Skip to main content
MedComm logoLink to MedComm
. 2025 Sep 16;6(10):e70375. doi: 10.1002/mco2.70375

G Protein‐Coupled Receptor Signaling: Implications and Therapeutic Development Advances in Cancers

Inamu Rashid Khan 1, Sana Khurshid 2, Saud Almawash 3, Rakesh Kumar 4, Ammira S Al‐Shabeeb Akil 5, Ajaz A Bhat 5,, Muzafar A Macha 2,
PMCID: PMC12441314  PMID: 40969301

ABSTRACT

G protein‐coupled receptors (GPCRs) are the largest and most diverse class of membrane proteins, mediating cellular responses to a wide range of extracellular stimuli. GPCRs initiate complex intracellular signaling networks that regulate vital physiological functions and are associated with numerous diseases, including various types of cancer. Their conserved seven‐transmembrane (7TM) structure enables these signaling networks by allowing interactions with multiple ligands and intracellular effectors. In several types of tumors, abnormal GPCR signaling promotes carcinogenesis by supporting immune evasion, cell proliferation, and therapeutic resistance. A significant research gap exists in fully understanding the molecular mechanisms behind pathway‐specific activation and biased ligand discovery of GPCRs, which could lead to the development of more effective therapies. This review examines the complexity of GPCRs, with a focus on their role in signaling through the differential activation of pathways regulated by β‐arrestin and G proteins. It discusses how targeted modulation of signaling outcomes by receptor mutants might offer therapeutic benefits in cancer treatment. The review also highlights emerging technologies, such as aptamers, PROTACs, and nanobodies, that more precisely target GPCRs. In addition to exploring receptor structure–function relationships and pathway selectivity, this review provides valuable insights into GPCR‐biased signaling and its implications in cancer biology.

Keywords: GPCRs, signaling, diseases, cancer, bias, therapeutics


G protein‐coupled receptors (GPCRs) are the dynamic protein family in the human genome. GPCR dysregulation aids in the advancement of cancer through increased migration, survival, and proliferation. Novel treatments that target GPCR signaling include orthosteric ligands, biased receptor mutants, PROTACs, and antibody–drug conjugates. Biased ligands offer improved treatment selectivity and reduced side effects by preferentially activating one pathway over another.

graphic file with name MCO2-6-e70375-g006.jpg

1. Introduction

The most extensive and diverse protein family encoded by the human genome is the G protein‐coupled receptors (GPCRs). They are found on the cell membrane and are essential for processing extracellular signals into vital biological responses [1]. GPCRs have evolved from a seven‐transmembrane (7TM) receptor with a common ancestor, resulting in a diverse superfamily of membrane receptors [2]. These receptors are encoded by around 1000 genes in the human genome, ranking them among the most prominent protein families [3, 4]. GPCRs are categorized into different classes based on similarities in the peptide chain patterns within the characteristic 7TM helical domains—the rhodopsin class (class A), secretin class (class B), adhesion class/glutamate class (class C), and frizzled/taste class (class F). The amino acid sequences of the human GPCR family serve to classify these subfamilies [5, 6]. The distinct structural features and ligand‐binding specificities in each GPCR subfamily are closely associated with their respective physiological functions [7].

GPCRs are selectively stabilized in specific conformations by biased ligands, which either stimulate or inhibit one or more alternative signaling pathways [8]. These ligands act on a broad spectrum of receptors, including adrenergic, dopaminergic, muscarinic, chemokine, and opioid receptors (ORs), each of which contributes to distinct physiological and pathological processes. A biased receptor refers to a genetically modified or mutant version of a receptor that, unlike the wild‐type (WT) receptor, can adopt a specific active conformation after ligand binding, selectively triggering a particular signaling pathway over others [9]. Among the key intracellular regulators of these signaling pathways are arrestins. Whereas β‐arrestins are widely distributed and act on a range of GPCRs, β‐arrestin 1 and 2 share around 80% sequence similarity and play overlapping and distinct functions in GPCR regulation [10, 11]. These are cytosolic adaptor proteins expressed in all cell types. Initially identified for their involvement in suppressing GPCR signaling by blocking contact with heterotrimeric G proteins, their roles have been discovered to go well beyond this inhibitory activity. β‐Arrestins block active GPCRs, facilitate receptor endocytosis, and activate downstream kinases, leading to the activation of specialized signaling pathways [12], which in turn elicit various physiological and cellular responses [12]. GPCRs play a pivotal role in cancer development by stimulating cell proliferation, survival, and migration in response to locally produced or circulating agonists. These agonists include chemokines, hormones, growth factors, and bioactive lipids, all of which contribute to critical tumor‐promoting processes such as angiogenesis, immune evasion, metastasis, and chronic inflammation [13, 14, 15]. Aberrant GPCR signaling has been observed in various cancers, resulting from receptor overexpression, constitutive activation, or excessive agonist production, and is associated with a poor prognosis and resistance to therapy. Upon agonist binding, GPCRs undergo rapid conformational changes that activate heterotrimeric G proteins, composed of Gα, Gβ, and Gγ subunits. The activated Gα subunit and Gβγ dimer modulate diverse intracellular effectors and second messengers, leading to altered gene expression and cellular behavior. In parallel, activated receptors are phosphorylated by GPCR kinases, which in turn recruit β‐arrestins. These adaptor proteins not only mediate receptor desensitization and internalization but also scaffold mitogenic signaling cascades such as mitogen‐activated protein kinase (MAPK)/ERK, PI3K/AKT, and JNK pathways, all of which are implicated in cancer progression [4, 16]. The pharmacological modulation of GPCR activity through orthosteric ligands—molecules that compete with endogenous ligands at the receptor's active site—offers a promising therapeutic strategy. Orthosteric agonists mimic the effects of natural ligands and can activate GPCR signaling, while antagonists block receptor activation by preventing ligand binding, thereby suppressing downstream oncogenic signaling. In some cases, inverse agonists can further inhibit constitutively active GPCRs by stabilizing their inactive conformations, reducing basal activity. This competitive binding mechanism enables precise control over aberrant GPCR activity, making GPCRs attractive drug targets for the treatment of multiple malignancies, including breast, prostate, ovarian, and gastrointestinal cancers [17, 18].

GPCRs are the targets of approximately 34% of US Food and Drug Administration (US FDA)‐approved medications, and the number of modulators in preclinical or clinical trials is increasing exponentially [19, 20, 21]. Ligands that preferentially activate one signaling route over another are thought to be biased and may improve therapeutic efficacy and safety [22, 23, 24]. Ligands that primarily activate G protein pathways enhance therapeutic outcomes by selectively activating G protein‐mediated signaling pathways while reducing or preventing β‐arrestin signaling. For instance, these biased ligands are engineered to specifically target G protein signaling at ORs, improving treatment efficacy [25, 26]. Various genetically biased altered D2 dopamine receptors, β‐arrestin‐biased mutants (A135R and M140D), and Gαi/o protein‐biased mutants (L125N and Y133L) have been extensively studied by predicting and modifying key residues crucial for interactions with G proteins or β‐arrestins, thereby revealing their distinct signaling pathways and functional properties [27]. In targeted protein degradation, proteolysis‐targeting chimeras (PROTACs) have provided an innovative approach to modify disease‐relevant GPCRs in cancers [28]. Recent advancements are enabling the creation of antibody–drug conjugates (ADCs) that target GPCRs, thereby expanding therapeutic options and facilitating more accurate tumor cell targeting [29, 30, 31, 32]. Collectively, the emerging modalities—nanobodies, aptamers, and gene therapy‐are expanding the therapeutic landscape for GPCR‐targeted interventions, offering new avenues for precision medicine [33, 34].

This review highlights the role of GPCRs in biased signaling networks, with a focus on their structure, signaling pathways, and therapeutic potential. It emphasizes the importance of signaling selectivity for drug development and explains how ligands can preferentially activate specific pathways, such as those mediated by β‐arrestins or G proteins. To enhance the understanding of the cellular mechanisms underlying biased signaling, the review discusses recent advances in structural biology, including the elucidation of GPCR structures and their complexes. It also examines the opportunities and challenges associated with translating these discoveries into more targeted and effective clinical therapies.

2. Classes of GPCRs, General Overview

The various classes of GPCRs emphasize the distinct roles of each class in cellular signaling. It also highlights the diseases associated with these receptors, showcasing their relevance in health and pathology (Figure 1). This comprehensive overview underscores the potential of targeting specific GPCR classes in therapeutic interventions.

FIGURE 1.

FIGURE 1

Diverse classes of GPCRs and their role in disease progression. This figure illustrates the classification of GPCRs and their critical roles in disease mechanisms. It emphasizes the relationship between ligand binding and the activation of specific signaling pathways, showcasing how each GPCR class uniquely contributes to pathological processes. The visual highlights the complexity of GPCR‐mediated responses and their significance as potential therapeutic targets, focusing on the interplay between ligand specificity and signaling diversity in various conditions. Abbreviations: GPCR—G protein‐coupled receptor, TSHR—thyroid‐stimulating hormone receptor, FSHR—follicle‐stimulating hormone receptor, CB1—cannabinoid receptor type 1, CB2—cannabinoid receptor type 2, MT1—melatonin receptor 1, MT2—melatonin receptor 2, ADGRB3—adhesion G protein‐coupled receptor B3, ADGRL3—adhesion G protein‐coupled receptor L3, GABA—gamma‐aminobutyric acid, mGluRs—metabotropic glutamate receptors, CaSR—calcium‐sensing receptor, SMO—smoothened receptor, FZ—frizzled receptor.

2.1. Class A GPCRs

Class A GPCRs are categorized into subgroups based on their specific ligand interactions. These subgroups include receptors for peptides, proteins, lipids, melatonin, nucleotides, steroids, sensory stimuli, and olfactory signals [19]. Not all class A GPCRs are considered orphan receptors. While class A GPCRs encompass a diverse range of receptors, only a subset remains “orphaned,” meaning their endogenous ligands have not yet been identified [35]. The intracellular domain's downstream effector proteins are activated when orthosteric ligands connect with the binding pocket created by the 7TM helices [36]. Many class A GPCRs have distinct ligand‐binding characteristics and activation mechanisms. A notable group is the glycoprotein hormone receptor subfamily, which includes essential receptors like FSHR, LHR, and TSHR, playing crucial roles in hormonal regulation and signaling [37, 38, 39]. Class A GPCRs comprise the odorant receptor (OR) and trace amine‐associated receptor families, two of the three categories of olfactory receptors that detect odorants. Olfactory sensory neurons were found to express specific molecules in extranasal tissues. These molecules play roles in various biological processes, indicating their potential as therapeutic and diagnostic targets [40]. Orphan GPCRs are a subset of class A GPCRs, where the “orphan” designation refers to the lack of a known ligand. Orphan GPCRs play a crucial role in regulating physiological processes and are linked to various human disorders. These include Alzheimer's disease, schizophrenia, type 2 diabetes, ADHD, cognitive decline, and different brain abnormalities, highlighting their clinical significance [41]. There are now about twelve structures known for orphan receptors. For instance, the ECL2 region functions as an inherent agonist for receptor activation [42, 43]. The development and metastasis of ovarian cancer are influenced by a large number of GPCRs and their particular ligands. Therefore, it would be highly beneficial to develop new chemotherapeutics to fully characterize the roles of these GPCRs during carcinogenesis [44]. Research on orphan GPCRs is still in its infancy, but promising opportunities emerge as more receptor structures are elucidated and detailed studies of constitutive activity and active conformations advance [45].

2.2. Class B GPCRs

Class B GPCRs are categorized into B1 secretin receptors and B2 adhesion receptors. Class B1 GPCRs undergo a sequence of conserved conformational changes within their 7‐transmembrane domain. The extracellular parts of TM6, TM7, and ECL3 move outward, while the extracellular part of TM1 moves inward, generating a V‐shaped pocket that enhances ligand binding [46]. The structural architecture of B1 receptors is comparable to their transmembrane domain, which has seven helices, and extracellular domain, which has 120–160 residues. These domains collaborate to facilitate ligand recognition [47]. The B1 receptor family activates various G protein subtypes to trigger downstream signaling, with Gs being the predominant subtype [48, 49]. These receptors are crucial for regulating several physiological functions, including blood sugar regulation, calcium homeostasis, and the motility and secretion of the gastrointestinal system [50, 51, 52]. The B2 adhesion GPCR (aGPCR) family includes 33 unique members, playing key roles in cellular signaling and communication. These receptors are involved in essential physiological functions, including endocrine regulation, reproduction, and cerebrovascular function. Mutations or abnormal expression in these receptors are associated with several diseases, including cancers and neurodevelopmental disorders [53].

2.3. Class C GPCRs

Class C GPCRs are classified into four groups according to the particular agonists they interact with: orphan receptors, amino acid receptors (such as γ‐aminobutyric acid receptors GABAB1/GABAB2, metabotropic glutamate receptors [mGluRs]), and taste receptors [54]. The calcium‐sensing receptor (CaSR), which predominantly couples with Gq, is a unique member of its subfamily [55]. The parathyroid glands and renal tubules are the primary sites of expression for the CaSR. It is essential for controlling the release of parathyroid hormone in response to blood calcium levels. Due to these actions, CaSR is considered a possible candidate gene for calcium nephrolithiasis [56]. Two members of the GABAB subfamily, GB1 and GB2, function as a heterodimer. GB1 binds the agonist, while GB2 plays a crucial role in coupling with the G protein heterotrimer [57, 58]. Both preclinical and clinical evidence suggest that the GABAB receptor holds significant potential as a therapeutic target for treating anxiety and depression. Identifying new GABAB receptor allosteric modulators and expanding knowledge of how specific intracellular GABAB receptor‐associated proteins influence GABAB receptor activity could lead to the development of GABAB receptor therapies for managing such disorders [59]. Taste GPCRs are in specialized taste receptor cells within taste buds. Type II taste GPCRs, such as monomeric bitter receptors and kokumi/CaSRs, are distinguished by their unique structure. On the other hand, type I taste GPCRs form heterodimeric complexes that function as receptors for sweet (TAS1R2/TAS1R3) and umami (TAS1R1/TAS1R3) tastes [60]. Activation of mGluR3 has been shown to have neuroprotective effects in astrocytes, while mGluR5 appears to exacerbate neurotoxicity associated with Aβ42 [61].

2.4. Class F GPCRs

Class F GPCRs differ from others in that they have a conserved 7TM domain linked to the N‐terminal cysteine‐rich domain (CRD) [62]. The CRD plays a crucial role in receptor activation and initiating downstream signaling, making it a key target for drug development [63]. Class F GPCRs encompass the smoothened (SMO) and frizzled (FZ) family receptors. Based on their different sensitivities for Wnt ligands and sequence similarities, the 10 members of the FZ receptor family are categorized into five subfamilies: FZ1/2/7, FZ3/6, FZ4, FZ5/8, and FZ9/10. The potential of these receptors as therapeutic targets is underscored by the fact that their alterations have been associated with several malignancies [64, 65, 66, 67]. Cancer, fibrosis, and neurodegeneration are among the many illnesses linked to FZs and their related Hedgehog (Hh) and Wnt signals [19]. SMO tends to have a straight TM6, but FZs show a kinked TM6 when activated. These different activation pathways may offer insights into targeting class F receptors to develop medications with enhanced pharmacological properties and selectivity [68]. SMO is integral to the Hh signaling pathway, playing a vital role in tissue repair and maintaining homeostasis [69]. In the activated state of SMO, compared with its inactive form, the intracellular end of TM6 shifts outward while TM5 moves inward [70]. The triggered SMO structure exhibits TM6 moving outward within the cell, reminiscent of structural changes in class A and B GPCRs [71].

In summary, GPCR classification into classes A, B, C, and F highlights their structural diversity and functional roles in key physiological processes, including neurotransmission, metabolism, and immune regulation. Dysregulation of these receptors contributes to cancer development, reinforcing their significance as therapeutic targets. A deeper understanding of class‐specific features can aid in designing targeted, biased ligands for more effective cancer therapies.

3. Signaling Role of Heterotrimeric G Proteins in Cellular Physiology

The G protein subtypes Gαq, Gαs, and Gαi illustrate their unique roles in cellular communication and showcases the variable signaling effects of each subtype. Gα, Gβ, and Gγ are the three subunits that make up the heterotrimeric G proteins, the primary signaling transducers in GPCR pathways [72]. Gαs subunits activate protein kinase A (PKA) and other targets by stimulating AC to convert ATP to cyclic adenosine monophosphate (cAMP). PKA phosphorylates a variety of effectors in the heart, including voltage‐gated L‐type Ca2+ channels, troponin I, myosin‐binding protein‐C, phospholamban, and the cardiac RYR2 (ryanodine receptor) [73]. Together, these subcellular elements are coordinated by βAR stimulation to enhance myofilament cross‐bridge cycling, thereby increasing both the relaxation rate (lusitropy) and the force of contraction (inotropy). Ca2+‐induced Ca2+ release occurs when action potential‐triggered L‐type Ca2+ channels initiate an inward Ca2+ entry, causing the RYR2 to release a considerable quantity of intracellular Ca2+. The RYR2 creates a Ca2+ spark, the primary intracellular Ca2+ source needed to develop and contract myofilament cross‐bridges. βAR activation affects the sinoatrial node ion channels, transporters, and hyperpolarization‐activated cyclic nucleotide‐gated channels to modulate heart rate (chronotropy) [74]. Gαq/11 catalyzes the conversion of membrane‐bound phosphatidylinositol 4,5‐bisphosphate to diacylglycerol and inositol 1,4,5‐trisphosphate (IP3) by activating phospholipase‐Cβ. The sarcoplasmic reticulum and nuclear envelope include intracellular Ca2+ release channels called cardiomyocyte IP3 receptors, which are activated by IP3. Calcineurin, a Ca2+ and calmodulin‐dependent serine/threonine protein phosphatase, and a nuclear pool of Ca2+‐calmodulin‐dependent protein kinase II, promote cardiac hypertrophic signaling. GPCR activation triggers IP3 production, which leads to Ca2+ release from the nuclear envelope through IP3 receptors, further contributing to the signaling process [75, 76]. The sarcoplasmic reticulum Ca2+ATPase pump that controls protein phospholamban is dephosphorylated when diacylglycerol and intracellular Ca2+ activate protein kinase C‐alpha (PKC‐α), which controls cardiac contractility by upregulating type 1 protein phosphatase [77]. Normal cardiac function depends on heterotrimeric G protein signaling; however, chronic, prolonged activation, especially of Gαs and Gαq/11, can lead to unfavorable ventricular remodeling and reduced cardiac performance. The detrimental effects of excessive G protein signaling on the heart include tachycardia and an increased risk of heart failure [78]. Gαi activation may help block Gαs signaling and have an inhibitory effect on AC activity. Therefore, following GPCR stimulation, negative feedback loops are essential for preserving cellular homeostasis. Regulators of G protein signaling (RGS) proteins act as GTPase‐activating proteins (GAPs), inhibiting GTP hydrolysis to Gα–GDP. By preventing GTP hydrolysis, these regulators stabilize the active Gα–GTP form [79]. This leads to reforming the heterotrimeric G protein complex, effectively terminating Gα‐mediated signaling (Figure 2).

FIGURE 2.

FIGURE 2

Distinct signaling branches of Gα proteins in GPCR pathways. The figure depicts the distinct signaling branches of Gαs, Gαq, and Gαi. This visual representation underscores the signaling associated with physiological impacts. Each branch highlights the subtle interplay between receptor activation and downstream effects. Understanding these signaling dynamics is crucial for harnessing GPCRs in therapeutic applications. Abbreviations: Gαs—stimulatory G protein alpha subunit, Gαi—inhibitory G protein alpha subunit, Gαq—G protein alpha q subunit, cAMP—cyclic adenosine monophosphate, PKA—protein kinase A, CREB—cAMP response element‐binding protein, PLC—phospholipase C, PIP2—phosphatidylinositol 4,5‐bisphosphate, DAG—diacylglycerol, IP3—inositol trisphosphate, NFAT—nuclear factor of activated T‐cells.

4. GPCR Signaling Bias

Biased signaling refers to the process in which GPCRs can adopt numerous active conformations, and different ligands can stabilize unique receptor states, leading to the selective activation of specific downstream signaling pathways [80]. In contrast to the conventional perspective, which holds that all pathways are activated with equal efficacy, biased signaling permits the preferential activation of one pathway (such as the G protein‐mediated pathway) over another (such as the arrestin‐mediated pathway) [81, 82]. The arrestins play crucial roles at the receptor level, including desensitization, trafficking, and signaling [83]. When specific ligands preferentially induce β‐arrestin recruitment and signaling over G protein activation, the idea of β‐arrestin‐biased signaling emerges. These β‐arrestin‐biased agonists can simultaneously promote β‐arrestin‐dependent pathways and inhibit G protein signaling, resulting in distinct cellular and physiological consequences [84]. While β‐arrestin‐biased ligands primarily regulate the conformational states of helix 7, it has been demonstrated that unbiased ligands bind to the receptor, primarily shifting the equilibrium toward the G protein‐specific active state of helix 6. This indicated that, contrary to what was previously thought, phosphorylation of the receptor itself does not always result in a switching of the receptor coupling to different G proteins [85, 86]. β‐Arrestins 1 and 2 serve distinct cellular roles. Their differential nucleocytoplasmic shuttling arises from the location of the nuclear export sequence, which is present on the C‐terminus of β‐arrestin 2 and the N‐terminus of β‐arrestin 1 and 2, underscoring their unique regulatory mechanisms [87]. Numerous conformational states of β‐arrestin: receptor complexes are made possible by extra interactions with the intracellular loops and core of active receptors. By preventing further G protein activation, β‐arrestins bind to receptors, blocking interactions with the Gα subunit and leading to receptor desensitization [88, 89]. As signaling transducers and multipurpose scaffold proteins, β‐arrestins are associated with various outcomes and are essential for controlling other cellular effects and intracellular signal transmission and amplification. Accordingly, they have diverse roles in the control and development of cancerous tumors via many signaling pathways, for example, MAPK and PI3K/AKT [90, 91]. It has been demonstrated that β‐arrestin 1 serves as a framework for cytoskeleton changes that affect the motility of tumor cells [92, 93, 94]. Various researchers have demonstrated that nuclear β‐arrestin 1 can ensemble transcriptional responses to environmental disturbances, indicating new functions for β‐arrestin 1 in cancer progression [95, 96, 97]. A recent clinical investigation suggested that β‐arrestin 2 is a potential target for novel therapeutic methods and a significant predictive factor in advanced ovarian cancer [98]. Similarly, a recent study using breast cancer cells showed that downregulating the expression of β‐arrestin 1 and β‐arrestin 2 tended to enhance cell invasion and proliferation, while upregulating their expression levels inhibited these processes [99]. By suppressing androgen receptor (AR) signaling, β‐arrestin 2 in prostate cancer prevents cell viability and proliferation [100, 101]. However, other research supports the notion that β‐arrestin 2 activity contributes to the growth of human malignancies; β‐arrestin 2 is highly expressed in several human cancers, such as renal cell carcinoma and breast cancer, and is associated with a poor patient survival rate [102, 103, 104]. To summarize, β‐arrestins initiate intracellular signaling cascades without the need for G protein support by combining intrinsic cellular pathways with GPCR signals. This combination enables the development of novel treatments targeting β‐arrestin‐mediated pathways, a phenomenon called arrestin‐biased agonism [22]. To comprehend the mechanics behind the role of β‐arrestins in cancer, further research should be conducted on the anticancer and tumor suppressor efficacy of β‐arrestin isoforms, which will reveal their functional specialization.

5. Mutational Activation of GPCR Signaling in Cancer

GPCRs and cellular transformation were first found to be directly related after the identification of the receptor for angiotensin (1–7) (MAS oncogene) in 1986 [14]. The GPCR protein family has since been linked to tumor growth and metastasis by molecular genetics, identifying significant GPCRs, their mutations, or altered expressions. Research has demonstrated that when a locally produced or circulating agonist activates GPCRs, it contributes to the growth of cancer cells. These agonists play a key role in inflammation‐related cancer, metastasis, and angiogenesis. Prior research has also examined the distinct GPCRs involved in cancer's functional activities, expression, and signaling [13, 14, 15]. A standard set of GPCRs was shown to be expressed by a particular type of cancer cell or tumor. Table 1 presents specific instances of a broad class of GPCRs that are highly expressed in various metastatic and primary tumor cells and are linked to the proliferation of tumor cells when triggered by circulating or locally generated ligands. Both cancer cells and the different cell types that comprise the tumor microenvironment (TME) exhibit high levels of GPCR expression. These receptors are crucial for mediating intercellular communication, a vital mechanism that influences the progression of tumors and cancer. Chemokine receptors stand out among the many GPCR families due to their role in cancer biology. These receptors can stimulate the growth of cancer cells, improve survival by preventing programmed cell death, and promote other characteristics of malignancy, including angiogenesis, invasion, and metastasis, by interacting with their ligands (Figure 3). They also serve as targets for unraveling tumor‐supportive connections in the microenvironment and as indicators of tumor behavior.

