Abstract
Purpose of review
CXCR2 is key stimulant of immune cell migration and recruitment, especially of neutrophils. Alleviating excessive neutrophil accumulation and infiltration could prevent prolonged tissue damage in inflammatory disorders. This review focuses on recent advances in our understanding of the role of CXCR2 in regulating neutrophil migration and the use of CXCR2 antagonists for therapeutic benefit in inflammatory disorders.
Recent findings
Recent studies have provided new insights into how CXCR2 signaling regulates hematopoietic cell mobilization and function in both health and disease. We also summarize several CXCR2 regulatory mechanisms during infection and inflammation such as via Wip1, T-bet, P-selectin glycoprotein ligand-1, granulocyte-colony-stimulating factor, and microbiome. Moreover, we provide an update of studies investigating CXCR2 blockade in the laboratory and in clinical trials.
Summary
Neutrophil homeostasis, migration, and recruitment must be precisely regulated. The CXCR2 signaling pathway is a potential target for modifying neutrophil dynamics in inflammatory disorders. We discuss the recent clinical use of CXCR2 antagonists for controlling inflammation.
Keywords: CXCR2, CXCR2 antagonist, infection, inflammation, neutrophils
INTRODUCTION
Chemokines are a large family of signaling proteins that mediate cellular migration, especially of immune cells. CXCR2 is mainly expressed on neutrophils, monocytes, natural killer cells, mast cells, and endothelial cells, and is the receptor for CXC chemokine ligands, especially CXCL8 [1]. CXCR2 mediates a G-protein-coupled receptor (GPCR) signaling cascade, with its activation starting with dissociation of the receptor from the G-protein, followed by release of the Gβ subunit from the Gα subunit [2]. It has been shown that CXCR2 is phosphorylated by GPCR kinases after activation, which triggers dynamin-mediated and clathrin-mediated receptor internalization mediated by β-arrestin1/2 and AP-2 [3]. Activated CXCR2 induces calcium release, activation of the Ras/MAPK, and PI3K signaling cascades, and it is involved in many immune responses including directed neutrophil migration [4]. Recently, Del Prete et al. [5■■] reported that CXCR2-mediated neutrophil recruitment to sites of inflammation can be regulated by CCRL2, a seven-transmembrane domain receptor that shares structural and functional similarities with atypical chemokine receptors. CCRL2/CXCR2 heterodimerization regulates membrane CXCR2 expression and function, providing a novel mechanism for its regulation. For example, CXCR2-mediated signaling was impaired in CCRL2-deficient neutrophils.
Neutrophils express over 30 receptor types including pattern recognition receptors, cytokine receptors, adhesion receptors, and GPCRs [6]. They play a central role in innate immunity via sensing various stimuli through these receptors, acting as the first line of host defense against infection. Neutrophils migrate to sites of infection to then control bacterial burden. A recent study suggests that neutrophil recruitment during inflammation proceeds in two phases: an early phase, mediated by short-lived signals, followed by an amplification phase to prolong neutrophil recruitment and activation, which is mediated by signaling cascades through leukotriene-B4 and CXCL8-family chemokines [7,8]. Excess neutrophil infiltration can enhance the inflammatory response and prolong tissue damage [9-11]. Balancing pathogen control and inflammation-related tissue injury is largely dependent upon neutrophil homeostasis as well as the degree of neutrophil activation in the presence of various stimuli. Regulating neutrophil CXCR2 and its ligand expression are potential therapeutic targets for controlling neutrophil recruitment and function in inflammatory disorders.
THE EFFECT OF CXCR2 SIGNALING ON HEMATOPOIETIC CELL MOBILIZATION AND RECRUITMENT
CXCR2 is known to be involved in neutrophil recruitment from peripheral blood to inflamed tissue. Chou et al. illustrated the amplification stage of neutrophil recruitment in an inflammatory arthritis modal and showed that cytokines such as IL-1β and chemokines such as CXCL12 released by activated neutrophils activate synovial cells. These activated synovial tissues then produced ligands for CXCR1 and CXCR2 to further promote sequential neutrophil activation [12]. CXCR2 is also responsible for neutrophil migration to tumors in mouse cancer models. Significantly, CXCR2 expression was significantly lower in tumor-associated neutrophils than those in the bone marrow and peripheral blood [13], with endogenous IFN-β inhibiting CXCR2-induced neutrophil recruitment by reducing CXCR2 ligand expression and chemokine gradients. Bian et al. recently reported myeloid-derived suppressor cells (MDSCs) with high CXCR2 expression that promoted tumor progression. These G-MDSCs were functionally different to mature neutrophils and accumulated in tumors to immunosuppress T cells [14■].
