Download PDF
Review  |  Open Access  |  22 May 2024

G protein-coupled receptors: a gateway to targeting oncogenic EVs?

Views: 436 |  Downloads: 45 |  Cited:  0
Extracell Vesicles Circ Nucleic Acids 2024;5:233-48.
10.20517/evcna.2024.10 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

Dysregulated intercellular communication is a key feature driving cancer progression. Recently, extracellular vesicles (EVs) have added a new channel to this dense communication network. Despite solid evidence that EVs are central mediators of dysregulated signaling in onco-pathological settings, this has yet to be translated into clinically actionable strategies. The heterogeneity of EV cargo molecules, plasticity of biogenesis routes, and large overlap with their role in physiological communication, complicate a potential targeting strategy. However, recent work has linked EV biology to perhaps the "most druggable" proteins - G protein-coupled receptors (GPCRs). GPCR targeting accounts for ~60% of drugs in development and more than a third of all currently approved drugs, spanning almost all areas of medicine. Although several GPCRs have been linked to cancer initiation and progression, relatively few agents have made it into oncological regimes, suggesting that their potential is underexploited. Herein, we examine the molecular mechanisms linking GPCRs to EV communication in cancer settings. We propose that GPCRs hold potential in the search for EV-targeting in oncology.

Keywords

G protein-coupled receptors (GPCRs), extracellular vesicles (EVs), oncogenic signaling, therapeutic targeting

INTRODUCTION

The notion that extracellular vesicles (EVs) drive tumor progression is backed up by ample evidence[1,2]. EVs have been implicated in several aspects of cancer, including tumor survival, angiogenesis, pre-metastatic niche formation, and drug resistance. EVs play an important role in intercellular communication through the transfer of the information contained in the vesicles to local or distant sites, maintaining the organism’s homeostasis. Tumors can hijack this network of communication, co-opting the EV machinery toward cargo selection and secretion of molecules that promote the establishment of a tumor-permissive local microenvironment. The EV-mediated crosstalk between cancer and stromal cells can, for example, suppress an antitumoral immune reaction[3], establish a pre-metastatic niche[4] or provide a means to expel targeting-agents[5]-mediating drug resistance.

Intercepting the EVs secreted from cancer cells has therefore often been proposed as a therapeutic strategy. This would be beneficial because EVs facilitate many cancer hallmarks[3-5]. However, developing a clinically feasible EV-targeting strategy has proven challenging. One might speculate that an approach to reduce the detrimental effects of cancer-released EVs could be to inhibit their biogenesis, secretion, or uptake by the recipient cells. Several studies have already shown that modulating certain proteins or lipids (e.g., Rab27a, nSMase2, farnesyl transferase, Ras, ceramide) involved in the biogenesis and secretion mechanisms of EVs effectively reduced the level of tumor EVs in the extracellular environment, thereby increasing sensitivity to chemotherapy and reducing tumor progression[6-13]. Unfortunately, none of these strategies have progressed beyond in vivo modeling.

Currently, the major hurdle is the development of drugs that selectively target tumor-derived EV biogenesis or secretion while sparing EV-mediated communication in physiological cell/tissue function[14]. It is not yet clear whether there are unique drivers of EV biogenesis in cancer cells or what these tumor-specific stimulants could be. With two remaining hurdles in mind - cancer specificity and druggability - attention is warranted to the emergence of studies linking EV biology to the lucrative family of G protein-coupled receptors (GPCRs). This large family of membrane receptors is involved in the regulation of a plethora of physiological and pathological processes[15-22]. In 2017, it was estimated that approximately 25-36% of all FDA- and EMA-approved drugs target a GPCR[23,24]. These receptors have an inherent druggability due to, for example, their favorable subcellular location, their dynamic structures, and their ability to activate multiple signaling pathways[25]. In addition, GPCRs have been identified as modulators of EV biogenesis, secretion, and uptake by recipient cells[26]. Concordantly, over the last few years, numerous studies have emerged regarding the GPCR contribution to EV-mediated cancer progression, which might be the targetable angle the EV community has been looking for. In this review, we provide an overview of how GPCR signaling and EV trafficking cooperate to support tumor survival and progression, and propose the potential for targeting GPCRs to modulate cancer-associated EVs as a therapeutic strategy.

G PROTEIN-COUPLED RECEPTORS AND THEIR ONCOMODULATORY PROPERTIES

GPCRs are the largest family of membrane-bound receptors (~ 800 members in humans)[22]. They are characterized by their seven transmembrane-spanning domains separated by alternating intracellular and extracellular loops. In humans, the GPCR family consists of four main classes based on their structural similarities: rhodopsin receptor-like (Class A), secretin and adhesion receptor-like (Class B), metabotropic glutamate receptor-like (Class C), and frizzled receptor-like (Class F)[27]. Depending on the type of GPCR, receptor activation is initiated upon ligand binding (including neurotransmitters, hormones, lipids, photons, and chemokines) to either an extracellular domain or a transmembrane domain, or a combination of the two [Figure 1].

G protein-coupled receptors: a gateway to targeting oncogenic EVs?

Figure 1. Schematic overview of GPCR signaling. (1) GPCR in its inactive state, where the heterotrimeric G protein complex, consisting of a Gα-subunit and a Gβγ-subunit, is still associated and GDP bound; (2) Ligand binding induces the GPCR to undergo a conformational change that initiates G protein coupling. The heterotrimeric G protein complex is activated by GDP exchange for GTP. For a constitutively active GPCR, the G protein constantly couples to the receptor; (3) Subsequently, the subunits dissociate and interact with their corresponding effector proteins, regulating downstream signaling; (4) GPCR signaling is downregulated by G protein-coupled receptor kinase (GRK)-mediated phosphorylation of the C-terminus. Subsequent arrestin binding initiates receptor endocytosis, which can lead to receptor degradation or recycling back to the plasma membrane, as well as endosomal signaling propagation. GPCR: G protein-coupled receptor; AC: adenylyl cyclase; PLCβ: phospholipase C-beta; GRK: G protein-coupled receptor kinase; Arr: arrestin; GDP: guanosine diphosphate; GTP: guanosine triphosphate.

In the classical model of GPCR functionality, ligand binding results in a conformational change of the transmembrane domains, causing it to couple to its heterotrimeric G protein, initiating Gα dissociation from its Gβγ subunits[27]. The heterotrimeric G proteins serve as transducers and scaffolding proteins that coordinate downstream signaling. In humans, 16 Gα, 5 Gβ, and 13 Gγ subunits have been identified, leading to a multitude of heterotrimeric combinations[27]. The Gα subunit may transduce signals independently of the other subunits, while the Gβγ subunits only signal as a single unit. Gα can be grouped into four major families: αs, αi, αq/11, and α12/13. Each of these interacts with different effector proteins, producing second messenger molecules that specifically activate different downstream signaling pathways. The Gαs and Gαi proteins can modulate the effector adenyl cyclase (AC) and subsequently the cAMP levels by stimulating (s) and inhibiting (i) AC, respectively[28,29]. The Gαq/11 proteins are involved in Ca2+ signaling through phospholipase C activation[30]. Lastly, Gα12/13 proteins are involved in activating small GTP-binding proteins[31]. Besides the canonical ligand-activated signaling, some GPCRs display constitutive activity, which is frequently associated with disease development, including cancer[32]. In these settings, overexpression of GPCRs is often apparent, which is most often associated with increased basal signaling.

To control GPCR-mediated signaling, negative feedback loops are in place to maintain biological homeostasis. Receptor desensitization takes place upon receptor phosphorylation by G protein-coupled receptor kinases, which allows for β-arrestin recruitment and subsequent internalization of the GPCR, downregulating signaling from the plasma membrane[22]. Once the GPCR is in the endosomal system, it can either be recycled back to the plasma membrane or targeted for lysosomal degradation. However, recently, it has become apparent that some GPCRs can also signal from their endosomal compartments upon internalization[33]. This changes the spatial-temporal profile of G protein signaling, generating a “second wave” of signaling [Figure 1.4]. This “second wave” of signaling can diverge from the conventionally described GPCR signaling mechanisms, potentially controlling unique physiological as well as pathological downstream effectors. However, such non-canonical signaling is outside the scope of the present review and has been discussed in detail elsewhere[34].

