Interleukin-8 in Melanoma Pathogenesis, Prognosis and Therapy—An Integrated View into Other Neoplasms and Chemokine Networks
Abstract
:1. Introduction
2. Melanoma Progression—An Interplay between Chemokines, Chemokine Receptors and Cellular Phenotypes within Tumor Microenvironment
3. Interleukin 8—A Major Key-Player in CM Pathogenesis
3.1. The Cellular Expression of IL-8 and IL-8Rs
3.2. Molecular Regulators of IL-8 Expression in CM
Cell Type | IL-8/References | Cell Type | IL-8Rs/References |
---|---|---|---|
Tumor cells | [28,30,59] | Tumor cells | [30,59] |
Melanocytes/melanoma | [60] | Melanocytes/melanoma | [60] |
Tumor stem cells | [71,79] | Keratinocytes | [63,84] |
Endothelial cells | [23] | Neurons and glial cells | [64] |
Epithelial cells | [80] | Hepathocytes | [65] |
Fibroblasts | [81] | Endothelial cells | [66] |
Cancer associated Fibroblasts | [82] | Epithelial cells | [67] |
Keratinocytes | [83,84] | Neutrophils | [30,68,74] |
Synovial cells | [61] | CD8+ T cells | [68,75] |
Smooth muscle cells | [62] | Mast cells | [68,75] |
Monocytes | [69,70] | Natural killer | [68,76] |
Macrophages | [72,85] | Myeloid derived suppressor cell | [68,77] |
T-cell lymphocytes | [73] | ||
Regulatory T cell | [78] |
3.3. IL-8 (CXCL8)/IL-8Rs(CXCR1,CXCR2) Axis in CM Progression
3.3.1. Tumor Cells and Immune Cells
IL-8 and Melanoma Cells
IL-8 and Immune Cells
3.3.2. Tumor Cells and Non-Immune Cellular Phenotypes
3.4. The Molecular Mechanisms behind IL-8-Mediated Pathways and Processes
3.4.1. Mitogen-Activated Protein Kinase (MAPK) Pathways/IL-8
3.4.2. Engulfment and Cell Motility Protein 1(ELMO1)/NF-kB/Snail/IL-8 and ELMO1/Dock180/RAC1/IL-8
3.4.3. Protein Kinase B (AKT)/IL-8
3.4.4. Beta-Catenin/Wnt/ IL-8
3.4.5. Vascular Endothelial Growth Factor (VEGF)/IL-8
3.4.6. Signal Transducer Activator of Transcription (STAT)/IL-8
3.4.7. ADRB2/PKA/IL-8
3.4.8. FKBinding Protein 51(FKBP51)/IL-8
3.5. IL-8-Mediated Pathways and Processes in the Prognostic and Therapy of CM
3.5.1. IL-8 Serum Levels
3.5.2. IL-8 and CM Therapy
Blocking IL-8 Expression
Blocking IL-8Rs
Combinatorials with Impact on IL-8/IL-8Rs Axis
Combinatorials with Impact on IL-8-Mediated Tumor Immune Cellular Milieu–MDSCs
Target | Therapeutic Strategy | Impact | Experimental Approach | References |
---|---|---|---|---|
IL-8 | mAb anti h-IL-8 (ABX-IL8) | Neutralize secreted IL-8; inhibit invasion, MMP-2 secretion, decrease vascularization | Human melanoma and animal models | [133] |
IL-8 neutralizing Abs IL-8 downregulated | Disorients DC migration, without impairing T-cell stimulation | Colon cell carcinoma tumors | [119] | |
IL-8Rs (CXCR1/2) | Inhibitors | |||
Low-molecular-weight antagonists, modified chemokines, antibodies directed against receptors | Inhibit tumor growth and angiogenesis | Human melanoma tumors in athymic nude mice | [196] | |
Antagonists to CXCR2 | Promotes tumor progression in vivo by impeding DC activation or recruitment | Colorectal cancer subtype | [208] | |
Ladarixin | Abrogates tumor cell motility, self-renewal, intratumor de novo-angiogenesis; induces apoptosis, polarizes M1 TAMs | Melanoma cells, xenografts and tumors | [198] | |
Reparixin | Reducing cancer stem cells by targeting their CXCR1 | Breast cancer clinical trial phase 3 | [197] | |
COMBINATORIALS IL-8/IL-8Rs agonists, mAbs, gene downregulators, tumor specific pathways/key molecules, chemotherapeuticals, immune cell modulators | Braf inhibitor/vemurafenib; MEK1/2 inhibitor/ trametinib | Decrease IL-8 and suppress tumor evolution | Melanoma cell subpopulations | [199] |
mAb (ABX-IL8) + MUC18 + DITC | Overcome resistance to chemotherapy and improve survival of patients | Metastatic melanoma | [194] | |
Si-IL-8 treatments + docetaxel | Downregulate IL-8 and potentiate chemotherapeutic agents | Ovarian tumor xenografts | [195] | |
mAbs anti-PD1/PD-L1- (nivolumab, pembrolizumab, atezolizumab) + mAbs antiCTLA-(Ipilimumab) | Tumor burden changes | Patients with melanoma and NSCLC | [206] | |
Reparixin + paclitaxel | Increases tumor sensibility to chemotherapy | HER-2 negative metastatic breast cancer | [211] | |
Inhibitors of CXCR1/2 + CXCL12/CXCR4 or CCR5 | Hinder recruitment of neutrophils in tumor microenvironment | Metastatic colorectal cancer | [213] | |
mAb anti-CD40 + gemcitabine; PI3Kg inhibitors+ nivolumab | Increases pro-inflammatory gene expression in TAMs, reprogramming of M2 to pro-inflammatory M1 phenotypes, anti-tumor activity | Pancreatic ductal carcinoma | [215] | |
Synergistic PI3K/mTOR and JAK2 /STAT5 inhibition | Reduced cancer cell number and tumor growth, decreased tumor seeding, metastasis, increased overall survival of the animals. | Breast cancer | [170] |
4. Conclusions and Perspectives
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Payne, A.S.; Cornelius, L.A. The Role of Chemokines in Melanoma Tumor Growth and Metastasis. J. Investig. Dermatol. 2002, 118, 915–922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balkwill, F. Cancer and the chemokine network. Nat. Rev. Cancer 2004, 4, 540–550. [Google Scholar] [CrossRef] [PubMed]
- Bonecchi, R.; Locati, M.; Mantovani, A. Chemokines and Cancer: A Fatal Attraction. Cancer Cell 2011, 19, 434–435. [Google Scholar] [CrossRef] [Green Version]
- Marcuzzi, E.; Angioni, R.; Molon, B.; Calì, B. Chemokines and chemokine receptors: Orchestrating tumor metastasization. Int. J. Mol. Sci. 2019, 20, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; Seethla, R.R.; Zhang, Q.; Gooding, W.; van Waes, C.; Hasegawa, G.; Ferris, L.R. Autocrine and paracrine chemokine receptor 7 activation in head and neck cancer: Implications for therapy. J. Natl. Cancer Inst. 2008, 100, 502–512. [Google Scholar] [CrossRef] [Green Version]
- Vilgelm, A.E.; Richmond, A. Chemokins modulate immune surveillance in tumorignesis, metastatsis, and response to immunotherapy. Front. Immunol. 2019, 10, 333. [Google Scholar] [CrossRef] [Green Version]
- Franciszkiewicz, K.; Boissonnas, A.; Boutet, M.; Combadière, C.; Mami-Chouaib, F. Role of chemokines and chemokine receptors in shaping the effector phase of the antitumor immune response. Cancer Res. 2012, 72, 6325–6332. [Google Scholar] [CrossRef] [Green Version]
- Speyer, C.L.; Ward, P.A. Role of endothelial chemokines and their receptors during inflammation. J. Investig. Surg. 2011, 24, 18–27. [Google Scholar] [CrossRef]
- Hussain, S.; Peng, B.; Cherian, M.; Song, J.W.; Ahirwar, D.K.; Ganju, R.K. The Roles of Stroma-Derived Chemokine inDifferent Stages of Cancer Metastases. Front. Immunol. 2020, 11, 598532. [Google Scholar] [CrossRef]
- Mishra, P.; Banerjee, D.; Ben-Baruch, A. Chemokines at the crossroads of tumor-fibroblast interactions that promote malignancy. J. Leukoc. Biol. 2011, 89, 31–39. [Google Scholar] [CrossRef]
- Strieter, R.M.; Burdick, M.D.; Gomperts, B.N.; Belperio, J.A.; Keane, M.P. CXC chemokines in angiogenesis. Cytokine Growth Factor Rev. 2005, 16, 593–609. [Google Scholar] [CrossRef] [Green Version]
- Suarez-Carmona, M.; Lesage, J.; Cataldo, D.; Gilles, C. EMT and inflammation: Inseparable actors of cancer progression. Mol. Oncol. 2017, 11, 805–823. [Google Scholar] [CrossRef]