TABLE 1.

Selected G protein‐coupled receptors, ligands, and signaling pathways involved in cancer.

Receptors Ligands Pathways Cancer type References
Protease‐activated receptors (PAR1 & 2) LPA Thrombin, trypsin, or TFLLRN (PAR1) or SLIGKV (PAR2) Lysophosphatidic acid (Gαq) Hippo/YAP pathways through Gαq or Gα12/13‐coupled receptor activation. Hippo pathway inhibition (by blocking Lats1/2 kinases)

Breast cancer

Colon cancer

[111]

[112]

Chemokine receptor (CXCR4) CXCL12, SDF‐1 PI3K, ERK1/2, PLC, JNK Akt, Src, PIP2, IP3, Ras, Raf HNSCC [105]
Frizzled (Fz) PAR1 Parathyroid receptor1 (PTHR1)

Wnt 3A (canonical pathway)

Thrombin or TFLLRN

PTH

Stabilization of β‐catenin and its transcription activity by canonical Wnt signaling

Colon cancer

Lung cancer

Prostate cancer

[113, 114]

[115, 116]

[117]

Prostaglandin receptors (PE2, PE4) PGE2 Cyclooxygenase pathway, PI3K (coupling to Gαi)

HNSCC

Breast cancer

Colon cancer

[118]

[105]

[119]

Lysophosphatidic acid receptors LPA1–6) LPA

Rho‐dependent pathways

β‐cantenin stabilization

Kruppel‐like factor 5

Colon cancer

Ovarian cancer

Prostate cancer

[120]

[121]

[105]

Sphingosine 1‐1‐phosphate receptor (S1PR) S1P Ras‐ERK, PI3K/‐Akt/‐Rac, Rho, STAT3 (coupling to Gαi) Glioma [122, 123]
Angiotensin II type 1 receptor Angiotensin II TNF‐α, ERK1/2, NF‐κB, STAT

Gastric cancer

Prostate cancer

[124]

[105]

Bradykinin receptor Type 1 and 2 (B1R, B2R) Kinins Gαq and cross‐talk with EGFR Ras, Raf, ERK HNSCC [118]

Abbreviations: LPA: lysophosphatidic acid, PGE2: prostaglandin E2, CXCL12: C‐X‐C motif chemokine ligand 12 (also SDF‐1), ERK: extracellular signal‐regulated kinase, PI3K: phosphoinositide 3‐kinase, PLC: phospholipase C, PIP2: phosphatidylinositol 4,5‐bisphosphate, IP3: inositol 1,4,5‐trisphosphate, TNF: tumor necrosis factor, EGFR: epidermal growth factor receptor, NF‐Κb: nuclear factor kappa‐light‐chain‐enhancer of activated B cells, STAT: signal transducer and activator of transcription, JNK: c‐Jun N‐terminal Kinase, HNSCC: head and neck squamous cell carcinoma.

FIGURE 3.

FIGURE 3

Roles of G proteins and GPCRs in cancer progression. Key G proteins and G protein‐coupled receptors (GPCRs) are highlighted as drivers of hallmark traits in cancer. Dysregulated signaling through these receptors supports tumor growth and progression by altering cellular processes and oncocrine networks within the tumor microenvironment. Abbreviations: GNAQ—G protein subunit alpha q, GNAS—G protein subunit alpha s, GNA12—G protein subunit alpha 12, GNA13—G protein subunit alpha 13, GNAI2—G protein subunit alpha i2, CXCR4—C‐X‐C chemokine receptor type 4, FZ7—frizzled class receptor 7, PAR1—protease‐activated receptor 1, VIPR2—vasoactive intestinal peptide receptor 2, P2Y2—purinergic receptor P2Y2, GPR68—G protein‐coupled receptor 68, LPAR1—lysophosphatidic acid receptor 1.

The intricate signaling networks that GPCRs mediate emphasize their importance as prospective therapeutic targets. It remains challenging to define GPCRs as cancer‐specific drivers due to the intricacy and variety of their expression and mutation patterns across numerous cancer types, despite the growing body of research on their role in cancer. On the other hand, hotspot mutations in G proteins have been shown to function as carcinogens in specific cancers. This review will examine the significance of G proteins and their role in cancer, highlighting their crucial role in disease development and potential as therapeutic targets.

5.1. Signaling Through Gαq G Proteins

The GNAQ oncogenes—a large number of the mitogenic signals acting on GPCRs—are triggered by growth factors and are transduced by heterotrimeric subunits of the Gαq family, which are encoded by GNA15, GNA14, GNAQ, and GNA11 and their associated receptors [105, 106]. Members of the Gαq family activate phospholipase Cβ (PLCB1‐4 4) and cleave PIP2 into diacylglycerol and IP3. By utilizing regulated kinases and transcriptional networks, the production of these second messengers triggers cytosolic calcium mobilization and initiates a range of downstream signaling cascades [107, 108, 109]. The original concept of mutating the Gαq subunit into a constitutively active, GTPase‐deficient type (Q209L and G207T) was the basis for earlier research investigating Ras oncogene mutations [110]. In addition to causing a malignant transformation in NIH3T3 cells, these Gαq mutants exhibited the ability to develop tumors in nude mice [110]. Recent attempts at cancer sequencing have indicated that activating mutations in Gα11 or Gαq are generally detected in more than 90% of uveal melanoma (UM), where they function as driving oncogenes, primarily at Q209. Since this initial finding, several clinical diseases, including Sturge‐Weber syndrome, congenital hemangiomas, and leptomeningeal melanocytic lesions, have been associated with abnormal Gαq activity [125, 126, 127, 128]. About 10% of cutaneous skin melanomas also include mutations in the Gαq family. Approximately 2500 persons in the US are affected by UM annually, making it the most common malignancy of the adult eye [128]. In UM, the oncogenic driver is the hyperactivation of the Gαq‐pathway, as almost 5% of UM patients without mutations in Gα11 or Gαq possess activating mutations in additional Gαq pathway levels that are cooperatively exclusive, for example, in Gαq‐coupled GPCR (CYSLTR2) or the downstream effector of Gαq PLCB4 [129, 130]. UM, resulting from CYSLTR2 mutation and hyperactivation, remains one of the most notable instances of a GPCR‐driven tumor type to date [131]. Investigations examining the mechanisms by which abnormal Gαq signaling contributes to carcinogenesis have shown that, in contrast to the transient activation of mitogenic kinases and second messengers following canonical Gαq stimulation, mutant Gαq needs an interaction between the protein‐Rho‐specific guanine nucleotide exchange factor (GEF) TRIO to maintain long‐term signaling [132]. TRIO phenocopy knockdown reduces Gαq expression in vivo in UM models based on their tumorigenic potential, stimulation of AP‐1‐mediated transcription, and activation of JNK and p38, as well as cell proliferation assays [132]. Notably, this signaling pathway activated a transcriptional coactivator, Yes‐associated protein (YAP), which is regulated by the Hippo pathway, without the assistance of PLCβ, the most well‐known Gαq target. It has been discovered that the Hippo pathway, which suppresses tumor growth and is widely involved in controlling organ growth and cell proliferation, is frequently altered in cancer [133]. It has been shown that YAP activity is essential for tumor development and proliferation in UM [134, 135]. According to recent research, focal adhesion kinase (FAK) regulates the activation of YAP, making it a crucial modulator of the oncogenic signaling pathway controlled by Gαq and a potential therapeutic target in UM105 patients. Like R183, Q209 is located in the GTP‐binding domain of Gαq, where mutations alter the inherent GTPase activity and enhance signaling activity. In contrast, compared with Q209 mutants, R183 mutants have a much lower expression potential. A class of other melanocytic neoplasms, including primary melanocytic tumors, Ota nevi, and blue nevi in the central nervous system, commonly contain activating mutations of Gαq. Moreover, they have also been connected to congenital hemangiomas [127, 136]. Numerous roles of Gαq in the immune system have been identified, including the potential to prevent tumor growth. In vitro survival and in vivo proliferation of B and T cells are improved by loss of GNAQ expression [137, 138, 139, 140]. Remarkably, recurrent loss‐of‐function (LOF) mutations at Y101or T96S or loss of Gαq expression affect the expression of the Gαq pathway in nearly 25% of natural killer (NK)/T/T cell lymphoma, an extremely malignant and highly aggressive subtype of non‐Hodgkin's lymphoma [140]. NK cells in mice specifically knocked out for Gαq showed an inherent survival advantage over WT NK cells, which is permanent, with the effects of Gαq deletion also observed in additional immune cells. Additionally, in Gαq‐low NK cell lines, WT Gαq expression induced apoptosis, which was inhibited by concurrent T96S Gαq expression, indicating that T96S Gαq functions as a dominantly negative mutant to enhance NK cell carcinogenicity [140]. The intricate molecular processes underlying Gαq‐driven carcinogenic signaling are highlighted by the propensity of Q209 and R183 Gαq hotspot mutations to occur in solid tumors, compared with the prevalence of Y101 and T96 in hematological cancers. This suggests a complex relationship between the tumor‐suppressive or carcinogenic functions of these mutations and the cellular environment in which they occur.

5.2. Signaling Through Gαs in Cancer

One of the most frequently mutated G proteins in human cancer is the GNAS oncogene, which codes for the Gαs protein [141]. Based on the description of mutations in several tumor types, GNAS occurs most frequently in appendix malignancies (70%) and pituitary tumors (27%), and it is mutated in around 5% of sequenced tumors [141]. Through TCGA, the most common mutations in GNAS are found in endometrial carcinomas (7.3%), colorectal malignancies (4.7%), adrenocortical carcinomas (5.5%), stomach adenocarcinomas (5.7%), esophageal carcinomas (4.9%), pancreatic adenocarcinomas (5.6%), and ulcerative carcinomas (5.7%). Most mutations in Gαs occur at a hotspot position (R201), resulting in the Gαs protein becoming constitutively active with impaired GTPase activity [142]. Q227 is a secondary hotspot; however, it is less common than R201 mutants. Studies investigating the Cholera toxin's mode of action were the first to propose the functional relevance of the R201 residue. It was discovered that the cholera toxin ADP‐ribosylates Gαs's R201, permanently blocking its GTPase activity, thereby activating its activation [143, 144]. Since then, Gαs crystallographic investigations have verified that the R201 residue is located in the GTP‐binding pocket and promotes GTP hydrolysis [145]. Adenylyl cyclases are stimulated when Gαs are activated, either by activating mutations or by stimulating Gαs‐coupled GPCRs. This results in the synthesis of cAMP, the activation of PKA, and additional signaling functions [146, 147]. Initially, several growth hormone‐secreting pituitary cancers with R201C/H mutations demonstrated the carcinogenic effects of Gαs [146, 148, 149]. Hormone‐insensitive growth resulted from sustained signaling triggered by mutant Gαs in these cancers, which bypassed the canonical necessity for growth hormone activation [142]. Indeed, around 7% of adrenocortical carcinomas in TCGA have a mutation in the Gαs subfamily (GNAS and GNAL). Surprisingly, a significant percentage of cancer types in TCGA exhibit extensive activation of the Gαs pathway, with gastrointestinal tract cancers showing a notable enrichment [150]. This is supported by the fact that 5.96% of stomach adenocarcinomas, 5.24% of colorectal cancers, and 5.03% of pancreatic cancers had mutations in the Gαs subfamily. Surprisingly, over 50% of patients with colorectal cancer exhibit abnormal Gs pathway activation, which includes downstream effectors of Gαs such as PKA and adenylyl cyclases, activating mutations or gene amplification of GNAS. Additionally, inflammatory prostaglandins (PGs) that accumulate locally due to COX2 overexpression lead to Gs‐coupled prostaglandin E2 (PGE2) receptors (PTGER2, 4) activation [151, 152]. Tumorigenesis and the development of colorectal cancer are thought to be facilitated by these genetic changes and autocrine activation, which are crucial in deregulating and activating the intestinal epithelium's PKA, MAPK, and Wnt pathways [153, 154, 155]. It is interesting to note that tumors with Gαs mutations frequently have a very mucinous appearance. This is consistent with research showing that mucin genes are upregulated in response to GNAS expression [156]. Among gastrointestinal mucinous neoplasms, GNAS mutations are prevalent in intraductal papillary mucinous neoplasms, a type of invasive pancreatic cystic neoplasm that has the potential to progress into appendiceal mucinous tumors or pancreatic adenocarcinoma, where Gαs‐promoted signaling is essential for tumor growth [156, 157, 158, 159]. Gαs and their downstream signaling pathways are crucial regulators within stem cell populations, where they prevent self‐renewal and play a role in the development of mucinous neoplasms. Notably, Gαs can have different functional effects depending on the cellular environment. Gαs can also function as a tumor suppressor in some stem‐like cell states; however, gain‐of‐function mutations in Gαs usually have pro‐oncogenic effects. The complex role of Gαs in cancer biology is further highlighted by the reality that LOF mutations or deletions of Gαs may expedite tumor initiation and progression by de‐repressing critical oncogenic pathways, including the Hippo signaling pathway and Sonic Hh [160, 161].

5.3. Gαi/o Protein‐Encoding Gene Mutations in Cancer

Activation of PI3K and MAPK signaling is among the many effectors through which the G proteins’ Gαi/o subfamily initiates signaling. Gαi inhibits adenylyl cyclase, which lowers cAMP in contrast to Gαs signaling [162]. Activation of the islet‐activating protein (IAP), also known as pertussis toxin, can indeed inhibit adenylyl cyclase activity, resulting in a reduction in cAMP levels. Treatment with IAP reversed this inhibition, leading to an increase in cAMP production. Adenylyl cyclase activity was increased, and Gαi was rendered inactive by IAP ADP‐ribosylation, as reported by various groups that studied the IAP substrate [163, 164, 165]. Constitutively active Gαi mutants have been proven to have the capacity to change cells; therefore, they are recognized as proto‐oncogenes, just like other G proteins [166, 167]. It has also been discovered that when expressed in cells, activated mutations originating from cancer of GNAO1, which codes for Gαo, induce anchorage‐dependent growth and oncogenic transformation [168]. Hotspot‐activating mutations in GNAI2, also known as the gip2 oncogene, have been identified in a variety of cancer types, encompassing adrenal and ovarian tumors, and have been demonstrated to cause dysfunction in a subgroup of growth hormone‐releasing pituitary adenomas. Nevertheless, because there are so few patients with this specific disorder, little research has been conducted on the impact of germline GNAI variants, which warrants additional investigation [169, 170]. Accordingly, it has been discovered that breast‐invasive cancer has an elevated level of GNAI2 (encoding Gαi2) [141]. The most frequently harmfully mutated class of GPCRs across malignancies is Gαi/o‐coupled receptors [150]. Furthermore, it has been found that Gαs‐activating mutations and Gαi/o‐coupled receptor inactivating mutations are mutually exclusive. This implies that they produce identical functional results, convergent on elevated cAMP activity [150]. The functional significance of these alterations has not yet been thoroughly investigated, despite a significant percentage of GPCR mutations in GI‐tract cancers being Gαi‐linked. The Gαi/o subfamily of G proteins is altered at indistinguishable frequencies in GI cancers as those of the Gαs subfamily [141]. In GI malignancies, corresponding to the frequent occurrence of Gαs‐pathway activation, elevated cAMP/PKA activity may be a common mechanism that causes tumors in these tissues and warrants further research from both signaling and therapeutic perspectives.

5.4. Gα12 Protein‐Encoding Mutations in GNA13 and GNA12 in Cancer

GNA12 and GNA13 encode the two α subunits that comprise the Gα12 subfamily of G proteins. Expression in nearly every tissue type, Gα12 and Gα13 are necessary for cytoskeletal remodeling, including cell proliferation, adhesion, polarity, migration, and as well as invasion [171, 172]. After research showed that Gα13 had vigorous transforming activity, it identified GNA13 and GNA12 as active mutants, collectively known as the gap oncogene [173, 174]. Since then, Gα12/13 have been extensively investigated as a factor driving cellular transformation, cancer development, and metastasis in various cell types [175, 176, 177]. Leukemia‐associated RhoGEF, PDZRhoGEF, and p115RhoGEF are among the members of the RhoGEF family that mediate signaling to Rho in response to Gα12/13 [178, 179, 180, 181]. Early research on how an active mutant of Gα12 fully transformed fibroblasts showed that Gα12 extensively promoted transcriptional activity through the serum response element and the c‐fos promoter element, dependent on Rho [182]. According to additional research, Gα12/13 can signal through several effectors, including MAPK, Radixin, and β‐catenin [171, 183]. Collectively, these signaling pathways regulate numerous transcriptional networks and cancer‐related cellular functions, including the activation of AP‐1, STAT3, and YAP [183, 184, 185]. Overexpression or mutation of Gα13/13 or Gα12/13‐linked GPCRs, such as thromboxane A2 receptor TA2R (TBXA2R) or protease‐activated receptor 1 (PAR1) (F2R), can induce aberrant Gα12/13‐driven signaling, which is transformative and dramatically increases the possibility for invasion of several cancer types [175, 186, 187, 188]. The elevated expression of Gα12/13 and Gα12/13‐coupled GPCRs, along with their effectors, such as Rho, is significant in various cancer types, including hepatocellular, prostate, and breast cancers [189, 190]. It has been discovered that the expression levels of GNA13, specifically elevated in several solid tumors, influence therapeutic resistance in squamous cell malignancies, including head and neck squamous cell carcinoma [191, 192, 193]. It is well known that many chemokine receptors, such as CXCR4, and receptors for bioactive lipids, such as LPAR and S1PR (S1PR2–5), can link to Gα12/13 and contribute to cancer's metastatic potential [193, 194]. Remarkably, it has been discovered that the Gα13/RhoA signaling axis has a tumor‐suppressive function in various hematological and lymphoid cancers, including diffuse large B cell lymphoma and Burkitt's lymphoma [195, 196]. Although it has not been adequately analyzed and identified as such by OncoKB188, GNA13 seems to have a possible hotspot mutation at R200 throughout TCGA PanCancer investigations. Only bladder urothelial carcinomas include this cluster of mutations, though their function remains unidentified [150]. Specifically, GNA13 is a significantly mutated gene in B‐cell lymphomas originating from germinal centers, which is prompted by a subpopulation of B lymphocytes with abnormal growth and dissemination that are firmly confined to germinal centers in lymphoid tissues and are highly regulated [197, 198]. Although the accurate processes by which the Gα13 signaling pathway's inactivation permits the formation of lymphomas are still unknown, numerous studies have shown that the reduction of the Gα13/RhoA axis causes B cells to produce higher amounts of phosphorylated AKT [197].

5.5. Mutations that Impact RGS and Gβγ in Cancer

Functional elements of G protein signaling, such as the βγ subunits of heterotrimeric G proteins and members of the RGS family, can also enhance or alter G protein‐driven signaling, leading to oncogenesis [199]. Due to their inherent GAP activity, members of the RGS family physiologically inhibit G proteins. They facilitate the rearranging and inactivation of the heterotrimeric G protein complex 5 and increase GTP hydrolysis by the Gα‐subunit. Numerous RGS protein mutations and transcriptomic dysregulation, particularly those resulting in LOF, have been identified in recent pan‐cancer analyses. This has promoted G protein activity through a mechanism of G protein signaling potentiation and a previously unknown tumor suppressive role [200, 201]. Numerous cancers, particularly hematological malignancies, have been found to harbor recurrent mutations in Gβ proteins, including GNB1 and GNB2, which lead to abnormal stimulation of the MAPK and PI3K/AKT pathways [202]. The impact of specific Gβ mutations may be determined by lineage‐dependent cellular predispositions, as demonstrated by the evident, unique mutations that group cancer types according to cell lineage. It has also been discovered that Gβγ subunits promote cell migration and metastasis. PREX1, a GEF that stimulates the small GTPase RAC, can be directly bound by Gβγ and cooperatively activated with PIP3 [203, 204]. Interestingly, Gβγ expression is required for PI3Kβ's transforming ability; p110 mutants with binding defects have been shown to significantly decrease cells' ability to transform and exhibit GPCR‐driven chemotaxis [205, 206]. This mutation dramatically reduced extravasation, matrix degradation, and macrophage‐stimulated invasion of tumor cells in breast cancer cells, which is consistent with these findings and suggests that Gβγ may play a potential function in paracrine communication among immune cells and tumor cells [207, 208]. In this regard, a growing body of research indicates that Gβγ signaling plays a crucial role in cancer metastasis and carcinogenesis, partly by promoting and modifying a prometastatic TME [209, 210].

In summary, mutations in GPCRs and their associated G proteins contribute significantly to cancer by driving constitutive signaling and promoting tumor growth, survival, and metastasis. Hotspot mutations in genes such as GNAS, GNAQ, and GNA13 underscore the oncogenic potential of aberrant GPCR signaling. Understanding these alterations offers promising avenues for targeted cancer therapies.

6. Cancer‐Associated GPCR‐Mediated Signaling Pathways

In both primary and metastatic tumor cells, various GPCR‐mediated signaling pathways are involved, linking their activation by circulating or locally generated ligands to the development of cancer. A brief description of some of these signaling pathways is given below.

6.1. Wnt Signaling

The signaling pathway and tissue homeostasis are mediated by the Fz receptor, also referred to as the Wnt receptor. Wnt proteins play a crucial role in both physiological development and malignant processes, such as cancer. By antagonizing the β‐catenin “destruction complex,” which is made up of adenomatosis polyposis coli (APC), E3 ubiquitin ligase part TrCP1, glycogen synthase kinase 3, Axin, and casein kinase 1, through the Fz‐lipoprotein‐related protein 5/6 (LRP5/6) receptor complex, the canonical Wnt signaling pathway stabilizes β‐catenin. In the absence of Wnt, the “destruction complex” persistently breaks down β‐catenin, the main effector of this pathway. After stabilizing, β‐catenin is transported to the nucleus of the cell, where it contributes to controlling proliferation and differentiation. LPA receptors (LPA1–6), PG receptors, PTHR1, and endothelin receptors (ET1‐4) are additional GPCRs actively participating in the β‐catenin stabilization pathway.

Due to either oncogenic alterations in its phosphorylation site at the N‐terminus or the mutational ablation of its negative regulators, Axin or APC, hyperactive stabilized β‐catenin is present in many cancers [211, 212]. Particularly in colon, breast, lung, oral, cervical, and hematological cancers, activated β‐catenin may be carcinogenic. Furthermore, Wnt signaling promotes carcinogenesis by affecting the TME through subtle interactions between invasive immune cells, such as leukocytes, and altered cells. Additionally, Wnt signaling contributes to the maintenance of cancer stem cells through the epithelial–mesenchymal transition (EMT). Although it is also transduced by Fz receptors, noncanonical Wnt signaling does not need β‐catenin/Tcf activity or the LRP5/6 coreceptor. Prototypes of this Wnt pathway include Wnt5A and Wnt5B [213]. Cancer, planar cell polarity, tissue regeneration, dorsoventral patterning, and convergent extension movements are among the developmental processes in vertebrates that are impacted by noncanonical Wnt signaling. Noncanonical signaling generally opposes canonical Wnt/β‐catenin signaling and facilitates the Rho‐associated protein kinase (ROCK) pathway, a key regulator of the cytoskeleton. The Wnt–Ca2+ pathway, which controls TAK1‐induced Nemo‐like Kinase and nuclear factor of activated T cells and is involved in cancer development, is another illustration of β‐catenin‐independent signaling. Stromal cells secreted growth factors within the TME, which impact Wnt/β‐catenin signaling. In colorectal cancer cells, for example, stimulation of hepatocyte growth factor has been shown to facilitate tyrosine‐based β‐catenin phosphorylation and its dissociation from Met through the PI3K pathway, promoting tumor development and invasion [214]. Additionally, PDGF, TGF‐β, and EGF stimulated β‐catenin translocation, leading to EMT [215]. In mouse models, vascular endothelial growth factor (VEGF)‐dependent angiogenesis was enhanced by Wnt/β‐catenin signaling [216]. The accurate role of G proteins remains an intriguing and unresolved issue in Fz‐mediated Wnt/β‐catenin signaling. Despite some studies suggesting that Wnt signaling is affected by G proteins [217, 218, 219], other studies have failed to demonstrate that G proteins are an essential component of Wnt/β‐catenin signaling [220, 221]. Research using transfection of MEF cells showed that Gαi and the Wnt/β‐catenin pathway did not interact [221]. G proteins have various effects on modulating Wnt/β‐catenin signaling; however, the study showed that they are insufficient to increase Wnt/β‐catenin signaling in MEF cells when exogenous Wnt3a is present. Gαs enhances Wnt/β‐catenin signaling, decreased by Gαq and Gα13, and unaffected by Gαi in the same experimental setup. G proteins are likely necessary for the Wnt/β‐catenin signaling pathway in all cell types, given their importance as key components. On the other hand, the scientists concluded that Gα proteins are not essential for pathway transduction and are not a component of the core Wnt/β‐catenin signaling pathway [221]. Consequently, the function of G proteins in Wnt signaling pathways remains a contested topic (Figure 4A).