Aside from regulating many physiological processes, dysregulated GPCR expression and/or signaling has been linked to several hallmarks of cancer (reviewed in[35]), including proliferation and survival[36], invasion and metastasis[37], angiogenesis[38], and immune cell evasion[39]. For instance, in breast cancer, the chemokine receptor CXCR4 is overexpressed and its natural ligand, CXCL12, was shown to be highly secreted near the organs that are the metastatic destination of the tumor cells, indicative of a key role in metastatic colonization[37]. Besides the CXCR4/CXCL12 axis, different chemokine receptors have also been shown to play a role in cancer[37,40-43]. Other examples of GPCRs involved in cancer progression are the sphingosine 1 phosphate receptors (S1PRs). In glioblastoma, sphingosine-1-phosphate (S1P), the natural ligand for the S1P receptors, stimulates tumor growth and invasiveness[36]. S1P stimulates cell proliferation of glioma cells by activating the S1PR1 and S1PR3 receptors that promote ERK signaling. Notably, the S1PR2 receptor appears to have the opposite effect, downregulating ERK activation, suggesting that the S1P-controlled GPCRs may confer contextual mitogenic regulation. Moreover, it was recently demonstrated that other GPCRs affect S1P signaling in glioma development[39]. The human cytomegalovirus (HCMV) encoded viral GPCR US28, linked to oncomodulation in several cancers, colocalizes with the S1P1 receptor (S1PR1) and recruits the sphingosine kinase 1 (SK1)[39,44]. The consequent SK1/S1PR1 signaling stimulates glioma proliferation and survival via AKT, cMYC, and STAT3 pathways. In addition to class A GPCRs, other receptor classes have been linked to cancer progression, including the adhesion[45,46] and metabotropic family[47,48]. In comparison to other clinical contexts such as heart disease, brain disorders, and allergies, the use of GPCR targeting agents lags in oncology, where efforts to develop therapeutics have traditionally focused on the smaller family of receptor tyrosine kinases. In the ever-present need for better treatment options for cancer patients, targeting GPCRs may provide unique novel avenues for therapeutic intervention.

GPCR AND EV INTERPLAY CONTRIBUTES TO ONCOMODULATION

In the last decade, it has become apparent that GPCRs also play roles in several aspects of EV biology, including biogenesis[49,50], sorting of cargo[51-53], secretion[54], and uptake by recipient cells[55,56]. In recent years, there has been an increase in studies that link the two in cancer contexts. In this review, we focus on the interplay between GPCRs and EVs in cancer, but that does not exclude that the EV-GPCR cooperation may be relevant for other pathologies as well; for a detailed convergence of EV and GPCR biology, readers are referred to our review[26]. GPCRs expressed in a cancerous donor cell may modulate EV secretion or alter EV cargo selection, promoting tumorigenesis when released into the extracellular environment. Alternatively, tumor-derived EVs may deliver molecular components that modulate GPCR signaling when taken up by recipient cells. EVs can also act as carriers for the receptor, horizontally transferring the GPCR from one cell to another, widening the receptor’s potential for modulating oncogenic signaling.

GPCRs modulate oncogenic EV biogenesis and secretion

Most, if not all, mammalian cells actively release EVs, a family of heterogeneous membrane-enclosed vesicles containing bioactive cargo. Generally, EVs can roughly be categorized into small EVs, when the particles have a diameter < 200 nm, and large EVs, when the particles have a diameter > 200 nm. When the mechanism of EV biogenesis is known, one can also make a distinction between exosomes and microvesicles, two subpopulations of small EVs[57]. Microvesicles originate via outward budding of the plasma membrane [Figure 2.I.a], while exosomes evolve from inward budding of the endosomal membrane, generating a multivesicular body containing intraluminal vesicles [Figure 2.I.b]. These vesicles will be recognized as exosomes upon release into the extracellular environment. Due to the overlap in characteristics between EV populations, it is technically challenging to distinguish them unless there is strong evidence on the exact route of biogenesis. Therefore, throughout this review, the general term EV has been chosen in the absence of robust experimental differentiation.

G protein-coupled receptors: a gateway to targeting oncogenic EVs?

Figure 2. Interplay between GPCRs and EVs during cancer development, highlighting their therapeutic potential. (I) Mutated or oncogenic GPCRs mediate abnormal signaling pathways that can affect protein sorting, leading to a change in EV cargo, or modulate tumor EV biogenesis and secretion, or a combination of the two. (II) EV-mediated functional horizontal transfer of GPCRs can propagate oncogenic signaling in the recipient cells. (III) Uptake of oncogenic EVs, either via endocytosis (c) or perhaps direct membrane fusion (d), can affect endogenously expressed GPCR signaling, contributing to cancer development. Additionally, GPCRs can modulate EV uptake, for example, via docking to the GPCR via its ligand (e). Thus, GPCRs pose an interesting point of interception to target tumor EV-mediated cancer development and progression. a: microvesicle secretion; b: exosome secretion; c: EV endocytosis; d: EV-plasma membrane fusion; e: GPCR-mediated EV docking. GPCR: G protein-coupled receptor; MVB: multivesicular body; EV: extracellular vesicle; EE: early endosome.

GPCR-mediated signaling has been linked to numerous aspects of EV biology. In fact, several studies describe GPCR-regulated tumor EV secretion [Table 1]. For example, the metabotropic glutamate receptor 3 (mGlu3) has been identified as a driver of EV secretion in breast cancer[58]. The activation of mGlu3 in MDA-MB-231 cells resulted in a Rab27a-dependent increased secretion of EVs, while silencing of Rab27a or inhibition of receptor signaling reduced EV release. Furthermore, these tumor EVs appear to carry mitochondrial DNA derived from the MDA-MB-231 cells, which promotes a pro-invasive behavior in recipient cells[58]. Similar results were obtained for the bombesin receptor subtype-3 (BRS-3), a GPCR highly expressed in various tumors, such as breast cancer, lung cancer, and prostate cancer[59,60]. Activation of BRS-3 in stably expressing HEK293 cells leads to a 50% increase in secretion of EVs compared to the unstimulated condition, while antagonist treatment reversed these effects, hinting at BRS-3 dependency[59]. Interestingly, BRS-3 itself was shown to be sorted into these EVs and horizontally transferred to recipient cells. Stimulation of the GPCR in the recipient cells resulted in elevated ERK phosphorylation, suggesting that BRS-3 retained its function upon transfer. These observations suggest that GPCRs can be responsible for the secretion of tumor-derived EVs as well as for their cargo composition including the horizontal transfer of the GPCRs themselves.

