Immune Cell Migration to Cancer
Abstract
:1. Basic Concepts of Immune Cell Movement
1.1. Immune Cells Employ Varying Modes of Migration
1.2. Adhesion Molecules Are Required for Immune Cell Migration
1.2.1. Integrin and Integrin Ligands
1.2.2. Selectin and Selectin Ligands
1.3. Migration Patterns of Specific Immune Cells
2. Adaptive Immune Cell Migration to Cancer
2.1. T Cell Migration to Cancer
2.1.1. T Cell Migration to Cancer in the Perspective of Integrins and Cell Adhesion Molecules
2.1.2. T Cell Migration to Cancer in the Perspective of Chemokines
CCL19, CCL21
CXCL9, CXCL10, CXCL11
CXCL12
CXCL13
Other Chemokines/Receptors
2.2. Migration of NK Cells and B Cells to Cancer
2.3. Other Mediators That Regulate Adaptive Immune Cell Migration to Cancer
3. Innate Immune Cell Migration to Cancer
3.1. TME-Derived Chemokines
3.1.1. CCL2
3.1.2. CXCL8, CXCL1, CXCL2, CXCL5
3.1.3. Other Chemokines
3.2. Non-Chemokine Mediators to Induce Tumor Infiltration of Myeloid Cells
3.2.1. Complement
3.2.2. VEGF
3.2.3. Leukotrienes (LTs)
3.2.4. Damage-Associated Molecular Patterns (DAMPs)
3.2.5. Platelet and Coagulation
3.2.6. Hypoxia and Acidosis
3.2.7. ICAM-1 and VCAM-1
4. Therapeutic Implications
5. Conclusions
Cancer | Functions | Outcome/Mechanism | |
---|---|---|---|
Cell adhesion molecules, Receptors | |||
ICAM-1/ICAM-2/ICAM-3, αLβ2 or αMβ2 | Colorectal cancer, Melanoma, Breast cancer, Pediatric osteosarcoma, Pancreatic cancer, Gastric cancer | T cell migration and adhesion | Antitumor/recruitment of antitumoral T cells to tumor microenvironment (TME) [12,13,14,15,53,54,55,56,63,64,65] |
Neutrophil/PMN-MDSC migration and adhesion | Protumor/recruitment of immunosuppressive neutrophils/PMN-MDSCs to TME [16,25,26,27,28], metastasis [406,407,408] Antitumor/recruitment and retention of tumoricidal neutrophils to/in TME [412,413] | ||
Monocyte/macrophage/M-MDSC migration and adhesion | Protumor/recruitment and retention of immunosuppressive monocytes/macrophages/M-MDSCs to/in TME [16,29] Antitumor/recruitment and retention of antitumor monocytes/macrophages to/in TME [413,414] | ||
VCAM-1, α4β1 | Renal cell carcinoma, Breast cancer | T cell migration | Protumor/disruption of T cell binding to tumor cells [60] |
Monocyte/macrophage adhesion/retention to tumor cells | Protumor/tumor survival at metastatic sites [409], bone metastasis by osteoclastogenesis [411] | ||
Selectins, Selectin ligands | Breast cancer, Melanoma | T cell trafficking to lymph nodes (LNs) and tumors, Tumor cell interaction with endothelium | Antitumor/generation of antitumor T cells in LNs [66], T cell infiltration into tumors [73,182] |
Protumor/suppression of T cell generation in LNs by L-selectin shedding/downregulation of T cells [70,71,72], Tumor cell extravasation [67,68] | |||
Chemokines, Chemokine receptors | |||
CCL19/21, CCR7 or CXCR7 | Lung carcinoma, Breast cancer, Cervical cancer, Gastric cancer | T cell migration to/in LNs and TME | Protumor/metastasis of CCR7+ tumors [84,85,86,87], Treg migration to TME [89], T cell sequestration by TME remodeling [91] Antitumor/generation and tumor infiltration of cytotoxic T cells [38,81], chemokine scavenge by CXCR7 [119] |
CXCL9/10/11, CXCR3 or CXCR7 | Colorectal carcinoma, Melanoma, Fibrosarcoma, Ovarian cancer | T cell migration to TME | Protumor/chemokine scavenge by CXCR7 [104,105], tumor infiltration of CXCR3+ Treg cells [106] Antitumor/activation and tumor infiltration of CXCR3+ T cells [94,95,96,97,98,99,100,101,102] |
CXCL12, CXCR4 or CXCR7 | Melanoma, Hepatocellular carcinoma, Ovarian cancer | T cell migration and localization to/in TME | Protumor/sequestration of T cells in TME [107], inhibition of tumor infiltration of T cells [108], recruitment of Treg cells and TAMs [106,110], chemokine scavenge by CXCR7 [120] Antitumor/tumor infiltration of T cells [109], retention and microlocalization of CD8+ T cells in tumors [111] |
CCL2, CCR2 | Lung carcinoma, Melanoma, Glioma, Inflammatory breast cancer | Monocyte/macrophage mobilization and localization to/in TME | Protumor/suppression of CD8+ T cells, and promotion of neovascularization and metastasis by M-MDSCs recruited to tumors and premetastatic sites [192,199,210,211,212,213,214,215,216,217,442,443,444,445,475] Antitumor/Tumor entrainment of neutrophils – tumor killing [412], antitumor monocyte recruitment to premetastatic lung [413], Recruitment of CCR2+ T cells and antigen presenting cells [449,450] |
CXCL1/2/5/8, CXCR2 | Melanoma, Gastric cancer, Lung cancer, Fibrosarcoma, Papilloma, Colon cancer | Neutrophil mobilization and localization to/in TME and CTCs | Protumor/suppression of CD8+ T cell activities and tissue infiltration, suppression of tumor cell senescence, promotion of tumor genomic instability, neovascularization, invasion, metastasis, and EMT by neutrophils/PMN-MDSCs recruited to tumors and premetastatic sites [187,192,202,212,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253,254,255,256,257,258,259,467,470,475], CTC-myeloid cell cluster [388,408] |
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Guak, H.; Krawczyk, C.M. Implications of cellular metabolism for immune cell migration. Immunology 2020, 161, 200–208. [Google Scholar] [CrossRef] [PubMed]
- Simula, L.; Fumagalli, M.; Vimeux, L.; Rajnpreht, I.; Icard, P.; Birsen, G.; An, D.; Pendino, F.; Rouault, A.; Bercovici, N.; et al. Mitochondrial metabolism sustains CD8(+) T cell migration for an efficient infiltration into solid tumors. Nat. Commun. 2024, 15, 2203. [Google Scholar] [CrossRef] [PubMed]
- SenGupta, S.; Parent, C.A.; Bear, J.E. The principles of directed cell migration. Nat. Rev. Mol. Cell. Biol. 2021, 22, 529–547. [Google Scholar] [CrossRef] [PubMed]
- Yamada, K.M.; Sixt, M. Mechanisms of 3D cell migration. Nat. Rev. Mol. Cell. Biol. 2019, 20, 738–752. [Google Scholar] [CrossRef] [PubMed]
- Moreau, H.D.; Piel, M.; Voituriez, R.; Lennon-Dumenil, A.M. Integrating Physical and Molecular Insights on Immune Cell Migration. Trends Immunol. 2018, 39, 632–643. [Google Scholar] [CrossRef] [PubMed]
- Merino-Casallo, F.; Gomez-Benito, M.J.; Hervas-Raluy, S.; Garcia-Aznar, J.M. Unravelling cell migration: Defining movement from the cell surface. Cell Adh. Migr. 2022, 16, 25–64. [Google Scholar] [CrossRef] [PubMed]
- Fowell, D.J.; Kim, M. The spatio-temporal control of effector T cell migration. Nat. Rev. Immunol. 2021, 21, 582–596. [Google Scholar] [CrossRef]
- Petrie, R.J.; Gavara, N.; Chadwick, R.S.; Yamada, K.M. Nonpolarized signaling reveals two distinct modes of 3D cell migration. J. Cell Biol. 2012, 197, 439–455. [Google Scholar] [CrossRef] [PubMed]
- Takagi, J.; Petre, B.M.; Walz, T.; Springer, T.A. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell 2002, 110, 599–611. [Google Scholar] [CrossRef]
- Tvaroska, I.; Kozmon, S.; Kona, J. Molecular Modeling Insights into the Structure and Behavior of Integrins: A Review. Cells 2023, 12, 324. [Google Scholar] [CrossRef]
- Smith, A.; Stanley, P.; Jones, K.; Svensson, L.; McDowall, A.; Hogg, N. The role of the integrin LFA-1 in T-lymphocyte migration. Immunol. Rev. 2007, 218, 135–146. [Google Scholar] [CrossRef]
