Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Feb 1.
Published in final edited form as: Cell Signal. 2020 Nov 27;78:109862. doi: 10.1016/j.cellsig.2020.109862

Biased Agonism at Chemokine Receptors

Dylan Scott Eiger a,b, Noelia Boldizsar a, Christopher Cole Honeycutt a, Julia Gardner a, Sudarshan Rajagopal a,b
PMCID: PMC7775275  NIHMSID: NIHMS1650054  PMID: 33249087

Abstract

In the human chemokine system, interactions between the approximately 50 known endogenous chemokine ligands and 20 known chemokine receptors (CKRs) regulate a wide range of cellular functions and biological processes including immune cell activation and homeostasis, development, angiogenesis, and neuromodulation. CKRs are a family of G protein-coupled receptors (GPCR), which represent the most common and versatile class of receptors in the human genome and the targets of approximately one third of all Food and Drug Administration-approved drugs. Chemokines and CKRs bind with significant promiscuity, as most CKRs can be activated by multiple chemokines and most chemokines can activate multiple CKRs. While these ligand-receptor interactions were previously regarded as redundant, it is now appreciated that many chemokine:CKR interactions display biased agonism, the phenomenon in which different ligands binding to the same receptor signal through different pathways with different efficacies, leading to distinct biological effects. Notably, these biased responses can be modulated through changes in ligand, receptor, and or the specific cellular context (system). In this review, we explore the biochemical mechanisms, functional consequences, and therapeutic potential of biased agonism in the chemokine system. An enhanced understanding of biased agonism in the chemokine system may prove transformative in the understanding of the mechanisms and consequences of biased signaling across all GPCR subtypes and aid in the development of biased pharmaceuticals with increased therapeutic efficacy and safer side effect profiles.

Keywords: Biased agonism, G protein-coupled receptors, Chemokine System

Chemokine System

Chemokine receptors (CKRs) are a subfamily of G protein-coupled receptors (GPCRs) that bind a group of small (8–12 kDa) and highly conserved chemotactic cytokines known as chemokines (1). The human chemokine system is composed of approximately 20 known CKRs and 50 known chemokines (Figure 1). The chemokines are classified into four subtypes (C, CC, CXC, CX3C) based on the number, positioning, and spacing of conserved N-terminal cysteine residues (2). Similarly, CKRs are organized and classified according to the ligands they bind (3). Chemokines are also categorized as homeostatic chemokines, which are constitutively expressed in a variety of specific tissues and cell types, and inflammatory chemokines, which are induced during immune responses primarily to recruit leukocytes to sites of inflammation (4). Homeostatic and inflammatory classifications of chemokines are not mutually exclusive, however, as some CKRs and chemokine ligands are involved in both normal and pathologic conditions. The role of CKR activation by chemokines was first recognized in the immune response, specifically as chemoattractants to direct leukocyte migration, a process known as chemotaxis (5, 6). While the functions and roles of chemokines in leukocytes are well known, it is now appreciated that chemokines and CKRs are also produced in a variety of non-leukocyte cell types, including epithelial cells, fibroblasts, endothelial cells, and neurons (7), and play a key role in a wide range of other cellular functions and biological processes including development, angiogenesis, neuromodulation, and immune cell homeostasis (811). For example, the expression of neuronal chemokine ligands and receptors has recently been shown to be involved in synaptic transmission and neuronal survival (12), as well as in guidance of central nervous system (CNS) cellular interactions via neuron-astrocyte, neuron-microglia, and neuron-neuron interactions (13).

FIGURE 1: The complexity of the human chemokine system.

FIGURE 1:

Chemokine receptors fall into five categories: CCRs, CXCRs, ACKRs, XCRs and CX3CRs. Chemokine ligands fall into four categories: CCLs, CXCLs, XCLs, CX3CLs. Lines connecting chemokine receptors to chemokines are colored for clarity.

Due to the chemokine system’s involvement in a wide variety of biological processes, it is unsurprising that chemokines and CKRs are implicated in various disease states including, but not limited to, autoimmune disorders, infectious diseases, hypersensitivity reactions, atherosclerosis, and cancer (1418). The role of the chemokine system in chronic inflammatory diseases is particularly important and chemokines play a central role in asthma, chronic obstructive pulmonary disease, inflammatory bowel disease, arthritis, multiple sclerosis, and psoriasis (19). Additionally, certain disorders are directly associated with mutations in the genes that encode CKRs, such as the Warts, Hypogammaglobulinemia, Immunodeficiency, and Myelokathexis (WHIM) Syndrome which is driven by an autosomal dominant truncation mutant in the receptor CXCR4 (20).

While the chemokine system is known to play a significant role in many disease states, there are relatively few drugs that target it directly. Chemokines and CKRs bind with significant promiscuity, wherein most CKRs can be activated by multiple chemokine ligands and most chemokines can activate multiple CKRs (21). This promiscuity was thought to lead to “redundancy” between chemokines and their receptors, serving as a mechanism for a robust physiologic response (22). As adequate chemokine levels are imperative for immune cell function, redundant chemokine signaling would provide sufficient signals to direct leukocyte chemotaxis and function that is relatively insensitive to variations in the concentration of any individual chemokine (23). However, we now appreciate that many of these ligands can have distinct signaling profiles at the same receptor and many receptors can have distinct signaling profiles when stimulated by the same ligand, a phenomenon referred to as “biased agonism” (22, 24). Biased signaling through differences in ligands, receptors and the cellular context (system) can have important effects on chemokine signaling and implications for drug development. Here, we review the current literature on biased signaling within the chemokine system to highlight the complex and multidimensional nature of biased agonism and the biochemical mechanisms that underlie it, the importance of biased agonism within the chemokine system and its physiologic effects, and the implications of biased signaling for drug development in the chemokine system.

G Protein-Coupled Receptor Signaling and Biased Agonism

CKRs are a subfamily of the rhodopsin class of GPCRs, the most common and versatile superfamily of receptors in the human genome (25) and the target of ~34% of all Food and Drug Administration (FDA) approved pharmaceutical drugs (26). Canonical GPCR signaling starts with agonist binding, upon which a GPCR undergoes conformational changes that induce the recruitment of heterotrimeric G proteins consisting of Gα, Gβ, and Gγ subunits. The guanosine diphosphate (GDP)-bound Gα subunit undergoes nucleotide exchange for guanosine triphosphate (GTP), leading to Gα activation and dissociation of the heterotrimeric complex into its Gα and Gβγ constituents (27). The Gα subunits are classified into four families based on sequence similarity: Gαs, Gαi/o, Gαq/11, and Gα12/13 (28). The activated Gα proteins typically regulate the production and subsequent signaling of secondary messengers, such as adenosine 3’,5’-cyclic monophosphate (cAMP), intracellular calcium, and inositol triphosphate (29). Most chemokine receptors signal through Gαi/o, which inhibits adenylyl cyclase and reduces intracellular concentrations of cAMP (30). There are various isoforms of the Gβ and Gγ subunits, and at chemokine receptors the Gβγ dimer has been shown to activate phosphoinositide-specific phospholipase Cβ (PLC) and phosphoinositide 3-kinase (PI3K). PLC then produces diacylglycerol (DAG), leading to the activation of protein kinase C (PKC), and inositol-triphosphate (IP3), which triggers calcium mobilization (31). The signaling messengers of the Gβγ dimer in chemokine receptors have been demonstrated to play a role in the promotion of leukocyte migration, among other functions (32).

Following G protein activation, G protein-coupled receptor kinases (GRKs) are recruited to the receptor and phosphorylate the receptor C-terminus and intracellular loops. This phosphorylation promotes the interaction of the receptor with the β-arrestins, which were first described for their function in the desensitization of G protein-mediated signaling through steric hindrance of the receptor (33). β-arrestins act as multifunctional adaptor proteins and are involved in a variety of other cellular process such as receptor internalization, transactivation, trafficking, and signaling (34). β-arrestins are capable of signaling through a variety of mediators including mitogen-activated protein kinases (MAPKs), nuclear factor κB (NF-κB), protein kinase B (Akt), and Src tyrosine kinase, thereby activating distinct signaling pathways independent of G protein-mediated signaling (3539). At some chemokine receptors, such as CXCR4 and CCR5, it has even been shown that in the absence of G protein coupling, β-arrestin-bound CKR complexes are still able to induce receptor endocytosis and signaling (40).

Biased Chemokine Signaling

With the ability of GPCRs to signal through multiple pathways such as different G proteins or β-arrestins, referred to as “pluridimensional efficacy” (41), it was soon discovered that many agonists were capable of signaling with different efficacies to their downstream effectors (42). Initially these examples were thought to be rare, but studies over the past fifteen years have demonstrated that, across a wide variety of GPCR subtypes, different ligands for the same receptor can activate specific and distinct signaling pathways downstream. These examples can range from mild bias, with slight differences in preferences for different G proteins, to completely biased ligands which preferentially activate G protein signaling pathways (G protein-biased) while not activating β-arrestin signaling (β-arrestin-biased) pathways, or vice versa (Figure 2AC).

FIGURE 2: Biased agonism at G Protein-Coupled Receptors.

FIGURE 2:

Signaling at G Protein-Coupled Receptors can be driven by ligands that are (A) balanced and equally activate both G protein and β-arrestins, (B) those that signal primarily through heterotrimeric G proteins (G protein-biased), or (C) those that signal primarily through β-arrestin adapter proteins (β-arrestin-biased). Biased agonism can be achieved through (D) biased ligands, (E) biased receptors, or (F) biased systems

Biased agonism within the chemokine subfamily was first described at CCR7, where it was demonstrated that the receptor’s two endogenous ligands—CCL19 and CCL21—both could activate G protein-dependent signaling cascades but only CCL19 was capable of activating β-arrestin dependent signaling (4346). Since then, numerous studies have demonstrated that the chemokine system is not redundant, but rather highly specific in its cellular outputs, part of which can be attributed to biased agonism (39). Functional and structural demonstrations of bias in chemokine signaling provide support for the notion that the promiscuity of the chemokine system is not redundant, but suggests high levels of signaling specificity by allowing individual chemokines and CKRs to induce functionally distinct biochemical, cellular, and physiologic outcomes (24). Biased agonism in the chemokine system can be a result of signaling through each component of the GPCR ternary complex: the ligand, the receptor, and the transducer (“system”) elements (Figure 2DF). With ligand bias, different ligands binding to the same receptor generate distinct responses. With receptor bias, the same ligand binding to different receptors generates distinct responses. With system bias, a different cellular context, e.g., differential transducer expression in a different cell type, results in a change in GPCR signaling. While the exact mechanisms and functional consequences of these layers of bias are incompletely understood, it is possible that this complexity allows for highly specific and coordinated physiologic responses. Additionally, the interplay between these individual sources of biased agonism can theoretically generate a multitude of permutations of immunophenotypes.

Ligand Bias

Ligand bias is arguably the most widely studied mechanism of bias in the chemokine system. Ligand bias refers to the phenomenon in which a molecule binds to a receptor and promotes an ensemble of receptor conformations that preferentially interact with some transducers over others (39). For example, the two endogenous ligands of CCR7, CCL19 and CCL21, induce G protein activation and signaling to similar extents, but CCL19 induces β-arrestin recruitment to CCR7 significantly more than CCL21 (4446). Additionally, receptor internalization and chemotaxis induced by CCL19, but not CCL21, are reduced following siRNA knock down of β-arrestin 2 and both β-arrestin 1 and β-arrestin 2, respectively (46). Other assays assessing β-arrestin trafficking, receptor desensitization, MAPK signaling, and receptor phosphorylation are significantly different following agonist stimulation with CCL19 or CCL21 (44), consistent with CCL19 acting as a β-arrestin biased ligand relative to CCL21. Similar to the ligands of CCR7, the endogenous ligands of CXCR3 also exhibit ligand bias. In previous work performed by our group, these endogenous ligands exhibit both varying levels and distinct patterns of interactions with β-arrestin (24). When stimulating CXCR3A, CXCL11 recruits β-arrestin at a significantly higher level compared to CXCL10, even though the two ligands activate Gαi to similar levels (47). Furthermore, CXCL11 demonstrates a class B pattern of β-arrestin recruitment, one of which promotes a stable β-arrestin:GPCR complex with trafficking of β-arrestin into endosomes, while CXCL9 and CXCL10 demonstrate a class A pattern of recruitment, which promotes a transient β-arrestin:GPCR complex limited to the plasma membrane (24). Multiple chemokine receptors and their endogenous ligands demonstrate similar degrees of biased agonism, including CCR1, CCR5, CCR10, CXCR1, and CXCR2 (24, 43, 4852).

Ligand bias has also been demonstrated to depend on the oligomeric state of chemokines. In vivo, chemokines homo-oligomerize and exist in an equilibrium between their monomeric and dimeric forms (53), with the monomer frequently serving as the active form that binds the receptor and the dimeric form bound to glycosaminoglycans (54). Changes in the overall oligomeric state of chemokines are critical for their cellular functions, reflected by the differential signaling profiles between their monomeric and dimeric forms (5558). Drury et al. found that at CXCR4 the monomeric form of CXCL12 is β-arrestin biased relative to its dimeric form, and this bias mediates increased levels of chemotaxis and differential MAPK activation kinetics (55). Modulation of the monomer-dimer equilibrium in vivo may consequently serve as another method for a system to finely tune chemokine signaling. Another mechanism that can regulate chemokine activity is post-translational modification, such as proteolytic processing, citrullination and glycosylation (59). While these modifications have been shown to regulate chemokine activity, it is largely unknown at this time as to whether they have an effect on ligand bias.

Beyond simple G protein vs β-arrestin bias, there are examples of bias between specific G protein subtypes or β-arrestin isoforms (60, 61). Various analogs of CCL5 analyzed by Lorenzen et al. at CCR5 signal through Gαi/o with similar efficacies, whereas they vary widely in their ability to signal through Gαq, suggesting that bias between G protein subtypes may affect signaling at receptors able to couple to multiple downstream transducer elements (60). While investigation into bias between Gα subtypes is difficult due to cross-talk between different G protein signaling pathways, bias between different G proteins presents an added layer of complexity to ligand bias in the chemokine system.

Receptor Bias

Receptor bias refers to a receptor that demonstrates preferential interaction with certain transducers over others (39). ACKR3, also known as CXCR7, was originally characterized as a “decoy” receptor that served to modulate CXCR4 signaling by either sequestering excess CXCL12 (as both ACKR3 and CXCR4 can bind CXCL12) or serving as a coreceptor with CXCR4, rather than activating specific downstream signaling pathways (6265). However, it was later discovered that ACKR3 not only acted as a decoy receptor, but also was capable of independently signaling via β-arrestins (66). CXCR4 and ACKR3 signal through different pathways despite being activated by the same ligand, with CXCR4 activating both G proteins and β-arrestins and ACKR3 signaling only via β-arrestins; therefore ACKR3 acts as a β-arrestin biased receptor relative to CXCR4 (67). Additional studies have shown that ACKR3 also serves as a receptor for endogenous opioids, contributing to circadian glucocorticoid oscillations (68) and acting as a decoy for opioids in the brain (69). Interestingly, recent work has demonstrated that β-arrestin recruitment to ACKR3 is dispensable for some receptor functionality. Montpas et al. show that both siRNA knockdown of β-arrestin 1 and or β-arrestin2 in HEK293 cells does not impact ACKR3 mediated degradation of CXCL11 or CXCL12 (70). They repeated these studies in β-arrestin 1 and 2 knockout murine embryonic fibroblasts (MEFs) and also saw that ACKR3 chemokine scavenging was unaffected. Similarly, Saaber et al. demonstrate that ACKR3 chemokine scavenging and endocytosis is dependent on receptor phosphorylation but independent of β-arrestins. Utilizing a variety of transgenic mice, they further demonstrate that β-arrestin is dispensable and receptor phosphorylation is indispensable in ACKR3 mediated migration of cortical interneurons (71). These findings demonstrate that ACKR3, while technically a β-arrestin-biased receptor as it does not signal via heterotrimeric G proteins, has functionality independent of the β-arrestins, highlighting the complexity of biased agonism at GPCRs.

Alternative splicing of GPCR transcripts further increases receptor diversity and the potential for receptor bias (72). Highly truncated receptor splice variants with only three transmembrane domains have been shown to heterodimerize with their wild type counterparts and retain them in the endoplasmic reticulum, while longer splice variants with a preserved seven transmembrane architecture have been shown to promote signaling via distinct signal transduction pathways (72). The isoforms of CXCR3 present an example of how alternative splicing can lead to endogenous receptor bias (47, 73). CXCR3 has three splice variants: CXCR3A, CXCR3B, and CXCR3-alt (73). CXCR3A and CXCR3B have nearly identical sequences, with the only difference being that CXCR3B possesses an extra 51 amino acids on its extracellular N terminal domain (73). CXCR3alt has both truncated forms of the third intracellular loop and transmembrane helices 6 and 7, resulting in a highly truncated 5 transmembrane receptor (73). CXCR3alt does not recruit β-arrestin or stimulate a Gαi response, but promotes modest extracellular signal-regulated kinase (ERK) phosphorylation and becomes internalized following ligand stimulation (73). The two functional CXCR3 splice variants CXCR3A and CXCR3B have significantly biased responses from one another (47, 73). CXCR3B is β-arrestin-biased relative to CXCR3A, as the two isoforms similarly recruit β-arrestin upon stimulation with CXCL11 but CXCR3A signals through Gαi to a significantly greater extent (47). Their specific interactions with β-arrestin also differ, as CXCR3A’s interaction with β-arrestin demonstrates a stable class B receptor pattern while CXCR3B’s demonstrates a transient interaction, consistent with class A receptor behavior (47). Smith et al. also found differences in receptor kinetics and mechanisms driving ERK phosphorylation and receptor internalization. Finally, the two variants display differential ligand induced transcriptional activity, as stimulation of CXCR3A, but not CXCR3B, with CXCL11 resulted in a robust response using serum response element (SRE) and serum response factor-response element (SRF) transcriptional reporters. These and other examples suggest that there are a number of finely tuned mechanisms that allow receptor bias to differentially regulate the response to chemokines.

