animal models suggest 5-HT2AR biased agonism for the β-arrestin-2 pathway may mitigate the psychoactive effects, and ultimately reduce the toxicity associated with the “trip” as well as improve drug tolerability and safety while still providing beneficial therapeutic effects (Figure 3) (e.g., antidepressant, procognitive, etc.).

Figure 3. G-protein- versus β-arrestin-biased signaling. (a) Gq-protein-biased signaling. (b) β-arrestin-2 (βarr2)-biased signaling. Extant literature indicates βarr2-biased signaling produces antidepressant effects without psychoactive effects (ie, head twitch response in rats).Reference Chisamore, Kaczmarek and Le12, Reference López-Giménez and González-Maeso58, Reference Schmid and Bohn85, Reference Wallach, Cao and Calkins120, Reference Turu, Soltész-Katona and Tóth137 Furthermore, it is observed that ERK and MAPK signaling are downregulated in individuals with depression, which suggests they may play a role in inducing antidepressant effects observed with psychedelics.Reference Chisamore, Kaczmarek and Le12, Reference Corrao, Pollicino, Maggio, Torres and Argano138 Created in BioRender. Le, G. (2025). https://BioRender.com/fvcgyje.

While biased agonism has demonstrated notable efficacy and safety benefits in preclinical models, translating these findings from in vitro and animal models to clinical populations remains uncertain and may present challenges due to fundamental differences in the complexity of human psychedelic experiences.Reference Urs, Gee and Pack123 The rodent head-twitch response only provides a behavioral model/strategy for inferring human hallucinogenic experiences; therefore, it may not capture some but not fully reflect the complex cognitive and affective components underlying the phenomenology of the human psychedelic experience.

In clinical research contexts, several ongoing clinical trials investigating psilocybin for treatment-resistant depression have employed risperidone—a 5-HT2A antagonist—to block the hallucinogenic effects that result from activation of the G protein signaling pathway, a strategy informed by well-established evidence from drug–drug interaction studies involving classical psychedelics.Reference Chisamore, Kaczmarek and Le12, Reference Halman, Kong, Sarris and Perkins118, Reference Husain, Blumberger and Castle119 In this way, it can be determined whether hallucinogenic experiences are necessary for the facilitation of antidepressant effects. More investigations in human studies are underway. To conclude, psychedelic analogues are being created by scientists with the ability to maintain their therapeutic potential for mood disorders and decrease psychedelic effects by controlling biased signaling pathways. The contemporary research aims to develop a new generation of serotonergic drugs which could deliver fast and enduring antidepressant treatment without harming cognitive abilities or causing psychotic-like adverse reactions.

Discussion

Herein, this narrative review synthesizes existing evidence, highlighting the potential evolution from conventional agonist–antagonist paradigms (i.e., “all-or-none” modulators) towards more nuanced pharmacological frameworks, notably biased agonism. The discovery of these ligand–receptor concepts have important implications for the development of psychiatric and neurological pharmacotherapeutics considering the degree and direction of receptor activation can be adjusted to maximize therapeutic benefit while minimizing adverse side effects (e.g., hallucinogenic effects, respiratory depression, etc.).Reference Kenakin6, Reference Michel and Charlton9, Reference Stahl124 Notwithstanding, it remains crucial to recognize the preliminary nature of the current body of evidence with regard to clinical translation.

Biased agonism (functional selectivity) has emerged as a promising approach to induce pathway-specific receptor signaling, thereby improving the therapeutic index—a ratio that compares the blood concentration at which a drug causes a therapeutic effect to the amount that causes death (in animal studies) or toxicity (in human studies).Reference Tamargo, Le Heuzey and Mabo125 As aforementioned, the development of oliceridine (TRV130) shows how biased G protein-mediated signaling at the mu-opioid receptor is able to maintain analgesia while reducing respiratory depression associated with activating the β-arrestin pathway.Reference DeWire, Yamashita and Rominger112, Reference Viscusi, Skobieranda, Soergel, Cook, Burt and Singla113 In addition, CB1-receptor-biased agonism has also been demonstrated to preserve pain relief and anti-inflammatory effects in preclinical settings without the marked sedation or psychomotor impairment.Reference Khajehali, Malone, Glass, Sexton, Christopoulos and Leach114

Clinical implications of biased agonism in psychiatry

Biased agonism represents a potential innovative strategy to transform psychiatric drug development, allowing selective modulation of intracellular signaling to engage the most beneficial intracellular downstream signaling cascades, thereby increasing therapeutic effects, while avoiding pathways associated with adverse effects.Reference Tamargo, Le Heuzey and Mabo125 In psychiatry, this paradigm is especially relevant for disorders where existing treatments may produce metabolic disruption, unwanted sedation, and/or extrapyramidal symptoms (e.g., impaired motor control).Reference Meshkat, Duffy and Tassone126

Notwithstanding, it is important to note that several examples of biased agonism discussed herein are currently supported predominantly by preclinical, indirect, and/or metabolic-linked outcomes rather than direct psychiatric endpoint evidence. While the current body of literature consists of compelling preclinical rationales and limited clinical evidence, inferences of direct psychiatric benefits remain speculative and hypothesis-generating, requiring rigorous clinical confirmation.