- Zhang, Y.; Wang, H. Integrin signalling and function in immune cells. Immunology 2012, 135, 268–275. [Google Scholar] [CrossRef] [PubMed]
- Walling, B.L.; Kim, M. LFA-1 in T Cell Migration and Differentiation. Front. Immunol. 2018, 9, 952. [Google Scholar] [CrossRef] [PubMed]
- Bui, T.M.; Wiesolek, H.L.; Sumagin, R. ICAM-1: A master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J. Leukoc. Biol. 2020, 108, 787–799. [Google Scholar] [CrossRef] [PubMed]
- Guerra-Espinosa, C.; Jimenez-Fernandez, M.; Sanchez-Madrid, F.; Serrador, J.M. ICAMs in Immunity, Intercellular Adhesion and Communication. Cells 2024, 13, 339. [Google Scholar] [CrossRef] [PubMed]
- Hidalgo, A.; Chang, J.; Jang, J.E.; Peired, A.J.; Chiang, E.Y.; Frenette, P.S. Heterotypic interactions enabled by polarized neutrophil microdomains mediate thromboinflammatory injury. Nat. Med. 2009, 15, 384–391. [Google Scholar] [CrossRef] [PubMed]
- Lim, K.; Hyun, Y.M.; Lambert-Emo, K.; Topham, D.J.; Kim, M. Visualization of integrin Mac-1 in vivo. J. Immunol. Methods 2015, 426, 120–127. [Google Scholar] [CrossRef] [PubMed]
- Phillipson, M.; Heit, B.; Colarusso, P.; Liu, L.; Ballantyne, C.M.; Kubes, P. Intraluminal crawling of neutrophils to emigration sites: A molecularly distinct process from adhesion in the recruitment cascade. J. Exp. Med. 2006, 203, 2569–2575. [Google Scholar] [CrossRef] [PubMed]
- Borsig, L. Selectins in cancer immunity. Glycobiology 2018, 28, 648–655. [Google Scholar] [CrossRef] [PubMed]
- Evans, S.S.; Repasky, E.A.; Fisher, D.T. Fever and the thermal regulation of immunity: The immune system feels the heat. Nat. Rev. Immunol. 2015, 15, 335–349. [Google Scholar] [CrossRef] [PubMed]
- Ebel, M.E.; Awe, O.; Kaplan, M.H.; Kansas, G.S. Diverse inflammatory cytokines induce selectin ligand expression on murine CD4 T cells via p38alpha MAPK. J. Immunol. 2015, 194, 5781–5788. [Google Scholar] [CrossRef] [PubMed]
- Lam, F.W.; Burns, A.R.; Smith, C.W.; Rumbaut, R.E. Platelets enhance neutrophil transendothelial migration via P-selectin glycoprotein ligand-1. Am. J. Physiol. Heart. Circ. Physiol. 2011, 300, H468–H475. [Google Scholar] [CrossRef] [PubMed]
- Ali, A.J.; Abuelela, A.F.; Merzaban, J.S. An Analysis of Trafficking Receptors Shows that CD44 and P-Selectin Glycoprotein Ligand-1 Collectively Control the Migration of Activated Human T-Cells. Front. Immunol. 2017, 8, 492. [Google Scholar] [CrossRef] [PubMed]
- Witz, I.P. The selectin-selectin ligand axis in tumor progression. Cancer Metastasis Rev. 2008, 27, 19–30. [Google Scholar] [CrossRef] [PubMed]
- Filippi, M.D. Neutrophil transendothelial migration: Updates and new perspectives. Blood 2019, 133, 2149–2158. [Google Scholar] [CrossRef] [PubMed]
- Liew, P.X.; Kubes, P. The Neutrophil’s Role During Health and Disease. Physiol. Rev. 2019, 99, 1223–1248. [Google Scholar] [CrossRef] [PubMed]
- Margraf, A.; Lowell, C.A.; Zarbock, A. Neutrophils in acute inflammation: Current concepts and translational implications. Blood 2022, 139, 2130–2144. [Google Scholar] [CrossRef] [PubMed]
- Summers, C.; Rankin, S.M.; Condliffe, A.M.; Singh, N.; Peters, A.M.; Chilvers, E.R. Neutrophil kinetics in health and disease. Trends Immunol. 2010, 31, 318–324. [Google Scholar] [CrossRef] [PubMed]
- Gerhardt, T.; Ley, K. Monocyte trafficking across the vessel wall. Cardiovasc. Res. 2015, 107, 321–330. [Google Scholar] [CrossRef] [PubMed]
- Karlmark, K.R.; Tacke, F.; Dunay, I.R. Monocytes in health and disease—Minireview. Eur. J. Microbiol. Immunol. 2012, 2, 97–102. [Google Scholar] [CrossRef] [PubMed]
- Worbs, T.; Hammerschmidt, S.I.; Forster, R. Dendritic cell migration in health and disease. Nat. Rev. Immunol. 2017, 17, 30–48. [Google Scholar] [CrossRef] [PubMed]
- Castellino, F.; Huang, A.Y.; Altan-Bonnet, G.; Stoll, S.; Scheinecker, C.; Germain, R.N. Chemokines enhance immunity by guiding naive CD8+ T cells to sites of CD4+ T cell-dendritic cell interaction. Nature 2006, 440, 890–895. [Google Scholar] [CrossRef] [PubMed]
- van Beek, J.J.P.; Flórez-Grau, G.; Gorris, M.A.J.; Mathan, T.S.M.; Schreibelt, G.; Bol, K.F.; Textor, J.; de Vries, I.J.M. Human pDCs Are Superior to cDC2s in Attracting Cytolytic Lymphocytes in Melanoma Patients Receiving DC Vaccination. Cell Rep. 2020, 30, 1027–1038. [Google Scholar] [CrossRef] [PubMed]
- Overstreet, M.G.; Gaylo, A.; Angermann, B.R.; Hughson, A.; Hyun, Y.M.; Lambert, K.; Acharya, M.; Billroth-Maclurg, A.C.; Rosenberg, A.F.; Topham, D.J.; et al. Inflammation-induced interstitial migration of effector CD4(+) T cells is dependent on integrin alphaV. Nat. Immunol. 2013, 14, 949–958. [Google Scholar] [CrossRef] [PubMed]
- Petrovic, A.; Alpdogan, O.; Willis, L.M.; Eng, J.M.; Greenberg, A.S.; Kappel, B.J.; Liu, C.; Murphy, G.J.; Heller, G.; van den Brink, M.R. LPAM (alpha 4 beta 7 integrin) is an important homing integrin on alloreactive T cells in the development of intestinal graft-versus-host disease. Blood 2004, 103, 1542–1547. [Google Scholar] [CrossRef] [PubMed]
- Pribila, J.T.; Quale, A.C.; Mueller, K.L.; Shimizu, Y. Integrins and T cell-mediated immunity. Annu. Rev. Immunol. 2004, 22, 157–180. [Google Scholar] [CrossRef] [PubMed]
- Topham, D.J.; Reilly, E.C. Tissue-Resident Memory CD8(+) T Cells: From Phenotype to Function. Front. Immunol. 2018, 9, 515. [Google Scholar] [CrossRef] [PubMed]
- Luther, S.A.; Bidgol, A.; Hargreaves, D.C.; Schmidt, A.; Xu, Y.; Paniyadi, J.; Matloubian, M.; Cyster, J.G. Differing activities of homeostatic chemokines CCL19, CCL21, and CXCL12 in lymphocyte and dendritic cell recruitment and lymphoid neogenesis. J. Immunol. 2002, 169, 424–433. [Google Scholar] [CrossRef] [PubMed]
- Matloubian, M.; Lo, C.G.; Cinamon, G.; Lesneski, M.J.; Xu, Y.; Brinkmann, V.; Allende, M.L.; Proia, R.L.; Cyster, J.G. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 2004, 427, 355–360. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Janeway, C. Immunobiology: The Immune System in Health and Disease, 6th ed.; Garland Science: New York, NY, USA, 2005; Volume 2, p. 154. [Google Scholar]
- Angeli, V.; Ginhoux, F.; Llodra, J.; Quemeneur, L.; Frenette, P.S.; Skobe, M.; Jessberger, R.; Merad, M.; Randolph, G.J. B cell-driven lymphangiogenesis in inflamed lymph nodes enhances dendritic cell mobilization. Immunity 2006, 24, 203–215. [Google Scholar] [CrossRef] [PubMed]
- Liao, S.; Ruddle, N.H. Synchrony of high endothelial venules and lymphatic vessels revealed by immunization. J. Immunol. 2006, 177, 3369–3379. [Google Scholar] [CrossRef] [PubMed]
- Randolph, G.J.; Angeli, V.; Swartz, M.A. Dendritic-cell trafficking to lymph nodes through lymphatic vessels. Nat. Rev. Immunol. 2005, 5, 617–628. [Google Scholar] [CrossRef] [PubMed]
- von Andrian, U.H.; M’Rini, C. In situ analysis of lymphocyte migration to lymph nodes. Cell Adhes. Commun. 1998, 6, 85–96. [Google Scholar] [CrossRef] [PubMed]
- Bannard, O.; Cyster, J.G. Germinal centers: Programmed for affinity maturation and antibody diversification. Curr. Opin. Immunol. 2017, 45, 21–30. [Google Scholar] [CrossRef] [PubMed]