FIGURE 4.

FIGURE 4

Wnt/β‐catenin pathway and dysregulation of GPCR signaling in cancer. (A) The Wnt/β‐catenin pathway is characteristic of the Frizzled (FZ) class F GPCRs, stimulated by Wnt (Wingless/Int1) proteins and activating G protein‐independent (DVL) and G protein‐dependent pathways. The canonical Wnt pathway activates β‐catenin signaling by binding Wnt to the Fz and LRP5/6 receptors. Without Wnt, β‐catenin is degraded by the “degradation complex.” Wnt binding disrupts this complex, stabilizing β‐catenin, facilitating its nuclear translocation, and promoting transcriptional activation via Lef/Tcf factors. Noncanonical Wnt signaling (e.g., Wnt5a) is mediated via Fz, affecting, among others, the activation of JNK and the cytoskeleton. (B) GPCR dysregulation in cancer is illustrated, showing its contribution to cancer hallmarks, including uncontrolled proliferation, evasion of apoptosis, and metastasis. Abbreviations: GSK3β—glycogen synthase kinase 3 beta, TCF/LEF—T‐cell factor/lymphoid enhancer‐binding factor, JNK—c‐Jun N‐terminal kinase, NF‐κB—nuclear factor kappa B, mTOR—mechanistic target of rapamycin, MAPK—mitogen‐activated protein kinase, ERK—extracellular signal‐regulated kinase, ROCK—Rho‐associated coiled‐coil containing protein kinase, RhoA—Ras homolog family member A, FAK—focal adhesion kinase, Src—proto‐oncogene tyrosine‐protein kinase Src, YAP—Yes‐associated protein, P38—p38 mitogen‐activated protein kinase.

6.2. Hippo Signaling Pathway

The oncoprotein is the transcriptional coactivator with a PDZ‐binding motif (TAZ)/Hippo– YAP pathway. It has become an essential conserved system that controls polarity, cell adhesion, mechanical and cytoskeletal proteins, organ size, and cell growth and transformation [222]. Cancer develops as a result of this mechanism being dysregulated. Two crucial downstream effectors of Hippo signaling that play a significant role in cancer are the protein YAP and its homolog, TAZ. Researchers are developing pharmacological inhibitors of TAZ and YAP, two key targets for cancer therapeutics. The oncogenic YAP pathway is triggered when the tumor‐suppressing Hippo pathway is disrupted, which limits the nuclear localization and transcriptional activity of YAP/TAZ. Once the Hippo enzymatic cascade is blocked, YAP/TAZ are released from their cytoplasmic anchoring point and migrate to the cell nucleus. They function as transcription coactivators in the nucleus by binding to transcription factors of the TEAD family, which activates downstream target genes and promotes oncogenicity. The Mst1–2–Lats1/2 kinase cascade of the Hippo pathway directly phosphorylates YAP/TAZ, which causes cytoplasmic retention through 14‐3‐3 binding. This encourages TrCP‐mediated ubiquitination and destruction of YAP/TAZ. Following the initial finding of LPA–YAP/TAZ and sphingosine 1‐phosphate (S1P) activity, GPCRs were identified as efficient YAP oncogenic pathway inducers in the pursuit of physiological YAP/TAZ [133, 223, 224]. It has been demonstrated that GPCRs linked to cell proliferation increase the transcriptional activity of the coactivator YAP [133, 135, 222, 225]. There are several methods in which GPCRs have been shown to control the Hippo pathway. Through the Gα12/13, Gαq, or Gαi routes, LPA and thrombin receptors lead to the activation of YAP/TAZ. However, epinephrine and glucagon receptors mediate the Gαs pathway, which inhibits the YAP/TAZ pathway. Through Gα12/13, It has been demonstrated that GPCRs reduce LATS activity. Consequently, YAP is released from LATS‐dependent suppression [133]. Based on the Gutkind group's study, activation of YAP via oncogenic mutations in Gαq through actin polymerization and a mechano‐sensing pathway instead of interfering with the Hippo‐suppressing pathway [123]. PKA mediates upstream signals by preventing the synthesis of actin fibers or by directly phosphorylating LATS1/2 [226, 227]. PKC seems to have particular effects on its isoforms; for instance, while novel PKC isoforms reduce YAP/TAZ activity, classical PKC isoforms increase it [228]. While MST1/2 does not appear to be a direct target of GPCR signaling, MAP4Ks‐mediated LATS1/2 phosphorylation responds to a range of GPCR ligands [229, 230]. Consequently, G protein and GPCR activity in Hippo signaling is most likely tissue‐dependently influenced by Rho GTPases, actin cytoskeleton remodeling, and protein kinases (including PKA and PKC). Nevertheless, the mechanism is still unknown [231]. Furthermore, according to current research, YAP/TAZ have been identified as the downstream effectors in the noncanonical Wnt signaling pathway, which includes Wnt–FZ/ROR, G12/13‐Rho GTPases, and Lats1/2. This pathway promotes TEAD‐mediated gene transcription and the carcinogenic effects of YAP/TAZ.[232]. Additionally, Hh ligands diminish YAP/TAZ through the SMO–Gαs–cAMP–PKA signaling pathway [160]. These results suggest that atypical GPCRs (FZ, SMO) regulate the Hippo pathway and the interactions between the Hippo pathway and other critical pathways in cancer progression. Moreover, GPCRs’ impact on the activity of YAP/TAZ, regulated by PKD and PI3K downstream of the insulin receptor, is enhanced in the presence of insulin [233]. It has also been shown that MAPK signaling modulates the Hippo pathway [234]. Given that YAP/TAZ‐induced transcriptional activities are heavily involved in cancer, weakening YAP and TAZ can be a feasible treatment and prevention strategy for a variety of cancers. A possible approach might be to lower the YAP dosage by using shRNA depletion. When the shRNA‐induced mortality of a large panel of human cancer cell lines was investigated, it was demonstrated that cancer cell lines with WNT signaling stimulation are especially susceptible to YAP knockdown [235]. Due to this, YAP suppression does not always correspond with its conduct, and blocking YAP may involve critical TEAD‐independent interactions that are particular to some tumor cells.

Additionally, medications that target GPCRs and G proteins may reduce YAP/TAZ activation and decrease the spread of cancer because the Hippo pathway is highly mediated by GPCR‐regulated downstream signaling. Gαs‐targeted substances, for instance, may similarly reduce YAP/TAZ function to epinephrine, dobutamine, and glucagon [236, 237]. Antagonizing or decreasing Gα12/13‐, Gαq/11‐, or Gαi/o‐mediated signals, as well as employing monoclonal antibodies (mAbs) specific for LPA or S1P and phosphatase‐resistant LPA analogs [238, 239], may decrease the YAP/TAZ function. It has recently been demonstrated that the cyclic depsipeptide FR900359 binds to mutant Gαq and inhibits the downstream effectors of MAPK and YAP [240, 241]. Moreover, it has been reported that phosphodiesterase inhibitors such as Rolipram or forskolin activate PKA and inhibit YAP/TAZ [160, 238]. Also, PKC inhibitors can reduce YAP/TAZ activity depending on the type of cell [242]. However, it has been demonstrated that several GPCR‐based drugs, such as dopamine and blockers, have significant effects on cardiac and psychological functions; as a result, adverse effects must be addressed before using these drugs in cancer treatment [243, 244] (Figure 5A).

FIGURE 5.

FIGURE 5

Key GPCR‐associated pathways in tumorigenesis. (A) Hippo signaling pathway: LATS1/2 kinases phosphorylate YAP/TAZ, thereby inactivating them and restricting their nuclear entry. Gαs activates Hippo (inhibiting YAP/TAZ), while Gα12/13, Gαi, and Gαq/11 activate YAP/TAZ, promoting tumorigenesis. (B) Hedgehog pathway: In the absence of Hh, PTCH inhibits SMO, thereby blocking GLI activation. When Hh binds to PTCH, SMO is activated, leading to the activation of Gαi signaling and, in turn, activating GLI‐driven transcription that promotes tumor growth. (C) β‐arrestin‐mediated signaling: β‐arrestins (β‐arr1, β‐arr2) modulate GPCR desensitization by inhibiting G proteins while activating G protein‐independent pathways. These include ERK1/2, PI3K/AKT, and Hedgehog signaling, which are critical for cancer progression. This panel highlights the dual role of β‐arrestins in regulating GPCR signaling and promoting tumor growth and metastasis. Abbreviations: MST1/2—mammalian sterile 20‐like kinase 1 and 2, LATS1/2—large tumor suppressor kinase 1 and 2, YAP—Yes‐associated protein, TAZ—transcriptional coactivator with PDZ‐binding motif, TEAD1—TEA domain transcription factor 1, SHh—sonic hedgehog, PTCH—patched receptor, GLI—glioma‐associated oncogene homolog, VEGF—vascular endothelial growth factor, CXCL8—C‐X‐C motif chemokine ligand 8.

6.3. The Hh Pathway

The Hh pathway is essential in stem cell maintenance and embryonic development, and is activated in tumors, and its deregulation may lead to neoplastic transformation [245]. The GPCR SMO, the tumor suppressor patched (PTCH) cell surface receptor, and the transcription factors for glioma‐associated oncogenes (GLI) are the primary elements of this cascade. PTCH inhibits SMO activity when it is quiescent (unstimulated), which prevents GLIs from activating and becoming ubiquitinated. To promote proliferation and survival during carcinogenesis, Hh releases SMO upon binding to PTCH, which stimulates Gαi signaling and activates GLIs. The GPCR PTCH and SMO are important targets for regulation, as inactivating PTCH mutations or activating SMO mutations are linked with medulloblastoma and basal cell carcinoma [246]. Despite mounting evidence of an SMO‐independent variant Hh route, SMO inhibitors have evolved to treat basal cell carcinoma. To address this, SMO/GLI multitarget antagonism is a practical approach to targeting medulloblastoma [247, 248] (Figure 5B).

6.4. GPCR–β‐Arrestin pathway

β‐Arrestins have emerged as key players in cancer‐related GPCR signaling, extending their traditional role in receptor desensitization. In various tumor types, they influence critical cellular behaviors such as cytoskeletal reorganization, migration, and transcriptional regulation. Their context‐ and isoform‐specific roles, particularly those of β‐arrestin 1 and β‐arrestin 2, vary across different cancer types and are influenced by the TME. Figure 5C illustrates how β‐arrestins interact with diverse intracellular effectors and signaling hubs, contributing to both tumor‐promoting and suppressive outcomes. Their involvement in processes such as metastasis, immune evasion, and nuclear signaling highlights their complexity in cancer biology. Continued investigation into β‐arrestin functions may help identify novel intervention points that are distinct from traditional GPCR–G protein signaling pathways.

6.5. Mitogen‐Activated Protein Kinases

MAPK signaling is essential for proliferation and survival. The proteins Ras and Raf, known to contain distinctive oncogenic alterations, are downstream of the layers of kinases that make up the MAPK pathways [249]. This pathway is linked to tumor‐promoting activity and is activated by the Gαq, Gαi, and Gβγ subunits that are regulated by GPCRs (Figure 4B).

6.6. PI3K–AKT–mTOR/NF‐κB

Cell migration, survival, growth, and metabolism are all influenced by the PI3K–AKT–mammalian target of rapamycin (mTOR)/NF‐κB pathway. It frequently exhibits dysregulation throughout tumor development, supporting proliferative signaling, preventing cell death, and facilitating immune evasion. The PI3K/Akt/NF‐κB axis can be activated by GPCR signaling through Gαq and Gβγ upon activation. This pathway enhances the expression of interleukin‐8 (CXCL8) and VEGF, which promotes key cancer hallmarks, including angiogenesis and metastasis. The EMT, a crucial step in metastasis, is mediated by the PI3K/Akt signaling pathway. For example, GPR120 stimulates the development of breast cancer [250]. These oncogenic signaling events are triggered by GPR137 (orphan, constitutively active), which causes ovarian cancer [251]. Alternatively, Gαi can activate the PI3K pathway through the chemokine receptor CXCR4, leading to NF‐κB activation and chemotaxis [252]. The serine/threonine kinase mTOR, which regulates cell growth, survival, and metabolism, particularly in cancer cells, can be activated by the protein kinase AKT. This cascade also receives signals that lead to the suppression of apoptosis. Gαs‐coupled GPCRs phosphorylate Raptor, a mTOR‐associated protein component of the mTOR complex (mTORC1), via PKA, which inhibits mTOR [253]. Therefore, one strategy to block the mTOR complex and reduce its proliferative function in cancer may be to activate Gαs‐coupled GPCRs (Figure 4B).

6.7. RhoGEFs (guanine exchange factors)

RhoGEFs are signaling mediators triggered by Gα12/13‐coupled GPCRs. By promoting tumor growth, angiogenesis, and metastasis, they activate key transcription factors associated with cancer. The Cholecystokinin B receptor (CCKBR) in pancreatic cancer signals through Gα12/13 when its ligand, Gastrin, activates it. Through the phosphorylation of FAK, the downstream effector ROCK of Gα12/13 is activated, leading to the phosphorylation of paxillin, a protein involved in cell adhesion and migration. Paxillin contributes to the EMT and proliferation, whereas FAK modulates downstream elements, including PI3K and Src [254]. aGPCRs have been shown to activate Gα12/13 proteins and are involved in cell adhesion to the extracellular matrix (ECM) and cell‐to‐cell communication. Among other malignancies, mutations and altered expression of aGPCRs have been identified in the prostate, colon, lung, and breast [255] (Figure 4B).

In summary, diverse GPCR‐mediated signaling pathways, such as Wnt, Hh, PI3K–AKT–mTOR, MAPK, and β‐arrestin pathways, play critical roles in tumorigenesis by regulating proliferation, survival, invasion, and immune modulation. The complexity and redundancy of these pathways underscore the challenges and opportunities in targeting GPCRs for cancer therapy. A deeper understanding of the pathway‐specific roles of GPCRs may guide the development of more effective, targeted interventions with reduced off‐target effects.

7. GPCRs in the Hallmarks of Cancer

GPCRs play essential roles in various physiological processes, and their dysregulation is increasingly recognized as a key driver in cancer. They contribute to major hallmarks of cancer, including uncontrolled proliferation, resistance to apoptosis, invasion, metastasis, angiogenesis, and immune evasion. Upon activation by ligands such as chemokines, hormones, and bioactive lipids, GPCRs trigger complex signaling cascades that reshape the TME and influence tumor–host interactions. This section focuses on their roles in migration, invasion, angiogenesis, and immune regulation.

7.1. Role in Migration, Invasion, and Metastasis

One of the most significant challenges in cancer treatment is metastasis, the process by which tumor cells migrate to other organs through the bloodstream or lymphatic vessels [256]. Instead of spreading randomly, tumor cells can selectively metastasize to particular organs with a higher prevalence [257]. By altering the cytoskeletal dynamics and structure of cells bearing receptors, chemokines are known to direct cell migration and facilitate metastasis. Furthermore, locally produced chemokines within the TME recruit leukocytes and macrophages, thereby increasing the cytokine‐rich environment and reducing the secretion of matrix metalloproteases (MMPs), which in turn promote the invasion, survival, and proliferation of cancer cells. Furthermore, organ‐specific metastasis is strongly correlated with GPCRs coupled to chemokine receptors in various types of cancer. Research has demonstrated that cancer cells with altered expression of chemokine GPCRs exploit chemokine migration, facilitating metastasis to multiple organs [258]. Among the most well‐known receptors for chemokines, CXCR4 is involved in migration, survival, and proliferation. It is frequently overexpressed in many types of cancer, which aids in metastasis. The widespread metastatic sites, like the bone marrow, liver, lungs, and lymph nodes, demonstrate that CXCL12/SDF‐1 is the chemokine ligand for CXCR4.[257]. It was shown that breast cancer cells express CXCR4 abundantly. Rac1 is activated by its activation through P‐REX1, which contributes to the spread of most breast‐type malignancies. Additionally, CXCR4 may link to G12/13, promoting metastasis in basal‐like breast cancer cells in a RhoA‐dependent manner [194]. Targeting the downstream signaling or the molecule regulating CXCR4 expression on tumor cells could provide therapeutic alternatives. It has been demonstrated that additional chemokine receptors, including CCR7 and CCR10, directly contribute to the proliferation, survival, and metastatic homing of cancer cells [259]. Additionally, several new investigations are underway to better understand the adhesion family of GPCRs and their potential contributions to the development and spread of cancer [260]. According to recent research, GPR116, a member of the mainly unidentified aGPCR family, activates the Gαq‐RhoA‐Rac1 pathway, contributing to the invasion and migration of breast cancer cells.

7.2. GPCRs in Tumor‐Induced Angiogenesis

Nag et al. [261] recently reviewed the different aspects of the cardinal GPCRs associated with tumor angiogenesis. Tumors release angiogenic substances that promote the migration and proliferation of endothelial cells, thereby forming new capillaries to meet the increased oxygen and nutritional needs of tumor cells. Numerous angiogenic substances, including S1P, PGs, thrombin, and chemokines, act on GPCRs expressed in endothelial cells [262, 263, 264]. Specific chemokines, including CCL5, CXCL8/IL‐8, and CCL2, can attract macrophages and leukocytes to the cancer site, where they might produce other angiogenic factors and VEGF that promote the formation of new blood vessels [263]. Moreover, the TME's inflammatory cytokines increase the production of COX‐2 and the local release of PGE2, which in turn increases the expression of proangiogenic VEGF, CXCL8, and CXCL5 in tumor and stromal cells [265]. By increasing the proliferation of endothelial cells, GPCRs and their ligands can directly stimulate angiogenesis. Alternatively, they can indirectly stimulate the production of other angiogenic factors and VEGF from immune, malignant, or stromal cells. Tumor vascularization enables pathways for invasion, metastasis, and the delivery of nutrients necessary for tumor growth.

7.3. TME Remodeling to Promote Immune Escape

The TME is a unique niche inhabited by a variety of cell types, including stromal cells, immune cells, cancer cells,  ECM, and diverse secreted substances, such as exosomes and microRNAs [266, 267]. Tumor growth, metastasis, and treatment outcomes are correlated with the altered TME landscape [268]. The TME score, an advanced TME infiltration pattern of GC, was recently established by evaluating cancer‐associated fibroblasts (CAFs) and 22 immune cell types, which are connected to pathologic and genomic features [266]. CAF biology and function have become a field of continuous, active research, and have been previously examined [269, 270]. The chemokines and their receptors are the most significant methods involved, and the compositions of infiltrating immune cells within the TME differed widely [271]. Interestingly, tumor, immunological, and stromal cells can all express chemokines and chemokine receptors [272]. The composition of TME immune cells was influenced by changes in chemokines and their receptors. To evade immune surveillance and eradication, they altered the immune responses, some of which were taken over by tumor cells [273]. The induction of immune cells, primarily dendritic cells (DCs), CD8+ T cells, NK cells, and M1 macrophages, triggered antitumor immune responses. GCs with elevated CXCR3 expression showed enhanced DC and T cell infiltration. The recruitment of DCs depends on the CXCR3/CXCL4 or CXCR3/CXCL4L1 axis, as these cells strongly stimulate T cells and activate the related humoral response, resulting in antitumor effects [274, 275]. Likewise, CXCR3 is essential for CD8+ T cell infiltration, which causes direct damage to tumor cells once the cells differentiate into cytotoxic CD8+ T cells [276, 277]. Furthermore, professional killer cells, or NK cells, are accumulated in the TME as a result of increased CXCL10 and CXCL12 signaling via CCR7 or CXCR3 [278, 279].

In contrast, chemokine signaling also plays a role in forming an immunosuppressive TME, allowing tumors to evade detection. This mechanism is primarily associated with the infiltration of several protumor immune cell types, including granulocytic (or PMN‐) myeloid‐derived suppressor cells (MDSCs), M2 macrophages, regulatory T (Treg) cells, and monocytic myeloid‐MDSCs [271, 280]. CCL22, produced mainly by tumor cells (or macrophage‐mediated), interacts with the receptor CCR4 on the surface of Treg cells to increase their number in the TME. Treg cells promote migration in response to CCL28 by another receptor, CCR10 [281, 282]. Furthermore, recruited monocytes can produce nonpolarized macrophages (M0) that can differentiate into two main subtypes: M1 and M2 macrophages. These two subtypes play very diverse roles in cancer. A variety of chemokine signals influences these transitions. Tumor‐derived chemokine releases, including CCL2, CCL3, CCL4, CCL5, CCL20, and CCL18, were necessary for active monocyte recruitment. Furthermore, blocking the CCL2‐CCR2 circuit resulted in an increase in M2 macrophages, while CCL11 favored macrophages toward an M2 phenotype [283, 284]. Other GPCRs also mediate the regulation of immunological responses. For example, through their respective GPCRs, EP1‐EP4 (PTGER1‐4), PGs can promote inflammation. The cyclooxygenases COX‐1 and COX‐2 produce PGs, especially PGE2. Nonsteroidal anti‐inflammatory medicines are examples of enzyme inhibitors used to reduce the incidence of some cancers and relieve pain. PGE2's ability to trigger inflammation by activating additional signaling pathways, including the Wnt, EGFR, and Toll‐like receptor/MyD88 pathway, has been thoroughly investigated [285, 286].

GPCRs play multifaceted roles in driving key hallmarks of cancer, including sustained proliferation, invasion, metastasis, and angiogenesis. Through interactions with chemokines, adhesion molecules, and diverse signaling pathways, GPCRs modulate both tumor cell behavior and the TME. Their ability to orchestrate organ‐specific metastasis and influence immune cell infiltration highlights their clinical relevance. Understanding these mechanisms offers promising therapeutic opportunities to disrupt cancer progression at multiple levels.

8. Recent Advances in Therapeutic Development Targeting GPCR Signaling Bias

Recent advances in therapeutic development targeting GPCR signaling bias have revolutionized the field by enabling more precise modulation of receptor pathways. Rather than uniformly activating or inhibiting all GPCR‐mediated signals, new strategies focus on selectively directing specific intracellular responses through the use of biased agonists and antagonists, PROTAC‐based modulation, mAbs, nanobodies, aptamers, and gene therapy. These innovative approaches, supported by breakthroughs in structural biology and pharmacology, are expanding the potential of GPCR‐targeted treatments for cancer and other complex diseases, offering improved efficacy and safety over traditional therapies.

8.1. Biased Agonists and Antagonists

Approximately 60% of medications in development and 36% of marketed treatments approved by the US FDA target human GPCRs, indicating that GPCRs are an essential target for many medicines. Furthermore, because many GPCRs are implicated in the development and spread of cancer, possibly impacting both treatment effectiveness and patient survival, GPCRs are regarded as some of the most promising targets for treatment in a wide range of solid malignancies. However, only a few GPCRs have been successfully utilized to create medications that block signaling pathways linked to cancer. More knowledge about GPCR biology reveals that functional selectivity and “biased agonism” exist; consequently, the concept of “one medication per GPCR target” is not as prevalent, and more attention is being paid to exploring different pharmacological options.

Understanding the mechanism of GPCR activation is crucial, as it can aid in the development of anticancer medications. There are numerous opportunities to develop innovative cancer treatments. Targeting GPCR signaling with agonists or antagonists and focusing on the unique interactions between GPCRs and their binding partners are two targeted methods for therapeutic development. These approaches aim to deliver anti‐neoplastic medications or toxins directly to malignant cells. For instance, an endocrine treatment for hormone‐responsive prostate cancer that targets the GnRH receptor lowers testosterone levels. This approach helps cure prostate cancer because promoting Prostate cancer proliferation requires the production of testosterone via a signaling pathway that starts with the release of GnRH from the hypothalamus [287]. An immunological method, such as direct vaccination administration, can restrict endogenous agonists from binding to a specific GPCR to achieve the intended neutralizing effect. Immunogen G17DT, a rare example, is being evaluated in a phase III trial to treat pancreatic cancer [288]. The atrasentan and the ETAR antagonist ZD4054 can effectively reduce the proliferation and invasion of cancer cells when combined with the monoclonal HER2‐specific antibody trastuzumab and the EGFR inhibitor gefitinib, respectively. However, atrasentan and ZD4054 are no longer used as anti‐tumor drugs and have been taken out of clinical trials due to their numerous adverse effects on patients [289]. Similar to this, the biased antagonist Vorapaxar can modulate PAR1, which is known to promote tumor invasion and angiogenesis. By blocking G protein activation without affecting β‐arrestin signaling, it potentially reduces thrombotic risks while attenuating malignant behavior in cancers like breast and prostate [290]. Biased ligands are being investigated for supportive care in oncology in addition to their direct antitumor effects. For instance, oliceridine (TRV130), a G protein‐biased μ‐OR agonist, offers potent analgesia with fewer respiratory and gastrointestinal side effects than morphine and may also reduce immunosuppression, which is a significant concern in maintaining immune surveillance during cancer therapy [291].