Psychedelic compounds, such as psilocybin, have generated significant interest in the field of psychiatry due to preliminary evidence suggesting rapid antidepressant effects in persons with treatment-resistant depressive disorders; however, uncertainty remains regarding whether the therapeutic benefits can be decoupled from hallucinogenic effects via biased signaling mechanisms.Reference Chisamore, Kaczmarek and Le12, Reference McIntyre115 As discussed, preclinical findings indicate that selectively activating the 5-HT2AR-linked β-arrestin-2 pathway may not only preserve the antidepressant effects but also reduce psychoactive effects in rodent depression-like models.Reference Wallach, Cao and Calkins120Reference Cao, Yu and Wang122 Furthermore, preclinical studies are evaluating other biased agonists, specifically of 5-HT1A receptors (e.g., NLX-101 and NLX-204), that display rapid-acting antidepressant properties and cognitive functioning benefits like ketamine without the dissociative side effects.Reference Papp, Gruca, Litwa, Lason, Newman-Tancredi and Depoortère127 Further research is necessary to determine whether the foregoing findings observed in animal models are maintained in human studies in terms of efficacy and safety. Furthermore, findings from recent research efforts with GLP-1 and the dual GIP/GLP-1 receptor agonists (i.e., tirzepatide) indicate potential metabolic benefits; however, indirect implications for psychiatric outcomes remain speculative wherein mood and cognitive benefits remain heterogeneous.Reference Coskun, Sloop and Loghin101, Reference Jones128

Notably, biased agonists may exert different effects in discrete brain regions as a result of region-specific variations in signaling protein expression. For example, recent evidence indicates that a β-arrestin-2-biased D2R ligand may elicit opposing antagonistic and agonistic effects in the striatum and cortex, respectively.Reference Urs, Gee and Pack123 This phenomenon has also been observed with 5-HT1AR-biased agonists. For example, although not U.S. FDA approved yet, NLX-101 and NLX-112 both exhibit biased agonism at 5-HT1ARs; however, they each exhibit differential properties and target distinct brain regions.Reference Newman-Tancredi, Depoortère, Kleven, Kołaczkowski and Zimmer129 Specifically, NLX-101 preferential activates cortical and brain stem 5-HT1ARs and has been observed to be potently active in rodent models of depression and respiratory control.Reference Newman-Tancredi, Depoortère, Kleven, Kołaczkowski and Zimmer129 In contrast, NLX-112 exhibits prominent activation of 5-HT1A autoreceptors in Raphe nuclei and motor-relevant pathways, and has shown promising activity in animal models of PD.Reference Newman-Tancredi, Depoortère, Kleven, Kołaczkowski and Zimmer129 The foregoing examples highlight region-specific nuances of biased agonism, which could be particularly valuable for different conditions (e.g., schizophrenia) wherein optimal treatment requires opposing or differential effects in different brain regions.

In psychiatry, several pressing unmet clinical needs remain unaddressed by existing pharmacotherapies such as treatment-resistant depression (TRD), cognitive dysfunction across affective and psychotic disorders, affective instability, medication-induced adverse events, as well as effectively and simultaneously addressing various clinical symptoms in highly comorbid disorders (e.g., depression and obesity). For example, as aforementioned, while interest has sparked for psilocybin’s rapid antidepressant effects, there remains debate in regard to its psychedelic effects and widespread applicability.Reference Chisamore, Kaczmarek and Le12, Reference López-Giménez and González-Maeso58, Reference McIntyre115, Reference McIntyre, Kwan and Mansur116 In line with this, preclinical evidence suggests that a β-arrestin-2-biased D2R ligand may not only maintain the rapid-antidepressant effects without the psychedelic “trip.”Reference Urs, Gee and Pack123 Separately, 5-HT1AR-biased agonists (e.g., NLX-204, NLX-101) also show promise in producing rapid antidepressant effects akin to ketamine without inducing dissociation.Reference Newman-Tancredi, Depoortère, Kleven, Kołaczkowski and Zimmer129 Notwithstanding the lack of clinical evidence on the foregoing agents, preliminary preclinical findings suggest that they may be uniquely positioned to meet the clinical demand for not only fast-acting, but also well-tolerated treatments in TRD. Such agents, exhibiting biased agonism, hold the potential to transform psychiatric pharmacotherapy towards enhancing mechanistic precision and patient-centered outcomes.

Future directions: emerging tools and frameworks

While the central aim of this narrative review is to critically evaluate the clinical and translational relevance of biased agonism for the potential development and improvement of psychiatric pharmacotherapy, it is of note that recent advances in ligand discovery have enabled the development of pathway-selective compounds. For example, tools such as structure–activity relationship (SAR) modeling have been applied to predict and optimize biased signaling profiles in the hopes of improving clinical symptom outcomes, especially for treatment-resistant mood disorders.Reference Pottie, Poulie and Simon130 Furthermore, quantitative models (e.g., ΔLog(Emax/EC50)) have also been proposed wherein drug discovery can apply the concept of biased ligand quantification and compare signaling bias across ligands in a large-scale, standardized manner.Reference Winpenny, Clark and Cawkill131 Although an in-depth review of these methodologies is beyond the scope of the present narrative, their continued development underscores the momentum of biased agonism as a translational development strategy.

Limitations of translational evidence

Notwithstanding recent efforts, it is pertinent that robust future clinical trials specifically designed to investigate agents exhibiting biased agonism and how selectively engaging specific pathways, while inhibiting others downstream, translates to meaningful long-term improvements in clinical symptom outcomes.Reference Urs, Gee and Pack123, Reference Ji and Tao132, Reference Kenakin133 In addition, rigorous clinical trials are needed to evaluate the long-term safety profile of these agents.