Table 1

Overview of GPCRs involved in EV-mediated cancer progression

GPCR locationGPCR involvedCancer typeOncogenic effectRef.
Class A
On EV-secreting cell
PAR2 (protease-activated receptor)Breast cancerPAR2 activation stimulates EV shedding from the plasma membrane. This process is regulated by MAPK, p38, and Rho signaling. These EVs also incorporate components that drive the invasion and migration of recipient cells[76]
H1R1 (histamine receptor)Cervical cancerActivation of H1R1 induces SNAP23-dependent fusion of the multivesicular body with the plasma membrane, resulting in increased EV secretion[77]
Incorporated into EVsCXCR4 (chemokine receptor)Murine hepatocarcinomaHorizontal transfer of CXCR4 via EVs from cells with high metastatic potential increases the motility of cells with a low metastatic potential[62]
CXCR4Acute myeloid leukemia (AML)AML cells secrete CXCR4-positive EVs that can be horizontally transferred to leukemia cells lacking CXCR4, increasing metastatic spread, growth, and bone marrow infiltration via the CXCL12-CXCR4 axis[63]
CXCR4GlioblastomaGlioblastoma-derived EVs carry CXCR4 monomers and dimers, as well as VEGF. Treatment of endothelial cells with these EVs promotes cell proliferation, motility and tube formation compared to control EVs[78]
CXCR4Breast cancerA liver-kidney-on-a-chip model shows breast cancer-derived EVs organ tropism. MCF7 and MDA-MB-231 cells secrete CXCR4+ EVs that drive liver-kidney tropism via an CXCL12 dependent gradient[79]
US28 (viral chemokine receptor)GlioblastomaUS28-positive EVs retain binding to chemokines in the extracellular environment. Further, the US28-EVs attenuate CX3CL1-CX3CR1 signaling in EV-treated HEK293T cells[66]
S1P2 (sphingolipid receptor)Breast cancerBreast cancer cell-derived EVs horizontally transfer an N-terminally processed form of S1P2 to fibroblast, where it constitutively activates ERK signaling[64]
CCR6, CX3CR1 (chemokine receptors)Lung, pancreatic and colorectal cancerTumor EVs shed by various cancer cells carry tumor cell markers CCR6 and CX3CR1. CCR6 was transferred to monocytes, promoting chemotaxis[80]
CCR1, CCR6, CXCR4 (chemokine receptors)Gastric cancerThe expression of chemokine receptors on EVs from gastric cancer patients seems to vary depending on the cancer stage (i.e., CCR6 expression goes up while CXCR4 expression goes down)[81]
On EV-receiving cell
CXCR2, CXCR4 (chemokine receptors)MelanomaEV-mediated skewing of neutrophils toward a tumor-promoting phenotype, yielding tumor cell survival and migration[70]
ACKR3, CXCR4 (chemokine receptors)MelanomaIncreased migration of non-osteotropic melanoma cells due to EV-mediated upregulated expression of ACKR3[71]
CXCR4Prostate cancerEVs mediate the transfer of PKM2 to bone marrow cells, inducing CXCL12 secretion and subsequent CXCR4-dependent migration of the cancerous cells to the bone marrow[72]
CXCR4Breast cancerPlatelets-derived microvesicles increase the expression of CXCR4 in recipient breast cancer cells, partially enhancing their chemo invasiveness[82]
ACKR3Colorectal cancerACKR3-overexpressing tumor cells secrete EVs containing miR146a-5p and miR155-5p. These microRNAs are endocytosed by cancer-associated fibroblasts which get activated via JAK2-STAT3/NF-B-dependent signaling. Subsequently, these fibroblasts secrete inflammatory cytokines, including CXCL12. CXCL12 activates ACKR3 on the tumor cells, inducing EMT and pre-metastatic niche formation[83]
CCR8 (chemokine receptor)Glioblastoma, malignant melanoma and lung carcinomaCCR8 facilitates the uptake of EVs carrying TMZ resistance[55]
PAR (protease-activated receptor)Triple-negative breast cancerEVs rich in MMP1 from TNBC cells with a high metastatic potential promote metastasis of low metastatic cells via PAR1-mediated EMT[73]
S1PR1, S1PR2 (sphingolipid receptor)Ovarian cancerTransfer of tumor-derived EVs rich in SPHK1 increases the S1P production, leading to S1PR1/2-dependent PD-L1 expression[74]
A2B (adenosine receptor)Head and neck squamous cell carcinoma Tumor EVs, rich in adenosine, promote the secretion of angiogenic factors via adenosine A2B signaling[84]
CCR2 (chemokine receptor)Breast cancerTumor EVs, decorated with CCL2, migrate and accumulate near cells expressing CCR2, driving metastatic spread[56]
A2A (adenosine receptor)Bladder, colorectal, prostate and breast cancerTumor EVs carrying CD39 and CD73 covert ATP into adenosine, which activates adenosine A2A signaling in immune cells, regulating an immune response[85]
EP2, EP4 (prostanoid receptors)Prostate cancerTumor-derived EVs, rich in PGE2, induce the expression of CD73 on dendritic cells. CD73 increases the extracellular levels of adenosine, which inhibits T cell functioning[86]
CX3CR1 (chemokine receptor)Prostate cancerTumor EV-treated fibroblast release EVs carrying membrane-bound CX3CL1 that promotes migration and invasion of cancer cells via the CX3CL1-CX3CR1 signaling axis[87]
LPAR1, LPAR3 (lysophosphatidic acid receptors)Not specified ATX bound EVs can sequester LPA and activated LPAR1/3 signaling. LPARs are often involved in tumor stroma interactions and metastasis[88,89]
PAR1Breast and pancreatic cancerTumor EVs carry activated factor X on the EVs through which they can activate PAR1 receptors, promoting metastasis and pre-metastatic niche formation[90]
Class B
On EV-secreting cell
CD97/ADGRE5 (adhesion receptor)Gastric cancerCD97 expression ensures incorporation of EV cargo that promotes tumor cell proliferation and invasion via MAPK signaling or increases metastatic effects[91,92]
Incorporated into EVsELTD1/ADGRL4 (adhesion receptor)Breast cancerAdhesion receptor ELTD1/ADGRL4, and its highly glycosylated form, are incorporated into EVs and horizontally transferred to endothelial cells, promoting endothelial sprouting[65]
Class C
On EV-secreting cell
mGLU3 (metabotropic glutamate receptor)Breast cancerReceptor activation results in a Rab27-dependent increase in tumor EV secretion, as well as incorporation of specific cargo that increases invasive behavior of the recipient cells[58]
GRM1 (metabotropic glutamate receptor)MelanomaMetabotropic glutamate receptor 1 GRM1 activation promotes the incorporation of cargo that modulates cell migration, invasion, and growth of GRM1-negative recipient cells[51]
Incorporated into EVsGPRC5C (metabotropic glutamate receptor)Pancreatic cancerGPRC5C is incorporated into EVs of cancer patients but not of healthy subjects, potentially serving as a biomarker. Note: in breast cancer, knockdown of GPRC5C promotes cell proliferation[93,94]
Class F
Incorporated into EVsFZD-10 (frizzled receptor)Colorectal and gastric cancer, hepatocarcinoma, cholangiocarcinoma FZD-10 protein incorporation into EVs is upregulated in cancer patients compared to healthy subjects. EVs carrying FZD-10 as cargo increase tumor cell proliferation and metastasis[95,96]
On EV-receiving cell
FZD (frizzled receptor)Breast cancerTyrosine kinases ROR1 and ROR2 are being transferred via EVs to ROR1/2-negative cancer cells, acting as co-receptors for the Wnt signaling pathway, driving cancer progression[97]
FZD-6 (frizzled receptor)Breast cancerCancer-associated fibroblast EVs get processed by breast cancer cells, promoting attachment of autocrine Wnt11. These EVs in turn promote the protrusive activity, motility and metastasis of the breast cancer cells[98]
FZD (frizzled receptor)Colorectal cancerCancer-associated fibroblast EVs rich in Wnt molecules dedifferentiate colorectal cancer cells via Wnt-FZD-dependent signaling, conferring chemotherapy resistance[99]
Orphan GPCRs
On EV-secreting cell
GPR143/OA1 (ocular albinism type 1 receptor)Melanoma, breast, colorectal cancerGPR143 regulates the ESCRT machinery, thereby determining the EV content and quantity. In mice, EVs secreted from GPR143-positive cells increased the migratory and invasive potentials[61]
On EV-secreting cell and incorporated into EVsBRS-3 (bombesin-like receptor)Lung, breast, and prostate cancerBRS-3 activation yields a 50% increase in EV secretion, which is Rho signaling-dependent. Further, BRS-3 was incorporated into the EVs and functionally transferred to recipient cells[59,60]

As briefly discussed above, GPCRs can also influence the composition of tumor EVs. Recently, the GPCR GPR143 was identified as a regulator of “endosomal sorting complex required for transport” (ESCRT)-dependent EV biogenesis[61]. Knockdown of GPR143 in MCF7 cells reduces the amount and alters the protein content of EVs. The downregulated proteins are involved in cell motility and invasion pathways. In addition, exposure of MCF7 cells to EVs derived from GPR143-overexpressing breast cancer cells (MCF-7) enhanced their migration and invasion compared to EVs from shRNA against GPR143 containing cells, while exposure of HUVEC cells to the former EVs promoted their vascular branching[61]. Interestingly, the ability of GPR143-dependent EVs to modulate cell motility was linked to specific integrins. This finding indicates that GPR143-mediated signaling may be responsible for the selection of the EV cargo. Comparable results were obtained for the metabotropic glutamate receptor 1 (GRM1)[51]. Expression of this GPCR in C81-61 melanoma cells does not increase the secretion of EVs; however, it does change the composition of these EVs[51]. C81-61 GRM-cells treated with EVs derived from C81-61 cells expressing GRM1 displayed an increase in migration and invasion, suggesting a role in cargo selection.