- Corrò, C.; Healy, M.E.; Engler, S.; Bodenmiller, B.; Li, Z.; Schraml, P.; Weber, A.; Frew, J.I.; Rechsteiner, M.; Moch, H. IL-8 and CXCR1 expression is associated with cancer stem cell-like properties of clear cell renal cancer. J. Pathol. 2019, 248, 377–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Sarrou, E.; Podgrabinska, S.; Cassella, M.; Mungamuri, S.K.; Feirt, N.; Gordon, R.; Nagi, C.S.; Wang, Y.; Entenberg, D.; et al. Tumor cell entry into the lymph node is controlled by CCL1 chemokine expressed by lymph node lymphatic sinuses. J. Exp. Med. 2013, 210, 1509–1528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bie, Y.; Ge, W.; Yang, Z.; Cheng, X.; Zhao, Z.; Li, S.; Wang, W.; Wang, Y.; Zhao, X.; Yin, Z.; et al. The Crucial Role of CXCL8 and its receptors in colorectal liver metastasis. Dis. Markers 2019, 2019, 8023460. [Google Scholar] [CrossRef]
- Psaila, B.; Lyden, D. The Metastatic Niche: Adapting the Foreign Soil Bethan. Nat. Rev. Cancer 2009, 9, 285–293. [Google Scholar] [CrossRef]
- Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodriguez, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
- Dong, C.; Slattery, M.J.; Liang, S.; Peng, H.H. Melanoma Cell Extravasation under Flow Conditions Is Modulated by Leukocytes and Endogenously Produced Interleukin. Mol. Cell Biomech. 2005, 2, 145–159. [Google Scholar]
- Moore, B.B.; Kunkel, S.L. Attracting Attention: Discovery of IL-8/CXCL8 and the Birth of the Chemokine Field. J. Immunol. 2019, 202, 3–4. [Google Scholar] [CrossRef] [Green Version]
- Caballero-Campo, P.; Domínguez, F.; Coloma, J.; Meseguer, M.; Rehomí, J.; Pellicer, A.; Simón, C. Hormonal and embryonic regulation of chemokines IL-8, MCP-1 and RANTES in the human endometrium during the window of implantation. Mol. Hum. Reprod. 2002, 8, 375–384. [Google Scholar] [CrossRef] [Green Version]
- Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil Extracellular Traps Kill Bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
- Li, A.; Dubey, S.; Varney, M.L.; Dave, B.J.; Singh, R.K. IL-8 Directly Enhanced Endothelial Cell Survival, Proliferation, and Matrix Metalloproteinases Production and Regulated Angiogenesis. J. Immunol. 2003, 170, 3233–3242. [Google Scholar] [CrossRef]
- Russo, R.C.; Garcia, C.C.; Teixeira, M.M.; Amaral, F.A. The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases. Expert Rev. Clin. Immunol. 2014, 10, 593–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skov, L.; Beurskens, F.J.; Zachariae, C.O.C.; Reitamo, S.; Teeling, J.; Satijn, D.; Knudsen, K.M.; Boot, E.P.J.; Hudson, D.; Baadsgaard, O.; et al. IL-8 as Antibody Therapeutic Target in Inflammatory Diseases: Reduction of Clinical Activity in Palmoplantar Pustulosis. J. Immunol. 2008, 181, 669–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gonzalez-Aparicio, M.; Alfaro, C. Significance of the IL-8 pathway for immunotherapy. Hum. Vaccin. Immunother. 2019, 16, 2312–2317. [Google Scholar] [CrossRef] [PubMed]
- Waugh, D.J.J.; Wilson, C. The interleukin-8 pathway in cancer. Clin. Cancer Res. 2008, 14, 6735–6741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, K. Interleukin-8 and human cancer biology. Cytokine Growth Factor Rev. 2001, 12, 375–391. [Google Scholar] [CrossRef]
- Emerging Roles of IL-8 in Cancer Etiology. Available online: kingfisherbiotech.com (accessed on 27 October 2021).
- Ha, H.; Debnath, B.; Neamati, N. Role of the CXCL8-CXCR1/2 axis in cancer and inflammatory diseases. Theranostics 2017, 7, 1543–1588. [Google Scholar] [CrossRef]
- Li, L.; Li, J.; Gao, M.; Fan, H.; Wang, Y.; Xu, X.; Chen, C.; Liu, J.; Kim, J.; Aliyari, R.; et al. Interleukin-8 as a Biomarker for Disease Prognosis of Coronavirus Disease-2019 Patients. Front. Immunol. 2021, 11, 602395. [Google Scholar] [CrossRef]
- Bar-Eli, M. Role of interleukin-8 in tumor growth and metastasis of human melanoma. Cancer Metastasis Rev. 1999, 18, 377–385. [Google Scholar] [CrossRef] [PubMed]
- Anghel, A.E.; Ene, C.D.; Nicolae, I.; Budu, V.A.; Constantin, C.; Neagu, M. Interleukin 8—Major player in cutaneous melanoma metastasic process. Rom. Biotechnol. Lett. 2015, 20, 10911–10920. [Google Scholar]
- Singh, R.K.; Varney, M.L. IL-8 expression in malignant melanoma: Implications in growth and metastasis. Histol. Histopathol. 2000, 15, 843–849. [Google Scholar] [CrossRef]
- Whiteman, D.C.; Green, A.C.; Olsen, C.M. The Growing Burden of Invasive Melanoma: Projections of Incidence Rates and Numbers of New Cases in Six Susceptible Populations through 2031. J. Investig. Dermatol. 2016, 136, 1161–1171. [Google Scholar] [CrossRef] [Green Version]
- Elder, D.E. Melanoma progression. Pathology 2016, 48, 147–154. [Google Scholar] [CrossRef]
- Curtin, J.A.; Fridlyand, J.; Kageshita, T.; Patel, H.N.; Busam, K.J.; Kutzner, H.; Cho, K.-H.; Aiba, S.; Bröcker, E.-B.; LeBoit, P.E.; et al. Distinct Sets of Genetic Alterations in Melanoma. N. Engl. J. Med. 2005, 353, 2135–2147. [Google Scholar] [CrossRef]
- Hodis, E.; Watson, I.R.; Kryukov, G.V.; Arold, S.T.; Imielinski, M.; Theurillat, J.-P.; Nickerson, E.; Auclair, D.; Li, L.; Place, C.; et al. A landscape of driver mutations in melanoma. Cell 2012, 150, 251–263. [Google Scholar] [CrossRef] [Green Version]
- Braeuer, R.R.; Watson, I.R.; Wu, C.-J.; Mobley, A.K.; Kamiya, T.; Shoshan, E.; Bar-Eli, M. Why is melanoma so metastatic? Pigment. Cell Melanoma Res. 2014, 27, 19–36. [Google Scholar] [CrossRef]
- Marzagalli, M.; Ebelt, N.D.; Manuel, E.R. Unraveling the crosstalk between melanoma and immune cells in the tumor microenvironment. Semin. Cancer Biol. 2019, 59, 236–250. [Google Scholar] [CrossRef] [PubMed]
- Adams, R.; Moser, B.; Karagiannis, S.N.; Lacy, K.E. Chemokine pathways in cutaneous melanoma: Their modulation by cancer and exploitation by the clinician. Cancers 2021, 13, 5625. [Google Scholar] [CrossRef] [PubMed]
- Lippitz, B.E. Cytokine patterns in patients with cancer: A systematic review. Lancet Oncol. 2013, 14, e218–e228. [Google Scholar] [CrossRef]
- Neagu, M.; Constantin, C.; Longo, C. Chemokines in the melanoma metastasis biomarkers portrait. J. Immunoass. Immunochem. 2015, 36, 559–566. [Google Scholar] [CrossRef] [PubMed]
- Jacquelot, N.; Duong, C.P.M.; Belz, G.T.; Zitvogel, L. Targeting Chemokines and Chemokine Receptors in Melanoma and Other Cancers. Front. Immunol. 2018, 9, 2480. [Google Scholar] [CrossRef] [PubMed]
- Amersi, F.F.; Terando, A.M.; Goto, Y.; Scolyer, R.A.; Thompson, J.F.; Tran, A.N.; Faries, M.B.; Morton, D.L.; Hoon, D.S.B. Activation of CCR9/CCL25 in cutaneous melanoma mediates preferential metastasis to the small intestine. Clin. Cancer Res. 2008, 14, 638–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murakami, T.; Maki, W.; Cardones, A.R.; Fang, H.; Kyi, A.T.; Nestle, F.O.; Hwang, S.T. Expression of CXC chemokine receptor-4 enhances the pulmonary metastatic potential of murine B16 melanoma cells. Cancer Res. 2002, 62, 7328–7334. [Google Scholar]
- Mendt, M.; Cardier, J.E. Activation of the CXCR4 chemokine receptor enhances biological functions associated with B16 melanoma liver metastasis. Melanoma Res. 2017, 27, 300–307. [Google Scholar] [CrossRef]
- Klein, A.; Sagi-Assif, O.; Meshel, T.; Telerman, A.; Izraely, S.; Ben-Menachem, S.; Bayry, J.; Marzese, D.M.; Ohe, S.; Hoon, D.S.B.; et al. CCR4 is a determinant of melanoma brain metastasis. Oncotarget 2017, 8, 31079–31091. [Google Scholar] [CrossRef] [Green Version]
