Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 80
1.
Nat Commun ; 15(1): 4611, 2024 May 30.
Article En | MEDLINE | ID: mdl-38816420

G protein-coupled receptors (GPCRs) are pivotal therapeutic targets, but their complex structure poses challenges for effective drug design. Nanobodies, or single-domain antibodies, have emerged as a promising therapeutic strategy to target GPCRs, offering advantages over traditional small molecules and antibodies. However, an incomplete understanding of the structural features enabling GPCR-nanobody interactions has limited their development. In this study, we investigate VUN701, a nanobody antagonist targeting the atypical chemokine receptor 3 (ACKR3). We determine that an extended CDR3 loop is required for ACKR3 binding. Uncommon in most nanobodies, an extended CDR3 is prevalent in GPCR-targeting nanobodies. Combining experimental and computational approaches, we map an inhibitory ACKR3-VUN701 interface and define a distinct conformational mechanism for GPCR inactivation. Our results provide insights into class A GPCR-nanobody selectivity and suggest a strategy for the development of these new therapeutic tools.


Receptors, CXCR , Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Humans , Receptors, CXCR/metabolism , Receptors, CXCR/genetics , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/chemistry , HEK293 Cells , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals
2.
Mol Med Rep ; 25(3)2022 03.
Article En | MEDLINE | ID: mdl-35014674

The C­X­C receptor (CXCR) 7 agonist, VUF11207, is a chemical compound that binds specifically to CXCR7, and negatively regulates C­X­C motif chemokine ligand 12 (CXCL12) and CXCR4­induced cellular events. Lipopolysaccharide (LPS) can induce inflammatory cytokines and pathological bone loss. LPS also induces expression of CXCL12, enhancing sensitivity to receptor activator of NF­κB ligand (RANKL) and tumor necrosis factor­α (TNF­α) in vivo. RANKL and TNF­α induce the differentiation of osteoclasts into osteoclast precursors and bone resorption. The current study was performed to examine the effects of a CXCR7 agonist on osteoclastogenesis and bone resorption induced by LPS in vivo. In addition, the mechanisms underlying these in vivo effects were investigated by in vitro experiments. Eight­week­old male C57BL/6J mice were subcutaneously injected over the calvariae with LPS alone or LPS and CXCR7 agonist. After sacrifice, the number of osteoclasts and the bone resorption area were measured. In vitro experiments were performed to investigate the effects of CXCL12 and CXCR7 agonist on osteoclastogenesis induced by RANKL and TNF­α. Mice injected with LPS and CXCR7 agonist showed significantly reduced osteoclastogenesis and bone resorption compared with mice injected with LPS alone. Moreover, the CXCR7 agonist inhibited CXCL12 enhancement of RANKL­ and TNF­α­induced osteoclastogenesis in vitro. Thus, CXCR7 agonist inhibited LPS­induced osteoclast­associated cytokines, such as RANKL and TNF­α, as well as RANKL­ and TNF­α­induced osteoclastogenesis in vitro by modulating CXCL12­mediated enhancement of osteoclastogenesis. In conclusion, CXCR7 agonist reduced CXCL12­mediated osteoclastogenesis and bone resorption.


Bone Resorption/metabolism , Chemokine CXCL12/antagonists & inhibitors , Osteogenesis/drug effects , Receptors, CXCR/antagonists & inhibitors , Animals , Biomarkers , Bone Resorption/diagnosis , Bone Resorption/drug therapy , Bone Resorption/etiology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Lipopolysaccharides/immunology , Male , Mice , Mitogen-Activated Protein Kinases , Phosphorylation , RANK Ligand/metabolism , X-Ray Microtomography
3.
Int J Mol Sci ; 22(24)2021 Dec 17.
Article En | MEDLINE | ID: mdl-34948374