8.2. PROTAC‐Based GPCR Modulation

In targeted protein degradation, PROTACs have revolutionized the field. They provide an innovative approach to modify disease‐relevant proteins, such as GPCRs, in cancer. By recruiting E3 ubiquitin ligases to the target protein, PROTACs induce the selective ubiquitination and subsequent proteasomal degradation of target proteins, in contrast to conventional small‐molecule inhibitors or antagonists that only inhibit receptor activation. Since GPCRs frequently cause tumors through aberrant or biased signaling pathways, this catalytic mode of action enables the complete elimination of both WT and mutant GPCRs from the cell surface [29]. According to recent research, it is feasible to create PROTACs that specifically target GPCRs linked to cancer, such as the estrogen receptor (ER) in breast cancer and the AR in prostate cancer, leading to strong anti‐tumor effects in preclinical models [292]. While most PROTAC development has focused on nuclear hormone receptors and kinases, proof‐of‐concept studies targeting membrane GPCRs are emerging, highlighting challenges such as cell permeability, receptor accessibility, and the selection of appropriate E3 ligases for efficient degradation [293]. Furthermore, in order to overcome compensatory signaling and drug resistance in advanced malignancies, dual‐targeting PROTACs that degrade several oncogenic drivers (such as EGFR and PARP) are being investigated [292]. The clinical translation of GPCR‐targeted PROTACs still faces several challenges, despite these advancements, including improving pharmacokinetics, optimizing ligand affinity, and achieving tissue‐selective degradation to reduce off‐target effects. However, a convincing method to specifically eradicate oncogenic GPCRs and associated pathological signaling networks is provided by the combination of PROTAC technology with the idea of biased GPCR signaling, opening the door for next‐generation cancer treatments.

8.3. mAbs Targeting GPCR

GPCRs are the targets of mAbs, which represent an attractive, rapidly evolving frontier in therapeutic development, particularly for cancer and immune‐related diseases. Due to several technical challenges, the development of mAbs against GPCRs has historically lagged behind small‐molecule methods, despite GPCRs being the most prominent family of membrane proteins and representing the targets for a significant portion of approved drugs. GPCRs’ intrinsic instability outside of their native lipid bilayer, their comparatively tiny and frequently conformationally dynamic extracellular regions, and the difficulty in generating high‐quality antigens that maintain the receptor's natural structure and function are some of these challenges [29, 30]. These challenges have begun to be addressed by advancements in structural biology, such as cryo‐electron microscopy, and novel antigen presentation techniques, including proteoliposomes and virus‐like particles, which have enabled the production of antibodies that can recognize native GPCR conformations [30]. By preventing ligand binding, locking the receptor in a specific conformation, or recruiting immune effector functions such as antibody‐dependent cellular cytotoxicity (ADCC), therapeutic mAbs can exert their effects. Interestingly, the defucosylated anti‐CCR4 antibody mogamulizumab is approved for use in cutaneous T‐cell lymphoma and adult T‐cell leukemia/lymphoma, where it facilitates the removal of tumor cells through enhanced ADCC [30, 294]. The therapeutic flexibility of this method is demonstrated by other mAbs, including erenumab, which prevent migraines by targeting the calcitonin gene‐related peptide receptor [295]. mAbs targeting chemokine receptors, such as CXCR4 and CCR5, are being investigated for their potential to prevent metastasis and modulate the TME in the context of cancer. Meanwhile, antibodies targeting aGPCRs, like GPRC5D, are advancing in clinical trials for multiple myeloma [29, 30]. Additionally, recent advancements are making it possible to create bispecific antibodies and ADCs that target GPCRs, thereby expanding the therapeutic options and enabling more accurate targeting of tumor cells. Moreover, structure‐guided antibody engineering allows the production of mAbs that can stabilize GPCRs in particular conformational or signaling states, potentially biasing receptor signaling in a manner advantageous for therapy [29, 30, 31, 32]. Even though challenges like tissue specificity, delivery, and overcoming tumor heterogeneity remain, the combination of advanced antibody engineering, structural insights, and creative antigen design is significantly accelerating the clinical translation of GPCR‐targeted mAbs. mAbs targeting GPCRs are therefore expected to play a significant role in immunotherapy and precision oncology in the years to come.

8.4. Emerging Strategies: Nanobody, Aptamers, and Gene Therapy

Emerging strategies for targeting GPCRs in cancer and other diseases are rapidly evolving beyond traditional small molecules and mAbs, with nanobodies, aptamers, and gene therapy offering unique advantages and novel mechanisms of action.

8.5. Nanobodies

Single‐domain antibody fragments, known as nanobodies, which are derived from camelid heavy‐chain antibodies, have garnered significant interest due to their small size, high stability, and ability to bind distinct or hidden GPCR epitopes that are often inaccessible to conventional antibodies [296, 297]. Their superior tissue penetration and rapid clearance make them attractive for both diagnostic imaging and targeted therapy [34, 296, 298]. In cancer models, nanobodies have been effectively employed to block GPCR signaling. They can also be engineered as targeting modules for chimeric antigen receptor (CAR)‐T cell therapies, drug conjugates, or photodynamic therapy agents.[34, 298, 299, 300] For instance, in photodynamic therapy, nanobody–photosensitizer conjugates have shown the ability to kill tumor cells that express GPCR selectively, and nanobody‐based CAR‐T cells have shown promise in the treatment of solid tumors [34, 299] By enhancing tumor selectivity and diminishing off‐target effects, nanobody–drug conjugates and nanobody‐functionalized nanoparticles further broaden the therapeutic toolkit [34, 298]

8.6. Aptamers

GPCRs can be bound by aptamers, which are short, single‐stranded nucleic acids that fold into particular three‐dimensional structures with great affinity and specificity [33]. Aptamers, selected via the SELEX process, offer several advantages, including reduced immunogenicity, ease of synthesis, and the ability to alter receptor activity either competitively or allosterically [19, 33]. Aptamers have demonstrated inhibitory action that targets GPCRs, such as CCR5 and MRGPRX2. Aptamer–siRNA chimeras have been developed to deliver gene‐silencing payloads to GPCR‐expressing cells with precision, exhibiting potential for both targeted gene therapy and direct receptor inhibition [33]. Additionally, aptamers can be designed to disrupt GPCR dimerization or oligomerization, providing another layer of regulatory control [19, 33, 297].

8.7. Gene Therapy

Additionally, gene therapy techniques are advancing, using both viral and nonviral vectors to alter GPCR function or expression at the genetic level. To downregulate oncogenic GPCRs, strategies include delivering genes encoding modified GPCRs, dominant‐negative receptor variants, or gene‐silencing constructs like siRNA or shRNA [301, 302].

8.8. Current Clinical Trials Landscape

GPCRs continue to be the most frequently used target class in medicine; 516 approved drugs targeting 121 distinct GPCRs across a wide range of diseases, and 30–50% of marketed drugs act through these receptors [303, 304]. In recent years, there has been a surge in interest in targeting GPCRs for cancer and other complex diseases, even though GPCRs have long been essential to treatments for neurological, metabolic, and cardiovascular disorders, such as bronchodilators for asthma, angiotensin receptor blockers for hypertension, and antipsychotics for mental health [303, 304, 305]. This shift is driven by increasing evidence that GPCRs control key cancer hallmarks, including immune evasion, metastasis, proliferation, and therapy resistance, and that approximately 20% of all malignancies have mutations or abnormal expression of GPCRs [303]. Through clinical trials, several GPCR‐targeted agents are making progress in oncology. Adenosine A2A receptor‐targeting minor molecule antagonist AZD4635 and ONC201, an imipramine that antagonizes the dopamine receptor D2 (DRD2) and has demonstrated efficacy in glioblastoma in phase II trials, are noteworthy examples [303]. Another example is the use of SMO antagonists, which block Hh signaling in basal cell carcinoma and other cancers [303, 305]. Additionally, biologics are gaining momentum. For example, mAbs and nanobodies that target chemokine receptors, such as CXCR4 and CCR4, are being used for metastatic malignancies, as well as novel therapeutic approaches, including peptide‐based medications and ADCs, which are currently undergoing clinical evaluation [305]. GPCR‐targeted medications are still the most common treatment for chronic diseases apart from cancer. While new agents for cardiovascular and respiratory disorders remain in high demand, drugs that target GLP‐1 and GIP receptors, for instance, are transforming the treatment of diabetes and obesity, and are expected to generate approximately $30 billion in sales by 2023 [303, 304]. Allosteric modulators and pathway‐biased ligands are recent clinical developments that aim to optimize GPCR signaling for increased efficacy and fewer side effects in both cancer and noncancer applications [303, 304]. The pipeline remains robust despite challenges, including the structural complexity of GPCRs and the need for more accurate biomarkers. As of 2025, 337 agents targeting 133 GPCRs, including 30 novel targets, were in clinical trials, with a focus on developing novel drug modalities and broadening indications [304]. The landscape of clinical trials for GPCR‐targeted therapeutics is becoming more diverse in terms of drug modalities, disease indications, and mechanistic sophistication. This includes biologics and small compounds targeting immune checkpoints, chemokine receptors, and orphan GPCRs in cancer; however, in other diseases, GPCRs continue to lead the way in drug discovery and development, with an ongoing stream of novel medicines and clinical advancements [303, 306].

In conclusion, the expanding arsenal of GPCR‐targeted therapeutics, including small‐molecule agonists and antagonists, biased ligands, mAbs, nanobodies, aptamers, gene therapies, and novel approaches like PROTACs, reflects the central and multifaceted role of GPCRs in cancer biology and treatment. These strategies exploit the ability of GPCRs to regulate tumor growth, metastasis, immune responses, and the TME, offering new avenues for more selective and effective anticancer treatments. Table 2 summarizes currently utilized anticancer drugs and antibodies targeting GPCRs and their respective targets, highlighting the clinical impact of this receptor family [29, 305, 307].

TABLE 2.

US FDA‐approved drugs and antibodies currently being used to treat various malignancies.

Drugs Receptor Cancer type Types of drugs Year of approval References
Cabergoline DRD1 Neuroendocrine tumors, pituitary tumors Small molecule 1996 [308]
Lanreotide SSTR Pancreatic cancer Peptide 2007 [309]
Degarelix GnRH Prostate cancer Peptide 2008 [310]
Plerixafor CXCR4 Multiple myeloma Small molecule 2008 [311, 312]
vismodegib (Erivedge) SMO Locally advanced and metastatic basal cell carcinoma Small molecule 2012 [313]
Raloxifene ER Breast cancer Small molecule 2014 [314]
Sonidegib (Odomzo) SMO Locally advanced and metastatic basal‐cell carcinoma Small molecule 2015 [315]
Mogamulizumab CCR4 Cutaneous/peripheral T‐cell lymphoma; adult T‐cell lymphoma mAb 2018 [294, 316]
Lutathera (Lutetium Lu 177 dotatate) SSTR Gastroenteropancreatic neuroendocrine tumors (GEP‐NETs) Peptide 2018 [317]
Gilteritinib Serotonin receptors Relapsed or refractory acute myeloid leukemia Small molecule 2018 [318]
Lutetium 177 dotatate SSTR2 Gastroenteropancreatic neuroendocrine tumors Radiolabeled peptide 2018 [319]

Abbreviations: DRD1: dopamine receptor D1, SSTR: somatostatin receptor, GnRH: gonadotropin‐releasing hormone receptor, CXCR4: C‐X‐C chemokine receptor 4, SMO: smoothened receptor, ER: estrogen receptor, CCR4: C‐C chemokine receptor type 4, mAb: monoclonal antibody.

In cancer and other diseases, combining gene therapy with nanobody or aptamer technologies, such as aptamer‐guided delivery of gene‐editing tools, holds promise for prolonged and selective regulation of GPCR signaling [19, 33]. Collectively, these emerging modalities—nanobodies, aptamers, and gene therapy—are expanding the therapeutic landscape for GPCR‐targeted interventions, offering new avenues for precision medicine, improved selectivity, and the potential to overcome resistance mechanisms associated with conventional therapies [33, 34].

9. Preclinical Animal Experiments on Biased GPCR Signaling

Recent years have seen significant advances in understanding biased signaling at GPCRs, particularly through the use of genetically modified mouse models (e.g., β‐arrestin 1 and β‐arrestin 2 knockout mice). These models have enabled the dissection of distinct signaling pathways activated by different ligands, revealing functional selectivity in vivo.

Mice lacking β‐arrestin 1 (β‐arrestin 1–KO mice) were very useful in identifying the potential for developing biased agonists at GPR109a receptors. Niacin (nicotinic acid), which activates GPR109a receptors, has been used for the treatment of cardiovascular disease for many years because it effectively lowers triglyceride levels and raises high‐density lipoprotein levels in the blood [9]. However, its therapeutic use is limited by the side effect of cutaneous flushing. The flushing response induced by niacin is alleviated in mice lacking β‐arrestin‐1 [320]. Moreover, further studies into the mechanism reveal that niacin‐activated GPR109a receptors signal through β‐arrestin 1 to activate phospholipase A2, thereby increasing arachidonic acid levels [321]. Although the cutaneous flushing is attenuated in the β‐arrestin‐1–KO mice, niacin remains efficacious in its ability to reduce serum free fatty acid levels. These in vivo studies suggest that biased ligands that activate GPR109a but do not recruit β‐arrestin‐1 could be helpful in treating dyslipidemia while avoiding the adverse side effect of cutaneous flushing [322]. The question remains, however, whether niacin's actions at GPR109a are the only means it has to lower serum fatty acid levels. A report by Lauring et al. [323] showed that mice lacking GPR109a were still responsive to niacin's lipid‐lowering properties. This underscores the difficulty of determining whether the effects of the agonist are bifurcated at specific signaling points or if there are additional targets that are not being accounted for when working with whole animal systems. Furthermore, the β‐arrestin 2–KO mice have proven to be very useful in identifying examples of functional selectivity in vivo, particularly pathways that utilize β‐arrestin 2 to promote GPCR‐mediated signaling. The first physiological phenotype reported for the β‐arrestin 2–KO mice was their enhanced responsiveness to morphine‐induced antinociception [324, 325], demonstrating a role for β‐arrestin 2 in negatively modulating μ‐OR responsiveness in vivo. Further studies showed that fentanyl and methadone did not reveal enhanced response profiles in the β‐arrestin 2–KO mice [326]. However, fentanyl and methadone were also known to mediate their analgesic effects through the μ‐OR [327]. These early studies suggest that there is a ligand‐directed bias at the MOR, such that the activity of some agonists, including morphine, is more influenced by the presence of β‐arrestin 2 than others. Additional studies using morphine in the β‐arrestin 2–KO mice suggest the contextual importance of receptor signaling. While morphine‐induced antinociception is negatively regulated by β‐arrestins, morphine‐induced constipation, respiratory suppression, and physical dependence appear to involve β‐arrestin 2, as these side effects are significantly diminished in the β‐arrestin 2–KO mice [327, 328, 329]. While these observations may imply a MOR‐β‐arrestin 2‐dependent signaling pathway, such a mechanism has yet to be mechanistically demonstrated in vivo. The development of MOR agonists that are biased toward G protein signaling and against β‐arrestin 2 recruitment may provide valuable tools for ascertaining whether biased signaling underlies the separation of the antinociceptive properties and side effects induced by opiate narcotics. The first opioid agonist identified as a functionally selective agonist is herkinorin [330]. Although it has limited bioavailability, it exhibits antinociceptive effects when injected into the paw of rats in the formalin test, without producing tolerance with repeated dosing [331]. A recently developed biased agonist (TRV130) has shown promising efficacy in pre‐clinical studies. This compound is potent in G protein‐mediated signaling but exhibits minimal efficacy in inducing β‐arrestin 2 recruitment in cell‐based assays. Importantly, in mouse and rat models, TRV130 exhibits analgesic potencies superior to those of morphine, yet it produces less gastrointestinal transit delay and has a lesser effect on respiratory parameters than morphine. TRV130 serves as a proof of concept demonstrating that the effects of an agonist that does not recruit β‐arrestin 2 can recapitulate the behaviors seen in morphine treated mice that lack β‐arrestin 2 and suggest that the development of G protein signaling‐biased MOR agonists may be a means to promote opioid analgesia while limiting specific side effects [291].

Additionally, PAR1 is a member of the four PARs, which also include PAR2‐4, that act as exquisite sensors for a select group of proteases [332]. PAR1 expression is increased in several cancers, including breast, colon, and lung cancer. A study using a xenograft model of breast carcinoma cells initially demonstrated a critical role for MMP‐1 derived from tumor‐infiltrating fibroblasts in the cleavage of PAR1, which appears to drive the migration and invasive behavior of cancer cells [333]. Under pathophysiological conditions in carcinomas, PAR1 is an oncogenic protein, which is a potent inducer of cancer cell migration, invasion, survival, and metastasis [334, 335, 336, 337, 338, 339]. Researchers have evaluated PAR1‐derived PZ‐128 as a potential PAR1 inhibitor to suppress breast cancer progression. PAR1 was topically expressed in PAR1‐null, estrogen‐sensitive MCF‐7 cells and tested for its ability to promote tumor‐growth and invasion in nude mice. PAR1 expression resulted in a 100% rate of tumor formation while PAR1‐null MCF‐7 cells did not form any palpable tumors [333]. These PAR1‐driven tumors could be significantly inhibited (62%, p < 0.01) with PZ‐128. PAR1‐dependent tumor growth was mediated by MMP1, as treatment with an MMP‐1 inhibitor resulted in an 82% inhibition of tumor growth. Likewise, PAR1‐expressing MCF‐7 cells formed metastatic lesions in the lungs with a 100% penetrance rate, similar to the highly metastatic MDA‐MB‐231. In contrast, the PAR1‐mutant R310E, which lacked signaling abilities, had undetectable metastatic lesions in the lungs [340]. Most strikingly, there was a similar reduction in the number of metastatic lesions with either treatment as monotherapy, PZ‐128, or MMP‐1 Inh (75 and 88%, respectively, p < 0.001) [335]. PAR1 is a poor prognostic marker in lung cancer that correlates with reduced survival in non‐small‐cell lung cancer (NSCLC) [341]. In 2007, bevacizumab (Avastin) was approved in combination with carboplatin and paclitaxel as the first‐line treatment for patients with unresectable, locally advanced, recurrent, or metastatic nonsquamous NSCLC [342]. A comparative efficacy study of PZ‐128 versus Avastin in a well‐established A549 xenograft model of lung adenocarcinoma injected subcutaneously in the flank of the mouse. PZ‐128 and Avastin significantly inhibited tumor growth (p < 0.01) with 75 and 67% inhibition, respectively (Table 3) [343]. Moreover the role of MMP‐1 in NSCLC is not well understood. To address the role of mouse homolog MMP‐1a, knockout mice were generated (MMP1a–KO) [344]. Lewis lung carcinoma (LLC1) is a highly tumorigenic cell line that expresses high levels of PAR1. Mouse LLC1 cells were implanted subcutaneously into the abdominal fat pad of MMP1a–KO and WT C57CL/6 mice. The progression of tumor growth was monitored over 26 days. The loss of stromal MMP1a resulted in a 50% reduction of tumor growth when compared with WT‐MMP1a mice (p < 0.001). There was a significant decrease (30%) (p < 0.01) in angiogenesis in the Mmp1a–KO mice when compared with WT mice.

TABLE 3.

Summary of preclinical studies carried out with PZ‐128 and the MMP1 Inhibitor in breast, lung, and ovarian cancer.

Cell Tumor model Type of assay Outcome References
MCF‐PAR1 Mammary fat pad NCR nu/nu mice Xenograft model (tumor size) >PAR1‐tumor incidence 100% PAR1‐null 0% after 6 weeks [333]
>PZ‐128 (10 mg/kg) inhibits 62% MMP‐1 Inh (5 mg/kg) inhibits 82% of tumor growth
MDA‐MB‐231 >Dual treatment with PZ‐128/taxotere (docetaxel) treatment day 2 98% inhibition [333]
Xenograft model (tumor size) TUNEL staining >Delayed dual treatment day 15 60% inhibition of tumor growth, 60% apoptotic area [335]
Western blot >pAkt as biomarker inhibited 54% by PZ‐128 after 5‐day treatment MMP‐1 Inh 5‐day treatment 61% Inhibition [335]
MDA‐MB‐231/GFP Metastasis to lung NCR nu/nu mice, tail vein Lung histology >Reduction in metastatic incidence by PZ‐128 75% MMP‐1 Inh 88% inhibition [335]
>Reduction in metastatic incidence by PZ‐128 75% MMP‐1 Inh 88% inhibition [335]
MCF7‐PAR1 (clone N55) >PAR1‐tumor incidence 100% [340]
MDA‐MB‐231 MDA‐MB‐231 tumor incidence 100% [340]
MCF7‐PAR1 (R310E) R310E tumor incidence 0% [340]
A549 NCR nu/nu mice Subcutaneous Xenograft model tumor size >75% Inhibition of tumor growth, PZ‐128 monotherapy (10 mg/kg) compared with Avastin (5 mg/kg) 67% inhibition of tumor growth [343]
OVCAR‐4 Intraperitoneal cavity Endothelial barrier >PZ‐128 (10 mg/kg i.p. every other day) reduced ascites formation by 60% [345]
SKOV‐3 Reduced ascites formation by 60% [345]
OVCAR‐4 Peritoneal carcinomatosis Histology >84–96% inhibition of angiogenesis monotherapy with MMP1 Inh and PZ‐128 [345]
>Dual treatment with PZ‐128 or MMP‐1 Inh/taxotere (docetaxel) inhibition of metastatic progression through the diaphragm and thoracic cavity [345]

Abbreviations: OVCAR: ovarian cancer cell line 4, SKOV‐3: serous ovarian adenocarcinoma cell line, MCF7: Michigan Cancer Foundation‐7, MDA‐MB‐231: M.D. Anderson—Metastatic Breast Cancer 231.

Ovarian cancer is still one of the deadliest gynecologic malignancies because of early metastatic spread into the abdominal cavity. Despite some advances, a taxane such as docetaxel remains the standard of care in combination with a platinum compound (cisplatin or carboplatin). A murine metastatic model of intraperitoneal (i.p.) ovarian cancer was conducted by injecting extremely aggressive and metastatic OVCAR‐4 cells into the i.p. cavity. All untreated (vehicle‐treated) mice had full‐blown metastasis through the diaphragm and into the lungs and mediastinum (Table 3) [345]. In contrast, dual therapy with PZ‐128 prevented invasion across the diaphragm into the thoracic cavity (p = 0.01) compared with docetaxel alone. Similar results were observed with MMP‐1 Inh (p = 0.003). These data strongly support an MMP1–PAR1 signaling pathway as an essential target for preventing the metastatic spread in ovarian cancer. Ovarian peritoneal carcinomas produce large volumes of ascitic fluid. Using OVCAR‐4 and SKOV‐3 peritoneal carcinoma models, there was significant inhibition (60%, p = 0.0017) of ascitic fluid accumulation using PZ‐128 as monotherapy (10 mg/kg every second day) when compared with vehicle control in an OVCAR‐4 peritoneal carcinoma model. Similar results were seen with SKOV‐3 cells. This effect on ascites accumulation may be a direct consequence of an MMP1–PAR1‐dependent alteration in endothelial barrier function. Taken together, PZ‐128 is an excellent therapeutic for ovarian cancer treatment [345].

In addition to these preclinical advances, several GPCR‐targeted agents have progressed into clinical trials across various cancer types, reflecting their translational potential. Notably, zibotentan and atrasentan, both small‐molecule endothelin A receptor antagonists, are being assessed in prostate cancer across multiple clinical trial phases [305]. In pancreatic and metastatic breast cancer, the Fz receptor‐targeting antibody vantictumab (OMP‐18R5) has shown potential, especially in combination regimens with chemotherapy. Similarly, G17DT, an immunogen targeting the cholecystokinin‐2 receptor, has progressed to phase III trials for pancreatic cancer [346]. In the context of head and neck cancer, vismodegib (GDC‐0449), a SMO receptor inhibitor, is under phase II investigation [347, 348]. Other notable agents include CCX872, a CCR2 antagonist for pancreatic cancer, and mogamulizumab (KW‐0761), an anti‐CCR4 mAb, which is in phase II trials for adult T‐cell leukemia/lymphoma [349]. Several trials also explore the efficacy of beta‐blockers targeting beta‐adrenergic receptors in breast and ovarian cancers, reflecting the utility of drug repurposing [305]. Collectively, these ongoing clinical efforts underscore the expanding landscape of GPCR‐targeted cancer therapeutics and their promise in enhancing precision oncology (Table 4).

