Chemokines and Cytokines in Immunotherapy of Melanoma and Other Tumors: From Biomarkers to Therapeutic Targets
Abstract
:1. Introduction
2. CXCL9
3. CXCL10
4. CXCL13
5. CXCL16
6. XCL1
7. CCL2
8. CCL4
9. CCL5
10. CCL22
11. IL-2
12. IL-4
13. IL-6
14. IL-12
15. IL-15
16. IL-17
17. The Drawbacks of Targeting Chemokines and Cytokines
18. Conclusions and Future Perspectives
Study | Model | Cell Lines/Mice | Agent | |
---|---|---|---|---|
CXCL10 | [28] | In vitro, in vivo | C57BL⁄6 mice B16F10 melanoma cells | adenovirus vector expressing human CXCL10 (AdCXCL10) |
[138] | In vitro, in vivo | nude mice A375 human melanoma cells | retrovirally transduced A375 human melanoma cells | |
CCL2 | [56] | In vitro, in vivo | tumor cell lines NA13, B16F10 and E0771 | RS504393, a small-molecule inhibitor of the CCL2 cognate receptor |
CCL4 | [65] | In vitro, in vivo | B16F10 tumor-bearing mice B16F10 melanoma cells EMT6 breast cancer cells | fusion protein of CCL4 and the collagen-binding domain (CBD) of the von Willebrand factor |
XCL1 | [49] | In vivo | C57Bl/6 mice bearing MC38 colon cancer or B16-OVA melanoma | Semliki Forest Virus (SFV)-based vectors encoding XCL1 |
[52] | In vivo | C57BL/6J mice B16-OVA cells | fusion protein consisting of an Ag peptide presented with MHC class I, an XCR1 ligand plus an immune adjuvant and polyinosinic:polycytidylic acids (poly(I:C)) | |
[51] | In vivo | C57BL/6J (B6) mice B16-OVA or B16 melanoma cells | Laser-assisted intradermal delivery of an OVA Ag fused to XCL1 | |
IL-15 | [112] | In vivo | C57BL/6J mice B16F10 cells | human IL-15 plasmid delivered by electroporation |
[109] | In vitro, in vivo | B6D2F1 mice 70Z/3-L leukemia cells | Lentivirally transduced cells producing soluble IL-15 | |
IL-2 | [85] | In vivo | Humans with metastatic melanoma | Intratumoral IL-2 +/− chemotherapy |
[86] | In vivo | Humans with metastatic melanoma | Intratumoral IL-2 | |
IL-6 | [102] | In vivo | KPC-Brca2 mice with Panc02, MT5 or KPC-luc cell lines | Intratumoral anti-IL-6 = anti-PD-L1 antibodies |
Study Phase | Patients # | Agent | Cytokine Delivery | ClinicalTrials.gov Identifier | |
---|---|---|---|---|---|
IL-15 | Phase I/Ib | 60 | Recombinant heterodimeric IL 5 (IL-15/sIL-15Ra) alone or with Spartalizumab | Subcutaneous | NCT04261439 |
Phase IIb | 147 | Fusion protein of IL-15 mutant bound to IL-15 receptor alpha/immunoglobulin G1 crystallizable fragment (Fc) In combination with Pembrolizumab, Nivolumab, Atezolizumab, Avelumab or Durvalumab | Subcutaneous | NCT03228667 | |
IL-6 | Phase II | 75 | Tocilizumab (anti-IL-6R) in combination with nivolumab and ipilimumab | Intravenous | NCT03999749 |
Phase II | 69 | Sarilumab (anti-IL-6R) in combination with ipilimumab, nivolumab and relatlimab | Subcutaneous | NCT05428007 | |
Phase II | 35 * | Tocilizumab (anti-IL-6R) in combination with nivolumab and ipilimumab | Subcutaneous | NCT04940299 | |
IL-12 | Phase I | 94 + | Monoclonal antibody-cytokine fusion protein IL12-L19L19 | Intravenous | NCT04471987 |
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef]
- Versluis, J.M.; Blankenstein, S.A.; Dimitriadis, P.; Wilmott, J.S.; Elens, R.; Blokx, W.A.M.; van Houdt, W.; Menzies, A.M.; Schrage, Y.M.; Wouters, M.W.J.M.; et al. Interferon-gamma signature as prognostic and predictive marker in macroscopic stage III melanoma. J. Immunother. Cancer 2024, 12, e008125. [Google Scholar] [CrossRef] [PubMed]
- Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef]
- Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.M.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.; Ciobanu, V.; et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515, 568–571. [Google Scholar] [CrossRef]
- Harlin, H.; Meng, Y.; Peterson, A.C.; Zha, Y.; Tretiakova, M.; Slingluff, C.; McKee, M.; Gajewski, T.F. Chemokine Expression in Melanoma Metastases Associated with CD8+ T-Cell Recruitment. Cancer Res. 2009, 69, 3077–3085. [Google Scholar] [CrossRef]
- Mikucki, M.E.; Fisher, D.T.; Matsuzaki, J.; Skitzki, J.J.; Gaulin, 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]
- Reschke, R.; Gajewski, T.F. CXCL9 and CXCL10 bring the heat to tumors. Sci. Immunol. 2022, 7, eabq6509. [Google Scholar] [CrossRef] [PubMed]
- Spranger, S.; Dai, D.; Horton, B.; Gajewski, T.F. Tumor-Residing Batf3 Dendritic Cells Are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 2017, 31, 711–723.e4. [Google Scholar] [CrossRef] [PubMed]
- Brewitz, A.; Eickhoff, S.; Dähling, S.; Quast, T.; Bedoui, S.; Kroczek, R.A.; Kurts, C.; Garbi, N.; Barchet, W.; Iannacone, M.; et al. CD8+ T Cells Orchestrate pDC-XCR1+ Dendritic Cell Spatial and Functional Cooperativity to Optimize Priming. Immunity 2017, 46, 205–219. [Google Scholar] [CrossRef]
- Kroczek, R.A.; Henn, V. The Role of XCR1 and its Ligand XCL1 in Antigen Cross-Presentation by Murine and Human Dendritic Cells. Front. Immunol. 2012, 3, 14. [Google Scholar] [CrossRef]
- Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef] [PubMed]
- Ziblat, A.; Horton, B.L.; Higgs, E.F.; Hatogai, K.; Martinez, A.; Shapiro, J.W.; Kim, D.E.; Zha, Y.; Sweis, R.F.; Gajewski, T.F. Batf3+ DCs and the 4-1BB/4-1BBL axis are required at the effector phase in the tumor microenvironment for PD-1/PD-L1 blockade efficacy. Cell Rep. 2024, 43, 114141. [Google Scholar] [CrossRef] [PubMed]
- Nagarsheth, N.; Wicha, M.S.; Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017, 17, 559–572. [Google Scholar] [CrossRef] [PubMed]
- Ding, Q.; Lu, P.; Xia, Y.; Ding, S.; Fan, Y.; Li, X.; Han, P.; Liu, J.; Tian, D.; Liu, M. CXCL9: Evidence and contradictions for its role in tumor progression. Cancer Med. 2016, 5, 3246–3259. [Google Scholar] [CrossRef] [PubMed]
- Wightman, S.C.; Uppal, A.; Pitroda, S.P.; Ganai, S.; Burnette, B.; Stack, M.; Oshima, G.; Khan, S.; Huang, X.; Posner, M.C.; et al. Oncogenic CXCL10 signalling drives metastasis development and poor clinical outcome. Br. J. Cancer 2015, 113, 327–335. [Google Scholar] [CrossRef] [PubMed]
- Amatschek, S.; Lucas, R.; Eger, A.; Pflueger, M.; Hundsberger, H.; Knoll, C.; Grosse-Kracht, S.; Schuett, W.; Koszik, F.; Maurer, D.; et al. CXCL9 induces chemotaxis, chemorepulsion and endothelial barrier disruption through CXCR3-mediated activation of melanoma cells. Br. J. Cancer 2011, 104, 469–479. [Google Scholar] [CrossRef] [PubMed]
- Marcovecchio, P.M.; Thomas, G.; Salek-Ardakani, S. CXCL9-expressing tumor-associated macrophages: New players in the fight against cancer. J. Immunother. Cancer 2021, 9, e002045. [Google Scholar] [CrossRef] [PubMed]
- Xiao, P.; Guo, Y.; Zhang, H.; Zhang, X.; Cheng, H.; Cao, Q.; Ke, Y. Myeloid-restricted ablation of Shp2 restrains melanoma growth by amplifying the reciprocal promotion of CXCL9 and IFN-γ production in tumor microenvironment. Oncogene 2018, 37, 5088–5100. [Google Scholar] [CrossRef]
- Karin, N. CXCR3 Ligands in Cancer and Autoimmunity, Chemoattraction of Effector T Cells, and Beyond. Front. Immunol. 2020, 11, 976. [Google Scholar] [CrossRef]
- Kuo, P.T.; Zeng, Z.; Salim, N.; Mattarollo, S.; Wells, J.W.; Leggatt, G.R. The Role of CXCR3 and Its Chemokine Ligands in Skin Disease and Cancer. Front. Med. 2018, 5, 271. [Google Scholar] [CrossRef]
- Bedognetti, D.; Spivey, T.L.; Zhao, Y.; Uccellini, L.; Tomei, S.; Dudley, M.E.; Ascierto, M.L.; De Giorgi, V.; Liu, Q.; Delogu, L.G.; et al. CXCR3/CCR5 pathways in metastatic melanoma patients treated with adoptive therapy and interleukin-2. Br. J. Cancer 2013, 109, 2412–2423. [Google Scholar] [CrossRef] [PubMed]
- House, I.G.; Savas, P.; Lai, J.; Chen, A.X.Y.; Oliver, A.J.; Teo, Z.L.; Todd, K.L.; Henderson, M.A.; Giuffrida, L.; Petley, E.V.; et al. Macrophage-Derived CXCL9 and CXCL10 Are Required for Antitumor Immune Responses Following Immune Checkpoint Blockade. Clin. Cancer Res. 2020, 26, 487–504. [Google Scholar] [CrossRef] [PubMed]
- Reschke, R.; Yu, J.; Flood, B.A.; Higgs, E.F.; Hatogai, K.; Gajewski, T.F. Immune cell and tumor cell-derived CXCL10 is indicative of immunotherapy response in metastatic melanoma. J. Immunother. Cancer 2021, 9, e003521. [Google Scholar] [CrossRef] [PubMed]
- Chow, M.T.; Ozga, A.J.; Servis, R.L.; Frederick, D.T.; Lo, J.A.; Fisher, D.E.; Freeman, G.J.; Boland, G.M.; Luster, A.D. Intratumoral Activity of the CXCR3 Chemokine System Is Required for the Efficacy of Anti-PD-1 Therapy. Immunity 2019, 50, 1498–1512.e5. [Google Scholar] [CrossRef] [PubMed]
- Yin, P.; Gui, L.; Wang, C.; Yan, J.; Liu, M.; Ji, L.; Wang, Y.; Ma, B.; Gao, W.-Q. Targeted Delivery of CXCL9 and OX40L by Mesenchymal Stem Cells Elicits Potent Antitumor Immunity. Mol. Ther. 2020, 28, 2553–2563. [Google Scholar] [CrossRef] [PubMed]
- Bagheri, H.; Pourhanifeh, M.H.; Derakhshan, M.; Mahjoubin-Tehran, M.; Ghasemi, F.; Mousavi, S.; Rafiei, R.; Abbaszadeh-Goudarzi, K.; Mirzaei, H.R. CXCL-10: A new candidate for melanoma therapy? Cell Oncol. 2020, 43, 353–365. [Google Scholar] [CrossRef] [PubMed]
- Chheda, Z.S.; Sharma, R.K.; Jala, V.R.; Luster, A.D.; Haribabu, B. Chemoattractant Receptors BLT1 and CXCR3 Regulate Antitumor Immunity by Facilitating CD8 + T Cell Migration into Tumors. J. Immunol. 2016, 197, 2016–2026. [Google Scholar] [CrossRef] [PubMed]
- Antonicelli, F.; Lorin, J.; Kurdykowski, S.; Gangloff, S.C.; Le Naour, R.; Sallenave, J.M.; Hornebeck, W.; Grange, F.; Bernard, P. CXCL10 reduces melanoma proliferation and invasiveness in vitro and in vivo: CXCL10 and melanoma progression. Br. J. Dermatol. 2011, 164, 720–728. [Google Scholar] [CrossRef]
- Zumwalt, T.J.; Arnold, M.; Goel, A.; Boland, C.R. Active secretion of CXCL10 and CCL5 from colorectal cancer microenvironments associates with GranzymeB+ CD8+ T-cell infiltration. Oncotarget 2015, 6, 2981–2991. [Google Scholar] [CrossRef]
- Barash, U.; Zohar, Y.; Wildbaum, G.; Beider, K.; Nagler, A.; Karin, N.; Ilan, N.; Vlodavsky, I. Heparanase enhances myeloma progression via CXCL10 downregulation. Leukemia 2014, 28, 2178–2187. [Google Scholar] [CrossRef]