Separately, given the novelty of biased agonism and complexity of GPCR signaling, the translation from preclinical to human clinical outcomes involves significant uncertainty. Heterogeneity across persons such as region-specific differences in receptor and effector molecule distributions, receptor dynamics, and genetic polymorphisms represent important translational barriers that must be systematically addressed in future research and initiatives. For example, individual genetic differences in receptor conformation or downstream effectors (e.g., G protein-coupled receptor kinases (GRK) polymorphism) may affect the degree to which a biased agonist reduces adverse effects or enhances therapeutic outcomes.Reference El Eid, Reynolds, Tomas and Jones110, Reference Kenakin133Reference Wang and Mao140

Conclusion

Biased agonism represents an innovative pharmacological concept with potential to improve safety profiles, and neurologic and psychiatric clinical outcomes by selectively modulating intracellular signaling pathways. Notwithstanding, substantial translational challenges remain which emphasizes the need for rigorous clinical validation and interdisciplinary collaboration—especially among pharmacologists, psychiatrists, and neuroscientists—to understand the complex relationships between the molecular signatures of GPCRs, and more importantly confirm the efficacy and safety in diverse patient populations. Understanding how different GPCR conformations turn cellular signaling into behavioral responses remains a key research focus in academia and industry. Discoveries from this field are expected to lead to the development of novel, safer, and more effective therapeutic strategies to improve clinical outcomes and quality of life in persons living with psychiatric disorders, especially in treatment-resistant populations.

Author contribution

Writing - original draft: S.B., S.J., H.K.L.; Writing - review & editing: K.T., D.O., H.G., P.K.L.; Conceptualization: G.H.L., R.S.M.; Investigation: S.W.

Financial support

This review article was not funded by any entity.

Competing interests

The authors declare none.

Disclosures

Dr. Roger S. McIntyre has received research grant support from CIHR/GACD/National Natural Science Foundation of China (NSFC) and the Milken Institute; speaker/consultation fees from Lundbeck, Janssen, Alkermes, Neumora Therapeutics, Boehringer Ingelheim, Sage, Biogen, Mitsubishi Tanabe, Purdue, Pfizer, Otsuka, Takeda, Neurocrine, Neurawell, Sunovion, Bausch Health, Axsome, Novo Nordisk, Kris, Sanofi, Eisai, Intra-Cellular, NewBridge Pharmaceuticals, Viatris, Abbvie, Bristol Myers Squibb (BMS) and Atai Life Sciences.

Dr. Jennifer Swainson has received speaker/ consultation fees from AbbVie, Bausch, Eisai, Elvium, Idorsia, Janssen, Novonordisk, Lundbeck, Otsuka,

Dr. Hernan F. Guillen-Burgos has received research grant support from the Ministry of Science, Technology, and Innovation (MinCiencias) in Colombia; UKRI in the United Kingdom; and speaker fees from Roche, Pfizer, Abbott, Synergy R&D.

Kayla M. Teopiz has received fees from Braxia Scientific Corp.

The remaining authors have no conflicts of interest to disclose, including G.H.L, S.W, S.B., D.K.O, H.K.Y.L, and P.K.L.