- Kawada, K.; Sonoshita, M.; Sakashita, H.; Takabayashi, A.; Yamaoka, Y.; Manabe, T.; Inaba, K.; Minato, N.; Oshima, M.; Taketo, M.M. Pivotal role of CXCR3 in melanoma cell metastasis to lymph nodes. Cancer Res. 2004, 64, 4010–4017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacquelot, N.; Enot, D.P.; Flament, C.; Vimond, N.; Blattner, C.; Pitt, J.M.; Yamazaki, T.; Roberti, M.P.; Daillère, R.; Vétizou, M.; et al. Chemokine receptor patterns in lymphocytes mirror metastatic spreading in melanoma. J. Clin. Investig. 2016, 126, 921–937. [Google Scholar] [CrossRef]
- Mullins, I.M.; Slingluff, C.L.; Lee, J.K.; Garbee, C.F.; Shu, J.; Anderson, S.G.; Mayer, M.E.; Knaus, W.A.; Mullins, D.W. CXC chemokine receptor 3 expression by activated CD8+ T cells is associated with survival in melanoma patients with stage III disease. Cancer Res. 2004, 64, 7697–7701. [Google Scholar] [CrossRef] [Green Version]
- Mikucki, M.E.; Fisher, D.T.; Matsuzaki, J.; Skitzki, J.J.; Galan, N.B.; Muhitch, J.B.; Ku, A.W.; Frelinger, J.G.; Odunsi, K.; Gajewski, T.F.; et al. Non-redundant requirement for CXCR3 signalling during tumoricidal T-cell trafficking across tumour vascular checkpoints. Nat. Commun. 2015, 6, 7458. [Google Scholar] [CrossRef]
- Merelli, B.; Massi, D.; Cattaneo, L.; Mandalà, M. Targeting the PD1/PD-L1 axis in melanoma: Biological rationale, clinical challenges and opportunities. Crit. Rev. Oncol. Hematol. 2014, 89, 140–165. [Google Scholar] [CrossRef] [Green Version]
- Koopmans, I.; Hendriks, M.A.J.M.; van Ginkel, R.J.; Samplonius, D.F.; Bremer, E.; Helfrich, W. Bispecific Antibody Approach for Improved Melanoma-Selective PD-L1 Immune Checkpoint Blockade. J. Invest. Dermatol. 2019, 139, 2343–2351. [Google Scholar] [CrossRef]
- Szegezdi, E.; Leverkus, M. Guiding the Killer and Bringing in Accomplices: Bispecific Antibody Treatment for Malignant Melanoma. J. Investig. Dermatol. 2016, 136, 362–364. [Google Scholar] [CrossRef] [Green Version]
- Sautès-Fridman, C.; Lawand, M.; Giraldo, N.A.; Kaplon, H.; Germain, C.; Fridman, W.H.; Dieu-Nosjean, M.-C. Tertiary lymphoid structures in cancers: Prognostic value, regulation, and manipulation for therapeutic intervention. Front. Immunol. 2016, 7, 407. [Google Scholar] [CrossRef] [Green Version]
- Yoshimura, T.; Matsushima, K.; Tanaka, S.; Robinson, E.A.; Appella, E.; Oppenheim, J.J.; Leonard, E.J. Purification of a human monocyte-derived neutrophil chemotactic factor that has peptide sequence similarity to other host defense cytokines. Proc. Natl. Acad. Sci. USA 1987, 84, 9233–9237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holmes, W.E.; Lee, J.; Kuang, W.J.; Rice, G.C.; Wood, W.I. Structure and functional expression of a human interleukin-8 receptor. Science 1991, 253, 1278–1280. [Google Scholar] [CrossRef]
- Liu, Q.; Li, A.; Tian, Y.; Wu, J.D.; Liu, Y.; Li, T.; Chen, Y.; Han, X.; Wu, K. The CXCL8-CXCR1/2 pathways in cancer. Cytokine Growth Factor Rev. 2016, 31, 61–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Varney, M.L.; Johansson, S.L.; Singh, R.K. Distinct expression of CXCL8 and its receptors CXCR1 and CXCR2 and their association with vessel density and aggressiveness in malignant melanoma. Am. J. Clin. Pathol. 2006, 125, 209–216. [Google Scholar] [CrossRef] [PubMed]
- Georganas, C.; Liu, H.; Perlman, H.; Hoffmann, A.; Thimmapaya, B.; Pope, R.M. Regulation of IL-6 and IL-8 Expression in Rheumatoid Arthritis Synovial Fibroblasts: The Dominant Role for NF-κB But Not C/EBPβ or c-Jun. J. Immunol. 2000, 165, 7199–7206. [Google Scholar] [CrossRef] [Green Version]
- Govindaraju, V.; Michoud, M.C.; Al-Chalabi, M.; Ferraro, P.; Powell, W.S.; Martin, J.G. Interleukin-8: Novel roles in human airway smooth muscle cell contraction and migration. Am. J. Physiol. Cell Physiol. 2006, 291, C957–C965. [Google Scholar] [CrossRef] [Green Version]
- Kroeze, K.L.; Boink, M.A.; Sampat-Sardjoepersad, S.C.; Waaijman, T.; Scheper, R.J.; Gibbs, S. Autocrine regulation of re-epithelialization after wounding by chemokine receptors CCR1, CCR10, CXCR1, CXCR2, and CXCR3. J. Investig. Dermatol. 2012, 132, 216–225. [Google Scholar] [CrossRef] [Green Version]
- Goczalik, I.; Ulbricht, E.; Hollborn, M.; Raap, M.; Uhlmann, S.; Weick, M.; Pannicke, T.; Wiedemann, P.; Bringmann, A.; Reichenbach, A.; et al. Expression of CXCL8, CXCR1, and CXCR2 in neurons and glial cells of the human and rabbit retina. Investig. Ophthalmol. Vis. Sci. 2008, 49, 4578–4589. [Google Scholar] [CrossRef] [Green Version]
- Nojima, H.; Konishi, T.; Freeman, C.M.; Schuster, R.M.; Japtok, L.; Kleuser, B.; Edwards, M.J.; Gulbins, E.; Lentsch, A.B. Chemokine receptors, CXCR1 and CXCR2, differentially regulate exosome release in hepatocytes. PLoS ONE 2016, 11, e0161443. [Google Scholar] [CrossRef] [PubMed]
- Li, A.; Dubey, S.; Varney, M.L.; Singh, R.K. Interleukin-8-induced proliferation, survival, and MMP production in CXCR1 and CXCR2 expressing human umbilical vein endothelial cells. Microvasc. Res. 2002, 64, 476–481. [Google Scholar] [CrossRef] [PubMed]
- Farkas, L.; Hahn, M.-C.; Schmoczer, M.; Jentsch, N.; Krätzel, K.; Pfeifer, M.; Schulz, C. Expression of CXC chemokine receptors 1 and 2 in human bronchial epithelial cells. Chest 2005, 128, 3724–3734. [Google Scholar] [CrossRef] [PubMed]
- Susek, K.H.; Karvouni, M.; Alici, E.; Lundqvist, A. The role of CXC chemokine receptors 1–4 on immune cells in the tumor microenvironment. Front. Immunol. 2018, 9, 2159. [Google Scholar] [CrossRef] [PubMed]
- Bosch, I.; Xhaja, K.; Estevez, L.; Raines, G.; Melichar, H.; Warke, R.V.; Fournier, M.V.; Ennis, F.A.; Rothman, A.L. Increased Production of Interleukin-8 in Primary Human Monocytes and in Human Epithelial and Endothelial Cell Lines after Dengue Virus Challenge. J. Virol. 2002, 76, 5588–5597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Geissmann, F.; Jung, S.; Littman, D.R. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 2003, 19, 71–82. [Google Scholar] [CrossRef] [Green Version]
- Jin, F.; Miao, Y.; Xu, P.; Qiu, X. IL-8 regulates the stemness properties of cancer stem cells in the small-cell lung cancer cell line H446. OncoTargets Ther. 2018, 11, 5723–5731. [Google Scholar] [CrossRef] [Green Version]
- Zheng, T.; Ma, G.; Tang, M.; Li, Z.; Xu, R. IL-8 secreted from M2 macrophages promoted prostate tumorigenesis via STAT3/MALAT1 pathway. Int. J. Mol. Sci. 2019, 20, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gasch, M.; Goroll, T.; Bauer, M.; Hinz, D.; Schütze, N.; Polte, T.; Kesper, D.; Simon, J.C.; Hackermüller, J.; Lehmann, I.; et al. Generation of IL-8 and IL-9 Producing CD4+ T cells is affected by Th17 polarizing conditions and AHR ligands. Mediat. Inflamm. 2014, 2014, 182549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, R.; Bao, C.; Huang, H.; Lin, F.; Yuan, Y.; Wang, S.; Jin, L.; Yang, T.; Shi, M.; Zhang, Z.; et al. Low expression of CXCR1/2 on neutrophils predicts poor survival in patients with hepatitis B virus-related acute-on-chronic liver failure. Sci. Rep. 2016, 6, 38714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lippert, U.; Zachmann, K.; Henz, B.M.; Neumann, C. Human T lymphocytes and mast cells differentially express and regulate extra- and intracellular CXCR1 and CXCR2. Exp. Dermatol. 2004, 13, 520–525. [Google Scholar] [CrossRef]
- Morohashi, H.; Miyawaki, T.; Nomura, H.; Kuno, K.; Murakami, S.; Matsushima, K.; Mukaida, N. Expression of both types of human interleukin-8 receptors on mature neutrophils, monocytes, and natural killer cells. J. Leukoc. Biol. 1995, 57, 180–187. [Google Scholar] [CrossRef]