- Arenberg, D.; White, E.; Burdick, M.; Strom, S.; Strieter, R. Improved survival in tumor-bearing SCID mice treated with interferon-γ-inducible protein 10 (IP-10/CXCL10). Cancer Immunol. Immunother. 2001, 50, 533–538. [Google Scholar] [CrossRef] [PubMed]
- Bronger, H.; Singer, J.; Windmüller, C.; Reuning, U.; Zech, D.; Delbridge, C.; Dorn, J.; Kiechle, M.; Schmalfeldt, B.; Schmitt, M.; et al. CXCL9 and CXCL10 predict survival and are regulated by cyclooxygenase inhibition in advanced serous ovarian cancer. Br. J. Cancer 2016, 115, 553–563. [Google Scholar] [CrossRef] [PubMed]
- Reschke, R.; Shapiro, J.W.; Yu, J.; Rouhani, S.J.; Olson, D.J.; Zha, Y.; Gajewski, T.F. Checkpoint blockade–induced dermatitis and colitis are dominated by tissue resident memory T cells and Th1/Tc1 cytokines. Cancer Immunol. Res. 2022, 10, 1167–1174. [Google Scholar] [CrossRef] [PubMed]
- Gao, S.-H.; Liu, S.-Z.; Wang, G.-Z.; Zhou, G.-B. CXCL13 in Cancer and Other Diseases: Biological Functions, Clinical Significance, and Therapeutic Opportunities. Life 2021, 11, 1282. [Google Scholar] [CrossRef] [PubMed]
- O’Connor, R.A.; Martinez, B.R.; Koppensteiner, L.; Mathieson, L.; Akram, A.R. Cancer-associated fibroblasts drive CXCL13 production in activated T cells via TGF-beta. Front. Immunol. 2023, 14, 1221532. [Google Scholar] [CrossRef] [PubMed]
- Groeneveld, C.S.; Fontugne, J.; Cabel, L.; Bernard-Pierrot, I.; Radvanyi, F.; Allory, Y.; de Reyniès, A. Tertiary lymphoid structures marker CXCL13 is associated with better survival for patients with advanced-stage bladder cancer treated with immunotherapy. Eur. J. Cancer 2021, 148, 181–189. [Google Scholar] [CrossRef]
- Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef] [PubMed]
- Yang, M.; Lu, J.; Zhang, G.; Wang, Y.; He, M.; Xu, Q.; Xu, C.; Liu, H. CXCL13 shapes immunoactive tumor microenvironment and enhances the efficacy of PD-1 checkpoint blockade in high-grade serous ovarian cancer. J. Immunother. Cancer 2021, 9, e001136. [Google Scholar] [CrossRef] [PubMed]
- Hojo, S.; Koizumi, K.; Tsuneyama, K.; Arita, Y.; Cui, Z.; Shinohara, K.; Minami, T.; Hashimoto, I.; Nakayama, T.; Sakurai, H.; et al. High-level expression of chemokine CXCL16 by tumor cells correlates with a good prognosis and increased tumor-infiltrating lymphocytes in colorectal cancer. Cancer Res. 2007, 67, 4725–4731. [Google Scholar] [CrossRef]
- Matsumura, S.; Wang, B.; Kawashima, N.; Braunstein, S.; Badura, M.; Cameron, T.O.; Babb, J.S.; Schneider, R.J.; Formenti, S.C.; Dustin, M.L.; et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J. Immunol. 2008, 181, 3099–3107. [Google Scholar] [CrossRef]
- Karaki, S.; Blanc, C.; Tran, T.; Galy-Fauroux, I.; Mougel, A.; Dransart, E.; Anson, M.; Tanchot, C.; Paolini, L.; Gruel, N.; et al. CXCR6 deficiency impairs cancer vaccine efficacy and CD8+ resident memory T-cell recruitment in head and neck and lung tumors. J. Immunother. Cancer 2021, 9, e001948. [Google Scholar] [CrossRef] [PubMed]
- Reschke, R.; Deitert, B.; Enk, A.H.; Hassel, J.C. The role of tissue-resident memory T cells as mediators for response and toxicity in immunotherapy-treated melanoma—Two sides of the same coin? Front. Immunol. 2024, 15, 1385781. [Google Scholar] [CrossRef] [PubMed]
- Reschke, R.; Gajewski, T.F. Tissue-resident memory T cells in immune-related adverse events: Friend or foe? Oncoimmunology 2023, 12, 2197358. [Google Scholar] [CrossRef]
- Hald, S.M.; Kiselev, Y.; Al-Saad, S.; Richardsen, E.; Johannessen, C.; Eilertsen, M.; Kilvaer, T.K.; Al-Shibli, K.; Andersen, S.; Busund, L.-T.; et al. Prognostic impact of CXCL16 and CXCR6 in non-small cell lung cancer: Combined high CXCL16 expression in tumor stroma and cancer cells yields improved survival. BMC Cancer 2015, 15, 441. [Google Scholar] [CrossRef] [PubMed]
- Cullen, R.; Germanov, E.; Shimaoka, T.; Johnston, B. Enhanced tumor metastasis in response to blockade of the chemokine receptor CXCR6 is overcome by NKT cell activation. J. Immunol. 2009, 183, 5807–5815. [Google Scholar] [CrossRef] [PubMed]
- Matsuo, K.; Kitahata, K.; Kawabata, F.; Kamei, M.; Hara, Y.; Takamura, S.; Oiso, N.; Kawada, A.; Yoshie, O.; Nakayama, T. A Highly Active Form of XCL1/Lymphotactin Functions as an Effective Adjuvant to Recruit Cross-Presenting Dendritic Cells for Induction of Effector and Memory CD8+ T Cells. Front. Immunol. 2018, 9, 2775. [Google Scholar] [CrossRef] [PubMed]
- Chandrasekar, B.; Mummidi, S.; Valente, A.J.; Patel, D.N.; Bailey, S.R.; Freeman, G.L.; Hatano, M.; Tokuhisa, T.; Jensen, L.E. The Pro-atherogenic Cytokine Interleukin-18 Induces CXCL16 Expression in Rat Aortic Smooth Muscle Cells via MyD88, Interleukin-1 Receptor-associated Kinase, Tumor Necrosis Factor Receptor-associated Factor 6, c-Src, Phosphatidylinositol 3-Kinase, Akt, c-Jun N-terminal Kinase, and Activator Protein-1 Signaling*. J. Biol. Chem. 2005, 280, 26263–26277. [Google Scholar] [CrossRef] [PubMed]
- Böttcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Sousa, C.R. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037.e14. [Google Scholar] [CrossRef] [PubMed]
- Sánchez-Paulete, A.R.; Teijeira, Á.; Quetglas, J.I.; Rodríguez-Ruiz, M.E.; Sánchez-Arráez, Á.; Labiano, S.; Etxeberria, I.; Azpilikueta, A.; Bolaños, E.; Ballesteros-Briones, M.C.; et al. Intratumoral Immunotherapy with XCL1 and sFlt3L Encoded in Recombinant Semliki Forest Virus–Derived Vectors Fosters Dendritic Cell–Mediated T-cell Cross-Priming. Cancer Res. 2018, 78, 6643–6654. [Google Scholar] [CrossRef]
- Chen, K.; Wu, Z.; Zhao, H.; Wang, Y.; Ge, Y.; Wang, D.; Li, Z.; An, C.; Liu, Y.; Wang, F.; et al. XCL1/Glypican-3 Fusion Gene Immunization Generates Potent Antitumor Cellular Immunity and Enhances Anti-PD-1 Efficacy. Cancer Immunol. Res. 2020, 8, 81–93. [Google Scholar] [CrossRef]
- Terhorst, D.; Fossum, E.; Baranska, A.; Tamoutounour, S.; Malosse, C.; Garbani, M.; Braun, R.; Lechat, E.; Crameri, R.; Bogen, B.; et al. Laser-assisted intradermal delivery of adjuvant-free vaccines targeting XCR1+ dendritic cells induces potent antitumoral responses. J. Immunol. 2015, 194, 5895–5902. [Google Scholar] [CrossRef] [PubMed]
- Mizumoto, Y.; Hemmi, H.; Katsuda, M.; Miyazawa, M.; Kitahata, Y.; Miyamoto, A.; Nakamori, M.; Ojima, T.; Matsuda, K.; Nakamura, M.; et al. Anticancer effects of chemokine-directed antigen delivery to a cross-presenting dendritic cell subset with immune checkpoint blockade. Br. J. Cancer 2020, 122, 1185–1193. [Google Scholar] [CrossRef] [PubMed]
- Matsushima, K.; Larsen, C.G.; DuBois, G.C.; Oppenheim, J.J. Purification and characterization of a novel monocyte chemotactic and activating factor produced by a human myelomonocytic cell line. J. Exp. Med. 1989, 169, 1485–1490. [Google Scholar] [CrossRef] [PubMed]
- Ohanna, M.; Giuliano, S.; Bonet, C.; Imbert, V.; Hofman, V.; Zangari, J.; Bille, K.; Robert, C.; Paillerets, B.B.-D.; Hofman, P.; et al. Senescent cells develop a PARP-1 and nuclear factor-{kappa}B-associated secretome (PNAS). Genes Dev. 2011, 25, 1245–1261. [Google Scholar] [CrossRef] [PubMed]
- Lim, S.Y.; Yuzhalin, A.E.; Gordon-Weeks, A.N.; Muschel, R.J. Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget 2016, 7, 28697–28710. [Google Scholar] [CrossRef] [PubMed]
- Tu, M.M.; Abdel-Hafiz, H.A.; Jones, R.T.; Jean, A.; Hoff, K.J.; Duex, J.E.; Chauca-Diaz, A.; Costello, J.C.; Dancik, G.M.; Tamburini, B.A.J.; et al. Inhibition of the CCL2 receptor, CCR2, enhances tumor response to immune checkpoint therapy. Commun. Biol. 2020, 3, 720. [Google Scholar] [CrossRef] [PubMed]
- Jin, J.; Lin, J.; Xu, A.; Lou, J.; Qian, C.; Li, X.; Wang, Y.; Yu, W.; Tao, H. CCL2: An Important Mediator Between Tumor Cells and Host Cells in Tumor Microenvironment. Front. Oncol. 2021, 11, 722916. [Google Scholar] [CrossRef]
- Li, X.; Yao, W.; Yuan, Y.; Chen, P.; Li, B.; Li, J.; Chu, R.; Song, H.; Xie, D.; Jiang, X.; et al. Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut 2017, 66, 157–167. [Google Scholar] [CrossRef] [PubMed]
- Fridlender, Z.G.; Buchlis, G.; Kapoor, V.; Cheng, G.; Sun, J.; Singhal, S.; Crisanti, M.C.; Wang, L.-C.S.; Heitjan, D.; Snyder, L.A.; et al. CCL2 Blockade Augments Cancer Immunotherapy. Cancer Res. 2010, 70, 109. [Google Scholar] [CrossRef]
- Wang, Y.; Zhang, X.; Yang, L.; Xue, J.; Hu, G. Blockade of CCL2 enhances immunotherapeutic effect of anti-PD1 in lung cancer. J. Bone Oncol. 2018, 11, 27–32. [Google Scholar] [CrossRef]
- Oliva, A.; Kinter, A.L.; Vaccarezza, M.; Rubbert, A.; Catanzaro, A.; Moir, S.; Monaco, J.; Ehler, L.; Mizell, S.; Jackson, R.; et al. Natural killer cells from human immunodeficiency virus (HIV)-infected individuals are an important source of CC-chemokines and suppress HIV-1 entry and replication in vitro. J. Clin. Investig. 1998, 102, 223–231. [Google Scholar] [CrossRef]
- Sektioglu, I.M.; Carretero, R.; Bulbuc, N.; Bald, T.; Tüting, T.; Rudensky, A.Y.; Hämmerling, G.J. Basophils Promote Tumor Rejection via Chemotaxis and Infiltration of CD8+ T Cells. Cancer Res. 2017, 77, 291–302. [Google Scholar] [CrossRef]
- Huang, B.; Han, W.; Sheng, Z.-F.; Shen, G.-L. Identification of immune-related biomarkers associated with tumorigenesis and prognosis in cutaneous melanoma patients. Cancer Cell Int. 2020, 20, 195. [Google Scholar] [CrossRef]
- Fairfax, B.P.; Taylor, C.A.; Watson, R.A.; Nassiri, I.; Danielli, S.; Fang, H.; Mahé, E.A.; Cooper, R.; Woodcock, V.; Traill, Z.; et al. Peripheral CD8+ T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma. Nat. Med. 2020, 26, 193–199. [Google Scholar] [CrossRef]
- Williford, J.-M.; Ishihara, J.; Ishihara, A.; Mansurov, A.; Hosseinchi, P.; Marchell, T.M.; Potin, L.; Swartz, M.A.; Hubbell, J.A. Recruitment of CD103+ dendritic cells via tumor-targeted chemokine delivery enhances efficacy of checkpoint inhibitor immunotherapy. Sci. Adv. 2019, 5, eaay1357. [Google Scholar] [CrossRef]