Taken together, these studies underscore that GPCRs form a potential therapeutic target when located on the EV-secreting cells themselves. Modulation of these receptors and/or their downstream signaling pathways may downregulate EV secretion or affect oncogenic cargo selection, attenuating cancer progression [Figure 2.I].

Tumor EVs incorporate and transfer functional GPCRs to recipient cells

Tumor cell migration and invasion - In addition to regulating EV secretion and composition, GPCRs can be included as molecular cargo within EVs [Table 1, Figure 2.II]. The metastatic potential of a tumor cell can be altered upon importing particular receptors, as they may affect whether and how the cell can then respond to extracellular stimuli. For instance, exposure to EVs isolated from mouse hepatocarcinoma cells with a high metastatic potential (Hca-F cells) increased the migratory and invasive capacity of hepatocarcinoma cells with low metastatic behavior (Hca-P cells)[62]. Transfer of chemokine receptor CXCR4 via EVs seemed particularly important in the EV-mediated transition to a more metastatic profile observed in Hca-P cells. These modified Hca-P cells significantly increased the expression of CXCR4. In line with this, CXCR4 knockdown in Hca-F cells yielded a lower level of CXCR4 in Hca-P cells and the same observation could be made upon inhibition of EV transfer, suggesting CXCR4 being trafficked between cells via EVs. Another study demonstrated that EVs are involved in acute myelogenous leukemia (AML)[63]. The EVs isolated from peripheral blood and bone marrow plasma were shown to have elevated protein levels of CXCR4 compared to samples of healthy individuals. Upon exposure of HL-60 cells to these EVs, CXCR4 levels significantly increased, as did the migratory potential toward CXCL12, suggesting EV-mediated transfer of a functional CXCR4 receptor.

Components undergo a contextual shift - Besides the direct transfer and uptake of functional GPCRs into recipient cells, some receptors undergo alterations during this process. This contextual switch could cause the GPCR to change conformation, specific G protein coupling, or downstream altered coupling to effector proteins. In breast cancer, the S1P2 receptor has been implicated in the stimulation of fibroblast cell proliferation via an EV-dependent manner[64]. MDA-MB-231 cell-derived EVs contain S1P2 receptors that are transferred to fibroblasts. Treatment of MEF fibroblasts with media S1P2-EV containing resulted in ERK1/2 activation and DNA synthesis. Interestingly, activating the endogenous S1P2 receptor with allosteric agonist CYM5520 on the recipient cells did not yield an increase in DNA synthesis according to the [3H]-thymidine incorporation assay. In addition, inhibition of S1P binding to S1P2 and subsequent receptor activation did not affect ERK1/2 activation in the EV-treated MEF cells, suggesting that the receptor obtained constitutive activity. These findings were validated by S1P2 knockdown in the donor cells, whose EVs failed to activate ERK1/2 in the fibroblasts. The transferred S1P2 receptor appeared to be structurally altered; upon uptake, it gets processed to a shorter form due to cleavage of the N-terminal domain, which results in a GPCR conformation that constitutively enables ERK1/2 activation. The consequences of proteolytic activation of this receptor in cancer progression have yet to be determined. However, it is tempting to speculate that increased fibroblast proliferation might result in myofibroblast transitioning, stromal cells known to be involved in cancer progression. Similarly, the adhesion GPCR, ADGLR4, formerly referred to as ELTD1, was recently identified in EVs[65]. This receptor is involved in vasculature formation and resistance to anti-angiogenic therapy. It has been demonstrated that ADGRL4/ELTD1 is taken up in EVs from HUVEC and HMEC cells mostly in its highly N-glycosylated form (ECD hyperglycosylated). Detachment of the hyperglycosylated ECD results in receptor activation, suggesting that the EVs may carry specifically the activated form of the GPCR. Upon exposure of healthy HUVEC cells with these ADGRL4/ELTD1-containing EVs, cell proliferation and endothelial sprouting significantly increased both in vitro and in vivo[65]. This suggests that ADGRL4/ELTD1 incorporation into EVs might increase the activity of the receptor and have a functional consequence on angiogenesis when transferred to endothelial cells.

Implications on viral pathogenesis - EVs have not only been implicated in the incorporation of endogenous GPCRs. Infecting pathogens like viruses can also encode GPCRs that can be expressed in tumorigenic cells, such as the HCMV-encoded US28 - a viral GPCR that has oncomodulatory properties in several cancers[44]. Recently, we discovered that this receptor is sorted into EVs secreted from U251 glioma cells[66]. Over the course of host-virus co-evolution, HCMV acquired the human chemokine C-X3-C motif receptor 1 (CX3CR1), which has since diverged into US28, displaying structural homology and allowing it to bind a broad spectrum of human chemokines[67]. Interestingly, US28-positive EVs retain their ability to bind these chemokines[66]. Context was provided by experiments showing that US28+ EVs derived from U251 cells could scavenge away chemokines from the human CX3CR1, reducing receptor activation in HEK293T cells. One could speculate that this could lead to a suppressed immune reaction by attenuating immune cell migration toward the site of infection. If, indeed, chemokine receptor-decorated EVs were shown to disrupt chemoattractant gradients sufficient to deflect immune cell patrol, this spurs curiosity about the spread of such a mechanism in other (patho)physiological contexts. One could imagine that in a cancer setting, for example, tumor cells (beyond those HMCV-infected) would benefit from a receptor-decorated EV “sponge” to control local chemoattractant gradients and hence an immune cell invitation list within the tumor microenvironment. EV-mediated horizontal transfer of US28 has yet to be investigated; however, such a process cannot be excluded as a potential way for viral spread or pre-metastatic niche formation. In fact, EV-mediated transfer of CXCR4[68] and CCR5[69] to increase viral spread has already been demonstrated in the human immunodeficiency virus.

Tumor-derived EVs mediate oncogenic GPCR signaling in recipient cells

GPCRs are not just interesting therapeutic targets as EV cargo molecules. EVs could also act as carriers of a host of ligands, co-receptors, or other molecules that go on to activate GPCRs, thereby eliciting oncogenic signal transduction in receiving cells. A considerable number of studies have indicated that tumor-specific EV cargo may influence GPCR signaling in recipient cells [Table 1, Figure 2.III].

Pre-metastatic niche formation - EVs have been shown to be important components in “tumor-education” of immune cells. A recent study demonstrated that MV3 melanoma-derived EVs can activate primary human neutrophils to adopt a pro-tumor/N2 phenotype, driving tumor progression [Figure 3A][70]. The tumor-released EVs induce neutrophil migration toward the tumor microenvironment through the chemokine receptor CXCR2/CXCL8 (IL8) axis, activating the PI3K-AKT pathway in the neutrophils. Activation of this signaling pathway resulted in an increased transcriptional level of CXCR4 in the EV-exposed neutrophils, which was abrogated upon treatment with the PI3K inhibitor LY294002. Furthermore, co-cultured EV-treated neutrophils yielded an increase in tumor cell viability compared to MV3 cells co-cultured with naïve neutrophils. Taken together, these data suggest that tumor-derived EVs may increase the transcriptional level of CXCR4 in neutrophils, skewing their phenotype to a pro-tumor polarization, allowing tumor cell survival.