TABLE 4.

Anti‐GPCRs drugs and antibodies under clinical trials.

Cancer Inhibitor Type of molecule receptor Receptor Phase References
PCa

Zibotentan (ZD4054)

Atrasentan (ABT‐627)

Small molecule Endothelin A receptor I, II, III [350, 351]
HNSCC GDC‐0449 (vismodegib) Small molecule

Smoothened receptor

(SMO)

II [347, 348]
OVC

GDC‐044 (vismodegib)

Propranolol (β‐blockers)

Small molecule

Endothelin receptor

β‐Adrenergic receptor

II, I [352]
PC

CCX872

(OMP‐18R5)

Vantictumab

G17DT

Small molecule

Antibodies

Immunogen

CCR2

Frizzled receptor

FZ7

Cholecystokinin‐2

receptor

I, III [346]
ATCL&L

KW‐0761

(mogamulizumab)

Antibodies CCR4 II [349]

Abbreviations: PCa: prostate cancer, HNSCC: head and neck cancer, OVC: ovarian cancer, PC: pancreatic cancer, ATCL&L: adult T‐cell leukemia and lymphoma

In summary, preclinical studies have demonstrated that biased GPCR signaling has a significant influence on tumor growth, metastasis, and drug response. Animal models using biased agonists and receptor mutants have provided compelling evidence for the therapeutic advantages of selectively modulating GPCR pathways. Supporting this, multiple GPCR‐targeting drugs are currently in clinical trials for cancers such as prostate, pancreatic, breast, and head and neck. These include endothelin receptor antagonists (zibotentan, atrasentan), Fz and SMO inhibitors (vantictumab, vismodegib), the immunogen G17DT, and repurposed beta‐blockers. These studies highlight the potential of targeting specific GPCR‐mediated signals to enhance efficacy and minimize side effects, thereby paving the way for the translation of biased GPCR modulation into clinical oncology.

10. Concluding Remarks and Future Perspectives

GPCRs play a crucial role in cellular communication by initiating multiple signaling pathways that regulate essential functions, including migration, differentiation, growth, and survival. By increasing cell proliferation, encouraging angiogenesis, facilitating invasion and metastasis, and supporting immune evasion, GPCRs aid in the development of cancerous tumors. The understanding of GPCR signaling has evolved recently from a straightforward binary paradigm to a more complex system, in which different GPCR conformations can activate distinct signaling pathways—a process referred to as biased signaling. This advanced understanding creates opportunities for creating cancer treatments that are more efficient and focused. Utilizing the immunomodulatory effects of GPCRs or altering tumor‐immune interactions could transform current immunotherapies to achieve durable clinical responses and prevent tumor recurrence. Research on these combinations is still ongoing, either as a way to treat cancer or as a preventative measure. As our understanding of the complex mechanisms underlying the genesis and spread of cancer advances, GPCRs will play an increasingly significant role as drivers and targets for innovative cancer treatments.

Recent research has clarified the intricate mechanisms behind GPCR‐mediated biased signaling. G protein‐dependent or β‐arrestin‐dependent pathways can be selectively activated by ligand interaction, thereby stabilizing particular GPCR conformations. While it affects multiple stages like tumor invasion, extravasation, and metastatic spread, this selective activation is essential to the development of cancer. To regulate cellular responses, for example, β‐arrestins play a crucial role in coordinating the timing, intensity, and spatial localization of GPCR‐mediated signaling. Furthermore, to predict ligand bias and comprehend the structural dynamics of GPCRs, computational methods such as machine learning models and molecular dynamics simulations have been used. The discovery of biased ligands has been made easier by experimental methods such as mass spectrometry, fluorescence resonance energy transfer, and 19F NMR, which have shed light on the conformational states of GPCRs. Such advances underscore the importance of combining experimental and computational approaches to fully comprehend the complexities of GPCR signaling in cancer.

To minimize side effects and increase therapeutic efficacy, biased ligands that can selectively regulate specific signaling pathways are essential for the future of GPCR‐targeted cancer therapy. Gaining a better knowledge of the structural and conformational dynamics of GPCRs is crucial to achieving this. High‐resolution insights into GPCR structures can be obtained through advancements in structural biology, such as time‐resolved X‐ray crystallography and cryo‐electron microscopy. Additionally, the use of generative AI models, such as GPCR‐BERT, shows potential for creating ligands with desirable bias profiles and predicting receptor conformations. To evaluate the effectiveness and safety of biased ligands, credible biomarkers and patient‐derived models must be developed, as there are still difficulties in transforming these findings into practical applications. Overcoming these obstacles and realizing the potential of biased signaling in cancer treatment will require ongoing interdisciplinary research that integrates structural biology, computational modeling, and pharmacology.

Author Contributions

I.R.K., S.K., M.A.M., and A.A.B. wrote the manuscript and generated figures. M.A.M. and A.A.B. contributed to the concept and design and critically edited the manuscript. S.A., R.K., A.A.A.S., A.A.B., and M.A.M. critically revised and edited the scientific content. All authors read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Ethics Statement

The authors have nothing to report.

Acknowledgments

Figures were created using BioRender. The Qatar National Library provides Open Access funding.

Khan I. R., Khurshid S., Almawash S., et al. “G Protein‐Coupled Receptor Signaling: Implications and Therapeutic Development Advances in Cancers.” MedComm 6, no. 10 (2025): e70375. 10.1002/mco2.70375

Funding: Sidra Medicine Precision Program provides research funding to Ajaz A. Bhat (SDR400190) and Ammira S. Al‐Shabeeb Akil (SDR400191). This study was also supported by the Ramalingaswami Re‐entry Fellowship (Grant number: D.O. NO.BT/HRD/35/02/2006) from the Department of Biotechnology, Government of India, New Delhi; Core Research Grant (CRG/2021/003805) from Anusandhan National Research Foundation (ANRF), formally called Science and Engineering Research Board (SERB), Government of India, New Delhi; Research Grant (ID No. 2022‐16465) from Indian Council of Medical Research (ICMR) Government of India; Central Council for Research in Unani Medicine (CCRUM), Ministry of Ayurveda, Yoga & Naturopathy, Unani, Siddha, and Homoeopathy (AYUSH) (TECH‐21013/20/2024‐CCRUM‐HQ), Government of India; and J&K Science Technology and Innovation Council, Department of Science and Technology (DST) (JKST&IC/SRE/936‐40), Government of J&K to Muzafar A. Macha. Promotion of University Research and Scientific Excellence (PURSE) (SR/PURSE/2022/121) grant from the Department of Science and Technology (DST), Government of India, New Delhi to the Islamic University of Science and Technology (IUST), Awantipora.

Inamu Rashid Khan and Sana Khurshid equaly contributed to first authorship.

Contributor Information

Ajaz A. Bhat, Email: abhat@sidra.org.

Muzafar A. Macha, Email: muzafar.macha@iust.ac.in, Email: muzafar.aiiims@gmail.com.

Data Availability Statement

The authors have nothing to report.