- Appay, V.; Rowland-Jones, S.L. RANTES: A versatile and controversial chemokine. Trends Immunol. 2001, 22, 83–87. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.-F.; Zhang, X.-L.; Wang, Y.-J.; Fang, Y.; Li, M.-L.; Liu, X.-Y.; Luo, H.-Y.; Tian, Y. The regulatory network of the chemokine CCL5 in colorectal cancer. Ann. Med. 2023, 55, 2205168. [Google Scholar] [CrossRef] [PubMed]
- Halama, N.; Zoernig, I.; Berthel, A.; Kahlert, C.; Klupp, F.; Suarez-Carmona, M.; Suetterlin, T.; Brand, K.; Krauss, J.; Lasitschka, F.; et al. Tumoral Immune Cell Exploitation in Colorectal Cancer Metastases Can Be Targeted Effectively by Anti-CCR5 Therapy in Cancer Patients. Cancer Cell 2016, 29, 587–601. [Google Scholar] [CrossRef] [PubMed]
- Jiao, X.; Nawab, O.; Patel, T.; Kossenkov, A.V.; Halama, N.; Jaeger, D.; Pestell, R.G. Recent Advances targeting CCR5 for Cancer and its Role in Immuno-Oncology. Cancer Res. 2019, 79, 4801–4807. [Google Scholar] [CrossRef]
- Yamashita, U.; Kuroda, E. Regulation of macrophage-derived chemokine (MDC, CCL22) production. Crit. Rev. Immunol. 2002, 22, 105–114. [Google Scholar] [CrossRef]
- Yashiro, T.; Nakano, S.; Nomura, K.; Uchida, Y.; Kasakura, K.; Nishiyama, C. A transcription factor PU.1 is critical for Ccl22 gene expression in dendritic cells and macrophages. Sci. Rep. 2019, 9, 1161. [Google Scholar] [CrossRef] [PubMed]
- Klarquist, J.; Tobin, K.; Farhangi Oskuei, P.; Henning, S.W.; Fernandez, M.F.; Dellacecca, E.R.; Navarro, F.C.; Eby, J.M.; Chatterjee, S.; Mehrotra, S.; et al. Ccl22 Diverts T Regulatory Cells and Controls the Growth of Melanoma. Cancer Res. 2016, 76, 6230–6240. [Google Scholar] [CrossRef] [PubMed]
- Mizukami, Y.; Kono, K.; Kawaguchi, Y.; Akaike, H.; Kamimura, K.; Sugai, H.; Fujii, H. CCL17 and CCL22 chemokines within tumor microenvironment are related to accumulation of Foxp3+ regulatory T cells in gastric cancer. Int. J. Cancer 2008, 122, 2286–2293. [Google Scholar] [CrossRef] [PubMed]
- Wiedemann, G.M.; Knott, M.M.L.; Vetter, V.K.; Rapp, M.; Haubner, S.; Fesseler, J.; Kühnemuth, B.; Layritz, P.; Thaler, R.; Kruger, S.; et al. Cancer cell-derived IL-1α induces CCL22 and the recruitment of regulatory T cells. Oncoimmunology 2016, 5, e1175794. [Google Scholar] [CrossRef] [PubMed]
- Sugiyama, D.; Nishikawa, H.; Maeda, Y.; Nishioka, M.; Tanemura, A.; Katayama, I.; Ezoe, S.; Kanakura, Y.; Sato, E.; Fukumori, Y.; et al. Anti-CCR4 mAb selectively depletes effector-type FoxP3+CD4+ regulatory T cells, evoking antitumor immune responses in humans. Proc. Natl. Acad. Sci. USA 2013, 110, 17945–17950. [Google Scholar] [CrossRef] [PubMed]
- Khabipov, A.; Trung, D.N.; van der Linde, J.; Miebach, L.; Lenz, M.; Erne, F.; von Bernstorff, W.; Schulze, T.; Kersting, S.; Bekeschus, S.; et al. CCR4 Blockade Diminishes Intratumoral Macrophage Recruitment and Augments Survival of Syngeneic Pancreatic Cancer-Bearing Mice. Biomedicines 2023, 11, 1517. [Google Scholar] [CrossRef] [PubMed]
- Tong, J.; Jiang, W.; Zhang, X.; Wang, R.; Qiao, T.; Song, Y.; Gao, D.; Yu, X.; Lv, Z.; Li, D. CCL22 and CCL26 are potential biomarkers for predicting distant metastasis in thyroid carcinoma. J. Int. Med. Res. 2022, 50, 3000605221139555. [Google Scholar] [CrossRef]
- Boyman, O.; Cho, J.-H.; Sprent, J. The role of interleukin-2 in memory CD8 cell differentiation. Adv. Exp. Med. Biol. 2010, 684, 28–41. [Google Scholar] [CrossRef] [PubMed]
- Malek, T.R. The biology of interleukin-2. Annu. Rev. Immunol. 2008, 26, 453–479. [Google Scholar] [CrossRef]
- Taniguchi, T.; Minami, Y. The IL-2/IL-2 receptor system: A current overview. Cell 1993, 73, 5–8. [Google Scholar] [CrossRef]
- Atkins, M.B.; Lotze, M.T.; Dutcher, J.P.; Fisher, R.I.; Weiss, G.; Margolin, K.; Abrams, J.; Sznol, M.; Parkinson, D.; Hawkins, M.; et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: Analysis of 270 patients treated between 1985 and 1993. J. Clin. Oncol. 1999, 17, 2105–2116. [Google Scholar] [CrossRef] [PubMed]
- Rosenberg, S.A.; Lotze, M.T.; Muul, L.M.; Leitman, S.; Chang, A.E.; Ettinghausen, S.E.; Matory, Y.L.; Skibber, J.M.; Shiloni, E.; Vetto, J.T.; et al. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N. Engl. J. Med. 1985, 313, 1485–1492. [Google Scholar] [CrossRef]
- Krieg, C.; Létourneau, S.; Pantaleo, G.; Boyman, O. Improved IL-2 immunotherapy by selective stimulation of IL-2 receptors on lymphocytes and endothelial cells. Proc. Natl. Acad. Sci. USA 2010, 107, 11906–11911. [Google Scholar] [CrossRef] [PubMed]
- Boyman, O.; Surh, C.D.; Sprent, J. Potential use of IL-2/anti-IL-2 antibody immune complexes for the treatment of cancer and autoimmune disease. Expert Opin. Biol. Ther. 2006, 6, 1323–1331. [Google Scholar] [CrossRef] [PubMed]