The role of CXCR2 in neutrophil migration from bone marrow to peripheral blood has also been well explored. ELR+ chemokines such as CXCL8 stimulated neutrophil mobilization in a CXCR1-dependent and CXCR2-dependent manner [15]. In addition, neutrophils lacking CXCR2 have impaired emigratory capacity [16]. Eash et al. [17] showed that CXCR2 signaling antagonized CXCR4 to regulate neutrophil release from the bone marrow. More recently, granulocyte-colony-stimulating factor (G-CSF) and CXCR2 have been shown to interact during inflammation. G-CSF is a well known neutrophil-mobilizing cytokine [18], and its mobilization function is believed to be mediated by CXCR2 ligands [19], as G-CSF failed to mobilize neutrophils in CXCR2-deficient mice. In an arthritis modal, anti-G-CSF receptor blockade not only significantly reduced cell adhesion in joints, but also reduced CXCR2-expressing circulating neutrophils [20■]. On the other hand, the activation of CRCR2 can also be modulated by G-CSF. Compared with G-CSF, CXCR2-induced neutrophil mobilization much more quickly [21,22], indicating a distinct mobilization mechanism. Bajrami et al. [23■■] revealed that Escherichia coli-induced G-CSF increases during the early stages of inflammation inhibited rapid CXCR2-mediated neutrophil mobilization. G-CSF was elevated after CXCR2 ligands macrophage inflammatory protein 2 and keratinocyte chemoattractant were expressed, and this elevation inhibited CXCR2-mediated neutrophil chemotaxis. Further, only CXCR2-induced neutrophil mobilization from the bone marrow was suppressed by G-CSF and not other chemoattractants such as leukotriene-B4, C5a, or fMLP. The regulatory function of G-CSF on CXCR2 signaling indicated that exaggerated neutrophil mobilization may be achieved by targeting G-CSF-elicited pathways.
CXCR2 signaling has also been implicated in the mobilization of hematopoietic stem/progenitor cells (HSPCs). Clinically, HSPCs are usually mobilized from the bone marrow to peripheral blood using a G-CSF regimen. The effect of CXCR2 on HSPC mobilization was first reported about 10 years ago [22]. More recently, Hoggatt et al. [24■■] reported an alternative HSPC mobilization regimen, a combination of GROb and AMD3100 (a CXCR2 agonist and a CXCR4 antagonist, respectively), which increased HSPC mobilization by elevating matrix metalloproteinase 9. Importantly, the new regimen displayed better engraftment and greater donor chimerism compared with the G-CSF regimen.
THE ROLE OF CXCR2 IN INFECTION AND INFLAMMATION
As noted above, neutrophils can migrate and recruit to inflamed sites under the control of CXCR2 during infection to exert their antimicrobial function. CXCR2 expression on neutrophils is downregulated during sepsis. Significantly, disruption of phospholipase D2 significantly upregulated CXCR2 expression in septic mice, increasing bactericidal activity, decreasing vital organ damage, and improving survival during experimental sepsis [25]. In another study, Shen et al. [26■] demonstrated that inhibition of wild-type p53-induced phosphatase 1 attenuated CXCR2 internalization and improved sepsis outcomes in mice. In addition, a negative relationship between neutrophil CXCR2 and Wip expression has also been observed in septic patients. Although CXCR2 signaling is clearly required for host defense responses, in a modal of Staphylococcus aureus-induced septic arthritis, delayed CXCR1/2 blockade effectively controlled the articular inflammatory damage without compromising antibacterial responses [27■].