System Bias

System bias is primarily regulated through the differential expression or function of receptor and transducer elements, which consequentially promote biased cellular outputs (39). These include every element of the signal transduction cascade, from proximal effectors such as GRKs, G proteins and β-arrestins, to other proteins that regulate second messenger responses, such as Regulator of G Protein Signaling (RGS) and Activator of G Protein Signaling (AGS) proteins, to all of the downstream signal transduction machinery. This phenomenon manifests as observed differences in signaling events between cell types. For example, the functional effects of mutations on CXCR6 receptor function and selectivity of Gi/o proteins are cell-type specific (74). In an F128Y mutant of CXCR6, the receptor’s dependence on Go proteins was diminished in HEK-293T cells, but resembled that of the wild type receptor in Jurkat E6–1 cells (74). Outside of the chemokine system, it has been shown that β-arrestin biased agonists for the dopamine receptor D2 demonstrate different signaling effects in the striatum and prefrontal cortex due to differential expression of GRK2 and β-arrestin 2 (75). While, to our knowledge, system bias in the chemokine system has not been rigorously and explicitly studied, changes in GRK expression have been proven to alter the functionality of numerous chemokine receptors (76). For example, murine T cells with a 50% reduction of GRK2 levels experienced significantly more migration, calcium flux, protein kinase B activity, and ERK phosphorylation upon stimulation with chemokine ligands than in wild type cells (77). Similarly, Arnon et al. created GRK2f/− mice carrying a T cell-specific or a B cell specific cre to study the effects of GRK2 on S1P receptor-1 function in lymphocyte migration (78). They found that T and B cell movement from the blood into lymphatic tissues was reduced in the absence of GRK2, and that B cells between the splenic marginal zone and follicle were also reduced. These findings have physiologic implications, as GRK expression is differentially modulated in various disease states. For example, the expression levels of GRK2 and GRK5 are elevated in the lungs of rats treated with IL-1β, while levels of GRK2 and GRK6 are reduced in mice in an experimental autoimmune encephalomyelitis (EAE) animal model (79). Furthermore, downregulated GRK2 and GRK6 have been found in patients with autoimmune diseases such as multiple sclerosis and rheumatoid arthritis, and dynamic expression of β-arrestin 1 has been found in mouse models of both EAE and an adjuvant-induced arthritis (8084).

It is inherently difficult to accurately recapitulate the dynamic in vivo expression levels of transducer elements across various cell types in vitro. However, considering the diversity of chemokine receptors and immune cells within mammals, and the dynamic nature of GPCRs and GPCR transducer elements in inflammatory conditions, system bias likely adds a significant layer of complexity to the chemokine system’s biased functionality (76, 80).

Biochemical Mechanisms of Biased Agonism

As discussed above, the ternary complex of ligand, receptor, and transducer serves as a model for the components driving biased agonism (39). A final cellular output is highly dependent on (1) the ligand, (2) the receptor, and (3) the transducer elements in the context of the cellular system. These three elements are the ultimate facilitators of biased signaling, and understanding the structural and functional bases of their interactions is integral to understanding potential mechanisms of biased agonism.

Ligand-Receptor Interactions

At the most fundamental level, ligand binding promotes an ensemble of receptor conformations that promote differential transducer coupling, leading to the selective activation of specific signaling pathways over others. Recent crystal structures and cryoEM have changed our understanding of how chemokines bind to their receptors (85). In the classic two-site model for chemokine binding, a chemokine binds its receptor through two distinct chemokine recognition sites (CRS), CRS1 and CRS2. CRS1 consists of an interaction between the sulfated tyrosines of the receptor N-terminus with the chemokine globular core, and CRS2 consists of an interaction between the transmembrane regions of the receptor with the chemokine N-terminus. Crystal structures have demonstrated an intervening CRS1.5 between CRS1 and CRS2 at a conserved Pro-Cys of the CKR N-terminus against the conserved disulfide of the chemokines (86, 87).

Affecting CRS2 by mutating chemokine N termini has been shown to affect biased signaling responses at CCR1, with data demonstrating that this region has a role in stabilizing active receptor conformations that contribute to biased agonism (51). CCR1 is known to bind at least nine different endogenous chemokines including CCL7, CCL8, and CCL15. Sanchez et al. showed that CCL7 and CCL8 demonstrate bias towards Gαi signaling relative to CCL15(Δ26), an N-terminal truncated version of CCL15 that renders it approximately the same length as CCL7 and CCL8. To elucidate the structural basis of this bias, Sanchez et al. created a chimeric chemokine, CCL15(N-CCL7), which consists of CCL15 with the N terminal region of CCL7. Using radioligand binding, CCL15(N-CCL7) had a binding affinity for CCR1 nearly identical to CCL7 and significantly lower than CCL15Δ(26). When looking at G protein activation, CCL15 (N-CCL7) demonstrated a signaling profile very similar to that of CCL15(Δ26) which suggests that the N-terminal regions of CCL7 and CCL15(Δ26) are equally capable of driving G protein activation. However, CCL15 (N-CCL7) shows an intermediate ability to recruit β-arrestin when compared to CCL15(Δ26) and CCL7, suggesting that the N-terminal regions of CCL7 and CCL15(Δ26) contribute to a chemokines ability to recruit β-arrestin (51). Sanchez et al. also found that shortening CCL15Δ(26) by only two residues, CCL15Δ(28), did not affect the chemokines affinity for the receptor, but drastically reduced its maximum ability to recruit β-arrestin without affecting its potency. Concurrently, this shortening increased its potency but not maximum ability to inhibit cAMP accumulation. These data demonstrate the importance of a chemokine’s N-terminal length and sequence identity not only in binding affinity, but also in stabilizing receptor conformations that differentially interact with both G proteins and β-arrestins.

Research exploring the endogenous ligands of CXCR3 suggests that each ligand exhibits differential binding interactions with the receptor, further implicating the CRS2 interface in a biased response. Cox et al. used radioligand binding studies to suggest that CXCL9 and CXCL10 bind at a separate site on the receptor than CXCL11 (88). Colvin et al. performed mutagenic studies on CXCR3 and elucidated which residues were implicated in this differential chemokine binding (89). Whereas deletion of the 16 proximal N terminal amino acids of the receptor resulted in diminished calcium flux, actin polymerization, and chemotaxis upon stimulation with CXCL10 and CXCL11, activity stimulated by CXCL9 was not affected (89). Together, these seemingly discordant results highlight the diversity in activation mechanisms at a single receptor and the importance of ligand and receptor identity, as well as the interactions between ligand, receptor, and transducer elements in determining the overall final cellular output.

Different receptors can display different binding patterns to the same ligand, further pointing to how this interaction contributes to biased signaling. For example, while the receptor CXCR4 and β-arrestin biased receptor ACKR3 are both capable of binding CXCL12, mutagenic studies have revealed that the ligand-receptor interface of CXCR4 is significantly different than that of ACKR3 (90, 91). Jaracz-Ros et al. created a variety of CXCL12 mutants and demonstrated that substitution of N-terminal amino acids decreased receptor affinity at both CXCR4 and ACKR3. However, while N-terminal truncations of CXCL12 similarly led to decreased receptor affinity at CXCR4, these truncation mutants were unable to bind to ACKR3. Interestingly, CXCL12 substitution mutants displayed a decreased ability to drive receptor internalization at CXCR4 when compared to WT CXCL12 but had no impact on internalization at ACKR3 (90). Similarly, these substitution mutants demonstrate no ability to recruit β-arrestin to CXCR4, but show slightly decreased potency and efficacy in β-arrestin recruitment at ACKR3 relative to WT CXCL12. Notably, both ACKR3 and CXCR4 bind CXCL12 through the receptor distal N-terminus wrapping around the chemokine, forming a novel CRS0.5 interface (86, 92).

A recent cryoEM structure of CCL20 bound to CCR6 further illustrates that chemokine receptors possess extremely diverse mechanisms of activation. Numerous class A GPCRs have been shown to exhibit a deep binding pocket mode of activation that involves interactions with the transmembrane receptor core and proximity to the toggle switch motif, a tryptophan residue that has been shown to contribute to conformational changes upon receptor activation (93). In contrast, CCL20 binds in a long, shallow extracellular site distal from the toggle switch (93). CCL20 does not require interactions between its N terminus and the 7 transmembrane receptor core to activate CCR6, but instead requires major interactions with extracellular loop 2 and the N terminus of the receptor (93). Wasilko et al. speculate that this differential binding pattern is due to CCL20’s short N terminal region, which consists of only 5 amino acids. Work from Riutta et al. further supports this notion as they manipulated various aspects of CCL20’s short N terminal region and determined that CCL20 binding and activation of CCR6 is highly tolerant to changes in the length and identity of the N terminus (94). Other chemokines or ligands with shorter N termini could prove to exhibit binding patterns similar to CCL20, emphasizing the notion that chemokine binding is extremely diverse and that attempting to generalize chemokine ligand-receptor interactions to a single model may be impossible. The interactions between the chemokine ligands and receptors have thus been proven to be multi-site, complex, and dynamic.

Phosphorylation Barcode

Receptor activation via ligand binding leads to a conformational change in the receptor which translates into a cellular response first through G protein recruitment and activation, followed by phosphorylation of the intracellular loops and C terminus of the receptor. This phosphorylation leads to the recruitment of the β-arrestins, which abrogate G protein signaling and also initiate β-arrestin dependent signaling cascades. Phosphorylation patterns of the receptor can vary based on the identity of the ligand used to activate the receptor and the kinases present in the cell, and these differential phosphorylation patterns can promote engagement with distinct transducer elements, and consequently generate a unique signaling cascade (Figure 3). Numerous studies support this model, known as the “barcode hypothesis,” as one mechanistic explanation that underlies biased agonism (9598).

FIGURE 3: Phosphorylation barcode promotes differential downstream signaling.

FIGURE 3:

Distinct patterns of G Protein-Coupled Receptor phosphorylation can promote differential downstream signaling cascades. (A) A phosphorylation barcode which preferentially drives β-arrestin activation of with effector “A” while (B) demonstrates a phosphorylation barcode which preferentially drives β-arrestin mediated activation of effector “B”.

Formation of the phosphorylation barcode is mediated by a number of kinases, most notably G protein-coupled receptor kinases, or GRKs (99). There are seven GRK isoforms, but only GRK2, 3, 5, and 6 are expressed ubiquitously and are primarily relevant for a discussion of bias in the chemokine system (99). GRKs are selective in the residues they phosphorylate, implicating them in the complexity required for a biased response. Peptide studies reveal that different classes of GRKs have varying affinities for phosphorylating sites within different proximities of acidic or basic residues (100). For example, GRK5 and GRK6 preferentially phosphorylate peptides with basic amino acids near the N terminal side of the target residues (101, 102). Furthermore, GRKs have also been shown to phosphorylate specific residues on the C terminal tails of chemokine receptors (103). Upon stimulation with CXCL12, CXCR4 is rapidly phosphorylated by GRK6 at Ser-324/5 and Ser-339, and slowly phosphorylated by GRK6 at Ser-330 (103). Data also suggests that phosphorylation sites at CXCR4 are hierarchical and sequential in nature, in which a certain order of phosphorylation is necessary to induce a response (104). Therefore, the pattern of phosphorylation mediated by distinct GRKs on the receptor C-terminal tail appears to be specific and coordinated.

Genetic knockdown of GRKs has connected specific classes of GRKs to distinct cellular functions both in vitro and in genetically modified mice (44, 47, 95, 105, 106). At CXCR4, for example, GRK2 and GRK6 have been shown to negatively regulate calcium mobilization. Additionally, ERK1/2 phosphorylation is negatively regulated by GRK2, but positively regulated by GRK3 and GRK6 (103). Furthermore, GRK3 is responsible for the phosphorylation of the two most distal phosphorylation sites on the CXCR4 C terminal tail, which have been implicated in β-arrestin recruitment (106). Similarly, Zidar et al. found that, at CCR7, CCL19-mediated β-arrestin recruitment was reduced following siRNA knockdown of GRK3 and GRK6, while CCL21-mediated β-arrestin recruitment was only reduced following GRK6 knockdown (44). Additionally, GRK6 knockdown led to a significant decrease in ERK 1/2 phosphorylation at both chemokines, while GRK2 and GRK3 knockdown led to an increase in ERK 1/2 phosphorylation only with CCL19 (44). Instances of differential involvement by GRKs also occur between the aforementioned splice variants of CXCR3 (47). CXCR3B driven recruitment of β-arrestin involves GRK2 and GRK3 but not GRK5 nor GRK6, while that of CXCRA involves all four kinases (47). These data are consistent with a model in which different expression levels of these kinases can influence a biased response. The utilization of different GRK isoforms by biased ligands and receptors to produce a phosphorylation barcode could lead to functionally distinct pools of β–arrestins, thus yielding important insights into the mechanism of biased agonism.

Receptor-Transducer Interactions

Upon stimulation with a ligand, a GPCR recruits transducer elements to induce signaling cascades. Potential for bias rests in the receptor-transducer interactions in a similar fashion as the ligand-receptor interaction; receptors have the potential to stabilize interactions with or conformations of transducer elements that lead to distinct signaling responses. Recent structural studies have provided important insights into how such structural changes can be linked to receptor activity and biased responses. Crystal structures of negative allosteric modulators bound to the chemokine receptors CCR2 (107) and CCR9 (108) have demonstrated an allosteric binding site on the intracellular surface. This binding site involves several conserved amino acid residues and is a common intracellular binding site in class A GPCRs (109). Notably at the type 1 angiotensin II receptor, Wingler et al. found that G-protein biased agonists stabilize an “open” conformation of the intracellular surface of the receptor, while β-arrestin biased agonists stabilize a more closed receptor intracellular conformation (110). This suggests that biased agonists promote differential interactions with transducers through the generation of specific receptor intracellular conformations, which could potentially be targeted with novel allosteric modulators. However, at this time there is little structural information on the mechanisms underlying biased agonism by chemokine receptors, although there is a large body of work that have studied how receptor-transducer interactions contribute to biased signaling.

Colvin et al. found that the endogenous ligands of CXCR3 promote signaling through different structural elements of the receptor (111). CXCL9- and CXCL10-induced internalization was dependent on the phosphorylation of the receptor C terminus, whereas CXCL11-induced internalization was dependent on phosphorylation of intracellular loop 3 (111). Similarly, the C terminal tail of CCR7 is required for chemotaxis, calcium flux, and ERK1/2 phosphorylation, but not receptor internalization nor recycling (112). Because biased ligands drive their distinct signaling profiles via the receptor, it can be postulated that biased signaling is also critically dependent on the differential interactions between the receptor and its transducers.

Distinct conformations of transducer elements can lead to varying cellular responses. Both synthetic and endogenous β-arrestin biased ligands of CXCR3A induced a conformational change in β-arrestin upon stimulation, suggesting that transducer conformation plays a role in their biased response (15, 47). Mutation of transducers has helped determine which structures are involved in specific signaling events. Cahill et al. demonstrated that β-arrestin assumes at least two distinct conformations when bound to a GPCR: a tail conformation or a core conformation (113). The “tail” conformation interacts with the C terminal tail of the receptor, while the “core” conformation involves the finger-loop region of β-arrestin and the seven transmembrane receptor core. A β-arrestin mutant lacking the finger loop region required for assuming the “core” conformation was unable to desensitize G protein mediated signaling, even though it was still able to induce receptor internalization and other β-arrestin dependent signaling effects (113). This directly connects specific conformations of transducer isoforms to cellular functions, suggesting that preferential stabilization of certain conformations could be the basis of a biased signaling response.

Furthermore, the varying functional effects of transducer conformations may be the basis of the functional selectivity of the two β-arrestin isoforms. These two isoforms, β-arrestin 1 (arrestin-2) and β-arrestin 2 (arrestin-3), despite possessing significant sequence similarity, have been shown to promote distinct cellular functions (114). For example, β-arrestin 2 is more heavily involved in receptor desensitization than β-arrestin 1 when interacting with the vasopressin 2 receptor, and β-arrestin 1 accumulates in the nucleus while β-arrestin 2 does not (115). β-arrestin1 and β-arrestin 2 have different conformations when in complex with a phosphorylated receptor, with the main difference resting in the interaction with the receptor core (115). Mutational analyses of the two isoforms suggest that the core interactions are the basis of their differential desensitization, supporting the notion of linking β-arrestin-receptor interactions to distinct functional consequences (113, 115).

Ligand-receptor interactions, receptor-transducer interactions, and the phosphorylation barcode each present unique insight into elucidating the means through which a biased response is produced. The mechanisms of biased agonism illustrate the significant potential for their involvement in the diverse functions within the chemokine system.

Functional Effects of Chemokine Bias

While much is known regarding the biochemical mechanisms underlying biased chemokine signaling, there is relatively less known about the functional consequences of such signaling. Table 1 lists selected endogenous and synthetic chemokine receptor ligands and their functional consequences at CKRs. It is difficult to completely assess ligand, receptor, and systems bias in vitro when considering the vast diversity of tissue types in which the chemokine system is expressed, as well as the variety of functions of the chemokine system under both homeostatic and inflammatory conditions. Therefore, it is important for future research to use in vivo examples of biased agonism to direct in vitro studies that aim to understand the mechanisms underlying this signaling. We highlight some examples below.

Table 1:

Selected studies of endogenous and synthetic chemokine ligands. Highlighted compounds are those at which biased agonism has been demonstrated.