References

  • 1. Insel P. A., Sriram K., Gorr M. W., et al., “GPCRomics: An Approach to Discover GPCR Drug Targets,” Trends in Pharmacological Sciences 40, no. 6 (2019): 378–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Calebiro D., Koszegi Z., Lanoiselée Y., Miljus T., and O'Brien S., “G Protein‐coupled Receptor‐G Protein Interactions: A Single‐molecule Perspective,” Physiological Reviews 101, no. 3 (2021): 857–906. [DOI] [PubMed] [Google Scholar]
  • 3. Zhang Y., Devries M. E., and Skolnick J., “Structure Modeling of all Identified G Protein‐coupled Receptors in the human Genome,” PLoS Computational Biology 2, no. 2 (2006): e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Rosenbaum D. M., Rasmussen S. G., and Kobilka B. K., “The Structure and Function of G‐protein‐coupled Receptors,” Nature 459, no. 7245 (2009): 356–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Fredriksson R., Lagerström M. C., Lundin L. G., and Schiöth H. B., “The G‐protein‐coupled Receptors in the human Genome Form Five Main Families. Phylogenetic Analysis, Paralogon Groups, and Fingerprints,” Molecular Pharmacology 63, no. 6 (2003): 1256–1272. [DOI] [PubMed] [Google Scholar]
  • 6. Shimada I., Ueda T., Kofuku Y., Eddy M. T., and Wüthrich K., “GPCR Drug Discovery: Integrating Solution NMR Data With Crystal and Cryo‐EM Structures,” Nature Reviews Drug Discovery 18, no. 1 (2019): 59–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Hauser A. S., Kooistra A. J., Munk C., et al., “GPCR Activation Mechanisms Across Classes and Macro/Microscales,” Nature Structural & Molecular Biology 28, no. 11 (2021): 879–888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Pupo A. S., Duarte D. A., Lima V., Teixeira L. B., Parreiras E. S. L. T., and Costa‐Neto C. M., “Recent Updates on GPCR Biased Agonism,” Pharmacological Research 112 (2016): 49–57. [DOI] [PubMed] [Google Scholar]
  • 9. Shenoy S. K., Drake M. T., Nelson C. D., et al., “beta‐arrestin‐dependent, G Protein‐independent ERK1/2 Activation by the beta2 Adrenergic Receptor,” The Journal of Biological Chemistry 281, no. 2 (2006): 1261–1273. [DOI] [PubMed] [Google Scholar]
  • 10. DeWire S. M., Ahn S., Lefkowitz R. J., and Shenoy S. K., “Beta‐arrestins and Cell Signaling,” Annual Review of Physiology 69 (2007): 483–510. [DOI] [PubMed] [Google Scholar]
  • 11. Srivastava A., Gupta B., Gupta C., and Shukla A. K., “Emerging Functional Divergence of β‐Arrestin Isoforms in GPCR Function,” Trends in Endocrinology and Metabolism: TEM 26, no. 11 (2015): 628–642. [DOI] [PubMed] [Google Scholar]
  • 12. Gether U., “Uncovering Molecular Mechanisms Involved in Activation of G Protein‐coupled Receptors,” Endocrine Reviews 21, no. 1 (2000): 90–113. [DOI] [PubMed] [Google Scholar]
  • 13. Lappano R. and Maggiolini M., “G Protein‐coupled Receptors: Novel Targets for Drug Discovery in Cancer,” Nature Reviews Drug Discovery 10, no. 1 (2011): 47–60. [DOI] [PubMed] [Google Scholar]
  • 14. Young D., Waitches G., Birchmeier C., Fasano O., and Wigler M., “Isolation and Characterization of a New Cellular Oncogene Encoding a Protein With Multiple Potential Transmembrane Domains,” Cell 45, no. 5 (1986): 711–719. [DOI] [PubMed] [Google Scholar]
  • 15. Li S., Huang S., and Peng S. B., “Overexpression of G Protein‐coupled Receptors in Cancer Cells: Involvement in Tumor Progression,” International Journal of Oncology 27, no. 5 (2005): 1329–1339. [PubMed] [Google Scholar]
  • 16. Pierce K. L., Premont R. T., and Lefkowitz R. J., “Seven‐transmembrane Receptors,” Nature Reviews Molecular Cell Biology 3, no. 9 (2002): 639–650. [DOI] [PubMed] [Google Scholar]
  • 17. Wootten D., Christopoulos A., and Sexton P. M., “Emerging Paradigms in GPCR Allostery: Implications for Drug Discovery,” Nature Reviews Drug Discovery 12, no. 8 (2013): 630–644. [DOI] [PubMed] [Google Scholar]
  • 18. Christopoulos A., “Advances in G Protein‐coupled Receptor Allostery: From Function to Structure,” Molecular Pharmacology 86, no. 5 (2014): 463–478. [DOI] [PubMed] [Google Scholar]
  • 19. Yang D., Zhou Q., Labroska V., et al., “G Protein‐coupled Receptors: Structure‐ and Function‐based Drug Discovery,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Casadó V. and Casadó‐Anguera V., “What Are the Current Trends in G Protein‐coupled Receptor Targeted Drug Discovery?,” Expert Opinion on Drug Discovery 18, no. 8 (2023): 815–820. [DOI] [PubMed] [Google Scholar]
  • 21. Hauser A. S., Attwood M. M., Rask‐Andersen M., Schiöth H. B., and Gloriam D. E., “Trends in GPCR Drug Discovery: New Agents, Targets and Indications,” Nature Reviews Drug Discovery 16, no. 12 (2017): 829–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Smith J. S., Lefkowitz R. J., and Rajagopal S., “Biased Signalling: From Simple Switches to Allosteric Microprocessors,” Nature Reviews Drug Discovery 17, no. 4 (2018): 243–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Tan L., Yan W., McCorvy J. D., and Cheng J., “Biased Ligands of G Protein‐Coupled Receptors (GPCRs): Structure‐Functional Selectivity Relationships (SFSRs) and Therapeutic Potential,” Journal of Medicinal Chemistry 61, no. 22 (2018): 9841–9878. [DOI] [PubMed] [Google Scholar]
  • 24. Kenakin T., “Biased Receptor Signaling in Drug Discovery,” Pharmacological Reviews 71, no. 2 (2019): 267–315. [DOI] [PubMed] [Google Scholar]
  • 25. Kieffer B. L., “Opioids: First Lessons From Knockout Mice,” Trends in Pharmacological Sciences 20, no. 1 (1999): 19–26. [DOI] [PubMed] [Google Scholar]
  • 26. Violin J. D., Crombie A. L., Soergel D. G., and Lark M. W., “Biased Ligands at G‐protein‐coupled Receptors: Promise and Progress,” Trends in Pharmacological Sciences 35, no. 7 (2014): 308–316. [DOI] [PubMed] [Google Scholar]
  • 27. Urs N. M., Peterson S. M., and Caron M. G., “New Concepts in Dopamine D(2) Receptor Biased Signaling and Implications for Schizophrenia Therapy,” Biological Psychiatry 81, no. 1 (2017): 78–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Békés M., Langley D. R., and Crews C. M., “PROTAC Targeted Protein Degraders: The Past Is Prologue,” Nature Reviews Drug Discovery 21, no. 3 (2022): 181–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Congreve M., de Graaf C., Swain N. A., and Tate C. G., “Impact of GPCR Structures on Drug Discovery,” Cell 181, no. 1 (2020): 81–91. [DOI] [PubMed] [Google Scholar]
  • 30. Hutchings C. J., Koglin M., Olson W. C., and Marshall F. H., “Opportunities for Therapeutic Antibodies Directed at G‐protein‐coupled Receptors,” Nature Reviews Drug Discovery 16, no. 9 (2017): 787–810. [DOI] [PubMed] [Google Scholar]
  • 31. Hutchings C. J., “Mini‐review: Antibody Therapeutics Targeting G Protein‐coupled Receptors and Ion Channels,” Antibody Therapeutics 3, no. 4 (2020): 257–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Hutchings C. J., “A Review of Antibody‐based Therapeutics Targeting G Protein‐coupled Receptors: An Update,” Expert Opinion on Biological Therapy 20, no. 8 (2020): 925–935. [DOI] [PubMed] [Google Scholar]
  • 33. Takahashi M., “Nucleic Acid Aptamers Emerging as Modulators of G‐Protein‐Coupled Receptors: Challenge to Difficult Cell Surface Proteins,” Cells 11, no. 11 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Yang E. Y. and Shah K., “Nanobodies: Next Generation of Cancer Diagnostics and Therapeutics,” Frontiers in Oncology 10 (2020): 1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Szwabowski G. L., Baker D. L., and Parrill A. L., “Application of Computational Methods for Class A GPCR Ligand Discovery,” Journal of Molecular Graphics & Modelling 121 (2023): 108434. [DOI] [PubMed] [Google Scholar]
  • 36. Unal H. and Karnik S. S., “Domain Coupling in GPCRs: The Engine for Induced Conformational Changes,” Trends in Pharmacological Sciences 33, no. 2 (2012): 79–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Duan J., Xu P., Zhang H., et al., “Mechanism of Hormone and Allosteric Agonist Mediated Activation of Follicle Stimulating Hormone Receptor,” Nature Communications 14, no. 1 (2023): 519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Duan J., Xu P., Cheng X., et al., “Structures of Full‐length Glycoprotein Hormone Receptor Signalling Complexes,” Nature 598, no. 7882 (2021): 688–692. [DOI] [PubMed] [Google Scholar]
  • 39. Faust B., Billesbølle C. B., Suomivuori C. M., et al., “Autoantibody Mimicry of Hormone Action at the Thyrotropin Receptor,” Nature 609, no. 7928 (2022): 846–853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Billesbølle C. B., de March C. A., van der Velden W. J. C., et al., “Structural Basis of Odorant Recognition by a human Odorant Receptor,” Nature 615, no. 7953 (2023): 742–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Martin A. L., Steurer M. A., and Aronstam R. S., “Constitutive Activity Among Orphan Class‐A G Protein Coupled Receptors,” PLoS ONE 10, no. 9 (2015): e0138463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Lin X., Chen B., Wu Y., et al., “Cryo‐EM Structures of Orphan GPR21 Signaling Complexes,” Nature Communications 14, no. 1 (2023): 216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Ye F., Wong T. S., Chen G., et al., “Cryo‐EM Structure of G‐protein‐coupled Receptor GPR17 in Complex With Inhibitory G Protein,” MedComm 3, no. 4 (2022): e159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Zhang Q., Madden N. E., Wong A. S. T., Chow B. K. C., and Lee L. T. O., “The Role of Endocrine G Protein‐Coupled Receptors in Ovarian Cancer Progression,” Frontiers in Endocrinology 8 (2017): 66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Banerjee A. and Ray S., “Molecular Interactions and Mutational Impact Upon Rhodopsin (G90→D90) for Hindering Dark Adaptation of Eye: A Comparative Structural Level Outlook for Signaling Mechanism in Night Blindness,” Mutation Research 814 (2019): 7–14. [DOI] [PubMed] [Google Scholar]
  • 46. Cary B. P., Zhang X., Cao J., et al., “New Insights Into the Structure and Function of Class B1 GPCRs,” Endocrine Reviews 44, no. 3 (2023): 492–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Cong Z., Chen L. N., Ma H., et al., “Molecular Insights Into Ago‐allosteric Modulation of the human Glucagon‐Like Peptide‐1 Receptor,” Nature Communications 12, no. 1 (2021): 3763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Wootten D., Simms J., Miller L. J., Christopoulos A., and Sexton P. M., “Polar Transmembrane Interactions Drive Formation of Ligand‐specific and Signal Pathway‐biased family B G Protein‐coupled Receptor Conformations,” Proceedings of the National Academy of Sciences of the United States of America 110, no. 13 (2013): 5211–5216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Cong Z., Liang Y. L., Zhou Q., et al., “Structural Perspective of Class B1 GPCR Signaling,” Trends in Pharmacological Sciences 3, no. 4 (2022): 321–334. [DOI] [PubMed] [Google Scholar]
  • 50. Zhang Y., Sun B., and Feng D., “Cryo‐EM Structure of the Activated GLP‐1 Receptor in Complex With a G Protein,” Nature 546, no. 7657 (2017): 248–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Sun W., Chen L. N., and Zhou Q., “A Unique Hormonal Recognition Feature of the human Glucagon‐Like Peptide‐2 Receptor,” Cell Research 30, no. 12 (2020): 1098–1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Liang Y. L., Khoshouei M., and Deganutti G., “Cryo‐EM Structure of the Active, G(s)‐protein Complexed, human CGRP Receptor,” Nature 561, no. 7724 (2018): 492–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Langenhan T., Piao X., and Monk K. R., “Adhesion G Protein‐coupled Receptors in Nervous System Development and Disease,” Nature Reviews Neuroscience 17, no. 9 (2016): 550–561. [DOI] [PubMed] [Google Scholar]
  • 54. Ellaithy A., Gonzalez‐Maeso J., Logothetis D. A., and Levitz J., “Structural and Biophysical Mechanisms of Class C G Protein‐Coupled Receptor Function,” Trends in Biochemical Sciences 45, no. 12 (2020): 1049–1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Lei I., Tian S., Gao W., et al., “Acetyl‐CoA Production by Specific Metabolites Promotes Cardiac Repair After Myocardial Infarction via Histone Acetylation,” Elife 10, (2021):. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Vezzoli G., Terranegra A., Rainone F., et al., “Calcium‐sensing Receptor and Calcium Kidney Stones,” Journal of Translational Medicine 9 (2011): 201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Shen C., Mao C., Xu C., et al., “Structural Basis of GABA(B) Receptor‐G(i) Protein Coupling,” Nature 594, no. 7864 (2021): 594–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Park J., Fu Z., Frangaj A., et al., “Structure of human GABA(B) Receptor in an Inactive state,” Nature 584, no. 7820 (2020): 304–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Felice D., Cryan J. F., and O'Leary O. F., “GABA(B) Receptors: Anxiety and Mood Disorders,” Current Topics in Behavioral Neurosciences 52 (2022): 241–265. [DOI] [PubMed] [Google Scholar]
  • 60. Ahmad R. and Dalziel J. E., “G Protein‐Coupled Receptors in Taste Physiology and Pharmacology,” Frontiers in Pharmacology 11 (2020): 587664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Abd‐Elrahman K. S., Sarasija S., and Ferguson S. S. G., “The Role of Neuroglial Metabotropic Glutamate Receptors in Alzheimer's Disease,” Current Neuropharmacology 21, no. 2 (2023): 273–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Zhang X., Dong S., and Xu F., “Structural and Druggability Landscape of Frizzled G Protein‐Coupled Receptors,” Trends in Biochemical Sciences 43, no. 12 (2018): 1033–1046. [DOI] [PubMed] [Google Scholar]
  • 63. Gurney A., Axelrod F., Bond C. J., et al., “Wnt Pathway Inhibition via the Targeting of Frizzled Receptors Results in Decreased Growth and Tumorigenicity of human Tumors,” Proceedings of the National Academy of Sciences of the United States of America 109, no. 29 (2012): 11717–11722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Anastas J. N. and Moon R. T., “WNT Signalling Pathways as Therapeutic Targets in Cancer,” Nature Reviews Cancer 13, no. 1 (2013): 11–26. [DOI] [PubMed] [Google Scholar]
  • 65. Nusse R. and Clevers H., “Wnt/β‐Catenin Signaling, Disease, and Emerging Therapeutic Modalities,” Cell 169, no. 6 (2017): 985–999. [DOI] [PubMed] [Google Scholar]
  • 66. Ueno K., Hazama S., Mitomori S., et al., “Down‐regulation of Frizzled‐7 Expression Decreases Survival, Invasion and Metastatic Capabilities of Colon Cancer Cells,” British Journal of Cancer 101, no. 8 (2009): 1374–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Wang Y., Zhao G., Condello S., et al., “Frizzled‐7 Identifies Platinum‐Tolerant Ovarian Cancer Cells Susceptible to Ferroptosis,” Cancer Research 81, no. 2 (2021): 384–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Turku A., Schihada H., Kozielewicz P., Bowin C. F., and Schulte G., “Residue 6.43 Defines Receptor Function in Class F GPCRs,” Nature Communications 12, no. 1 (2021): 3919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Briscoe J. and Thérond P. P., “The Mechanisms of Hedgehog Signalling and Its Roles in Development and Disease,” Nature Reviews Molecular Cell Biology 14, no. 7 (2013): 416–429. [DOI] [PubMed] [Google Scholar]
  • 70. Kozielewicz P., Turku A., and Schulte G., “Molecular Pharmacology of Class F Receptor Activation,” Molecular Pharmacology 97, no. 2 (2020): 62–71. [DOI] [PubMed] [Google Scholar]
  • 71. Shao Z., Yan W., Chapman K., et al., “Structure of an Allosteric Modulator Bound to the CB1 Cannabinoid Receptor,” Nature Chemical Biology 15, no. 12 (2019): 1199–1205. [DOI] [PubMed] [Google Scholar]
  • 72. Jiang H., Galtes D., Wang J., and Rockman H. A., “G Protein‐coupled Receptor Signaling: Transducers and Effectors,” American Journal of Physiology Cell Physiology 323, no. 3 (2022): C731–c748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Tilley D. G., “G Protein‐dependent and G Protein‐independent Signaling Pathways and Their Impact on Cardiac Function,” Circulation Research 109, no. 2 (2011): 217–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Cheng H., Lederer W. J., and Cannell M. B., “Calcium Sparks: Elementary Events Underlying Excitation‐contraction Coupling in Heart Muscle,” Science (New York, NY) 262, no. 5134 (1993): 740–744. [DOI] [PubMed] [Google Scholar]
  • 75. Nakayama H., Bodi I., Maillet M., et al., “The IP3 Receptor Regulates Cardiac Hypertrophy in Response to Select Stimuli,” Circulation Research 107, no. 5 (2010): 659–666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Wu X., Zhang T., Bossuyt J., et al., “Local InsP3‐dependent Perinuclear Ca2+ Signaling in Cardiac Myocyte Excitation‐transcription Coupling,” The Journal of Clinical Investigation 116, no. 3 (2006): 675–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Braz J. C., Gregory K., Pathak A., et al., “PKC‐alpha Regulates Cardiac Contractility and Propensity Toward Heart Failure,” Nature Medicine 10, no. 3 (2004): 248–254. [DOI] [PubMed] [Google Scholar]
  • 78. Rockman H. A., Koch W. J., and Lefkowitz R. J., “Seven‐transmembrane‐spanning Receptors and Heart Function,” Nature 415, no. 6868 (2002): 206–212. [DOI] [PubMed] [Google Scholar]
  • 79. Zhong H. and Neubig R. R., “Regulator of G Protein Signaling Proteins: Novel Multifunctional Drug Targets,” The Journal of Pharmacology and Experimental Therapeutics 297, no. 3 (2001): 837–845. [PubMed] [Google Scholar]
  • 80. Yang L. K., Hou Z. S., and Tao Y. X., “Biased Signaling in Naturally Occurring Mutations of G Protein‐coupled Receptors Associated With Diverse human Diseases,” Biochimica Et Biophysica Acta Molecular Basis of Disease 1867, no. 1 (2021): 165973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Borah M. P., Trakroo D., Soni N., Kumari P., and Baidya M., “Exploring Bias in GPCR Signaling and Its Implication in Drug Development: A One‐Sided Affair,” Biochemistry 64, no. 1 (2025): 1–14. [DOI] [PubMed] [Google Scholar]
  • 82. Gurevich V. V. and Gurevich E. V., “Biased GPCR Signaling: Possible Mechanisms and Inherent Limitations,” Pharmacology & Therapeutics 211 (2020): 107540. [DOI] [PubMed] [Google Scholar]
  • 83. Lefkowitz R. J., Rajagopal K., and Whalen E. J., “New Roles for Beta‐arrestins in Cell Signaling: Not Just for Seven‐transmembrane Receptors,” Molecular Cell 24, no. 5 (2006): 643–652. [DOI] [PubMed] [Google Scholar]
  • 84. Jean‐Charles P. Y., Kaur S., and Shenoy S. K., “G Protein‐Coupled Receptor Signaling through β‐Arrestin‐Dependent Mechanisms,” Journal of Cardiovascular Pharmacology 70, no. 3 (2017): 142–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Liu J. J., Horst R., Katritch V., Stevens R. C., and Wüthrich K., “Biased Signaling Pathways in β2‐adrenergic Receptor Characterized by 19F‐NMR,” Science (New York, NY) 335, no. 6072 (2012): 1106–1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Woo A. Y., Jozwiak K., Toll L., et al., “Tyrosine 308 Is Necessary for Ligand‐directed Gs Protein‐biased Signaling of β2‐adrenoceptor,” The Journal of Biological Chemistry 289, no. 28 (2014): 19351–19363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Wang P., Wu Y., Ge X., Ma L., and Pei G., “Subcellular Localization of Beta‐arrestins Is Determined by Their Intact N Domain and the Nuclear Export Signal at the C Terminus,” The Journal of Biological Chemistry 278, no. 13 (2003): 11648–11653. [DOI] [PubMed] [Google Scholar]
  • 88. Shukla A. K., Westfield G. H., Xiao K., et al., “Visualization of Arrestin Recruitment by a G‐protein‐coupled Receptor,” Nature 512, no. 7513 (2014): 218–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Maharana J., Sano F. K., Sarma P., et al., “Molecular Insights Into atypical Modes of β‐arrestin Interaction With Seven Transmembrane Receptors,” Science (New York, NY) 383, no. 6678 (2024): 101–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Bagnato A. and Rosanò L., “New Routes in GPCR/β‐Arrestin‐Driven Signaling in Cancer Progression and Metastasis,” Frontiers in Pharmacology 10 (2019): 114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Song Q., Ji Q., and Li Q., “The Role and Mechanism of Βarrestins in Cancer Invasion and Metastasis (Review),” International Journal of Molecular Medicine 41, no. 2 (2018): 631–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Zoudilova M., Kumar P., Ge L., Wang P., Bokoch G. M., and DeFea K. A., “β‐Arrestin‐dependent Regulation of the Cofilin Pathway Downstream of Protease‐activated Receptor‐2*,” Journal of Biological Chemistry 282 (2007): 20634–20646.. [DOI] [PubMed] [Google Scholar]
  • 93. Min J. and Defea K., “β‐arrestin‐dependent Actin Reorganization: Bringing the Right Players Together at the Leading Edge,” Molecular Pharmacology 80, no. 5 (2011): 760–768. [DOI] [PubMed] [Google Scholar]
  • 94. Ma X., Espana‐Serrano L., Kim W.‐J., Thayele Purayil H., Nie Z., and Daaka Y., “βArrestin1 Regulates the Guanine Nucleotide Exchange Factor RasGRF2 Expression and the Small GTPase Rac‐mediated Formation of Membrane Protrusion and Cell Motility *,” Journal of Biological Chemistry 289, no. 19 (2014): 13638–13650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Kang J., Shi Y., Xiang B., et al., “A Nuclear Function of β‐Arrestin1 in GPCR Signaling: Regulation of Histone Acetylation and Gene Transcription,” Cell 123, no. 5 (2005): 833–847. [DOI] [PubMed] [Google Scholar]
  • 96. Shi Y., Feng Y., Kang J., et al., “Critical Regulation of CD4+ T Cell Survival and Autoimmunity by Beta‐arrestin 1,” Nature Immunology 8, no. 8 (2007): 817–824. [DOI] [PubMed] [Google Scholar]
  • 97. Yang Y., Guo Y., Tan S., et al., “Β‐Arrestin1 Enhances Hepatocellular Carcinogenesis Through Inflammation‐mediated Akt Signalling,” Nature Communications 6 (2015): 7369. [DOI] [PubMed] [Google Scholar]
  • 98. Czogalla B., Partenheimer A., and Jeschke U., “β‐arrestin 2 Is a Prognostic Factor for Survival of Ovarian Cancer Patients Upregulating Cell Proliferation,” Frontiers in Endocrinology 11 (2020): 554733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Bostanabad S. Y., Noyan S., Dedeoglu B. G., and Gurdal H., “Overexpression of β‐Arrestins Inhibits Proliferation and Motility in Triple Negative Breast Cancer Cells,” Scientific Reports 11, no. 1 (2021): 1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Duan X., Kong Z., Liu Y., et al., “β‐Arrestin2 Contributes to Cell Viability and Proliferation via the Down‐Regulation of FOXO1 in Castration‐Resistant Prostate Cancer,” Journal of Cellular Physiology 230, no. 10 (2015): 2371–2381. [DOI] [PubMed] [Google Scholar]
  • 101. Lakshmikanthan V., Zou L., Kim J. I., et al., “Identification of betaArrestin2 as a Corepressor of Androgen Receptor Signaling in Prostate Cancer,” Proceedings of the National Academy of Sciences of the United States of America 106, no. 23 (2009): 9379–9384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Sun Y., Cheng Z., Ma L., and Pei G., “β‐Arrestin2 Is Critically Involved in CXCR4‐mediated Chemotaxis, and This Is Mediated by Its Enhancement of p38 MAPK Activation*,” Journal of Biological Chemistry 277, no. 51 (2002): 49212–49219. [DOI] [PubMed] [Google Scholar]
  • 103. Ge L., Shenoy S. K., Lefkowitz R. J., and DeFea K., “Constitutive Protease‐activated Receptor‐2‐mediated Migration of MDA MB‐231 Breast Cancer Cells Requires both Beta‐arrestin‐1 and ‐2,” The Journal of Biological Chemistry 279, no. 53 (2004): 55419–55424. [DOI] [PubMed] [Google Scholar]
  • 104. Gol S., Pena R. N., Rothschild M. F., Tor M., and Estany J., “A Polymorphism in the Fatty Acid Desaturase‐2 Gene Is Associated With the Arachidonic Acid Metabolism in Pigs,” Scientific Reports 8, no. 1 (2018): 14336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Dorsam R. T. and Gutkind J. S., “G‐protein‐coupled Receptors and Cancer,” Nature Reviews Cancer 7, no. 2 (2007): 79–94. [DOI] [PubMed] [Google Scholar]
  • 106. van Biesen T., Luttrell L. M., Hawes B. E., and Lefkowitz R. J., “Mitogenic Signaling via G Protein‐coupled Receptors,” Endocrine Reviews 17, no. 6 (1996): 698–714. [DOI] [PubMed] [Google Scholar]
  • 107. Prevarskaya N., Skryma R., and Shuba Y., “Calcium in Tumour Metastasis: New Roles for Known Actors,” Nature Reviews Cancer 11, no. 8 (2011): 609–618. [DOI] [PubMed] [Google Scholar]
  • 108. Griner E. M. and Kazanietz M. G., “Protein Kinase C and Other Diacylglycerol Effectors in Cancer,” Nature Reviews Cancer 7, no. 4 (2007): 281–294. [DOI] [PubMed] [Google Scholar]
  • 109. Rozengurt E., “Mitogenic Signaling Pathways Induced by G Protein‐coupled Receptors,” Journal of Cellular Physiology 213, no. 3 (2007): 589–602. [DOI] [PubMed] [Google Scholar]
  • 110. Kalinec G., Nazarali A. J., Hermouet S., Xu N., and Gutkind J. S., “Mutated Alpha Subunit of the Gq Protein Induces Malignant Transformation in NIH 3T3 Cells,” Molecular and Cellular Biology 12, no. 10 (1992): 4687–4693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Arora P., Cuevas B. D., Russo A., Johnson G. L., and Trejo J., “Persistent Transactivation of EGFR and ErbB2/HER2 by Protease‐activated Receptor‐1 Promotes Breast Carcinoma Cell Invasion,” Oncogene 27, no. 32 (2008): 4434–4445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Arora P., Ricks T. K., and Trejo J., “Protease‐activated Receptor Signalling, Endocytic Sorting and Dysregulation in Cancer,” Journal of Cell Science 120, no. 6 (2007): 921–928. [DOI] [PubMed] [Google Scholar]
  • 113. Clevers H., “Wnt/Beta‐catenin Signaling in Development and Disease,” Cell 127, no. 3 (2006): 469–480. [DOI] [PubMed] [Google Scholar]
  • 114. Lai S. L., Chien A. J., and Moon R. T., “Wnt/Fz Signaling and the Cytoskeleton: Potential Roles in Tumorigenesis,” Cell Research 19, no. 5 (2009): 532–545. [DOI] [PubMed] [Google Scholar]
  • 115. Gugger M., White R., Song S., et al., “GPR87 is an Overexpressed G‐protein Coupled Receptor in Squamous Cell Carcinoma of the Lung,” Disease Markers 24, no. 1 (2008): 41–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Lustig B. and Behrens J., “The Wnt Signaling Pathway and Its Role in Tumor Development,” Journal of Cancer Research and Clinical Oncology 129, no. 4 (2003): 199–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Schwartz G. G., “Prostate Cancer, Serum Parathyroid Hormone, and the Progression of Skeletal Metastases,” Cancer Epidemiology, Biomarkers & Prevention: a Publication of the American Association for Cancer Research, Cosponsored By the American Society of Preventive Oncology 17, no. 3 (2008): 478–483. [DOI] [PubMed] [Google Scholar]
  • 118. Gschwind A., Hart S., Fischer O. M., and Ullrich A., “TACE Cleavage of Proamphiregulin Regulates GPCR‐induced Proliferation and Motility of Cancer Cells,” The EMBO Journal 22, no. 10 (2003): 2411–2421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Greenhough A., Smartt H. J., Moore A. E., et al., “The COX‐2/PGE2 Pathway: Key Roles in the Hallmarks of Cancer and Adaptation to the Tumour Microenvironment,” Carcinogenesis 30, no. 3 (2009): 377–386. [DOI] [PubMed] [Google Scholar]
  • 120. Contos J. J., Ishii I., and Chun J., “Lysophosphatidic Acid Receptors,” Molecular Pharmacology 58, no. 6 (2000): 1188–1196. [DOI] [PubMed] [Google Scholar]
  • 121. Sanchez‐Vega F., Mina M., Armenia J., et al., “Oncogenic Signaling Pathways in the Cancer Genome Atlas,” Cell 173, no. 2 (2018): 321–337.e310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Sudol M., Bork P., Einbond A., et al., “Characterization of the Mammalian YAP (Yes‐associated protein) Gene and Its Role in Defining a Novel Protein Module, the WW Domain,” The Journal of Biological Chemistry 270, no. 24 (1995): 14733–14741. [DOI] [PubMed] [Google Scholar]
