Fact-checked by Grok 2 weeks ago

Functional selectivity

Functional selectivity, also known as biased agonism, refers to the capacity of a to stabilize distinct conformational states of a receptor—most commonly G protein-coupled receptors (GPCRs)—thereby preferentially activating certain intracellular signaling pathways over others that the same receptor can couple to, resulting in pathway-specific cellular responses rather than uniform activation across all possible effectors. This concept challenges classical receptor theory, which posited a single intrinsic for a ligand-receptor interaction independent of the downstream response measured. In essence, a functionally selective ligand can act as an for one pathway (e.g., G protein-mediated modulation) while functioning as an or even for another (e.g., β-arrestin recruitment), all at the same receptor subtype. The mechanisms underlying functional selectivity involve ligand-induced conformational changes in the receptor that alter the efficiency of coupling to diverse transducers, such as heterotrimeric G proteins (e.g., G_s, G_{i/o}, G_{q/11}) or β-arrestins, influenced by factors like receptor phosphorylation patterns, cellular context, and effector availability. For instance, biased ligands may promote specific microswitch transitions in the receptor's intracellular loops, favoring G protein engagement over arrestin binding, or vice versa, as revealed by structural studies of GPCR-ligand complexes. This selectivity arises not from receptor subtype differences but from the ligand's ability to "traffic" the receptor toward particular active states, a process first evidenced in systems like the β_2-adrenergic and angiotensin AT_1 receptors in the early 2000s. Quantitative pharmacology models, such as the operational model of agonism extended for bias, enable measurement of this phenomenon through metrics like the bias factor (τ/KA ratios across pathways), highlighting its dependence on assay conditions. Notable examples abound in pharmacologically relevant GPCRs, including opioid receptors where G protein-biased agonists like (TRV130; FDA-approved in 2020) provide analgesia with reduced respiratory depression and gastrointestinal side effects compared to balanced agonists like . Similarly, in dopamine D_2 receptors, β-arrestin-biased ligands engage Akt/GSK3β pathways implicated in effects, while G protein-biased ones may contribute to motor side effects. Functional selectivity has also been observed in serotonin 5-HT_{2A} receptors, where hallucinogenic agonists induce unique signaling profiles distinct from non-hallucinogenic ones, potentially involving differential β-arrestin recruitment. These pathway-specific outcomes underscore the therapeutic potential of designing biased ligands to decouple beneficial effects from adverse ones, revolutionizing for conditions like , psychiatric disorders, and . Ongoing research emphasizes the need for advanced assays and structural insights to accurately quantify and harness bias, as misinterpretation can arise from low-efficacy ligands mimicking selectivity.

Definition and Background

Core Definition

Functional selectivity, also known as biased or ligand bias, refers to the capacity of a to stabilize specific conformations of a receptor that lead to the preferential activation of certain downstream signaling pathways while relatively sparing others, rather than uniformly engaging all possible pathways associated with that receptor. This phenomenon arises from the inherent conformational dynamics of receptors, where ligands act as allosteric modulators influencing the receptor's active states to produce pathway-specific outcomes. Central to understanding functional selectivity are several key pharmacological concepts. Receptor conformational dynamics describe how ligands induce or stabilize distinct receptor states, each capable of coupling to different effectors. quantifies a ligand's ability to activate a given signaling pathway, often represented by the maximum response it elicits, while potency indicates the concentration required to achieve half of that maximum effect. In the context of G protein-coupled receptors (GPCRs), which are a primary where this occurs, these properties allow ligands to exhibit varied signaling profiles. A quantitative measure of functional selectivity is the bias factor, which compares the relative activation of two pathways by a relative to a reference . This is calculated using the operational model of , where the bias factor (ΔΔlog(τ/K_A)) is given by: \Delta \Delta \log\left(\frac{\tau}{K_A}\right) = \log\left(\frac{\tau_A}{K_{A_A}}\right) - \log\left(\frac{\tau_B}{K_{A_B}}\right) Here, τ represents the (transducer ratio, reflecting the system's capacity to produce a response), and K_A is the equilibrium of the ligand-receptor complex for each pathway (A and B). Functional selectivity differs fundamentally from traditional notions of full agonism and partial agonism. Full agonism involves ligands that maximally activate all available receptor-mediated pathways with balanced , whereas partial agonism produces submaximal activation across those pathways without preferential bias. In contrast, functionally selective ligands decouple pathway activation, enabling targeted therapeutic effects by avoiding unwanted side effects from non-preferred pathways.