G protein-coupled receptors: a gateway to targeting oncogenic EVs?

Figure 3. Illustrative examples of EV-GPCR oncomodulation. (A) MV3 melanoma cell-derived EVs activate primary neutrophils to adopt a pro-tumor/N2-phenotype by increasing the expression of chemokine receptor CXCR4. This allows the neutrophils to migrate toward the tumor cells in a CXCL12-dependent manner, promoting tumor cell survival; (B) Osteopathic melanoma LCP-derived EVs alter the osteotropism of other melanoma cells. EV exposure of SK-Mel28 and WM-266 cells induces plasma membrane ACKR3 expression, resulting in a CXCL12-dependent tumor cell migration toward the bone; (C) EVs derived from prostate cancer cells are rich in PKM2, a kinase that induces CXCL12 production and secretion in bone marrow stem cells. Increased CXCL12 section induces migration of cancer cells toward the bone marrow in a chemokine receptor CXCR4-dependent manner; (D) Breast cancer cells can increase the migration and invasion of other breast cancer cells via EV-mediated transfer of MMP1. MMP1 is a protease that can activate PAR1 receptors on the receiving breast cancer cells in an autocrine manner, resulting in increased migration. PKM2: pyruvate kinase muscle isozyme 2; MMP1: matrix metalloprotease 1; BMSCs: bone marrow stem cells.

Bone metastases rarely occur in patients suffering from malignant melanoma. However, when they do, it severely worsens prognosis. The effects of melanoma-derived EVs on the propensity of non-osteotropic cells to be attracted toward bone (osteotropism) have been investigated. EVs derived from osteotropic LCP melanoma cells appear to stimulate the osteotropic behavior of SK-Mel28 and WM-266, non-osteotropic tumor cells, toward bone in a CXCL12-stimulated manner through the atypical chemokine receptor 3 (ACKR3)/CXCR4 axis [Figure 3B][71]. Exposure to LCP-derived EVs upregulates the level of plasma membrane-localized ACKR3 in SK-Mel28 and WM-266 cells. In contrast, plasma membrane levels of CXCR4, also activated by CXCL12, are not affected after exposure to LCP-derived EVs. The tumor-derived EVs did not carry CXCR4 nor ACKR3 protein, suggesting the transfer of other molecules that induce ACKR3 expression. However, silencing of either of the GPCRs attenuates the migratory properties these non-osteotropic cells obtain after EV treatment. This proves that the gained osteotropism of SK-Mel28 and WM-266 cells is driven by the CXCL12/CXCR4/ACKR3 axis, which appears to be stimulated in the recipient cells through EV-mediated upregulation of plasma membrane ACKR3. Similar results were reported in the context of prostate cancer, where tumor EVs promote pre-metastatic niche formation, enhancing bone metastasis[72]. In this case, prostate cancer PC-3 and C4-2B cell-derived EVs are rich in pyruvate kinase muscle isozyme M2 (PKM2) and transfer this protein to bone marrow stem cells (BMSCs). In BMSCs, the increase in protein levels of PKM2 upregulates CXCL12 production in a HIF-1α-dependent manner[72]. This attracts the prostatic cancer cells through CXCR4 and enhances their growth in bone marrow [Figure 3C]. CXCL12 is a clinically targetable protein, but blocking this chemokine comes with various immune-related adverse events. Thus, targeting the GPCR involved could be a reasonable approach to counter the EV-mediated metastasis.

EVs in establishing a tumor microenvironment - Apart from tumor-stromal crosstalk, EV-mediated communication can also occur between the tumor cells themselves. In triple-negative breast cancer (TNBC), tumor EVs have been shown to enhance the metastatic potential of other TNBC cells[73]. MDA-MB-231-HM cells with high metastatic potential secrete EVs enriched in matrix metalloprotease 1 (MMP-1). MMP-1 activates PAR1, a protease-activated GPCR involved in migration and invasion, on the recipient MDA-MB-231 cells by cleaving the N-terminal domain. It was hypothesized that MMP1 is packed into the EVs of the donor cell and transferred to the receiving cells. These cells then release the protease into the extracellular environment, so it can activate PAR1 in an autocrine manner, promoting migration and invasion of TNBC [Figure 3D][73]. Another interesting study demonstrated that ovarian cancer-derived EVs are packed with SPHK1/SK1, which catalyzes the phosphorylation of S1P[74]. S1P was shown to elevate programmed cell death ligand 1 (PD-L1) expression on three different ovarian cell lines - OVCAR5, HeyA8, and OVCAR4, driving T cell exhaustion. Moreover, it was demonstrated that silencing S1PR1 and S1PR2, two GPCRs that are activated via S1P binding, yielded a significant reduction in PD-L1 expression in OVACR5 cells[74]. One might hypothesize that EVs transfer SPHK1/SK1 to cells in the tumor microenvironment, elevating their production of S1P. In turn, S1P could activate its receptors on the recipient cells, leading to the reported PD-L1 overexpression and subsequently immune evasion.

GPCRs as cellular anchors - Modulation of GPCR signaling pathways within the recipient cells is not the only mode of EV-mediated action. GPCRs can also be used as docking sites, connecting EVs with the recipient cells. In glioblastoma, it was shown that the chemokine receptor CCR8 acts as a docking site for tumor-derived EVs[55]. CCR8 binds to the soluble ligand CCL18, which in turn binds to glycans on the EVs; thereby, CCL18 acts as a bridging molecule between the cells and the EVs. A “chemokine cloud” model where glycans cause chemokine retention near the EVs by constantly engaging with them, trafficking the EVs toward cells expressing the chemokine receptor, was postulated as the mode of action[55,75]. This study shows CCR8’s involvement in the EV-mediated transfer of chemoresistance-determining factors between glioblastoma cells[55]. CCR8 appears to ensure EV uptake and cargo transfer, enhancing cell growth and protecting the cells against temozolomide, a chemotherapeutic. The latter was counteracted by blocking CCR8, re-sensitizing the recipient GBM8 cells to temozolomide in vitro and in vivo. These are promising findings that strengthen the notion of targeting GPCRs as a therapeutic approach to counteract tumor EV-mediated effects [Figure 2.III.e].

CLINICAL APPLICATIONS

The field of EV therapeutic applications is experiencing significant development. Pre-clinical data suggest that EVs hold promise in many aspects of biomedicine, ranging from diagnostic monitoring to active treatment. Traditionally, cancer diagnosis involves invasive biopsy procedures, whereas clinically informative circulating EVs can be obtained via liquid biopsies, such as urine or blood. By carrying GPCRs and/or their ligands, EVs could open new avenues as biomarkers for cancer diagnosis or treatment monitoring. For example, in the case where EVs obtained from blood and plasma samples of AML patients showed increased levels of protein CXCR4 and CXCL12 compared to EVs from healthy control samples[63]. Similarly, EVs derived from breast and prostate cancer cells are packed with MMP1 and PKM2, respectively, compared to EVs derived from their healthy counterparts[72,73]. EVs are also currently being explored as drug delivery systems due to their many biocompatible features. In comparison to conventional drug delivery systems, EVs might overcome the hurdle of cytotoxicity and immunogenicity due to their natural origins. One could, therefore, envisage that EVs loaded with drugs targeting oncogenic GPCR signaling hubs may elicit an antitumor response. Many hurdles still need to be overcome, including EV production and standardization at scale, or large clinical trials for biomarker incorporation into treatment regimes. In parallel, given the wide range of GPCR physiological functions, any potential EV-GPCR targeting strategy must be rigorously tested to determine whether a sufficient therapeutic window exists. Nevertheless, the field holds excitement for the promise of EVs in next-generation therapeutics, from which GPCR and EV researchers may benefit.