Inflammation, including neutrophil adhesion to lesional vascular endothelial cells, contributes to deep vein thrombosis. Yago et al.[28■■] demonstrated that synergy between P-selectin glycoprotein ligand-1 and CXCR2 on rolling neutrophils increased neutrophil adhesion and procoagulant neutrophil extracellular trap (NET) release, increasing thrombus frequency and size. Gollomp et al.[29■] investigated the molecular mechanism of NETs during thrombosis development in a heparin-induced thrombocytopenia model. They showed that neutrophils migrated via a CXCR2-dependent mechanism to accumulate in thrombi, and CXCR2 antagonist therapy reduced neutrophil counts in thrombi with clinical significance.
The role of CXCR2 signaling in other inflammatory diseases has also been investigated in recent years. CXCR2 has been considered as a potential pharmacological target in controlling inflammatory damage in the pathogenesis of chronic obstructive pulmonary disease (COPD) [30,31], asthma [32], arthritis [20■], and hepatitis [33,34]. Hoegl et al. [35■] reported that reduced CXCR2 chemokines were related to attenuated endotoxin-induced lung injury in Tbet−/− mice. There are also intriguing associations between the microbiome and susceptibility to amebiasis infection, with dysbiosis increasing the susceptibility and severity of amebiasis infection, with fewer infiltrating cecal neutrophils and diminished CXCR2 expression [36■■]. These results suggest that the interaction between neutrophils and the microbiome may be a promising target in colitis treatment and emphasize the role of CXCR2 in regulating intestinal neutrophil recruitment.
CXCR2 BLOCKADE IN ACUTE INFLAMMATION
Several attempts have been made to regulate neutrophil migration and recruitment to control inflammatory diseases. The role of CXCR2 in regulating neutrophil recruitment is now better established, especially through the use of CXCR2 knockout strategies [37]. Hence, blocking CXCR2 signaling, including with CXCR2 antagonists, chemokine analogs, and CXCR2 receptor antibodies, has been applied to inflammatory diseases [37]. Several different CXCR2 antagonist structures exist: diarylurea and its analogs, compounds in which urea is replaced with squaramide, pyrimidine-based compounds, and bicyclic pyrimidine compounds. Of these, the diarylurea compounds were the first to be described. These different structures provide varying improvements in oral bioavailability, receptor selectivity, and tolerance [38].
Some popular CXCR2 antagonists are listed in Table 1, and those in clinical trials are summarized in Table 2. The types of CXCR2 blockers are wide with similarly diverse indications, including COPD, bronchiectasis, asthma, nephropathy, pancreatitis, and cancer. Taking respiratory inflammation as an example, AZD5069 has been reported to reduce airway neutrophilia [39■,40]. However, safety remains of concern, with clinical results showing that even though neutrophil immunoprotection was not compromised during CXCR2 inhibition [39■], this neutrophil-targeted therapy still increased infection susceptibility.
Table 1.
Name | Structure | Model | Therapeutic use or clinical trial | Reference |
---|---|---|---|---|
Danirixin | 1-[4-chloro-2-hydroxy-3-[(3S)-piperidin-3-yl] sulfonylphenyl]-3-(3-fluoro-2-methylphenyl)urea | Rat | Phase II for COPD | [41-43] |
SCH527123 (MK-7123) | 2-hydroxy-N,N-dimethyl-3-[[2-[[(1R)-1-(5-methylfuran-2-yl)propyl]amino]-3,4-dioxocyclobuten-1-yl]amino]benzamide | Mouse, rat | Phase II for COPD, asthma, traumatic brain injury, breast cancer, pancreatic cancer, acute-on-chronic liver failure | [44-46] |
Reparixin | (2R)-2-[4-(2-methylpropyl)phenyl]-N-methylsulfonylpropanamide | Rat, mouse, and pig | Phase II for kidney and lung transplantation, metastatic breast cancer; Phase III for pancreatic islet transplantation in type 1 diabetes |
[47-51] |
AZD5069 | N-(2-((2,3-difluorobenzyl)thio)-6-(((2R,3S)-3,4-dihydroxybutan-2-yl)oxy)pyrimidin-4-yl)azetidine-1-sulfonamide | Monkey | Phases Ib and II for head and neck cancer, metastatic castration-resistant prostate cancer; Phase II for asthma, COPD, bronchiectasis, coronary artery disease |
[39■,52-55] |
AZD8309 | (R)-5-[[(2,3-difluorophenyl)methyl]thio]-7-[(2-hydroxy-1-methylethyl)amino]-thiazolo[4,5-d]pyrimidin-2(3H)-one | Mouse | Phase I for rheumatoid arthritis, COPD, pancreatitis | [56,57] |
G31P | 6-Chloronicotinamide N-oxide 4a | Mouse, rat, and guinea pig | Aspiration pneumonia; ischemia and reperfusion injury; lung cancer; mastitis lesions; diabetic nephropathy; UAN kidneys; hepatocellular carcinoma; prostate cancer; ulcerative colitis | [58-61] |
Ladarixin | (1,1-dioxo-1λ6-thiomorpholin-4-yl)-{6-[3-(4-fluoro-phenyl)-5-methyl-isoxazol-4-ylmethoxy]-pyridin-3-yl}-methanone | Mouse | Type 1 diabetes | [62] |
COPD, chronic obstructive pulmonary disease; UAN, uric acid nephropathy.