Historical Context

The concept of functional selectivity in G protein-coupled receptors (GPCRs) emerged from early observations in the and 1990s, particularly through studies on opioid receptors that revealed pathway-specific signaling effects. For instance, research on mu-opioid receptor variants in rat brain tissue demonstrated the interconversion between mu and delta receptor forms, suggesting inherent differences in their functional responses to ligands. Subsequent work in the mid-1990s highlighted agonist-dependent switching of mu-opioid receptor coupling from inhibitory Gαi/o proteins to stimulatory Gαs proteins, leading to varied physiological outcomes such as analgesia versus excitation. These findings indicated that receptors could produce differential signaling based on ligand interaction, challenging the uniform activation model prevalent at the time. The terminology to describe these phenomena developed in the 1990s, with Terry Kenakin introducing the concept of "agonist trafficking" in 1995 to explain how ligands direct receptors toward specific intracellular pathways, often through selective G protein coupling. This idea gained traction as evidence accumulated across receptor systems, including opioids and serotonin receptors. By 2007, the term "functional selectivity" was formalized in pharmacological literature to encompass this ligand-directed bias more broadly, emphasizing its implications for quantitative pharmacology and drug design. The marked a , driven by advances in that enabled systematic detection of biased signaling profiles and that began elucidating receptor conformations. This culminated indirectly in the 2012 awarded to and for their work on GPCRs, which provided foundational insights into receptor activation states supportive of functional selectivity. More recently, since 2017, cryo-electron microscopy (cryo-EM) structures of GPCR-G protein complexes have confirmed the existence of biased conformations, visualizing how ligands stabilize distinct active states. Bias factor quantification emerged as a key analytical tool from these historical studies, allowing researchers to measure the degree of pathway preference relative to reference agonists.

Comparison to Traditional Concepts

Traditional Receptor Selectivity

Traditional receptor selectivity in refers to the classical model where a ligand's specificity for a particular receptor subtype is primarily determined by its affinity to that receptor's orthosteric site, assuming uniform activation of downstream signaling upon . This framework emerged in the early as part of receptor theory, emphasizing that drug effects result from reversible interactions at discrete sites on proteins. A foundational element of this model is A.J. Clark's occupancy theory, proposed in 1926, which posits that the intensity of a pharmacological response is directly proportional to the fraction of receptors occupied by an , following the . Clark's approach treated receptors as independent units where leads to a maximal response once fully occupied, without differentiating between ligand types beyond their concentration and . To address observations of partial agonists that elicited submaximal responses despite full occupancy, E.J. Ariëns extended the theory in 1954 by introducing the concept of intrinsic efficacy, which separates a ligand's for the receptor from its capacity to induce a conformational change necessary for activation. In this view, full agonists possess high intrinsic efficacy (approaching 1), while partial agonists have lower values, but selectivity remains tied to differences across receptor subtypes. Key measurements in traditional selectivity include dose-response curves, where potency is assessed via the half-maximal inhibitory concentration (IC50) or the equilibrium dissociation constant (d), often reported as pd or pKi (-log d or -log Ki) to quantify binding strength. Selectivity ratios are calculated as the quotient of affinities for non-target versus receptors (e.g., Kioff-target / Kitarget), with ratios greater than 100-fold typically indicating high subtype specificity to minimize off-target effects. The model assumes that orthosteric at a specific site triggers a singular, predictable cellular response proportional to and , without variation in downstream pathways. However, subsequent research has exposed limitations in this uniform activation assumption, as demonstrated by functional selectivity studies.

Distinguishing Features

Functional selectivity differs operationally from traditional receptor selectivity, which primarily assesses a ligand's for distinct receptor subtypes through assays, such as radioligand experiments that measure of a labeled from receptor sites. In contrast, functional selectivity evaluates a ligand's ability to produce differential efficacies across multiple intracellular signaling pathways activated by the same receptor, often using pathway-specific functional assays like those quantifying β-arrestin recruitment versus G protein-mediated responses, such as accumulation or production. These distinctions became prominent in pharmacological research during the early as evidence accumulated for ligand-biased signaling. Conceptually, traditional selectivity aligns with the "lock-and-key" model, where ligands bind uniformly to receptor subtypes to elicit consistent responses, whereas functional selectivity embodies a of multiple receptor active states, allowing ligands to stabilize conformations that preferentially activate certain pathways over . This shift is quantified using extensions of the operational model of , originally proposed by Black and Leff in 1983, which incorporates parameters for (τ) and bias factors to compare signaling outputs across pathways relative to a reference ligand. One key advantage of functional selectivity is its potential to achieve - or cell-type-specific effects by exploiting pathway biases inherent to different physiological contexts, without relying on receptor subtype differences. However, detecting and characterizing it poses challenges, as it necessitates parallel assays across multiple signaling pathways to identify and quantify bias, rather than single-endpoint measurements used in traditional approaches.