CONCLUDING REMARKS

Reports of marked upregulation of EV release from cancer cells catapulted research interest in the field. Evidence of the plethora of cancer-supporting roles continues to accumulate - positioning EV-mediated communication as the target of therapeutic interjection. Yet, being a diverse and malleable communication channel, the right choice of molecular target remains elusive. In parallel, pharmacological research in the last decades has heralded the GPCR family as their muse: not only becoming the largest family of approved drug targets, but also the system by which many new pharmacological strategies are probed and developed. And crucially, where many paradigm shifts in our fundamental understanding of cellular signaling have been discovered - with repercussions far beyond the receptors themselves. While estimates vary on both the number of GPCRs and the number of drugs that target these, what is clear is the unique therapeutic potential housed within this family of proteins. Numerous factors underlie this - the vast number of receptors within the family, their structural druggability, interaction with key cellular mediators, and their presence in the plasma membrane. To therapeutically intercept oncogenic EV-mediated communication, a rational and actionable molecular target that regulates key steps within bidirectional onco-EV channels is required. The studies collated herein demonstrate that, ultimately, there may be therapeutic benefits in cross-targeting EVs through GPCRs, as they represent nodes at crucial points of the EV life cycle in various oncological settings.

DECLARATIONS

Acknowledgments

Figures were created with BioRender.com.

Authors’ contribution

Conceptualization: Crudden C, Siderius M, Smit MJ

Collection and collation of literature: Di Niro L, Glynn T

Figure/Table preparation: Di Niro L, Linders AC, Glynn T

Writing-preparation of initial draft: Di Niro L, Glynn T, Crudden C

Writing-review and editing: Crudden C, Linders AC, Pegtel DM, Siderius M, Smit MJ

Financial support: Smit MJ

Availability of data and materials

Not applicable.

Financial support and sponsorship

L Di Niro and AC Linders are employed on The Netherlands Health Research Organisation (ZonMw) grant (ZonMw Open Competition; 09120012110079) to MJ Smit and DM Pegtel; The laboratory of MJ Smit is additionally supported by the European Union (H2020-MSCA Program, grant agreement 860229, ONCORNET2.0); C Crudden is supported by a ZonMw Veni Fellowship (ZonMw Veni; 09150162010212).

Conflict of interest

All authors declare that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

REFERENCES

1. Han L, Lam EW, Sun Y. Extracellular vesicles in the tumor microenvironment: old stories, but new tales. Mol Cancer 2019;18:59.

2. Marar C, Starich B, Wirtz D. Extracellular vesicles in immunomodulation and tumor progression. Nat Immunol 2021;22:560-70.

3. Sharma P, Diergaarde B, Ferrone S, Kirkwood JM, Whiteside TL. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Sci Rep 2020;10:92.

4. Morrissey SM, Zhang F, Ding C, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab 2021;33:2040-2058.e10.

5. Xavier CPR, Belisario DC, Rebelo R, et al. The role of extracellular vesicles in the transfer of drug resistance competences to cancer cells. Drug Resist Updat 2022;62:100833.

6. Datta A, Kim H, McGee L, et al. High-throughput screening identified selective inhibitors of exosome biogenesis and secretion: a drug repurposing strategy for advanced cancer. Sci Rep 2018;8:8161.

7. Datta A, Kim H, Lal M, et al. Manumycin A suppresses exosome biogenesis and secretion via targeted inhibition of Ras/Raf/ERK1/2 signaling and hnRNP H1 in castration-resistant prostate cancer cells. Cancer Lett 2017;408:73-81.

8. Fonseka P, Chitti SV, Sanwlani R, Mathivanan S. Sulfisoxazole does not inhibit the secretion of small extracellular vesicles. Nat Commun 2021;12:977.

9. Trajkovic K, Hsu C, Chiantia S, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008;319:1244-7.

10. Cao YL, Zhuang T, Xing BH, Li N, Li Q. Exosomal DNMT1 mediates cisplatin resistance in ovarian cancer. Cell Biochem Funct 2017;35:296-303.

11. Roseblade A, Luk F, Ung A, Bebawy M. Targeting microparticle biogenesis: a novel approach to the circumvention of cancer multidrug resistance. Curr Cancer Drug Targets 2015;15:205-14.

12. Kosgodage US, Trindade RP, Thompson PR, Inal JM, Lange S. Chloramidine/bisindolylmaleimide-I-mediated inhibition of exosome and microvesicle release and enhanced efficacy of cancer chemotherapy. Int J Mol Sci 2017;18:1007.

13. Li B, Antonyak MA, Zhang J, Cerione RA. RhoA triggers a specific signaling pathway that generates transforming microvesicles in cancer cells. Oncogene 2012;31:4740-9.

14. Chitti SV, Nedeva C, Manickam R, Fonseka P, Mathivanan S. Extracellular vesicles as drug targets and delivery vehicles for cancer therapy. Pharmaceutics 2022;14:2822.

15. Korbecki J, Grochans S, Gutowska I, Barczak K, Baranowska-Bosiacka I. CC chemokines in a tumor: a review of pro-cancer and anti-cancer properties of receptors CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 Ligands. Int J Mol Sci 2020;21:7619.

16. Moreno P, Mantey SA, Lee SH, Ramos-Álvarez I, Moody TW, Jensen RT. A possible new target in lung-cancer cells: The orphan receptor, bombesin receptor subtype-3. Peptides 2018;101:213-26.

17. Ma B, Zhu J, Su J, et al. The role of GPR110 in lung cancer progression. Ann Transl Med 2020;8:745.

18. Billington CK, Penn RB. Signaling and regulation of G protein-coupled receptors in airway smooth muscle. Respir Res 2003;4:2.

19. Cannavo A, Liccardo D, Koch WJ. Targeting cardiac β-adrenergic signaling via GRK2 inhibition for heart failure therapy. Front Physiol 2013;4:264.

20. Sun GC, Ho WY, Chen BR, et al. GPCR dimerization in brainstem nuclei contributes to the development of hypertension. Br J Pharmacol 2015;172:2507-18.

21. Nickols HH, Conn PJ. Development of allosteric modulators of GPCRs for treatment of CNS disorders. Neurobiol Dis 2014;61:55-71.

22. Rosenbaum DM, Rasmussen SG, Kobilka BK. The structure and function of G-protein-coupled receptors. Nature 2009;459:356-63.

23. Hauser AS, Attwood MM, Rask-Andersen M, Schiöth HB, Gloriam DE. Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov 2017;16:829-42.

24. Sriram K, Insel PA. G protein-coupled receptors as targets for approved drugs: how many targets and how many drugs? Mol Pharmacol 2018;93:251-8.

25. Congreve M, de Graaf C, Swain NA, Tate CG. Impact of GPCR structures on drug discovery. Cell 2020;181:81-91.

26. Bebelman MP, Crudden C, Pegtel DM, Smit MJ. The convergence of extracellular vesicle and GPCR biology. Trends Pharmacol Sci 2020;41:627-40.

27. Wootten D, Christopoulos A, Marti-Solano M, Babu MM, Sexton PM. Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat Rev Mol Cell Biol 2018;19:638-53.

28. Taussig R, Iñiguez-Lluhi JA, Gilman AG. Inhibition of adenylyl cyclase by Gi alpha. Science 1993;261:218-21.

29. Northup JK, Sternweis PC, Smigel MD, Schleifer LS, Ross EM, Gilman AG. Purification of the regulatory component of adenylate cyclase. Proc Natl Acad Sci U S A 1980;77:6516-20.

30. Smrcka AV, Hepler JR, Brown KO, Sternweis PC. Regulation of polyphosphoinositide-specific phospholipase C activity by purified Gq. Science 1991;251:804-7.

31. Kozasa T, Jiang X, Hart MJ, et al. p115 RhoGEF, a GTPase activating protein for Galpha12 and Galpha13. Science 1998;280:2109-11.

32. Seifert R, Wenzel-Seifert K. Constitutive activity of G-protein-coupled receptors: cause of disease and common property of wild-type receptors. Naunyn Schmiedebergs Arch Pharmacol 2002;366:381-416.