Receptor Endogenous ligands Synthetic ligands Synthetic ligand characterization System Assays References
CXCR1 CXCL6
CXCL8
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
(24, 170)
(R)-Ketoprofen Antagonist In vitro Chemotaxis (171)
Reparixin (Repertaxin) Allosteric antagonist In vitro Chemotaxis (172, 173)
In vivo Inflammation
Reperfusion injury
Blood pressure
DF 2156A Allosteric antagonist In vitro G protein signaling
Chemotaxis
Cell proliferation
(174)
In vivo Angiogenesis
Hepatic reperfusion injury
SX-517 Antagonist In vitro G protein signaling (175)
In vivo Inflammation
CXCR2 CXCL1
CXCL2
CXCL3
CXCL5
CXCL6
CXCL7
CXCL8
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
(24, 170)
SB 455821 Antagonist In vitro Chemotaxis (176)
In vivo Neutrophil migration
Reparixin (Repertaxin) Allosteric antagonist In vitro Chemotaxis (172, 173)
In vivo Inflammation
Reperfusion injury
Blood pressure
DF 2156A Allosteric antagonist In vitro G protein signaling
Chemotaxis
Cell proliferation
(174)
In vivo Angiogenesis
Hepatic reperfusion injury
Navarixin (MK-7123) Antagonist In vivo COPD Solid tumors (177179)
Danirixin Antagonist In vitro G protein signaling (180182)
In vivo Neutrophil migration and activation
COPD
SB225002 Antagonist In vitro Neutrophil
chemotaxis
(183, 184)
In vivo Neutrophil margination
Colitis
Inflammatory bowel syndrome
SX-517 Antagonist In vitro G protein signaling (175)
In vivo Inflammation
AZD5069 Antagonist In vitro Neutrophil
chemotaxis
(185)
In vivo Acute lung inflammation
CXCR3 CXCL4
CXCL9
CXCL10
CXCL11
CCL7 (Antagonist)
CCL11 (Antagonist)
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
(24, 89, 111, 119, 186, 187)
In vivo T cell polarization
T cell localization
VUF1066
VUF11418
Agonist
Agonist
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
(188, 189)
In vivo GRK engagement
Chemotaxis
Chemotaxis
Inflammation
FAUC1036
FAUC1104
Allosteric agonist
Allosteric agonist
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
(190)
BD103
BD064
cRAMX3
Allosteric modulator
Allosteric modulator
Antagonist
In vitro G protein signaling
β-arrestin recruitment
(191)
SCH546738 Antagonist In vitro G protein signaling
Chemotaxis
(192, 193)
In vivo Mouse collagen induced arthritis
Rat experimental autoimmune encephalitis
Rat Cardiac transplantation
8-azaquinazolinone derivative Allosteric modulator In vitro G protein signaling
β-arrestin recruitment
(194)
CXCR 4 Monomeric/d imeric
CXCL1
In vitro G protein signaling
β-arrestin
recruitment
Chemotaxis
(55, 195, 196)
EPI-X4 (Antagonist) In vivo Cancer metastasis
Hematopoietic stem cell mobilization
Plerixafor (AMD3100) Allosteric antagonist In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
(136, 137,168, 197)
X4-2-6 Allosteric antagonist In vivo Hematopoietic stem cell mobilization
TG-0054 (burixafor) Antagonist In vivo Hematopoietic stem cell mobilization
Choroid neovasculariz ation
Inflammation Cardiac function
(198200)
AMD11070 (mavorixafor) Antagonist In vivo X4-trophic
HIV-1 infection
Inflammation
WHIM syndrome
Melanoma
(201205)
MSX-122 Antagonist In vitro G protein signaling
Chemotaxis
Angiogenesis
(206, 207)
In vivo Inflammation
Breast cancer metastasis
Solid tumors
CTCE-9908 Antagonist In vivo Solid tumors (208)
POL-6326 Antagonist In vivo Hematopoietic stem cell mobilization (209)
RSVM
ASLW
Agonist Superagonist In vitro Chemotaxis
Receptor cell surface expression
(210)
ATI-2341 Allosteric agonist In vitro G protein signaling
β-arrestin recruitment
GRK engagement
Chemotaxis
Neutrophil mobilization
Receptor internalization
(211213)
DV1-K-(DV3) Antagonist In vitro G protein signaling
Chemotaxis
X4-tropic HIV-1 infection
(214)
T22 Antagonist In vitro G protein signaling
X4-tropic HIV-1 infection
(215)
POL6326 (Balixafortide) Antagonist In vivo Hematopoietic stem cell mobilization
HER2
negative, locally recurrent or metastatic breast cancer
(216218)
T140 Inverse agonist In vitro X4-tropic HIV-1 infection (219)
CXCL12-T140 chimera Agonist In vitro Chemotaxis (220)
CXCL12-IT1t chimera Antagonist In vitro G protein signaling
β-arrestin recruitment
Chemotaxis
(221)
PZ-218 Antagonist In vitro G protein signaling
Chemotaxis
(222, 223)
In vivo Leukocytosis
508MCI Antagonist In vitro G protein signaling (224)
In vivo Inflammation
Tumor metastasis
GSK812397 Antagonist In vitro Chemotaxis
X4-tropic HIV-1 infection
(225)
TIQ-15 Antagonist In vitro G protein signaling
β-arrestin recruitment
X4-tropic HIV-1 infection
(226)
IT1t Allosteric antagonist In vitro Chemotaxis (227, 228)
In vivo X4-tropic HIV-1 infection
ALX40–4C Antagonist In vitro
In vivo
X4-tropic HIV infection (229, 230)
CXCR5 CXCL13 (231, 232)
CXCR6 CXCL16 (233, 234)
Small molecules with exo-[3.3.1]azabicyclononane core Antagonist In vitro G protein signaling
β-arrestin recruitment
(235)
In vivo Hepatocellular carcinoma
CCR1 CCL2
CCL3
CCL4
CCL5
CCL7
CCL8
CCL13
CCL14
CCL15
CCL16
CCL23
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
(24, 61)
J113863
UCB35625
Antagonist
Antagonist
In vitro Eosinophil morphology
Receptor internalization
Chemotaxis
(236239)
In vivo Experimental murine arthritis
Met-CCL5 (Met-RANTES) Antagonist In vitro Chemotaxis (240242)
In vivo Experimental rat and murine arthritis
Rabies virus
CCX354 Antagonist In vitro Chemotaxis (243)
In vivo Inflammation
Rheumatoid arthritis
CP-481,715 Antagonist In vitro G protein signaling
Chemotaxis
(244, 245)
MLN3897 Antagonist In vivo Rheumatoid arthritis (246)
BX471 Antagonist In vivo Multiple sclerosis (245)
AZD-4818 Antagonist In vivo COPD (245)
CCX9588 Antagonist In vitro Chemotaxis (247)
LMD-559 Agonist In vitro G protein signaling
Chemotaxis
(248)
CCR2 CCL2
CCL7
CCL8
CCL11
CCL13
CCL16
G protein signaling
β-arrestin recruitment
Receptor internalization
(249)
J113863
UCB35625
Agonist
Agonist
In vitro
G protein signaling
β-arrestin recruitment
Chemotaxis
(169)
CAS 445479-97-0 Antagonist In vitro Cell proliferation
Cell migration
Invasion
(250)
INCB3344 Antagonist In vitro G protein signaling
β-arrestin recruitment
Chemotaxis
(251, 252)
N-(2-((1-(4-(3methoxyphenyl)cyclohex yl)piperidin-4-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)benzami de Antagonist (253)
CCR2-RA Antagonist In vitro G protein signaling
β-arrestin recruitment
(252)
15a Antagonist In vivo Murine atherogenesis (254)
MK-0812 Antagonist In vitro
Cell morphology (255)
In vivo Monocyte recruitment
CCR3 CCL5
CCL7
CCL8
CCL11
CCL13
CCL15
CCL24
CCL26
CCL28
In vitro G protein signaling Receptor internalization
Chemotaxis
(238, 256)
J113863
UCB35625
Antagonist
Antagonist
In vitro Eosinophil morphologic change
Receptor internalization
CCR3-tropic
HIV-1 infection
Chemotaxis
(236238)
CH0076989 Agonist In vitro Eosinophil shape change
Receptor internalization
Chemotaxis
(238)
SB-328437 Antagonist In vitro G protein signaling
Eosinophil chemotaxis
(257)
R321 Antagonist In vitro G protein signaling
β-arrestin recruitment
Chemotaxis
Receptor internalization
Eosinophil recruitment to lungs
Murine airway hyperresponsi veness
(258)
GW766994 Antagonist In vivo Asthma
Eosinophilic bronchitis
(259)
CCR4 CCL17
CCL22
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
(52, 260262)
In vivo Inflammation
MDC67
Compounds 1–4
Antagonist
Antagonist
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
(52)
Compound 8a Antagonist In vitro Chemotaxis (263)
In vivo Allergic asthma in mice
CCR5 CCL2
CCL3
CCL3L1
CCL4
CCL5
CCL8
CCL11
CCL13
CCL14
CCL16
In vitro G protein signaling
β-arrestin recruitment
(24, 242)
In vivo Receptor internalization
Inflammation
Rabies virus
Maraviroc Antagonist In vivo R5-tropic HIV-1 infection (134)
Aplaviroc Antagonist In vitro R5-tropic HIV-1 infection (264, 265)
In vivo R5-tropic HIV-1 infection
AOP-RANTES Agonist In vitro G protein signaling
Receptor internalization
R5-tropic HIV-1 infection
(160)
PSC-RANTES
5P14-RANTES
5P12-RANTES
Superagonist
Agonist
Agonist
In vitro G protein signaling
Receptor internalization
R5-tropic HIV-1 infection
(162)
TAK-652
TAK-779
Antagonist
Antagonist
In vitro R5-tropic HIV-1 infection (266, 267)
Vicriviroc Antagonist In vitro
In vivo
R5-tropic HIV-1 infection (268)
J113863
UCB35625
Agonist
Agonist
In vitro G protein signaling
β-arrestin
recruitment
Chemotaxis
(169)
YM-370749 Agonist In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
R5-tropic HIV-1 infection
(269)
Met-CCL5 (Met-RANTES) Antagonist In vitro Chemotaxis (240242)
In vivo Experimental rat and murine arthritis
Rabies virus
SCH-351125 (Ancriviroc) Antagonist In vitro G protein signaling (270272)
In vivo Rheumatoid arthritis
E-913 Antagonist In vitro G protein signaling (270)
CCR6 CCL20 In vitro Receptor
Internalization
β-arrestin recruitment
ERK phosphorylatio n
(273277)
In vivo Actin polymerization
Graft-versus-host disease
Colorectal
Cancer
CCL20 S64C Agonist In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
(278)
In vivo IL-23 dependent murine model of psoriasis
CCR7 CCL19
CCL21
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
GRK engagement
Chemotaxis β2 integrin activation
(44, 45, 129, 279)
In vivo Naïve T cell recirculation in secondary lymphoid tissue
Cmp2105
CS-1
CS-2
Navarixin
Allosteric antagonist
Allosteric antagonist
Allosteric antagonist
Allosteric antagonist
In vitro Crystal structure
β-arrestin recruitment
Thermal shift assay
(280)
Cosalane Antagonist In vitro β-arrestin recruitment
Chemotaxis
(281)
CCR8 CCL1
CCL8
CCL16
CCL18
(282, 283)
LMD-559 Agonist In vitro G protein signaling
Chemotaxis
(248)
R243 Antagonist In vitro G protein signaling
Macrophage aggregation
(284)
In vivo Cytokine secretion
Hapten induced colitis
Peritoneal adhesions
Napthalene-sulfonamide derivatives Allosteric antagonists In vitro G protein signaling
Chemotaxis
(285)
CCR9 CCL25 (286)
Vercirnon Allosteric antagonist In vitro Crystal structure
G protein signaling
(108, 287, 288)
In vivo Crohn’s disease
CCX8037 Antagonist In vitro Chemotaxis (289)
In vivo Intestinal T cell accumulation
CCX282-B CCX025 Antagonist
Antagonist
In vitro G protein signaling
Chemotaxis
(290, 291)
In vivo Crohn’s disease
Ulcerative colitis
CCR10 CCL27
CCL28
In vitro G protein signaling
β-arrestin recruitment
Receptor internalization
Chemotaxis
(24, 292)
Eut-22 Antagonist In vitro G protein signaling
Chemotaxis
(293)
In vivo Murine contact hypersensitivity
CX3CR1 CX3CL1 (294)
JMS-17–2 Antagonist In vitro Chemotaxis
Pancreatic ductal adenocarcino ma motility and viability
(295, 296)
In vivo Breast cancer metastasis
KAND567 Antagonist In vivo Atherosclerosis
Myocardial infarction
(297)
XCR1/CCXCR1 XCL1
XCL2
(298)
ACKR1/DARC CXCL5
CXCL6
CXCL8
CXCL11
CCL2
CCL5
CCL7
CCL11
CCL13
CCL14
CCL17
(299)
ACKR2 CCL2
CCL3
CCL4
CCL5
CCL7
CCL8
CCL11
CCL13
CCL14
CCL17
CCL22
In vitro G protein signaling
Receptor internalization
(300, 301)
CCL14(9–74) Agonist In vitro G protein signaling
Actin rearrangement
Receptor internalization
(300, 301)
ACKR3/CXCR7 CXCL11
CXCL12
In vitro G protein signaling
β-arrestin recruitment
Chemotaxis
(62, 63, 66, 299)
TC14012 Allosteric antagonist In vitro β-arrestin recruitment (302)
GSLW Allosteric agonist In vitro β-arrestin recruitment
Receptor internalization
(303)
FC313 Allosteric agonist In vitro β-arrestin recruitment (304)
CXCL11_12 chimera Agonist In vitro Selective
ACKR3 agonist, over
CXCR3 and CXCR4
(305, 306)
AMD3100 Allosteric agonist In vitro β-arrestin recruitment (307)
VUF11207
VUF11403
Agonist In vitro β-arrestin recruitment
Receptor internalization
(308)
CCX77 Agonist In vitro β-arrestin recruitment
Receptor internalization
Chemotaxis
(309312)
In vivo Trans-endothelial migration
Tumor growth
Tumor growth and metastasis
Tumor angiogenesis
X4-tropic HIV-1 infection c
CCX777 Agonist In vitro β-arrestin recruitment (86)
ACKR4/CCR11 CCL19
CCL20
CCL21
CCL25
(299, 313)

CXCR3

Mechanisms and functional effects of biased agonism are especially well-characterized at CXCR3 in activated CD8+ T cells. CXCL11 promotes significantly greater chemotactic response when compared to CXCL9 and CXCL10 (89, 116). Synthetic small molecule agonists, VUF10661 and VUF11418, similarly display biased agonism at CXCR3, with VUF10661 characterized as β-arrestin-biased and VUF11418 as G protein-biased (116, 117). Notably, Smith et al. showed in a murine model of allergic contact hypersensitivity that VUF10661 potentiates a dinitrofluorobenzene-induced inflammatory response, while VUF11418 does not (116). This increase in inflammation derived from VUF10661 was lost when these experiments were repeated in β-arrestin 2 knock out (KO) mice. Additionally, a greater number of CD3+ T cells and, specifically, CD3+ β-arrestin+ CXCR3+ T cells, were found in tissue biopsies derived from patients with allergic contact dermatitis than from nonlesional controls. However, in vitro chemotaxis assays further revealed that both β-arrestin dependent phosphorylation of Akt and G protein signaling contribute to CXCR3-mediated chemotaxis (116). The exact consequences of G protein and β-arrestin bias for chemotaxis are therefore still unclear. While VUF10661 induces the greatest chemotactic response, G protein signaling is additionally necessary for any chemotactic response, as T cell migration controlled by CXCL9, CXCL10, and CXCL11 is abrogated following pretreatment with pertussis toxin, an inhibitor of Gαi (116, 118). It is possible that β-arrestin and G protein dependent signaling cascades that contribute to the overall chemotactic response are interrelated.

Biased signaling at CXCR3 has also been found to direct T cell polarization and function (119). In a murine model of EAE, Zohar et al. found that CXCL10 induces polarization of T helper (Th) cells into Th1/Th17 cells, promoting inflammation, whereas CXCL11 promotes Th polarization into IL-10-producing T regulatory (Treg) cells, dampening inflammation. The authors also demonstrate distinct downstream signaling cascades for the endogenous ligands, with induction of T cell polarization by CXCL10 via phosphorylation of STAT1, STAT4, and STAT5, and by CXCL11 via phosphorylation of STAT3 and STAT6 (120). Additionally, CXCR3 signaling directs T cell localization by limiting Th cells to the perivascular space in the central nervous system, thereby increasing interaction between Treg and Th cells, and attenuating inflammation, demyelination, and axonal damage (121). CXCR3−/− mice demonstrate a more disorganized perivascular distribution of Th cells and experience increased tissue damage and less recovery in EAE than WT CXCR3 mice (121). Biased signaling has not been directly implicated in this disease process; however, there is evidence demonstrating that in murine EAE, CXCL9, CXCL10, and CXCL11 are expressed in different spatial and cellular locations within the central nervous system, suggesting a functional role of biased agonism at CXCR3 (121).

CXCR1 and CXCR2

There is also evidence demonstrating different cellular signaling induced by CXCL8 at its endogenous receptors, CXCR1 and CXCR2, both of which are primarily involved in directing neutrophil chemotaxis to sites of infection and initiating its cytotoxic effects (56). Interestingly, CXCL8 can exist as a monomer, dimer, or a combination of these two states in vivo. Nasser et al. provide evidence for biased signaling in this system in that the monomeric form of CXCL8 is more effective at driving calcium mobilization, chemotaxis and exocytosis than the dimeric form. They also discovered that this ligand bias is present at CXCR1, but not at CXCR2, suggesting an interaction between both ligand and receptor bias. To our knowledge, the functional consequences of this ligand and receptor bias have not been rigorously explored in vivo.

CXCR4

Similarly, at CXCR4, which is implicated in tumor cell trafficking, differential signaling by the monomeric and dimeric forms of its endogenous ligand CXCL12 demonstrated functional consequences in a murine model of colorectal carcinoma or melanoma (55). Previous research has shown that locally produced CXCL12 limits migration potential of cells expressing CXCR4 by either desensitizing these cells to endogenous ligand or exceeding optimal levels for chemotaxis by nullifying endogenous CXCL12 gradients produced in distant tissues (122124). Drury et al. demonstrate that the monomeric form of CXCL12 is much more efficacious in stimulating filamentousactin polymerization and inducing chemotaxis than the dimeric form (55). From this in vitro evidence, one could postulate that the dimeric form of CXCL12 would inhibit metastasis and the monomeric form would promote metastasis. However, the authors found that exogenous administration of either WT CXCL12, a preferentially monomeric form, or preferentially dimeric form all inhibited tumor metastasis to similar extents. The reasons underlying this discrepancy between in vitro and in vivo work is possibly due to systems bias where the function of exogenously administered CXCL12 may assume a different function in vivo.

Furthermore, CXCR4 also exhibits receptor bias in WHIM syndrome which is most commonly due to a gain of function receptor C terminal truncation mutant (125). The truncated CXCR4 receptor found in these patients promotes more G protein dependent signaling cascades compared to the WT receptor, presumably due to the decreased ability to recruit the GRKs and β-arrestins and desensitize the receptor. However, there is evidence demonstrating that, even in this truncated receptor, β-arrestin is still able to engage the receptor via intracellular loop 3 and that β-arrestins engage in different signaling cascades than those in the WT receptor. Lagane et al. demonstrate using CXCR4 mutants derived from patients with WHIM syndrome that receptor desensitization and internalization is regulated by the C terminal tail of CXCR4 while β-arrestin-mediated signaling, like ERK1/2 activation, is primarily mediated by its interaction with ICL3 (125). The authors also show that WT CXCR4 can homodimerize and also heterodimerize with mutant CXCR4 receptors; these heterodimers are able to further enhance chemotaxis due to reduced internalization of both the mutant and WT CXCR4 and prolonged β-arrestin dependent signaling as measured by ERK 1/2 phosphorylation (125). This study highlights the many complex levels of bias that can exist in the chemokine system, where a biased receptor can not only direct different signaling patterns, but influence those of wild-type receptors. Studies have also shown, among others, CCR5 homodimerization shortly after synthesis in the endoplasmic reticulum, CXCR4 homodimer formation and heterodimer formation with CCR2, and CXCR2 homodimerization (126128). Future research in this area could uncover unidentified sources of biased signaling, which could explain many of the non-redundant functional effects seen within the chemokine system.

CCR7

There is evidence of biased agonism at CCR7 between its endogenous ligands CCL19 and CCL21 and their role in lymphocyte extravasation and lymphocyte trafficking to and within secondary lymphoid organs (45, 129). Förster et al. demonstrated the importance of CCR7 in these signaling pathways by generating CCR7 deficient mice, which had impaired entry and retention of naïve T cells, dendritic cells (DCs), and B cells in multiple secondary lymphoid organs (129). CCR7-dependent signaling is therefore likely necessary for effective T cell-B cell and T cell-DC interactions.

Kohout et al. further characterize biased signaling at CCR7 by examining receptor desensitization and ERK1/2 activation in response to CCL19 and CCL21 (45). The endogenous ligands promote nearly identical amounts of G protein signaling, but only CCL19 induces substantial receptor desensitization via receptor C terminal phosphorylation and β-arrestin recruitment (45). Although these ligands induce similar calcium mobilization and leukocyte migration potential in vitro, CCL19, but not CCL21, induces rapid CCR7 internalization (130). Resultingly, pretreatment of T lymphocytes with CCL19 inhibits a chemotactic response to either CCL19 or CCL21, while cells retain full chemotactic potential following pretreatment with CCL21 (130).