Underlying Mechanisms

Molecular Foundations

G protein-coupled receptors (GPCRs) and other receptors maintain a among multiple conformational states, forming an ensemble that underlies functional selectivity at the molecular level. Ligands function as stabilizers within this ensemble, preferentially shifting the receptor toward conformations that favor specific downstream interactions. This conformational selection arises from the intrinsic flexibility of the receptor structure, particularly in the transmembrane helices and intracellular domains. A hallmark of these conformational changes is the movement of transmembrane helix 6 (TM6), which adopts an outward displacement to accommodate coupling, thereby opening an intracellular binding pocket. Conformations stabilized by biased ligands may involve nuanced differences in TM6 positioning and other structural elements that differentially favor or β-arrestin recruitment, both typically featuring outward TM6 displacement. Ligand-induced allosteric modulation propagates structural changes across the receptor, influencing key intracellular elements that dictate pathway choice. The intracellular 2 (ICL2) stabilizes active conformations by forming interactions that guide effector , while 8 contributes to selectivity through its positioning and formations, which fine-tune the intracellular interface. These allosteric effects ensure that binding at the orthosteric site elicits targeted responses without uniform of all possible pathways. Biophysical tools, including (NMR) , have elucidated these ligand-receptor complexes by capturing dynamic shifts in . For instance, 19F-NMR studies from the 2010s revealed that biased ligands alter the equilibrium of transmembrane helices, such as TM6, to produce distinct active states in GPCRs. Complementary techniques like double electron-electron resonance (DEER) further quantify these conformational distances, confirming the heterogeneity of stabilized complexes. The quantitative foundation of functional selectivity lies in the landscapes governing conformational transitions. Biased ligands reshape this landscape by differentially modulating the energies of specific states, as described by for the difference: \Delta G = -RT \ln K where \Delta G represents the free energy change, R is the , T is the absolute temperature, and K is the between conformational states. This framework highlights how ligands lower the energy of favored conformations relative to others, promoting selective signaling. The determination of the β2-adrenergic receptor in 2007 provided an initial structural blueprint for understanding these conformational ensembles in GPCRs. Recent cryo-EM structures (as of 2025) and computational simulations have further elucidated these biased conformations, highlighting allosteric networks influencing effector selectivity.

Pathway-Specific

Functional selectivity manifests as pathway-specific , where ligands differentially engage intracellular signaling cascades downstream of the same receptor, leading to biased cellular responses. In G protein-coupled receptors (GPCRs), core pathways include G protein-mediated signaling, such as Gs coupling that elevates cyclic AMP () levels, Gi/o coupling that inhibits to decrease cAMP, and Gq/11 coupling that activates to produce (IP3) and diacylglycerol. Complementing these are β-arrestin-mediated pathways, which promote (MAPK)/extracellular signal-regulated kinase (ERK) and contribute to receptor desensitization and . Bias mechanisms arise from ligand-induced recruitment of specific effectors, where distinct agonists stabilize receptor states that preferentially interact with either s or β-arrestins. A key feature involves phospho-switch residues on the receptor's C-terminal tail, which, upon by kinases (GRKs), modulate coupling specificity between and β-arrestin pathways. These events create a "" that influences effector selectivity, enabling ligands to favor one pathway over another. Pathway crosstalk and occur through divergence at early signaling steps, notably varying GRK patterns that dictate downstream outcomes, such as enhanced β-arrestin signaling versus sustained activity. Quantitative models, including the operational model of , assess this by calculating transduction ratios (τ/KA), where τ represents and KA is the equilibrium dissociation constant, allowing comparison of pathway activation across ligands. Dose-response analyses further quantify by evaluating shifts in potency and ratios between pathways, revealing effects in divergent signaling. Beyond GPCRs, similar pathway-specific bias is observed in receptor tyrosine kinases (RTKs), where scaffold proteins such as proteoglycans and adhesion molecules regulate signaling specificity by directing receptor trafficking and complex assembly, leading to differential activation of downstream cascades like MAPK or PI3K pathways.