Table 2.
CXCR2 blockade | Clinical trial | Identifier |
---|---|---|
AZD5069 | Phases Ib and II for head and neck cancer | |
Phases Ib and II for metastatic castration-resistant prostate cancer | ||
Phases Ib and II for metastatic pancreatic Ductal adenocarcinoma | ||
Phase II for asthma | ||
Phase II for COPD | ||
Phase II for bronchiectasis | ||
Danirixin | Phase II for COPD | |
SCH527123 (MK-7123) | For solid tumors (nonsmall-cell lung cancer, castration-resistant prostate cancer, and microsatellite stable colorectal cancer) | |
AZD8309 | The effect on cells and inflammatory biomarkers after nasal challenge with LPS | |
Reparixin | Phase I for metastatic breast cancer | |
Phase II/III for pancreatic islet transplantation in type 1 diabetes mellitus | ||
Phase II for ischemia-reperfusion injury in liver transplantation | ||
Phase II for breast cancer | ||
Phase II for pancreatectomy for chronic pancreatitis | ||
Phase II for ischemia-reperfusion injury kidney diseases | ||
Phase II for ischemia-reperfusion injury lung transplantation | ||
Ladarixin | Phase II for new-onset type 1 diabetes |
COPD, chronic obstructive pulmonary disease; LPS, lipopolysaccharide.
CONCLUSION
It is now well established that CXCR2 is a crucial regulator of hematopoietic cell homeostasis, migration, and recruitment, and is implicated in a variety of inflammatory diseases and cancer. Therefore, CXCR2 and its related signaling pathways could be promising pharmacological targets in these diseases. Efforts still need to be made to fully understand the pathophysiological role of CXCR2 signaling in health and disease and to define optimal regimens with improved compounds with ideal pharmacokinetics, treatment timing, and clinical responses.
KEY POINTS.
CXCR2 is a crucial regulator of hematopoietic cell homeostasis, migration, and recruitment, and is implicated in a variety of inflammatory diseases and cancer.
CXCR2 expression and activation are tightly regulated during infection and inflammation.
CXCR2 and the related signaling pathways could be promising pharmacological targets in various infections and inflammatory diseases.
Efforts still need to be made to fully understand the pathophysiological role of CXCR2 signaling in health and disease and to define optimal regimens with improved compounds with ideal pharmacokinetics, treatment timing, and clinical responses.
Acknowledgments
Financial support and sponsorship
Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences (2018RC31002, 2017PT31033); the Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Sciences (2017-I2M-1-015); State Key Laboratory of Experimental Hematology Research Grant (157-Z18-01); NIH grants (R01AI103142, R01HL092020, and P01 HL095489) and a grant from FAMRI (CIA 123008).
Footnotes
Conflicts of interest
There are no conflicts of interest.