When comparing CCL19−/− mice, plt/plt mice (deficient in both CCL19 and CCL21), and plt/CCL19 mice (lacking CCL19 and one CCL21 allele) to WT mice, Britschgi et al. found that deficiency of both CCL19 and CCL21 is required for abnormal localization of DCs in lymph nodes (131). Interestingly, they also discovered that CCL19 was not needed for DC migration, maturation, and T-cell priming (131). These results suggest that CCL19 is dispensable for certain aspects of DC function regulated by CCR7. It is unclear if CCL19 is sufficient to support normal DC function in the absence of CCL21, and if CCL19 and CCL21 have both overlapping and distinct physiologic roles. Taken together, these studies indicate the importance of biased signaling at CCR7 in the trafficking of a wide array of immune cells, as well as their interactions within secondary lymphoid tissues.

Targeting Chemokine System Biased Agonism in Drug Development

As opposed to the other GPCRs that have one or perhaps a few endogenous ligands, most CKRs have a number of endogenous ligands, many of which are biased, have additional receptor targets, and have variable tissue expression. This primary example of naturally occurring biased agonism may unveil a variety of new drug targets which exploit this biochemical phenomenon, but it may also uncover key information regarding the mechanisms and structural determinants underlying biased agonism across all GPCRs. An enhanced understanding of the mechanisms that control biased signaling in GPCRs may lead to the development of pharmaceutical drugs with increased efficacy and reduced side effect profile by selectively activating therapeutic signaling pathways. Biased therapeutics at other GPCR subtypes, including the type 1 angiotensin II receptor (AT1R), μ-opioid receptor, κ-opioid receptor, D2 dopamine receptor have demonstrated that differential activation of G-protein and β-arrestin signaling pathways has the potential to improve efficacy and avoid unwanted side effects. However, the in vivo effects of these compounds can be difficult to predict, even with detailed studies involving knockout animals and comparisons to balanced agonists and antagonists.

Due to the wide range of biological processes and pathologies in which the chemokine system is implicated, identification of biased drugs targeting chemokine ligands and receptors is of significant interest in clinical research. Yet, the success of drug development to target the chemokine system and its associated pathologies has been limited. Although clinical trials over the past decade have attempted to target over 50% of CKRs (132), there are currently only three FDA approved drugs on the market that target CKRs. Maraviroc is a CCR5 antagonist that inhibits R5-tropic HIV-1 entry into cells (133, 134). Plerixafor (AMD3100) is a CXCR4 antagonist first identified to block X4-tropic HIV-1 infection, but is now FDA-approved for use in autologous hematopoietic stem cell (HSC) transplantation in patient with Non-Hodgkin’s lymphoma or multiple myeloma (135138). CXCR4 signaling promotes the retention of HSCs in the bone marrow and AMD3100, as an antagonist of CXCR4, mobilizes HSCs into the peripheral blood, a crucial step for conferring the therapeutic benefit of autologous transplantation (139). Finally, mogamulizumab is a CCR4 antibody used for treating cutaneous T-cell lymphoma (CTCL) and adult T-cell leukemia/lymphoma (ATLL) (140, 141). Currently, phase 3 clinical trials are under way to evaluate additional chemokine antagonists including the CCR5 antagonist leronimab and CXCR4 antagonists balixafortide and mavorixafor (142). Studies for leronimab are investigating the safety and effectiveness of using it with and without an optimized antiretroviral therapy (ART) regimen and for the safety and effectiveness of leronimab monotherapy for the maintenance of R5-tropic HIV-1 suppression over 48 weeks (143, 144). Balixafortide is currently being studied for its efficacy, safety and tolerability when given intravenously with eribulin, a common breast cancer chemotherapy drug, versus eribulin alone in the treatment of HER2 negative, locally recurrent or metastatic breast cancer (145). Mavorixafor is being examined for its efficacy in participants with WHIM syndrome as assessed by increasing levels of circulating neutrophils compared with placebo, as well as its safety and tolerability (146). Continued studies will determine whether these drugs represent promise for therapeutic benefit in their respective disease states.

CCR5 in HIV

A large component of drug development in the chemokine system has been focused on inhibition of HIV-1 entry and infection. R5-tropic HIV-1, the predominant form of the virus during initial transmission and infection, requires CCR5 as a co-receptor to CD4 for entry into monocytes and macrophages (147151). X4-tropic virus, which appears later and is responsible for the serious reduction in Th cell count, requires CXCR4 as a co-receptor to CD4 (147151). A surge in research on chemokine receptor biology occurred following the findings that CCR5Δ32 homozygous mutant individuals are resistant to HIV infection, and CCR5Δ32 heterozygous mutant individuals had a slower progression to AIDS after infection (152156). These small populations of individuals have truncated forms of CCR5 that fail to reach the cell surface and, therefore, cannot be utilized by HIV to enter the cell. This led to the development of maraviroc, a CCR5 antagonist which is currently used for combination antiretroviral therapy in adults infected with R5-tropic HIV-1 (157, 158). Drugs which induce receptor internalization of CCR5 or CXCR4, a canonically β-arrestin driven process, may therefore provide significant therapeutic benefit. Interestingly, while there have been worries that such a pharmacological approach would lead to deleterious immunological side effects, CCR5Δ32 mutant individuals do not appear to be immunocompromised (159). However, this finding does not suggest that a drug of this type would not have serious side effect, and future research should rigorously study these details to determine if this is a viable therapeutic strategy.

While endogenous CCR5 ligands CCL3, CCL4, and CCL5 (RANTES) and CCL8 display limited degrees of bias (43), many modified chemokine analogs for CCR5 do exhibit a high degree of bias. AOP-RANTES is β-arrestin-biased agonist, which rapidly induces CCR5 internalization, and inhibits recycling to the cell surface, retaining a large amount of receptor in endosomes (160). Notably, it has been shown that the potency of AOP-RANTES and CCL5 to inhibit R5-tropic HIV-1 infection was correlated with the degree of CCR5 internalization and inhibition of recycling, which supports the notion that these processes represent therapeutic targets to prevent HIV-1 infection (160). Unfortunately, AOP-RANTES simultaneously promoted the intracellular replication of HIV-1 via a mechanism downstream of viral entry that is sensitive to pertussis toxin, suggesting a role for the Gαi subunit in viral replication (161). PSC-RANTES, is a balanced CCR5 super-agonist that is 50 times more potent than AOP-RANTES for HIV infection inhibition in vitro and acts via a mechanism of long-term intracellular sequestration of CCR5 (162). While PSC-RANTES serves as a potential topical agent to prevent HIV transmission at mucosal barriers, CCR5 induced receptor internalization occurs alongside other CCR5 direct signaling pathways which have been shown to result in intense mucosal inflammation and paradoxically enhance HIV infection (163). 5P14-RANTES is a β-arrestin-biased agonist that induces no detectable G protein activation but does induce receptor internalization, however, not to the extent of PSC-RANTES. PSC-RANTES and 5P14-RANTES also differ in their post-endocytic trafficking of CCR5, a spatiotemporal form of bias that could have significant implications for their ability to inhibit HIV infection (164). PSC-RANTES directs internalized receptor first into the endosome recycling compartment (ERC) and then into the trans-Golgi network (TGN), resulting in long term sequestration, while 5P14-RANTES directs CCR5 only to the ERC, resulting in short term sequestration (164). Bönsch et al. suggest that duration of β-arrestin association with CCR5 is key to the differential post-endocytic receptor trafficking, with PSC-RANTES inducing robust and long-term β-arrestin recruitment to CCR5 and 5P14-RANTES inducing recruitment that is more transient in nature (164).

Altogether, the past roughly 20 years of research on the importance of CCR5 and CXCR4 for HIV entry and infection of leukocytes reveals a pathological state in which biased signaling could provide significant therapeutic benefit. However, it is still not clear exactly to what extent G protein signaling and β-arrestin recruitment, and their associated downstream signaling cascades affect HIV entry and infection. Additionally, the side effect profiles of synthetic ligands which induce receptor internalization have not been determined, an important question for future research to address in order to develop clinically efficacious drugs.

CXCR3 and T cell-mediated inflammation

Similar efforts to target the biased signaling properties of the chemokine system have been focused on CXCR3. CXCR3 is an important mediator of Th1 cell function and plays a role in natural killer cell migration and inhibition of angiogenesis (165). CXCR3 is rapidly expressed on naïve T cells following their activation and remains highly expressed on Th1-type CD4+ T cells and effector CD8+ T cells (166). CXCR3, regulated by its three interferon-inducible endogenous ligands, CXCL9, CXCL10, and CXCL11, promotes T cell trafficking to peripheral sites of inflammation, as well as lymphoid tissues, facilitating the interaction of T cells with antigen presenting cells (166). CXCR3 and its endogenous ligands have been implicated in a variety of disease processes and pathological states, including allergic contact dermatitis, autoinflammation and autoimmunity, atherosclerosis, and cancer metastasis (116, 167). As previously mentioned, Smith et al. demonstrate the pharmacologic potential of biased signaling at CXCR3 using a mouse model of allergic contact hypersensitivity (116). Their finding that both G protein signaling and β-arrestin-mediated signaling are necessary for full chemotactic function suggests that drugs designed to increase migration of CXCR3+ T cells, such as those used in cancer immunotherapy, could promote signaling through the β-arrestin, Gαi, or potentially both (116). On the other hand, a more complicated picture arises for development of drugs designed to inhibit migration of CXCR3+ T cells to reduce inflammation (116). Pertussis toxin treatment completely abrogates T cell chemotaxis, whereas Akt inhibition and β-arrestin 2 knock out (KO) T cells display only reduced chemotaxis (116). Therefore, further research is needed to determine the most efficacious mechanism of action for drugs designed to promote or inhibit migration of CXCR3+ T cells. Karin et al. suggest the possibility of using stabilized chemokines as biological drugs for autoimmunity and graft-versus-host disease (GVHD) (119). Endogenous chemokines have relatively short half-lives, and therefore their administration alone does not provide significant therapeutic benefit. Based on their finding that CXCL11 induces T cell polarization toward IL-10-producing Treg cells, whereas CXCL9 and CXCL10 induce T cell polarization toward Th1/Th17 cells, Karin et al. propose that exogenously stabilized chemokines could be developed as biological drugs at CXCR3 that selectively activate FOXP3+ Treg or Tr1 cells over proinflammatory cells, taking advantage of the phenomenon of biased agonism (119).

Other CKRs

Lastly, the pharmacologic potential of biased signaling has been implicated at various other CKRs. At CXCR4, drug tolerance has been seen to occur in response to administration of AMD3100, with increasing CXCR4 expression on the surface of hematopoietic stem cells (HSCs), promoting their rehoming to the bone marrow (168). Hitchinson et al. found that such drug tolerance was avoided in vitro with administration of X4-2-6, a β-arrestin-biased peptide antagonist (168). X4-2-6 forms a ternary complex with endogenous CXCL12 and CXCR4, causing the N terminus of CXCL12 to detach from CXCR4. This N terminal interaction is important for inducing the conformational change in CXCR4 that promotes GTP loading of Gαi, and X4-2-6 therefore inhibits G protein signaling (168). Receptor C terminal phosphorylation, β-arrestin recruitment, and receptor internalization are retained, creating a biased signaling profile that demonstrates decreased drug tolerance (168). These findings suggest the potential of biased allosteric modulators to provide therapeutic benefit to patients who develop tolerance to unbiased antagonists. At CCR2 and CCR5, synthetic enantiomers J113863 and UCB35625 exhibit differential G protein signaling and β-arrestin recruitment, as well as functional effects on L1.2 cell (a mouse leukemia cell line) chemotaxis (169). This represents the first description of chemokine receptor ligands displaying enantioselective properties, a finding of particular pharmacologic significance as many drugs are typically racemic mixtures (169).

Conclusion

The discovery of alternative signaling pathways beyond G protein signaling, specifically those driven by β-arrestin, have led to a renewed appreciation of the complexity and non-redundant nature of the chemokine system. The number of possible cellular phenotypes driven by the chemokine signaling are staggering when considering the diversity of ligands, receptors, and cell and tissue types implicated in this system. Rather than being designed to simply produce robust outputs, numerous functional and structural studies suggest that the chemokine system is capable of driving highly specific and directed signaling processes to coordinate immune cell function. Considering that CKRs are found in many non-immune related tissue types, the complexity of the biased signaling within this system is likely underappreciated. An enhanced understanding of the pluridimensional efficacy within the chemokine system presents an opportunity to determine the fundamental principles underlying biased agonism across all GPCRs. Additionally, the phenomenon of biased signaling within the chemokine system presents a potential target for future drug development. Within the complex signaling mechanisms of the chemokine system lies the potential for greater druggability, with more focused pharmacological effects and possibly fewer harmful side effects. Although biased agonism has been widely observed using both endogenous and synthetic chemokine ligands, understanding the mechanisms and functional effects of this complex signaling in vivo must progress in order to develop promising therapeutics. Many of the problems faced in pharmaceutical research on chemokine-targeted therapeutics relate to an apparent lack of translation between cellular, animal, and human models, as well as an incomplete understanding of the biochemical and physiological implications of the chemokine system bias. Advances in our understanding of the structural and functional determinants of biased agonism may be necessary before the promise of more effective therapeutics aimed at treating CKR- and GPCR-related pathologies can be fully realized.

Highlights.

  • Biased agonism within the chemokine system drives highly specific cellular outputs

  • Chemokine system is a model for biased agonism at G protein-coupled receptors

  • Utilizing biased agonism in the chemokine system for potential therapeutic purposes

ACKNOWLEDGEMENTS

This work was supported by the National Institute of General Medical Sciences T32GM00717 (D.S.E) and R01GM122798 (S.R); the Duke University Medical Scientist Training Program (D.S.E); and the American Heart Association Predoctoral Fellowship 20PRE35120592 (D.S.E).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