Peritonitis and peritonitis-associated sepsis are characterized by an increased formation of platelet-neutrophil complexes (PNCs), which contribute to an excessive migration of polymorphonuclear neutrophils (PMN) into the inflamed tissue. An important neutrophilic mechanism to capture and kill invading pathogens is the formation of neutrophil extracellular traps (NETs). Formation of PNCs and NETs are essential to eliminate pathogens, but also lead to aggravated tissue damage. The chemokine receptors CXCR4 and CXCR7 on platelets and PMNs have been shown to play a pivotal role in inflammation. Thereby, CXCR4 and CXCR7 were linked with functional adenosine A2B receptor (Adora2b) signaling. We evaluated the effects of selective CXCR4 and CXCR7 inhibition on PNCs and NETs in zymosan- and fecal-induced sepsis. We determined the formation of PNCs in the blood and, in addition, their infiltration into various organs in wild-type and Adora2b-/- mice by flow cytometry and histological methods. Further, we evaluated NET formation in both mouse lines and the impact of Adora2b signaling on it. We hypothesized that the protective effects of CXCR4 and CXCR7 antagonism on PNC and NET formation are linked with Adora2b signaling. We observed an elevated CXCR4 and CXCR7 expression in circulating platelets and PMNs during acute inflammation. Specific CXCR4 and CXCR7 inhibition reduced PNC formation in the blood, respectively, in the peritoneal, lung, and liver tissue in wild-type mice, while no protective anti-inflammatory effects were observed in Adora2b-/- animals. In vitro, CXCR4 and CXCR7 antagonism dampened PNC and NET formation with human platelets and PMNs, confirming our in vivo data. In conclusion, our study reveals new protective aspects of the pharmacological modulation of CXCR4 and CXCR7 on PNC and NET formation during acute inflammation.


Extracellular Traps/drug effects , Receptor, Adenosine A2B/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Cells, Cultured , Extracellular Traps/metabolism , Humans , Male , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Receptors, CXCR/metabolism , Receptors, CXCR4/metabolism
4.
Biomed Pharmacother ; 144: 112363, 2021 Dec.
Article En | MEDLINE | ID: mdl-34794236

Antagonism of the chemokine receptor CXCR7 has shown promising effects in diverse disease areas through modulation of its ligands, CXCL11 and CXCL12. Preclinical data of the first-in-class CXCR7 antagonist, ACT-1004-1239, showed efficacy in animal models of multiple sclerosis and acute lung injury. In healthy humans, single-dose administration of ACT-1004-1239 revealed a favorable clinical profile. Here, we report the target engagement of ACT-1004-1239 in healthy mice and humans after multiple doses using CXCL11 and CXCL12 as biomarkers. In addition, safety/tolerability, concentration-QTc relationship, and pharmacokinetics (PK) were assessed in a randomized, double-blind, placebo-controlled Phase 1 clinical study. Multiple-dose ACT-1004-1239 dose-dependently increased CXCL12 plasma concentration across the investigated dose range in mice and humans (mice: 1-100 mg/kg b.i.d.; humans: 30-200 mg o.d.) when compared to vehicle/placebo demonstrating target engagement. Mouse and human PK/PD models predicted that CXCL12 concentration approached a plateau within these dose ranges. In humans, ACT-1004-1239 was rapidly absorbed (tmax: 1.75-3.01 h) and the terminal t1/2 was approximately 19 h. Steady-state conditions were reached by Day 3 with an accumulation index of 1.2. Female subjects had overall higher exposure compared to males. Multiple-dose ACT-1004-1239 was well tolerated up to 200 mg once daily in humans. There was no evidence of ACT-1004-1239-mediated QTc interval prolongation. Overall, multiple oral doses of ACT-1004-1239 showed target engagement with CXCR7 in healthy mice and humans, therefore, assessment of CXCL12 as translational tool for further investigations in patients is warranted. Favorable safety/tolerability and PK profiles allow for further clinical development.


Oxazoles/pharmacology , Piperidines/pharmacology , Receptors, CXCR/antagonists & inhibitors , Administration, Oral , Animals , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Long QT Syndrome/chemically induced , Male , Mice , Mice, Inbred DBA , Oxazoles/administration & dosage , Piperidines/administration & dosage , Sex Characteristics
5.
Bioorg Med Chem Lett ; 50: 128320, 2021 10 15.
Article En | MEDLINE | ID: mdl-34400299

The atypical chemokine receptor C-X-C chemokine receptor type 7 (CXCR7) is an attractive therapeutic target for a variety of cardiac and immunological diseases. As a strategy to mitigate known risks associated with the development of higher molecular weight, basic compounds, a series of pyrrolidinyl-azolopyrazines were identified as promising small-molecule CXCR7 modulators. Using a highly enabled parallel medicinal chemistry strategy, structure-activity relationship studies geared towards a reduction in lipophilicity and incorporation of saturated heterocycles led to the identification of representative tool compound 20. Notably, compound 20 maintained good potency against CXCR7 with a suitable balance of physicochemical properties to support in vivo pharmacokinetic studies.