- Li, B.H.; Garstka, M.A.; Li, Z.F. Chemokines and their receptors promoting the recruitment of myeloid-derived suppressor cells into the tumor. Mol. Immunol. 2020, 117, 201–215. [Google Scholar] [CrossRef]
- Himmel, M.E.; Crome, S.Q.; Ivison, S.; Piccirillo, C.; Steiner, T.S.; Levings, M.K. Human CD4+FOXP3+ regulatory T cells produce CXCL8 and recruit neutrophils. Eur. J. Immunol. 2011, 41, 306–312. [Google Scholar] [CrossRef]
- Babashah, S. Cancer Stem Cells: Emerging Concepts and Future Perspectives in Translational Oncology; Springer: Cham, Switzerland, 2015. [Google Scholar] [CrossRef]
- Rossi, O.; Karczewski, J.; Stolte, E.H.; Brummer, R.J.M.; van Nieuwenhoven, M.A.; Meijerink, M.; van Neerven, J.R.J.; van Ijzendoorn, S.C.D.; van Baarlen, P.; Wells, J.M. Vectorial secretion of interleukin-8 mediates autocrine signalling in intestinal epithelial cells via apically located CXCR1. BMC Res. Notes 2013, 6, 6431–6438. [Google Scholar] [CrossRef] [Green Version]
- Larsen, C.G.; Anderson, A.O.; Oppenheim, J.J.; Matsushima, K. Production of interleukin-8 by human dermal fibroblasts and keratinocytes in response to interleukin-1 or tumour necrosis factor. Immunology 1989, 68, 31–36. [Google Scholar]
- Zhai, J.; Shen, J.; Xie, G.; Wu, J.; He, M.; Gao, L.; Zhang, Y.; Yao, X.; Shen, L. Cancer-associated fibroblasts-derived IL-8 mediates resistance to cisplatin in human gastric cancer. Cancer Lett. 2019, 454, 37–43. [Google Scholar] [CrossRef]
- Nakamura, Y.; Kano, R.; Hasegawa, A.; Watanabe, S. Interleukin-8 and tumor necrosis factor alpha production in human epidermal keratinocytes induced by Trichophyton mentagrophytes. Clin. Diagn. Lab. Immunol. 2002, 9, 935–937. [Google Scholar] [CrossRef] [Green Version]
- Jiang, W.G.; Sanders, A.J.; Ruge, F.; Harding, K.G. Influence of interleukin-8 (IL-8) and IL-8 receptors on the migration of human keratinocytes, the role of plc-γ and potential clinical implications. Exp. Ther. Med. 2012, 3, 231–236. [Google Scholar] [CrossRef] [Green Version]
- Zimmermann, H.W.; Seidler, S.; Gassler, N.; Nattermann, J.; Luedde, T.; Trautwein, C.; Tacke, F. Interleukin-8 is activated in patients with chronic liver diseases and associated with hepatic macrophage accumulation in human liver fibrosis. PLoS ONE 2011, 6, e21381. [Google Scholar] [CrossRef]
- Yang, W.; Wang, D.; Richmond, A. Role of clathrin-mediated endocytosis in CXCR2 sequestration, resensitization, and signal transduction. J. Biol. Chem. 1999, 274, 11328–11333. [Google Scholar] [CrossRef] [Green Version]
- Brat, D.J.; Bellail, A.C.; Van Meir, E.G. The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro. Oncol. 2005, 7, 122–133. [Google Scholar] [CrossRef]
- Weathington, N.M.; Van Houwelingen, A.H.; Noerager, B.D.; Jackson, P.L.; Kraneveld, A.D.; Galin, F.S.; Folkerts, G.; Nijkamp, F.P.; Blalock, J.E. A novel peptide CXCR ligand derived from extracellular matrix degradation during airway inflammation. Nat. Med. 2006, 12, 317–323. [Google Scholar] [CrossRef] [PubMed]
- Bernhagen, J.; Krohn, R.; Lue, H.; Gregory, J.L.; Zernecke, A.; Koenen, R.R.; Dewor, M.; Georgiev, I.; Schober, A.; Leng, L.; et al. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat. Med. 2007, 13, 587–596. [Google Scholar] [CrossRef]
- Roebuck, K.A. Regulation of Interleukin-8 Gene Expression. J. Interferon Cytokine Res. 1999, 438, 429–438. [Google Scholar] [CrossRef] [PubMed]
- Shoshan, E.; Braeuer, R.R.; Kamiya, T.; Mobley, A.K.; Huang, L.; Vasquez, M.E.; Velazquez-Torres, G.; Chakravarti, N.; Ivan, C.; Prieto, V.; et al. NFAT1 directly regulates IL8 and MMP3 to promote melanoma tumor growth and metastasis. Cancer Res. 2016, 76, 3145–3155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, S.; DeGuzman, A.; Bucana, C.D.; Fidler, I.J. Nuclear factor-κB activity correlates with growth, angiogenesis, and metastasis of human melanoma cells in nude mice. Clin. Cancer Res. 2000, 6, 2573–2581. [Google Scholar] [PubMed]
- Giles, K.M.; Brown, R.A.M.; Ganda, C.; Podgorny, M.J.; Candy, P.A.; Wintle, L.C.; Richardson, K.L.; Kalinowski, F.C.; Stuart, L.M.; Epis, M.R.; et al. MicroRNA-7-5p inhibits melanoma cell proliferation and metastasis by suppressing RelA/NF-κB. Oncotarget 2016, 7, 31663–31680. [Google Scholar] [CrossRef] [Green Version]
- Sinars, C.R.; Cheung-Flynn, J.; Rimerman, R.A.; Scammell, J.G.; Smith, D.F.; Clardy, J. Structure of the large FK506-binding protein FKBP51, an Hsp90-binding protein and a component of steroid receptor complexes. Proc. Natl. Acad. Sci. USA 2003, 100, 868–873. [Google Scholar] [CrossRef] [Green Version]
- Wilson, B.J.; Saab, K.R.; Ma, J.; Schatton, T.; Pütz, P.; Zhan, Q.; Murphy, G.F.; Gasser, M.; Waaga-Gasser, A.M.; Frank, N.Y.; et al. ABCB5 maintains melanoma-initiating cells through a proinflammatory cytokine signaling circuit. Cancer Res. 2014, 74, 4196–4207. [Google Scholar] [CrossRef] [Green Version]
- Wu, G.D.; Lai, E.J.; Huang, N.; Wen, X. Oct-1 and CCAAT/enhancer-binding protein (C/EBP) bind to overlapping elements within the interleukin-8 promoter. The role of oct-1 as a transcriptional repressor. J. Biol. Chem. 1997, 272, 2396–2403. [Google Scholar] [CrossRef] [Green Version]
- Huang, S.M.; McCance, D.J. Down Regulation of the Interleukin-8 Promoter by Human Papillomavirus Type 16 E6 and E7 through Effects on CREB Binding Protein/p300 and P/CAF. J. Virol. 2002, 76, 8710–8721. [Google Scholar] [CrossRef] [Green Version]
- Jenkins, M.J.; Brinckerhoff, C.E.; Mullins, D.W. CXCR3 Signaling in BRAFWT melanoma increases IL-8 expression and tumorigenicity. PLoS ONE 2015, 10, e0121140. [Google Scholar] [CrossRef] [Green Version]
- Timani, K.A.; Gyorffy, B.; Liu, Y.; Mohammad, S.K.; He, J.J. Tip110/SART3 regulates IL-8 expression and predicts the clinical outcomes in melanoma. Mol. Cancer 2018, 17, 8–13. [Google Scholar] [CrossRef] [PubMed]
- Ostrand-Rosenberg, S. Immune surveillance: A balance between protumor and antitumor immunity. Curr. Opin. Genet. Dev. 2008, 18, 11–18. [Google Scholar] [CrossRef] [Green Version]
- Miniati, A.; Weng, Z.; Zhang, B.; Therianou, A.; Vasiadi, M.; Nicolaidou, E.; Stratigos, A.J.; Antoniou, C.; Theoharides, T.C. Stimulated human melanocytes express and release interleukin-8, which is inhibited by luteolin: Relevance to early vitiligo. Clin. Exp. Dermatol. 2014, 39, 54–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, R.K.; Varney, M.L.; Bucana, C.D.; Johansson, S.L. Expression of interleukin-8 in primary and metastatic malignant melanoma of the skin. Melanoma Res. 1999, 9, 383–387. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.K.; Gutman, M.; Reich, R.; Bar-Eli, M. Ultraviolet B Irradiation Promotes Tumorigenic and Metastatic Properties in Primary Cutaneous Melanoma via Induction of Interleukin 8. Cancer Res. 1995, 55, 3669–3674. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, N.; Couts, K.L.; Luo, Y.; Fujita, M. Understanding melanoma stem cells. Melanoma Manag. 2015, 2, 179–188. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Yang, X.; Wang, L.; Zhang, C. Interplay between inflammatory tumor microenvironment and cancer stem cells (Review). Oncol. Lett. 2018, 16, 679–686. [Google Scholar] [CrossRef] [PubMed]
- Rohaan, M.W.; van den Berg, J.H.; Kvistborg, P.; Haanen, J.B.A.G. Adoptive transfer of tumor-infiltrating lymphocytes in melanoma: A viable treatment option. J. Immunother. Cancer 2018, 6, 102. [Google Scholar] [CrossRef] [PubMed]