DECLARATION OF INTEREST

None

REFERENCES

  • 1.Rossi D, Zlotnik A. The Biology of Chemokines and their Receptors. Annual Review of Immunology. 2000;18(1):217–42. [DOI] [PubMed] [Google Scholar]
  • 2.Scholten DJ, Canals M, Maussang D, Roumen L, Smit MJ, Wijtmans M, et al. Pharmacological modulation of chemokine receptor function. Br J Pharmacol. 2012;165(6):1617–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Allen SJ, Crown SE, Handel TM. Chemokine: receptor structure, interactions, and antagonism. Annu Rev Immunol. 2007;25:787–820. [DOI] [PubMed] [Google Scholar]
  • 4.Zlotnik A, Burkhardt AM, Homey B. Homeostatic chemokine receptors and organ-specific metastasis. Nat Rev Immunol. 2011;11(9):597–606. [DOI] [PubMed] [Google Scholar]
  • 5.Rajagopalan L, Rajarathnam K. Structural basis of chemokine receptor function--a model for binding affinity and ligand selectivity. Biosci Rep. 2006;26(5):325–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hughes CE, Nibbs RJB. A guide to chemokines and their receptors. FEBS J. 2018;285(16):2944–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Russo RC, Garcia CC, Teixeira MM. Anti-inflammatory drug development: Broad or specific chemokine receptor antagonists? Curr Opin Drug Discov Devel. 2010;13(4):414–27. [PubMed] [Google Scholar]
  • 8.Rosenkilde MM, Schwartz TW. The chemokine system -- a major regulator of angiogenesis in health and disease. APMIS. 2004;112(7–8):481–95. [DOI] [PubMed] [Google Scholar]
  • 9.Rostène W, Kitabgi P, Parsadaniantz SM. Chemokines: a new class of neuromodulator? Nature Reviews Neuroscience. 2007;8(11):895–903. [DOI] [PubMed] [Google Scholar]
  • 10.Sokol CL, Luster AD. The chemokine system in innate immunity. Cold Spring Harb Perspect Biol. 2015;7(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cha YR, Fujita M, Butler M, Isogai S, Kochhan E, Siekmann AF, et al. Chemokine signaling directs trunk lymphatic network formation along the preexisting blood vasculature. Dev Cell. 2012;22(4):824–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Meucci O, Fatatis A, Simen AA, Bushell TJ, Gray PW, Miller RJ. Chemokines regulate hippocampal neuronal signaling and gp120 neurotoxicity. Proc Natl Acad Sci U S A. 1998;95(24):14500–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.de Haas AH, van Weering HR, de Jong EK, Boddeke HW, Biber KP. Neuronal chemokines: versatile messengers in central nervous system cell interaction. Mol Neurobiol. 2007;36(2):137–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wang Z, Shang H, Jiang Y. Chemokines and Chemokine Receptors: Accomplices for Human Immunodeficiency Virus Infection and Latency. Front Immunol. 2017;8:1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Smith JS, Rajagopal S, Atwater AR. Chemokine Signaling in Allergic Contact Dermatitis: Toward Targeted Therapies. Dermatitis. 2018;29(4):179–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Balkwill FR. The chemokine system and cancer. J Pathol. 2012;226(2):148–57. [DOI] [PubMed] [Google Scholar]
  • 17.Antonia AL, Gibbs KD, Trahair ED, Pittman KJ, Martin AT, Schott BH, et al. Pathogen Evasion of Chemokine Response Through Suppression of CXCL10. Frontiers in Cellular and Infection Microbiology. 2019;9(280). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zernecke A, Weber C. Chemokines in Atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2014;34(4):742–50. [DOI] [PubMed] [Google Scholar]
  • 19.Koelink PJ, Overbeek SA, Braber S, de Kruijf P, Folkerts G, Smit MJ, et al. Targeting chemokine receptors in chronic inflammatory diseases: an extensive review. Pharmacol Ther. 2012;133(1):1–18. [DOI] [PubMed] [Google Scholar]
  • 20.Hernandez PA, Gorlin RJ, Lukens JN, Taniuchi S, Bohinjec J, Francois F, et al. Mutations in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined immunodeficiency disease. Nat Genet. 2003;34(1):70–4. [DOI] [PubMed] [Google Scholar]
  • 21.Kleist AB, Getschman AE, Ziarek JJ, Nevins AM, Gauthier PA, Chevigné A, et al. New paradigms in chemokine receptor signal transduction: Moving beyond the two-site model. Biochem Pharmacol. 2016;114:53–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Mantovani A The chemokine system: redundancy for robust outputs. Immunol Today. 1999;20(6):254–7. [DOI] [PubMed] [Google Scholar]
  • 23.Devalaraja MN, Richmond A. Multiple chemotactic factors: fine control or redundancy? Trends Pharmacol Sci. 1999;20(4):151–6. [DOI] [PubMed] [Google Scholar]
  • 24.Rajagopal S, Bassoni DL, Campbell JJ, Gerard NP, Gerard C, Wehrman TS. Biased agonism as a mechanism for differential signaling by chemokine receptors. J Biol Chem. 2013;288(49):35039–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pierce KL, Premont RT, Lefkowitz RJ. Seven-transmembrane receptors. Nat Rev Mol Cell Biol. 2002;3(9):639–50. [DOI] [PubMed] [Google Scholar]
  • 26.Hauser AS, Attwood MM, Rask-Andersen M, Schioth HB, Gloriam DE. Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov. 2017;16(12):829–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Rajagopal S, Rajagopal K, Lefkowitz RJ. Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nat Rev Drug Discov. 2010;9(5):373–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Neves SR, Ram PT, Iyengar R. G Protein Pathways. Science. 2002;296(5573):1636–9. [DOI] [PubMed] [Google Scholar]
  • 29.Steen A, Larsen O, Thiele S, Rosenkilde MM. Biased and g protein-independent signaling of chemokine receptors. Front Immunol. 2014;5:277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Reiter E, Lefkowitz RJ. GRKs and beta-arrestins: roles in receptor silencing, trafficking and signaling. Trends Endocrinol Metab. 2006;17(4):159–65. [DOI] [PubMed] [Google Scholar]
  • 31.Legler DF, Thelen M. New insights in chemokine signaling. F1000Res. 2018;7:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Arai H, Tsou CL, Charo IF. Chemotaxis in a lymphocyte cell line transfected with C-C chemokine receptor 2B: evidence that directed migration is mediated by betagamma dimers released by activation of Galphai-coupled receptors. Proc Natl Acad Sci U S A. 1997;94(26):14495–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lohse M, Benovic J, Codina J, Caron M, Lefkowitz R. beta-Arrestin: a protein that regulates beta-adrenergic receptor function. Science. 1990;248(4962):1547–50. [DOI] [PubMed] [Google Scholar]
  • 34.Smith JS, Rajagopal S. The beta-Arrestins: Multifunctional Regulators of G Protein-coupled Receptors. J Biol Chem. 2016;291(17):8969–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Violin JD, Lefkowitz RJ. Beta-arrestin-biased ligands at seven-transmembrane receptors. Trends Pharmacol Sci. 2007;28(8):416–22. [DOI] [PubMed] [Google Scholar]
  • 36.van Gastel J, Hendrickx JO, Leysen H, Santos-Otte P, Luttrell LM, Martin B, et al. beta-Arrestin Based Receptor Signaling Paradigms: Potential Therapeutic Targets for Complex Age-Related Disorders. Front Pharmacol. 2018;9:1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lefkowitz RJ, Rajagopal K, Whalen EJ. New roles for beta-arrestins in cell signaling: not just for seven-transmembrane receptors. Mol Cell. 2006;24(5):643–52. [DOI] [PubMed] [Google Scholar]
  • 38.DeWire SM, Ahn S, Lefkowitz RJ, Shenoy SK. Beta-arrestins and cell signaling. Annu Rev Physiol. 2007;69:483–510. [DOI] [PubMed] [Google Scholar]
  • 39.Smith JS, Lefkowitz RJ, Rajagopal S. Biased signalling: from simple switches to allosteric microprocessors. Nat Rev Drug Discov. 2018;17(4):243–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Liebick M, Henze S, Vogt V, Oppermann M. Functional consequences of chemically-induced beta-arrestin binding to chemokine receptors CXCR4 and CCR5 in the absence of ligand stimulation. Cell Signal. 2017;38:201–11. [DOI] [PubMed] [Google Scholar]
  • 41.Galandrin S, Bouvier M. Distinct signaling profiles of beta1 and beta2 adrenergic receptor ligands toward adenylyl cyclase and mitogen-activated protein kinase reveals the pluridimensionality of efficacy. Mol Pharmacol. 2006;70(5):1575–84. [DOI] [PubMed] [Google Scholar]
  • 42.Kenakin T Agonist-receptor efficacy. II. Agonist trafficking of receptor signals. Trends Pharmacol Sci. 1995;16(7):232–8. [DOI] [PubMed] [Google Scholar]
  • 43.Corbisier J, Galès C, Huszagh A, Parmentier M, Springael JY. Biased signaling at chemokine receptors. J Biol Chem. 2015;290(15):9542–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Zidar DA, Violin JD, Whalen EJ, Lefkowitz RJ. Selective engagement of G protein coupled receptor kinases (GRKs) encodes distinct functions of biased ligands. Proc Natl Acad Sci U S A. 2009;106(24):9649–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kohout TA, Nicholas SL, Perry SJ, Reinhart G, Junger S, Struthers RS. Differential desensitization, receptor phosphorylation, beta-arrestin recruitment, and ERK1/2 activation by the two endogenous ligands for the CC chemokine receptor 7. J Biol Chem. 2004;279(22):23214–22. [DOI] [PubMed] [Google Scholar]
  • 46.Byers MA, Calloway PA, Shannon L, Cunningham HD, Smith S, Li F, et al. Arrestin 3 mediates endocytosis of CCR7 following ligation of CCL19 but not CCL21. J Immunol. 2008;181(7):4723–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Smith JS, Alagesan P, Desai NK, Pack TF, Wu JH, Inoue A, et al. C-X-C Motif Chemokine Receptor 3 Splice Variants Differentially Activate Beta-Arrestins to Regulate Downstream Signaling Pathways. Mol Pharmacol. 2017;92(2):136–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Berchiche YA, Gravel S, Pelletier ME, St-Onge G, Heveker N. Different effects of the different natural CC chemokine receptor 2b ligands on beta-arrestin recruitment, Gαi signaling, and receptor internalization. Mol Pharmacol. 2011;79(3):488–98. [DOI] [PubMed] [Google Scholar]
  • 49.Watts AO, Scholten DJ, Heitman LH, Vischer HF, Leurs R. Label-free impedance responses of endogenous and synthetic chemokine receptor CXCR3 agonists correlate with Gi-protein pathway activation. Biochem Biophys Res Commun. 2012;419(2):412–8. [DOI] [PubMed] [Google Scholar]
  • 50.O’Boyle G, Brain JG, Kirby JA, Ali S. Chemokine-mediated inflammation: Identification of a possible regulatory role for CCR2. Mol Immunol. 2007;44(8):1944–53. [DOI] [PubMed] [Google Scholar]
  • 51.Sanchez J, Lane JR, Canals M, Stone MJ. Influence of Chemokine N-Terminal Modification on Biased Agonism at the Chemokine Receptor CCR1. Int J Mol Sci. 2019;20(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ajram L, Begg M, Slack R, Cryan J, Hall D, Hodgson S, et al. Internalization of the chemokine receptor CCR4 can be evoked by orthosteric and allosteric receptor antagonists. Eur J Pharmacol. 2014;729:75–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ludeman JP, Stone MJ. The structural role of receptor tyrosine sulfation in chemokine recognition. Br J Pharmacol. 2014;171(5):1167–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Proudfoot AEI, Johnson Z, Bonvin P, Handel TM. Glycosaminoglycan Interactions with Chemokines Add Complexity to a Complex System. Pharmaceuticals (Basel). 2017;10(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Drury LJ, Ziarek JJ, Gravel S, Veldkamp CT, Takekoshi T, Hwang ST, et al. Monomeric and dimeric CXCL12 inhibit metastasis through distinct CXCR4 interactions and signaling pathways. Proc Natl Acad Sci U S A. 2011;108(43):17655–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Nasser MW, Raghuwanshi SK, Grant DJ, Jala VR, Rajarathnam K, Richardson RM. Differential activation and regulation of CXCR1 and CXCR2 by CXCL8 monomer and dimer. J Immunol. 2009;183(5):3425–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ziarek JJ, Kleist AB, London N, Raveh B, Montpas N, Bonneterre J, et al. Structural basis for chemokine recognition by a G protein-coupled receptor and implications for receptor activation. Sci Signal. 2017;10(471). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Veldkamp CT, Seibert C, Peterson FC, De la Cruz NB, Haugner JC, Basnet H, et al. Structural basis of CXCR4 sulfotyrosine recognition by the chemokine SDF-1/CXCL12. Sci Signal. 2008;1(37):ra4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mortier A, Gouwy M, Van Damme J, Proost P. Effect of posttranslational processing on the in vitro and in vivo activity of chemokines. Exp Cell Res. 2011;317(5):642–54. [DOI] [PubMed] [Google Scholar]
  • 60.Lorenzen E, Ceraudo E, Berchiche YA, Rico CA, Furstenberg A, Sakmar TP, et al. G protein subtype-specific signaling bias in a series of CCR5 chemokine analogs. Sci Signal. 2018;11(552). [DOI] [PubMed] [Google Scholar]
  • 61.Tian Y, New DC, Yung LY, Allen RA, Slocombe PM, Twomey BM, et al. Differential chemokine activation of CC chemokine receptor 1-regulated pathways: ligand selective activation of Galpha 14-coupled pathways. Eur J Immunol. 2004;34(3):785–95. [DOI] [PubMed] [Google Scholar]
  • 62.Burns JM, Summers BC, Wang Y, Melikian A, Berahovich R, Miao Z, et al. A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J Exp Med. 2006;203(9):2201–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Balabanian K, Lagane B, Infantino S, Chow KY, Harriague J, Moepps B, et al. The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem. 2005;280(42):35760–6. [DOI] [PubMed] [Google Scholar]
  • 64.Levoye A, Balabanian K, Baleux F, Bachelerie F, Lagane B. CXCR7 heterodimerizes with CXCR4 and regulates CXCL12-mediated G protein signaling. Blood. 2009;113(24):6085–93. [DOI] [PubMed] [Google Scholar]
  • 65.Décaillot FM, Kazmi MA, Lin Y, Ray-Saha S, Sakmar TP, Sachdev P. CXCR7/CXCR4 heterodimer constitutively recruits beta-arrestin to enhance cell migration. J Biol Chem. 2011;286(37):32188–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Rajagopal S, Kim J, Ahn S, Craig S, Lam CM, Gerard NP, et al. Beta-arrestin-but not G protein-mediated signaling by the “decoy” receptor CXCR7. Proc Natl Acad Sci U S A. 2010;107(2):628–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wang Y, Li G, Stanco A, Long JE, Crawford D, Potter GB, et al. CXCR4 and CXCR7 have distinct functions in regulating interneuron migration. Neuron. 2011;69(1):61–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ikeda Y, Kumagai H, Skach A, Sato M, Yanagisawa M. Modulation of Circadian Glucocorticoid Oscillation via Adrenal Opioid-CXCR7 Signaling Alters Emotional Behavior. Cell. 2013;155(6):1323–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Meyrath M, Szpakowska M, Zeiner J, Massotte L, Merz MP, Benkel T, et al. The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides. Nature communications. 2020;11(1):3033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Montpas N, St-Onge G, Nama N, Rhainds D, Benredjem B, Girard M, et al. Ligand-specific conformational transitions and intracellular transport are required for atypical chemokine receptor 3-mediated chemokine scavenging. J Biol Chem. 2018;293(3):893–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Saaber F, Schutz D, Miess E, Abe P, Desikan S, Ashok Kumar P, et al. ACKR3 Regulation of Neuronal Migration Requires ACKR3 Phosphorylation, but Not beta-Arrestin. Cell Rep. 2019;26(6):1473–88 e9. [DOI] [PubMed] [Google Scholar]
  • 72.Wise H The roles played by highly truncated splice variants of G protein-coupled receptors. J Mol Signal. 2012;7(1):13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Berchiche YA, Sakmar TP. CXC Chemokine Receptor 3 Alternative Splice Variants Selectively Activate Different Signaling Pathways. Mol Pharmacol. 2016;90(4):483–95. [DOI] [PubMed] [Google Scholar]
  • 74.Singh SP, Foley JF, Zhang HH, Hurt DE, Richards JL, Smith CS, et al. Selectivity in the Use of Gi/o Proteins Is Determined by the DRF Motif in CXCR6 and Is Cell-Type Specific. Mol Pharmacol. 2015;88(5):894–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Urs NM, Gee SM, Pack TF, McCorvy JD, Evron T, Snyder JC, et al. Distinct cortical and striatal actions of a β-arrestin-biased dopamine D2 receptor ligand reveal unique antipsychotic-like properties. Proc Natl Acad Sci U S A. 2016;113(50):E8178–E86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Vroon A, Heijnen CJ, Kavelaars A. GRKs and arrestins: regulators of migration and inflammation. J Leukoc Biol. 2006;80(6):1214–21. [DOI] [PubMed] [Google Scholar]
  • 77.Vroon A, Heijnen CJ, Lombardi MS, Cobelens PM, Mayor F, Caron MG, et al. Reduced GRK2 level in T cells potentiates chemotaxis and signaling in response to CCL4. J Leukoc Biol. 2004;75(5):901–9. [DOI] [PubMed] [Google Scholar]
  • 78.Arnon TI, Xu Y, Lo C, Pham T, An J, Coughlin S, et al. GRK2-dependent S1PR1 desensitization is required for lymphocytes to overcome their attraction to blood. Science. 2011;333(6051):1898–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Mak JCW, Hisada T, Salmon M, Barnes PJ, Chung KF. Glucocorticoids reverse IL-1beta-induced impairment of beta-adrenoceptor-mediated relaxation and up-regulation of G-protein-coupled receptor kinases. British journal of pharmacology. 2002;135(4):987–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lombardi MS, Kavelaars A, Cobelens PM, Schmidt RE, Schedlowski M, Heijnen CJ. Adjuvant arthritis induces down-regulation of G protein-coupled receptor kinases in the immune system. J Immunol. 2001;166(3):1635–40. [DOI] [PubMed] [Google Scholar]
  • 81.Lombardi MS, Kavelaars A, Schedlowski M, Bijlsma JW, Okihara KL, Van de Pol M, et al. Decreased expression and activity of G-protein-coupled receptor kinases in peripheral blood mononuclear cells of patients with rheumatoid arthritis. FASEB J. 1999;13(6):715–25. [DOI] [PubMed] [Google Scholar]
  • 82.Tutunea-Fatan E, Caetano FA, Gros R, Ferguson SS. GRK2 targeted knockdown results in spontaneous hypertension, and altered vascular GPCR signaling. J Biol Chem. 2015;290(8):5141–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Keys JR, Zhou RH, Harris DM, Druckman CA, Eckhart AD. Vascular smooth muscle overexpression of G protein-coupled receptor kinase 5 elevates blood pressure, which segregates with sex and is dependent on Gi-mediated signaling. Circulation. 2005;112(8):1145–53. [DOI] [PubMed] [Google Scholar]
  • 84.Vroon A, Lombardi MS, Kavelaars A, Heijnen CJ. Changes in the G-protein-coupled receptor desensitization machinery during relapsing-progressive experimental allergic encephalomyelitis. J Neuroimmunol. 2003;137(1–2):79–86. [DOI] [PubMed] [Google Scholar]