Drug Discovery , Immunologic Factors/chemical synthesis , Immunologic Factors/pharmacology , Receptors, CXCR/antagonists & inhibitors , Animals , Drug Delivery Systems , Drug Design , Immunologic Factors/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Signal Transduction , Structure-Activity Relationship
6.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article En | MEDLINE | ID: mdl-34298991

Chemokines are chemotactic cytokines that promote cancer growth, metastasis, and regulate resistance to chemotherapy. Stromal cell-derived factor 1 (SDF1) also known as C-X-C motif chemokine 12 (CXCL12), a prognostic factor, is an extracellular homeostatic chemokine that is the natural ligand for chemokine receptors C-X-C chemokine receptor type 4 (CXCR4), also known as fusin or cluster of differentiation 184 (CD184) and chemokine receptor type 7 (CXCR7). CXCR4 is the most widely expressed rhodopsin-like G protein coupled chemokine receptor (GPCR). The CXCL12-CXCR4 axis is involved in tumor growth, invasion, angiogenesis, and metastasis in colorectal cancer (CRC). CXCR7, recently termed as atypical chemokine receptor 3 (ACKR3), is amongst the G protein coupled cell surface receptor family that is also commonly expressed in a large variety of cancer cells. CXCR7, like CXCR4, regulates immunity, angiogenesis, stem cell trafficking, cell growth and organ-specific metastases. CXCR4 and CXCR7 are expressed individually or together, depending on the tumor type. When expressed together, CXCR4 and CXCR7 can form homo- or hetero-dimers. Homo- and hetero-dimerization of CXCL12 and its receptors CXCR4 and CXCR7 alter their signaling activity. Only few drugs have been approved for clinical use targeting CXCL12-CXCR4/CXCR7 axis. Several CXCR4 inhibitors are in clinical trials for solid tumor treatment with limited success whereas CXCR7-specific inhibitors are still in preclinical studies for CRC. This review focuses on current knowledge of chemokine CXCL12 and its receptors CXCR4 and CXCR7, with emphasis on targeting the CXCL12-CXCR4/CXCR7 axis as a treatment strategy for CRC.


Chemokine CXCL12/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Signal Transduction/drug effects , Animals , Chemokine CXCL12/antagonists & inhibitors , Colorectal Neoplasms/pathology , Dimerization , Humans , Neoplasm Metastasis , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction/genetics
8.
Med Oncol ; 38(5): 58, 2021 Apr 13.
Article En | MEDLINE | ID: mdl-33847822

C-X-C chemokine receptor 7 (CXCR7), a novel receptor of C-X-C motif chemokine ligand 12 (CXCL12), is associated with the occurrence and metastasis of various malignant tumours. However, the role, function and underlying mechanisms of CXCR7 expression in cervical cancer remain undefined. The expression level of CXCR7 was evaluated in cervical cancer samples by immunohistochemistry and real-time PCR analyses. Western blot analysis was used to examine the expression level of CXCR7 in cervical cancer cell lines. HeLa cells were genetically silenced or pharmacologically inhibited for CXCR7 or CXCR4. Transwell and CCK-8 assays were used to examine cell migration and proliferation. The expression levels of MMP2, MMP9, TIMP-1 and TIMP-2 in HeLa cells were assessed by western blot or real-time PCR. HeLa cells silenced for CXCR7 were subcutaneously injected into nude mice to form tumours. The expression of CXCR7 in nude mice was investigated by immunohistochemical staining. Tumour volumes and weights were measured. The in vivo expression levels of MMP2, MMP9, TIMP-1 and TIMP-2 were determined by western blot analysis and real-time PCR. CXCR7 was overexpressed in cervical cancer tissues and cell lines. CXCL12 was highly expressed in cervical cancer lines. CXCR7 silencing or CCX733 treatment rather than CXCR4 silencing or AMD3100 treatment suppressed the proliferation, migration and invasion of cervical cancer cells stimulated by CXCL12. In a xenograft tumour model, CXCR7 silencing or CCX733 treatment inhibited the volumes and weights of xenograft tumours. In addition, downregulation of CXCR7 decreased the expression levels of MMP2 and MMP9 but increased the expression levels of TIMP-1 and TIMP-2 in vivo. These data support the finding that the downregulation of CXCR7 suppresses the proliferation and metastasis of cervical cancer cells. Inhibition of CXCR7 may be a potential targeted therapy for cervical cancer.