REFERENCES AND RECOMMENDED READING
Papers of particular interest, published within the annual period of review, have been highlighted as:
■ of special interest
■■ of outstanding interest
- 1.Manzo A, Caporali R, Montecucco C, Pitzalis C. Role of chemokines and chemokine receptors in regulating specific leukocyte trafficking in the immune/inflammatory response. Clin Exp Rheumatol 2003; 21:501–508. [PubMed] [Google Scholar]
- 2.Wu Y, Wang S, Farooq SM, et al. A chemokine receptor CXCR2 macromolecular complex regulates neutrophil functions in inflammatory diseases. J Biol Chem 2012; 287:5744–5755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Raghuwanshi SK, Su Y, Singh V, et al. The chemokine receptors CXCR1 and CXCR2 couple to distinct G protein-coupled receptor kinases to mediate and regulate leukocyte functions. J Immunol 2012; 189:2824–2832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Thelen M, Munoz LM, Rodriguez-Frade JM, Mellado M. Chemokine receptor oligomerization: functional considerations. Curr Opin Pharmacol 2010; 10:38–43. [DOI] [PubMed] [Google Scholar]
- 5.■■.Del Prete A, Martinez-Munoz L, Mazzon C, et al. The atypical receptor CCRL2 is required for CXCR2-dependent neutrophil recruitment and tissue damage. Blood 2017; 130:1223–1234.The article revealed that CCRL2 could constitutively form homodimers and heterodimers with CXCR2. CCRL2/CXCR2 heterodimerization regulated membrane expression and promoted CXCR2 function, providing a novel mechanism for CXCR2 functional regulation.
- 6.Futosi K, Fodor S, Mocsai A. Neutrophil cell surface receptors and their intracellular signal transduction pathways. Int Immunopharmacol 2013; 17:638–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sadik CD, Luster AD. Lipid–cytokine–chemokine cascades orchestrate leukocyte recruitment in inflammation. J Leukoc Biol 2012; 91:207–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.de Oliveira S, Rosowski EE, Huttenlocher A. Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol 2016; 16:378–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol 2013; 13:159–175. [DOI] [PubMed] [Google Scholar]
- 10.Borregaard N Neutrophils, from marrow to microbes. Immunity 2010; 33:657–670. [DOI] [PubMed] [Google Scholar]
- 11.Caielli S, Banchereau J, Pascual V. Neutrophils come of age in chronic inflammation. Curr Opin Immunol 2012; 24:671–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chou RC, Kim ND, Sadik CD, et al. Lipid–cytokine–chemokine cascade drives neutrophil recruitment in a murine model of inflammatory arthritis. Immunity 2010; 33:266–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jablonska J, Wu CF, Andzinski L, et al. CXCR2-mediated tumor-associated neutrophil recruitment is regulated by IFN-beta. Int J Cancer 2014; 134:1346–1358. [DOI] [PubMed] [Google Scholar]
- 14.■.Bian Z, Shi L, Venkataramani M, et al. Tumor conditions induce bone marrow expansion of granulocytic, but not monocytic, immunosuppressive leukocytes with increased CXCR2 expression in mice. Eur J Immunol 2018; 48:532–542.The article demonstrated that G-myeloid-derived suppressor cells (G-MDSCs) in a tumor modal, isolated using a Percoll gradient, had increased CXCR2 expression. G-MDSCs expressing elevated CXCR2 guided egress out of the bone marrow and produced arginase-1 and reactive oxygen species upon encountering antigen-activated T cells.
- 15.Burdon PC, Martin C, Rankin SM. Migration across the sinusoidal endothelium regulates neutrophil mobilization in response to ELR + CXC chemokines. Br J Haematol 2008; 142:100–108. [DOI] [PubMed] [Google Scholar]
- 16.Burdon PC, Martin C, Rankin SM. The CXC chemokine MIP-2 stimulates neutrophil mobilization from the rat bone marrow in a CD49d-dependent manner. Blood 2005; 105:2543–2548. [DOI] [PubMed] [Google Scholar]
- 17.Eash KJ, Greenbaum AM, Gopalan PK, Link DC. CXCR2 and CXCR4 antagonistically regulate neutrophil trafficking from murine bone marrow. J Clin Invest 2010; 120:2423–2431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Eyles JL, Hickey MJ, Norman MU, et al. A key role for G-CSF-induced neutrophil production and trafficking during inflammatory arthritis. Blood 2008; 112:5193–5201. [DOI] [PubMed] [Google Scholar]
- 19.Kohler A, De Filippo K, Hasenberg M, et al. G-CSF-mediated thrombopoietin release triggers neutrophil motility and mobilization from bone marrow via induction of Cxcr2 ligands. Blood 2011; 117:4349–4357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.■.Campbell IK, Leong D, Edwards KM, et al. Therapeutic targeting of the G-CSF receptor reduces neutrophil trafficking and joint inflammation in antibody-mediated inflammatory arthritis. J Immunol 2016; 197:4392–4402.The article described the effect of granulocyte-colony-stimulating factor (G-CSF) receptor blockade in a therapeutic model of inflammatory joint disease.