33. Thomsen ARB, Jensen DD, Hicks GA, Bunnett NW. Therapeutic targeting of endosomal G-protein-coupled receptors. Trends Pharmacol Sci 2018;39:879-91.

34. Crilly SE, Puthenveedu MA. Compartmentalized GPCR signaling from intracellular membranes. J Membr Biol 2021;254:259-71.

35. O'Hayre M, Degese MS, Gutkind JS. Novel insights into G protein and G protein-coupled receptor signaling in cancer. Curr Opin Cell Biol 2014;27:126-35.

36. Young N, Van Brocklyn JR. Roles of sphingosine-1-phosphate (S1P) receptors in malignant behavior of glioma cells. Differential effects of S1P2 on cell migration and invasiveness. Exp Cell Res 2007;313:1615-27.

37. Müller A, Homey B, Soto H, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature 2001;410:50-6.

38. Favara DM, Liebscher I, Jazayeri A, et al. Elevated expression of the adhesion GPCR ADGRL4/ELTD1 promotes endothelial sprouting angiogenesis without activating canonical GPCR signalling. Sci Rep 2021;11:8870.

39. Bergkamp ND, van Senten JR, Brink HJ, et al. A virally encoded GPCR drives glioblastoma through feed-forward activation of the SK1-S1P1 signaling axis. Sci Signal 2023;16:eade6737.

40. Gabellini C, Trisciuoglio D, Desideri M, et al. Functional activity of CXCL8 receptors, CXCR1 and CXCR2, on human malignant melanoma progression. Eur J Cancer 2009;45:2618-27.

41. Benelli R, Stigliani S, Minghelli S, Carlone S, Ferrari N. Impact of CXCL1 overexpression on growth and invasion of prostate cancer cell. Prostate 2013;73:941-51.

42. Amersi FF, Terando AM, Goto Y, et al. Activation of CCR9/CCL25 in cutaneous melanoma mediates preferential metastasis to the small intestine. Clin Cancer Res 2008;14:638-45.

43. Ploenes T, Scholtes B, Krohn A, et al. CC-chemokine ligand 18 induces epithelial to mesenchymal transition in lung cancer A549 cells and elevates the invasive potential. PLoS One 2013;8:e53068.

44. Heukers R, Fan TS, de Wit RH, et al. The constitutive activity of the virally encoded chemokine receptor US28 accelerates glioblastoma growth. Oncogene 2018;37:4110-21.

45. Vallon M, Essler M. Proteolytically processed soluble tumor endothelial marker (TEM) 5 mediates endothelial cell survival during angiogenesis by linking integrin alpha(v)beta3 to glycosaminoglycans. J Biol Chem 2006;281:34179-88.

46. Yasinska IM, Sakhnevych SS, Pavlova L, et al. The Tim-3-Galectin-9 pathway and its regulatory mechanisms in human breast cancer. Front Immunol 2019;10:1594.

47. Sexton RE, Hachem AH, Assi AA, Bukhsh MA, Gorski DH, Speyer CL. Metabotropic glutamate receptor-1 regulates inflammation in triple negative breast cancer. Sci Rep 2018;8:16008.

48. Xiao B, Chen D, Zhou Q, et al. Glutamate metabotropic receptor 4 (GRM4) inhibits cell proliferation, migration and invasion in breast cancer and is regulated by miR-328-3p and miR-370-3p. BMC Cancer 2019;19:891.

49. Kajimoto T, Okada T, Miya S, Zhang L, Nakamura S. Ongoing activation of sphingosine 1-phosphate receptors mediates maturation of exosomal multivesicular endosomes. Nat Commun 2013;4:2712.

50. Burgoyne T, Jolly R, Martin-Martin B, et al. Expression of OA1 limits the fusion of a subset of MVBs with lysosomes - a mechanism potentially involved in the initial biogenesis of melanosomes. J Cell Sci 2013;126:5143-52.

51. Isola AL, Eddy K, Zembrzuski K, Goydos JS, Chen S. Exosomes released by metabotropic glutamate receptor 1 (GRM1) expressing melanoma cells increase cell migration and invasiveness. Oncotarget 2018;9:1187-99.

52. Pironti G, Strachan RT, Abraham D, et al. Circulating exosomes induced by cardiac pressure overload contain functional angiotensin II type 1 receptors. Circulation 2015;131:2120-30.

53. Brown M, Johnson LA, Leone DA, et al. Lymphatic exosomes promote dendritic cell migration along guidance cues. J Cell Biol 2018;217:2205-21.

54. Nojima H, Konishi T, Freeman CM, et al. Chemokine receptors, CXCR1 and CXCR2, differentially regulate exosome release in hepatocytes. PLoS One 2016;11:e0161443.

55. Berenguer J, Lagerweij T, Zhao XW, et al. Glycosylated extracellular vesicles released by glioblastoma cells are decorated by CCL18 allowing for cellular uptake via chemokine receptor CCR8. J Extracell Vesicles 2018;7:1446660.

56. Lima LG, Ham S, Shin H, et al. Tumor microenvironmental cytokines bound to cancer exosomes determine uptake by cytokine receptor-expressing cells and biodistribution. Nat Commun 2021;12:3543.

57. Welsh JA, Goberdhan DCI, O'Driscoll L, et al. MISEV Consortium. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles 2024;13:e12404.

58. Rabas N, Palmer S, Mitchell L, et al. PINK1 drives production of mtDNA-containing extracellular vesicles to promote invasiveness. J Cell Biol 2021;220:e202006049.

59. Wang Z, Wu L, Wang H, Zhang Y, Xiao H. Agonist-induced extracellular vesicles contribute to the transfer of functional bombesin receptor-subtype 3 to recipient cells. Cell Mol Life Sci 2022;79:72.

60. Moreno P, Ramos-Álvarez I, Moody TW, Jensen RT. Bombesin related peptides/receptors and their promising therapeutic roles in cancer imaging, targeting and treatment. Expert Opin Ther Targets 2016;20:1055-73.

61. Lee YJ, Shin KJ, Jang HJ, et al. GPR143 controls ESCRT-dependent exosome biogenesis and promotes cancer metastasis. Dev Cell 2023;58:320-34.e8.

62. Li M, Lu Y, Xu Y, et al. Horizontal transfer of exosomal CXCR4 promotes murine hepatocarcinoma cell migration, invasion and lymphangiogenesis. Gene 2018;676:101-9.

63. Kalinkovich A, Tavor S, Avigdor A, et al. Functional CXCR4-expressing microparticles and SDF-1 correlate with circulating acute myelogenous leukemia cells. Cancer Res 2006;66:11013-20.

64. El Buri A, Adams DR, Smith D, et al. The sphingosine 1-phosphate receptor 2 is shed in exosomes from breast cancer cells and is N-terminally processed to a short constitutively active form that promotes extracellular signal regulated kinase activation and DNA synthesis in fibroblasts. Oncotarget 2018;9:29453-67.

65. Sheldon H, Zhang W, Bridges E, et al. ELTD1 is present in extracellular vesicles derived from endothelial cells as a cleaved extracellular domain which induces in vivo angiogenesis. J Extracell Bio 2022;1:e52.

66. Bebelman MP, Setiawan IM, Bergkamp ND, et al. Exosomal release of the virus-encoded chemokine receptor US28 contributes to chemokine scavenging. iScience 2023;26:107412.

67. Scarborough JA, Paul JR, Spencer JV. Evolution of the ability to modulate host chemokine networks via gene duplication in human cytomegalovirus (HCMV). Infect Genet Evol 2017;51:46-53.

68. Tsukamoto T. Transcriptional gene silencing limits CXCR4-associated depletion of bone marrow CD34+ cells in HIV-1 infection. AIDS 2018;32:1737-47.

69. Mack M, Kleinschmidt A, Brühl H, et al. Transfer of the chemokine receptor CCR5 between cells by membrane-derived microparticles: a mechanism for cellular human immunodeficiency virus 1 infection. Nat Med 2000;6:769-75.