Carcinoma, Squamous Cell/pathology , Chemokine CXCL12/physiology , Neoplasm Proteins/physiology , Receptors, CXCR/physiology , Signal Transduction/physiology , Uterine Cervical Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/drug therapy , Cell Division , Cell Line , Cell Line, Tumor , Cervix Uteri/cytology , Epithelial Cells/metabolism , Female , Humans , Matrix Metalloproteinases/biosynthesis , Mice , Mice, Nude , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Receptors, CXCR4/physiology , Uterine Cervical Neoplasms/drug therapy , Xenograft Model Antitumor Assays
9.
FASEB J ; 35(3): e21431, 2021 03.
Article En | MEDLINE | ID: mdl-33595155

Current strategies for the treatment of demyelinating diseases such as multiple sclerosis (MS) are based on anti-inflammatory or immunomodulatory drugs. Those drugs have the potential to reduce the frequency of new lesions but do not directly promote remyelination in the damaged central nervous system (CNS). Targeting CXCR7 (ACKR3) has been postulated as a potential therapeutic approach in demyelinating diseases, leading to both immunomodulation by reducing leukocyte infiltrates and promyelination by enhancing myelin repair. ACT-1004-1239 is a potent, selective, insurmountable, and orally available first-in-class CXCR7 receptor antagonist. The effect of ACT-1004-1239 was evaluated in the myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) and the cuprizone-induced demyelination mouse models. In addition, ACT-1004-1239 was assessed in a rat oligodendrocyte precursor cell (OPC) differentiation assay in vitro. In the MOG-induced EAE model, ACT-1004-1239 treatment (10-100 mg/kg, twice daily, orally) showed a significant dose-dependent reduction in disease clinical scores, resulting in increased survival. At the highest dose tested (100 mg/kg, twice daily), ACT-1004-1239 delayed disease onset and significantly reduced immune cell infiltrates into the CNS and plasma neurofilament light chain concentration. Treatment with ACT-1004-1239 dose-dependently increased plasma CXCL12 concentration, which correlated with a reduction of the cumulative disease score. Furthermore, in the cuprizone model, ACT-1004-1239 treatment significantly increased the number of mature myelinating oligodendrocytes and enhanced myelination in vivo. In vitro, ACT-1004-1239 promoted the maturation of OPCs into myelinating oligodendrocytes. These results provide evidence that ACT-1004-1239 both reduces neuroinflammation and enhances myelin repair substantiating the rationale to explore its therapeutic potential in a clinical setting.


Cuprizone/pharmacology , Immunomodulation/drug effects , Myelin Sheath/drug effects , Receptors, CXCR/antagonists & inhibitors , Animals , Cell Differentiation/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunomodulation/immunology , Inflammation/drug therapy , Male , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Myelin Sheath/pathology , Myelin-Oligodendrocyte Glycoprotein/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , Stem Cells/cytology
10.
Clin Pharmacol Ther ; 109(6): 1648-1659, 2021 06.
Article En | MEDLINE | ID: mdl-33406277

The C-X-C chemokine receptor 7 (CXCR7) has evolved as a promising, druggable target mainly in the immunology and oncology fields modulating plasma concentrations of its ligands CXCL11 and CXCL12 through receptor-mediated internalization. This "scavenging" activity creates concentration gradients of these ligands between blood vessels and tissues that drive directional cell migration. This randomized, double-blind, placebo-controlled first-in-human study assessed the safety, tolerability, pharmacokinetics, and pharmacodynamics of ACT-1004-1239, a first-in-class drug candidate small-molecule CXCR7 antagonist. Food effect and absolute bioavailability assessments were also integrated in this multipurpose study. Healthy male subjects received single ascending oral doses of ACT-1004-1239 (n = 36) or placebo (n = 12). At each of six dose levels (1-200 mg), repeated blood sampling was done over 144 hours for pharmacokinetic/pharmacodynamic assessments using CXCL11 and CXCL12 as biomarkers of target engagement. ACT-1004-1239 was safe and well tolerated up to the highest tested dose of 200 mg. CXCL12 plasma concentrations dose-dependently increased and more than doubled compared with baseline, indicating target engagement, whereas CXCL11 concentrations remained unchanged. An indirect-response pharmacokinetic/pharmacodynamic model well described the relationship between ACT-1004-1239 and CXCL12 concentrations across the full dose range, supporting once-daily dosing for future clinical studies. At doses ≥ 10 mg, time to reach maximum plasma concentration ranged from 1.3 to 3.0 hours and terminal elimination half-life from 17.8 to 23.6 hours. The exposure increase across the dose range was essentially dose-proportional and no relevant food effect on pharmacokinetics was determined. The absolute bioavailability was 53.0% based on radioactivity data after oral vs. intravenous 14 C-radiolabeled microtracer administration of ACT-1004-1239. Overall, these comprehensive data support further clinical development of ACT-1004-1239.