- 21.Fibbe WE, Pruijt JF, Velders GA, et al. Biology of IL-8-induced stem cell mobilization. Ann N Y Acad Sci 1999; 872:71–82. [DOI] [PubMed] [Google Scholar]
- 22.Pelus LM, Fukuda S. Peripheral blood stem cell mobilization: the CXCR2 ligand GRObeta rapidly mobilizes hematopoietic stem cells with enhanced engraftment properties. Exp Hematol 2006; 34:1010–1020. [DOI] [PubMed] [Google Scholar]
- 23.■■.Bajrami B, Zhu H, Kwak HJ, et al. G-CSF maintains controlled neutrophil mobilization during acute inflammation by negatively regulating CXCR2 signaling. J Exp Med 2016; 213:1999–2018.The study revealed that G-CSF can negatively regulate CXCR2 signaling and inhibit CXCR2-mediated neutrophil mobilization from the bone marrow.
- 24.■■.Hoggatt J, Singh P, Tate TA, et al. Rapid mobilization reveals a highly engraftable hematopoietic stem cell. Cell 2018; 172:191–204.The study developed a rapid stem cell mobilization regimen utilizing a unique CXCR2 agonist, GROβ, and the CXCR4 antagonist, AMD3100. Hematopoietic stem cells collected using this mobilization regimen displayed a higher engraftment efficiency than those mobilized by G-CSF.
- 25.Lee SK, Kim SD, Kook M, et al. Phospholipase D2 drives mortality in sepsis by inhibiting neutrophil extracellular trap formation and down-regulating CXCR2. J Exp Med 2015; 212:1381–1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.■.Shen XF, Zhao Y, Cao K, et al. Wip1 deficiency promotes neutrophil recruitment to the infection site and improves sepsis outcome. Front Immunol 2017; 8:1023.The study revealed the negative association between Wip1 and CXCR2 in a sepsis modal.
- 27.■.Boff D, Oliveira VLS, Queiroz CM Junior, et al. CXCR2 is critical for bacterial control and development of joint damage and pain in Staphylococcus aureus-induced septic arthritis in mouse. Eur J Immunol 2018; 48:454–463.The study investigated the role of CXCR2 in the control of infection, hypernociception, and tissue damage in Staphylococcus aureus-induced septic arthritis in mice. CXCR2 was critical for bacterial control and development of joint damage. Treatment with DF2156A, noncompetitive antagonist of CXCR1/2, effectively controlled tissue inflammation and dysfunction but its effects were highly dependent on the timing of treatment.
- 28.■■.Yago T, Liu Z, Ahamed J, McEver RP. Cooperative PSGL-1 and CXCR2 signaling in neutrophils promotes deep vein thrombosis in mice. Blood 2018; 132:1426–1437.The study illustrated the signals that mediate the release procoagulant factor by neutrophils and the cooperation between P-selectin glycoprotein ligand-1 and CXCR2 on rolling neutrophils during thrombosis.
- 29.■.Gollomp K, Kim M, Johnston I, et al. Neutrophil accumulation and NET release contribute to thrombosis in HIT. JCI Insight 2018; 3:e99445.The article described CXCR2-dependent neutrophil migration in thrombosis in heparin-induced thrombocytopenia.