Key Examples

G Protein-Coupled Receptors

Functional selectivity, also known as biased agonism, is prominently observed in G protein-coupled receptors (GPCRs), where ligands can preferentially activate specific signaling pathways such as G protein-mediated effects over β-arrestin recruitment, leading to tailored physiological outcomes. In GPCRs, this selectivity arises from ligand-induced stabilization of distinct receptor conformations that differentially engage downstream effectors, allowing for dissociation of therapeutic benefits from adverse effects. A key example is the mu-opioid receptor (MOR), where traditional agonists like recruit both and β-arrestin pathways, promoting analgesia via signaling but also engaging β-arrestin to cause respiratory depression. In contrast, -biased agonists such as selectively activate pathways to enhance analgesia while minimizing β-arrestin-mediated side effects like respiratory suppression. , approved by the FDA on August 7, 2020, for acute , represents a clinical advancement in this approach. At the II type 1 receptor (AT1R), saralasin, a /, activates both Gq protein and β-arrestin pathways with lower efficacy than full agonists, contributing to and cardiac . The biased ligand TRV027, however, displays β-arrestin bias, favoring and cardioprotective effects without the hypertrophic responses associated with Gq signaling. This selectivity has been explored in models, where TRV027 unloads the heart while preserving renal function. For the β2-adrenergic receptor (β2AR), demonstrates β-arrestin bias despite its primary role as a β-blocker, promoting cardioprotective signaling independent of activation. Quantitative bias assessments using bioluminescence resonance energy transfer (BRET) assays reveal that recruits β-arrestin with high efficacy, enhancing contractility in cardiac and without the tachycardic effects of pathways. This bias contributes to 's efficacy in chronic treatment. The D2 receptor (D2R) illustrates partial functional selectivity with aripiprazole, which shows bias toward Gi/o protein signaling over β-arrestin recruitment, supporting effects by stabilizing in hyperdopaminergic states. This Gi/o preference allows aripiprazole to act as a under high tone and an under low tone, reducing extrapyramidal side effects compared to unbiased antagonists. As of 2025, ongoing research continues to develop new biased ligands, with clinical trials evaluating additional G-protein-biased opioids to further mitigate side effects beyond .

Ion Channel and Enzyme Receptors

Functional selectivity manifests in receptors through ligand-induced conformational changes that differentially modulate permeability, desensitization kinetics, and downstream signaling, distinct from the pathway biases observed in G protein-coupled receptors. In the family, particularly the homomeric α7 subtype (α7 nAChR), which exhibits high calcium permeability and rapid desensitization, s can preferentially enhance calcium influx while altering desensitization profiles. For instance, the selective α7 PNU-282987 activates the receptor to promote calcium-dependent in retinal cells, supporting and survival. Noncanonical s further demonstrate differential desensitization states, with recovery rates varying based on structural features, underscoring conformational selectivity in control. Enzyme-linked receptors, such as receptor s, exhibit functional selectivity via ligand-dependent dimerization and adaptor protein , leading to biased activation of signaling cascades. In the (), natural ligands like (EGF) and transforming growth factor-α (TGF-α) stabilize distinct receptor dimer conformations, directing trafficking and pathway outcomes. EGF preferentially engages the Grb2-SOS complex to activate the /MAPK pathway, promoting , whereas TGF-α biases toward Shc-PI3K , enhancing Akt-mediated signaling. This selectivity arises from differences in ligand-receptor binding kinetics and allosteric coupling, allowing pathway-specific responses despite shared orthosteric sites. Similarly, in the , a with isoforms IR-A and IR-B, ligands and modulators can bias between metabolic (e.g., IRS-PI3K-glucose uptake) and mitogenic (e.g., Shc-MAPK-cell growth) signaling; for example, engineered insulin dimers act as partial agonists, favoring metabolic effects with reduced mitogenic potential. agonists further illustrate this by allosterically modulating the receptor to selectively enhance IRS-1 over Shc, demonstrating isoform-specific bias in 2010s structural studies. Emerging evidence in nuclear receptors, which function enzymatically in , reveals functional selectivity through ligand-specific coactivator recruitment. For γ (PPARγ), partial and full agonists induce distinct conformational ensembles that differentially engage coactivators like PGC-1α, biasing toward anti-diabetic (e.g., ) or anti-inflammatory profiles over full agonism's side effects. This mechanism, akin to biased agonism in membrane receptors, allows selective modulation of coregulator binding motifs (LXXLL), as shown in structural analyses of ligand-bound complexes.