Chemokine CXCL12/blood , Receptors, CXCR/antagonists & inhibitors , Adult , Biological Availability , Biomarkers/blood , Chemokine CXCL11/blood , Dose-Response Relationship, Drug , Food-Drug Interactions , Half-Life , Healthy Volunteers , Humans , Male , Middle Aged , Young Adult
11.
J Med Chem ; 63(24): 15864-15882, 2020 12 24.
Article En | MEDLINE | ID: mdl-33314938

The chemokine receptor CXCR7, also known as ACKR3, is a seven-transmembrane G-protein-coupled receptor (GPCR) involved in various pathologies such as neurological diseases, autoimmune diseases, and cancers. By binding and scavenging the chemokines CXCL11 and CXCL12, CXCR7 regulates their extracellular levels. From an original high-throughput screening campaign emerged hit 3 among others. The hit-to-lead optimization led to the discovery of a novel chemotype series exemplified by the trans racemic compound 11i. This series provided CXCR7 antagonists that block CXCL11- and CXCL12-induced ß-arrestin recruitment. Further structural modifications on the trisubstituted piperidine scaffold of 11i yielded compounds with high CXCR7 antagonistic activities and balanced ADMET properties. The effort described herein culminated in the discovery of ACT-1004-1239 (28f). Biological characterization of ACT-1004-1239 demonstrated that it is a potent, insurmountable antagonist. Oral administration of ACT-1004-1239 in mice up to 100 mg/kg led to a dose-dependent increase of plasma CXCL12 concentration.


Piperidines/chemistry , Receptors, CXCR/antagonists & inhibitors , Administration, Oral , Amides/chemistry , Amines/chemistry , Animals , Chemokine CXCL12/blood , Crystallography, X-Ray , Dogs , Drug Evaluation, Preclinical , Half-Life , Humans , Inhibitory Concentration 50 , Mice , Molecular Conformation , Piperidines/metabolism , Piperidines/pharmacokinetics , Protein Binding , Rats , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Structure-Activity Relationship
12.
J Cell Mol Med ; 24(18): 10589-10603, 2020 09.
Article En | MEDLINE | ID: mdl-32761803

Low-energy shock wave (LESW) has been recognized as a promising non-invasive intervention to prevent the organs or tissues against ischaemia reperfusion injury (IRI), whereas its effect on kidney injury is rarely explored. To investigate the protective role of pretreatment with LESW on renal IRI in rats, animals were randomly divided into Sham, LESW, IRI and LESW + IRI groups. At 4, 12, 24 hours and 3 and 7 days after reperfusion, serum samples and renal tissues were harvested for performing the analysis of renal function, histopathology, immunohistochemistry, flow cytometry and Western blot, as well as enzyme-linked immunosorbent assay. Moreover, circulating endothelial progenitor cells (EPCs) were isolated, labelled with fluorescent dye and injected by tail vein. The fluorescent signals of EPCs were detected using fluorescence microscope and in vivo imaging system to track the distribution of injected circulating EPCs. Results showed that pretreatment with LESW could significantly reduce kidney injury biomarkers, tubular damage, and cell apoptosis, and promote cell proliferation and vascularization in IRI kidneys. The renoprotective role of LESW pretreatment would be attributed to the remarkably increased EPCs in the treated kidneys, part of which were recruited from circulation through SDF-1/CXCR7 pathway. In conclusion, pretreatment with LESW could increase the recruitment of circulating EPCs to attenuate and repair renal IRI.


Endothelial Progenitor Cells/physiology , Extracorporeal Shockwave Therapy , Kidney/blood supply , Reperfusion Injury/prevention & control , Animals , Apoptosis , Cell Movement , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Chemokine CXCL12/physiology , Extracorporeal Shockwave Therapy/methods , Fluorescent Dyes/pharmacokinetics , In Situ Nick-End Labeling , Kidney/pathology , Kidney/physiology , Male , Microscopy, Fluorescence , Microvessels/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/biosynthesis , Receptors, CXCR/genetics , Receptors, CXCR/physiology , Regeneration , Reperfusion Injury/blood , Reperfusion Injury/pathology , Signal Transduction , Time Factors
13.
Front Immunol ; 11: 407, 2020.
Article En | MEDLINE | ID: mdl-32210974

