ABSTRACT
The discovery of biased agonism as a pharmacological technique that enables specific receptor pathway activation through ligands transformed receptor pharmacology. The analysis evaluates the fundamental mechanisms and medical implications alongside the development hurdles for biased agonist drug discovery. The research analyzed PubMed alongside Scopus and Web of Science databases for articles between 2000 and 2024 under keywords that included “biased agonism” “GPCR signalling” and “functional selectivity.” The review included articles about biased agonist mechanistic aspects together with clinical applications and regulatory frameworks. The study demonstrates how biased agonism enables therapeutic benefits through reduced unwanted effects specifically within opioid and adrenergic signalling pathways. Clinical studies of oliceridine and carvedilol demonstrate proof-of-concept yet their transition from laboratory research to clinical applications faces ongoing challenges. The context-dependent nature of receptor signalling together with methodological inconsistencies prevents accurate translation. Our analysis reveals the main challenges of assay inconsistency together with regulatory ambiguity and the reduction of complex signalling to simple dichotomies. New technologies which include cryo-EM organoids and AI-driven ligand screening systems provide predictive frameworks to scientific research. The practice of precision pharmacotherapy shows great potential through biased agonism. Biased agonism requires collaboration between different fields together with improved biomarkers and revised regulatory standards to achieve full therapeutic benefits.
INTRODUCTION
According to traditional receptor theory agonist binding produces equivalent activation of every linked signalling pathway. Biased agonists produce different receptor shapes that lead to the activation of particular intracellular signalling molecules (Kenakin, 2011). The first description of biased agonism occurred in G-Protein-Coupled Receptors (GPCRs) since they represent the largest drug-targeted receptor family in pharmacology.
The dual-pathway activation (e.g., G protein vs. β-arrestin) opens avenues to fine-tune therapeutic responses. Through biased activation of μ-opioid receptors oliceridine achieves analgesic effects through G protein pathways while reducing adverse effects related to β-arrestin activation (DeWire et al., 2013). The method now applies to drug development beyond GPCRs through Receptor Tyrosine Kinases (RTKs) and nuclear receptors (Rittiner et al., 2023). In this review aims to explain the mechanistic basis of biased agonism, evaluate current methodologies to quantify it, highlight clinically validated biased agonists, examine its therapeutic potential and pitfalls, and suggest future research directions.
METHODOLOGY
This is a narrative review. A literature search was conducted using PubMed, Scopus, and Web of Science (2000-2024) with keywords: biased agonism, GPCR signalling, functional selectivity, β-arrestin, drug discovery. This review included peer-reviewed English-language articles focusing on mechanistic and preclinical or clinical aspects of biased agonists. The research excluded studies written in non-English languages together with preprints and non-peer-reviewed literature. This review was not registered in PROSPERO because it fails to meet systematic review criteria.
MECHANISTIC BASIS OF BIASED AGONISM
Biased agonism occurs through ligand-induced stabilization of receptor conformations that activate particular intracellular signalling pathways (Van der Westhuizen et al., 2014). Research indicates that traditional views about receptors having a single “active” conformation do not apply because receptors instead exist in equilibrium between various active and inactive states. The chemical structure along with binding kinetics of ligands determines which receptor conformations become preferred to interact with specific effectors.
GPCRs function as the fundamental model of biased agonism because one receptor interacts with both G proteins and β-arrestins among its downstream effectors.
The rapid second-messenger signals cyclic AMP (cAMP), Inositol Trisphosphate (IP3) and calcium mobilization originate from G protein interactions.
The GPCR signalling process is desensitized through β-arrestin interaction which leads to receptor internalization alongside the activation of ERK1/2 p38 MAPK and JNK pathways without G protein involvement (Luttrell and Lefkowitz, 2002).
The structural mechanisms behind biased agonism have become clearer through recent advances in cryo-Electron Microscopy (cryo-EM) combined with molecular dynamics simulations. The techniques reveal that different ligands produce distinct receptor shapes which lead to particular effector coupling patterns (Shukla et al., 2011; Weis and Kobilka, 2018).