References

Katritch, V, Cherezov, V, Stevens, RC. Diversity and modularity of G protein-coupled receptor structures. Trends Pharmacol Sci. 2012;33(1):1727.Google Scholar
Insel, PA, Sriram, K, Gorr, MW, et al. GPCRomics: an approach to discover GPCR drug targets. Trends Pharmacol Sci. 2019;40(6):378387.Google Scholar
Kurose, H, Kim, SG. Recent advances of GPCR studies: an old-fashioned perspective? Biol Pharm Bull. 2022;45(6):669674.Google Scholar
Sriram, K, Insel, PA. G protein-coupled receptors as targets for approved drugs: how many targets and how many drugs? Mol Pharmacol. 2018;93(4):251258.Google Scholar
Rosenbaum, DM, Rasmussen, SGF, Kobilka, BK. The structure and function of G-protein-coupled receptors. Nature. 2009;459:356363.Google Scholar
Kenakin, T. Functional selectivity and biased receptor signaling. J Pharmacol Exp Ther. 2011;336(2):296302.Google Scholar
Berg, KA, Clarke, WP. Making sense of pharmacology: inverse Agonism and functional selectivity. Int J Neuropsychopharmacol. 2018;21(10):962977.Google Scholar
Sum, CS, Murphy, BJ, Li, Z, et al. Pharmacological characterization of GPCR agonists, antagonists, allosteric modulators and biased ligands from HTS hits to Lead optimization. In: Markossian, S, Grossman, A, Arkin, M, et al., eds. Assay Guidance Manual. Bethesda: Eli Lilly & Company and the National Center for Advancing Translational Sciences; 2019. https://www.ncbi.nlm.nih.gov/books/NBK549462/Google Scholar
Michel, MC, Charlton, SJ. Biased Agonism in drug discovery-is it too soon to choose a path? Mol Pharmacol. 2018;93(4):259265.Google Scholar
Andresen, BT. A pharmacological primer of biased Agonism. Endocr Metab Immune Disord Drug Targets. 2011;11(2):9298.Google Scholar
Wootten, D, Christopoulos, A, Marti-Solano, M, Madan Babu, M, Sexton, PM. Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat Rev Mol Cell Biol. 2018;19:638653.Google Scholar
Chisamore, N, Kaczmarek, E, Le, GH, et al. Neurobiology of the antidepressant effects of serotonergic psychedelics: a narrative review. Curr Treat Options Psych. 2024;11:90105.Google Scholar
Kenakin, T. Overview of receptor interactions of agonists and antagonists. Curr Protoc Pharmacol. 2008;42(1):4.1.14.1.24.Google Scholar
Bardal, ST, Waechter, JE, Martin, DS. Chapter 1 - basic principles and pharmacodynamics. In: Applied Pharmacology. W.B. Saunders; 2011:316. https://doi.org/10.1016/B978-1-4377-0310-8.00001-4Google Scholar
Williams, J. Enzyme inhibition and induction. Anaesth Intensive Care Med. 2008;9(4):165166. https://doi.org/10.1016/j.mpaic.2008.02.004Google Scholar
Zhu, BT. The competitive and noncompetitive antagonism of receptor-mediated drug actions in the presence of spare receptors. J Pharmacol Toxicol Methods. 1993;29(2):8591.Google Scholar
Stephenson, RP. A modification of receptor theory. Br J Pharmacol. 1997;120(Suppl 1):106120.Google Scholar
Ferner, RE, Aronson, JK. Cato Guldberg and Peter Waage, the history of the law of mass action, and its relevance to clinical pharmacology. Br J Clin Pharmacol. 2016;81(1):5255.Google Scholar
Kenakin, T. The mass action equation in pharmacology. Br J Clin Pharmacol. 2016;81(1):4151.Google Scholar
Nadeem, MS, Riaz, MN, Hosawi, SBI, et al. Chapter 10 - mechanism of action of antipsychotics and antimanics. In: How Synthetic Drugs Work: Insights into Molecular Pharmacology of Classic and New Pharmaceuticals. Academic Press. 2023:215253. https://doi.org/10.1016/B978-0-323-99855-0.00010-5.Google Scholar
Thomas, K, Olanzapine, SA. StatPearls. Treasure Island: StatPearls Publishing;2025. https://www.ncbi.nlm.nih.gov/books/NBK532903/Google Scholar
Lista, AD, Sirimaturos, M. Pharmacokinetic and Pharmacodynamic principles for toxicology. Crit Care Clin. 2021;37(3):475486.Google Scholar
Arias, HR, Bhumireddy, P, Bouzat, C. Molecular mechanisms and binding site locations for noncompetitive antagonists of nicotinic acetylcholine receptors. Int J Biochem Cell Biol. 2006;38(8):12541276.Google Scholar
Delaune, KP, Alsayouri, K. Physiology, noncompetitive inhibitor. In: StatPearls. Treasure Island: StatPearls Publishing; 2025. https://www.ncbi.nlm.nih.gov/books/NBK545242/Google Scholar
Jelen, LA, Young, AH, Stone, JM. Ketamine: a tale of two enantiomers. J Psychopharmacol. 2020;35(2):102123.Google Scholar
Zorumski, CF, Izumi, Y, Ketamine, MS. NMDA receptors and beyond. J Neurosci. 2016;36(44):1115811164.Google Scholar
Krystal, JH, Abdallah, CG, Sanacora, G, Charney, DS, Duman, RS. Ketamine: a paradigm shift for depression research and treatment. Neuron. 2019;101:774778.Google Scholar
Mion, G, Villevieille, T. Ketamine pharmacology: an update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther. 2013;19(6):370380.Google Scholar
Autry, AE, Adachi, M, Nosyreva, E, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011;475:9195.Google Scholar
Yang, C, Shirayama, Y, Jc, Z, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5:e632Google Scholar
Smith, KL, Rao, RR, Velázquez-Sánchez, C, et al. The uncompetitive N-methyl-D-aspartate antagonist Memantine reduces binge-like eating, food-seeking behavior, and compulsive eating: role of the nucleus Accumbens Shell. Neuropsychopharmacol. 2015;40(5):11631171.Google Scholar
Traynelis, SF, Wollmuth, LP, McBain, CJ, et al. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev. 2010;62(3):405496.Google Scholar
Chen, HS, Lipton, SA. Mechanism of memantine block of NMDA-activated channels in rat retinal ganglion cells: uncompetitive antagonism. J Physiol. 1997;499:2746.Google Scholar
Lipton, SA. The molecular basis of memantine action in Alzheimer’s disease and other neurologic disorders: low-affinity, uncompetitive antagonism. Curr Alzheimer Res. 2005;2(2):155165.Google Scholar
Xia, P, Chen, HV, Zhang, D, Lipton, SA. Memantine preferentially blocks Extrasynaptic over synaptic NMDA receptor currents in hippocampal Autapses. J Neurosci. 2010;30(33):1124611250.Google Scholar
Folch, J, Busquets, O, Ettcheto, M, et al. Memantine for the treatment of dementia: a review on its current and future applications. J Alzheimers Dis. 2018;2(3):12231240.Google Scholar
Puranik, N, Song, M. Glutamate: molecular mechanisms and Signaling pathway in Alzheimer’s disease, a potential therapeutic target. Molecules. 2024;29(23):5744Google Scholar
McCarthy, B, Bunn, H, Santalucia, M, Wilmouth, C, Muzyk, A, Smith, CM. Dextromethorphan-bupropion (Auvelity) for the treatment of major depressive disorder. Clin Psychopharmacol Neurosci. 2023;21(4):609616.Google Scholar