  • 85.Kufareva I, Gustavsson M, Zheng Y, Stephens BS, Handel TM. What Do Structures Tell Us About Chemokine Receptor Function and Antagonism? Annu Rev Biophys. 2017;46:175–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Gustavsson M, Wang L, van Gils N, Stephens BS, Zhang P, Schall TJ, et al. Structural basis of ligand interaction with atypical chemokine receptor 3. Nat Commun. 2017;8:14135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Burg JS, Ingram JR, Venkatakrishnan AJ, Jude KM, Dukkipati A, Feinberg EN, et al. Structural biology. Structural basis for chemokine recognition and activation of a viral G protein-coupled receptor. Science. 2015;347(6226):1113–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Cox MA, Jenh CH, Gonsiorek W, Fine J, Narula SK, Zavodny PJ, et al. Human interferon-inducible 10-kDa protein and human interferon-inducible T cell alpha chemoattractant are allotopic ligands for human CXCR3: differential binding to receptor states. Mol Pharmacol. 2001;59(4):707–15. [DOI] [PubMed] [Google Scholar]
  • 89.Colvin RA, Campanella GS, Manice LA, Luster AD. CXCR3 requires tyrosine sulfation for ligand binding and a second extracellular loop arginine residue for ligand-induced chemotaxis. Mol Cell Biol. 2006;26(15):5838–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Jaracz-Ros A, Bernadat G, Cutolo P, Gallego C, Gustavsson M, Cecon E, et al. Differential activity and selectivity of N-terminal modified CXCL12 chemokines at the CXCR4 and ACKR3 receptors. J Leukoc Biol. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Szpakowska M, Nevins AM, Meyrath M, Rhainds D, D’huys T, Guité-Vinet F, et al. Different contributions of chemokine N-terminal features attest to a different ligand binding mode and a bias towards activation of ACKR3/CXCR7 compared with CXCR4 and CXCR3. Br J Pharmacol. 2018;175(9):1419–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ngo T, Stephens BS, Gustavsson M, Holden LG, Abagyan R, Handel TM, et al. Crosslinking-guided geometry of a complete CXC receptor-chemokine complex and the basis of chemokine subfamily selectivity. PLoS Biol. 2020;18(4):e3000656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wasilko DJ, Johnson ZL, Ammirati M, Che Y, Griffor MC, Han S, et al. Structural basis for chemokine receptor CCR6 activation by the endogenous protein ligand CCL20. Nat Commun. 2020;11(1):3031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Riutta SJ, Larsen O, Getschman AE, Rosenkilde MM, Hwang ST, Volkman BF. Mutational analysis of CCL20 reveals flexibility of N-terminal amino acid composition and length. J Leukoc Biol. 2018;104(2):423–34. [DOI] [PubMed] [Google Scholar]
  • 95.Nobles KN, Xiao K, Ahn S, Shukla AK, Lam CM, Rajagopal S, et al. Distinct phosphorylation sites on the β(2)-adrenergic receptor establish a barcode that encodes differential functions of β-arrestin. Sci Signal. 2011;4(185):ra51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Doll C, Konietzko J, Pöll F, Koch T, Höllt V, Schulz S. Agonist-selective patterns of μ-opioid receptor phosphorylation revealed by phosphosite-specific antibodies. Br J Pharmacol. 2011;164(2):298–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Butcher AJ, Prihandoko R, Kong KC, McWilliams P, Edwards JM, Bottrill A, et al. Differential G-protein-coupled receptor phosphorylation provides evidence for a signaling bar code. J Biol Chem. 2011;286(13):11506–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kim J, Ahn S, Ren XR, Whalen EJ, Reiter E, Wei H, et al. Functional antagonism of different G protein-coupled receptor kinases for beta-arrestin-mediated angiotensin II receptor signaling. Proc Natl Acad Sci U S A. 2005;102(5):1442–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Komolov KE, Benovic JL. G protein-coupled receptor kinases: Past, present and future. Cell Signal. 2018;41:17–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pitcher JA, Freedman NJ, Lefkowitz RJ. G protein-coupled receptor kinases. Annu Rev Biochem. 1998;67:653–92. [DOI] [PubMed] [Google Scholar]
  • 101.Loudon RP, Benovic JL. Expression, purification, and characterization of the G protein-coupled receptor kinase GRK6. J Biol Chem. 1994;269(36):22691–7. [PubMed] [Google Scholar]
  • 102.Kunapuli P, Onorato JJ, Hosey MM, Benovic JL. Expression, purification, and characterization of the G protein-coupled receptor kinase GRK5. J Biol Chem. 1994;269(2):1099–105. [PubMed] [Google Scholar]
  • 103.Busillo JM, Armando S, Sengupta R, Meucci O, Bouvier M, Benovic JL. Sitespecific phosphorylation of CXCR4 is dynamically regulated by multiple kinases and results in differential modulation of CXCR4 signaling. J Biol Chem. 2010;285(10):780517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mueller W, Schütz D, Nagel F, Schulz S, Stumm R. Hierarchical organization of multi-site phosphorylation at the CXCR4 C terminus. PLoS One. 2013;8(5):e64975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Raghuwanshi SK, Su Y, Singh V, Haynes K, Richmond A, Richardson RM. The chemokine receptors CXCR1 and CXCR2 couple to distinct G protein-coupled receptor kinases to mediate and regulate leukocyte functions. J Immunol. 2012;189(6):2824–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Luo J, Busillo JM, Stumm R, Benovic JL. G Protein-Coupled Receptor Kinase 3 and Protein Kinase C Phosphorylate the Distal C-Terminal Tail of the Chemokine Receptor CXCR4 and Mediate Recruitment of. Mol Pharmacol. 2017;91(6):554–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zheng Y, Qin L, Zacarias NV, de Vries H, Han GW, Gustavsson M, et al. Structure of CC chemokine receptor 2 with orthosteric and allosteric antagonists. Nature. 2016;540(7633):458–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Oswald C, Rappas M, Kean J, Dore AS, Errey JC, Bennett K, et al. Intracellular allosteric antagonism of the CCR9 receptor. Nature. 2016;540(7633):462–5. [DOI] [PubMed] [Google Scholar]
  • 109.Ortiz Zacarias NV, Lenselink EB, AP IJ, Handel TM, Heitman LH. Intracellular Receptor Modulation: Novel Approach to Target GPCRs. Trends Pharmacol Sci. 2018;39(6):547–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wingler LM, Elgeti M, Hilger D, Latorraca NR, Lerch MT, Staus DP, et al. Angiotensin Analogs with Divergent Bias Stabilize Distinct Receptor Conformations. Cell. 2019;176(3):468–78.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Colvin RA, Campanella GS, Sun J, Luster AD. Intracellular domains of CXCR3 that mediate CXCL9, CXCL10, and CXCL11 function. J Biol Chem. 2004;279(29):30219–27. [DOI] [PubMed] [Google Scholar]
  • 112.Otero C, Eisele PS, Schaeuble K, Groettrup M, Legler DF. Distinct motifs in the chemokine receptor CCR7 regulate signal transduction, receptor trafficking and chemotaxis. J Cell Sci. 2008;121(Pt 16):2759–67. [DOI] [PubMed] [Google Scholar]
  • 113.Cahill TJ, Thomsen AR, Tarrasch JT, Plouffe B, Nguyen AH, Yang F, et al. Distinct conformations of GPCR-β-arrestin complexes mediate desensitization, signaling, and endocytosis. Proc Natl Acad Sci U S A. 2017;114(10):2562–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Srivastava A, Gupta B, Gupta C, Shukla AK. Emerging Functional Divergence of β-Arrestin Isoforms in GPCR Function. Trends Endocrinol Metab. 2015;26(11):628–42. [DOI] [PubMed] [Google Scholar]
  • 115.Ghosh E, Dwivedi H, Baidya M, Srivastava A, Kumari P, Stepniewski T, et al. Conformational Sensors and Domain Swapping Reveal Structural and Functional Differences between β-Arrestin Isoforms. Cell Rep. 2019;28(13):3287–99.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Smith JS, Nicholson LT, Suwanpradid J, Glenn RA, Knape NM, Alagesan P, et al. Biased agonists of the chemokine receptor CXCR3 differentially control chemotaxis and inflammation. Sci Signal. 2018;11(555). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Scholten DJ, Canals M, Wijtmans M, de Munnik S, Nguyen P, Verzijl D, et al. Pharmacological characterization of a small-molecule agonist for the chemokine receptor CXCR3. Br J Pharmacol. 2012;166(3):898–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Smit MJ, Verdijk P, van der Raaij-Helmer EM, Navis M, Hensbergen PJ, Leurs R, et al. CXCR3-mediated chemotaxis of human T cells is regulated by a Gi- and phospholipase C-dependent pathway and not via activation of MEK/p44/p42 MAPK nor Akt/PI-3 kinase. Blood. 2003;102(6):1959–65. [DOI] [PubMed] [Google Scholar]
  • 119.Karin N, Wildbaum G, Thelen M. Biased signaling pathways via CXCR3 control the development and function of CD4+ T cell subsets. Journal of Leukocyte Biology. 2016;99(6):857–62. [DOI] [PubMed] [Google Scholar]
  • 120.Zohar Y, Wildbaum G, Novak R, Salzman AL, Thelen M, Alon R, et al. CXCL11-dependent induction of FOXP3-negative regulatory T cells suppresses autoimmune encephalomyelitis. J Clin Invest. 2014;124(5):2009–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Muller M, Carter SL, Hofer MJ, Manders P, Getts DR, Getts MT, et al. CXCR3 signaling reduces the severity of experimental autoimmune encephalomyelitis by controlling the parenchymal distribution of effector and regulatory T cells in the central nervous system. J Immunol. 2007;179(5):2774–86. [DOI] [PubMed] [Google Scholar]
  • 122.Wendt MK, Johanesen PA, Kang-Decker N, Binion DG, Shah V, Dwinell MB. Silencing of epithelial CXCL12 expression by DNA hypermethylation promotes colonic carcinoma metastasis. Oncogene. 2006;25(36):4986–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Wendt MK, Cooper AN, Dwinell MB. Epigenetic silencing of CXCL12 increases the metastatic potential of mammary carcinoma cells. Oncogene. 2008;27(10):1461–71. [DOI] [PubMed] [Google Scholar]
  • 124.Wendt MK, Drury LJ, Vongsa RA, Dwinell MB. Constitutive CXCL12 expression induces anoikis in colorectal carcinoma cells. Gastroenterology. 2008;135(2):508–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lagane B, Chow KY, Balabanian K, Levoye A, Harriague J, Planchenault T, et al. CXCR4 dimerization and beta-arrestin-mediated signaling account for the enhanced chemotaxis to CXCL12 in WHIM syndrome. Blood. 2008;112(1):34–44. [DOI] [PubMed] [Google Scholar]
  • 126.Issafras H, Angers S, Bulenger S, Blanpain C, Parmentier M, Labbe-Jullie C, et al. Constitutive agonist-independent CCR5 oligomerization and antibody-mediated clustering occurring at physiological levels of receptors. J Biol Chem. 2002;277(38):34666–73. [DOI] [PubMed] [Google Scholar]
  • 127.Percherancier Y, Berchiche YA, Slight I, Volkmer-Engert R, Tamamura H, Fujii N, et al. Bioluminescence resonance energy transfer reveals ligand-induced conformational changes in CXCR4 homo- and heterodimers. J Biol Chem. 2005;280(11):9895–903. [DOI] [PubMed] [Google Scholar]
  • 128.Trettel F, Di Bartolomeo S, Lauro C, Catalano M, Ciotti MT, Limatola C. Ligand-independent CXCR2 dimerization. J Biol Chem. 2003;278(42):40980–8. [DOI] [PubMed] [Google Scholar]
  • 129.Forster R, Schubel A, Breitfeld D, Kremmer E, Renner-Muller I, Wolf E, et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell. 1999;99(1):23–33. [DOI] [PubMed] [Google Scholar]
  • 130.Bardi G, Lipp M, Baggiolini M, Loetscher P. The T cell chemokine receptor CCR7 is internalized on stimulation with ELC, but not with SLC. Eur J Immunol. 2001;31(11):3291–7. [DOI] [PubMed] [Google Scholar]
  • 131.Britschgi MR, Favre S, Luther SA. CCL21 is sufficient to mediate DC migration, maturation and function in the absence of CCL19. Eur J Immunol. 2010;40(5):1266–71. [DOI] [PubMed] [Google Scholar]
  • 132.Zweemer AJ, Toraskar J, Heitman LH, AP IJ. Bias in chemokine receptor signalling. Trends Immunol. 2014;35(6):243–52. [DOI] [PubMed] [Google Scholar]
  • 133.Lieberman-Blum SS, Fung HB, Bandres JC. Maraviroc: a CCR5-receptor antagonist for the treatment of HIV-1 infection. Clin Ther. 2008;30(7):1228–50. [DOI] [PubMed] [Google Scholar]
  • 134.Fatkenheuer G, Pozniak AL, Johnson MA, Plettenberg A, Staszewski S, Hoepelman AI, et al. Efficacy of short-term monotherapy with maraviroc, a new CCR5 antagonist, in patients infected with HIV-1. Nat Med. 2005;11(11):1170–2. [DOI] [PubMed] [Google Scholar]
  • 135.De Clercq E The AMD3100 story: the path to the discovery of a stem cell mobilizer (Mozobil). Biochem Pharmacol. 2009;77(11):1655–64. [DOI] [PubMed] [Google Scholar]
  • 136.De Clercq E, Yamamoto N, Pauwels R, Balzarini J, Witvrouw M, De Vreese K, et al. Highly potent and selective inhibition of human immunodeficiency virus by the bicyclam derivative JM3100. Antimicrob Agents Chemother. 1994;38(4):668–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Donzella GA, Schols D, Lin SW, Este JA, Nagashima KA, Maddon PJ, et al. AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 co-receptor. Nat Med. 1998;4(1):72–7. [DOI] [PubMed] [Google Scholar]
  • 138.De Clercq E Mozobil(R) (Plerixafor, AMD3100), 10 years after its approval by the US Food and Drug Administration. Antivir Chem Chemother. 2019;27:2040206619829382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Fricker SP. Physiology and pharmacology of plerixafor. Transfus Med Hemother. 2013;40(4):237–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ollila TA, Sahin I, Olszewski AJ. Mogamulizumab: a new tool for management of cutaneous T-cell lymphoma. Onco Targets Ther. 2019;12:1085–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Mullard A FDA approves second GPCR-targeted antibody. Nat Rev Drug Discov. 2018;17(9):613. [DOI] [PubMed] [Google Scholar]
  • 142.Miao M, De Clercq E, Li G. Clinical significance of chemokine receptor antagonists. Expert Opin Drug Metab Toxicol. 2020;16(1):11–30. [DOI] [PubMed] [Google Scholar]
  • 143.Swaminath G, Xiang Y, Lee TW, Steenhuis J, Parnot C, Kobilka BK. Sequential binding of agonists to the beta2 adrenoceptor. Kinetic evidence for intermediate conformational states. J Biol Chem. 2004;279(1):686–91. [DOI] [PubMed] [Google Scholar]
  • 144.Xiang Y, Kobilka BK. Myocyte adrenoceptor signaling pathways. Science. 2003;300(5625):1530–2. [DOI] [PubMed] [Google Scholar]
  • 145.Neumann L, Wohland T, Whelan RJ, Zare RN, Kobilka BK. Functional immobilization of a ligand-activated G-protein-coupled receptor. Chembiochem. 2002;3(10):993–8. [DOI] [PubMed] [Google Scholar]
  • 146.Efficacy and Safety Study of Mavorixafor in Participants With Warts, Hypogammaglobulinemia, Infections, and Myelokathexis (WHIM) Syndrome. https://ClinicalTrials.gov/show/NCT03995108.
  • 147.Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, et al. CC CKR5: a RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science. 1996;272(5270):1955–8. [DOI] [PubMed] [Google Scholar]
  • 148.Choe H, Farzan M, Sun Y, Sullivan N, Rollins B, Ponath PD, et al. The beta-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell. 1996;85(7):1135–48. [DOI] [PubMed] [Google Scholar]
  • 149.Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, et al. Identification of a major co-receptor for primary isolates of HIV-1. Nature. 1996;381(6584):661–6. [DOI] [PubMed] [Google Scholar]
  • 150.Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature. 1996;381(6584):667–73. [DOI] [PubMed] [Google Scholar]
  • 151.Doranz BJ, Rucker J, Yi Y, Smyth RJ, Samson M, Peiper SC, et al. A dual-tropic primary HIV-1 isolate that uses fusin and the beta-chemokine receptors CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell. 1996;85(7):1149–58. [DOI] [PubMed] [Google Scholar]
  • 152.Liu R, Paxton WA, Choe S, Ceradini D, Martin SR, Horuk R, et al. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell. 1996;86(3):367–77. [DOI] [PubMed] [Google Scholar]
  • 153.Samson M, Libert F, Doranz BJ, Rucker J, Liesnard C, Farber CM, et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature. 1996;382(6593):722–5. [DOI] [PubMed] [Google Scholar]
  • 154.Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science 1996;273(5283):1856–62. [DOI] [PubMed] [Google Scholar]
  • 155.Huang Y, Paxton WA, Wolinsky SM, Neumann AU, Zhang L, He T, et al. The role of a mutant CCR5 allele in HIV-1 transmission and disease progression. Nat Med. 1996;2(11):1240–3. [DOI] [PubMed] [Google Scholar]
  • 156.Paxton WA, Martin SR, Tse D, O’Brien TR, Skurnick J, VanDevanter NL, et al. Relative resistance to HIV-1 infection of CD4 lymphocytes from persons who remain uninfected despite multiple high-risk sexual exposure. Nat Med. 1996;2(4):412–7. [DOI] [PubMed] [Google Scholar]
  • 157.Veljkovic N, Vucicevic J, Tassini S, Glisic S, Veljkovic V, Radi M. Preclinical discovery and development of maraviroc for the treatment of HIV. Expert Opin Drug Discov. 2015;10(6):671–84. [DOI] [PubMed] [Google Scholar]
  • 158.De Luca A, Pezzotti P, Boucher C, Doring M, Incardona F, Kaiser R, et al. Clinical use, efficacy, and durability of maraviroc for antiretroviral therapy in routine care: A European survey. PLoS One. 2019;14(11):e0225381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Hogan CM, Hammer SM. Host determinants in HIV infection and disease. Part 2: genetic factors and implications for antiretroviral therapeutics. Ann Intern Med. 2001;134(10):978–96. [DOI] [PubMed] [Google Scholar]
  • 160.Mack M, Luckow B, Nelson PJ, Cihak J, Simmons G, Clapham PR, et al. Aminooxypentane-RANTES induces CCR5 internalization but inhibits recycling: a novel inhibitory mechanism of HIV infectivity. J Exp Med. 1998;187(8):1215–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Marozsan AJ, Torre VS, Johnson M, Ball SC, Cross JV, Templeton DJ, et al. Mechanisms involved in stimulation of human immunodeficiency virus type 1 replication by aminooxypentane RANTES. J Virol. 2001;75(18):8624–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Gaertner H, Cerini F, Escola JM, Kuenzi G, Melotti A, Offord R, et al. Highly potent, fully recombinant anti-HIV chemokines: reengineering a low-cost microbicide. Proc Natl Acad Sci U S A. 2008;105(46):17706–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Galvin SR, Cohen MS. The role of sexually transmitted diseases in HIV transmission. Nat Rev Microbiol. 2004;2(1):33–42. [DOI] [PubMed] [Google Scholar]