70. Guimarães-Bastos D, Frony AC, Barja-Fidalgo C, Moraes JA. Melanoma-derived extracellular vesicles skew neutrophils into a pro-tumor phenotype. J Leukoc Biol 2022;111:585-96.

71. Mannavola F, Tucci M, Felici C, Passarelli A, D'Oronzo S, Silvestris F. Tumor-derived exosomes promote the in vitro osteotropism of melanoma cells by activating the SDF-1/CXCR4/CXCR7 axis. J Transl Med 2019;17:230.

72. Dai J, Escara-Wilke J, Keller JM, et al. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J Exp Med 2019;216:2883-99.

73. Zhu Y, Tao Z, Chen Y, et al. Exosomal MMP-1 transfers metastasis potential in triple-negative breast cancer through PAR1-mediated EMT. Breast Cancer Res Treat 2022;193:65-81.

74. Gupta P, Kadamberi IP, Mittal S, et al. Tumor derived extracellular vesicles drive T cell exhaustion in tumor microenvironment through sphingosine mediated signaling and impacting immunotherapy outcomes in ovarian cancer. Adv Sci 2022;9:e2104452.

75. Graham GJ, Handel TM, Proudfoot AEI. Leukocyte adhesion: reconceptualizing chemokine presentation by glycosaminoglycans. Trends Immunol 2019;40:472-81.

76. Das K, Prasad R, Singh A, et al. Protease-activated receptor 2 promotes actomyosin dependent transforming microvesicles generation from human breast cancer. Mol Carcinog 2018;57:1707-22.

77. Verweij FJ, Bebelman MP, Jimenez CR, et al. Quantifying exosome secretion from single cells reveals a modulatory role for GPCR signaling. J Cell Biol 2018;217:1129-42.

78. Giusti I, Delle Monache S, Di Francesco M, et al. From glioblastoma to endothelial cells through extracellular vesicles: messages for angiogenesis. Tumour Biol 2016;37:12743-53.

79. Tian H, Pang J, Qin K, et al. A novel tissue-based liver-kidney-on-a-chip can mimic liver tropism of extracellular vesicles derived from breast cancer cells. Biotechnol J 2020;15:e1900107.

80. Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, et al. Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes. Cancer Immunol Immun 2006;55:808-18.

81. Baran J, Baj-Krzyworzeka M, Weglarczyk K, et al. Circulating tumour-derived microvesicles in plasma of gastric cancer patients. Cancer Immunol Immun 2010;59:841-50.

82. Janowska-Wieczorek A, Marquez-Curtis LA, Wysoczynski M, Ratajczak MZ. Enhancing effect of platelet-derived microvesicles on the invasive potential of breast cancer cells. Transfusion 2006;46:1199-209.

83. Wang D, Wang X, Song Y, et al. Exosomal miR-146a-5p and miR-155-5p promote CXCL12/CXCR7-induced metastasis of colorectal cancer by crosstalk with cancer-associated fibroblasts. Cell Death Dis 2022;13:380.

84. Ludwig N, Yerneni SS, Azambuja JH, et al. Tumor-derived exosomes promote angiogenesis via adenosine A2B receptor signaling. Angiogenesis 2020;23:599-610.

85. Clayton A, Al-Taei S, Webber J, Mason MD, Tabi Z. Cancer exosomes express CD39 and CD73, which suppress T cells through adenosine production. J Immunol 2011;187:676-83.

86. Salimu J, Webber J, Gurney M, Al-Taei S, Clayton A, Tabi Z. Dominant immunosuppression of dendritic cell function by prostate-cancer-derived exosomes. J Extracell Vesicles 2017;6:1368823.

87. Castellana D, Zobairi F, Martinez MC, et al. Membrane microvesicles as actors in the establishment of a favorable prostatic tumoral niche: a role for activated fibroblasts and CX3CL1-CX3CR1 axis. Cancer Res 2009;69:785-93.

88. Jethwa SA, Leah EJ, Zhang Q, et al. Exosomes bind to autotaxin and act as a physiological delivery mechanism to stimulate LPA receptor signalling in cells. J Cell Sci 2016;129:3948-57.

89. Muinonen-Martin AJ, Susanto O, Zhang Q, et al. Melanoma cells break down LPA to establish local gradients that drive chemotactic dispersal. PLoS Biol 2014;12:e1001966.

90. Che SPY, Park JY, Stokol T. Tissue factor-expressing tumor-derived extracellular vesicles activate quiescent endothelial cells via protease-activated receptor-1. Front Oncol 2017;7:261.

91. Li C, Liu DR, Li GG, et al. CD97 promotes gastric cancer cell proliferation and invasion through exosome-mediated MAPK signaling pathway. World J Gastroenterol 2015;21:6215-28.

92. Liu D, Li C, Trojanowicz B, et al. CD97 promotion of gastric carcinoma lymphatic metastasis is exosome dependent. Gastric Cancer 2016;19:754-66.

93. Yoshioka Y, Shimomura M, Saito K, et al. Circulating cancer-associated extracellular vesicles as early detection and recurrence biomarkers for pancreatic cancer. Cancer Sci 2022;113:3498-509.

94. Yamaga R, Ikeda K, Boele J, et al. Systemic identification of estrogen-regulated genes in breast cancer cells through cap analysis of gene expression mapping. Biochem Biophys Res Commun 2014;447:531-6.

95. Scavo MP, Cigliano A, Depalo N, et al. Frizzled-10 extracellular vesicles plasma concentration is associated with tumoral progression in patients with colorectal and gastric cancer. J Oncol 2019;2019:2715968.

96. Scavo MP, Depalo N, Rizzi F, et al. FZD10 carried by exosomes sustains cancer cell proliferation. Cells 2019;8:777.

97. Irmer B, Efing J, Reitnauer LE, et al. Extracellular vesicle-associated tyrosine kinase-like orphan receptors ROR1 and ROR2 promote breast cancer progression. Cell Commun Signal 2023;21:171.

98. Luga V, Zhang L, Viloria-Petit AM, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell 2012;151:1542-56.

99. Hu YB, Yan C, Mu L, et al. Exosomal Wnt-induced dedifferentiation of colorectal cancer cells contributes to chemotherapy resistance. Oncogene 2019;38:1951-65.

Cite This Article

Export citation file: BibTeX | EndNote | RIS

OAE Style

Di Niro L, Linders AC, Glynn T, Pegtel DM, Siderius M, Crudden C, Smit MJ. G protein-coupled receptors: a gateway to targeting oncogenic EVs?. Extracell Vesicles Circ Nucleic Acids 2024;5:233-48. http://dx.doi.org/10.20517/evcna.2024.10

AMA Style

Di Niro L, Linders AC, Glynn T, Pegtel DM, Siderius M, Crudden C, Smit MJ. G protein-coupled receptors: a gateway to targeting oncogenic EVs?. Extracellular Vesicles and Circulating Nucleic Acids. 2024; 5(2): 233-48. http://dx.doi.org/10.20517/evcna.2024.10

Chicago/Turabian Style

Lotte Di Niro, Amber C. Linders, Thomas Glynn, D. Michiel Pegtel, Marco Siderius, Caitrin Crudden, Martine J. Smit. 2024. "G protein-coupled receptors: a gateway to targeting oncogenic EVs?" Extracellular Vesicles and Circulating Nucleic Acids. 5, no.2: 233-48. http://dx.doi.org/10.20517/evcna.2024.10

ACS Style

Di Niro, L.; Linders AC.; Glynn T.; Pegtel DM.; Siderius M.; Crudden C.; Smit MJ. G protein-coupled receptors: a gateway to targeting oncogenic EVs?. Extracell. Vesicles. Circ. Nucleic. Acids. 2024, 5, 233-48. http://dx.doi.org/10.20517/evcna.2024.10

About This Article

Special Issue

This article belongs to the Special Issue EV Insight
© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
436
Downloads
45
Citations
0
Comments
0
13

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Extracellular Vesicles and Circulating Nucleic Acids
ISSN 2767-6641 (Online)
Follow Us

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/