- Cyster, J.G.; Allen, C.D.C. B Cell Responses: Cell Interaction Dynamics and Decisions. Cell 2019, 177, 524–540. [Google Scholar] [CrossRef] [PubMed]
- Denton, A.E.; Linterman, M.A. Stromal networking: Cellular connections in the germinal centre. Curr. Opin. Immunol. 2017, 45, 103–111. [Google Scholar] [CrossRef] [PubMed]
- Dhenni, R.; Phan, T.G. The geography of memory B cell reactivation in vaccine-induced immunity and in autoimmune disease relapses. Immunol. Rev. 2020, 296, 62–86. [Google Scholar] [CrossRef] [PubMed]
- Weisel, F.J.; Zuccarino-Catania, G.V.; Chikina, M.; Shlomchik, M.J. A Temporal Switch in the Germinal Center Determines Differential Output of Memory B and Plasma Cells. Immunity 2016, 44, 116–130. [Google Scholar] [CrossRef] [PubMed]
- Leon, B.; Ballesteros-Tato, A.; Misra, R.S.; Wojciechowski, W.; Lund, F.E. Unraveling effector functions of B cells during infection: The hidden world beyond antibody production. Infect. Disord. Drug Targets 2012, 12, 213–221. [Google Scholar] [CrossRef]
- Van Meerhaeghe, T.; Neel, A.; Brouard, S.; Degauque, N. Regulation of CD8 T cell by B-cells: A narrative review. Front. Immunol. 2023, 14, 1125605. [Google Scholar] [CrossRef]
- Fisher, D.T.; Chen, Q.; Skitzki, J.J.; Muhitch, J.B.; Zhou, L.; Appenheimer, M.M.; Vardam, T.D.; Weis, E.L.; Passanese, J.; Wang, W.C.; et al. IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. J. Clin. Investig. 2011, 121, 3846–3859. [Google Scholar] [CrossRef] [PubMed]
- Harjunpaa, H.; Llort Asens, M.; Guenther, C.; Fagerholm, S.C. Cell Adhesion Molecules and Their Roles and Regulation in the Immune and Tumor Microenvironment. Front. Immunol. 2019, 10, 1078. [Google Scholar] [CrossRef] [PubMed]
- Kantari-Mimoun, C.; Barrin, S.; Vimeux, L.; Haghiri, S.; Gervais, C.; Joaquina, S.; Mittelstaet, J.; Mockel-Tenbrinck, N.; Kinkhabwala, A.; Damotte, D.; et al. CAR T-cell Entry into Tumor Islets Is a Two-Step Process Dependent on IFNgamma and ICAM-1. Cancer Immunol. Res. 2021, 9, 1425–1438. [Google Scholar] [CrossRef] [PubMed]
- Hickman, A.; Koetsier, J.; Kurtanich, T.; Nielsen, M.C.; Winn, G.; Wang, Y.; Bentebibel, S.E.; Shi, L.; Punt, S.; Williams, L.; et al. LFA-1 activation enriches tumor-specific T cells in a cold tumor model and synergizes with CTLA-4 blockade. J. Clin. Investig. 2022, 132, e154152. [Google Scholar] [CrossRef] [PubMed]
- Mirlekar, B.; Pylayeva-Gupta, Y. IL-12 Family Cytokines in Cancer and Immunotherapy. Cancers 2021, 13, 167. [Google Scholar] [CrossRef] [PubMed]
- He, R.; Hou, S.; Liu, C.; Zhang, A.; Bai, Q.; Han, M.; Yang, Y.; Wei, G.; Shen, T.; Yang, X.; et al. Follicular CXCR5- expressing CD8(+) T cells curtail chronic viral infection. Nature 2016, 537, 412–428. [Google Scholar] [CrossRef] [PubMed]
- Philip, M.; Fairchild, L.; Sun, L.; Horste, E.L.; Camara, S.; Shakiba, M.; Scott, A.C.; Viale, A.; Lauer, P.; Merghoub, T.; et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 2017, 545, 452–456. [Google Scholar] [CrossRef] [PubMed]
- Wu, T.C. The role of vascular cell adhesion molecule-1 in tumor immune evasion. Cancer Res. 2007, 67, 6003–6006. [Google Scholar] [CrossRef]
- VanHeyst, K.A.; Choi, S.H.; Kingsley, D.T.; Huang, A.Y. Ectopic Tumor VCAM-1 Expression in Cancer Metastasis and Therapy Resistance. Cells 2022, 11, 3922. [Google Scholar] [CrossRef]
- Chen, Q.; Massague, J. Molecular pathways: VCAM-1 as a potential therapeutic target in metastasis. Clin. Cancer Res. 2012, 18, 5520–5525. [Google Scholar] [CrossRef]
- Hiraoka, N.; Yamazaki-Itoh, R.; Ino, Y.; Mizuguchi, Y.; Yamada, T.; Hirohashi, S.; Kanai, Y. CXCL17 and ICAM2 are associated with a potential anti-tumor immune response in early intraepithelial stages of human pancreatic carcinogenesis. Gastroenterology 2011, 140, 310–321. [Google Scholar] [CrossRef] [PubMed]
- Tang, X.; Huang, J.; Jiang, Y.; Qiu, J.; Li, T.; Li, W.; Chen, Z.; Huang, Z.; Yu, X.; Yang, T.; et al. Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis. J. Transl. Med. 2023, 21, 670. [Google Scholar] [CrossRef] [PubMed]
- da Silva, S.F.; Murta, E.F.; Michelin, M.A. ICAM2 is related to good prognosis in dendritic cell immunotherapy for cancer. Immunotherapy 2024, 16, 173–185. [Google Scholar] [CrossRef] [PubMed]
- Sallusto, F.; Lenig, D.; Forster, R.; Lipp, M.; Lanzavecchia, A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999, 401, 708–712. [Google Scholar] [CrossRef] [PubMed]
- Borsig, L.; Wong, R.; Hynes, R.O.; Varki, N.M.; Varki, A. Synergistic effects of L- and P-selectin in facilitating tumor metastasis can involve non-mucin ligands and implicate leukocytes as enhancers of metastasis. Proc. Natl. Acad. Sci. USA 2002, 99, 2193–2198. [Google Scholar] [CrossRef] [PubMed]
- Hauselmann, I.; Roblek, M.; Protsyuk, D.; Huck, V.; Knopfova, L.; Grassle, S.; Bauer, A.T.; Schneider, S.W.; Borsig, L. Monocyte Induction of E-Selectin-Mediated Endothelial Activation Releases VE-Cadherin Junctions to Promote Tumor Cell Extravasation in the Metastasis Cascade. Cancer Res. 2016, 76, 5302–5312. [Google Scholar] [CrossRef] [PubMed]
- Laubli, H.; Borsig, L. Selectins promote tumor metastasis. Semin. Cancer Biol. 2010, 20, 169–177. [Google Scholar] [CrossRef] [PubMed]
- Hanson, E.M.; Clements, V.K.; Sinha, P.; Ilkovitch, D.; Ostrand-Rosenberg, S. Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J. Immunol. 2009, 183, 937–944. [Google Scholar] [CrossRef] [PubMed]
- Ku, A.W.; Muhitch, J.B.; Powers, C.A.; Diehl, M.; Kim, M.; Fisher, D.T.; Sharda, A.P.; Clements, V.K.; O’Loughlin, K.; Minderman, H.; et al. Tumor-induced MDSC act via remote control to inhibit L-selectin-dependent adaptive immunity in lymph nodes. eLife 2016, 5, e17375. [Google Scholar] [CrossRef] [PubMed]
- Parker, K.H.; Sinha, P.; Horn, L.A.; Clements, V.K.; Yang, H.; Li, J.; Tracey, K.J.; Ostrand-Rosenberg, S. HMGB1 enhances immune suppression by facilitating the differentiation and suppressive activity of myeloid-derived suppressor cells. Cancer Res. 2014, 74, 5723–5733. [Google Scholar] [CrossRef] [PubMed]
- Watson, H.A.; Durairaj, R.R.P.; Ohme, J.; Alatsatianos, M.; Almutairi, H.; Mohammed, R.N.; Vigar, M.; Reed, S.G.; Paisey, S.J.; Marshall, C.; et al. L-Selectin Enhanced T Cells Improve the Efficacy of Cancer Immunotherapy. Front. Immunol. 2019, 10, 1321. [Google Scholar] [CrossRef] [PubMed]
- Yayan, J.; Franke, K.J.; Berger, M.; Windisch, W.; Rasche, K. Adhesion, metastasis, and inhibition of cancer cells: A comprehensive review. Mol. Biol. Rep. 2024, 51, 165. [Google Scholar] [CrossRef] [PubMed]
- Janiszewska, M.; Primi, M.C.; Izard, T. Cell adhesion in cancer: Beyond the migration of single cells. J. Biol. Chem. 2020, 295, 2495–2505. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Caron, C.; Chan, Y.Y.; Lee, C.K.; Xu, X.; Zhang, J.; Masubuchi, T.; Wu, C.; Bui, J.D.; Hui, E. cis-B7:CD28 interactions at invaginated synaptic membranes provide CD28 co-stimulation and promote CD8(+) T cell function and anti-tumor immunity. Immunity 2023, 56, 1187–1203. [Google Scholar] [CrossRef] [PubMed]
- Cho, E.; Singh, R.; Han, C.; Kim, S.H.; Kim, K.H.; Park, B.M.; Shin, D.H.; Han, S.; Kim, Y.H.; Kwon, B.S.; et al. 4-1BB-4-1BBL cis-interaction contributes to the survival of self-reactive CD8(+) T cell. Cell. Mol. Immunol. 2023, 20, 1077–1080. [Google Scholar] [CrossRef] [PubMed]