- David, J.M.; Dominguez, C.; Hamilton, D.H.; Palena, C. The IL-8/IL-8R axis: A double agent in tumor immune resistance. Vaccines 2016, 4, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, H.H.; Liang, S.; Henderson, A.J.; Dong, C. Regulation of interleukin-8 expression in melanoma-stimulated neutrophil inflammatory response. Exp. Cell Res. 2007, 313, 551–559. [Google Scholar] [CrossRef] [Green Version]
- Jaillon, S.; Ponzetta, A.; Di Mitri, D.; Santoni, A.; Bonecchi, R.; Mantovani, A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat. Rev. Cancer 2020, 20, 485–503. [Google Scholar] [CrossRef] [PubMed]
- Schedel, F.; Mayer-Hain, S.; Pappelbaum, K.I.; Metze, D.; Stock, M.; Goerge, T.; Loser, K.; Sunderkötter, C.; Luger, T.A.; Weishaupt, C. Evidence and impact of neutrophil extracellular traps in malignant melanoma. Pigment Cell Melanoma Res. 2020, 33, 63–73. [Google Scholar] [CrossRef] [Green Version]
- Hussein, M.R. Tumour-associated macrophages and melanoma tumourigenesis: Integrating the complexity. Int. J. Exp. Pathol. 2006, 87, 163–176. [Google Scholar] [CrossRef]
- Koch, A.E.; Polverini, P.J.; Kunkel, S.L.; Harlow, L.A.; DiPietro, L.A.; Elner, V.M.; Elner, S.G.; Strieter, R.M. Interleukin-8 as a macrophage-derived mediator of angiogenesis. Science 1992, 258, 1798–1801. [Google Scholar] [CrossRef]
- Xu, H.; Lai, W.; Zhang, Y.; Liu, L.; Luo, X.; Zeng, Y.; Wu, H.; Lan, Q.; Chu, Z. Tumor-associated macrophage-derived IL-6 and IL-8 enhance invasive activity of LoVo cells induced by PRL-3 in a KCNN4 channel-dependent manner. BMC Cancer 2014, 14, 330. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, M.K.; Sinha, P.; Clements, V.K.; Rodriguez, P.; Ostrand-Rosenberg, S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010, 70, 68–77. [Google Scholar] [CrossRef] [Green Version]
- Pan, P.Y.; Ma, G.; Weber, K.J.; Ozao-Choy, J.; Wang, G.; Yin, B.; Divino, C.M.; Chen, S.H. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010, 70, 99–108. [Google Scholar] [CrossRef] [Green Version]
- Tobin, R.P.; Jordan, K.R.; Kapoor, P.; Spongberg, E.; Davis, D.; Vorwald, V.M.; Couts, K.L.; Gao, D.; Smith, D.E.; Borgers, J.S.W.; et al. IL-6 and IL-8 Are Linked with Myeloid-Derived Suppressor Cell Accumulation and Correlate with Poor Clinical Outcomes in Melanoma Patients. Front. Oncol. 2019, 9, 1223. [Google Scholar] [CrossRef] [Green Version]
- Shurin, M.R.; Naiditch, H.; Zhong, H.; Shurin, G.V. Regulatory dendritic cells: New targets for cancer immunotherapy. Cancer Biol. Ther. 2011, 11, 988–992. [Google Scholar] [CrossRef] [Green Version]
- Tucci, M.; Passarelli, A.; Mannavola, F.; Felici, C.; Stucci, L.S.; Cives, M.; Silvestris, F.M. Immune System Evasion as Hallmark of Melanoma Progression: The Role of Dendritic Cells. Front. Oncol. 2019, 9, 1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alfaro, C.; Suárez, N.; Martínez-Forero, I.; Palazón, A.; Rouzaut, A.; Solano, S.; Feijoo, E.; Gúrpide, A.; Bolanús, E.; Erro, L.; et al. Carcinoma-derived Interleukin-8 disorients dendritic cell migration without impairing T-cell stimulation. PLoS ONE 2011, 6, e17922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kondo, S.; Yoneta, A.; Yazawa, H.; Kamada, A.; Jimbow, K. Downregulation of CXCR-2 but not CXCR-1 expression by human keratinocytes by UVB. J. Cell. Physiol. 2000, 182, 366–370. [Google Scholar] [CrossRef]
- Jobe, N.P.; Rösel, D.; Dvořánková, B.; Kodet, O.; Lacina, L.; Mateu, R.; Smetana, K.; Brábek, J. Simultaneous blocking of IL-6 and IL-8 is sufficient to fully inhibit CAF-induced human melanoma cell invasiveness. Histochem. Cell Biol. 2016, 146, 205–217. [Google Scholar] [CrossRef]
- Li, A.; Varney, M.L.; Valasek, J.; Godfrey, M.; Dave, B.J.; Singh, R.K. Autocrine role of interleukin-8 in induction of endothelial cell proliferation, survival, migration and MMP-2 production and angiogenesis. Angiogenesis 2005, 8, 63–71. [Google Scholar] [CrossRef]
- Ramjeesingh, R.; Leung, R.; Siu, C.H. Interleukin-8 secreted by endothelial cells induces chemotaxis of melanoma cells through the chemokine receptor CXCR1. FASEB J. 2003, 17, 1292–1294. [Google Scholar] [CrossRef]
- Griffith, J.W.; Sokol, C.L.; Luster, A.D. Chemokines and chemokine receptors: Positioning cells for host defense and immunity. Annu. Rev. Immunol. 2014, 32, 659–702. [Google Scholar] [CrossRef] [Green Version]
- Nestle, F.O.; Di Meglio, P.; Qin, J.Z.; Nickoloff, B.J. Skin immune sentinels in health and disease. Nat. Rev. Immunol. 2009, 9, 679–691. [Google Scholar] [CrossRef] [Green Version]
- Fernando, R.I.; Castillo, M.D.; Litzinger, M.; Hamilton, D.H.; Palena, C. IL-8 signaling plays a critical role in the epithelial-mesenchymal transition of human carcinoma cells. Cancer Res. 2011, 71, 5296–5306. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Z.; Wang, S.; Lin, Y.; Miao, Y.; Zeng, Y.; Nie, Y.; Guo, P.; Jiang, G.; Wu, J. Epithelial-mesenchymal transition in cancer: Role of the IL-8/IL-8R axis. Oncol. Lett. 2017, 13, 4577–4584. [Google Scholar] [CrossRef] [Green Version]
- Gabellini, C.; Trisciuoglio, D.; Desideri, M.; Candiloro, A.; Ragazzoni, Y.; Orlandi, A.; Zupi, G.; Del Bufalo, D. Functional activity of CXCL8 receptors, CXCR1 and CXCR2, on human malignant melanoma progression. Eur. J. Cancer 2009, 45, 2618–2627. [Google Scholar] [CrossRef]
- Wu, S.; Singh, S.; Varney, M.L.; Kindle, S.; Singh, R.K. Modulation of CXCL-8 expression in human melanoma cells regulates tumor growth, angiogenesis, invasion, and metastasis. Cancer Med. 2012, 1, 306–317. [Google Scholar] [CrossRef]
- Singh, S.; Sadanandam, A.; Varney, M.L.; Nannuru, K.C.; Singh, R.K. Small interfering RNA-mediated CXCR1 or CXCR2 knock-down inhibits melanoma tumor growth and invasion. Int. J. Cancer 2010, 126, 328–336. [Google Scholar] [CrossRef]
- Varney, M.L.; Li, A.; Dave, B.J.; Bucana, C.D.; Johansson, S.L.; Singh, R.K. Expression of CXCR1 and CXCR2 receptors in malignant melanoma with different metastatic potential and their role in interleukin-8 (CXCL-8) -mediated modulation of metastatic phenotype. Clin. Exp. Metastasis 2003, 20, 723–731. [Google Scholar] [CrossRef]
- Singh, S.; Nannuru, K.C.; Sadanandam, A.; Varney, M.L.; Singh, R.K. CXCR1 and CXCR2 enhances human melanoma tumourigenesis, growth and invasion. Br. J. Cancer. 2009, 100, 1638–1646. [Google Scholar] [CrossRef] [Green Version]
- Huang, S.; Mills, L.; Mian, B.; Tellez, C.; McCarty, M.; Yang, X.D.; Gudas, J.M.; Bar-Eli, M. Fully humanized neutralizing antibodies to interleukin-8 (ABX-IL8) inhibit angiogenesis, tumor growth, and metastasis of human melanoma. Am. J. Pathol. 2002, 161, 125–134. [Google Scholar] [CrossRef] [Green Version]
- Singh, R.K.; Gutman, M.; Radinsky, R.; Bucana, C.D.; Fidler, I.J. Expression of Interleukin 8 Correlates with the Metastatic Potential of Human Melanoma Cells in Nude Mice. Cancer Res. 1994, 54, 3242–3247. [Google Scholar] [PubMed]
- Singh, S.; Varney, M.; Singh, R.K. Host CXCR2-dependent regulation of melanoma growth, angiogenesis, and experimental lung metastasis. Cancer Res. 2009, 69, 411–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Savoia, P.; Fava, P.; Casoni, F.; Cremona, O. Targeting the ERK signaling pathway in melanoma. Int. J. Mol. Sci. 2019, 20, 1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Venkatakrishnan, G.; Salgia, R.; Groopman, J.E. Chemokine Receptors CXCR-1/2 Activate Mitogen-activated Protein Kinase via the Epidermal Growth Factor Receptor in Ovarian Cancer Cells. JBC 2000, 275, 6868–6875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luppi, F.; Longo, A.M.; de Boer, W.I.; Rabe, K.F.; Hiemstra, P.S. Interleukin-8 stimulates cell proliferation in non-small cell lung cancer through epidermal growth factor receptor transactivation. Lung Cancer 2007, 56, 25–33. [Google Scholar] [CrossRef]