Implications for Research and Therapy

Therapeutic Opportunities

Functional selectivity, also known as biased , offers significant therapeutic opportunities by allowing ligands to preferentially activate specific signaling pathways downstream of receptors, thereby enhancing while minimizing off-target effects and adverse reactions. In precision medicine, this pathway bias can reduce side effects associated with traditional agonists; for instance, G protein-biased agonists at the mu-opioid receptor (MOR), such as (TRV130; approved by the FDA in 2020 for the management of acute pain severe enough to require an intravenous ), provide analgesia with decreased risk of respiratory compared to balanced agonists like , as they avoid β-arrestin-mediated pathways linked to tolerance and reward. Similarly, for (GLP-1R) agonists in management, biased variants that favor G signaling over β-arrestin recruitment have shown potential to maintain glycemic control while attenuating gastrointestinal side effects like , which affect up to 50% of patients on conventional therapies. Drug design strategies leveraging functional selectivity include structure-based screening and computational modeling to identify biased ligands that exploit conformational differences in receptor states. For example, cryo-EM and have guided the development of ligands with tailored bias profiles, enabling for pathway-specific activation. Clinical translation is exemplified by TRV027, a β-arrestin-biased II type 1 receptor (AT1R) tested in phase 2 trials for COVID-19-associated in the early 2020s, where it aimed to enhance and cardiac contractility without the vasoconstrictive side effects of balanced s, although the trials did not demonstrate significant clinical benefits, such as improved oxygen-free days. These approaches underscore the shift toward designing drugs that fine-tune receptor outputs for optimized therapeutic windows. In disease-specific applications, functional selectivity holds promise across multiple therapeutic areas. In , β-arrestin-biased β1-adrenergic receptor (β1AR) agonists like improve outcomes in by promoting cardioprotective signaling, such as ERK activation, while mitigating arrhythmogenic effects from pathways. For neurology, in , β-arrestin-biased D2 agonists selectively modulate pathways, reducing dyskinesia risks associated with chronic levodopa use through preferential avoidance of certain couplings. Looking ahead, the integration of with functional selectivity profiling promises paradigms, where patient-specific genetic variations in receptor or transducer expression (e.g., polymorphisms in GPCR genes) guide the selection of biased ligands to match individual signaling biases, potentially maximizing therapeutic response and safety in heterogeneous populations. This approach could revolutionize treatments for complex disorders like , , and neurodegeneration by enabling predictive bias matching based on genomic data.

Methodological Challenges

One major challenge in studying functional selectivity lies in the limitations of available assays, which often fail to capture the full spectrum of signaling pathways without introducing biases. Traditional assays like the [35S]-GTPγS binding assay effectively detect activation but cannot distinguish between different Gα subtypes, restricting their utility for pathway-specific analysis. To address this, orthogonal readouts are essential, such as ELISA for Gαs/i-mediated signaling versus Tango assays for β-arrestin recruitment, allowing comparative assessment of biased agonism across pathways. However, these assays are prone to cell-type dependency, where signaling outcomes vary due to differences in receptor localization, coupling partners, and tissue context, complicating direct comparisons between systems. Additionally, overexpression artifacts in engineered cell lines, such as HEK293 or cells, can inflate receptor density and potency (e.g., 5–10-fold lower values compared to endogenous expression), leading to non-physiological results that obscure true ligand bias. Quantifying functional selectivity introduces further pitfalls, particularly in the variability of bias factors across experimental systems. The bias factor, derived from differences in coefficients like ΔΔlog(τ/KA), measures pathway preference but is sensitive to system-specific factors such as receptor and efficiency, resulting in inconsistent values that hinder cross-study comparisons. For instance, like CCL3L1 exhibit bias factors of 23.7 toward CCR5 internalization over IP1 production in U373 cells, but these shift in other cell types due to varying signaling profiles. Statistical approaches, including global fitting to the Black-Leff operational model, help estimate τ/KA independently of receptor , but they require precise maximal response (E_m) determination and can suffer from parameter identifiability issues in skewed datasets. Translating observations of functional selectivity to contexts reveals significant gaps, as biased signaling in systems often fails to predict physiological outcomes. Species differences exacerbate this, with analogs like PTH(1-34) showing divergent bone formation responses in mice versus humans due to variations in receptor expression and downstream effectors. , biased ligands interact with endogenous hormones and native tissues, leading to "unbalanced" effects not seen , such as mixed behaviors in osteoblasts. Regulatory hurdles compound these issues, as the unpredictable nature of β-arrestin versus pathways demands extensive validation, slowing approval of biased therapeutics. Emerging solutions aim to mitigate these challenges through advanced cellular engineering and computational tools. /Cas9-edited cell lines, such as those lacking specific β-arrestins (e.g., βArr1/2 knockouts in HEK293 cells), enable pathway-specific dissection of GPCR signaling, revealing how deletions shift ERK1/2 activation toward dependence while avoiding siRNA off-target effects. Similarly, post-2020 AI-driven methods like AlphaFold2 and AlphaFold-Multimer predict GPCR conformations with high accuracy (e.g., TM domain RMSD ~1 Å), generating state-specific models for active/inactive transitions that inform ligand-induced functional selectivity without relying solely on experimental structures. These approaches, including AlphaFold3 for receptor-ligand co-folding, facilitate of biased agonists by accounting for induced-fit dynamics.