- Baker, C.M.; Comrie, W.A.; Hyun, Y.M.; Chung, H.L.; Fedorchuk, C.A.; Lim, K.; Brakebusch, C.; McGrath, J.L.; Waugh, R.E.; Meier-Schellersheim, M.; et al. Opposing roles for RhoH GTPase during T-cell migration and activation. Proc. Natl. Acad. Sci. USA 2012, 109, 10474–10479. [Google Scholar] [CrossRef] [PubMed]
- Lawson, C.D.; Ridley, A.J. Rho GTPase signaling complexes in cell migration and invasion. J. Cell Biol. 2018, 217, 447–457. [Google Scholar] [CrossRef]
- Gowhari Shabgah, A.; Al-Obaidi, Z.M.J.; Sulaiman Rahman, H.; Kamal Abdelbasset, W.; Suksatan, W.; Bokov, D.O.; Thangavelu, L.; Turki Jalil, A.; Jadidi-Niaragh, F.; Mohammadi, H.; et al. Does CCL19 act as a double-edged sword in cancer development? Clin. Exp. Immunol. 2022, 207, 164–175. [Google Scholar] [CrossRef] [PubMed]
- Cheng, H.W.; Onder, L.; Cupovic, J.; Boesch, M.; Novkovic, M.; Pikor, N.; Tarantino, I.; Rodriguez, R.; Schneider, T.; Jochum, W.; et al. CCL19-producing fibroblastic stromal cells restrain lung carcinoma growth by promoting local antitumor T-cell responses. J. Allergy Clin. Immunol. 2018, 142, 1257–1271. [Google Scholar] [CrossRef]
- Zhang, Q.; Sun, L.; Yin, L.; Ming, J.; Zhang, S.; Luo, W.; Qiu, X. CCL19/CCR7 upregulates heparanase via specificity protein-1 (Sp1) to promote invasion of cell in lung cancer. Tumour Biol. 2013, 34, 2703–2708. [Google Scholar] [CrossRef]
- Jayatilleke, K.M.; Hulett, M.D. Heparanase and the hallmarks of cancer. J. Transl. Med. 2020, 18, 453. [Google Scholar] [CrossRef] [PubMed]
- Xu, B.; Zhou, M.; Qiu, W.; Ye, J.; Feng, Q. CCR7 mediates human breast cancer cell invasion, migration by inducing epithelial-mesenchymal transition and suppressing apoptosis through AKT pathway. Cancer Med. 2017, 6, 1062–1071. [Google Scholar] [CrossRef] [PubMed]
- Kotiyal, S.; Bhattacharya, S. Events of Molecular Changes in Epithelial-Mesenchymal Transition. Crit. Rev. Eukaryot Gene Expr. 2016, 26, 163–171. [Google Scholar] [CrossRef] [PubMed]
- Li, K.; Xu, B.; Xu, G.; Liu, R. CCR7 regulates Twist to induce the epithelial-mesenchymal transition in pancreatic ductal adenocarcinoma. Tumour Biol. 2016, 37, 419–424. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Wang, Y.; Cao, Y.; Zhang, X.; Zhao, H. Increased CCL19 expression is associated with progression in cervical cancer. Oncotarget 2017, 8, 73817–73825. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Seethala, R.R.; Zhang, Q.; Gooding, W.; van Waes, C.; Hasegawa, H.; Ferris, R.L. 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] [PubMed]
- Xu, D.; Liu, X.; Ke, S.; Guo, Y.; Zhu, C.; Cao, H. CCL19/CCR7 drives regulatory T cell migration and indicates poor prognosis in gastric cancer. BMC Cancer 2023, 23, 464. [Google Scholar] [CrossRef] [PubMed]
- Han, S.J.; Jain, P.; Gilad, Y.; Xia, Y.; Sung, N.; Park, M.J.; Dean, A.M.; Lanz, R.B.; Xu, J.; Dacso, C.C.; et al. Steroid receptor coactivator 3 is a key modulator of regulatory T cell-mediated tumor evasion. Proc. Natl. Acad. Sci. USA 2023, 120, e2221707120. [Google Scholar] [CrossRef] [PubMed]
- Shields, J.D.; Kourtis, I.C.; Tomei, A.A.; Roberts, J.M.; Swartz, M.A. Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science 2010, 328, 749–752. [Google Scholar] [CrossRef] [PubMed]
- Groom, J.R.; Luster, A.D. CXCR3 ligands: Redundant, collaborative and antagonistic functions. Immunol. Cell. Biol. 2011, 89, 207–215. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Zhang, Y.; Wang, S.; Ni, H.; Zhao, P.; Chen, G.; Xu, B.; Yuan, L. The role of CXCR3 and its ligands in cancer. Front. Oncol. 2022, 12, 1022688. [Google Scholar] [CrossRef] [PubMed]
- Maurice, N.J.; McElrath, M.J.; Andersen-Nissen, E.; Frahm, N.; Prlic, M. CXCR3 enables recruitment and site-specific bystander activation of memory CD8(+) T cells. Nat. Commun. 2019, 10, 4987. [Google Scholar] [CrossRef] [PubMed]
- Groom, J.R.; Luster, A.D. CXCR3 in T cell function. Exp. Cell Res. 2011, 317, 620–631. [Google Scholar] [CrossRef] [PubMed]
- Ozga, A.J.; Chow, M.T.; Luster, A.D. Chemokines and the immune response to cancer. Immunity 2021, 54, 859–874. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.Y.; Nguyen, B.; Mukhopadhyay, A.; Han, M.; Zhang, J.; Gujar, R.; Salazar, J.; Hermiz, R.; Svenson, L.; Browning, E.; et al. Amplification of the CXCR3/CXCL9 axis via intratumoral electroporation of plasmid CXCL9 synergizes with plasmid IL-12 therapy to elicit robust anti-tumor immunity. Mol. Ther. Oncolytics 2022, 25, 174–188. [Google Scholar] [CrossRef] [PubMed]
- Gorbachev, A.V.; Kobayashi, H.; Kudo, D.; Tannenbaum, C.S.; Finke, J.H.; Shu, S.; Farber, J.M.; Fairchild, R.L. CXC chemokine ligand 9/monokine induced by IFN-gamma production by tumor cells is critical for T cell-mediated suppression of cutaneous tumors. J. Immunol. 2007, 178, 2278–2286. [Google Scholar] [CrossRef] [PubMed]
- Nagarsheth, N.; Peng, D.; Kryczek, I.; Wu, K.; Li, W.; Zhao, E.; Zhao, L.; Wei, S.; Frankel, T.; Vatan, L.; et al. PRC2 Epigenetically Silences Th1-Type Chemokines to Suppress Effector T-Cell Trafficking in Colon Cancer. Cancer Res. 2016, 76, 275–282. [Google Scholar] [CrossRef] [PubMed]
- Peng, D.; Kryczek, I.; Nagarsheth, N.; Zhao, L.; Wei, S.; Wang, W.; Sun, Y.; Zhao, E.; Vatan, L.; Szeliga, W.; et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 2015, 527, 249–253. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.H.; Nieman, L.T.; Spurrell, M.; Jorgji, V.; Elmelech, L.; Richieri, P.; Xu, K.H.; Madhu, R.; Parikh, M.; Zamora, I.; et al. Human lung cancer harbors spatially organized stem-immunity hubs associated with response to immunotherapy. Nat. Immunol. 2024, 25, 644–658. [Google Scholar] [CrossRef]
- Meiser, P.; Knolle, M.A.; Hirschberger, A.; de Almeida, G.P.; Bayerl, F.; Lacher, S.; Pedde, A.M.; Flommersfeld, S.; Honninger, J.; Stark, L.; et al. A distinct stimulatory cDC1 subpopulation amplifies CD8(+) T cell responses in tumors for protective anti-cancer immunity. Cancer Cell 2023, 41, 1498–1515. [Google Scholar] [CrossRef]
- Zipin-Roitman, A.; Meshel, T.; Sagi-Assif, O.; Shalmon, B.; Avivi, C.; Pfeffer, R.M.; Witz, I.P.; Ben-Baruch, A. CXCL10 promotes invasion-related properties in human colorectal carcinoma cells. Cancer Res. 2007, 67, 3396–3405. [Google Scholar] [CrossRef] [PubMed]
- Burns, J.M.; Summers, B.C.; Wang, Y.; Melikian, A.; Berahovich, R.; Miao, Z.; Penfold, M.E.; Sunshine, M.J.; Littman, D.R.; Kuo, C.J.; et al. A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J. Exp. Med. 2006, 203, 2201–2213. [Google Scholar] [CrossRef] [PubMed]
- Naumann, U.; Cameroni, E.; Pruenster, M.; Mahabaleshwar, H.; Raz, E.; Zerwes, H.G.; Rot, A.; Thelen, M. CXCR7 functions as a scavenger for CXCL12 and CXCL11. PLoS ONE 2010, 5, e9175. [Google Scholar] [CrossRef] [PubMed]
- Li, A.; Herbst, R.H.; Canner, D.; Schenkel, J.M.; Smith, O.C.; Kim, J.Y.; Hillman, M.; Bhutkar, A.; Cuoco, M.S.; Rappazzo, C.G.; et al. IL-33 Signaling Alters Regulatory T Cell Diversity in Support of Tumor Development. Cell Rep. 2019, 29, 2998–3008. [Google Scholar] [CrossRef] [PubMed]