The binding kinetics between ligands and receptors can be monitored through NMR spectroscopy to show how they affect the degree and direction of signalling bias (Mangliket al., 2015). Non-orthosteric receptor conformations become stabilized by allosteric modulators which can either enhance or inhibit bias (Christopoulos, 2014).
The cellular environment plays a vital role in the modulation process. Different factors including receptor density and G protein subtypes and scaffolding proteins and membrane lipid composition determine both the magnitude and type of biased signalling (Smith and Rajagopal, 2016; Thomsen et al., 2016). Research shows that the duration a ligand stays on a receptor surface determines receptor conformational changes which lead to signalling results (Copeland, 2016).
Biased agonism arises from the dynamic interactions between receptors and ligands and effectors within cellular environments rather than being a ligand property. The drug development process needs to include this mechanistic complexity to achieve reproducible and clinically meaningful results (Eddy et al., 2018).
QUANTIFICATION AND MEASUREMENT OF BIASED AGONISM
Biased agonism is quantified by comparing ligand-induced responses across different signalling pathways:
- Operational Model of Agonism (Black-Leff Model),
- Transduction Coefficient,
- Equiactive Comparison: comparing ligand responses at the same effect level (Michel et al., 2020).
BRET, FRET and impedance-based label-free assays serve as tools for functional bias profiling. Standardized protocols remain essential because assay system variability such as receptor expression levels generates biased results (Dahan et al., 2020). Machine learning tools predict bias from ligand-receptor interaction datasets (Rodríguez et al., 2021).
CLINICALLY RELEVANT BIASED AGONISTS
Oliceridine
The FDA approved oliceridine (TRV130) as a μ-opioid Receptor (MOR) agonist to treat moderate-to-severe acute pain in adults when traditional pain treatments fail (U.S. FDA, 2020). Oliceridine differs from traditional opioids because it activates G protein-biased agonism at the μ-opioid receptor (Table 2). The μ-opioid receptor agonist oliceridine activates G protein pathways that produce analgesia without triggering β-arrestin-2 activation which leads to adverse effects such as respiratory depression and constipation and opioid tolerance (DeWire et al., 2013). The drug’s selective signalling properties are thought to provide enhanced safety benefits.
Feature | G Protein Pathway | β-Arrestin Pathway |
---|---|---|
Activation | cAMP, Ca²⁺ | ERK, JNK, receptor internalization |
Signal Duration | Transient | Sustained. |
Therapeutic Role | Primary response | Modulatory/counter-regulatory |
Associated with | Analgesia, cardiac effects | Desensitization, side effects |
Tools for detection | cAMP assay, calcium flux | BRET/FRET, phosphorylation assays |
Drug | Target Receptor | Bias Direction | Clinical Indication | Status |
---|---|---|---|---|
Oliceridine | μ-Opioid receptor | G protein-biased | Acute pain | Approved |
Carvedilol | β1-Adrenergic receptor | β-arrestin-biased | Heart failure | Approved |
TRV027 | AT1R (angiotensin) | β-arrestin-biased | Acute HF | Failed Phase II |
Biased 5-HT2A ligands | Serotonin receptor | G protein-biased | Mood disorders | Preclinical |
Biased D2 agonists | Dopamine receptor | G protein-biased | Schizophrenia | Early Clinical |
Preclinical experiments on animals revealed that oliceridine produced strong pain relief together with substantially decreased respiratory and gastrointestinal adverse effects when compared to morphine administration (DeWire et al., 2013; Manglik et al., 2016). The experimental findings created hope that biased agonism could lead to a breakthrough in opioid treatment methods. The pharmacokinetic profile of oliceridine shows rapid intravenous delivery which starts pain relief within 2-5 min while having a short elimination period between 1.3 to 3 hr. The drug undergoes primary metabolism through CYP3A4 and CYP2D6 enzymes while showing 77% plasma protein binding according to the U.S. FDA (2020). The compound leaves the body through liver processing along with urinary waste.