- 30.Lerner CA, Lei W, Sundar IK, Rahman I. Genetic ablation of CXCR2 protects against cigarette smoke-induced lung inflammation and injury. Front Pharmacol 2016; 7:391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Pedersen F, Waschki B, Marwitz S, et al. Neutrophil extracellular trap formation is regulated by CXCR2 in COPD neutrophils. Eur Respir J 2018; 51:1700970. [DOI] [PubMed] [Google Scholar]
- 32.Hosoki K, Rajarathnam K, Sur S. Attenuation of murine allergic airway inflammation with a CXCR1/CXCR2 chemokine receptor inhibitor. Clin Exp Allergy 2018; doi: 10.1111/cea.13275. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ye D, Yang K, Zang S, et al. Lipocalin-2 mediates nonalcoholic steatohepatitis by promoting neutrophil-macrophage crosstalk via the induction of CXCR2.J Hepatol 2016; 65:988–997. [DOI] [PubMed] [Google Scholar]
- 34.Groepper C, Rufinatscha K, Schroder N, et al. HCV modifies EGF signalling and upregulates production of CXCR2 ligands: role in inflammation and antiviral immune response. J Hepatol 2018; 69:594–602. [DOI] [PubMed] [Google Scholar]
- 35.■.Hoegl S, Ehrentraut H, Brodsky KS, et al. NK cells regulate CXCR2+ neutrophil recruitment during acute lung injury. J Leukoc Biol 2017; 101:471–480.The article demonstrated that mature Tbet+ natural killer cells were critical for the production of pulmonary CXCR2-mediated polymorphonuclear neutrophil recruitment.
- 36.■■.Watanabe K, Gilchrist CA, Uddin MJ, et al. Microbiome-mediated neutrophil recruitment via CXCR2 and protection from amebic colitis. PLoS Pathog 2017; 13:e1006513.The study revealed that dysbiosis increased the severity of amebic colitis due to decreased surface expression of CXCR2 on neutrophils and decreased CXCR2-mediated neutrophil recruitment to the gut. Blockage of CXCR2 signaling increased the susceptibility of mice to amebiasis.
- 37.Boppana NB, Devarajan A, Gopal K, et al. Blockade of CXCR2 signalling: a potential therapeutic target for preventing neutrophil-mediated inflammatory diseases. Exp Biol Med 2014; 239:509–518. [DOI] [PubMed] [Google Scholar]
- 38.Nicholls DJ, Wiley K, Dainty I, et al. Pharmacological characterization of AZD5069, a slowly reversible CXC chemokine receptor 2 antagonist. J Pharmacol Exp Ther 2015; 353:340–350. [DOI] [PubMed] [Google Scholar]
- 39.■.Uddin M, Betts C, Robinson I, et al. The chemokine CXCR2 antagonist (AZD5069) preserves neutrophil-mediated host immunity in nonhuman primates. Haematologica 2017; 102:e65–e68.The article found that AZD5069 effectively inhibited neutrophil recruitment without impairing neutrophil-mediated host immunity.
- 40.Jurcevic S, Humfrey C, Uddin M, et al. The effect of a selective CXCR2 antagonist (AZD5069) on human blood neutrophil count and innate immune functions. Br J Clin Pharmacol 2015; 80:1324–1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lazaar AL, Miller BE, Tabberer M, et al. Effect of the CXCR2 antagonist danirixin on symptoms and health status in COPD. Eur Respir J 2018; 79:809–816. [DOI] [PubMed] [Google Scholar]
- 42.Busch-Petersen J, Carpenter DC, Burman M, et al. Danirixin: a reversible and selective antagonist of the CXC chemokine receptor 2. J Pharmacol Exp Ther 2017; 362:338–346. [DOI] [PubMed] [Google Scholar]
- 43.Bloomer JC, Ambery C, Miller BE, et al. Identification and characterisation of a salt form of Danirixin with reduced pharmacokinetic variability in patient populations. Eur J Pharm Biopharm 2017; 117:224–231. [DOI] [PubMed] [Google Scholar]
- 44.Fu S, Chen X, Lin HJ, Lin J. Inhibition of interleukin 8/CX-C chemokine receptor 1,/2 signaling reduces malignant features in human pancreatic cancer cells. Int J Oncol 2018; 53:349–357. [DOI] [PubMed] [Google Scholar]
- 45.Khanam A, Trehanpati N, Riese P, et al. Blockade of neutrophil’s chemokine receptors CXCR1/2 abrogate liver damage in acute-on-chronic liver failure. Front Immunol 2017; 8:464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Todd CM, Salter BM, Murphy DM, et al. The effects of a CXCR1/CXCR2 antagonist on neutrophil migration in mild atopic asthmatic subjects. Pulm Pharmacol Ther 2016; 41:34–39. [DOI] [PubMed] [Google Scholar]