- Vianello, F.; Papeta, N.; Chen, T.; Kraft, P.; White, N.; Hart, W.K.; Kircher, M.F.; Swart, E.; Rhee, S.; Palu, G.; et al. Murine B16 melanomas expressing high levels of the chemokine stromal-derived factor-1/CXCL12 induce tumor-specific T cell chemorepulsion and escape from immune control. J. Immunol. 2006, 176, 2902–2914. [Google Scholar] [CrossRef] [PubMed]
- Garg, B.; Giri, B.; Modi, S.; Sethi, V.; Castro, I.; Umland, O.; Ban, Y.; Lavania, S.; Dawra, R.; Banerjee, S.; et al. NFkappaB in Pancreatic Stellate Cells Reduces Infiltration of Tumors by Cytotoxic T Cells and Killing of Cancer Cells, via Up-regulation of CXCL12. Gastroenterology 2018, 155, 880–891. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.N.; Schmidt, M.O.; Sharif, G.M.; Vietsch, E.E.; Kiliti, A.J.; Barefoot, M.E.; Riegel, A.T.; Wellstein, A. Impaired CXCL12 signaling contributes to resistance of pancreatic cancer subpopulations to T cell-mediated cytotoxicity. Oncoimmunology 2022, 11, 2027136. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Feng, W.; Sun, M.; Huang, W.; Wang, G.; Chen, X.; Yin, Y.; Chen, X.; Zhang, B.; Nie, Y.; et al. TGF-beta1-Induced SOX18 Elevation Promotes Hepatocellular Carcinoma Progression and Metastasis Through Transcriptionally Upregulating PD-L1 and CXCL12. Gastroenterology 2024. [Google Scholar] [CrossRef] [PubMed]
- Steele, M.M.; Jaiswal, A.; Delclaux, I.; Dryg, I.D.; Murugan, D.; Femel, J.; Son, S.; du Bois, H.; Hill, C.; Leachman, S.A.; et al. T cell egress via lymphatic vessels is tuned by antigen encounter and limits tumor control. Nat. Immunol. 2023, 24, 664–675. [Google Scholar] [CrossRef] [PubMed]
- Hussain, M.; Liu, J.; Wang, G.Z.; Zhou, G.B. CXCL13 Signaling in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2021, 1302, 71–90. [Google Scholar] [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]
- Ukita, M.; Hamanishi, J.; Yoshitomi, H.; Yamanoi, K.; Takamatsu, S.; Ueda, A.; Suzuki, H.; Hosoe, Y.; Furutake, Y.; Taki, M.; et al. CXCL13-producing CD4+ T cells accumulate in the early phase of tertiary lymphoid structures in ovarian cancer. JCI Insight 2022, 7, e157215. [Google Scholar] [CrossRef] [PubMed]
- Groeneveld, C.S.; Fontugne, J.; Cabel, L.; Bernard-Pierrot, I.; Radvanyi, F.; Allory, Y.; de Reynies, 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] [PubMed]
- Ren, J.; Lan, T.; Liu, T.; Liu, Y.; Shao, B.; Men, K.; Ma, Y.; Liang, X.; Wei, Y.Q.; Luo, M.; et al. CXCL13 as a Novel Immune Checkpoint for Regulatory B Cells and Its Role in Tumor Metastasis. J. Immunol. 2022, 208, 2425–2435. [Google Scholar] [CrossRef] [PubMed]
- Samus, M.; Rot, A. Atypical chemokine receptors in cancer. Cytokine 2024, 176, 156504. [Google Scholar] [CrossRef] [PubMed]
- Feng, L.Y.; Ou, Z.L.; Wu, F.Y.; Shen, Z.Z.; Shao, Z.M. Involvement of a novel chemokine decoy receptor CCX-CKR in breast cancer growth, metastasis and patient survival. Clin. Cancer Res. 2009, 15, 2962–2970. [Google Scholar] [CrossRef] [PubMed]
- Luker, K.E.; Lewin, S.A.; Mihalko, L.A.; Schmidt, B.T.; Winkler, J.S.; Coggins, N.L.; Thomas, D.G.; Luker, G.D. Scavenging of CXCL12 by CXCR7 promotes tumor growth and metastasis of CXCR4-positive breast cancer cells. Oncogene 2012, 31, 4750–4758. [Google Scholar] [CrossRef] [PubMed]
- Glass, W.G.; Lim, J.K.; Cholera, R.; Pletnev, A.G.; Gao, J.L.; Murphy, P.M. Chemokine receptor CCR5 promotes leukocyte trafficking to the brain and survival in West Nile virus infection. J. Exp. Med. 2005, 202, 1087–1098. [Google Scholar] [CrossRef] [PubMed]
- Huffman, A.P.; Lin, J.H.; Kim, S.I.; Byrne, K.T.; Vonderheide, R.H. CCL5 mediates CD40-driven CD4+ T cell tumor infiltration and immunity. JCI Insight 2020, 5, e137263. [Google Scholar] [CrossRef] [PubMed]
- Aldinucci, D.; Borghese, C.; Casagrande, N. The CCL5/CCR5 Axis in Cancer Progression. Cancers 2020, 12, 1765. [Google Scholar] [CrossRef] [PubMed]
- Sugasawa, H.; Ichikura, T.; Kinoshita, M.; Ono, S.; Majima, T.; Tsujimoto, H.; Chochi, K.; Hiroi, S.; Takayama, E.; Saitoh, D.; et al. Gastric cancer cells exploit CD4+ cell-derived CCL5 for their growth and prevention of CD8+ cell-involved tumor elimination. Int. J. Cancer 2008, 122, 2535–2541. [Google Scholar] [CrossRef] [PubMed]
- Chang, L.Y.; Lin, Y.C.; Mahalingam, J.; Huang, C.T.; Chen, T.W.; Kang, C.W.; Peng, H.M.; Chu, Y.Y.; Chiang, J.M.; Dutta, A.; et al. Tumor-derived chemokine CCL5 enhances TGF-beta-mediated killing of CD8(+) T cells in colon cancer by T-regulatory cells. Cancer Res. 2012, 72, 1092–1102. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Qin, J.; Zhong, L.; Gong, L.; Zhang, B.; Zhang, Y.; Gao, W.Q. CCL5-Mediated Th2 Immune Polarization Promotes Metastasis in Luminal Breast Cancer. Cancer Res. 2015, 75, 4312–4321. [Google Scholar] [CrossRef] [PubMed]
- Nesbeth, Y.; Scarlett, U.; Cubillos-Ruiz, J.; Martinez, D.; Engle, X.; Turk, M.J.; Conejo-Garcia, J.R. CCL5-mediated endogenous antitumor immunity elicited by adoptively transferred lymphocytes and dendritic cell depletion. Cancer Res. 2009, 69, 6331–6338. [Google Scholar] [CrossRef] [PubMed]
- Jacobs, C.; Shah, S.; Lu, W.C.; Ray, H.; Wang, J.; Hockaden, N.; Sandusky, G.; Nephew, K.P.; Lu, X.; Cao, S.; et al. HSF1 Inhibits Antitumor Immune Activity in Breast Cancer by Suppressing CCL5 to Block CD8+ T-cell Recruitment. Cancer Res. 2024, 84, 276–290. [Google Scholar] [CrossRef] [PubMed]
- Gowhari Shabgah, A.; Qasim, M.T.; Mojtaba Mostafavi, S.; Olegovna Zekiy, A.; Ezzatifar, F.; Ahmadi, M.; Mohammadian Haftcheshmeh, S.; Gholizadeh Navashenaq, J. CXC chemokine ligand 16: A Swiss army knife chemokine in cancer. Expert Rev. Mol. Med. 2021, 23, e4. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- Di Pilato, M.; Kfuri-Rubens, R.; Pruessmann, J.N.; Ozga, A.J.; Messemaker, M.; Cadilha, B.L.; Sivakumar, R.; Cianciaruso, C.; Warner, R.D.; Marangoni, F.; et al. CXCR6 positions cytotoxic T cells to receive critical survival signals in the tumor microenvironment. Cell 2021, 184, 4512–4530. [Google Scholar] [CrossRef]
- Parsonage, G.; Machado, L.R.; Hui, J.W.; McLarnon, A.; Schmaler, T.; Balasothy, M.; To, K.F.; Vlantis, A.C.; van Hasselt, C.A.; Lo, K.W.; et al. CXCR6 and CCR5 localize T lymphocyte subsets in nasopharyngeal carcinoma. Am. J. Pathol. 2012, 180, 1215–1222. [Google Scholar] [CrossRef] [PubMed]
- Muthuswamy, R.; McGray, A.R.; Battaglia, S.; He, W.; Miliotto, A.; Eppolito, C.; Matsuzaki, J.; Takemasa, T.; Koya, R.; Chodon, T.; et al. CXCR6 by increasing retention of memory CD8+ T cells in the ovarian tumor microenvironment promotes immunosurveillance and control of ovarian cancer. J. Immunother. Cancer 2021, 9, e003329. [Google Scholar] [CrossRef] [PubMed]
- Broquet, A.; Gourain, V.; Goronflot, T.; Le Mabecque, V.; Sinha, D.; Ashayeripanah, M.; Jacqueline, C.; Martin, P.; Davieau, M.; Boutin, L.; et al. Sepsis-trained macrophages promote antitumoral tissue-resident T cells. Nat. Immunol. 2024, 25, 802–819. [Google Scholar] [CrossRef] [PubMed]