  • 123. Feng X., Degese M. S., Iglesias‐Bartolome R., et al., “Hippo‐independent Activation of YAP by the GNAQ Uveal Melanoma Oncogene Through a Trio‐regulated Rho GTPase Signaling Circuitry,” Cancer Cell 25, no. 6 (2014): 831–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Baud V. and Karin M., “Is NF‐kappaB a Good Target for Cancer Therapy? Hopes and Pitfalls,” Nature Reviews Drug Discovery 8, no. 1 (2009): 33–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Van Raamsdonk C. D., Bezrookove V., Green G., et al., “Frequent Somatic Mutations of GNAQ in Uveal Melanoma and Blue Naevi,” Nature 457, no. 7229 (2009): 599–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Van Raamsdonk C. D., Griewank K. G., and Crosby M. B., “Mutations in GNA11 in Uveal Melanoma,” The New England Journal of Medicine 363, no. 23 (2010): 2191–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Ayturk U. M., Couto J. A., and Hann S., “Somatic Activating Mutations in GNAQ and GNA11 Are Associated With Congenital Hemangioma,” American Journal of Human Genetics 98, no. 4 (2016): 789–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Comi A. M., “Sturge‐Weber Syndrome,” Handbook of Clinical Neurology 132 (2015): 157–168. [DOI] [PubMed] [Google Scholar]
  • 129. Robertson A. G., Shih J., Yau C., et al., “Integrative Analysis Identifies Four Molecular and Clinical Subsets in Uveal Melanoma,” Cancer Cell 32, no. 2 (2017): 204–220.e215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Johansson P., Aoude L. G., Wadt K., et al., “Deep Sequencing of Uveal Melanoma Identifies a Recurrent Mutation in PLCB4,” Oncotarget 7, no. 4 (2016): 4624–4631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Moore A. R., Ceraudo E., Sher J. J., et al., “Recurrent Activating Mutations of G‐protein‐coupled Receptor CYSLTR2 in Uveal Melanoma,” Nature Genetics 48, no. 6 (2016): 675–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Vaqué J. P., Dorsam R. T., Feng X., et al., “A Genome‐wide RNAi Screen Reveals a Trio‐regulated Rho GTPase Circuitry Transducing Mitogenic Signals Initiated by G Protein‐coupled Receptors,” Molecular Cell 49, no. 1 (2013): 94–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Pan D., “The Hippo Signaling Pathway in Development and Cancer,” Developmental Cell 19, no. 4 (2010): 491–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Feng X., Degese Maria S., and Iglesias‐Bartolome R., “Hippo‐Independent Activation of YAP by the GNAQ Uveal Melanoma Oncogene Through a Trio‐Regulated Rho GTPase Signaling Circuitry,” Cancer Cell 25, no. 6 (2014): 831–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Yu F. X., Luo J., and Mo J. S., “Mutant Gq/11 Promote Uveal Melanoma Tumorigenesis by Activating YAP,” Cancer Cell 25, no. 6 (2014): 822–830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Küsters‐Vandevelde H. V., van Engen‐van Grunsven I. A., Küsters B., et al., “Improved Discrimination of Melanotic Schwannoma From Melanocytic Lesions by Combined Morphological and GNAQ Mutational Analysis,” Acta Neuropathologica 120, no. 6 (2010): 755–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Misra R. S., Shi G., Moreno‐Garcia M. E., et al., “G Alpha Q‐containing G Proteins Regulate B Cell Selection and Survival and Are Required to Prevent B Cell‐dependent Autoimmunity,” The Journal of Experimental Medicine 207, no. 8 (2010): 1775–1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. He Y., Yuan X., Li Y., et al., “Loss of Gαq Impairs Regulatory B‐cell Function,” Arthritis Research & Therapy 20, no. 1 (2018): 186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Wang D., Zhang Y., He Y., Li Y., Lund F. E., and Shi G., “The Deficiency of Gαq Leads to Enhanced T‐cell Survival,” Immunology and Cell Biology 92, no. 9 (2014): 781–790. [DOI] [PubMed] [Google Scholar]
  • 140. Li Z., Zhang X., Xue W., et al., “Recurrent GNAQ Mutation Encoding T96S in Natural Killer/T Cell Lymphoma,” Nature Communications 10, no. 1 (2019): 4209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Wu V., Yeerna H., Nohata N., et al., “Illuminating the Onco‐GPCRome: Novel G Protein‐coupled Receptor‐driven Oncocrine Networks and Targets for Cancer Immunotherapy,” The Journal of Biological Chemistry 294, no. 29 (2019): 11062–11086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Landis C. A., Masters S. B., Spada A., Pace A. M., Bourne H. R., and Vallar L., “GTPase Inhibiting Mutations Activate the Alpha Chain of Gs and Stimulate Adenylyl Cyclase in human Pituitary Tumours,” Nature 340, no. 6236 (1989): 692–696. [DOI] [PubMed] [Google Scholar]
  • 143. Cassel D. and Pfeuffer T., “Mechanism of Cholera Toxin Action: Covalent Modification of the Guanyl Nucleotide‐binding Protein of the Adenylate Cyclase System,” Proceedings of the National Academy of Sciences of the United States of America 75, no. 6 (1978): 2669–2673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Gill D. M. and Meren R., “ADP‐ribosylation of Membrane Proteins Catalyzed by Cholera Toxin: Basis of the Activation of Adenylate Cyclase,” Proceedings of the National Academy of Sciences of the United States of America 75, no. 7 (1978): 3050–3054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Sunahara R. K., Tesmer J. J., Gilman A. G., and Sprang S. R., “Crystal Structure of the Adenylyl Cyclase Activator Gsalpha,” Science (New York, NY) 278, no. 5345 (1997): 1943–1947. [DOI] [PubMed] [Google Scholar]
  • 146. O'Hayre M., Vázquez‐Prado J., Kufareva I., et al., “The Emerging Mutational Landscape of G Proteins and G‐protein‐coupled Receptors in Cancer,” Nature Reviews Cancer 13, no. 6 (2013): 412–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Hu Q. and Shokat K. M., “Disease‐Causing Mutations in the G Protein Gαs Subvert the Roles of GDP and GTP,” Cell 173, no. 5 (2018): 1254–1264.e1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Vallar L., Spada A., and Giannattasio G., “Altered Gs and Adenylate Cyclase Activity in human GH‐secreting Pituitary Adenomas,” Nature 330, no. 6148 (1987): 566–568. [DOI] [PubMed] [Google Scholar]
  • 149. Drews R. T., Gravel R. A., and Collu R., “Identification of G Protein Alpha Subunit Mutations in human Growth Hormone (GH)‐ and GH/Prolactin‐secreting Pituitary Tumors by Single‐strand Conformation Polymorphism (SSCP) Analysis,” Molecular and Cellular Endocrinology 87, no. 1‐3 (1992): 125–129. [DOI] [PubMed] [Google Scholar]
  • 150. Raimondi F., Inoue A., Kadji F. M. N., et al., “Rare, Functional, Somatic Variants in Gene Families Linked to Cancer Genes: GPCR Signaling as a Paradigm,” Oncogene 38, no. 38 (2019): 6491–6506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Gao J., Aksoy B. A., Dogrusoz U., et al., “Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal,” Science Signaling 6, no. 269 (2013): pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Cerami E., Gao J., Dogrusoz U., et al., “The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data,” Cancer Discovery 2, no. 5 (2012): 401–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Castellone M. D., Teramoto H., Williams B. O., Druey K. M., and Gutkind J. S., “Prostaglandin E2 Promotes Colon Cancer Cell Growth Through a Gs‐axin‐beta‐catenin Signaling Axis,” Science (New York, NY) 310, no. 5753 (2005): 1504–1510. [DOI] [PubMed] [Google Scholar]
  • 154. Nomura R., Saito T., Mitomi H., et al., “GNAS Mutation as an Alternative Mechanism of Activation of the Wnt/β‐catenin Signaling Pathway in Gastric Adenocarcinoma of the Fundic Gland Type,” Human Pathology (2014): 45. [DOI] [PubMed] [Google Scholar]
  • 155. Wood L. D., Parsons D. W., Jones S., et al., “The Genomic Landscapes of Human Breast and Colorectal Cancers,” Science (New York, NY) 318, no. 5853 (2007): 1108–1113. [DOI] [PubMed] [Google Scholar]
  • 156. Nishikawa G., Sekine S., Ogawa R., et al., “Frequent GNAS Mutations in Low‐grade Appendiceal Mucinous Neoplasms,” British Journal of Cancer 108, no. 4 (2013): 951–958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Furukawa T., Kuboki Y., Tanji E., et al., “Whole‐exome Sequencing Uncovers Frequent GNAS Mutations in Intraductal Papillary Mucinous Neoplasms of the Pancreas,” Scientific Reports 1 (2011): 161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Takano S., Fukasawa M., Maekawa S., et al., “Deep Sequencing of Cancer‐related Genes Revealed GNAS Mutations to be Associated With Intraductal Papillary Mucinous Neoplasms and Its Main Pancreatic Duct Dilation,” PLoS ONE 9, no. 6 (2014): e98718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Alakus H., Babicky M. L., Ghosh P., et al., “Genome‐wide Mutational Landscape of Mucinous Carcinomatosis Peritonei of Appendiceal Origin,” Genome Medicine 6, no. 5 (2014): 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Rao R., Salloum R., Xin M., and Lu Q. R., “The G Protein Gαs Acts as a Tumor Suppressor in Sonic Hedgehog Signaling‐driven Tumorigenesis,” Cell Cycle (Georgetown, Tex) 15, no. 10 (2016): 1325–1330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Cowley G. S., Weir B. A., Vazquez F., et al., “Parallel Genome‐scale Loss of Function Screens in 216 Cancer Cell Lines for the Identification of Context‐specific Genetic Dependencies,” Scientific Data 1 (2014): 140035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Taussig R., Iñiguez‐Lluhi J. A., and Gilman A. G., “Inhibition of Adenylyl Cyclase by Gi alpha,” Science (New York, NY) 261, no. 5118 (1993): 218–221. [DOI] [PubMed] [Google Scholar]
  • 163. Bokoch G. M., Katada T., Northup J. K., Ui M., and Gilman A. G., “Purification and Properties of the Inhibitory Guanine Nucleotide‐binding Regulatory Component of Adenylate Cyclase,” The Journal of Biological Chemistry 259, no. 6 (1984): 3560–3567. [PubMed] [Google Scholar]
  • 164. Katada T. and Ui M., “Direct Modification of the Membrane Adenylate Cyclase System by Islet‐activating Protein due to ADP‐ribosylation of a Membrane Protein,” Proceedings of the National Academy of Sciences of the United States of America 79, no. 10 (1982): 3129–3133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Codina J., Hildebrandt J., Iyengar R., Birnbaumer L., Sekura R. D., and Manclark C. R., “Pertussis Toxin Substrate, the Putative Ni Component of Adenylyl Cyclases, Is an Alpha Beta Heterodimer Regulated by Guanine Nucleotide and Magnesium,” Proceedings of the National Academy of Sciences of the United States of America 80, no. 14 (1983): 4276–4280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Dhanasekaran N., Tsim S. T., Dermott J. M., and Onesime D., “Regulation of Cell Proliferation by G Proteins,” Oncogene 17, no. 11 Reviews (1998): 1383–1394. [DOI] [PubMed] [Google Scholar]
  • 167. Pace A. M., Wong Y. H., and Bourne H. R., “A Mutant Alpha Subunit of Gi2 Induces Neoplastic Transformation of Rat‐1 Cells,” Proceedings of the National Academy of Sciences of the United States of America 88, no. 16 (1991): 7031–7035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Garcia‐Marcos M., Ghosh P., and Farquhar M. G., “Molecular Basis of a Novel Oncogenic Mutation in GNAO1,” Oncogene 30, no. 23 (2011): 2691–2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Lyons J., Landis C. A., Harsh G., et al., “Two G Protein Oncogenes in human Endocrine Tumors,” Science (New York, NY) 249, no. 4969 (1990): 655–659. [DOI] [PubMed] [Google Scholar]
  • 170. Demir H., Donner I., Kivipelto L., et al., “Mutation Analysis of Inhibitory Guanine Nucleotide Binding Protein Alpha (GNAI) Loci in Young and Familial Pituitary Adenomas,” PLoS ONE 9, no. 10 (2014): e109897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Juneja J. and Casey P. J., “Role of G12 Proteins in Oncogenesis and Metastasis,” British Journal of Pharmacology 158, no. 1 (2009): 32–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Spicher K., Kalkbrenner F., Zobel A., et al., “G12 and G13 Alpha‐subunits Are Immunochemically Detectable in Most Membranes of Various Mammalian Cells and Tissues,” Biochemical and Biophysical Research Communications 198, no. 3 (1994): 906–914. [DOI] [PubMed] [Google Scholar]
  • 173. Xu N., Voyno‐Yasenetskaya T., and Gutkind J. S., “Potent Transforming Activity of the G13 Alpha Subunit Defines a Novel family of Oncogenes,” Biochemical and Biophysical Research Communications 201, no. 2 (1994): 603–609. [DOI] [PubMed] [Google Scholar]
  • 174. Vara Prasad M. V., Shore S. K., and Dhanasekaran N., “Activated Mutant of G Alpha 13 Induces Egr‐1, c‐fos, and Transformation in NIH 3T3 Cells,” Oncogene 9, no. 8 (1994): 2425–2429. [PubMed] [Google Scholar]
  • 175. Kelly P., Moeller B. J., Juneja J., et al., “The G12 family of Heterotrimeric G Proteins Promotes Breast Cancer Invasion and Metastasis,” Proceedings of the National Academy of Sciences of the United States of America 103, no. 21 (2006): 8173–8178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Gan C. P., Patel V., Mikelis C. M., et al., “Heterotrimeric G‐protein Alpha‐12 (Gα12) Subunit Promotes Oral Cancer Metastasis,” Oncotarget 5, no. 20 (2014): 9626–9640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Marinissen M. J. and Gutkind J. S., “G‐protein‐coupled Receptors and Signaling Networks: Emerging Paradigms,” Trends in Pharmacological Sciences 22, no. 7 (2001): 368–376. [DOI] [PubMed] [Google Scholar]
  • 178. Hart M. J., Jiang X., Kozasa T., et al., “Direct Stimulation of the Guanine Nucleotide Exchange Activity of p115 RhoGEF by Galpha13,” Science (New York, NY) 280, no. 5372 (1998): 2112–2114. [DOI] [PubMed] [Google Scholar]
  • 179. Fukuhara S., Chikumi H., and Gutkind J. S., “Leukemia‐associated Rho Guanine Nucleotide Exchange Factor (LARG) Links Heterotrimeric G Proteins of the G(12) family to Rho,” FEBS Letters 485, no. 2‐3 (2000): 183–188. [DOI] [PubMed] [Google Scholar]
  • 180. Kozasa T., Jiang X., Hart M. J., et al., “p115 RhoGEF, a GTPase Activating Protein for Gα12and Gα13 ,” Science (New York, NY) 280, no. 5372 (1998): 2109–2111. [DOI] [PubMed] [Google Scholar]
  • 181. Mikelis C. M., Palmby T. R., Simaan M., et al., “PDZ‐RhoGEF and LARG Are Essential for Embryonic Development and Provide a Link Between Thrombin and LPA Receptors and Rho Activation,” The Journal of Biological Chemistry 288, no. 17 (2013): 12232–12243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Fromm C., Coso O. A., Montaner S., Xu N., and Gutkind J. S., “The Small GTP‐binding Protein Rho Links G Protein‐coupled Receptors and Galpha12 to the Serum Response Element and to Cellular Transformation,” Proceedings of the National Academy of Sciences of the United States of America 94, no. 19 (1997): 10098–10103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Radhika V. and Dhanasekaran N., “Transforming G Proteins,” Oncogene 20, no. 13 (2001): 1607–1614. [DOI] [PubMed] [Google Scholar]
  • 184. Rasheed S. A. K., Subramanyan L. V., Lim W. K., Udayappan U. K., Wang M., and Casey P. J., “The Emerging Roles of Gα12/13 Proteins on the Hallmarks of Cancer in Solid Tumors,” Oncogene 41, no. 2 (2022): 147–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Goldsmith Z. G. and Dhanasekaran D. N., “G Protein Regulation of MAPK Networks,” Oncogene 26, no. 22 (2007): 3122–3142. [DOI] [PubMed] [Google Scholar]
  • 186. Kelly P., Stemmle L. N., Madden J. F., Fields T. A., Daaka Y., and Casey P. J., “A Role for the G12 family of Heterotrimeric G Proteins in Prostate Cancer Invasion,” The Journal of Biological Chemistry 281, no. 36 (2006): 26483–26490. [DOI] [PubMed] [Google Scholar]
  • 187. Bartolomé R. A., Wright N., Molina‐Ortiz I., Sánchez‐Luque F. J., and Teixidó J., “Activated G(alpha)13 Impairs Cell Invasiveness Through p190RhoGAP‐mediated Inhibition of RhoA Activity,” Cancer Research 68, no. 20 (2008): 8221–8230. [DOI] [PubMed] [Google Scholar]
  • 188. Bian D., Mahanivong C., Yu J., et al., “The G12/13‐RhoA Signaling Pathway Contributes to Efficient Lysophosphatidic Acid‐stimulated Cell Migration,” Oncogene 25, no. 15 (2006): 2234–2244. [DOI] [PubMed] [Google Scholar]
  • 189. Xu Y., Rong J., Duan S., et al., “High Expression of GNA13 Is Associated With Poor Prognosis in Hepatocellular Carcinoma,” Scientific Reports 6 (2016): 35948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Daaka Y., “G Proteins in Cancer: The Prostate Cancer Paradigm,” Science's STKE: Signal Transduction Knowledge Environment 2004, no. 216 (2004): re2. [DOI] [PubMed] [Google Scholar]
  • 191. Rasheed S. A. K., Leong H. S., Lakshmanan M., et al., “GNA13 expression Promotes Drug Resistance and Tumor‐initiating Phenotypes in Squamous Cell Cancers,” Oncogene 37, no. 10 (2018): 1340–1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Zhang J. X., Yun M., Xu Y., et al., “GNA13 as a Prognostic Factor and Mediator of Gastric Cancer Progression,” Oncotarget 7, no. 4 (2016): 4414–4427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Smith M. C., Luker K. E., Garbow J. R., et al., “CXCR4 regulates Growth of both Primary and Metastatic Breast Cancer,” Cancer Research 64, no. 23 (2004): 8604–8612. [DOI] [PubMed] [Google Scholar]
  • 194. Yagi H., Tan W., Dillenburg‐Pilla P., et al., “A Synthetic Biology Approach Reveals a CXCR4‐G13‐Rho Signaling Axis Driving Transendothelial Migration of Metastatic Breast Cancer Cells,” Science Signaling 4, no. 191 (2011): ra60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. O'Hayre M., Inoue A., and Kufareva I., “Inactivating Mutations in GNA13 and RHOA in Burkitt's Lymphoma and Diffuse Large B‐cell Lymphoma: A Tumor Suppressor Function for the Gα13/RhoA Axis in B Cells,” Oncogene 35, no. 29 (2016): 3771–3780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Love C., Sun Z., Jima D., et al., “The Genetic Landscape of Mutations in Burkitt Lymphoma,” Nature Genetics 44, no. 12 (2012): 1321–1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Muppidi J. R., Schmitz R., Green J. A., et al., “Loss of Signalling via Gα13 in Germinal Centre B‐cell‐derived Lymphoma,” Nature 516, no. 7530 (2014): 254–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Healy J. A., Nugent A., Rempel R. E., et al., “GNA13 loss in Germinal Center B Cells Leads to Impaired Apoptosis and Promotes Lymphoma in Vivo,” Blood 127, no. 22 (2016): 2723–2731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Hurst J. H. and Hooks S. B., “Regulator of G‐protein Signaling (RGS) Proteins in Cancer Biology,” Biochemical Pharmacology 78, no. 10 (2009): 1289–1297. [DOI] [PubMed] [Google Scholar]
  • 200. DiGiacomo V., Maziarz M., Luebbers A., Norris J. M., Laksono P., and Garcia‐Marcos M., “Probing the Mutational Landscape of Regulators of G Protein Signaling Proteins in Cancer,” Science Signaling no. 617 (2020): 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Sethakorn N. and Dulin N. O., “RGS Expression in Cancer: Oncomining the Cancer Microarray Data,” Journal of Receptor and Signal Transduction Research 33, no. 3 (2013): 166–171. [DOI] [PubMed] [Google Scholar]
  • 202. Zimmermannova O., Doktorova E., Stuchly J., et al., “An Activating Mutation of GNB1 Is Associated With Resistance to Tyrosine Kinase Inhibitors in ETV6‐ABL1‐positive Leukemia,” Oncogene 36, no. 43 (2017): 5985–5994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Barber M. A., Donald S., Thelen S., Anderson K. E., Thelen M., and Welch H. C. E., “Membrane Translocation of P‐Rex1 Is Mediated by G Protein Βγ Subunits and Phosphoinositide 3‐Kinase*,” Journal of Biological Chemistry 282, no. 41 (2007): 29967–29976. [DOI] [PubMed] [Google Scholar]
  • 204. Welch H. C., Coadwell W. J., Ellson C. D., et al., “P‐Rex1, a PtdIns(3,4,5)P3‐ and Gbetagamma‐regulated Guanine‐nucleotide Exchange Factor for Rac,” Cell 108, no. 6 (2002): 809–821. [DOI] [PubMed] [Google Scholar]
  • 205. Khalil B. D., Hsueh C., Cao Y., et al., “GPCR Signaling Mediates Tumor Metastasis via PI3Kβ,” Cancer Research 76, no. 10 (2016): 2944–2953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Dbouk H. A., Vadas O., Shymanets A., et al., “G Protein‐coupled Receptor‐mediated Activation of p110β by Gβγ Is Required for Cellular Transformation and Invasiveness,” Science Signaling 5, no. 253 (2012): ra89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Tang X., Sun Z., Runne C., et al., “A Critical Role of Gbetagamma in Tumorigenesis and Metastasis of Breast Cancer,” The Journal of Biological Chemistry 286, no. 15 (2011): 13244–13254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Vázquez‐Prado J., Bracho‐Valdés I., Cervantes‐Villagrana R. D., and Reyes‐Cruz G., “Gβγ Pathways in Cell Polarity and Migration Linked to Oncogenic GPCR Signaling: Potential Relevance in Tumor Microenvironment,” Molecular Pharmacology 90, no. 5 (2016): 573–586. [DOI] [PubMed] [Google Scholar]
  • 209. Bookout A. L., Finney A. E., Guo R., Peppel K., Koch W. J., and Daaka Y., “Targeting Gbetagamma Signaling to Inhibit Prostate Tumor Formation and Growth,” The Journal of Biological Chemistry 278, no. 39 (2003): 37569–37573. [DOI] [PubMed] [Google Scholar]
  • 210. Sosa M. S., Lopez‐Haber C., Yang C., et al., “Identification of the Rac‐GEF P‐Rex1 as an Essential Mediator of ErbB Signaling in Breast Cancer,” Molecular Cell 40, no. 6 (2010): 877–892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Korinek V., Barker N., Morin P. J., et al., “Constitutive Transcriptional Activation by a Beta‐catenin‐Tcf Complex in APC‐/‐ colon Carcinoma,” Science (New York, NY) 275, no. 5307 (1997): 1784–1787. [DOI] [PubMed] [Google Scholar]
  • 212. Valenta T., Hausmann G., and Basler K., “The Many Faces and Functions of β‐catenin,” The EMBO Journal 31, no. 12 (2012): 2714–2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. van Amerongen R., “Alternative Wnt Pathways and Receptors,” Cold Spring Harbor Perspectives in Biology 4 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Rasola A., Fassetta M., De Bacco F., et al., “A Positive Feedback Loop Between Hepatocyte Growth Factor Receptor and Beta‐catenin Sustains Colorectal Cancer Cell Invasive Growth,” Oncogene 26, no. 7 (2007): 1078–1087. [DOI] [PubMed] [Google Scholar]
  • 215. Yang L., Lin C., and Liu Z. R., “P68 RNA Helicase Mediates PDGF‐induced Epithelial Mesenchymal Transition by Displacing Axin From Beta‐catenin,” Cell 127, no. 1 (2006): 139–155. [DOI] [PubMed] [Google Scholar]
  • 216. Birdsey G. M., Shah A. V., and Dufton N., “The Endothelial Transcription Factor ERG Promotes Vascular Stability and Growth Through Wnt/β‐catenin Signaling,” Developmental Cell 32, no. 1 (2015): 82–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Katanaev V. L., Ponzielli R., Sémériva M., and Tomlinson A., “Trimeric G Protein‐dependent Frizzled Signaling in Drosophila,” Cell 120, no. 1 (2005): 111–122. [DOI] [PubMed] [Google Scholar]
  • 218. Liu T., DeCostanzo A. J., Liu X., et al., “G Protein Signaling From Activated Rat Frizzled‐1 to the β‐Catenin‐Lef‐Tcf Pathway,” Science (New York, NY) 292, no. 5522 (2001): 1718–1722. [DOI] [PubMed] [Google Scholar]
  • 219. Slusarski D. C., Corces V. G., and Moon R. T., “Interaction of Wnt and a Frizzled Homologue Triggers G‐protein‐linked Phosphatidylinositol Signalling,” Nature 390, no. 6658 (1997): 410–413. [DOI] [PubMed] [Google Scholar]
  • 220. Chen M. and He X., “APC Deficiency Leads to β‐Catenin Stabilization and Signaling Independent of LRP5/6,” Developmental Cell 49, no. 6 (2019): 825–826. [DOI] [PubMed] [Google Scholar]
  • 221. Regard J. B., Cherman N., Palmer D., et al., “Wnt/β‐catenin Signaling Is Differentially Regulated by Gα Proteins and Contributes to Fibrous Dysplasia,” Proceedings of the National Academy of Sciences of the United States of America 108, no. 50 (2011): 20101–20106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Miller E., Yang J., DeRan M., et al., “Identification of Serum‐derived Sphingosine‐1‐phosphate as a Small Molecule Regulator of YAP,” Chemistry & Biology 19, no. 8 (2012): 955–962. [DOI] [PubMed] [Google Scholar]
  • 223. Yu F. X., Zhao B., Panupinthu N., et al., “Regulation of the Hippo‐YAP Pathway by G‐Protein‐Coupled Receptor Signaling,” Cell 187, no. 6 (2024): 1563–1564. [DOI] [PubMed] [Google Scholar]
  • 224. Li H., Li Q., Dang K., et al., “YAP/TAZ Activation Drives Uveal Melanoma Initiation and Progression,” Cell Reports 29, no. 10 (2019): 3200–3211.e3204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Ramos A. and Camargo F. D., “The Hippo Signaling Pathway and Stem Cell Biology,” Trends in Cell Biology 22, no. 7 (2012): 339–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Kim M., Kim M., Lee S., et al., “Lim DS: CAMP/PKA Signalling Reinforces the LATS‐YAP Pathway to Fully Suppress YAP in Response to Actin Cytoskeletal Changes,” The EMBO Journal 32, no. 11 (2013): 1543–1555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Iglesias‐Bartolome R., Torres D., Marone R., et al., “Inactivation of a Gαs‐PKA Tumor Suppressor Pathway in Skin Stem Cells Initiates Basal‐cell Carcinogenesis,” Nature Cell Biology 17 (2015): 793–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Gong R., Hong A. W., Plouffe S. W., et al., “Opposing Roles of Conventional and Novel PKC Isoforms in Hippo‐YAP Pathway Regulation,” Cell Research 25, no. 8 (2015): 985–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Yu F. X., Zhao B., Panupinthu N., et al., “Regulation of the Hippo‐YAP Pathway by G‐protein‐coupled Receptor Signaling,” Cell 150, no. 4 (2012): 780–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Meng Z., Moroishi T., Mottier‐Pavie V., et al., “MAP4K family Kinases Act in Parallel to MST1/2 to Activate LATS1/2 in the Hippo Pathway,” Nature Communications 6, no. 1 (2015): 8357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Yu F. X., Zhao B., and Guan K. L., “Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer,” Cell 163, no. 4 (2015): 811–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Park H. W., Kim Y. C., Yu B., et al., “Alternative Wnt Signaling Activates YAP/TAZ,” Cell 162, no. 4 (2015): 780–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Hao F., Xu Q., Zhao Y., et al., “Insulin Receptor and GPCR Crosstalk Stimulates YAP via PI3K and PKD in Pancreatic Cancer Cells,” Molecular Cancer Research: MCR 15, no. 7 (2017): 929–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Feng R., Gong J., Wu L., et al., “MAPK and Hippo Signaling Pathways Crosstalk via the RAF‐1/MST‐2 Interaction in Malignant Melanoma,” Oncology Reports 38, no. 2 (2017): 1199–1205. [DOI] [PubMed] [Google Scholar]
  • 235. Rosenbluh J., Nijhawan D., Cox A. G., et al., “Β‐Catenin‐driven Cancers Require a YAP1 Transcriptional Complex for Survival and Tumorigenesis,” Cell 151, no. 7 (2012): 1457–1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Dethlefsen C., Hansen L. S., Lillelund C., et al., “Exercise‐Induced Catecholamines Activate the Hippo Tumor Suppressor Pathway to Reduce Risks of Breast Cancer Development,” Cancer Research 77, no. 18 (2017): 4894–4904. [DOI] [PubMed] [Google Scholar]
  • 237. Bao Y., Nakagawa K., Yang Z., et al., “A Cell‐based Assay to Screen Stimulators of the Hippo Pathway Reveals the Inhibitory Effect of Dobutamine on the YAP‐dependent Gene Transcription,” Journal of Biochemistry 150, no. 2 (2011): 199–208. [DOI] [PubMed] [Google Scholar]
  • 238. Park H. W. and Guan K.‐L., “Regulation of the Hippo Pathway and Implications for Anticancer Drug Development,” Trends in Pharmacological Sciences 34, no. 10 (2013): 581–589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Nieto Gutierrez A. and McDonald P. H., “GPCRs: Emerging Anti‐cancer Drug Targets,” Cellular Signalling 41 (2018): 65–74. [DOI] [PubMed] [Google Scholar]