Our previous studies revealed a pivotal role of the chemokine stromal cell-derived factor (SDF)-1 and its receptors CXCR4 and CXCR7 on migratory behavior of polymorphonuclear granulocytes (PMNs) in pulmonary inflammation. Thereby, the SDF-1-CXCR4/CXCR7-axis was linked with adenosine signaling. However, the role of the SDF-1 receptors CXCR4 and CXCR7 in acute inflammatory peritonitis and peritonitis-related sepsis still remained unknown. The presented study provides new insight on the mechanism of a selective inhibition of CXCR4 (AMD3100) and CXCR7 (CCX771) in two models of peritonitis and peritonitis-related sepsis by injection of zymosan and fecal solution. We observed an increased expression of SDF-1, CXCR4, and CXCR7 in peritoneal tissue and various organs during acute inflammatory peritonitis. Selective inhibition of CXCR4 and CXCR7 reduced PMN accumulation in the peritoneal fluid and infiltration of neutrophils in lung and liver tissue in both models. Both inhibitors had no anti-inflammatory effects in A2B knockout animals (A2B-/-). AMD3100 and CCX771 treatment reduced capillary leakage and increased formation of tight junctions as a marker for microvascular permeability in wild type animals. In contrast, both inhibitors failed to improve capillary leakage in A2B-/- animals, highlighting the impact of the A2B-receptor in SDF-1 mediated signaling. After inflammation, the CXCR4 and CXCR7 antagonist induced an enhanced expression of the protective A2B adenosine receptor and an increased activation of cAMP (cyclic adenosine mono phosphate) response element-binding protein (CREB), as downstream signaling pathway of A2B. The CXCR4- and CXCR7-inhibitor reduced the release of cytokines in wild type animals via decreased intracellular phosphorylation of ERK and NFκB p65. In vitro, CXCR4 and CXCR7 antagonism diminished the chemokine release of human cells and increased cellular integrity by enhancing the expression of tight junctions. These protective effects were linked with functional A2B-receptor signaling, confirming our in vivo data. In conclusion, our study revealed new protective aspects of the pharmacological modulation of the SDF-1-CXCR4/CXCR7-axis during acute peritoneal inflammation in terms of the two hallmarks PMN migration and barrier integrity. Both anti-inflammatory effects were linked with functional adenosine A2B-receptor signaling.


Benzylamines/therapeutic use , Cyclams/therapeutic use , Neutrophils/immunology , Peritonitis/drug therapy , Receptor, Adenosine A2B/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Sepsis/drug therapy , Acute Disease , Animals , Benzylamines/pharmacology , Capillary Permeability , Chemokine CXCL12/metabolism , Cyclams/pharmacology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2B/genetics , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction
14.
J Cell Mol Med ; 24(4): 2464-2474, 2020 02.
Article En | MEDLINE | ID: mdl-31904910

Adult stem cells have a major role in endometrial physiology, including remodelling and repair. However, they also have a critical role in the development and progression of endometriosis. Bone marrow-derived stem cells engraft eutopic endometrium and endometriotic lesions, differentiating to both stromal and epithelial cell fates. Using a mouse bone marrow transplantation model, we show that bone marrow-derived cells engrafting endometriosis express CXCR4 and CXCR7. Targeting either receptor by the administration of small molecule receptor antagonists AMD3100 or CCX771, respectively, reduced BM-derived stem cell recruitment into endometriosis implants. Endometriosis lesion size was decreased compared to vehicle controls after treatment with each antagonist in both an early growth and established lesion treatment model. Endometriosis lesion size was not effected when the local effects of CXCL12 were abrogated using uterine-specific CXCL12 null mice, suggesting an effect primarily on bone marrow cell migration rather than a direct endometrial effect. Antagonist treatment also decreased hallmarks of endometriosis physiopathology such as pro-inflammatory cytokine production and vascularization. CXCR4 and CXCR7 antagonists are potential novel, non-hormonal therapies for endometriosis.


Benzylamines/pharmacology , Bone Marrow Cells/drug effects , Cell Movement/drug effects , Cyclams/pharmacology , Endometriosis/drug therapy , Endometrium/drug effects , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR/antagonists & inhibitors , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , Endometriosis/metabolism , Endometrium/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Uterus/drug effects , Uterus/metabolism
15.
Cytokine ; 125: 154809, 2020 01.
Article En | MEDLINE | ID: mdl-31437604