The positive results from preclinical studies failed to translate into substantial benefits during clinical trial phases. The Phase 3 clinical trials APOLLO-1 and APOLLO-2 showed that oliceridine delivered analgesia equivalent to morphine for postoperative pain management (Viscusi et al., 2019; Singla et al., 2019). The clinical trials showed a possible decrease in respiratory safety complications alongside reduced gastrointestinal side effects although these results failed to reach statistical significance. Oliceridine preserves its potential for abuse despite being classified as a Schedule II controlled substance by the U.S. FDA (U.S. FDA, 2020). The adverse effects of oliceridine include nausea, vomiting, sedation and respiratory depression but their intensity may be lower in particular clinical contexts.
The clinical advantages of oliceridine as a G protein-biased opioid approved by the FDA are restricted because it does not offer substantial benefits over conventional opioids. Oliceridine serves only as a short-term treatment under hospital supervision due to its fast-acting IV formulation. The drug functions primarily to validate biased agonism concepts rather than provide a revolutionary pain management solution. Research into new biased ligands continues because they show promise to improve both efficacy and safety while maintaining high selectivity (Manglik et al., 2016; Kliewer et al., 2020).
Carvedilol
Carvedilol functions as a non-selective β-adrenergic receptor blocker which provides additional α₁-adrenergic blocking properties and serves as a common medication for heart failure and hypertension treatment. Research has elevated carvedilol’s status to a β-arrestin-biased ligand at the β₁-Adrenergic Receptor (β₁-AR) in addition to its established function as a G Protein-Coupled Receptor (GPCR) antagonist (Table 1). Carvedilol functions differently from standard β-blockers since it blocks G protein pathways while stimulating β-arrestin-dependent pathways particularly EGFR transactivation which results in its beneficial cardioprotective effects (Wisler et al., 2007).
Carvedilol blocks the Gs protein-coupled signalling cascade that produces cyclic AMP thus decreasing heart rate and contractility which helps patients with reduced ejection fraction heart failure. Unlike other β-blockers carvedilol both blocks the β₁-AR from G protein signalling and recruits β-arrestin to activate downstream pathways that do not need G proteins. The activation of ERK1/2 pathways through EGFR transactivation occurs as part of β-arrestin signalling which leads to cardiomyocyte survival and prevents cell death (Kim et al., 2008). The drug functions beyond antagonist properties to exhibit functional selectivity which positions it as an early clinical application of biased ligands.
Research demonstrates carvedilol triggers β-arrestin-mediated signalling to enhance cardiac remodeling processes and reduce oxidative stress while protecting myocytes from death particularly during β-adrenergic receptor overactivation found in heart failure patients (Noma et al., 2007). These therapeutic advantages operate independently from blood pressure reduction and do not apply to all β-blockers. Research comparing carvedilol to metoprolol in animal experiments confirmed that carvedilol specifically triggered β-arrestin-dependent ERK signalling because of its unique signalling bias (Wisler et al., 2007; Kim et al., 2008).
Carvedilol comes in oral form and its bioavailability reaches 25-35% before undergoing extensive first-pass metabolism through CYP2D6 and CYP2C9 enzymes. The half-life of carvedilol ranges between 7 to 10 hr and it binds heavily to proteins while primarily eliminating through bile and feces. The drug presents as a racemic mixture where the S-enantiomer performs β-blockade functions and the R-enantiomer provides more α₁-blocking activity (Rehsia and Dhalla, 2010). Its dual pharmacological properties create an effective treatment solution for lowering blood pressure and managing afterload while reducing sympathetic nervous system activity.
The medical literature shows carvedilol lowers death rates together with hospital readmissions among patients who have chronic heart failure based on results from both COPERNICUS and COMET trials. The biased signalling properties of carvedilol have gained recognition as vital contributors to its protective effects on the heart although its clinical benefits are traditionally linked to its dual β- and α-blocking activities (Lymperopoulos et al., 2013). Research continues to develop β-arrestin-biased ligands beyond carvedilol to optimize heart protection while minimizing standard β-blocker adverse effects.
The therapeutic application of carvedilol demonstrates how biased agonism functions through blocking dangerous G protein-mediated β₁-AR signalling while simultaneously activating protective β-arrestin pathways. The drug’s distinct pharmacological characteristics introduce a new concept of adrenergic drug action which will guide development of future cardio-selective agents that use signalling bias advantages.