  • 240. Onken M. D., Makepeace C. M., Kaltenbronn K. M., et al., “Targeting Nucleotide Exchange to Inhibit Constitutively Active G Protein α Subunits in Cancer Cells,” Science Signaling 11, no. 546 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Annala S., Feng X., Shridhar N., et al., “Direct Targeting of Gα≪Sub≫Q≪/Sub≫ and Gα≪Sub≫11≪/Sub≫ Oncoproteins in Cancer Cells,” Science Signaling 12 (2019): eaau5948. [DOI] [PubMed] [Google Scholar]
  • 242. Chen X., Wu Q., Depeille P., et al., “RasGRP3 Mediates MAPK Pathway Activation in GNAQ Mutant Uveal Melanoma,” Cancer Cell 31, no. 5 (2017): 685–696.e686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Borcherding D. C., Tong W., Hugo E. R., et al., “Expression and Therapeutic Targeting of Dopamine Receptor‐1 (D1R) in Breast Cancer,” Oncogene 35, no. 24 (2016): 3103–3113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Sever R. and Brugge J. S., “Signal Transduction in Cancer,” Cold Spring Harbor Perspectives in Medicine 5, no. 4 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Sari I. N., Phi L. T. H., Jun N., Wijaya Y. T., Lee S., and Kwon H. Y., “Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells,” Cells 7, no. 11 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Liu X., Ding C., Tan W., and Zhang A., “Medulloblastoma: Molecular Understanding, Treatment Evolution, and New Developments,” Pharmacology & Therapeutics 210 (2020): 107516. [DOI] [PubMed] [Google Scholar]
  • 247. Didiasova M., Schaefer L., and Wygrecka M., “Targeting GLI Transcription Factors in Cancer,” Molecules (Basel, Switzerland) 23, no. 5 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Lospinoso Severini L., Quaglio D., and Basili I., “A Smo/Gli Multitarget Hedgehog Pathway Inhibitor Impairs Tumor Growth,” Cancers 11, no. 10 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Hanahan D. and Weinberg R. A., “Hallmarks of Cancer: The next Generation,” Cell 144, no. 5 (2011): 646–674. [DOI] [PubMed] [Google Scholar]
  • 250. Zhang M. and Qiu S., “Activation of GPR120 Promotes the Metastasis of Breast Cancer Through the PI3K/Akt/NF‐κB Signaling Pathway,” Anti‐Cancer Drugs 30, no. 3 (2019): 260–270. [DOI] [PubMed] [Google Scholar]
  • 251. Zhang L.‐Q., Yang S.‐Q., Qu X.‐D., Chen X.‐J., Lu H.‐S., and Wang Y., “GRP137 promotes Cell Proliferation and Metastasis Through Regulation of the PI3K/AKT Pathway in human Ovarian Cancer,” Tumori Journal 104, no. 5 (2018): 330–337. [DOI] [PubMed] [Google Scholar]
  • 252. Smit M. J., Schlecht‐Louf G., Neves M., et al., “The CXCL12/CXCR4/ACKR3 Axis in the Tumor Microenvironment: Signaling, Crosstalk, and Therapeutic Targeting,” Annual Review of Pharmacology and Toxicology 61 (2021): 541–563. [DOI] [PubMed] [Google Scholar]
  • 253. Jewell J. L., Fu V., Hong A. W., et al., “GPCR Signaling Inhibits mTORC1 via PKA Phosphorylation of Raptor,” Elife 8, (2019):. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Mu G., Ding Q., Li H., et al., “Gastrin Stimulates Pancreatic Cancer Cell Directional Migration by Activating the Gα12/13‐RhoA‐ROCK Signaling Pathway,” Experimental & Molecular Medicine 50, no. 5 (2018): 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Gad A. A. and Balenga N., “The Emerging Role of Adhesion GPCRs in Cancer,” ACS Pharmacology & Translational Science 3, no. 1 (2020): 29–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Chambers A. F., Groom A. C., and MacDonald I. C., “Dissemination and Growth of Cancer Cells in Metastatic Sites,” Nature Reviews Cancer 2, no. 8 (2002): 563–572. [DOI] [PubMed] [Google Scholar]
  • 257. Müller A., Homey B., Soto H., et al., “Involvement of Chemokine Receptors in Breast Cancer Metastasis,” Nature 410, no. 6824 (2001): 50–56. [DOI] [PubMed] [Google Scholar]
  • 258. Balkwill F., “Cancer and the Chemokine Network,” Nature Reviews Cancer 4, no. 7 (2004): 540–550. [DOI] [PubMed] [Google Scholar]
  • 259. Zlotnik A., Burkhardt A. M., and Homey B., “Homeostatic Chemokine Receptors and Organ‐specific Metastasis,” Nature Reviews Immunology 11, no. 9 (2011): 597–606. [DOI] [PubMed] [Google Scholar]
  • 260. Tang X., Jin R., Qu G., et al., “GPR116, an Adhesion G‐protein‐coupled Receptor, Promotes Breast Cancer Metastasis via the Gαq‐p63RhoGEF‐Rho GTPase Pathway,” Cancer Research 73, no. 20 (2013): 6206–6218. [DOI] [PubMed] [Google Scholar]
  • 261. Nag J. K., Malka H., Appasamy P., Sedley S., and Bar‐Shavit R., “GPCR Partners as Cancer Driver Genes: Association With PH‐Signal Proteins in a Distinctive Signaling Network,” International Journal of Molecular Sciences 22, no. 16 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Moore B. B., Arenberg D. A., Addison C. L., Keane M. P., Polverini P. J., and Strieter R. M., “CXC Chemokines Mechanism of Action in Regulating Tumor Angiogenesis,” Angiogenesis 2, no. 2 (1998): 123–134. [DOI] [PubMed] [Google Scholar]
  • 263. Richard D. E., Vouret‐Craviari V., and Pouysségur J., “Angiogenesis and G‐protein‐coupled Receptors: Signals That Bridge the Gap,” Oncogene 20, no. 13 (2001): 1556–1562. [DOI] [PubMed] [Google Scholar]
  • 264. Wang D. and Dubois R. N., “Prostaglandins and Cancer,” Gut 55, no. 1 (2006): 115–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Iñiguez M. A., Rodríguez A., Volpert O. V., Fresno M., and Redondo J. M., “Cyclooxygenase‐2: A Therapeutic Target in Angiogenesis,” Trends in Molecular Medicine 9, no. 2 (2003): 73–78. [DOI] [PubMed] [Google Scholar]
  • 266. Zeng D., Li M., Zhou R., et al., “Tumor Microenvironment Characterization in Gastric Cancer Identifies Prognostic and Immunotherapeutically Relevant Gene Signatures,” Cancer Immunology Research 7, no. 5 (2019): 737–750. [DOI] [PubMed] [Google Scholar]
  • 267. Rojas A., Araya P., Gonzalez I., and Morales E., “Gastric Tumor Microenvironment,” Advances in Experimental Medicine and Biology 1226 (2020): 23–35. [DOI] [PubMed] [Google Scholar]
  • 268. Quail D. F. and Joyce J. A., “Microenvironmental Regulation of Tumor Progression and Metastasis,” Nature Medicine 19, no. 11 (2013): 1423–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Chiu K. J., Chiou H. C., Huang C. H., et al., “Natural Compounds Targeting Cancer‐Associated Fibroblasts Against Digestive System Tumor Progression: Therapeutic Insights,” Biomedicines 10, no. 3 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Peltier A., Seban R. D., Buvat I., Bidard F. C., and Mechta‐Grigoriou F., “Fibroblast Heterogeneity in Solid Tumors: From Single Cell Analysis to Whole‐body Imaging,” Seminars in Cancer Biology 86, no. Pt 3 (2022): 262–272. [DOI] [PubMed] [Google Scholar]
  • 271. Nagarsheth N., Wicha M. S., and Zou W., “Chemokines in the Cancer Microenvironment and Their Relevance in Cancer Immunotherapy,” Nature Reviews Immunology 17, no. 9 (2017): 559–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Gorbachev A. V. and Fairchild R. L., “Regulation of Chemokine Expression in the Tumor Microenvironment,” Critical Reviews in Immunology 34, no. 2 (2014): 103–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273. Shen H., Gu X., Li X., et al., “C5aR1 shapes a Non‐inflammatory Tumor Microenvironment and Mediates Immune Evasion in Gastric Cancer,” Biomolecules & Biomedicine 23, no. 3 (2023): 392–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Chen F., Yin S., Niu L., et al., “Expression of the Chemokine Receptor CXCR3 Correlates With Dendritic Cell Recruitment and Prognosis in Gastric Cancer,” Genetic Testing and Molecular Biomarkers 22, no. 1 (2018): 35–42. [DOI] [PubMed] [Google Scholar]
  • 275. Sebrell T. A., Hashimi M., Sidar B., et al., “A Novel Gastric Spheroid Co‐culture Model Reveals Chemokine‐Dependent Recruitment of Human Dendritic Cells to the Gastric Epithelium,” Cellular and Molecular Gastroenterology and Hepatology 8, no. 1 (2019): 157–171.e153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276. Maimela N. R., Liu S., and Zhang Y., “Fates of CD8+ T Cells in Tumor Microenvironment,” Computational and Structural Biotechnology Journal 17 (2019): 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277. Slaney C. Y., Kershaw M. H., and Darcy P. K., “Trafficking of T Cells Into Tumors,” Cancer Research 74, no. 24 (2014): 7168–7174. [DOI] [PubMed] [Google Scholar]
  • 278. Ishigami S., Natsugoe S., Tokuda K., et al., “Clinical Impact of Intratumoral Natural Killer Cell and Dendritic Cell Infiltration in Gastric Cancer,” Cancer Letters 159, no. 1 (2000): 103–108. [DOI] [PubMed] [Google Scholar]
  • 279. Bald T., Krummel M. F., Smyth M. J., and Barry K. C., “The NK Cell‐cancer Cycle: Advances and New Challenges in NK Cell‐based Immunotherapies,” Nature Immunology 21, no. 8 (2020): 835–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Bikfalvi A. and Billottet C., “The CC and CXC Chemokines: Major Regulators of Tumor Progression and the Tumor Microenvironment,” American Journal of Physiology Cell Physiology 318, no. 3 (2020): C542–c554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281. Mizukami Y., Kono K., Kawaguchi Y., et al., “CCL17 and CCL22 Chemokines Within Tumor Microenvironment Are Related to Accumulation of Foxp3+ Regulatory T Cells in Gastric Cancer,” International Journal of Cancer 122, no. 10 (2008): 2286–2293. [DOI] [PubMed] [Google Scholar]
  • 282. Liu X., Zhang Z., and Zhao G., “Recent Advances in the Study of Regulatory T Cells in Gastric Cancer,” International Immunopharmacology 73 (2019): 560–567. [DOI] [PubMed] [Google Scholar]
  • 283. Parisi L., Gini E., Baci D., et al., “Macrophage Polarization in Chronic Inflammatory Diseases: Killers or Builders?,” Journal of Immunology Research 2018 (2018): 8917804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284. Ruytinx P., Proost P., Van Damme J., and Struyf S., “Chemokine‐Induced Macrophage Polarization in Inflammatory Conditions,” Frontiers in Immunology 9 (2018): 1930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285. Echizen K., Hirose O., Maeda Y., and Oshima M., “Inflammation in Gastric Cancer: Interplay of the COX‐2/Prostaglandin E2 and Toll‐Like Receptor/MyD88 Pathways,” Cancer Science 107, no. 4 (2016): 391–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Oshima H. and Oshima M., “The Role of PGE2‐associated Inflammatory Responses in Gastric Cancer Development,” Seminars in Immunopathology 35, no. 2 (2013): 139–150. [DOI] [PubMed] [Google Scholar]
  • 287. Labrie F., Bélanger A., Luu‐The V., et al., “Gonadotropin‐releasing Hormone Agonists in the Treatment of Prostate Cancer,” Endocrine Reviews 26, no. 3 (2005): 361–379. [DOI] [PubMed] [Google Scholar]
  • 288. Meyer T., Caplin M. E., Palmer D. H., et al., “A Phase Ib/IIa Trial to Evaluate the CCK2 Receptor Antagonist Z‐360 in Combination With Gemcitabine in Patients With Advanced Pancreatic Cancer,” European Journal of Cancer (Oxford, England: 1990) 46, no. 3 (2010): 526–533. [DOI] [PubMed] [Google Scholar]
  • 289. Fischgräbe J., Götte M., Michels K., Kiesel L., and Wülfing P., “Targeting Endothelin A Receptor Enhances Anti‐proliferative and Anti‐invasive Effects of the HER2 Antibody Trastuzumab in HER2‐overexpressing Breast Cancer Cells,” International Journal of Cancer 127, no. 3 (2010): 696–706. [DOI] [PubMed] [Google Scholar]
  • 290. Liu X., Yu J., Song S., Yue X., and Li Q., “Protease‐activated Receptor‐1 (PAR‐1): A Promising Molecular Target for Cancer,” Oncotarget 8, no. 63 (2017): 107334–107345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. DeWire S. M., Yamashita D. S., Rominger D. H., et al., “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, no. 3 (2013): 708–717. [DOI] [PubMed] [Google Scholar]
  • 292. Li J., Chen X., Lu A., and Liang C., “Targeted Protein Degradation in Cancers: Orthodox PROTACs and Beyond,” Innovation (Cambridge (Mass)) 4, no. 3 (2023): 100413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293. Saca V. R., Huber T., and Sakmar T. P., “G Protein‐coupled Receptor–targeted Proteolysis‐targeting Chimeras in Cancer Therapeutics,” Molecular Pharmacology 107, no. 2 (2025): 100013. [DOI] [PubMed] [Google Scholar]
  • 294. Ishida T., Joh T., Uike N., et al., “Defucosylated Anti‐CCR4 Monoclonal Antibody (KW‐0761) for Relapsed Adult T‐cell Leukemia‐lymphoma: A Multicenter Phase II Study,” Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 30, no. 8 (2012): 837–842. [DOI] [PubMed] [Google Scholar]
  • 295. Garg S., Vij M., Edward N., and Vij B., “Erenumab: A Novel Calcitonin Gene‐related Peptide Receptor Antagonist Developed Specifically for Migraine Prevention,” Journal of Anaesthesiology, Clinical Pharmacology 36, no. 1 (2020): 104–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Kang W., Ding C., Zheng D., et al., “Nanobody Conjugates for Targeted Cancer Therapy and Imaging,” Technology in Cancer Research & Treatment 20 (2021): 15330338211010117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297. Manglik A., Kobilka B. K., and Steyaert J., “Nanobodies to Study G Protein‐Coupled Receptor Structure and Function,” Annual Review of Pharmacology and Toxicology 57 (2017): 19–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298. Liu L., Tu B., Sun Y., et al., “Nanobody‐based Drug Delivery Systems for Cancer Therapy,” Journal of Controlled Release: Official Journal of the Controlled Release Society 381 (2025): 113562. [DOI] [PubMed] [Google Scholar]
  • 299. De Groof T. W. M., Mashayekhi V., Fan T. S., et al., “Nanobody‐Targeted Photodynamic Therapy Selectively Kills Viral GPCR‐Expressing Glioblastoma Cells,” Molecular Pharmaceutics 16, no. 7 (2019): 3145–3156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300. Bao C., Gao Q., Li L.‐L., et al., “The Application of Nanobody in CAR‐T Therapy,” Biomolecules 11, no. 2 (2021): 238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301. Zhang J., Feng H., Xu S., and Feng P., “Hijacking GPCRs by Viral Pathogens and Tumor,” Biochemical Pharmacology 114 (2016): 69–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. van Senten J. R., Fan T. S., Siderius M., and Smit M. J., “Viral G Protein‐coupled Receptors as Modulators of Cancer Hallmarks,” Pharmacological Research 156 (2020): 104804. [DOI] [PubMed] [Google Scholar]
  • 303. Schalop L. and Allen J., GPCRs, Desirable Therapeutic Targets in Oncology (Drug Discovery and Development, 2017). [Google Scholar]
  • 304. Lorente J. S., Sokolov A. V., Ferguson G., Schiöth H. B., Hauser A. S., and Gloriam D. E., “GPCR Drug Discovery: New Agents, Targets and Indications,” Nature Reviews Drug Discovery (2025). [DOI] [PubMed] [Google Scholar]
  • 305. Usman S., Khawer M., Rafique S., Naz Z., and Saleem K., “The Current Status of Anti‐GPCR Drugs Against Different Cancers,” Journal of Pharmaceutical Analysis 10, no. 6 (2020): 517–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306. McBrien C. and O'Connell D. J., “The Use of Biologics for Targeting GPCRs in Metastatic Cancers,” Biotechniques 14, no. 1 (2025): 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307. Perez Almeria C. V., Setiawan I. M., Siderius M., and Smit M. J., “G Protein‐coupled Receptors as Promising Targets in Cancer,” Current Opinion in Endocrine and Metabolic Research 16 (2021): 119–127. [Google Scholar]
  • 308. Lin S. J., Wu Z. R., Cao L., et al., “Pituitary Tumor Suppression by Combination of Cabergoline and Chloroquine,” The Journal of Clinical Endocrinology & Metabolism 102, no. 10 (2017): 3692–3703. [DOI] [PubMed] [Google Scholar]
  • 309. Caplin M. E., Pavel M., Ćwikła J. B., et al., “Lanreotide in Metastatic Enteropancreatic Neuroendocrine Tumors,” The New England Journal of Medicine 371, no. 3 (2014): 224–233. [DOI] [PubMed] [Google Scholar]
  • 310. Van Poppel H., Tombal B., de la Rosette J. J., Persson B.‐E., Jensen J.‐K., and Kold Olesen T., “Degarelix: A Novel Gonadotropin‐Releasing Hormone (GnRH) Receptor Blocker—Results From a 1‐yr, Multicentre, Randomised, Phase 2 Dosage‐Finding Study in the Treatment of Prostate Cancer,” European Urology 54, no. 4 (2008): 805–815. [DOI] [PubMed] [Google Scholar]
  • 311. De Clercq E., “Mozobil® (Plerixafor, AMD3100), 10 Years After Its Approval by the US Food and Drug Administration,” Antiviral Chemistry & Chemotherapy 27 (2019): 2040206619829382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312. Lor K. W., Helmons P. J., Belew H., Lane J. R., and Ball E. D., “Plerixafor as First‐ and Second‐line Strategies for Autologous Stem Cell Mobilization in Patients With non‐Hodgkin's Lymphoma or Multiple Myeloma,” Pharmacotherapy 32, no. 7 (2012): 596–603. [DOI] [PubMed] [Google Scholar]
  • 313. Fellner C., “Vismodegib (erivedge) for Advanced Basal Cell Carcinoma,” P & T: A Peer‐Reviewed Journal for Formulary Management 37, no. 12 (2012): 670–682. [PMC free article] [PubMed] [Google Scholar]
  • 314. O'Donnell E. F., Koch D. C., Bisson W. H., Jang H. S., and Kolluri S. K., “The Aryl Hydrocarbon Receptor Mediates Raloxifene‐induced Apoptosis in Estrogen Receptor‐negative Hepatoma and Breast Cancer Cells,” Cell Death & Disease 5, no. 1 (2014): e1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315. Strosberg J., El‐Haddad G., Wolin E., et al., “Phase 3 Trial of (177)Lu‐Dotatate for Midgut Neuroendocrine Tumors,” The New England Journal of Medicine 376, no. 2 (2017): 125–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316. de Lartigue J., “Mogamulizumab for the Treatment of Adult T‐cell Leukemia/Lymphoma,” Drugs of Today (Barcelona, Spain: 1998) 48, no. 10 (2012): 655–660. [DOI] [PubMed] [Google Scholar]
  • 317. Klotz L., Boccon‐Gibod L., Shore N. D., et al., “The Efficacy and Safety of degarelix: A 12‐month, Comparative, Randomized, Open‐label, Parallel‐group Phase III Study in Patients With Prostate Cancer,” BJU International 102, no. 11 (2008): 1531–1538. [DOI] [PubMed] [Google Scholar]
  • 318. Pulte E. D., Norsworthy K. J., Wang Y., et al., “FDA Approval Summary: Gilteritinib for Relapsed or Refractory Acute Myeloid Leukemia With a FLT3 Mutation,” Clinical Cancer Research: an Official Journal of the American Association for Cancer Research 27, no. 13 (2021): 3515–3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319. Das S., Al‐Toubah T., El‐Haddad G., and Strosberg J., “(177)Lu‐DOTATATE for the Treatment of Gastroenteropancreatic Neuroendocrine Tumors,” Expert Review of Gastroenterology & Hepatology 13, no. 11 (2019): 1023–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. Walters R. W., Shukla A. K., Kovacs J. J., et al., “beta‐Arrestin1 Mediates Nicotinic Acid‐induced Flushing, but Not Its Antilipolytic Effect, in Mice,” The Journal of Clinical Investigation 119, no. 5 (2009): 1312–1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321. Kamanna V. S., Ganji S. H., and Kashyap M. L., “The Mechanism and Mitigation of Niacin‐induced Flushing,” International Journal of Clinical Practice 63, no. 9 (2009): 1369–1377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322. DeWire S. M. and Violin J. D., “Biased Ligands for Better Cardiovascular Drugs: Dissecting G‐protein‐coupled Receptor Pharmacology,” Circulation Research 109, no. 2 (2011): 205–216. [DOI] [PubMed] [Google Scholar]
  • 323. Lauring B., Taggart A. K., Tata J. R., et al., “Niacin Lipid Efficacy Is Independent of both the Niacin Receptor GPR109A and Free Fatty Acid Suppression,” Science Translational Medicine 4, no. 148 (2012): 148ra115. [DOI] [PubMed] [Google Scholar]
  • 324. Bohn L. M., Lefkowitz R. J., Gainetdinov R. R., Peppel K., Caron M. G., and Lin F. T., “Enhanced Morphine Analgesia in Mice Lacking Beta‐arrestin 2,” Science (New York, NY) 286, no. 5449 (1999): 2495–2498. [DOI] [PubMed] [Google Scholar]
  • 325. Bohn L. M., Gainetdinov R. R., Lin F. T., Lefkowitz R. J., and Caron M. G., “Mu‐opioid Receptor Desensitization by Beta‐arrestin‐2 Determines Morphine Tolerance but Not Dependence,” Nature 408, no. 6813 (2000): 720–723. [DOI] [PubMed] [Google Scholar]
  • 326. Bohn L. M., Dykstra L. A., Lefkowitz R. J., Caron M. G., and Barak L. S., “Relative Opioid Efficacy Is Determined by the Complements of the G Protein‐coupled Receptor Desensitization Machinery,” Molecular Pharmacology 66, no. 1 (2004): 106–112. [DOI] [PubMed] [Google Scholar]
  • 327. Raehal K. M. and Bohn L. M., “The Role of Beta‐arrestin2 in the Severity of Antinociceptive Tolerance and Physical Dependence Induced by Different Opioid Pain Therapeutics,” Neuropharmacology 60, no. 1 (2011): 58–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328. Raehal K. M., Walker J. K., and Bohn L. M., “Morphine Side Effects in Beta‐arrestin 2 Knockout Mice,” The Journal of Pharmacology and Experimental Therapeutics 314, no. 3 (2005): 1195–1201. [DOI] [PubMed] [Google Scholar]
  • 329. Bohn L. M. and Raehal K. M., “Opioid Receptor Signaling: Relevance for Gastrointestinal Therapy,” Current Opinion in Pharmacology 6, no. 6 (2006): 559–563. [DOI] [PubMed] [Google Scholar]
  • 330. Groer C. E., Tidgewell K., Moyer R. A., et al., “An Opioid Agonist That Does Not Induce Mu‐opioid Receptor–arrestin Interactions or Receptor Internalization,” Molecular Pharmacology 71, no. 2 (2007): 549–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331. Lamb K., Tidgewell K., Simpson D. S., Bohn L. M., and Prisinzano T. E., “Antinociceptive Effects of Herkinorin, a MOP Receptor Agonist Derived From Salvinorin A in the Formalin Test in Rats: New Concepts in Mu Opioid Receptor Pharmacology: From a Symposium on New Concepts in Mu‐opioid Pharmacology,” Drug and Alcohol Dependence 121, no. 3 (2012): 181–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332. Hollenberg M. D. and Compton S. J., “International Union of Pharmacology. XXVIII. Proteinase‐activated Receptors,” Pharmacological Reviews 54, no. 2 (2002): 203–217. [DOI] [PubMed] [Google Scholar]
  • 333. Boire A., Covic L., Agarwal A., Jacques S., Sherifi S., and Kuliopulos A., “PAR1 is a Matrix Metalloprotease‐1 Receptor That Promotes Invasion and Tumorigenesis of Breast Cancer Cells,” Cell 120, no. 3 (2005): 303–313. [DOI] [PubMed] [Google Scholar]
  • 334. Kamath L., Meydani A., Foss F., and Kuliopulos A., “Signaling From Protease‐activated Receptor‐1 Inhibits Migration and Invasion of Breast Cancer Cells,” Cancer Research 61, no. 15 (2001): 5933–5940. [PubMed] [Google Scholar]
  • 335. Yang E., Boire A., Agarwal A., et al., “Blockade of PAR1 Signaling With Cell‐penetrating Pepducins Inhibits Akt Survival Pathways in Breast Cancer Cells and Suppresses Tumor Survival and Metastasis,” Cancer Research 69, no. 15 (2009): 6223–6231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336. Even‐Ram S., Uziely B., Cohen P., et al., “Thrombin Receptor Overexpression in Malignant and Physiological Invasion Processes,” Nature Medicine 4, no. 8 (1998): 909–914. [DOI] [PubMed] [Google Scholar]
  • 337. Even‐Ram S. C., Maoz M., Pokroy E., et al., “Tumor Cell Invasion Is Promoted by Activation of Protease Activated Receptor‐1 in Cooperation With the Alpha Vbeta 5 Integrin,” The Journal of Biological Chemistry 276, no. 14 (2001): 10952–10962. [DOI] [PubMed] [Google Scholar]
  • 338. Martin C. B., Mahon G. M., Klinger M. B., et al., “The Thrombin Receptor, PAR‐1, Causes Transformation by Activation of Rho‐mediated Signaling Pathways,” Oncogene 20, no. 16 (2001): 1953–1963. [DOI] [PubMed] [Google Scholar]
  • 339. Yin Y. J., Salah Z., Maoz M., et al., “Oncogenic Transformation Induces Tumor Angiogenesis: A Role for PAR1 Activation,” FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology 17, no. 2 (2003): 163–174. [DOI] [PubMed] [Google Scholar]
  • 340. Yang E., Cisowski J., Nguyen N., et al., “Dysregulated Protease Activated Receptor 1 (PAR1) Promotes Metastatic Phenotype in Breast Cancer Through HMGA2,” Oncogene 35, no. 12 (2016): 1529–1540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341. Ghio P., Cappia S., Selvaggi G., et al., “Prognostic Role of Protease‐activated Receptors 1 and 4 in Resected Stage IB Non‐small‐cell Lung Cancer,” Clinical Lung Cancer 7, no. 6 (2006): 395–400. [DOI] [PubMed] [Google Scholar]
  • 342. Cohen M. H., Gootenberg J., Keegan P., and Pazdur R., “FDA Drug Approval Summary: Bevacizumab (Avastin) plus Carboplatin and Paclitaxel as First‐line Treatment of Advanced/Metastatic Recurrent Nonsquamous Non‐small Cell Lung Cancer,” The Oncologist 12, no. 6 (2007): 713–718. [DOI] [PubMed] [Google Scholar]
  • 343. Cisowski J., O'Callaghan K., Kuliopulos A., et al., “Targeting Protease‐activated Receptor‐1 With Cell‐penetrating Pepducins in Lung Cancer,” The American Journal of Pathology 179, no. 1 (2011): 513–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344. Foley C. J. and Kuliopulos A., “Mouse Matrix Metalloprotease‐1a (Mmp1a) Gives New Insight Into MMP Function,” Journal of Cellular Physiology 229, no. 12 (2014): 1875–1880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345. Agarwal A., Covic L., Sevigny L. M., et al., “Targeting a Metalloprotease‐PAR1 Signaling System With Cell‐penetrating Pepducins Inhibits Angiogenesis, Ascites, and Progression of Ovarian Cancer,” Molecular Cancer Therapeutics 7, no. 9 (2008): 2746–2757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346. Boland A. J., O'Kane A. A., Buick R., Longley D. B., and Scott C. J., “Antibody Therapy in Pancreatic Cancer: MAb‐ye We're Onto Something?,” Biochimica Et Biophysica Acta (BBA)—Reviews on Cancer 1876, no. 1 (2021): 188557. [DOI] [PubMed] [Google Scholar]
  • 347. Booker B. E., Steg A. D., Kovac S., Landen C. N., and Amm H. M., “The Use of Hedgehog Antagonists in Cancer Therapy: A Comparison of Clinical Outcomes and Gene Expression Analyses,” Cancer Biology & Therapy 21, no. 10 (2020): 873–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348. Hehlgans S., Booms P., Güllülü Ö., et al., “Radiation Sensitization of Basal Cell and Head and Neck Squamous Cell Carcinoma by the Hedgehog Pathway Inhibitor Vismodegib,” International Journal of Molecular Sciences 19, no. 9 (2018): 2485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349. Ogura M., Ishida T., Hatake K., et al., “Multicenter Phase II Study of mogamulizumab (KW‐0761), a Defucosylated Anti‐cc Chemokine Receptor 4 Antibody, in Patients With Relapsed Peripheral T‐cell Lymphoma and Cutaneous T‐cell Lymphoma,” Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 32, no. 11 (2014): 1157–1163. [DOI] [PubMed] [Google Scholar]
  • 350. Miller K., Moul J. W., Gleave M., et al., “Phase III, Randomized, Placebo‐controlled Study of Once‐daily Oral Zibotentan (ZD4054) in Patients With Non‐metastatic Castration‐resistant Prostate Cancer,” Prostate Cancer and Prostatic Diseases 16, no. 2 (2013): 187–192. [DOI] [PubMed] [Google Scholar]
  • 351. Younis I. R., George D. J., McManus T. J., et al., “Clinical Pharmacology of an Atrasentan and Docetaxel Regimen in Men With Hormone‐refractory Prostate Cancer,” Cancer Chemotherapy and Pharmacology 73, no. 5 (2014): 991–997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352. Kaye S. B., Fehrenbacher L., Holloway R., et al., “A Phase II, Randomized, Placebo‐controlled Study of vismodegib as Maintenance Therapy in Patients With Ovarian Cancer in Second or Third Complete Remission,” Clinical Cancer Research: an Official Journal of the American Association for Cancer Research 18, no. 23 (2012): 6509–6518. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The authors have nothing to report.


Articles from MedComm are provided here courtesy of Wiley

RESOURCES