The chemokine, CXCL11, is highly expressed in different solid tumors and controls tumor growth, metastasis, and lymphocyte infiltration. Although of potential clinical interest, it is presently unknown whether these tumor-promoting activities involve the CXCL11 receptors, CXCR3 and/or CXCR7. This issue is further intrigued by the fact that CXCR3 exists in the two functionally divergent splice variants, CXCR3A and CXCR3B, which exert pro- and anti-tumorigenic influences, respectively. To unravel the role of the various CXCL11 receptors in tumor progression, we have now defined their role in CXCL11-induced chemotaxis of the tumor cell lines, A549, C33-A, DLD-1, MDA-MB-231, and PC-3. CXCL11-induced cell migration was either sensitive to the CXCR3 antagonist, ÀMG487 (DLD-1), the CXCR7 antagonist, CCX771 (C33-A, PC-3), or both (A549, MDA-231). Moreover, in C33-A and PC-3 cells, but not in the other tumor cells, pharmacological activation and inhibition of CXCR3B prevented and potentiated CXCL11-induced cell migration, respectively. Both immunocytochemistry and Western blot analysis finally revealed that the observed cell type specific organization of the CXCL11 system is not the result of differences in expression levels or subcellular location of CXCL11 receptors. Our findings imply that the therapeutic use of CXCR3 antagonists in cancer patients requires exact knowledge of the organization of the CXCR3 system in the respective tumor.


Cell Movement/drug effects , Chemokine CXCL11/metabolism , Receptors, CXCR3/metabolism , Receptors, CXCR/metabolism , Acetamides/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Disease Progression , Humans , Pyrimidinones/pharmacology , RNA Splicing , Real-Time Polymerase Chain Reaction , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR3/antagonists & inhibitors , Receptors, CXCR3/genetics , Signal Transduction/genetics , Signal Transduction/immunology
16.
Mol Pharmacol ; 96(6): 737-752, 2019 12.
Article En | MEDLINE | ID: mdl-31548340

The two G protein-coupled receptors (GPCRs) C-X-C chemokine receptor type 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) are part of the class A chemokine GPCR family and represent important drug targets for human immunodeficiency virus (HIV) infection, cancer, and inflammation diseases. CXCR4 is one of only three chemokine receptors with a US Food and Drug Administration approved therapeutic agent, the small-molecule modulator AMD3100. In this review, known modulators of the two receptors are discussed in detail. Initially, the structural relationship between receptors and ligands is reviewed on the basis of common structural motifs and available crystal structures. To date, no atypical chemokine receptor has been crystallized, which makes ligand design and predictions for these receptors more difficult. Next, the selectivity, receptor activation, and the resulting ligand-induced signaling output of chemokines and other peptide ligands are reviewed. Binding of pepducins, a class of lipid-peptides whose basis is the internal loop of a GPCR, to CXCR4 is also discussed. Finally, small-molecule modulators of CXCR4 and ACKR3 are reviewed. These modulators have led to the development of radio- and fluorescently labeled tool compounds, enabling the visualization of ligand binding and receptor characterization both in vitro and in vivo. SIGNIFICANCE STATEMENT: To investigate the pharmacological modulation of CXCR4 and ACKR3, significant effort has been focused on the discovery and development of a range of ligands, including small-molecule modulators, pepducins, and synthetic peptides. Imaging tools, such as fluorescent probes, also play a pivotal role in the field of drug discovery. This review aims to provide an overview of the aforementioned modulators that facilitate the study of CXCR4 and ACKR3 receptors.


Receptors, CXCR4/physiology , Receptors, CXCR/physiology , Amino Acid Sequence , Animals , Benzylamines , Cyclams , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacology , Humans , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, CXCR/agonists , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors
17.
Mol Pharmacol ; 96(6): 753-764, 2019 12.
Article En | MEDLINE | ID: mdl-31481460

Dysregulation of the chemokine system is implicated in a number of autoimmune and inflammatory diseases, as well as cancer. Modulation of chemokine receptor function is a very promising approach for therapeutic intervention. Despite interest from academic groups and pharmaceutical companies, there are currently few approved medicines targeting chemokine receptors. Monoclonal antibodies (mAbs) and antibody-based molecules have been successfully applied in the clinical therapy of cancer and represent a potential new class of therapeutics targeting chemokine receptors belonging to the class of G protein-coupled receptors (GPCRs). Besides conventional mAbs, single-domain antibodies and antibody scaffolds are also gaining attention as promising therapeutics. In this review, we provide an extensive overview of mAbs, single-domain antibodies, and other antibody fragments targeting CXCR4 and ACKR3, formerly referred to as CXCR7. We discuss their unique properties and advantages over small-molecule compounds, and also refer to the molecules in preclinical and clinical development. We focus on single-domain antibodies and scaffolds and their utilization in GPCR research. Additionally, structural analysis of antibody binding to CXCR4 is discussed. SIGNIFICANCE STATEMENT: Modulating the function of GPCRs, and particularly chemokine receptors, draws high interest. A comprehensive review is provided for monoclonal antibodies, antibody fragments, and variants directed at CXCR4 and ACKR3. Their advantageous functional properties, versatile applications as research tools, and use in the clinic are discussed.


Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/metabolism , Animals , Drug Delivery Systems/methods , Humans , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR4/antagonists & inhibitors
18.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2246-2256, 2019 09 01.
Article En | MEDLINE | ID: mdl-31059778

Development of cholangiocarcinoma (CCA) is dependent on a cross-talk with stromal cells, which release different chemokines including CXCL12, that interacts with two different receptors, CXCR4 and CXCR7. The aim of the present study was to investigate the role of CXCR7 in CCA cells. CXCR7 is overexpressed by different CCA cell lines and in human CCA specimens. Knock-down of CXCR7 in HuCCT-1 cells reduced migration, invasion, and CXCL12-induced adhesion to collagen I. Survival of CCA was also reduced in CXCR7-silenced cells. The ability of CXCL12 to induce cell migration and survival was also blocked by CCX733, a CXCR7 antagonist. Similar effects of CXCR7 activation were observed in CCLP-1 cells and in primary iCCA cells. Enrichment of tumor stem-like cells by a 3D culture system resulted in increased CXCR7 expression compared to cells grown in monolayers, and genetic knockdown of CXCR7 robustly reduced sphere formation both in HuCCT-1 and in CCLP-1 cells. In HuCCT-1 cells CXCR7 was found to interact with ß-arrestin 2, which was necessary to mediate CXCL12-induced migration, but not survival. In conclusion, CXCR7 is widely expressed in CCA, and contributes to the aggressive phenotype of CCA cells, inducing cell migration, invasion, adhesion, survival, growth and stem cell-like features. Cell migration induced by CXCR7 requires interaction with ß-arrestin 2.


Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Receptors, CXCR/metabolism , Bile Duct Neoplasms/metabolism , Cell Movement , Cell Survival , Chemokine CXCL12/metabolism , Cholangiocarcinoma/metabolism , Female , Humans , Liver/metabolism , Liver/pathology , Male , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/genetics , Tumor Cells, Cultured , beta-Arrestin 2/metabolism
19.
Front Immunol ; 10: 379, 2019.
Article En | MEDLINE | ID: mdl-30894861

Immunotherapy is a clinically validated treatment for many cancers to boost the immune system against tumor growth and dissemination. Several strategies are used to harness immune cells: monoclonal antibodies against tumor antigens, immune checkpoint inhibitors, vaccination, adoptive cell therapies (e.g., CAR-T cells) and cytokine administration. In the last decades, it is emerging that the chemokine system represents a potential target for immunotherapy. Chemokines, a large family of cytokines with chemotactic activity, and their cognate receptors are expressed by both cancer and stromal cells. Their altered expression in malignancies dictates leukocyte recruitment and activation, angiogenesis, cancer cell proliferation, and metastasis in all the stages of the disease. Here, we review first attempts to inhibit the chemokine system in cancer as a monotherapy or in combination with canonical or immuno-mediated therapies. We also provide recent findings about the role in cancer of atypical chemokine receptors that could become future targets for immunotherapy.


Chemokines, CC/metabolism , Chemokines, CXC/metabolism , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Receptors, CCR/metabolism , Receptors, CXCR/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Benzylamines , Cyclams , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Humans , Molecular Targeted Therapy/methods , Receptors, CCR/antagonists & inhibitors , Receptors, CXCR/antagonists & inhibitors
20.
Org Lett ; 21(6): 1622-1626, 2019 03 15.
Article En | MEDLINE | ID: mdl-30779584

CXCR7 plays an emerging role in several physiological processes. A linear peptide, amantamide (1), was isolated from marine cyanobacteria, and the structure was determined by NMR and mass spectrometry. The total synthesis was achieved by solid-phase method. After screening two biological target libraries, 1 was identified as a selective CXCR7 agonist. The selective activation of CXCR7 by 1 could provide the basis for developing CXCR7-targeted therapeutics and deciphering the role of CXCR7 in different diseases.


Amides/pharmacology , Cyanobacteria/chemistry , Peptides/chemistry , Receptors, CXCR/antagonists & inhibitors , Amides/chemistry , Molecular Structure , Receptors, CXCR/chemistry
...