TRV027
Other Examples
Biased agonism offers several benefits during drug discovery processes
- Enhanced Therapeutic Selectivity: Tailored pathway activation reduces side effects
- Reduced Tolerance: The absence of β-arrestin-mediated desensitization leads to sustained efficacy.
- Drug Repurposing: Biased signalling can rescue previously abandoned ligands.
- Precision Medicine: Biased ligands allow context-specific receptor targeting.
- Intellectual Property Advantages: Novel scaffolds enable extended patent lifespans (Christopoulos, 2002).
Pitfalls and Limitations
- In vitro vs. In vivo Disconnect: Ligand bias often fails to predict clinical efficacy,
- Context Dependence: Cell type, receptor reserve and assay systems influence the results of bias experiments (Onaran and Costa, 2012),
- Over-Simplification: Pathway categorization into beneficial or harmful types can lead to errors since β-arrestins exhibit detrimental effects in particular situations,
- Regulatory Barriers: The drug approval systems do not have established criteria for pathway-selectivity (Iijima et al., 2020),
- Interpretation Risks: Small differences in bias may be over-interpreted.
Emerging Trends and Future Directions
- AI/ML Tools: Predicting ligand bias using structural fingerprints and deep learning,
- Allosteric Modulators: Targeting non-orthosteric sites to promote functional selectivity,
- Beyond GPCRs: Biased signalling now observed in RTKs, cytokine receptors, and nuclear receptors,
- Organoids and Organ-on-Chip: Enhancing physiological relevance of functional assays. (Low et al., 2021),
- Regulatory Innovations: Emphasis on novel biomarkers, patient stratification, and adaptive trials for biased ligands.
CONCLUSION
The drug discovery approach of biased agonism transforms drug development by providing improved selectivity and efficacy. Its clinical application remains limited due to methodological inconsistencies and regulatory gaps as well as the complex nature of receptor signalling. Future success will depend on Standardized functional assays, Robust translational models, Clear regulatory guidelines, Interdisciplinary collaboration, Biased agonism will serve as a fundamental element in the development of upcoming precision pharmacotherapy treatments.
Cite this article:
Ananthy V, Palanisamy PR, Umamaheswari S. Biased Agonism in Drug Discovery: Clinical Promise and Pitfalls. J Young Pharm. 2025;17(3):532-6.
ACKNOWLEDGEMENT
The authors express deepest gratitude to their colleagues and students who shared their valuable insights about GPCR signalling and biased agonism which influenced the content of this review.
References
- Christopoulos A.. (2002) Allosteric binding sites on cell-surface receptors: Novel targets for drug discovery. Nature Reviews. Drug Discovery 1: 198-210 https://doi.org/10.1038/nrd746 | Google Scholar
- Christopoulos A.. (2014) Advances in G protein-coupled receptor allostery: From function to structure. Molecular Pharmacology 86: 463-478 https://doi.org/10.1124/mol.114.094342 | Google Scholar
- Copeland R. A.. (2016) The drug-target residence time model: A 10-year retrospective. Nature Reviews. Drug Discovery 15: 87-95 https://doi.org/10.1038/nrd.2015.18 | Google Scholar
- Dahan A., van Dam C. J., Niesters M.. (2020) Translational issues in biased opioid pharmacology. British Journal of Anaesthesia 125: e212-e226 https://doi.org/10.1016/j.bja.2020.02.018 | Google Scholar