- Richardsen, E.; Ness, N.; Melbo-Jorgensen, C.; Johannesen, C.; Grindstad, T.; Nordbakken, C.; Al-Saad, S.; Andersen, S.; Donnem, T.; Nordby, Y.; et al. The prognostic significance of CXCL16 and its receptor C-X-C chemokine receptor 6 in prostate cancer. Am. J. Pathol. 2015, 185, 2722–2730. [Google Scholar] [CrossRef] [PubMed]
- Allaoui, R.; Bergenfelz, C.; Mohlin, S.; Hagerling, C.; Salari, K.; Werb, Z.; Anderson, R.L.; Ethier, S.P.; Jirstrom, K.; Pahlman, S.; et al. Cancer-associated fibroblast-secreted CXCL16 attracts monocytes to promote stroma activation in triple-negative breast cancers. Nat. Commun. 2016, 7, 13050. [Google Scholar] [CrossRef] [PubMed]
- Mujal, A.M.; Delconte, R.B.; Sun, J.C. Natural Killer Cells: From Innate to Adaptive Features. Annu. Rev. Immunol. 2021, 39, 417–447. [Google Scholar] [CrossRef] [PubMed]
- Pockley, A.G.; Vaupel, P.; Multhoff, G. NK cell-based therapeutics for lung cancer. Expert Opin. Biol. Ther. 2020, 20, 23–33. [Google Scholar] [CrossRef] [PubMed]
- Muntasell, A.; Rojo, F.; Servitja, S.; Rubio-Perez, C.; Cabo, M.; Tamborero, D.; Costa-Garcia, M.; Martinez-Garcia, M.; Menendez, S.; Vazquez, I.; et al. NK Cell Infiltrates and HLA Class I Expression in Primary HER2(+) Breast Cancer Predict and Uncouple Pathological Response and Disease-free Survival. Clin. Cancer Res. 2019, 25, 1535–1545. [Google Scholar] [CrossRef] [PubMed]
- Liu, P.; Chen, L.; Zhang, H. Natural Killer Cells in Liver Disease and Hepatocellular Carcinoma and the NK Cell-Based Immunotherapy. J. Immunol. Res. 2018, 2018, 1206737. [Google Scholar] [CrossRef] [PubMed]
- Ali, T.H.; Pisanti, S.; Ciaglia, E.; Mortarini, R.; Anichini, A.; Garofalo, C.; Tallerico, R.; Santinami, M.; Gulletta, E.; Ietto, C.; et al. Enrichment of CD56(dim)KIR + CD57 + highly cytotoxic NK cells in tumour-infiltrated lymph nodes of melanoma patients. Nat. Commun. 2014, 5, 5639. [Google Scholar] [CrossRef] [PubMed]
- Terren, I.; Orrantia, A.; Mikelez-Alonso, I.; Vitalle, J.; Zenarruzabeitia, O.; Borrego, F. NK Cell-Based Immunotherapy in Renal Cell Carcinoma. Cancers (Basel) 2020, 12, 316. [Google Scholar] [CrossRef] [PubMed]
- Ran, G.H.; Lin, Y.Q.; Tian, L.; Zhang, T.; Yan, D.M.; Yu, J.H.; Deng, Y.C. Natural killer cell homing and trafficking in tissues and tumors: From biology to application. Signal Transduct Target Ther. 2022, 7, 205. [Google Scholar] [CrossRef] [PubMed]
- Braun, S.E.; Chen, K.; Foster, R.G.; Kim, C.H.; Hromas, R.; Kaplan, M.H.; Broxmeyer, H.E.; Cornetta, K. The CC chemokine CK beta-11/MIP-3 beta/ELC/Exodus 3 mediates tumor rejection of murine breast cancer cells through NK cells. J. Immunol. 2000, 164, 4025–4031. [Google Scholar] [CrossRef] [PubMed]
- Bottcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037. [Google Scholar] [CrossRef] [PubMed]
- Wolf, N.K.; Kissiov, D.U.; Raulet, D.H. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat. Rev. Immunol. 2023, 23, 90–105. [Google Scholar] [CrossRef] [PubMed]
- Cozar, B.; Greppi, M.; Carpentier, S.; Narni-Mancinelli, E.; Chiossone, L.; Vivier, E. Tumor-Infiltrating Natural Killer Cells. Cancer Discov. 2021, 11, 34–44. [Google Scholar] [CrossRef] [PubMed]
- Seliger, B.; Koehl, U. Underlying mechanisms of evasion from NK cells as rationale for improvement of NK cell-based immunotherapies. Front. Immunol. 2022, 13, 910595. [Google Scholar] [CrossRef] [PubMed]
- Mauri, C.; Bosma, A. Immune regulatory function of B cells. Annu. Rev. Immunol. 2012, 30, 221–241. [Google Scholar] [CrossRef] [PubMed]
- Berraondo, P.; Minute, L.; Ajona, D.; Corrales, L.; Melero, I.; Pio, R. Innate immune mediators in cancer: Between defense and resistance. Immunol. Rev. 2016, 274, 290–306. [Google Scholar] [CrossRef] [PubMed]
- Reis, E.S.; Mastellos, D.C.; Ricklin, D.; Mantovani, A.; Lambris, J.D. Complement in cancer: Untangling an intricate relationship. Nat. Rev. Immunol. 2018, 18, 5–18. [Google Scholar] [CrossRef] [PubMed]
- Hajishengallis, G.; Reis, E.S.; Mastellos, D.C.; Ricklin, D.; Lambris, J.D. Novel mechanisms and functions of complement. Nat. Immunol. 2017, 18, 1288–1298. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.; Zhao, Q.; Liao, J.Y.; Song, E.; Xia, Q.; Pan, J.; Li, Y.; Li, J.; Zhou, B.; Ye, Y.; et al. Complement Signals Determine Opposite Effects of B Cells in Chemotherapy-Induced Immunity. Cell 2020, 180, 1081–1097. [Google Scholar] [CrossRef] [PubMed]
- Mueller, C.G.; Boix, C.; Kwan, W.H.; Daussy, C.; Fournier, E.; Fridman, W.H.; Molina, T.J. Critical role of monocytes to support normal B cell and diffuse large B cell lymphoma survival and proliferation. J. Leukoc. Biol. 2007, 82, 567–575. [Google Scholar] [CrossRef]
- Workel, H.H.; Lubbers, J.M.; Arnold, R.; Prins, T.M.; van der Vlies, P.; de Lange, K.; Bosse, T.; van Gool, I.C.; Eggink, F.A.; Wouters, M.C.A.; et al. A Transcriptionally Distinct CXCL13(+)CD103(+)CD8(+) T-cell Population Is Associated with B-cell Recruitment and Neoantigen Load in Human Cancer. Cancer Immunol. Res. 2019, 7, 784–796. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Tan, Y.; Sun, F.; Hou, L.; Zhang, C.; Ge, T.; Yu, H.; Wu, C.; Zhu, Y.; Duan, L.; et al. Single-cell transcriptome and antigen-immunoglobin analysis reveals the diversity of B cells in non-small cell lung cancer. Genome Biol. 2020, 21, 152. [Google Scholar] [CrossRef] [PubMed]
- Hu, Q.; Hong, Y.; Qi, P.; Lu, G.; Mai, X.; Xu, S.; He, X.; Guo, Y.; Gao, L.; Jing, Z.; et al. Atlas of breast cancer infiltrated B-lymphocytes revealed by paired single-cell RNA-sequencing and antigen receptor profiling. Nat. Commun. 2021, 12, 2186. [Google Scholar] [CrossRef] [PubMed]
- Dieci, M.V.; Radosevic-Robin, N.; Fineberg, S.; van den Eynden, G.; Ternes, N.; Penault-Llorca, F.; Pruneri, G.; D’Alfonso, T.M.; Demaria, S.; Castaneda, C.; et al. Update on tumor-infiltrating lymphocytes (TILs) in breast cancer, including recommendations to assess TILs in residual disease after neoadjuvant therapy and in carcinoma in situ: A report of the International Immuno-Oncology Biomarker Working Group on Breast Cancer. Semin. Cancer Biol. 2018, 52, 16–25. [Google Scholar] [PubMed]
- Jiang, H.; Hegde, S.; DeNardo, D.G. Tumor-associated fibrosis as a regulator of tumor immunity and response to immunotherapy. Cancer Immunol. Immunother. 2017, 66, 1037–1048. [Google Scholar] [CrossRef] [PubMed]
- DuChez, B.J.; Doyle, A.D.; Dimitriadis, E.K.; Yamada, K.M. Durotaxis by Human Cancer Cells. Biophys. J. 2019, 116, 670–683. [Google Scholar] [CrossRef]
- Yang, S.; Plotnikov, S.V. Mechanosensitive Regulation of Fibrosis. Cells 2021, 10, 994. [Google Scholar] [CrossRef]
- Sunyer, R.; Conte, V.; Escribano, J.; Elosegui-Artola, A.; Labernadie, A.; Valon, L.; Navajas, D.; Garcia-Aznar, J.M.; Munoz, J.J.; Roca-Cusachs, P.; et al. Collective cell durotaxis emerges from long-range intercellular force transmission. Science 2016, 353, 1157–1161. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Liu, Y.; Hai, R.; Liao, W.; Luo, X. The role of circadian clocks in cancer: Mechanisms and clinical implications. Genes Dis. 2023, 10, 1279–1290. [Google Scholar] [CrossRef] [PubMed]