Tate, CG. A crystal clear solution for determining G-protein-coupled receptor structures. Trends Biochem Sci. 2012;37(9):343352.Google Scholar
Watts, SW, Townsend, RR, Neubig, RR. How new developments in pharmacology receptor theory are changing (our understanding of) hypertension therapy. Am J Hypertens. 2023;37(4):248260.Google Scholar
Edinoff, AN, Kaplan, LA, Khan, S, et al. Full opioid agonists and tramadol: pharmacological and clinical considerations. Anesth Pain Med. 2021;11(4):e119156Google Scholar
Dhaliwal, A, Gupta, M. Physiology, opioid receptor. In: StatPearls. Treasure Island: StatPearls Publishing; 2025. https://www.ncbi.nlm.nih.gov/books/NBK546642/Google Scholar
Keith, DE, Murray, SR, Zaki, PA, et al. Morphine activates opioid receptors without causing their rapid internalization. J Biol Chem. 1996;271(32):1902119024.Google Scholar
Ricarte, A, Dalton, JAR, Giraldo, J. Structural assessment of agonist efficacy in the μ-opioid receptor: morphine and fentanyl elicit different activation patterns. J Chem Inf Model. 2021;61(3):12511274.Google Scholar
Levinstein, MR, De Oliveira, PA, Casajuana-Martin, N, et al. Unique pharmacodynamic properties and low abuse liability of the μ-opioid receptor ligand (S)-methadone. Mol Psychiatry. 2024;29(3):624632.Google Scholar
Sandilands, EA, Bateman, DN. Opioids. Medicine. 2016;44(3):187–19.Google Scholar
Mohr, P, Masopust, J, Kopeček, M. Dopamine receptor partial agonists: do they differ in their clinical efficacy? Front Psychiatry. 2022;12:781946Google Scholar
Webster, L, Gudin, J, Raffa, RB, et al. Understanding buprenorphine for use in chronic pain: expert opinion. Pain Med. 2020;21(4):714723.Google Scholar
Citrome, L. Brexpiprazole: a new dopamine D₂receptor partial agonist for the treatment of schizophrenia and major depressive disorder. Drugs Today (Barc). 2015;51(7):397414.Google Scholar
Citrome, L. Cariprazine for the treatment of schizophrenia: a review of this dopamine D3-preferring D3/D2 receptor partial agonist. Clin Schizophr Relat Psychoses. 2016;10(2):109119.Google Scholar
Ragguett, RM, McIntyre, RS. Cariprazine for the treatment of bipolar depression: a review. Expert Rev Neurother. 2019;19(4):317323.Google Scholar
Tuplin, EW, Holahan, MR. Aripiprazole, a drug that displays partial Agonism and functional selectivity. Curr Neuropharmacol. 2017;15(8):11921207.Google Scholar
McIntyre, RS, Soczynska, JK, Woldeyohannes, HO, Miranda, A, Konarski, JZ. Aripiprazole: pharmacology and evidence in bipolar disorder. Expert Opin Pharmacother. 2007;8(7):10011009.Google Scholar
Sciascio, GD, Riva, MA. Aripiprazole: from pharmacological profile to clinical use. Neuropsychiatr Dis Treat. 2015;11:26352647.Google Scholar
Lieberman, JA. Dopamine partial agonists: a new class of antipsychotic. CNS Drugs. 2004;18(4):251267.Google Scholar
de Bartolomeis, A, Tomasetti, C, Iasevoli, F. Update on the mechanism of action of Aripiprazole: translational insights into antipsychotic strategies beyond dopamine receptor antagonism. CNS Drugs. 2015;29:773799.Google Scholar
Costa, T, Herz, A. Antagonists with negative intrinsic activity at delta opioid receptors coupled to GTP-binding proteins. Proc Natl Acad Sci U S A. 1989;86(19):73217325.Google Scholar
López-Giménez, JF, González-Maeso, J. Hallucinogens and serotonin 5-HT2A receptor-mediated Signaling pathways. Curr Top Behav Neurosci. 2018;36:4573.Google Scholar
Vollenweider, FX, Vollenweider-Scherpenhuyzen, MF, Bäbler, A, Vogel, H, Hell, D. Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2 agonist action. Neuroreport. 1998;9(17):38973902.Google Scholar
Zhang, G, Stackman, RW. The role of serotonin 5-HT2A receptors in memory and cognition. Front Pharmacol. 2015;6:225Google Scholar
Casey, AB, Cui, M, Booth, RG, Canal, CE. Selective" serotonin 5-HT2A receptor antagonists. Biochem Pharmacol. 2022;200:115028Google Scholar
Cummings, J, Isaacson, S, Mills, R, et al. Pimavanserin for patients with Parkinson’s disease psychosis: a randomised, placebo-controlled phase 3 trial. Lancet. 2014;383(9916):533540.Google Scholar
Muneta-Arrate, I, Diez-Alarcia, R, Horrillo, I, Meana, JJ. Pimavanserin exhibits serotonin 5-HT2A receptor inverse agonism for Gαi1- and neutral antagonism for gαq/11-proteins in human brain cortex. Eur Neuropsychopharmacol. 2020;36:8389.Google Scholar
Meltzer, KR, Cao, TV, Schad, JF, King, H, Stoll, ST, Standley, PR. In vitro modeling of repetitive motion injury and myofascial release. J Bodyw Mov Ther. 2010;14(2):162171.Google Scholar
Rissardo, JP, Durante, Í, Sharon, I, Fornari Caprara, AL. Pimavanserin and Parkinson’s disease psychosis: a narrative review. Brain Sci. 2022;12(10):1286Google Scholar
Schrage, R, De Min, A, Hochheiser, K, Kostenis, E, Mohr, K. Superagonism at G protein-coupled receptors and beyond. Br J Pharmacol. 2015;173(20):30183027.Google Scholar
Brown, KE. Revisiting CD28 Superagonist TGN1412 as potential therapeutic for Pediatric B cell Leukemia: a review. Diseases. 2018;6(2):41Google Scholar
Smith, NJ, Bennett, KA, Milligan, G. When simple agonism is not enough: emerging modalities of GPCR ligands. Mol Cell Endocrinol. 2011;331(2):241247.Google Scholar
Miess, E, Gondin, AB, Yousuf, A, et al. Multisite phosphorylation is required for sustained interaction with GRKs and arrestins during rapid μ-opioid receptor desensitization. Sci Signal. 2018;11(539):eaas9609Google Scholar
Lowe, JD, Sanderson, HS, Cooke, AE, et al. Role of G protein-coupled receptor kinases 2 and 3 in μ-opioid receptor desensitization and internalization. Mol Pharmacol. 2015;88(2):347356.Google Scholar
Malcolm, NJ, Palkovic, B, Sprague, DJ, et al. Mu-opioid receptor selective superagonists produce prolonged respiratory depression. iScience. 2023;26(7):107121Google Scholar
Vandeputte, MM, Van Uytfanghe, K, Layle, NK, St Germaine, DM, Iula, DM, Stove, CP. Synthesis, chemical characterization, and μ-opioid receptor activity assessment of the emerging group of “Nitazene” 2-Benzylbenzimidazole synthetic opioids. ACS Chem Neurosci. 2021;12(7):12411251.Google Scholar
Hilger, D. The role of structural dynamics in GPCR-mediated signaling. FEBS J. 2021;288(8):24612489.Google Scholar
Calebiro, D, Koszegi, Z, Lanoiselée, Y, Milijus, T, O’Brien, S. G protein-coupled receptor-G protein interactions: a single-molecule perspective. Physiol Rev. 2021;101(3):857906.Google Scholar