  • 164.Bonsch C, Munteanu M, Rossitto-Borlat I, Furstenberg A, Hartley O. Potent Anti-HIV Chemokine Analogs Direct Post-Endocytic Sorting of CCR5. PLoS One. 2015;10(4):e0125396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Metzemaekers M, Vanheule V, Janssens R, Struyf S, Proost P. Overview of the Mechanisms that May Contribute to the Non-Redundant Activities of Interferon-Inducible CXC Chemokine Receptor 3 Ligands. Front Immunol. 2017;8:1970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Groom JR, Luster AD. CXCR3 in T cell function. Exp Cell Res. 2011;317(5):620–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Kuo PT, Zeng Z, Salim N, Mattarollo S, Wells JW, Leggatt GR. The Role of CXCR3 and Its Chemokine Ligands in Skin Disease and Cancer. Front Med (Lausanne). 2018;5:271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Hitchinson B, Eby JM, Gao X, Guite-Vinet F, Ziarek JJ, Abdelkarim H, et al. Biased antagonism of CXCR4 avoids antagonist tolerance. Sci Signal. 2018;11(552). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Corbisier J, Huszagh A, Galés C, Parmentier M, Springael J-Y. Partial agonist and biased signaling properties of the synthetic enantiomers J113863/UCB35625 at chemokine receptors CCR2 and CCR5. 2016:jbc.M116.757559. [DOI] [PMC free article] [PubMed]
  • 170.Feniger-Barish R, Ran M, Zaslaver A, Ben-Baruch A. Differential modes of regulation of cxc chemokine-induced internalization and recycling of human CXCR1 and CXCR2. Cytokine. 1999;11(12):996–1009. [DOI] [PubMed] [Google Scholar]
  • 171.Allegretti M, Bertini R, Cesta MC, Bizzarri C, Di Bitondo R, Di Cioccio V, et al. 2-Arylpropionic CXC chemokine receptor 1 (CXCR1) ligands as novel noncompetitive CXCL8 inhibitors. J Med Chem. 2005;48(13):4312–31. [DOI] [PubMed] [Google Scholar]
  • 172.Bertini R, Allegretti M, Bizzarri C, Moriconi A, Locati M, Zampella G, et al. Noncompetitive allosteric inhibitors of the inflammatory chemokine receptors CXCR1 and CXCR2: prevention of reperfusion injury. Proc Natl Acad Sci U S A. 2004;101(32):11791–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Kim HY, Choi JH, Kang YJ, Park SY, Choi HC, Kim HS. Reparixin, an inhibitor of CXCR1 and CXCR2 receptor activation, attenuates blood pressure and hypertension-related mediators expression in spontaneously hypertensive rats. Biol Pharm Bull. 2011;34(1):120–7. [DOI] [PubMed] [Google Scholar]
  • 174.Bertini R, Barcelos LS, Beccari AR, Cavalieri B, Moriconi A, Bizzarri C, et al. Receptor binding mode and pharmacological characterization of a potent and selective dual CXCR1/CXCR2 non-competitive allosteric inhibitor. Br J Pharmacol. 2012;165(2):436–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Maeda DY, Peck AM, Schuler AD, Quinn MT, Kirpotina LN, Wicomb WN, et al. Discovery of 2-[5-(4-Fluorophenylcarbamoyl)pyridin-2-ylsulfanylmethyl]phenylboronic Acid (SX-517): Noncompetitive Boronic Acid Antagonist of CXCR1 and CXCR2. J Med Chem. 2014;57(20):8378–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Matzer SP, Zombou J, Sarau HM, Rollinghoff M, Beuscher HU. A synthetic, non-peptide CXCR2 antagonist blocks MIP-2-induced neutrophil migration in mice. Immunobiology. 2004;209(3):225–33. [DOI] [PubMed] [Google Scholar]
  • 177.Rennard SI, Dale DC, Donohue JF, Kanniess F, Magnussen H, Sutherland ER, et al. CXCR2 Antagonist MK-7123. A Phase 2 Proof-of-Concept Trial for Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2015;191(9):1001–11. [DOI] [PubMed] [Google Scholar]
  • 178.Efficacy and Safety Study of Navarixin (MK-7123) in Combination With Pembrolizumab (MK-3475) in Adults With Selected Advanced/Metastatic Solid Tumors (MK-7123–034): https://ClinicalTrials.gov/show/NCT03473925; 2018. [
  • 179.Dwyer MP, Yu Y, Chao J, Aki C, Chao J, Biju P, et al. Discovery of 2-hydroxy-N,N-dimethyl-3-{2-[[(R)-1-(5- methylfuran-2-yl)propyl]amino]-3,4-dioxocyclobut-1-enylamino}benzamide (SCH 527123): a potent, orally bioavailable CXCR2/CXCR1 receptor antagonist. J Med Chem. 2006;49(26):7603–6. [DOI] [PubMed] [Google Scholar]
  • 180.Busch-Petersen J, Carpenter DC, Burman M, Foley J, Hunsberger GE, Kilian DJ, et al. Danirixin: A Reversible and Selective Antagonist of the CXC Chemokine Receptor 2. J Pharmacol Exp Ther. 2017;362(2):338–46. [DOI] [PubMed] [Google Scholar]
  • 181.Lazaar AL, Miller BE, Tabberer M, Yonchuk J, Leidy N, Ambery C, et al. Effect of the CXCR2 antagonist danirixin on symptoms and health status in COPD. Eur Respir J. 2018;52(4). [DOI] [PubMed] [Google Scholar]
  • 182.Miller BE, Mistry S, Smart K, Connolly P, Carpenter DC, Cooray H, et al. The pharmacokinetics and pharmacodynamics of danirixin (GSK1325756)--a selective CXCR2 antagonist --in healthy adult subjects. BMC Pharmacol Toxicol. 2015;16:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Bento AF, Leite DF, Claudino RF, Hara DB, Leal PC, Calixto JB. The selective nonpeptide CXCR2 antagonist SB225002 ameliorates acute experimental colitis in mice. J Leukoc Biol. 2008;84(4):1213–21. [DOI] [PubMed] [Google Scholar]
  • 184.White JR, Lee JM, Young PR, Hertzberg RP, Jurewicz AJ, Chaikin MA, et al. Identification of a potent, selective non-peptide CXCR2 antagonist that inhibits interleukin-8-induced neutrophil migration. J Biol Chem. 1998;273(17):10095–8. [DOI] [PubMed] [Google Scholar]
  • 185.Nicholls DJ, Wiley K, Dainty I, MacIntosh F, Phillips C, Gaw A, et al. Pharmacological characterization of AZD5069, a slowly reversible CXC chemokine receptor 2 antagonist. J Pharmacol Exp Ther. 2015;353(2):340–50. [DOI] [PubMed] [Google Scholar]
  • 186.Sauty A, Colvin RA, Wagner L, Rochat S, Spertini F, Luster AD. CXCR3 internalization following T cell-endothelial cell contact: preferential role of IFN-inducible T cell alpha chemoattractant (CXCL11). J Immunol. 2001;167(12):7084–93. [DOI] [PubMed] [Google Scholar]
  • 187.Müller M, Carter SL, Hofer MJ, Manders P, Getts DR, Getts MT, et al. CXCR3 Signaling Reduces the Severity of Experimental Autoimmune Encephalomyelitis by Controlling the Parenchymal Distribution of Effector and Regulatory T Cells in the Central Nervous System. The Journal of Immunology. 2007;179(5):2774–86. [DOI] [PubMed] [Google Scholar]
  • 188.Scholten D, Canals M, Wijtmans M, De Munnik S, Nguyen P, Verzijl D, et al. Pharmacological characterization of a small-molecule agonist for the chemokine receptor CXCR3. 2012;166(3):898–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Smith JS, Nicholson LT, Suwanpradid J, Glenn RA, Knape NM, Alagesan P, et al. Biased agonists of the chemokine receptor CXCR3 differentially control chemotaxis and inflammation. Science Signaling. 2018;11(555):eaaq1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Milanos L, Brox R, Frank T, Poklukar G, Palmisano R, Waibel R, et al. Discovery and Characterization of Biased Allosteric Agonists of the Chemokine Receptor CXCR3. Journal of Medicinal Chemistry. 2016;59(5):2222–43. [DOI] [PubMed] [Google Scholar]
  • 191.Brox R, Milanos L, Saleh N, Baumeister P, Buschauer A, Hofmann D, et al. Molecular Mechanisms of Biased and Probe-Dependent Signaling at CXC-Motif Chemokine Receptor CXCR3 Induced by Negative Allosteric Modulators. Mol Pharmacol. 2018;93(4):309–22. [DOI] [PubMed] [Google Scholar]
  • 192.Boye K, Billottet C, Pujol N, Alves ID, Bikfalvi A. Ligand activation induces different conformational changes in CXCR3 receptor isoforms as evidenced by plasmon waveguide resonance (PWR). Sci Rep. 2017;7(1):10703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Jenh CH, Cox MA, Cui L, Reich EP, Sullivan L, Chen SC, et al. A selective and potent CXCR3 antagonist SCH 546738 attenuates the development of autoimmune diseases and delays graft rejection. BMC Immunol. 2012;13:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Bernat V, Brox R, Heinrich MR, Auberson YP, Tschammer N. Ligand-biased and probe-dependent modulation of chemokine receptor CXCR3 signaling by negative allosteric modulators. ChemMedChem. 2015;10(3):566–74. [DOI] [PubMed] [Google Scholar]
  • 195.Hesselgesser J, Ng HP, Liang M, Zheng W, May K, Bauman JG, et al. Identification and characterization of small molecule functional antagonists of the CCR1 chemokine receptor. J Biol Chem. 1998;273(25):15687–92. [DOI] [PubMed] [Google Scholar]
  • 196.Zirafi O, Kim KA, Standker L, Mohr KB, Sauter D, Heigele A, et al. Discovery and characterization of an endogenous CXCR4 antagonist. Cell Rep. 2015;11(5):737–47. [DOI] [PubMed] [Google Scholar]
  • 197.Burger JA, Stewart DJ. CXCR4 chemokine receptor antagonists: perspectives in SCLC. Expert Opin Investig Drugs. 2009;18(4):481–90. [DOI] [PubMed] [Google Scholar]
  • 198.Safety and PK/PD of TG-0054 in Multiple Myeloma, Non-Hodgkin Lymphoma and Hodgkin Disease Patients: https://ClinicalTrials.gov/show/NCT01018979; 2015. [
  • 199.Shelke NB, Kadam R, Tyagi P, Rao VR, Kompella UB. Intravitreal Poly(L-lactide) Microparticles Sustain Retinal and Choroidal Delivery of TG-0054, a Hydrophilic Drug Intended for Neovascular Diseases. Drug Deliv Transl Res. 2011;1(1):76–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Hsu WT, Jui HY, Huang YH, Su MY, Wu YW, Tseng WY, et al. CXCR4 Antagonist TG-0054 Mobilizes Mesenchymal Stem Cells, Attenuates Inflammation, and Preserves Cardiac Systolic Function in a Porcine Model of Myocardial Infarction. Cell Transplant. 2015;24(7):1313–28. [DOI] [PubMed] [Google Scholar]
  • 201.Safety and Activity of the Oral HIV Entry Inhibitor AMD11070 in HIV Infected Patients: https://ClinicalTrials.gov/show/NCT00089466; 2012. [
  • 202.Bridger GJ, Skerlj RT, Hernandez-Abad PE, Bogucki DE, Wang Z, Zhou Y, et al. Synthesis and structure-activity relationships of azamacrocyclic C-X-C chemokine receptor 4 antagonists: analogues containing a single azamacrocyclic ring are potent inhibitors of T-cell tropic (X4) HIV-1 replication. J Med Chem. 2010;53(3):1250–60. [DOI] [PubMed] [Google Scholar]
  • 203.Skerlj RT, Bridger GJ, Kaller A, McEachern EJ, Crawford JB, Zhou Y, et al. Discovery of novel small molecule orally bioavailable C-X-C chemokine receptor 4 antagonists that are potent inhibitors of T-tropic (X4) HIV-1 replication. J Med Chem. 2010;53(8):3376–88. [DOI] [PubMed] [Google Scholar]
  • 204.A Dose Determination and Safety Study of X4P-001 (Mavorixafor) in Participants With Warts, Hypogammaglobulinemia, Infections, and Myelokathexis (WHIM) Syndrome: https://ClinicalTrials.gov/show/NCT03005327; 2019. [
  • 205.X4P-001 and Pembrolizumab in Patients With Advanced Melanoma: https://ClinicalTrials.gov/show/NCT02823405; 2019. [
  • 206.MSX-122 Administered Orally in Patients With Refractory Metastatic or Locally Advanced Solid Tumors: https://ClinicalTrials.gov/show/NCT00591682; 2008. [
  • 207.Liang Z, Zhan W, Zhu A, Yoon Y, Lin S, Sasaki M, et al. Development of a unique small molecule modulator of CXCR4. PLoS One. 2012;7(4):e34038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Wong D, Korz W. Translating an Antagonist of Chemokine Receptor CXCR4: from bench to bedside. Clin Cancer Res. 2008;14(24):7975–80. [DOI] [PubMed] [Google Scholar]
  • 209.Safety and Efficacy of POL6326 for Mobilization/Transplant of Sibling Donor in Patients With Hematologic Malignancies: https://ClinicalTrials.gov/show/NCT01413568; 2011. [
  • 210.Sachpatzidis A, Benton BK, Manfredi JP, Wang H, Hamilton A, Dohlman HG, et al. Identification of allosteric peptide agonists of CXCR4. J Biol Chem. 2003;278(2):896907. [DOI] [PubMed] [Google Scholar]
  • 211.Quoyer J, Janz JM, Luo J, Ren Y, Armando S, Lukashova V, et al. Pepducin targeting the C-X-C chemokine receptor type 4 acts as a biased agonist favoring activation of the inhibitory G protein. Proc Natl Acad Sci U S A. 2013;110(52):E5088–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Tchernychev B, Ren Y, Sachdev P, Janz JM, Haggis L, O’Shea A, et al. Discovery of a CXCR4 agonist pepducin that mobilizes bone marrow hematopoietic cells. Proc Natl Acad Sci U S A. 2010;107(51):22255–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Dimond P, Carlson K, Bouvier M, Gerard C, Xu L, Covic L, et al. G protein-coupled receptor modulation with pepducins: moving closer to the clinic. Ann N Y Acad Sci. 2011;1226:34–49. [DOI] [PubMed] [Google Scholar]
  • 214.Xu Y, Duggineni S, Espitia S, Richman DD, An J, Huang Z. A synthetic bivalent ligand of CXCR4 inhibits HIV infection. Biochem Biophys Res Commun. 2013;435(4):646–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Murakami T, Nakajima T, Koyanagi Y, Tachibana K, Fujii N, Tamamura H, et al. A small molecule CXCR4 inhibitor that blocks T cell line-tropic HIV-1 infection. J Exp Med. 1997;186(8):1389–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Karpova D, Brauninger S, Wiercinska E, Kramer A, Stock B, Graff J, et al. Mobilization of hematopoietic stem cells with the novel CXCR4 antagonist POL6326 (balixafortide) in healthy volunteers-results of a dose escalation trial. J Transl Med. 2017;15(1):2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Pernas S, Martin M, Kaufman PA, Gil-Martin M, Gomez Pardo P, Lopez-Tarruella S, et al. Balixafortide plus eribulin in HER2-negative metastatic breast cancer: a phase 1, single-arm, dose-escalation trial. Lancet Oncol. 2018;19(6):812–24. [DOI] [PubMed] [Google Scholar]
  • 218.Pivotal Study in HER2 Negative, Locally Recurrent or Metastatic Breast Cancer: https://ClinicalTrials.gov/show/NCT03786094; 2018. [
  • 219.Tamamura H, Xu Y, Hattori T, Zhang X, Arakaki R, Kanbara K, et al. A low-molecular-weight inhibitor against the chemokine receptor CXCR4: a strong anti-HIV peptide T140. Biochem Biophys Res Commun. 1998;253(3):877–82. [DOI] [PubMed] [Google Scholar]
  • 220.Lefrancois M, Lefebvre MR, Saint-Onge G, Boulais PE, Lamothe S, Leduc R, et al. Agonists for the Chemokine Receptor CXCR4. ACS Med Chem Lett. 2011;2(8):597602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Mona CE, Besserer-Offroy E, Cabana J, Leduc R, Lavigne P, Heveker N, et al. Design, synthesis, and biological evaluation of CXCR4 ligands. Org Biomol Chem. 2016;14(43):10298–311. [DOI] [PubMed] [Google Scholar]
  • 222.Kaneider NC, Agarwal A, Leger AJ, Kuliopulos A. Reversing systemic inflammatory response syndrome with chemokine receptor pepducins. Nat Med. 2005;11(6):661–5. [DOI] [PubMed] [Google Scholar]
  • 223.O’Callaghan K, Lee L, Nguyen N, Hsieh MY, Kaneider NC, Klein AK, et al. Targeting CXCR4 with cell-penetrating pepducins in lymphoma and lymphocytic leukemia. Blood. 2012;119(7):1717–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Zhu A, Zhan W, Liang Z, Yoon Y, Yang H, Grossniklaus HE, et al. Dipyrimidine amines: a novel class of chemokine receptor type 4 antagonists with high specificity. J Med Chem. 2010;53(24):8556–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Jenkinson S, Thomson M, McCoy D, Edelstein M, Danehower S, Lawrence W, et al. Blockade of X4-tropic HIV-1 cellular entry by GSK812397, a potent noncompetitive CXCR4 receptor antagonist. Antimicrob Agents Chemother. 2010;54(2):817–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Truax VM, Zhao H, Katzman BM, Prosser AR, Alcaraz AA, Saindane MT, et al. Discovery of tetrahydroisoquinoline-based CXCR4 antagonists. ACS Med Chem Lett. 2013;4(11):1025–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Thoma G, Streiff MB, Kovarik J, Glickman F, Wagner T, Beerli C, et al. Orally bioavailable isothioureas block function of the chemokine receptor CXCR4 in vitro and in vivo. J Med Chem. 2008;51(24):7915–20. [DOI] [PubMed] [Google Scholar]
  • 228.Wu B, Chien EY, Mol CD, Fenalti G, Liu W, Katritch V, et al. Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists. Science. 2010;330(6007):1066–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Zhou N, Fang J, Acheampong E, Mukhtar M, Pomerantz RJ. Binding of ALX40–4C to APJ, a CNS-based receptor, inhibits its utilization as a co-receptor by HIV-1. Virology. 2003;312(1):196–203. [DOI] [PubMed] [Google Scholar]
  • 230.Doranz BJ, Filion LG, Diaz-Mitoma F, Sitar DS, Sahai J, Baribaud F, et al. Safe use of the CXCR4 inhibitor ALX40–4C in humans. AIDS Res Hum Retroviruses. 2001;17(6):475–86. [DOI] [PubMed] [Google Scholar]
  • 231.Chu F, Li HS, Liu X, Cao J, Ma W, Ma Y, et al. CXCR5(+)CD8(+) T cells are a distinct functional subset with an antitumor activity. Leukemia. 2019;33(11):2640–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Kazanietz MG, Durando M, Cooke M. CXCL13 and Its Receptor CXCR5 in Cancer: Inflammation, Immune Response, and Beyond. Front Endocrinol (Lausanne). 2019;10:471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Ashhurst AS, Florido M, Lin LCW, Quan D, Armitage E, Stifter SA, et al. CXCR6-Deficiency Improves the Control of Pulmonary Mycobacterium tuberculosis and Influenza Infection Independent of T-Lymphocyte Recruitment to the Lungs. Front Immunol. 2019;10:339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Wein AN, McMaster SR, Takamura S, Dunbar PR, Cartwright EK, Hayward SL, et al. CXCR6 regulates localization of tissue-resident memory CD8 T cells to the airways. J Exp Med. 2019;216(12):2748–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Peddibhotla S, Hershberger PM, Jason Kirby R, Sugarman E, Maloney PR, Hampton Sessions E, et al. Discovery of small molecule antagonists of chemokine receptor CXCR6 that arrest tumor growth in SK-HEP-1 mouse xenografts as a model of hepatocellular carcinoma. Bioorg Med Chem Lett. 2020;30(4):126899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Sabroe I, Peck MJ, Van Keulen BJ, Jorritsma A, Simmons G, Clapham PR, et al. A small molecule antagonist of chemokine receptors CCR1 and CCR3. Potent inhibition of eosinophil function and CCR3-mediated HIV-1 entry. J Biol Chem. 2000;275(34):25985–92. [DOI] [PubMed] [Google Scholar]
  • 237.de Mendonca FL, da Fonseca PC, Phillips RM, Saldanha JW, Williams TJ, Pease JE. Site-directed mutagenesis of CC chemokine receptor 1 reveals the mechanism of action of UCB 35625, a small molecule chemokine receptor antagonist. J Biol Chem. 2005;280(6):4808–16. [DOI] [PubMed] [Google Scholar]
  • 238.Wise EL, Duchesnes C, da Fonseca PC, Allen RA, Williams TJ, Pease JE. Small molecule receptor agonists and antagonists of CCR3 provide insight into mechanisms of chemokine receptor activation. J Biol Chem. 2007;282(38):27935–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Amat M, Benjamim CF, Williams LM, Prats N, Terricabras E, Beleta J, et al. Pharmacological blockade of CCR1 ameliorates murine arthritis and alters cytokine networks in vivo. Br J Pharmacol. 2006;149(6):666–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Shahrara S, Proudfoot AE, Woods JM, Ruth JH, Amin MA, Park CC, et al. Amelioration of rat adjuvant-induced arthritis by Met-RANTES. Arthritis Rheum. 2005;52(6):1907–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Plater-Zyberk C, Hoogewerf AJ, Proudfoot AE, Power CA, Wells TN. Effect of a CC chemokine receptor antagonist on collagen induced arthritis in DBA/1 mice. Immunol Lett. 1997;57(1–3):117–20. [DOI] [PubMed] [Google Scholar]