- Dummer, R.; Rochlitz, C.; Velu, T.; Acres, B.; Limacher, J.-M.; Bleuzen, P.; Lacoste, G.; Slos, P.; Romero, P.; Urosevic, M. Intralesional adenovirus-mediated interleukin-2 gene transfer for advanced solid cancers and melanoma. Mol. Ther. 2008, 16, 985–994. [Google Scholar] [CrossRef] [PubMed]
- Weide, B.; Derhovanessian, E.; Pflugfelder, A.; Eigentler, T.K.; Radny, P.; Zelba, H.; Pföhler, C.; Pawelec, G.; Garbe, C. High response rate after intratumoral treatment with interleukin-2: Results from a phase 2 study in 51 patients with metastasized melanoma. Cancer 2010, 116, 4139–4146. [Google Scholar] [CrossRef] [PubMed]
- Gattinoni, L.; Powell, D.J.; Rosenberg, S.A.; Restifo, N.P. Adoptive immunotherapy for cancer: Building on success. Nat. Rev. Immunol. 2006, 6, 383–393. [Google Scholar] [CrossRef] [PubMed]
- Goff, S.L.; Dudley, M.E.; Citrin, D.E.; Somerville, R.P.; Wunderlich, J.R.; Danforth, D.N.; Zlott, D.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; et al. Randomized, Prospective Evaluation Comparing Intensity of Lymphodepletion Before Adoptive Transfer of Tumor-Infiltrating Lymphocytes for Patients with Metastatic Melanoma. J. Clin. Oncol. 2016, 34, 2389–2397. [Google Scholar] [CrossRef] [PubMed]
- Chesney, J.; Lewis, K.D.; Kluger, H.; Hamid, O.; Whitman, E.; Thomas, S.; Wermke, M.; Cusnir, M.; Domingo-Musibay, E.; Phan, G.Q.; et al. Efficacy and safety of lifileucel, a one-time autologous tumor-infiltrating lymphocyte (TIL) cell therapy, in patients with advanced melanoma after progression on immune checkpoint inhibitors and targeted therapies: Pooled analysis of consecutive cohorts of the C-144-01 study. J. Immunother. Cancer 2022, 10, e005755. [Google Scholar] [CrossRef]
- Mullard, A. FDA approves first tumour-infiltrating lymphocyte (TIL) therapy, bolstering hopes for cell therapies in solid cancers. Nat. Rev. Drug Discov. 2024, 23, 238. [Google Scholar] [CrossRef]
- Diab, A.; Gogas, H.; Sandhu, S.; Long, G.V.; Ascierto, P.A.; Larkin, J.; Sznol, M.; Franke, F.; Ciuleanu, T.E.; Pereira, C.; et al. Bempegaldesleukin Plus Nivolumab in Untreated Advanced Melanoma: The Open-Label, Phase III PIVOT IO 001 Trial Results. J. Clin. Oncol. 2023, 41, 4756–4767. [Google Scholar] [CrossRef] [PubMed]
- Saif, A.; Rossi, A.J.; Sarnaik, A.; Hernandez, J.M.; Zager, J.S. Efficacy of Neoadjuvant Intratumoral Darleukin/Fibromun (L19IL2 + L19TNF) in Patients with Clinical Stage IIIB/C Melanoma (Neo-DREAM). Ann. Surg. Oncol. 2022, 29, 3377–3378. [Google Scholar] [CrossRef] [PubMed]
- Neoadjuvant Daromun Meets Primary End Point of RFS in Melanoma. Targeted Oncology 2023. Available online: https://www.targetedonc.com/view/neoadjuvant-daromun-meets-primary-end-point-of-rfs-in-melanoma (accessed on 8 May 2024).
- Humeau, J.; Le Naour, J.; Galluzzi, L.; Kroemer, G.; Pol, J.G. Trial watch: Intratumoral immunotherapy. OncoImmunology 2021, 10, 1984677. [Google Scholar] [CrossRef] [PubMed]
- Luzina, I.G.; Keegan, A.D.; Heller, N.M.; Rook, G.A.W.; Shea-Donohue, T.; Atamas, S.P. Regulation of inflammation by interleukin-4: A review of “alternatives”. J. Leukoc. Biol. 2012, 92, 753–764. [Google Scholar] [CrossRef] [PubMed]
- LaMarche, N.M.; Hegde, S.; Park, M.D.; Maier, B.B.; Troncoso, L.; Le Berichel, J.; Hamon, P.; Belabed, M.; Mattiuz, R.; Hennequin, C.; et al. An IL-4 signalling axis in bone marrow drives pro-tumorigenic myelopoiesis. Nature 2024, 625, 166–174. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Gong, C.; Mao, H.; Li, Z.; Fang, Z.; Chen, Q.; Lin, M.; Jiang, X.; Hu, Y.; Wang, W.; et al. E2F1/SP3/STAT6 axis is required for IL-4-induced epithelial-mesenchymal transition of colorectal cancer cells. Int. J. Oncol. 2018, 53, 567–578. [Google Scholar] [CrossRef] [PubMed]
- Horn, F.; Henze, C.; Heidrich, K. Interleukin-6 Signal Transduction and Lymphocyte Function. Immunobiology 2000, 202, 151–167. [Google Scholar] [CrossRef]
- Taga, T.; Kishimoto, T. Gp130 and the interleukin-6 family of cytokines. Annu. Rev. Immunol. 1997, 15, 797–819. [Google Scholar] [CrossRef] [PubMed]
- Rouhani, S.J.; Trujillo, J.A.; Pyzer, A.R.; Yu, J.; Fessler, J.; Cabanov, A.; Higgs, E.; Cron, K.; Zha, Y.; Lu, Y.; et al. Severe COVID-19 infection is associated with aberrant cytokine production by infected lung epithelial cells rather than by systemic immune dysfunction. Res. Sq. 2021, 3, 1083825. [Google Scholar] [CrossRef]
- Taniguchi, K.; Karin, M. IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Semin. Immunol. 2014, 26, 54–74. [Google Scholar] [CrossRef]
- Mace, T.A.; Shakya, R.; Pitarresi, J.R.; Swanson, B.; McQuinn, C.W.; Loftus, S.; Nordquist, E.; Cruz-Monserrate, Z.; Yu, L.; Young, G.; et al. IL-6 and PD-L1 antibody blockade combination therapy reduces tumour progression in murine models of pancreatic cancer. Gut 2018, 67, 320–332. [Google Scholar] [CrossRef]