- DeWire S. M., Yamashita D. S., Rominger D. H., Liu G. X., Cowan C. L., Graczyk T. M., Chen X.-T., Pitis P. M., Gotchev D., Yuan C., Koblish M., Lark M. W., Violin J. D., et al. (2013) A G protein-biased ligand at the μ-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine. The Journal of Pharmacology and Experimental Therapeutics 344: 708-717 https://doi.org/10.1124/jpet.112.201616 | Google Scholar
- Eddy M. T., Lee M.-Y., Gao Z.-G., White K. L., Didenko T., Horst R., Audet M., Stanczak P., McClary K. M., Han G. W., Jacobson K. A., Stevens R. C., Wüthrich K., et al. (2018) Allosteric coupling of drug binding and intracellular signalling in the A2A adenosine receptor. Cell 172: 68-80.e12 https://doi.org/10.1016/j.cell.2017.12.004 | Google Scholar
- Felker G. M., Butler J., Collins S. P., Cotter G., Davison B. A., Ezekowitz J. A., Page R. L., Ponikowski P., et al. (2017) Effect of TRV027, a biased ligand of the angiotensin II type 1 receptor, on clinical outcomes in acute heart failure: A randomized clinical trial. JAMA Cardiology 2: 229-235 https://doi.org/10.1001/jamacardio.2016.5085 | Google Scholar
- Iijima M., Mori T., Funao T.. (2020) Regulatory challenges in evaluating G protein-coupled receptor-biased agonists, 25(2). Drug Discovery Today : 389-394 https://doi.org/10.1016/j.drudis.2019.12.002 | Google Scholar
- Kenakin T.. (2011) Functional selectivity and biased receptor signalling. The Journal of Pharmacology and Experimental Therapeutics 336: 296-302 https://doi.org/10.1124/jpet.110.173948 | Google Scholar
- Kim I.-M., Tilley D. G., Chen J., Salazar N. C., Whalen E. J., Violin J. D., Rockman H. A., et al. (2008) β-blockers alprenolol and carvedilol stimulate β-arrestin-mediated EGFR transactivation. Proceedings of the National Academy of Sciences of the United States of America 105: 14555-14560 https://doi.org/10.1073/pnas.0804745105 | Google Scholar
- Kliewer A., Gillis A., Hill R., Schmiedel F., Bailey C., Kelly E., Henderson G., Christie M. J., Schulz S., et al. (2020) Morphine-induced respiratory depression is independent of β-arrestin2 signalling. British Journal of Pharmacology 177: 2923-2931 https://doi.org/10.1111/bph.15004 | Google Scholar
- Low L. A., Mummery C. L., Berridge B. R., Austin C. P., Tagle D. A.. (2021) Organs-on-chips: Into the next decade. Nature Reviews. Drug Discovery 20: 345-361 https://doi.org/10.1038/s41573-020-0079-3 | Google Scholar
- Luttrell L. M., Lefkowitz R. J.. (2002) The role of β-arrestins in the termination and transduction of G-protein-coupled receptor signals. Journal of Cell Science 115: 455-465 https://doi.org/10.1242/jcs.115.3.455 | Google Scholar
- Lymperopoulos A., Rengo G., Koch W. J.. (2013) GRK2 inhibition in heart failure: Something old, something new. Current Pharmaceutical Design 18: 186-191 https://doi.org/10.2174/138161212798919374 | Google Scholar
- Manglik A., Kruse A. C., Kobilka T. S., Thian F. S., Mathiesen J. M., Sunahara R. K., Pardo L., Weis W., Kobilka B. K., Kobilka B. K., et al. (2015) Crystal structure of the μ-opioid receptor bound to a morphinan antagonist. Nature 485: 321-326 https://doi.org/10.1038/nature10954 | Google Scholar
- Michel M. C., Charlton S. J.. (2020) Biased agonism in drug discovery-The problem of systematic errors. Trends in Pharmacological Sciences 41: 323-324 https://doi.org/10.1016/j.tips.2020.02.008 | Google Scholar
- Noma T., Lemaire A., Naga Prasad S. V., Barki-Harrington L., Tilley D. G., Chen J., Le Corvoisier P., Violin J. D., Wei H., Lefkowitz R. J., Rockman H. A., et al. (2007) Beta-arrestin-mediated beta1-adrenergic receptor transactivation of the EGFR confers cardioprotection. The Journal of Clinical Investigation 117: 2445-2458 https://doi.org/10.1172/JCI31901 | Google Scholar