References

  1. [1]
  2. [2]
  3. [3]
  4. [4]
  5. [5]
  6. [6]
    Quantifying Functional Selectivity & Agonist Bias
    Utilizing a “transduction coefficient” term, log(τ/KA), this scale can statistically evaluate selective agonist effects in a manner that can theoretically ...
  7. [7]
    Biased agonism: An emerging paradigm in GPCR drug discovery
    This review will discuss the current understanding of some of the key aspects of biased signaling that are related to these questions, including mechanistic ...Missing: 2020-2025 | Show results with:2020-2025
  8. [8]
    How New Developments in Pharmacology Receptor Theory Are ...
    The goal of this review is to provide an up-to-date summary of drug receptor theory. This is followed by a discussion of the drug classes recognized for ...
  9. [9]
    Why classical receptor theory, which ignores allostery, can ...
    Feb 11, 2024 · The classical theory of receptor action has been used for decades as a powerful tool to estimate molecular determinants of ligand-induced receptor activation.Abstract · INTRODUCTION · RESULTS · DISCUSSION
  10. [10]
    The reaction between acetyl choline and muscle cells - Clark - 1926
    The reaction between acetyl choline and muscle cells. A. J. Clark,. A. J. ... https://doi.org/10.1113/jphysiol.1926.sp002314. Citations: 188. About. Related ...Missing: acetylcholine | Show results with:acetylcholine
  11. [11]
    100 years of modelling ligand–receptor binding and response: A ...
    Jan 23, 2020 · The idea of occupancy is first implied by Clark's (1926b) observation that the action of ACh is proportional to the degree of “combination ...
  12. [12]
    Affinity and intrinsic activity in the theory of competitive inhibition. I ...
    Affinity and intrinsic activity in the theory of competitive inhibition. I. Problems and theory. ... 1954 Sep 1;99(1):32-49. Author. E J ARIENS. PMID: 13229418.Missing: receptor | Show results with:receptor
  13. [13]
    A Receptor Model With Binding Affinity, Activation Efficacy, and ...
    It is also similar to the intrinsic activity αA introduced by Ariëns (1954), which is the ratio of the maximum response produced by the partial agonist to that ...
  14. [14]
    Receptor theory of drug action | Deranged Physiology
    Dec 28, 2017 · Occupancy theory rests on the concept that the proportion of occupied receptors is related to the effect of the drug (eg. for full agonists and ...
  15. [15]
    Affinity, potency, efficacy, and selectivity of neurokinin A analogs at ...
    Oct 25, 2018 · The endogenous peptide, NKA, lacked selectivity with an NK1/NK2 Ki ratio = 20 and NK1/NK2 EC50 ratio = 1. Of the compounds selected for ...
  16. [16]
    The Different Ways through Which Specificity Works in Orthosteric ...
    In orthosteric drugs, high specificity implies high affinity and selectivity, to avoid unwanted side effects. This points to drugs whose shape and chemistry ...
  17. [17]
    Making Sense of Pharmacology: Inverse Agonism and Functional ...
    The second topic discussed is functional selectivity, also commonly referred to as biased agonism. Traditional receptor theory also posited that intrinsic ...
  18. [18]
    Radioligand binding assays and their analysis - PubMed
    The affinity and selectivity of an unlabeled ligand to compete for the binding of a fixed concentration of a radiolabeled ligand to a receptor are determined ...Missing: traditional | Show results with:traditional
  19. [19]
    Functional selectivity and classical concepts of quantitative ...
    At the extreme, functionally selective ligands may be both agonists and antagonists at different functions mediated by the same receptor. Data illustrating this ...
  20. [20]
    Operational models of pharmacological agonism - Journals
    The result is a general model that explicity describes agonism by three parameters: an agonist-receptor dissociation constant, KA; the total receptor ...
  21. [21]
    Structural insights into biased G protein-coupled receptor ... - PNAS
    Cell-based studies suggest that functional selectivity arises as a result of distinct conformational states of the receptor stabilized by the ligands (12, 13).
  22. [22]
    GPCR activation mechanisms across classes and macro/microscales