- Chan, L.-P.; Liu, C.; Chiang, F.-Y.; Wang, L.-F.; Lee, K.-W.; Chen, W.-T.; Kuo, P.-L.; Liang, C.-H. IL-8 promotes inflammatory mediators and stimulates activation of p38 MAPK/ERK-NF-κB pathway and reduction of JNK in HNSCC. Oncotarget 2017, 8, 56375–56388. [Google Scholar] [CrossRef] [Green Version]
- Martínez-Limón, A.; Joaquin, M.; Caballero, M.; Posas, F.; de Nadal, E. The p38 pathway: From biology to cancer therapy. Int. J. Mol. Sci. 2020, 21, 1913. [Google Scholar] [CrossRef] [Green Version]
- Desai, S.; Laskar, S.; Pandey, B.N. Autocrine IL-8 and VEGF mediate epithelial-mesenchymal transition and invasiveness via p38/JNK-ATF-2 signalling in A549 lung cancer cells. Cell. Signal. 2013, 25, 1780–1791. [Google Scholar] [CrossRef]
- Bogucka, K.; Pompaiah, M.; Marini, F.; Binder, H.; Harms, G.; Kaulich, M.; Klein, M.; Michel, C.; Radsak, M.P.; Rosigkeit, S.; et al. ERK3/MAPK6 controls IL-8 production and chemotaxis. eLife 2020, 9, e52511. [Google Scholar] [CrossRef]
- Vandamme, N.; Denecker, G.; Bruneel, K.; Blancke, G.; Akay, O.; Taminau, J.; De Coninck, J.; De Smedt, E.; Skrypek, N.; Loocke, W.V.; et al. The EMT transcription factor ZEB2 promotes proliferation of primary and metastatic melanoma while suppressing an invasive, mesenchymal-like phenotype. Cancer Res. 2020, 80, 2983–2995. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Shi, L.; Lu, S.; Sun, X.; Liu, Y.; Li, H.; Wang, X.; Gabb, C.; Zhang, G.; Wang, Y. Autocrine IL-8 promotes F-actin polymerization and mediate mesenchymal transition via ELMO1-NF-κB-Snail signaling in glioma. Cancer Biol. Ther. 2015, 16, 283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, H.; Zhang, Y.; Zhou, Z.; Guo, Y.; Huang, X.; Westover, K.D.; Zhng, Z.; Chen, B.; Hua, Y.; Li, S.; et al. Intergrated analysis of ELMO1, serves as a link between tumour mutation burden and epithelial-mesenchymal transition in hepatocellular carcinoma. EBioMedicine 2019, 46, 105–118. [Google Scholar] [CrossRef]
- Franke, T.F. PI3K/Akt: Getting it right matters. Oncogene 2008, 27, 6473–6488. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Tang, C.; Cao, H.; Li, K.; Pang, X.; Zhong, L.; Dang, W.; Tang, H.; Huang, Y.; Wei, L.; et al. Activation of IL-8 via PI3K/AKT-dependent pathway is involved in leptin-mediated epithelial-mesenchymal transition in human breast cancer cells. Cancer Biol. Ther. 2015, 16, 1220–1230. [Google Scholar] [CrossRef] [Green Version]
- Sun, F.; Wang, J.; Sun, Q.; Li, F.; Gao, H.; Xu, L.; Zhang, J.; Sun, X.; Tian, Y.; Zhao, Q.; et al. Interleukin-8 promotes integrin β3 upregulation and cell invasion through PI3K/Akt pathway in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2019, 38, 449. [Google Scholar] [CrossRef]
- Yan, W.; Han, P.; Zhou, Z.; Tu, W.; Liao, J.; Li, P.; Liu, M.; Tian, D.; Fu, Y. Netrin-1 induces epithelial-mesenchymal transition and promotes hepatocellular carcinoma invasiveness. Dig. Dis. Sci. 2014, 59, 1213–1221. [Google Scholar] [CrossRef]
- Zhou, N.; Lu, F.; Liu, C.; Xu, K.; Huang, J.; Yu, D.; Bi, L. IL-8 induces the epithelial-mesenchymal transition of renal cell carcinoma cells through the activation of AKT signaling. Oncol. Let. 2016, 12, 1915–1920. [Google Scholar] [CrossRef] [PubMed]
- MacManus, C.F.; Pettigrew, J.; Seaton, A.; Wilson, C.; Maxwell, P.J.; Berlingeri, S.; Purcell, C.; McGurk, M.; Johnston, P.G.; Waugh, D.J.J. Interleukin-8 signaling promotes translational regulation of cyclin D in androgen-independent prostate cancer cells. Mol. Cancer Res. 2007, 5, 737–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kovacs, D.; Migliano, E.; Muscardin, L.; Silipo, V.; Picardo, M.; Bellei, B. The role of WNTβ-catenin signaling pathway in melanoma. Oncotarget 2016, 7, 43295–43314. [Google Scholar] [CrossRef] [Green Version]
- Larue, L.; Delmas, V. The WNT/Beta-catenin pathway in melanoma. Front. Biosci. 2006, 11, 733–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.; Wang, Y.; Fan, C.; Xiao, X.; Zhang, Q.; Xu, T.; Jiang, C. Interleukin-8/β-catenin mediates epithelial—Mesenchymal transition in ameloblastoma. Oral Dis. 2019, 8, 1964–1971. [Google Scholar] [CrossRef] [PubMed]
- Wen, J.; Zhao, Z.; Huang, L.; Wang, L.; Miao, Y.; Wu, J. IL-8 promotes cell migration through regulating EMT by activating the Wnt/β-catenin pathway in ovarian cancer. J. Cell. Mol. Med. 2020, 24, 1588–1598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mills, C.N.; Nowsheen, S.; Bonner, A.J.; Yang, E.S. Emerging Roles of Glycogen Synthase Kinase 3 in the Treatment of Brain Tumors. Front. Mol. Neurosci. 2011, 4, 47. [Google Scholar] [CrossRef] [Green Version]
- Sun, Y.; Ai, J.Z.; Jin, X.; Liu, L.R.; Lin, T.H.; Xu, H.; Wei, Q.; Yang, L. IL-8 protects prostate cancer cells from GSK-3β-induced oxidative stress by activating the mTOR signaling pathway. Prostate 2019, 79, 1180–1190. [Google Scholar] [CrossRef] [PubMed]
- Poon, E.; Harris, A.L.; Ashcroft, M. Targeting the hypoxia-inducible factor (HIF) pathway in cancer. Expert Rev. Mol. Med. 2009, 11, E26. [Google Scholar] [CrossRef] [Green Version]
- Quaresmini, D.; Guida, M. Neoangiogenesis in Melanoma: An Issue in Biology and Systemic Treatment. Front. Immunol. 2020, 11, 584903. [Google Scholar] [CrossRef]
- Arany, Z.; Foo, S.; Ma, Y.; Ruas, J.L.; Bommi-Reddy, A.; Girnun, G.; Cooper, M.; Laznik, D.; Chinsomboon, J.; Rangwala, S.M.; et al. HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1α. Nature 2008, 451, 1008–1012. [Google Scholar] [CrossRef] [Green Version]
- Torisu, H.; Ono, M.; Kiryu, H.; Furue, M.; Ohmoto, Y.; Nakayama, J.; Nishioka, Y.; Sone, S.; Kuwano, M. Macrophage infiltration correlates with tumor stage and angiogenesis in human malignant melanoma: Possible involvement of TNFα and IL-1α. Int. J. Cancer 2000, 85, 182–188. [Google Scholar] [CrossRef]
- Martin, D.; Galisteo, R.; Gutkind, J.S. CXCL8/IL8 stimulates vascular endothelial growth factor (VEGF) expression and the autocrine activation of VEGFR2 in endothelial cells by activating NFκB through the CBM (Carma3/Bcl10/Malt1) complex. J. Biol. Chem. 2009, 284, 6038–6042. [Google Scholar] [CrossRef] [Green Version]
- Lesinski, G.B. The potential for targeting the STAT3 pathway as a novel therapy for melanoma. Futur. Oncol. 2013, 9, 925–927. [Google Scholar] [CrossRef] [Green Version]
- Swoboda, A.; Soukup, R.; Eckel, O.; Kinslechner, K.; Wingelhofer, B.; Schörghofer, D.; Sternberg, C.; Pham, T.T.; Vallianou, M.; Horvath, J.; et al. STAT3 promotes melanoma metastasis by CEBP-induced repression of the MITF pathway. Oncogene 2021, 40, 1091–1105. [Google Scholar] [CrossRef]
- Piperi, C.; Samaras, V.; Levidou, G.; Kavantzas, N.; Boviatsis, E.; Petraki, K.; Grivas, A.; Barbatis, K.; Varsos, V.; Patsouris, E.; et al. Prognostic significance of IL-8-STAT-3 pathway in astrocytomas: Correlation with IL-6, VEGF and microvessel morphometry. Cytokine 2011, 55, 387–395. [Google Scholar] [CrossRef] [PubMed]
- Xu, Q.; Ma, H.; Chang, H.; Feng, Z.; Zhang, C.; Yang, X. The interaction of interleukin-8 and PTEN inactivation promotes the malignant progression of head and neck squamous cell carcinoma via the STAT3 pathway. Cell Death Dis. 2020, 11, 405. [Google Scholar] [CrossRef]