Eiger, DS, Pham, U, Gardner, J, Hicks, C, Rajagopal, S. GPCR systems pharmacology: a different perspective on the development of biased therapeutics. Am J Physiol Cell Physiol. 2022;322(5):C887C895.Google Scholar
Lamichhane, R, Liu, JJ, White, KL, et al. Biased Signaling of the G-protein-coupled receptor β2AR is governed by conformational exchange kinetics. Structure. 2020;28(3):371377.e3.Google Scholar
Ehlert, FJ. Analysis of biased Agonism. Prog Mol Biol Transl Sci. 2018;160:63104.Google Scholar
Franco, R, Aguinaga, D, Jiménez, J, Lillo, J, Martínez-Pinilla, E, Navarro, G. Biased receptor functionality versus biased agonism in G-protein-coupled receptors. Biomol Concepts. 2018;9(1):143154.Google Scholar
Chang, SD, Bruchas, MR. Functional selectivity at GPCRs: new opportunities in psychiatric drug discovery. Neuropsychopharmacol. 2014;39(1):248249.Google Scholar
Yang, D, Zhou, Q, Laborska, V, et al. G protein-coupled receptors: structure- and function-based drug discovery. Sig Transduct Target Ther. 2021;6:7Google Scholar
Niswender, CM, Conn, PJ. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharmacol Toxicol. 2010;50:295322.Google Scholar
Lamberts, JT, Traynor, JR. Opioid receptor interacting proteins and the control of opioid signaling. Curr Pharm Des. 2013;19(42):73337347.Google Scholar
Bruchas, MR, Chavkin, C. Kinase cascades and ligand-directed signaling at the kappa opioid receptor. Psychopharmacology (Berl). 2010;210:137147.Google Scholar
Bruchas, MR, Schindler, AG, Shankar, H, et al. Selective p38α MAPK deletion in serotonergic neurons produces stress resilience in models of depression and addiction. Neuron. 2011;71:498511.Google Scholar
Schmid, CL, Bohn, LM. Serotonin, but not N-methyltryptamines, activates the serotonin 2 a receptor via a ß-arrestin2/Src/Akt signaling complex in vivo. J Neurosci. 2010;30:1351313524.Google Scholar
Urs, NM, Snyder, JC, Jacobsen, JP, Peterson, SM, Caron, MG. Deletion of GSK3β in D2R-expressing neurons reveals distinct roles for β-arrestin signaling in antipsychotic and lithium action. Proc Natl Acad Sci USA. 2012;109:2073220737.Google Scholar
Urs, NM, Bido, S, Peterson, SM, et al. Targeting β-arrestin2 in the treatment of l-DOPA–induced dyskinesia in Parkinson’s disease. Proc Natl Acad Sci USA. 2012;109(49):2119621201.Google Scholar
Kurtzhals, P, Flindt Kreiner, F, Singh Bindra, R. The role of weight control in the management of type 2 diabetes mellitus: perspectives on semaglutide. Diabetes Res Clin Pract. 2023;203:110881Google Scholar
Badulescu, S, Tabassum, A, Le, GH, et al. Glucagon-like peptide 1 agonist and effects on reward behaviour: a systematic review. Physiol Behav. 2024;283:114622Google Scholar
De Giorgi, R, Ghenciulescu, A, Dziwisz, O, et al. An analysis on the role of glucagon-like peptide-1 receptor agonists in cognitive and mental health disorders. Nat Mental Health. 2025;3:354373.Google Scholar
Zheng, YJ, Soegiharto, C, Au, HCT, et al. A systematic review on the role of glucagon-like peptide-1 receptor agonists on alcohol-related behaviors: potential therapeutic strategy for alcohol use disorder. Acta Neuropsychiatr. 2025;37:e51Google Scholar
Lee, S, Li, M, Le, GH, et al. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) as treatment for nicotine cessation in psychiatric populations: a systematic review. Ann Gen Psychiatry. 2024;23(1):45Google Scholar
Cooper, DH, Ramachandra, R, Ceban, F, et al. Glucagon-like peptide 1 (GLP-1) receptor agonists as a protective factor for incident depression in patients with diabetes mellitus: a systematic review. J Psychiatr Res. 2023;164:8089.Google Scholar
McIntyre, RS. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and suicidality: what do we know and future vistas. Expert Opin Drug Saf. 2024;23(5):539542.Google Scholar
Segal, Y, Gunturu, S. Psychological issues associated with obesity. In: StatPearls. Treasure Island: StatPearls Publishing; 2025. https://www.ncbi.nlm.nih.gov/books/NBK603747/Google Scholar
Busili, A, Kumar, K, Kudrna, L, Busaily, I. The risk factors for mental health disorders in patients with type 2 diabetes: an umbrella review of systematic reviews with and without meta-analysis. Heliyon. 2024;10(7):e28782Google Scholar
Chen, X, Zhao, P, Wang, W, Guo, L, Pan, Q. The antidepressant effects of GLP-1 receptor agonists: a systematic review and meta-analysis. Am J Geriatr Psychiatry. 2024;32(1):117127.Google Scholar
Camkurt, MA, Lavagnino, L, Zhang, XY, Teixeira, AL. Liraglutide for psychiatric disorders: clinical evidence and challenges. Horm Mol Biol Clin Investig. 2018;36(2):/j/hmbci.2018.36.issue-2/hmbci-2018-0031/hmbci-2018-0031.xml. https://doi.org/10.1515/hmbci-2018-0031.Google Scholar
Gunturu, S. The potential role of GLP-1 agonists in psychiatric disorders: a paradigm shift in mental health treatment. Indian J Psychol Med. 2024;46(3):193195.Google Scholar
McIntyre, RS, Rasgon, N, Goldberg, J, et al. The effect of glucagon-like peptide-1 and glucose dependent insulinotropic polypeptide receptor agonists on neurogenesis, differentiation, and plasticity (Neuro-GDP): potential mechanistically informed therapeutics in the treatment and prevention of mental disorders. CNS Spectr. 2025;30(1):e23Google Scholar
Coskun, T, Sloop, KW, Loghin, C, et al. LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet. 2018;392(10160):21802193.Google Scholar
Andraos, J, Muhar, H, Smith, SR. Beyond glycemia: comparing tirzepatide to GLP-1 analogues. Rev Endocr Metab Disord. 2023;24(6):10891101.Google Scholar
Chuang, MH, Chen, JY, Wang, HY, Jiang, ZH, Wu, VC. Clinical outcomes of Tirzepatide or GLP-1 receptor agonists in individuals with type 2 diabetes. JAMA Netw Open. 2024;7(8):e2427258Google Scholar
Anthamatten, A, Henry-Okafor, Q. Strategies to address the metabolic burden of psychotropic medications. J for Nurse Practitioners. 2025;21(4):105346Google Scholar
Tobaiqy, M, Elkout, H. Psychiatric adverse events associated with semaglutide, liraglutide and tirzepatide: a pharmacovigilance analysis of individual case safety reports submitted to the EudraVigilance database. Int J Clin Pharm. 2024;46:488495.Google Scholar
Willard, FS, Douros, JD, Gabe, MB, et al. Tirzepatide is an imbalanced and biased dual GIP and GLP-1 receptor agonist. JCI Insight. 2020;5(17):e140532Google Scholar
Zheng, Z, Zong, Y, Ma, Y, et al. Glucagon-like peptide-1 receptor: mechanisms and advances in therapy. Sig Transduct Target Ther. 2024;9:234Google Scholar