    Nov 10, 2021 · We present a GPCR superfamily-wide molecular mechanistic map of activation, and link determinants to ligand-binding, G-protein coupling, transduction and ...
  23. [23]
    G protein-coupled receptors (GPCRs): advances in structures ...
    Apr 10, 2024 · Over the past 30 years, the widespread use of X-ray and Cryo-EM has facilitated the characterization of GPCR-orthosteric ligand complexes, with ...
  24. [24]
    Ligand-Induced Modulation of the Free-Energy Landscape of G ...
    Compelling evidence herein referred to as 'functional selectivity' shows that ligands with varied efficacies can stabilize different GPCR conformations that may ...
  25. [25]
    Crystal structure of the human β2 adrenergic G-protein ... - Nature
    Oct 21, 2007 · Here we report a structure of the human β 2 adrenoceptor (β 2 AR), which was crystallized in a lipid environment when bound to an inverse agonist.
  26. [26]
  27. [27]
  28. [28]
  29. [29]
  30. [30]
  31. [31]
    Functional selectivity profiling of the angiotensin II type 1 receptor ...
    Dec 4, 2018 · G protein–coupled receptors (GPCRs) are important therapeutic targets that exhibit functional selectivity (biased signaling), in which different ...
  32. [32]
    Olinvyk (oliceridine) FDA Approval History - Drugs.com
    Sep 17, 2020 · FDA Approved: Yes (First approved August 7, 2020) ; Brand name: Olinvyk ; Generic name: oliceridine ; Dosage form: Injection ; Previous Name: Olinvo
  33. [33]
    Distinct Mechanisms of β-Arrestin–Biased Agonist and Blocker of ...
    Nov 28, 2022 · TRV027-engaged AT1R prevented AA and associated mortality by distinct molecular mechanisms compared with the AT1R blocker, Olmesartan.
  34. [34]
    Biased ligand of the angiotensin II type 1 receptor in patients with ...
    Aug 7, 2017 · TRV027 is a novel 'biased' ligand of the angiotensin II type 1 receptor (AT1R), selectively antagonizing the negative effects of angiotensin II.
  35. [35]
    How Carvedilol activates β2-adrenoceptors | Nature Communications
    Nov 19, 2022 · The beta-arrestin-biased beta-adrenergic receptor blocker carvedilol enhances skeletal muscle contractility. Proc. Natl Acad. Sci. USA 117 ...
  36. [36]
    β-arrestin–biased signaling through the β2-adrenergic receptor ...
    Jun 27, 2016 · Carvedilol, biochemically characterized as a β-arrestin–biased agonist, can promote β-arrestin–mediated processes, such as receptor ...
  37. [37]
    The β-arrestin-biased β-adrenergic receptor blocker carvedilol ...
    May 15, 2020 · Carvedilol, classically defined as a βAR antagonist, is widely used for the treatment of chronic systolic heart failure and hypertension, and ...Missing: BRET | Show results with:BRET
  38. [38]
    Aripiprazole has Functionally Selective Actions at Dopamine D2 ...
    Mar 22, 2006 · The results are consistent with the hypothesis that aripiprazole is a functionally selective D 2 ligand rather than a simple partial agonist.
  39. [39]
    Discovery of G Protein-biased D2 Dopamine Receptor Partial Agonists
    We unexpectedly discovered a G protein-biased agonist of D2R, compound 1, which is the first G protein-biased D2R agonist from the aripiprazole scaffold.
  40. [40]
    Alpha7 nicotinic acetylcholine receptor agonist promotes retinal ...
    May 11, 2017 · In this study, we investigated whether a highly selective α7-nAChR agonist (PNU-282987) promotes RGC survival and functional recovery and ...
  41. [41]
    α7 nicotinic acetylcholine receptor and depression
    The α7 nAChR agonist PNU-282987 does not generally produce robust antidepressant-like effects when administered alone, but it potentiates behavioral effects ...
  42. [42]
    Differential Activation and Desensitization States Promoted by ...
    DPPs activate α7 receptors, causing rapid desensitization. Desensitization recovery varies, and noncanonical agonists promote unique conformational changes. ...
  43. [43]
    EGFR Ligands Differentially Stabilize Receptor Dimers to Specify ...
    Oct 19, 2017 · Our results show how biased agonism or functional selectivity can occur with EGFR and its natural human ligands. We explain the structural ...
  44. [44]
    Quantification of ligand and mutation-induced bias in EGFR ... - Nature