- Laino, A.S.; Woods, D.; Vassallo, M.; Qian, X.; Tang, H.; Wind-Rotolo, M.; Weber, J. Serum interleukin-6 and C-reactive protein are associated with survival in melanoma patients receiving immune checkpoint inhibition. J. Immunother. Cancer 2020, 8, e000842. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, K.G.; Vrabel, M.R.; Mantooth, S.M.; Hopkins, J.J.; Wagner, E.S.; Gabaldon, T.A.; Zaharoff, D.A. Localized Interleukin-12 for Cancer Immunotherapy. Front. Immunol. 2020, 11, 575597. [Google Scholar] [CrossRef] [PubMed]
- Reschke, R.; Olson, D.J. Leveraging STING, Batf3 Dendritic Cells, CXCR3 Ligands, and Other Components Related to Innate Immunity to Induce A “Hot” Tumor Microenvironment that Is Responsive to Immunotherapy. Cancers 2022, 14, 2458. [Google Scholar] [CrossRef] [PubMed]
- Burton, J.D.; Bamford, R.N.; Peters, C.; Grant, A.J.; Kurys, G.; Goldman, C.K.; Brennan, J.; Roessler, E.; Waldmann, T.A. A lymphokine, provisionally designated interleukin T and produced by a human adult T-cell leukemia line, stimulates T-cell proliferation and the induction of lymphokine-activated killer cells. Proc. Natl. Acad. Sci. USA 1994, 91, 4935–4939. [Google Scholar] [CrossRef]
- Tagaya, Y.; Bamford, R.N.; DeFilippis, A.P.; Waldmann, T.A. IL-15: A pleiotropic cytokine with diverse receptor/signaling pathways whose expression is controlled at multiple levels. Immunity 1996, 4, 329–336. [Google Scholar] [CrossRef] [PubMed]
- Waldmann, T.A. The biology of interleukin-2 and interleukin-15: Implications for cancer therapy and vaccine design. Nat. Rev. Immunol. 2006, 6, 595–601. [Google Scholar] [CrossRef]
- Berger, A.; Colpitts, S.J.; Seabrook, M.S.S.; Furlonger, C.L.; Bendix, M.B.; Moreau, J.M.; McKillop, W.M.; Medin, J.A.; Paige, C.J. Interleukin-15 in cancer immunotherapy: IL-15 receptor complex versus soluble IL-15 in a cancer cell-delivered murine leukemia model. J. Immunother. Cancer 2019, 7, 355. [Google Scholar] [CrossRef]
- Wang, X.; Zhao, X.-Y. Transcription Factors Associated with IL-15 Cytokine Signaling during NK Cell Development. Front. Immunol. 2021, 12, 610789. [Google Scholar] [CrossRef]
- Dubsky, P.; Saito, H.; Leogier, M.; Dantin, C.; Connolly, J.E.; Banchereau, J.; Palucka, A.K. IL-15-induced human DC efficiently prime melanoma-specific naive CD8+ T cells to differentiate into CTL. Eur. J. Immunol. 2007, 37, 1678–1690. [Google Scholar] [CrossRef]
- Marrero, B.; Shirley, S.; Heller, R. Delivery of Interleukin-15 to B16 Melanoma by Electroporation Leads to Tumor Regression and Long-term Survival. Technol. Cancer Res. Treat. 2014, 13, 551–560. [Google Scholar] [CrossRef] [PubMed]
- Robinson, T.O.; Schluns, K.S. The potential and promise of IL-15 in immuno-oncogenic therapies. Immunol. Lett. 2017, 190, 159–168. [Google Scholar] [CrossRef] [PubMed]
- Knudson, K.M.; Hodge, J.W.; Schlom, J.; Gameiro, S.R. Rationale for IL-15 superagonists in cancer immunotherapy. Expert Opin. Biol. Ther. 2020, 20, 705–709. [Google Scholar] [CrossRef] [PubMed]
- Kuwabara, T.; Ishikawa, F.; Kondo, M.; Kakiuchi, T. The Role of IL-17 and Related Cytokines in Inflammatory Autoimmune Diseases. Mediat. Inflamm. 2017, 2017, 3908061. [Google Scholar] [CrossRef] [PubMed]
- Khan, D.; Ansar Ahmed, S. Regulation of IL-17 in autoimmune diseases by transcriptional factors and microRNAs. Front. Genet. 2015, 6, 236. [Google Scholar] [CrossRef] [PubMed]
- Váraljai, R.; Zimmer, L.; Al-Matary, Y.; Kaptein, P.; Albrecht, L.J.; Shannan, B.; Brase, J.C.; Gusenleitner, D.; Amaral, T.; Wyss, N.; et al. Interleukin 17 signaling supports clinical benefit of dual CTLA-4 and PD-1 checkpoint inhibition in melanoma. Nat. Cancer 2023, 4, 1292–1308. [Google Scholar] [CrossRef] [PubMed]
- Martin-Orozco, N.; Muranski, P.; Chung, Y.; Yang, X.O.; Yamazaki, T.; Lu, S.; Hwu, P.; Restifo, N.P.; Overwijk, W.W.; Dong, C. T helper 17 cells promote cytotoxic T cell activation in tumor immunity. Immunity 2009, 31, 787–798. [Google Scholar] [CrossRef] [PubMed]
- Kuen, D.-S.; Kim, B.-S.; Chung, Y. IL-17-Producing Cells in Tumor Immunity: Friends or Foes? Immune Netw. 2020, 20, e6. [Google Scholar] [CrossRef] [PubMed]
- Ito, T. Recent advances in the pathogenesis of autoimmune hair loss disease alopecia areata. Clin. Dev. Immunol. 2013, 2013, 348546. [Google Scholar] [CrossRef]
- Boniface, K.; Jacquemin, C.; Darrigade, A.-S.; Dessarthe, B.; Martins, C.; Boukhedouni, N.; Vernisse, C.; Grasseau, A.; Thiolat, D.; Rambert, J.; et al. Vitiligo Skin Is Imprinted with Resident Memory CD8 T Cells Expressing CXCR3. J. Investig. Dermatol. 2018, 138, 355–364. [Google Scholar] [CrossRef]
- Loos, T.; Dekeyzer, L.; Struyf, S.; Schutyser, E.; Gijsbers, K.; Gouwy, M.; Fraeyman, A.; Put, W.; Ronsse, I.; Grillet, B.; et al. TLR ligands and cytokines induce CXCR3 ligands in endothelial cells: Enhanced CXCL9 in autoimmune arthritis. Lab. Investig. 2006, 86, 902–916. [Google Scholar] [CrossRef] [PubMed]
- Antonelli, A.; Ferrari, S.M.; Corrado, A.; Ferrannini, E.; Fallahi, P. CXCR3, CXCL10 and type 1 diabetes. Cytokine Growth Factor. Rev. 2014, 25, 57–65. [Google Scholar] [CrossRef] [PubMed]