- Nicholas, C.; Lesinki, G.B. The Jak-STAT Signal Transduction Pathway in Melanoma. Break. Melanoma Res. 2011, 2009, 283–306. [Google Scholar] [CrossRef] [Green Version]
- Wu, H.; Goel, V.; Haluska, F.G. PTEN signaling pathways in melanoma. Oncogene 2003, 22, 3113–3122. [Google Scholar] [CrossRef] [Green Version]
- Guo, Y.; Zang, Y.; Lv, L.; Cai, F.; Qian, T.; Zhang, G.; Feng, Q. IL-8 promotes proliferation and inhibition of apoptosis via STAT3/AKT/NF-κB pathway in prostate cancer. Mol. Med. Rep. 2017, 16, 9035–9042. [Google Scholar] [CrossRef] [Green Version]
- Britschgi, A.; Andraos, R.; Brinkhaus, H.; Klebba, I.; Romanet, V.; Müller, U.; Murakami, M.; Radimerski, T.; Bentires-Alj, M. JAK2/STAT5 Inhibition Circumvents Resistance to PI3K/mTOR Blockade: A Rationale for Cotargeting These Pathways in Metastatic Breast Cancer. Cancer Cell 2012, 22, 796–811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Repasky, E.A.; Eng, J.; Hylander, B.L. Stress, Metabolism and Cancer: Integrated Pathways Contributing to Immune Suppression. Cancer J. 2015, 21, 97–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shahzad, M.M.K.; Arevalo, J.M.; Armaiz-Pena, G.N.; Lu, C.; Stone, R.L.; Moreno-Smith, M.; Nishimura, M.; Lee, J.-W.; Jennings, N.B.; Basttsford-Miller, J.; et al. Stress effects on FosB- and interleukin-8 (IL8)-driven ovarian cancer growth and metastasis. J. Biol. Chem. 2010, 285, 35462–35470. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, S.K.; Bhardwaj, A.; Arora, S.; Tyagi, N.; Singh, A.P.; Carter, J.E.; Scammell, J.G.; Fodstad, Ø.; Singh, S. Interleukin-8 is a key mediator of FKBP51-induced melanoma growth, angiogenesis and metastasis. Br. J. Cancer 2015, 112, 1772–1781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hähle, A.; Merz, S.; Meyners, C.; Hausch, F. The many faces of FKBP51. Biomolecules 2019, 9, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Romano, S.; D’Angelillo, A.; Pacelli, R.; Staibano, S.; De Luna, E.; Bisogni, R.; Eskelinen, E.L.; Mascolo, M.; Calı, G.; Arra, C.; et al. Role of FK506-binding protein 51 in the control of apoptosis of irradiated melanoma cells. Cell Death Differ. 2010, 17, 145–157. [Google Scholar] [CrossRef] [PubMed]
- Romano, S.; Mallardo, M.; Romano, M.F. FKBP51 and the NF-κB regulatory pathway in cancer. Curr. Opin. Pharmacol. 2011, 11, 288–293. [Google Scholar] [CrossRef] [PubMed]
- Romano, S.; Staibano, S.; Greco, A.; Brunetti, A.; Nappo, G.; Ilardi, G.; Martinelli, R.; Sorrentino, A.; Di Pace, A.; Mascolo, M.; et al. FK506 binding protein 51 positively regulates melanoma stemness and metastatic potential. Cell Death Dis. 2013, 4, e578-10. [Google Scholar] [CrossRef] [Green Version]
- Popa, I.L.; Milac, A.L.; Sima, L.E.; Alexandru, P.R.; Pastrama, F.; Munteanu, C.V.A.; Negroiu, G. Cross-talk between dopachrome tautomerase and caveolin-1 is melanoma cell phenotype-specific and potentially involved in tumor progression. J. Biol. Chem. 2016, 291, 12481–12500. [Google Scholar] [CrossRef] [Green Version]
- Eddy, K.; Shah, R.; Chen, S. Decoding Melanoma Development and Progression: Identification of Therapeutic Vulnerabilities. Front. Oncol. 2021, 10, 626129. [Google Scholar] [CrossRef]
- Bartlett, E.K.; Karakousis, G.C. Current staging and prognostic factors in melanoma. Surg. Oncol. Clin. N. Am. 2015, 24, 215–227. [Google Scholar] [CrossRef]
- Kučera, J.; Strnadová, K.; Dvořánková, B.; Lacina, L.; Krajsová, I.; Štork, J.; Kovářová, H.; Skalníková, H.K.; Vodička, P.; Motlík, J.; et al. Serum proteomic analysis of melanoma patients with immunohistochemical profiling of primary melanomas and cultured cells: Pilot study. Oncol. Rep. 2019, 42, 1793–1804. [Google Scholar] [CrossRef]
- Yurkovetsky, Z.R.; Kirkwood, J.M.; Edington, H.D.; Marrangoni, A.M.; Velikokhatnaya, L.; Winans, M.T.; Gorelik, E.; Lokshin, A.E. Multiplex analysis of serum cytokines in melanoma patients treated with interferon-α2b. Clin. Cancer Res. 2007, 13, 2422–2428. [Google Scholar] [CrossRef] [Green Version]
- Li, X.-J.; Peng, L.-X.; Shao, J.-Y.; Lu, W.-H.; Zhang, J.-X.; Chen, S.; Chen, Z.-Y.; Xiang, Y.-Q.; Bao, Y.-N.; Zheng, F.-J.; et al. As an independent unfavorable prognostic factor, IL-8 promotes metastasis of nasopharyngeal carcinoma through induction of epithelial-mesenchymal transition and activation of AKT signaling. Carcinogenesis 2012, 33, 1302–1309. [Google Scholar] [CrossRef] [Green Version]
- Shahzad, A.; Knapp, M.; Lang, I.; Köhler, G. Interleukin 8 (IL-8)-a universal biomarker? Int. Arch. Med. 2010, 3, 2–5. [Google Scholar] [CrossRef] [Green Version]
- Ene, C.D.; Anghel, A.E.; Neagu, M.; Nicolae, I.C. 25-OH Vitamin D and Interleukin-8: Emerging Biomarkers in Cutaneous Melanoma Development and Progression. Mediat. Inflamm. 2015, 2015, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Ugurel, S.; Rappl, G.; Tilgen, W.; Reinhold, U. Increased Serum Concentration of Angiogenic Factors in Malignant Melanoma Patients Correlates with Tumor Progression and Survival. J. Clin.Oncol. 2001, 19, 577–583. [Google Scholar] [CrossRef]
- Schalper, K.A.; Carleton, M.; Zhou, M.; Chen, T.; Feng, Y.; Huang, S.-P.; Walsh, A.M.; Baxi, V.; Pandya, D.; Baradet, T.; et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat. Med. 2020, 26, 688–692. [Google Scholar] [CrossRef]
- Sanmamed, M.F.; Carranza-Rua, O.; Alfaro, C.; Oñate, C.; Martín-Algarra, S.; Perez, G.; Landazuri, S.F.; Gonzalez, A.; Gross, S.; Rodriguez, I.; et al. Serum interleukin-8 reflects tumor burden and treatment response across malignancies of multiple tissue origins. Clin. Cancer Res. 2014, 20, 5697–5707. [Google Scholar] [CrossRef] [Green Version]
- Kaczanowska, S.; Joseph, A.M.; Davila, E. TLR agonists: Our best frenemy in cancer immunotherapy. J. Leukoc. Biol. 2013, 93, 847–863. [Google Scholar] [CrossRef] [Green Version]
- Suarez-Kelly, L.P.; Levine, K.M.; Olencki, T.E.; Martin del Campo, S.E.; Streacker, E.A.; Brooks, T.R.; Karpa, V.I.; Markowitz, J.; Bingman, A.K.; Geyer, S.M.; et al. A pilot study of interferon-alpha-2b dose reduction in the adjuvant therapy of high-risk melanoma. Cancer Immunol. Immunother. 2019, 68, 619–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Trolio, R.; Simeone, E.; Di Lorenzo, G.; Grimaldi, A.M.; Romano, A.; Ayala, F.; Caracò, C.; Mozzillo, N.; Ascierto, P.A. Update on PEG-interferon α-2b as adjuvant therapy in melanoma. Anticancer Res. 2012, 32, 3901–3910. [Google Scholar] [PubMed]
- Davar, D.; Ding, F.; Saul, M.; Sander, C.; Tarhini, A.A.; Kirkwood, J.M.; Tawbi, H.A. High-dose interleukin-2 (HD IL-2) for advanced melanoma: A single center experience from the University of Pittsburgh Cancer Institute. J. Immunother. Cancer 2017, 5, 74. [Google Scholar] [CrossRef] [PubMed]
- Alfaro, C.; Sanmamed, M.F.; Rodríguez-Ruiz, M.E.; Teijeira, A.; Oñate, C.; González, A.; Ponz, M.; Schalper, K.A.; Pérez-Gracia, J.L.; Melero, I. Interleukin-8 in cancer pathogenesis, treatment and follow-up. Cancer Treat. Rev. 2017, 60, 24–31. [Google Scholar] [CrossRef]
- Zigler, M.; Villares, G.J.; Lev, D.C.; Melnikova, V.O.; Bar-Eli, M. Tumor immunotherapy in melanoma: Strategies for overcoming mechanisms of resistance and escape. Am. J. Clin. Dermatol. 2008, 9, 307–311. [Google Scholar] [CrossRef]