    Nov 21, 2023 · ... functional selectivity in EGFR signaling. Experiments were ... G protein-coupled receptor kinases (GRKs) orchestrate biased agonism at the beta2- ...
  45. [45]
    Functionally selective signaling and broad metabolic benefits by ...
    Feb 17, 2022 · ... insulin receptor (IR) tyrosine kinase is achievable, and to explore ... functional selectivity. As a result, inherent liabilities (e.g. ...
  46. [46]
    Agonistic aptamer to the insulin receptor leads to biased ... - PubMed
    Sep 18, 2015 · Agonistic aptamer to the insulin receptor leads to biased signaling and functional selectivity through allosteric modulation. Nucleic Acids ...
  47. [47]
    Agonists of the Nuclear Receptor PPARγ Can Produce Biased ...
    Nov 18, 2024 · Biased signaling and ligand bias, often termed functional selectivity ... biased agonism. Our data support the idea that partial agonists, and ...
  48. [48]
    A structural mechanism of nuclear receptor biased agonism - PubMed
    Dec 13, 2022 · ... coactivator complexes ... Keywords: biased agonism; fluorescence anisotropy; functional selectivity; nuclear receptor; selective modulator.
  49. [49]
    Untangling the complexity of opioid receptor function - Nature
    Sep 24, 2018 · Since their discovery in the 1970's, there have been major advances in our understanding of the endogenous opioid systems that these drugs ...
  50. [50]
    Autocrine selection of a GLP-1R G-protein biased agonist ... - Nature
    Dec 1, 2015 · Glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists have emerged as treatment options for type 2 diabetes mellitus (T2DM).Missing: opportunities | Show results with:opportunities
  51. [51]
    Targeting GLP-1 receptor trafficking to improve agonist efficacy
    Apr 23, 2018 · Nausea is a side effect which affects 30–50% of patients taking GLP-1R agonists at clinically licensed doses, with higher doses glycemically ...
  52. [52]
    G protein-coupled receptors: structure- and function-based drug ...
    Jan 8, 2021 · The structure of GPCRs is a crucial determinant for understanding the molecular mechanisms underlying ligand recognition and receptor activation ...<|separator|>
  53. [53]
    Heterotrimeric Gq proteins act as a switch for GRK5/6 selectivity ...
    Jan 25, 2022 · TRV027 was developed as a β-arrestin-biased AT1R ligand that induces preferential engagement of Gq over β-arrestin and is tested in a clinical ...<|control11|><|separator|>
  54. [54]
    Gαi is required for carvedilol-induced β1 adrenergic receptor β ...
    Nov 22, 2017 · Some βAR ligands, such as carvedilol, stimulate βAR signaling preferentially through β-arrestin, a concept known as β-arrestin-biased agonism.<|control11|><|separator|>
  55. [55]
    Bias analyses of preclinical and clinical D2 dopamine ligands
    Dec 24, 2014 · These unique responses provide opportunities for biased or functionally selective ligands to preferentially modulate one signaling pathway over ...
  56. [56]
    Intracellular GPCR modulators enable precision pharmacology
    May 12, 2025 · GPCR biased signaling also known as functional selectivity refers to the phenomenon where a given ligand (such as a drug or endogenous ...
  57. [57]
    Approaches to Assess Functional Selectivity in GPCRs - NIH
    This report presents our method and offers tips for evaluating G protein signaling in endogenous tissues.
  58. [58]
    Translating in vitro ligand bias into in vivo efficacy - PMC - NIH
    May 7, 2017 · Abstract. It is increasingly apparent that ligand structure influences both the efficiency with which G protein-coupled receptors (GPCRs) ...Missing: gaps | Show results with:gaps
  59. [59]
    Manifold roles of β-arrestins in GPCR signaling elucidated ... - Science
    Sep 25, 2018 · Studies based on CRISPR/Cas9-generated cell lines suggested that β-arrestins are dispensable for ERK1/2 activation. Luttrell et al. compared the ...β-Arrestin--Dependent... · Materials And Methods · Bret Assay Of Gα Activation...
  60. [60]
    AI meets physics in computational structure-based drug discovery ...
    Jul 3, 2025 · In this review, we discuss the recent innovation in artificial intelligence- (AI) and physics-based computational methodologies that advance SBDD for GPCRs.