- Onaran H. O., Costa T.. (2012) Agonist efficacy and allosteric models. European Journal of Pharmacology 739: 37-51 https://doi.org/10.1016/j.ejphar.2013.04.040 | Google Scholar
- Rehsia N. S., Dhalla N. S.. (2010) Mechanisms of the beneficial effects of beta-adrenoceptor antagonists in congestive heart failure. Experimental and Clinical Cardiology 15: e86-e95 https://doi.org/10.1016/j.ejphar.2013.04.040 | Google Scholar
- Rittiner J. E., Korb E., Jin J., Coughlin S. R.. (2023) Biased signalling beyond GPCRs: Ligand bias in receptor tyrosine kinases and cytokine receptors. Nature Reviews. Drug Discovery 22: 143-161 https://doi.org/10.1038/s41573-022-00589-w | Google Scholar
- Rodríguez D., Janz R., Perez-Nueno V. I., Mestres J.. (2021) Machine learning approaches for bias detection in G protein-coupled receptor ligands. Current Opinion in Structural Biology 69: 169-177 https://doi.org/10.1016/j.sbi.2021.03.001 | Google Scholar
- Schmid C. L., Kennedy N. M., Ross N. C., Lovell K. M., Yue Z., Morgenweck J., et al. https://doi.org/10.1016/j.sbi.2021.03.001 | Google Scholar
- Schmid C. L., Kennedy N. M., Ross N. C., Lovell K. M., Yue Z., Morgenweck J., Cameron M. D., Bannister T. D., Bohn L. M., et al. (2017) Bias factor and therapeutic window correlate to predict safer opioid analgesics. Cell 171: 1165–1175.e13 https://doi.org/10.1016/j.cell.2017.10.035 | Google Scholar
- Shukla A. K., Xiao K., Lefkowitz R. J.. (2011) Emerging paradigms of β-arrestin-dependent GPCR signalling. Trends in Biochemical Sciences 36: 457-469 https://doi.org/10.1016/j.tibs.2011.06.003 | Google Scholar
- Singla N., Minkowitz H. S., Soergel D. G., Burt D. A., Subach R. A.. (2019) A randomized, double-blind, placebo- and active-controlled phase III trial of oliceridine (TRV130), a G-protein selective μ-opioid receptor agonist, for moderate to severe acute pain following abdominoplasty. Pain Practice 19: 715-731 https://doi.org/10.1111/papr.12810 | Google Scholar
- Smith J. S., Rajagopal S.. (2016) The β-arrestins: Multifunctional regulators of G protein-coupled receptors. Journal of Biological Chemistry 291: 8969-8977 https://doi.org/10.1074/jbc.R115.713313 | Google Scholar
- Thomsen A. R. B., Plouffe B., Cahill T. J., Shukla A. K., Tarrasch J. T., Dosey A. M., Kahsai A. W., Strachan R. T., Pani B., Mahoney J. P., Huang L., Breton B., Heydenreich F. M., Sunahara R. K., Skiniotis G., Bouvier M., Lefkowitz R. J., et al. (2016) GPCR-G protein-β-arrestin super-complex mediates sustained G protein signalling. Cell 166: 907-919 https://doi.org/10.1016/j.cell.2016.07.004 | Google Scholar
- . (2020) Olinvyk (oliceridine) injection prescribing information. https://doi.org/10.1016/j.cell.2016.07.004 | Google Scholar
- Viscusi E. R., Skobieranda F., Soergel D. G., Cook E., Burt D. A.. (2019) APOLLO-1: A randomized, placebo- and active-controlled phase III study evaluating oliceridine (TRV130), a G-protein-selective μ-opioid receptor agonist, for management of moderate to severe acute pain following bunionectomy. Regional Anesthesia and Pain Medicine 44: 492-500 https://doi.org/10.1136/rapm-2018-100231 | Google Scholar
- Weis W. I., Kobilka B. K.. (2018) The molecular basis of G protein-coupled receptor activation. Annual Review of Biochemistry 87: 897-919 https://doi.org/10.1146/annurev-biochem-060614-033910 | Google Scholar
- Wisler J. W., DeWire S. M., Whalen E. J., Violin J. D., Drake M. T., Ahn S., Lefkowitz R. J., et al. (2007) A unique mechanism of β-blocker action: Carvedilol stimulates β-arrestin signalling. Nature 447: 1114-1118 https://doi.org/10.1038/nature05879 | Google Scholar