- Merritt, W.M.; Lin, Y.G.; Spannuth, W.A.; Fletcher, M.S.; Kamat, A.A.; Han, L.Y.; Landen, C.N.; Jennings, N.; De Geest, K.; Langley, R.R.; et al. Effect of interleukin-8 gene silencing with liposome-encapsulated small interfering RNA on ovarian cancer cell growth. J. Natl. Cancer Inst. 2008, 100, 359–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, B.; Singh, S.; Varney, M.L.; Singh, R.K. Targeting CXCR1/CXCR2 receptor antagonism in malignant melanoma. Expert Opin. Ther. Targets 2010, 14, 435–442. [Google Scholar] [CrossRef] [Green Version]
- Goldstein, L.J.; Perez, R.P.; Yardley, D.; Han, L.K.; Reuben, J.M.; Gao, H.; McCanna, S.; Butler, B.; Ruffini, P.A.; Liu, Y.; et al. A window-of-opportunity trial of the CXCR1/2 inhibitor reparixin in operable HER-2-negative breast cancer. Breast Cancer Res. 2020, 22, 4. [Google Scholar] [CrossRef] [PubMed]
- Kemp, D.M.; Pidich, A.; Larijani, M.; Jonas, R.; Lash, E.; Sato, T.; Terai, M.; De Pizzol, M.; Allegretti, M.; Igoucheva, O.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hartman, M.L.; Rozanski, M.; Osrodek, M.; Zalesna, I.; Czyz, M. Vemurafenib and trametinib reduce expression of CTGF and IL-8 in V600EBRAF melanoma cells. Lab. Investig. 2017, 97, 217–227. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Han, X.L. Anti–PD-1/PD-L1 therapy of human cancer: Past, present, and future. J. Clin. Investig. 2015, 125, 3384–3391. [Google Scholar] [CrossRef] [Green Version]
- Topalian, S.L.; Sznol, M.; McDermott, D.F.; Kluger, H.M.; Carvajal, R.D.; Sharfman, W.H.; Brahmer, J.R.; Lawrence, D.P.; Atkins, M.B.; Powderly, J.D.; et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J. Clin. Oncol. 2014, 32, 1020–1030. [Google Scholar] [CrossRef]
- Hodi, F.S.; Hwu, W.-J.; Kefford, R.; Weber, J.S.; Daud, A.; Hamid, O.; Patnaik, A.; Ribas, A.; Robert, C.; Gangadhar, T.C.; et al. Evaluation of immune-related response criteria and RECIST v1.1 in patients with advanced melanoma treated with Pembrolizumab. J. Clin. Oncol. 2016, 34, 1510–1517. [Google Scholar] [CrossRef]
- Sullivan, R.J.; Hamid, O.; Gonzalez, R.; Infante, J.R.; Patel, M.R.; Hodi, F.S.; Lewis, K.D.; Tawbi, H.A.; Hernandez, G.; Wongchenko, M.J.; et al. Atezolizumab plus cobimetinib and vemurafenib in BRAF-mutated melanoma patients. Nat. Med. 2019, 25, 929–935. [Google Scholar] [CrossRef]
- Lipson, E.J.; Drake, C.G. Ipilimumab: An Anti-CTLA-4 Antibody for Metastatic Melanoma. Clin. Cancer Res. 2011, 17, 6958–6962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scherpereel, A.; Mazieres, J.; Greillier, L.; Lantuejoul, S.; Dô, P.; Bylicki, O.; Monnet, I.; Corre, R.; Audigier-Valette, C.; Locatelli-Sanchez, M.; et al. Nivolumab or nivolumab plus ipilimumab in patients with relapsed malignant pleural mesothelioma (IFCT-1501 MAPS2): A multicentre, open-label, randomised, non-comparative, phase 2 trial. Lancet Oncol. 2019, 20, 239–253. [Google Scholar] [CrossRef]
- Sanmamed, M.F.; Perez-Gracia, J.L.; Schalper, K.A.; Fusco, J.P.; Gonzalez, A.; Rodriguez-Ruiz, M.E.; Oñate, C.; Perez, G.; Alfaro, C.; Martín-Algarra, S.; et al. Changes in serum interleukin-8 (IL-8) levels reflect and predict response to anti-PD-1 treatment in melanoma and non-small-cell lung cancer patients. Ann. Oncol. 2017, 28, 1988–1995. [Google Scholar] [CrossRef] [PubMed]
- Umansky, V.; Sevko, A.; Gebhardt, C.; Utikal, J. Myeloide Suppressorzellen (MDSC) beim malignen Melanom. JDDG J. Ger. Soc. Dermatol. 2014, 12, 1021–1027. [Google Scholar] [CrossRef] [Green Version]
- Li, E.; Yang, X.; Du, Y.; Wang, G.; Chan, D.W.; Wu, D.; Xu, P.; Ni, P.; Xu, D.; Hu, Y. CXCL8 Associated Dendritic Cell Activation Marker Expression and Recruitment as Indicators of Favorable Outcomes in Colorectal Cancer. Front. Immunol. 2021, 12, 667177. [Google Scholar] [CrossRef]
- Chen, M.B.; Hajal, C.; Benjamin, D.C.; Yu, C.; Azizgolshani, H.; Hynes, R.O.; Kamm, R.D. Inflamed neutrophils sequestered at entrapped tumor cells via chemotactic confinement promote tumor cell extravasation. Proc. Natl. Acad. Sci. USA 2018, 115, 7022–7027. [Google Scholar] [CrossRef] [Green Version]
- Huh, S.J.; Liang, S.; Sharma, A.; Dong, C.; Robertson, G.P. Transiently entrapped circulating tumor cells interact with neutrophils to facilitate lung metastasis development. Cancer Res. 2010, 70, 6071–6082. [Google Scholar] [CrossRef] [Green Version]
- Schott, A.F.; Goldstein, L.J.; Cristofanilli, M.; Ruffini, P.A.; McCanna, S.; Reuben, J.M.; Perez, R.P.; Kato, G.; Wicha, M. Phase Ib pilot study to evaluate reparixin in combination with weekly paclitaxel in patients with HER-2–negative metastatic breast cancer. Clin. Cancer Res. 2017, 23, 5358–5365. [Google Scholar] [CrossRef] [Green Version]
- Nywening, T.M.; Belt, B.A.; Cullinan, D.R.; Panni, R.Z.; Han, B.J.; Sanford, D.E.; Jacobs, R.C.; Ye, J.; Patel, A.A.; Gillanders, W.E.; et al. Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut 2018, 67, 1112–1123. [Google Scholar] [CrossRef] [Green Version]
- Shi, Y.; Tomczak, K.; Li, J.; Ochieng, J.K.; Lee, Y.; Haymaker, C. Next-Generation Immunotherapies to Improve Anticancer Immunity. Front. Pharmacol. 2021, 11, 566401. [Google Scholar] [CrossRef]
- Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 1947. [Google Scholar] [CrossRef]
- Beatty, G.L.; Torigian, D.A.; Chiorean, E.G.; Saboury, B.; Brothers, A.; Alavi, A.; Troxel, A.B.; Sun, W.; Teitelbaum, U.R.; Vonderheide, R.H.; et al. A Phase I Study of an Agonist CD40 Monoclonal Antibody (CP-870,893) in Combination with Gemcitabine in Patients with Advanced Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. 2013, 19, 6286–6295. [Google Scholar] [CrossRef] [Green Version]
- Kaneda, M.M.; Messer, K.S.; Ralainirina, N.; Li, H.; Leem, C.; Gorjestani, S.; Gyunghwi, W.; Nguyen, A.V.; Figueiredo, C.C.; Foubert, P.; et al. PI3Kγ is a molecular switch that controls immune suppression. Nature 2016, 539, 437–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carminati, L.; Pinessi, D.; Borsotti, P.; Minoli, L.; Giavazzi, R.; D’Incalci, M.; Belotti, D.; Taraboletti, G. Antimetastatic and antiangiogenic activity of trabectedin in cutaneous melanoma. OUP 2018, 40, 303–312. [Google Scholar] [CrossRef] [PubMed]
- Etzerodt, A.; Tsalkitzi, K.; Maniecki, M.; Damsky, W.; Delfini, M.; Baudoin, E.; Moulin, M.; Bosenberg, M.; Graversen, J.H.; Auphan-Anezin, N.; et al. Specific targeting of CD163+ TAMs mobilizes inflammatory monocytes and promotes T cell-mediated tumor regression. J. Exp. Med. 2019, 216, 2394–2411. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Filimon, A.; Preda, I.A.; Boloca, A.F.; Negroiu, G. Interleukin-8 in Melanoma Pathogenesis, Prognosis and Therapy—An Integrated View into Other Neoplasms and Chemokine Networks. Cells 2022, 11, 120. https://doi.org/10.3390/cells11010120
Filimon A, Preda IA, Boloca AF, Negroiu G. Interleukin-8 in Melanoma Pathogenesis, Prognosis and Therapy—An Integrated View into Other Neoplasms and Chemokine Networks. Cells. 2022; 11(1):120. https://doi.org/10.3390/cells11010120
Chicago/Turabian StyleFilimon, Anca, Iulia A. Preda, Adina F. Boloca, and Gabriela Negroiu. 2022. "Interleukin-8 in Melanoma Pathogenesis, Prognosis and Therapy—An Integrated View into Other Neoplasms and Chemokine Networks" Cells 11, no. 1: 120. https://doi.org/10.3390/cells11010120
APA StyleFilimon, A., Preda, I. A., Boloca, A. F., & Negroiu, G. (2022). Interleukin-8 in Melanoma Pathogenesis, Prognosis and Therapy—An Integrated View into Other Neoplasms and Chemokine Networks. Cells, 11(1), 120. https://doi.org/10.3390/cells11010120