- Reschke, R.; Gussek, P.; Boldt, A.; Sack, U.; Köhl, U.; Lordick, F.; Gora, T.; Kreuz, M.; Reiche, K.; Simon, J.-C.; et al. Distinct Immune Signatures Indicative of Treatment Response and Immune-Related Adverse Events in Melanoma Patients under Immune Checkpoint Inhibitor Therapy. Int. J. Mol. Sci. 2021, 22, 8017. [Google Scholar] [CrossRef] [PubMed]
- Fukumoto, N.; Shimaoka, T.; Fujimura, H.; Sakoda, S.; Tanaka, M.; Kita, T.; Yonehara, S. Critical roles of CXC chemokine ligand 16/scavenger receptor that binds phosphatidylserine and oxidized lipoprotein in the pathogenesis of both acute and adoptive transfer experimental autoimmune encephalomyelitis. J. Immunol. 2004, 173, 1620–1627. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.J.; Sun, H.J.; Song, Y.S.; Yoo, S.-K.; Kim, Y.A.; Seo, J.-S.; Park, Y.J.; Cho, S.W. CXCL16 positively correlated with M2-macrophage infiltration, enhanced angiogenesis, and poor prognosis in thyroid cancer. Sci. Rep. 2019, 9, 13288. [Google Scholar] [CrossRef] [PubMed]
- Hu, W.; Liu, Y.; Zhou, W.; Si, L.; Ren, L. CXCL16 and CXCR6 are coexpressed in human lung cancer in vivo and mediate the invasion of lung cancer cell lines in vitro. PLoS ONE 2014, 9, e99056. [Google Scholar] [CrossRef] [PubMed]
- Allard-Chamard, H.; Mishra, H.K.; Nandi, M.; Mayhue, M.; Menendez, A.; Ilangumaran, S.; Ramanathan, S. Interleukin-15 in autoimmunity. Cytokine 2020, 136, 155258. [Google Scholar] [CrossRef] [PubMed]
- Lin, S.; Huang, G.; Xiao, Y.; Sun, W.; Jiang, Y.; Deng, Q.; Peng, M.; Wei, X.; Ye, W.; Li, B.; et al. CD215+ Myeloid Cells Respond to Interleukin 15 Stimulation and Promote Tumor Progression. Front. Immunol. 2017, 8, 1713. [Google Scholar] [CrossRef] [PubMed]
- Schlecker, E.; Stojanovic, A.; Eisen, C.; Quack, C.; Falk, C.S.; Umansky, V.; Cerwenka, A.; Xu, M.; Hadinoto, V.; Appanna, R.; et al. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J. Immunol. 2012, 189, 5602–5611. [Google Scholar] [CrossRef]
- Li, L.; Liu, Y.-D.; Zhan, Y.-T.; Zhu, Y.-H.; Li, Y.; Xie, D.; Guan, X. High levels of CCL2 or CCL4 in the tumor microenvironment predict unfavorable survival in lung adenocarcinoma. Thorac. Cancer 2018, 9, 775–784. [Google Scholar] [CrossRef]
- Hua, F.; Tian, Y. CCL4 promotes the cell proliferation, invasion and migration of endometrial carcinoma by targeting the VEGF-A signal pathway. Int. J. Clin. Exp. Pathol. 2017, 10, 11288–11299. [Google Scholar] [PubMed]
- Zhang, T.; Somasundaram, R.; Berencsi, K.; Caputo, L.; Gimotty, P.; Rani, P.; Guerry, D.; Swoboda, R.; Herlyn, D. Migration of cytotoxic T lymphocytes toward melanoma cells in three-dimensional organotypic culture is dependent on CCL2 and CCR4. Eur. J. Immunol. 2006, 36, 457–467. [Google Scholar] [CrossRef] [PubMed]
- Hu, K.; Xiong, J.; Ji, K.; Sun, H.; Wang, J.; Liu, H. Recombined CC chemokine ligand 2 into B16 cells induces production of Th2-dominant [correction of dominanted] cytokines and inhibits melanoma metastasis. Immunol. Lett. 2007, 113, 19–28. [Google Scholar] [CrossRef] [PubMed]
- Reschke, R.; Gussek, P.; Ziemer, M. Identifying High-Risk Tumors within AJCC Stage IB–III Melanomas Using a Seven-Marker Immunohistochemical Signature. Cancers 2021, 13, 2902. [Google Scholar] [CrossRef] [PubMed]
- Meyer, S.; Buser, L.; Haferkamp, S.; Berneburg, M.; Maisch, T.; Klinkhammer-Schalke, M.; Pauer, A.; Vogt, T.; Garbe, C. Identification of high-risk patients with a seven-biomarker prognostic signature for adjuvant treatment trial recruitment in American Joint Committee on Cancer v8 stage I–IIA cutaneous melanoma. Eur. J. Cancer 2023, 182, 77–86. [Google Scholar] [CrossRef] [PubMed]
- Reschke, R.; Dumann, K.; Ziemer, M. Risk Stratification and Clinical Characteristics of Patients with Late Recurrence of Melanoma (>10 Years). J. Clin. Med. 2022, 11, 2026. [Google Scholar] [CrossRef]
- Feldman, A.L.; Friedl, J.; Lans, T.E.; Libutti, S.K.; Lorang, D.; Miller, M.S.; Turner, E.M.; Hewitt, S.M.; Alexander, H.R. Retroviral gene transfer of interferon-inducible protein 10 inhibits growth of human melanoma xenografts. Int. J. Cancer 2002, 99, 149–153. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 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
Reschke, R.; Enk, A.H.; Hassel, J.C. Chemokines and Cytokines in Immunotherapy of Melanoma and Other Tumors: From Biomarkers to Therapeutic Targets. Int. J. Mol. Sci. 2024, 25, 6532. https://doi.org/10.3390/ijms25126532
Reschke R, Enk AH, Hassel JC. Chemokines and Cytokines in Immunotherapy of Melanoma and Other Tumors: From Biomarkers to Therapeutic Targets. International Journal of Molecular Sciences. 2024; 25(12):6532. https://doi.org/10.3390/ijms25126532
Chicago/Turabian StyleReschke, Robin, Alexander H. Enk, and Jessica C. Hassel. 2024. "Chemokines and Cytokines in Immunotherapy of Melanoma and Other Tumors: From Biomarkers to Therapeutic Targets" International Journal of Molecular Sciences 25, no. 12: 6532. https://doi.org/10.3390/ijms25126532