Baggio, LL, Drucker, DJ. Glucagon-like peptide-1 receptors in the brain: controlling food intake and body weight. J Clin Invest. 2014;124(10):42234226.Google Scholar
Rajan, TM, Menon, V. Psychiatric disorders and obesity: a review of association studies. J Postgrad Med. 2017;63(3):182190.Google Scholar
El Eid, L, Reynolds, CA, Tomas, A, Jones, B. Biased agonism and polymorphic variation at the GLP-1 receptor: implications for the development of personalised therapeutics. Pharmacol Res. 2022;184:106411Google Scholar
Luttrell, LM, Maudsley, S, Bohn, LM. Fulfilling the promise of “biased” G protein-coupled receptor Agonism. Mol Pharmacol. 2015;88(3):579588.Google Scholar
DeWire, SM, Yamashita, DS, Rominger, DH, et al. A G protein-biased ligand at the μ-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine. J Pharmacol Exp Ther. 2013;344(3):708717.Google Scholar
Viscusi, ER, Skobieranda, F, Soergel, DG, Cook, E, Burt, DA, Singla, N. APOLLO-1: a randomized placebo and active-controlled phase III study of oliceridine (TRV130), a G protein–biased ligand at the μ-opioid receptor, for management of moderate to severe acute pain following bunionectomy. J Pain Res. 2019;12:927943.Google Scholar
Khajehali, E, Malone, DT, Glass, M, Sexton, PM, Christopoulos, A, Leach, K. Biased Agonism and biased allosteric modulation at the CB1 cannabinoid receptor. Mol Pharmacol. 2015;88(2):368379.Google Scholar
McIntyre, RS. Is the psychedelic experience an essential aspect of the therapeutic effect of serotonergic psychedelics? Conceptual, discovery, development and implementation implications for psilocybin and related agents. Expert Opin Drug Saf. 2023;22(10):885889.Google Scholar
McIntyre, RS, Kwan, ATH, Mansur, RB, et al. Psychedelics for the treatment of psychiatric disorders: interpreting and translating available evidence and guidance for future research. Am J Psychiatry. 2025;182(1):2132.Google Scholar
Rosenblat, JD, Leon-Carlyle, M, Ali, S, Husain, MI, McIntyre, RS. Antidepressant effects of psilocybin in the absence of psychedelic effects. Am J Psychiatry. 2023;180(5):395396.Google Scholar
Halman, A, Kong, G, Sarris, J, Perkins, D. Drug-drug interactions involving classic psychedelics: a systematic review. J Psychopharmacol. 2024;38(1):318.Google Scholar
Husain, MI, Blumberger, DM, Castle, DJ, et al. Psilocybin for treatment-resistant depression without psychedelic effects: study protocol for a 4-week, double-blind, proof-of-concept randomised controlled trial. BJPsych Open. 2023;9(4):e134Google Scholar
Wallach, J, Cao, AB, Calkins, MM, et al. Identification of 5-HT2A receptor signaling pathways associated with psychedelic potential. Nat Commun. 2023;14(1):8221Google Scholar
Lewis, V, Bonniwell, EM, Lanham, JK, et al. A non-hallucinogenic LSD analog with therapeutic potential for mood disorders. Cell Rep. 2023;42(3):112203Google Scholar
Cao, D, Yu, J, Wang, H, et al. Structure-based discovery of nonhallucinogenic psychedelic analogs. Science. 2022;375(6579):403411.Google Scholar
Urs, NM, Gee, SM, Pack, TF, et al. Distinct cortical and striatal actions of a β-arrestin-biased dopamine D2 receptor ligand reveal unique antipsychotic-like properties. Proc Natl Acad Sci U S A. 2016;113(50):E8178E8186.Google Scholar
Stahl, SM. Stahl’s essential psychopharmacology: Neuroscientific basis and practical applications. Mens Sana Monogr. 2010;8(1):146–15.Google Scholar
Tamargo, J, Le Heuzey, JY, Mabo, P. Narrow therapeutic index drugs: a clinical pharmacological consideration to flecainide. Eur J Clin Pharmacol. 2015;71(5):549567.Google Scholar
Meshkat, S, Duffy, SF, Tassone, KV, et al. Increased odds of metabolic syndrome among adults with depressive symptoms or antidepressant use. Transl Psychiatry. 2025;15:68Google Scholar
Papp, M, Gruca, P, Litwa, E, Lason, M, Newman-Tancredi, A, Depoortère, R. The 5-HT1A receptor biased agonists, NLX-204 and NLX-101, like ketamine, elicit rapid-acting antidepressant activity in the rat chronic mild stress model via cortical mechanisms. J Psychopharmacol. 2024;38(7):661671.Google Scholar
Jones, B. The therapeutic potential of GLP-1 receptor biased agonism. Br J Pharmacol. 2022;179(4):492510.Google Scholar
Newman-Tancredi, A, Depoortère, RY, Kleven, MS, Kołaczkowski, M, Zimmer, L. Translating biased agonists from molecules to medications: serotonin 5-HT1A receptor functional selectivity for CNS disorders. Pharmacol Ther. 2022;229:107937Google Scholar
Pottie, E, Poulie, CBM, Simon, IA, et al. Structure-activity assessment and in-depth analysis of biased Agonism in a set of Phenylalkylamine 5-HT2A receptor agonists. ACS Chem Neurosci. 2023;14(15):27272742.Google Scholar
Winpenny, D, Clark, M, Cawkill, D. Biased ligand quantification in drug discovery: from theory to high throughput screening to identify new biased μ opioid receptor agonists. Br J Pharmacol. 2016;173(8):13931403.Google Scholar
Ji, RL, Tao, YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther. 2025;268:108804Google Scholar
Kenakin, T. Biased receptor Signaling in drug discovery. Pharmacol Rev. 2019;71(2):267315.Google Scholar
Rankovic, Z, Brust, TF, Bohn, LM. Biased agonism: an emerging paradigm in GPCR drug discovery. Bioorg Med Chem Lett. 2016;26(2):241250.Google Scholar
Raehal, KM, Schmid, CL, Groer, CE, Bohn, LM. Functional selectivity at the μ-opioid receptor: implications for understanding opioid analgesia and tolerance. Pharmacol Rev. 2011;63(4):10011019.Google Scholar
Yang, LK, Hou, ZS, Tao, YX. Biased signaling in naturally occurring mutations of G protein-coupled receptors associated with diverse human diseases. Biochim Biophys Acta Mol Basis Dis. 2021;1867(1):165973Google Scholar
Turu, G, Soltész-Katona, E, Tóth, AD, et al. Biased coupling to β-Arrestin of two common variants of the CB2 cannabinoid receptor. Front Endocrinol (Lausanne). 2021;12:714561Google Scholar
Corrao, S, Pollicino, C, Maggio, , Torres, A, Argano, C. Tirzepatide against obesity and insulin-resistance: pathophysiological aspects and clinical evidence. Front Endocrinol (Lausanne). 2024;15:1402583Google Scholar
Jean-Charles, PY, Kaur, S, Shenoy, SK. G protein-coupled receptor Signaling through β-Arrestin-dependent mechanisms. J Cardiovasc Pharmacol. 2017;70(3):142158.Google Scholar
Wang, JQ, Mao, L. The ERK pathway: molecular mechanisms and treatment of depression. Mol Neurobiol. 2019;56(9):61976205.Google Scholar
Figure 0