  • 242.Huang Y, Jiao S, Tao X, Tang Q, Jiao W, Xiao J, et al. Met-CCL5 represents an immunotherapy strategy to ameliorate rabies virus infection. J Neuroinflammation. 2014;11:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Dairaghi DJ, Zhang P, Wang Y, Seitz LC, Johnson DA, Miao S, et al. Pharmacokinetic and pharmacodynamic evaluation of the novel CCR1 antagonist CCX354 in healthy human subjects: implications for selection of clinical dose. Clin Pharmacol Ther. 2011;89(5):726–34. [DOI] [PubMed] [Google Scholar]
  • 244.Gladue RP, Tylaska LA, Brissette WH, Lira PD, Kath JC, Poss CS, et al. CP-481,715, a potent and selective CCR1 antagonist with potential therapeutic implications for inflammatory diseases. J Biol Chem. 2003;278(42):40473–80. [DOI] [PubMed] [Google Scholar]
  • 245.Gladue RP, Brown MF, Zwillich SH. CCR1 antagonists: what have we learned from clinical trials. Curr Top Med Chem. 2010;10(13):1268–77. [DOI] [PubMed] [Google Scholar]
  • 246.Vergunst CE, Gerlag DM, von Moltke L, Karol M, Wyant T, Chi X, et al. MLN3897 plus methotrexate in patients with rheumatoid arthritis: safety, efficacy, pharmacokinetics, and pharmacodynamics of an oral CCR1 antagonist in a phase IIa, double-blind, placebo-controlled, randomized, proof-of-concept study. Arthritis Rheum. 2009;60(12):3572–81. [DOI] [PubMed] [Google Scholar]
  • 247.Tak PP, Balanescu A, Tseluyko V, Bojin S, Drescher E, Dairaghi D, et al. Chemokine receptor CCR1 antagonist CCX354-C treatment for rheumatoid arthritis: CARAT-2, a randomised, placebo controlled clinical trial. Ann Rheum Dis. 2013;72(3):337–44. [DOI] [PubMed] [Google Scholar]
  • 248.Jensen PC, Thiele S, Steen A, Elder A, Kolbeck R, Ghosh S, et al. Reversed binding of a small molecule ligand in homologous chemokine receptors - differential role of extracellular loop 2. Br J Pharmacol. 2012;166(1):258–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Berchiche YA, Gravel S, Pelletier ME, St-Onge G, Heveker N. Different effects of the different natural CC chemokine receptor 2b ligands on beta-arrestin recruitment, Galphai signaling, and receptor internalization. Mol Pharmacol. 2011;79(3):488–98. [DOI] [PubMed] [Google Scholar]
  • 250.An J, Xue Y, Long M, Zhang G, Zhang J, Su H. Targeting CCR2 with its antagonist suppresses viability, motility and invasion by downregulating MMP-9 expression in non-small cell lung cancer cells. Oncotarget. 2017;8(24):39230–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Shin N, Baribaud F, Wang K, Yang G, Wynn R, Covington MB, et al. Pharmacological characterization of INCB3344, a small molecule antagonist of human CCR2. Biochem Biophys Res Commun. 2009;387(2):251–5. [DOI] [PubMed] [Google Scholar]
  • 252.Zweemer AJ, Nederpelt I, Vrieling H, Hafith S, Doornbos ML, de Vries H, et al. Multiple binding sites for small-molecule antagonists at the CC chemokine receptor 2. Mol Pharmacol. 2013;84(4):551–61. [DOI] [PubMed] [Google Scholar]
  • 253.Vilums M, Zweemer AJ, Dekkers S, Askar Y, de Vries H, Saunders J, et al. Design and synthesis of novel small molecule CCR2 antagonists: evaluation of 4aminopiperidine derivatives. Bioorg Med Chem Lett. 2014;24(23):5377–80. [DOI] [PubMed] [Google Scholar]
  • 254.Bot I, Ortiz Zacarias NV, de Witte WE, de Vries H, van Santbrink PJ, van der Velden D, et al. A novel CCR2 antagonist inhibits atherogenesis in apoE deficient mice by achieving high receptor occupancy. Sci Rep. 2017;7(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Wisniewski T, Bayne E, Flanagan J, Shao Q, Wnek R, Matheravidathu S, et al. Assessment of chemokine receptor function on monocytes in whole blood: In vitro and ex vivo evaluations of a CCR2 antagonist. J Immunol Methods. 2010;352(1–2):101–10. [DOI] [PubMed] [Google Scholar]
  • 256.Mueller A, Mahmoud NG, Goedecke MC, McKeating JA, Strange PG. Pharmacological characterization of the chemokine receptor, CCR5. Br J Pharmacol. 2002;135(4):1033–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.White JR, Lee JM, Dede K, Imburgia CS, Jurewicz AJ, Chan G, et al. Identification of potent, selective non-peptide CC chemokine receptor-3 antagonist that inhibits eotaxin-, eotaxin-2-, and monocyte chemotactic protein-4-induced eosinophil migration. J Biol Chem. 2000;275(47):36626–31. [DOI] [PubMed] [Google Scholar]
  • 258.Grozdanovic M, Laffey KG, Abdelkarim H, Hitchinson B, Harijith A, Moon HG, et al. Novel peptide nanoparticle-biased antagonist of CCR3 blocks eosinophil recruitment and airway hyperresponsiveness. J Allergy Clin Immunol. 2019;143(2):669–80 e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Neighbour H, Boulet LP, Lemiere C, Sehmi R, Leigh R, Sousa AR, et al. Safety and efficacy of an oral CCR3 antagonist in patients with asthma and eosinophilic bronchitis: a randomized, placebo-controlled clinical trial. Clin Exp Allergy. 2014;44(4):508–16. [DOI] [PubMed] [Google Scholar]
  • 260.Viney JM, Andrew DP, Phillips RM, Meiser A, Patel P, Lennartz-Walker M, et al. Distinct conformations of the chemokine receptor CCR4 with implications for its targeting in allergy. J Immunol. 2014;192(7):3419–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Bonner K, Pease JE, Corrigan CJ, Clark P, Kay AB. CCL17/thymus and activation-regulated chemokine induces calcitonin gene-related peptide in human airway epithelial cells through CCR4. J Allergy Clin Immunol. 2013;132(4):942-50 e1-3. [DOI] [PubMed] [Google Scholar]
  • 262.Mariani M, Lang R, Binda E, Panina-Bordignon P, D’Ambrosio D. Dominance of CCL22 over CCL17 in induction of chemokine receptor CCR4 desensitization and internalization on human Th2 cells. Eur J Immunol. 2004;34(1):231–40. [DOI] [PubMed] [Google Scholar]
  • 263.Zhang Y, Wu Y, Qi H, Xiao J, Gong H, Zhang Y, et al. A new antagonist for CCR4 attenuates allergic lung inflammation in a mouse model of asthma. Sci Rep. 2017;7(1):15038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Watson C, Jenkinson S, Kazmierski W, Kenakin T. The CCR5 receptor-based mechanism of action of 873140, a potent allosteric noncompetitive HIV entry inhibitor. Mol Pharmacol. 2005;67(4):1268–82. [DOI] [PubMed] [Google Scholar]
  • 265.Nichols WG, Steel HM, Bonny T, Adkison K, Curtis L, Millard J, et al. Hepatotoxicity observed in clinical trials of aplaviroc (GW873140). Antimicrob Agents Chemother. 2008;52(3):858–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Baba M, Takashima K, Miyake H, Kanzaki N, Teshima K, Wang X, et al. TAK-652 inhibits CCR5-mediated human immunodeficiency virus type 1 infection in vitro and has favorable pharmacokinetics in humans. Antimicrob Agents Chemother. 2005;49(11):4584–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Este JA. TAK-779 (Takeda). Curr Opin Investig Drugs. 2001;2(3):354–6. [PubMed] [Google Scholar]
  • 268.Kummerle T, Lehmann C, Hartmann P, Wyen C, Fatkenheuer G. Vicriviroc: a CCR5 antagonist for treatment-experienced patients with HIV-1 infection. Expert Opin Investig Drugs. 2009;18(11):1773–85. [DOI] [PubMed] [Google Scholar]
  • 269.Saita Y, Kodama E, Orita M, Kondo M, Miyazaki T, Sudo K, et al. Structural basis for the interaction of CCR5 with a small molecule, functionally selective CCR5 agonist. J Immunol. 2006;177(5):3116–22. [DOI] [PubMed] [Google Scholar]
  • 270.Saita Y, Kondo M, Shimizu Y. Species selectivity of small-molecular antagonists for the CCR5 chemokine receptor. Int Immunopharmacol. 2007;7(12):1528–34. [DOI] [PubMed] [Google Scholar]
  • 271.Palani A, Shapiro S, Josien H, Bara T, Clader JW, Greenlee WJ, et al. Synthesis, SAR, and biological evaluation of oximino-piperidino-piperidine amides. 1. Orally bioavailable CCR5 receptor antagonists with potent anti-HIV activity. J Med Chem. 2002;45(14):3143–60. [DOI] [PubMed] [Google Scholar]
  • 272.van Kuijk AW, Vergunst CE, Gerlag DM, Bresnihan B, Gomez-Reino JJ, Rouzier R, et al. CCR5 blockade in rheumatoid arthritis: a randomised, double-blind, placebo-controlled clinical trial. Ann Rheum Dis. 2010;69(11):2013–6. [DOI] [PubMed] [Google Scholar]
  • 273.Ghadjar P, Rubie C, Aebersold DM, Keilholz U. The chemokine CCL20 and its receptor CCR6 in human malignancy with focus on colorectal cancer. Int J Cancer. 2009;125(4):741–5. [DOI] [PubMed] [Google Scholar]
  • 274.Frick VO, Rubie C, Kolsch K, Wagner M, Ghadjar P, Graeber S, et al. CCR6/CCL20 chemokine expression profile in distinct colorectal malignancies. Scand J Immunol. 2013;78(3):298–305. [DOI] [PubMed] [Google Scholar]
  • 275.Shimizu Y, Murata H, Kashii Y, Hirano K, Kunitani H, Higuchi K, et al. CC-chemokine receptor 6 and its ligand macrophage inflammatory protein 3alpha might be involved in the amplification of local necroinflammatory response in the liver. Hepatology. 2001;34(2):311–9. [DOI] [PubMed] [Google Scholar]
  • 276.Varona R, Cadenas V, Gomez L, Martinez AC, Marquez G. CCR6 regulates CD4+ T-cell-mediated acute graft-versus-host disease responses. Blood. 2005;106(1):18–26. [DOI] [PubMed] [Google Scholar]
  • 277.Lu MY, Lu SS, Chang SL, Liao F. The Phosphorylation of CCR6 on Distinct Ser/Thr Residues in the Carboxyl Terminus Differentially Regulates Biological Function. Front Immunol. 2018;9:415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Getschman AE, Imai Y, Larsen O, Peterson FC, Wu X, Rosenkilde MM, et al. Protein engineering of the chemokine CCL20 prevents psoriasiform dermatitis in an IL-23-dependent murine model. Proc Natl Acad Sci U S A. 2017;114(47):12460–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Campbell JJ, Bowman EP, Murphy K, Youngman KR, Siani MA, Thompson DA, et al. 6-C-kine (SLC), a lymphocyte adhesion-triggering chemokine expressed by high endothelium, is an agonist for the MIP-3beta receptor CCR7. J Cell Biol. 1998;141(4):1053–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Jaeger K, Bruenle S, Weinert T, Guba W, Muehle J, Miyazaki T, et al. Structural Basis for Allosteric Ligand Recognition in the Human CC Chemokine Receptor 7. Cell. 2019;178(5):1222–30 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Hull-Ryde EA, Porter MA, Fowler KA, Kireev D, Li K, Simpson CD, et al. Identification of Cosalane as an Inhibitor of Human and Murine CC-Chemokine Receptor 7 Signaling via a High-Throughput Screen. SLAS Discov. 2018;23(10):1083–91. [DOI] [PubMed] [Google Scholar]
  • 282.Blanco-Perez F, Kato Y, Gonzalez-Menendez I, Laino J, Ohbayashi M, Burggraf M, et al. CCR8 leads to eosinophil migration and regulates neutrophil migration in murine allergic enteritis. Sci Rep. 2019;9(1):9608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Barsheshet Y, Wildbaum G, Levy E, Vitenshtein A, Akinseye C, Griggs J, et al. CCR8(+)FOXp3(+) Treg cells as master drivers of immune regulation. Proc Natl Acad Sci U S A. 2017;114(23):6086–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Oshio T, Kawashima R, Kawamura YI, Hagiwara T, Mizutani N, Okada T, et al. Chemokine receptor CCR8 is required for lipopolysaccharide-triggered cytokine production in mouse peritoneal macrophages. PLoS One. 2014;9(4):e94445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Rummel PC, Arfelt KN, Baumann L, Jenkins TJ, Thiele S, Luttichau HR, et al. Molecular requirements for inhibition of the chemokine receptor CCR8--probe-dependent allosteric interactions. Br J Pharmacol. 2012;167(6):1206–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Cosorich I, McGuire HM, Warren J, Danta M, King C. CCR9 Expressing T Helper and T Follicular Helper Cells Exhibit Site-Specific Identities During Inflammatory Disease. Front Immunol. 2018;9:2899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Arseneau KO, Cominelli F. Vercirnon for the treatment of Crohn’s disease. Expert Opin Investig Drugs. 2013;22(7):907–13. [DOI] [PubMed] [Google Scholar]
  • 288.Feagan BG, Sandborn WJ, D’Haens G, Lee SD, Allez M, Fedorak RN, et al. Randomised clinical trial: vercirnon, an oral CCR9 antagonist, vs. placebo as induction therapy in active Crohn’s disease. Aliment Pharmacol Ther. 2015;42(10):1170–81. [DOI] [PubMed] [Google Scholar]
  • 289.Tubo NJ, Wurbel MA, Charvat TT, Schall TJ, Walters MJ, Campbell JJ. A systemically-administered small molecule antagonist of CCR9 acts as a tissue-selective inhibitor of lymphocyte trafficking. PLoS One. 2012;7(11):e50498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Walters MJ, Wang Y, Lai N, Baumgart T, Zhao BN, Dairaghi DJ, et al. Characterization of CCX282-B, an orally bioavailable antagonist of the CCR9 chemokine receptor, for treatment of inflammatory bowel disease. J Pharmacol Exp Ther. 2010;335(1):61–9. [DOI] [PubMed] [Google Scholar]
  • 291.Bekker P, Ebsworth K, Walters MJ, Berahovich RD, Ertl LS, Charvat TT, et al. CCR9 Antagonists in the Treatment of Ulcerative Colitis. Mediators Inflamm. 2015;2015:628340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Wu Q, Chen JX, Chen Y, Cai LL, Wang XZ, Guo WH, et al. The chemokine receptor CCR10 promotes inflammation-driven hepatocarcinogenesis via PI3K/Akt pathway activation. Cell Death Dis. 2018;9(2):232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Abeywardane A, Caviness G, Choi Y, Cogan D, Gao A, Goldberg D, et al. N-Arylsulfonyl-alpha-amino carboxamides are potent and selective inhibitors of the chemokine receptor CCR10 that show efficacy in the murine DNFB model of contact hypersensitivity. Bioorg Med Chem Lett. 2016;26(21):5277–83. [DOI] [PubMed] [Google Scholar]
  • 294.Lee M, Lee Y, Song J, Lee J, Chang SY. Tissue-specific Role of CX3CR1 Expressing Immune Cells and Their Relationships with Human Disease. Immune Netw. 2018;18(1):e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Shen F, Zhang Y, Jernigan DL, Feng X, Yan J, Garcia FU, et al. Novel Small-Molecule CX3CR1 Antagonist Impairs Metastatic Seeding and Colonization of Breast Cancer Cells. Mol Cancer Res. 2016;14(6):518–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Stout MC, Narayan S, Pillet ES, Salvino JM, Campbell PM. Inhibition of CX3CR1 reduces cell motility and viability in pancreatic adenocarcinoma epithelial cells. Biochem Biophys Res Commun. 2018;495(3):2264–9. [DOI] [PubMed] [Google Scholar]
  • 297.Abdelmoaty S, Arthur H, Spyridopoulos I, Wagberg M, Danielson RF, Pernow J, et al. 5234KAND567, the first selective small molecule CX3CR1 antagonist in clinical development, medates anti-inflammatory cardioprotective effects in rodent models of atherosclerosis and myocardial infarction. European Heart Journal. 2019;40. [Google Scholar]
  • 298.Ohta T, Sugiyama M, Hemmi H, Yamazaki C, Okura S, Sasaki I, et al. Crucial roles of XCR1-expressing dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune homeostasis. Sci Rep. 2016;6:23505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Bonecchi R, Graham GJ. Atypical Chemokine Receptors and Their Roles in the Resolution of the Inflammatory Response. Front Immunol. 2016;7:224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Savino B, Borroni EM, Torres NM, Proost P, Struyf S, Mortier A, et al. Recognition versus adaptive up-regulation and degradation of CC chemokines by the chemokine decoy receptor D6 are determined by their N-terminal sequence. J Biol Chem. 2009;284(38):26207–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Borroni EM, Cancellieri C, Vacchini A, Benureau Y, Lagane B, Bachelerie F, et al. beta-arrestin-dependent activation of the cofilin pathway is required for the scavenging activity of the atypical chemokine receptor D6. Sci Signal. 2013;6(273):ra30 1–11, S1-3. [DOI] [PubMed] [Google Scholar]
  • 302.Gravel S, Malouf C, Boulais PE, Berchiche YA, Oishi S, Fujii N, et al. The peptidomimetic CXCR4 antagonist TC14012 recruits beta-arrestin to CXCR7: roles of receptor domains. J Biol Chem. 2010;285(49):37939–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Ehrlich A, Ray P, Luker KE, Lolis EJ, Luker GD. Allosteric peptide regulators of chemokine receptors CXCR4 and CXCR7. Biochem Pharmacol. 2013;86(9):1263–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Oishi S, Kuroyanagi T, Kubo T, Montpas N, Yoshikawa Y, Misu R, et al. Development of novel CXC chemokine receptor 7 (CXCR7) ligands: selectivity switch from CXCR4 antagonists with a cyclic pentapeptide scaffold. J Med Chem. 2015;58(13):5218–25. [DOI] [PubMed] [Google Scholar]
  • 305.Puddinu V, Casella S, Radice E, Thelen S, Dirnhofer S, Bertoni F, et al. ACKR3 expression on diffuse large B cell lymphoma is required for tumor spreading and tissue infiltration. Oncotarget. 2017;8(49):85068–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Ameti R, Melgrati S, Radice E, Cameroni E, Hub E, Thelen S, et al. Characterization of a chimeric chemokine as a specific ligand for ACKR3. J Leukoc Biol. 2018;104(2):391–400. [DOI] [PubMed] [Google Scholar]
  • 307.Kalatskaya I, Berchiche YA, Gravel S, Limberg BJ, Rosenbaum JS, Heveker N. AMD3100 is a CXCR7 ligand with allosteric agonist properties. Mol Pharmacol. 2009;75(5):1240–7. [DOI] [PubMed] [Google Scholar]
  • 308.Wijtmans M, Maussang D, Sirci F, Scholten DJ, Canals M, Mujic-Delic A, et al. Synthesis, modeling and functional activity of substituted styrene-amides as small-molecule CXCR7 agonists. Eur J Med Chem. 2012;51:184–92. [DOI] [PubMed] [Google Scholar]
  • 309.Zabel BA, Wang Y, Lewen S, Berahovich RD, Penfold ME, Zhang P, et al. Elucidation of CXCR7-mediated signaling events and inhibition of CXCR4-mediated tumor cell transendothelial migration by CXCR7 ligands. J Immunol. 2009;183(5):3204–11. [DOI] [PubMed] [Google Scholar]
  • 310.Zabel BA, Lewen S, Berahovich RD, Jaen JC, Schall TJ. The novel chemokine receptor CXCR7 regulates trans-endothelial migration of cancer cells. Mol Cancer. 2011;10:73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Yamada K, Maishi N, Akiyama K, Towfik Alam M, Ohga N, Kawamoto T, et al. CXCL12-CXCR7 axis is important for tumor endothelial cell angiogenic property. Int J Cancer. 2015;137(12):2825–36. [DOI] [PubMed] [Google Scholar]
  • 312.D’Huys T, Claes S, Van Loy T, Schols D. CXCR7/ACKR3-targeting ligands interfere with X7 HIV-1 and HIV-2 entry and replication in human host cells. Heliyon. 2018;4(3):e00557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Matti C, D’Uonnolo G, Artinger M, Melgrati S, Salnikov A, Thelen S, et al. CCL20 is a novel ligand for the scavenging atypical chemokine receptor 4. J Leukoc Biol. 2020;107(6):1137–54. [DOI] [PubMed] [Google Scholar]

RESOURCES