- 47.Shih CH, Chiang TB, Wang WJ. Synergistic suppression of a disintegrin acurhagin-C in combination with AZD4547 and reparixin on terminating development for human osteosarcoma MG-63 cell. Biochem Biophys Res Commun 2017; 492:513–519. [DOI] [PubMed] [Google Scholar]
- 48.de Oliveira THC, Marques PE, Poosti F, et al. Intravital microscopic evaluation of the effects of a CXCR2 antagonist in a model of liver ischemia reperfusion injury in mice. Front Immunol 2017; 8:1917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Che J, Wang Z, Sheng H, et al. Ligand-based pharmacophore model for the discovery of novel CXCR2 antagonists as anticancer metastatic agents. R Soc Open Sci 2018; 5:180176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.French BM, Sendil S, Sepuru KM, et al. Interleukin-8 mediates neutrophil–endothelial interactions in pig-to-human xenogeneic models. Xenotransplantation 2018; 25:e12385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Jia D, Li L, Andrew S, et al. An autocrine inflammatory forward-feedback loop after chemotherapy withdrawal facilitates the repopulation of drug-resistant breast cancer cells. Cell Death Dis 2017; 8:e2932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Watz H, Uddin M, Pedersen F, et al. Effects of the CXCR2 antagonist AZD5069 on lung neutrophil recruitment in asthma. Pulm Pharmacol Ther 2017; 45:121–123. [DOI] [PubMed] [Google Scholar]
- 53.O’Byrne PM, Metev H, Puu M, et al. Efficacy and safety of a CXCR2 antagonist, AZD5069, in patients with uncontrolled persistent asthma: a randomised, double-blind, placebo-controlled trial. Lancet Respir Med 2016; 4:797–806. [DOI] [PubMed] [Google Scholar]
- 54.Cullberg M, Arfvidsson C, Larsson B, et al. Pharmacokinetics of the oral selective CXCR2 antagonist AZD5069: a summary of eight phase I studies in healthy volunteers. Drugs R D 2018; 18:149–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Joseph JP, Reyes E, Guzman J, et al. CXCR2 Inhibition – a novel approach to treating CoronAry heart DiseAse (CICADA): study protocol for a randomised controlled trial. Trials 2017; 18:473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Malla SR, Karrman Mardh C, Gunther A, et al. Effect of oral administration of AZD8309, a CXCR2 antagonist, on the severity of experimental pancreatitis. Pancreatology 2016; 16:761–769. [DOI] [PubMed] [Google Scholar]
- 57.Virtala R, Ekman AK, Jansson L, et al. Airway inflammation evaluated in a human nasal lipopolysaccharide challenge model by investigating the effect of a CXCR2 inhibitor. Clin Exp Allergy 2012; 42:590–596. [DOI] [PubMed] [Google Scholar]
- 58.Ye Y, Zhang Y, Wang B, et al. CXCR1/CXCR2 antagonist G31P inhibits nephritis in a mouse model of uric acid nephropathy. Biomed Pharmacother 2018; 107:1142–1150. [DOI] [PubMed] [Google Scholar]
- 59.Walana W, Wang JJ, Yabasin IB, et al. IL-8 analogue CXCL8 (3-72) K11R/G31P, modulates LPS-induced inflammation via AKT1-NF-kbeta and ERK1/2-AP-1 pathways in THP-1 monocytes. Hum Immunol 2018; 79:809–816. [DOI] [PubMed] [Google Scholar]
- 60.Cui S, Zhu Y, Du J, et al. CXCL8 antagonist improves diabetic nephropathy in male mice with diabetes and attenuates high glucose-induced mesangial injury. Endocrinology 2017; 158:1671–1684. [DOI] [PubMed] [Google Scholar]
- 61.Walana W, Ye Y, Li M, et al. IL-8 antagonist, CXCL8(3-72)K11R/G31P coupled with probiotic exhibit variably enhanced therapeutic potential in ameliorating ulcerative colitis. Biomed Pharmacother 2018; 103:253–261. [DOI] [PubMed] [Google Scholar]
- 62.Kemp DM, Pidich A, Larijani M, et al. Ladarixin, a dual CXCR1/2 inhibitor, attenuates experimental melanomas harboring different molecular defects by affecting malignant cells and tumor microenvironment. Oncotarget 2017; 8:14428–14442. [DOI] [PMC free article] [PubMed] [Google Scholar]