Figure 1. G-protein-coupled receptor-biased agonism. Left: Balanced agonist that activates both the G-protein- and β-arrestin-mediated pathways. Middle: G-protein-biased agonist that selectively activates the G-protein intracellular pathway. Right: βarrestin-biased agonist that selectively activates the β-arrestin intracellular pathway. Created in BioRender. Le, G. (2025). https://BioRender.com/3tmy5ld.

Figure 1

Figure 2. Tirzepatide’s hypothesized mechanism of action. Once bound to the GIP/GLP-1 receptor, tirzepatide selectively activates the G-protein intracellular pathway, specifically the Gɑs-protein-mediated signaling pathway. This ligand–receptor interaction results in receptor conformational change and activation. Subsequently, the activated receptor exchanges the guanosine diphosphate (GDP) on the ɑs-subunit to guanosine triphosphate (GTP), which activates the G-protein, leading to the dissociation of the ɑ-subunit. The activated ɑs-subunit binds to and stimulates the activation of its effector protein adenylyl cyclase (AC). Activated AC catalyzes the conversion of adenosine triphosphate (ATP) to cyclic adenosine monophosphate (cAMP). cAMP will then bind to and activate protein kinase A (PKA), which (1) activates glucose metabolism to increase the release of insulin and (2) initiates insulin gene transcription for insulin synthesis.136 Created in BioRender. Le, G. (2025). https://BioRender.com/ce23rn8.

Figure 2

Figure 3. G-protein- versus β-arrestin-biased signaling. (a) Gq-protein-biased signaling. (b) β-arrestin-2 (βarr2)-biased signaling. Extant literature indicates βarr2-biased signaling produces antidepressant effects without psychoactive effects (ie, head twitch response in rats).12,58,85,120,137 Furthermore, it is observed that ERK and MAPK signaling are downregulated in individuals with depression, which suggests they may play a role in inducing antidepressant effects observed with psychedelics.12,138 Created in BioRender. Le, G. (2025). https://BioRender.com/fvcgyje.