- He, W.; Holtkamp, S.; Hergenhan, S.M.; Kraus, K.; de Juan, A.; Weber, J.; Bradfield, P.; Grenier, J.M.P.; Pelletier, J.; Druzd, D.; et al. Circadian Expression of Migratory Factors Establishes Lineage-Specific Signatures that Guide the Homing of Leukocyte Subsets to Tissues. Immunity 2018, 49, 1175–1190. [Google Scholar] [CrossRef] [PubMed]
- Scheiermann, C.; Kunisaki, Y.; Frenette, P.S. Circadian control of the immune system. Nat. Rev. Immunol. 2013, 13, 190–198. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Zhao, Q.; Zhang, Y.; Liu, Z.; Zheng, Z.; Liu, S.; Meng, L.; Xin, Y.; Jiang, X. Targeting hypoxia in the tumor microenvironment: A potential strategy to improve cancer immunotherapy. J. Exp. Clin. Cancer Res. 2021, 40, 24. [Google Scholar] [CrossRef] [PubMed]
- Parent, C.A.; Devreotes, P.N. A cell’s sense of direction. Science 1999, 284, 765–770. [Google Scholar] [CrossRef]
- Lanitis, E.; Irving, M.; Coukos, G. Targeting the tumor vasculature to enhance T cell activity. Curr. Opin. Immunol. 2015, 33, 55–63. [Google Scholar] [CrossRef] [PubMed]
- Kolaczkowska, E.; Kubes, P. Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 2013, 13, 159–175. [Google Scholar] [CrossRef] [PubMed]
- Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [CrossRef] [PubMed]
- Shi, C.; Pamer, E.G. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 2011, 11, 762–774. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Hossain, M.; Thanabalasuriar, A.; Gunzer, M.; Meininger, C.; Kubes, P. Visualizing the function and fate of neutrophils in sterile injury and repair. Science 2017, 358, 111–116. [Google Scholar] [CrossRef] [PubMed]
- Lim, K.; Hyun, Y.M.; Lambert-Emo, K.; Capece, T.; Bae, S.; Miller, R.; Topham, D.J.; Kim, M. Neutrophil trails guide influenza-specific CD8(+) T cells in the airways. Science 2015, 349, aaa4352. [Google Scholar] [CrossRef] [PubMed]
- Lim, K.; Kim, T.H.; Trzeciak, A.; Amitrano, A.M.; Reilly, E.C.; Prizant, H.; Fowell, D.J.; Topham, D.J.; Kim, M. In situ neutrophil efferocytosis shapes T cell immunity to influenza infection. Nat. Immunol. 2020, 21, 1046–1057. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef] [PubMed]
- Geiger, R.; Rieckmann, J.C.; Wolf, T.; Basso, C.; Feng, Y.; Fuhrer, T.; Kogadeeva, M.; Picotti, P.; Meissner, F.; Mann, M.; et al. L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity. Cell 2016, 167, 829–842. [Google Scholar] [CrossRef] [PubMed]
- Norian, L.A.; Rodriguez, P.C.; O’Mara, L.A.; Zabaleta, J.; Ochoa, A.C.; Cella, M.; Allen, P.M. Tumor-infiltrating regulatory dendritic cells inhibit CD8+ T cell function via L-arginine metabolism. Cancer Res. 2009, 69, 3086–3094. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [PubMed]
- Huang, B.; Pan, P.Y.; Li, Q.; Sato, A.I.; Levy, D.E.; Bromberg, J.; Divino, C.M.; Chen, S.H. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006, 66, 1123–1131. [Google Scholar] [CrossRef] [PubMed]
- Gehad, A.E.; Lichtman, M.K.; Schmults, C.D.; Teague, J.E.; Calarese, A.W.; Jiang, Y.; Watanabe, R.; Clark, R.A. Nitric oxide-producing myeloid-derived suppressor cells inhibit vascular E-selectin expression in human squamous cell carcinomas. J. Investig. Dermatol. 2012, 132, 2642–2651. [Google Scholar] [CrossRef]
- Lin, E.Y.; Li, J.F.; Gnatovskiy, L.; Deng, Y.; Zhu, L.; Grzesik, D.A.; Qian, H.; Xue, X.N.; Pollard, J.W. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006, 66, 11238–11246. [Google Scholar] [CrossRef] [PubMed]
- Nozawa, H.; Chiu, C.; Hanahan, D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc. Natl. Acad. Sci. USA 2006, 103, 12493–12498. [Google Scholar] [CrossRef] [PubMed]
- Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef] [PubMed]
- Deryugina, E.I.; Quigley, J.P. Pleiotropic roles of matrix metalloproteinases in tumor angiogenesis: Contrasting, overlapping and compensatory functions. Biochim. Biophys. Acta 2010, 1803, 103–120. [Google Scholar] [CrossRef] [PubMed]
- Toh, B.; Wang, X.; Keeble, J.; Sim, W.J.; Khoo, K.; Wong, W.C.; Kato, M.; Prevost-Blondel, A.; Thiery, J.P.; Abastado, J.P. Mesenchymal transition and dissemination of cancer cells is driven by myeloid-derived suppressor cells infiltrating the primary tumor. PLoS Biol. 2011, 9, e1001162. [Google Scholar] [CrossRef] [PubMed]
- Kitamura, T.; Kometani, K.; Hashida, H.; Matsunaga, A.; Miyoshi, H.; Hosogi, H.; Aoki, M.; Oshima, M.; Hattori, M.; Takabayashi, A.; et al. SMAD4-deficient intestinal tumors recruit CCR1+ myeloid cells that promote invasion. Nat. Genet. 2007, 39, 467–475. [Google Scholar] [CrossRef] [PubMed]
- El Rayes, T.; Catena, R.; Lee, S.; Stawowczyk, M.; Joshi, N.; Fischbach, C.; Powell, C.A.; Dannenberg, A.J.; Altorki, N.K.; Gao, D.; et al. Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc. Natl. Acad. Sci. USA 2015, 112, 16000–16005. [Google Scholar] [CrossRef]
- Coussens, L.M.; Tinkle, C.L.; Hanahan, D.; Werb, Z. MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell 2000, 103, 481–490. [Google Scholar] [CrossRef] [PubMed]
- Khan, I.Z.; Del Guezzo, C.A.; Shao, A.; Cho, J.; Du, R.; Cohen, A.O.; Owens, D.M. The CD200-CD200R axis promotes squamous cell carcinoma metastasis via regulation of Cathepsin K. Cancer Res. 2021, 81, 5021–5032. [Google Scholar] [CrossRef]
- Lu, Z.; Zou, J.; Li, S.; Topper, M.J.; Tao, Y.; Zhang, H.; Jiao, X.; Xie, W.; Kong, X.; Vaz, M.; et al. Epigenetic therapy inhibits metastases by disrupting premetastatic niches. Nature 2020, 579, 284–290. [Google Scholar] [CrossRef]
- Chen, Z.; Zhang, G.; Ren, X.; Yao, Z.; Zhou, Q.; Ren, X.; Chen, S.; Xu, L.; Sun, K.; Zeng, Q.; et al. Cross-talk between myeloid and B cells shapes the distinct microenvironments of primary and secondary liver cancer. Cancer Res. 2023, 83, 3544–3561. [Google Scholar] [CrossRef]
- Laubli, H.; Spanaus, K.S.; Borsig, L. Selectin-mediated activation of endothelial cells induces expression of CCL5 and promotes metastasis through recruitment of monocytes. Blood 2009, 114, 4583–4591. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef]
- Cook, R.S.; Jacobsen, K.M.; Wofford, A.M.; DeRyckere, D.; Stanford, J.; Prieto, A.L.; Redente, E.; Sandahl, M.; Hunter, D.M.; Strunk, K.E.; et al. MerTK inhibition in tumor leukocytes decreases tumor growth and metastasis. J. Clin. Investig. 2013, 123, 3231–3242. [Google Scholar] [CrossRef] [PubMed]
- Ng, M.S.F.; Kwok, I.; Tan, L.; Shi, C.; Cerezo-Wallis, D.; Tan, Y.; Leong, K.; Calvo, G.F.; Yang, K.; Zhang, Y.; et al. Deterministic reprogramming of neutrophils within tumors. Science 2024, 383, eadf6493. [Google Scholar] [CrossRef]
- Sattiraju, A.; Kang, S.; Giotti, B.; Chen, Z.; Marallano, V.J.; Brusco, C.; Ramakrishnan, A.; Shen, L.; Tsankov, A.M.; Hambardzumyan, D.; et al. Hypoxic niches attract and sequester tumor-associated macrophages and cytotoxic T cells and reprogram them for immunosuppression. Immunity 2023, 56, 1825–1843. [Google Scholar] [CrossRef] [PubMed]
- Pekarek, L.A.; Starr, B.A.; Toledano, A.Y.; Schreiber, H. Inhibition of tumor growth by elimination of granulocytes. J. Exp. Med. 1995, 181, 435–440. [Google Scholar] [CrossRef] [PubMed]
- Lin, E.Y.; Nguyen, A.V.; Russell, R.G.; Pollard, J.W. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J. Exp. Med. 2001, 193, 727–740. [Google Scholar] [CrossRef]
- Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
- Trinchieri, G. Cancer and inflammation: An old intuition with rapidly evolving new concepts. Annu. Rev. Immunol. 2012, 30, 677–706. [Google Scholar] [CrossRef]
- Qian, B.Z.; Li, J.; Zhang, H.; Kitamura, T.; Zhang, J.; Campion, L.R.; Kaiser, E.A.; Snyder, L.A.; Pollard, J.W. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 2011, 475, 222–225. [Google Scholar] [CrossRef] [PubMed]
- Varn, F.S.; Wang, Y.; Mullins, D.W.; Fiering, S.; Cheng, C. Systematic Pan-Cancer Analysis Reveals Immune Cell Interactions in the Tumor Microenvironment. Cancer Res. 2017, 77, 1271–1282. [Google Scholar] [CrossRef] [PubMed]
- Ajith, A.; Mamouni, K.; Horuzsko, D.D.; Musa, A.; Dzutsev, A.K.; Fang, J.R.; Chadli, A.; Zhu, X.; Lebedyeva, I.; Trinchieri, G.; et al. Targeting TREM1 augments antitumor T cell immunity by inhibiting myeloid-derived suppressor cells and restraining anti-PD-1 resistance. J. Clin. Invest. 2023, 133, e167951. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Zhao, Q.; Tan, L.; Wu, X.; Huang, R.; Zuo, Y.; Chen, L.; Yang, J.; Zhang, Z.X.; Ruan, W.; et al. Neutralizing IL-8 potentiates immune checkpoint blockade efficacy for glioma. Cancer Cell 2023, 41, 693–710. [Google Scholar] [CrossRef]
- Mei, Y.; Zhu, Y.; Yong, K.S.M.; Hanafi, Z.B.; Gong, H.; Liu, Y.; Teo, H.Y.; Hussain, M.; Song, Y.; Chen, Q.; et al. IL-37 dampens immunosuppressive functions of MDSCs via metabolic reprogramming in the tumor microenvironment. Cell Rep. 2024, 43, 113835. [Google Scholar] [CrossRef] [PubMed]
- Monteran., L.; Ershaid, N.; Scharff, Y.; Zoabi, Y.; Sanalla, T.; Ding, Y.; Pavlovsky, A.; Zait, Y.; Langer, M.; Caller, T. Combining TIGIT blockage with MDSC inhibition hinders breast cancer bone metastasis by activating anti-tumor immunity. Cancer Discov. 2024. epub ahead of print. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Liu, Y.D.; Zhan, Y.T.; Zhu, Y.H.; Li, Y.; Xie, D.; Guan, X.Y. 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]
- 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]
- Chang, A.L.; Miska, J.; Wainwright, D.A.; Dey, M.; Rivetta, C.V.; Yu, D.; Kanojia, D.; Pituch, K.C.; Qiao, J.; Pytel, P.; et al. CCL2 Produced by the Glioma Microenvironment Is Essential for the Recruitment of Regulatory T Cells and Myeloid-Derived Suppressor Cells. Cancer Res. 2016, 76, 5671–5682. [Google Scholar] [CrossRef]
- Sawanobori, Y.; Ueha, S.; Kurachi, M.; Shimaoka, T.; Talmadge, J.E.; Abe, J.; Shono, Y.; Kitabatake, M.; Kakimi, K.; Mukaida, N.; et al. Chemokine-mediated rapid turnover of myeloid-derived suppressor cells in tumor-bearing mice. Blood 2008, 111, 5457–5466. [Google Scholar] [CrossRef]
- Lesokhin, A.M.; Hohl, T.M.; Kitano, S.; Cortez, C.; Hirschhorn-Cymerman, D.; Avogadri, F.; Rizzuto, G.A.; Lazarus, J.J.; Pamer, E.G.; Houghton, A.N.; et al. Monocytic CCR2(+) myeloid-derived suppressor cells promote immune escape by limiting activated CD8 T-cell infiltration into the tumor microenvironment. Cancer Res. 2012, 72, 876–886. [Google Scholar] [CrossRef] [PubMed]
- Nesbit, M.; Schaider, H.; Miller, T.H.; Herlyn, M. Low-level monocyte chemoattractant protein-1 stimulation of monocytes leads to tumor formation in nontumorigenic melanoma cells. J. Immunol. 2001, 166, 6483–6490. [Google Scholar] [CrossRef] [PubMed]
- Rogic, A.; Pant, I.; Grumolato, L.; Fernandez-Rodriguez, R.; Edwards, A.; Das, S.; Sun, A.; Yao, S.; Qiao, R.; Jaffer, S.; et al. High endogenous CCL2 expression promotes the aggressive phenotype of human inflammatory breast cancer. Nat. Commun. 2021, 12, 6889. [Google Scholar] [CrossRef] [PubMed]
- Platten, M.; Kretz, A.; Naumann, U.; Aulwurm, S.; Egashira, K.; Isenmann, S.; Weller, M. Monocyte chemoattractant protein-1 increases microglical infiltration and aggressiveness of gliomas. Ann. Neurol. 2003, 54, 388–392. [Google Scholar] [CrossRef] [PubMed]
- Moreno Ayala, M.A.; Campbell, T.F.; Zhang, C.; Dahan, N.; Bockman, A.; Prakash, V.; Feng, L.; Sher, T.; DuPage, M. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8(+) T cell antitumor immunity. Immunity 2023, 56, 1613–1630. [Google Scholar] [CrossRef] [PubMed]
- Spehlmann, M.E.; Dann, S.M.; Hruz, P.; Hanson, E.; McCole, D.F.; Eckmann, L. CXCR2-dependent mucosal neutrophil influx protects against colitis-associated diarrhea caused by an attaching/effacing lesion-forming bacterial pathogen. J. Immunol. 2009, 183, 3332–3343. [Google Scholar] [CrossRef] [PubMed]
- Lazennec, G.; Rajarathnam, K.; Richmond, A. CXCR2 chemokine receptor—A master regulator in cancer and physiology. Trends Mol. Med. 2024, 30, 37–55. [Google Scholar] [CrossRef]
- Cui, S.; Chen, X.; Li, J.; Wang, W.; Meng, D.; Zhu, S.; Shen, S. Endothelial CXCR2 deficiency attenuates renal inflammation and glycocalyx shedding through NF-kappaB signaling in diabetic kidney disease. Cell Commun. Signal. 2024, 22, 191. [Google Scholar] [CrossRef]
- Li, D.J.; Zhong, Z.J.; Wang, X.L.; Wei, N.; Zhao, S.J.; Shan, T.T.; Liu, Y.P.; Yu, Y.Q. Chemokine receptor CXCR2 in primary sensory neurons of trigeminal ganglion mediates orofacial itch. Front. Mol. Neurosci. 2023, 16, 1279237. [Google Scholar] [CrossRef]
- Li, Y.; He, Y.; Butler, W.; Xu, L.; Chang, Y.; Lei, K.; Zhang, H.; Zhou, Y.; Gao, A.C.; Zhang, Q.; et al. Targeting cellular heterogeneity with CXCR2 blockade for the treatment of therapy-resistant prostate cancer. Sci. Transl. Med. 2019, 11, eaax0428. [Google Scholar] [CrossRef]
- Gungabeesoon, J.; Gort-Freitas, N.A.; Kiss, M.; Bolli, E.; Messemaker, M.; Siwicki, M.; Hicham, M.; Bill, R.; Koch, P.; Cianciaruso, C.; et al. A neutrophil response linked to tumor control in immunotherapy. Cell 2023, 186, 1448–1463. [Google Scholar] [CrossRef] [PubMed]
- Cheng, W.L.; Wang, C.S.; Huang, Y.H.; Tsai, M.M.; Liang, Y.; Lin, K.H. Overexpression of CXCL1 and its receptor CXCR2 promote tumor invasion in gastric cancer. Ann. Oncol. 2011, 22, 2267–2276. [Google Scholar] [CrossRef] [PubMed]
- Yuen, K.C.; Liu, L.F.; Gupta, V.; Madireddi, S.; Keerthivasan, S.; Li, C.; Rishipathak, D.; Williams, P.; Kadel, E.E.; Koeppen, H.; et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat. Med. 2020, 26, 693–698. [Google Scholar] [CrossRef] [PubMed]
- Van Meir, E.; Ceska, M.; Effenberger, F.; Walz, A.; Grouzmann, E.; Desbaillets, I.; Frei, K.; Fontana, A.; de Tribolet, N. Interleukin-8 is produced in neoplastic and infectious diseases of the human central nervous system. Cancer Res. 1992, 52, 4297–4305. [Google Scholar] [PubMed]
- Schadendorf, D.; Moller, A.; Algermissen, B.; Worm, M.; Sticherling, M.; Czarnetzki, B.M. IL-8 produced by human malignant melanoma cells in vitro is an essential autocrine growth factor. J. Immunol. 1993, 151, 2667–2675. [Google Scholar] [CrossRef] [PubMed]
- Bellocq, A.; Antoine, M.; Flahault, A.; Philippe, C.; Crestani, B.; Bernaudin, J.F.; Mayaud, C.; Milleron, B.; Baud, L.; Cadranel, J. Neutrophil alveolitis in bronchioloalveolar carcinoma: Induction by tumor-derived interleukin-8 and relation to clinical outcome. Am. J. Pathol. 1998, 152, 83–92. [Google Scholar] [PubMed]
- Olivera, I.; Sanz-Pamplona, R.; Bolaños, E.; Rodriguez, I.; Etxeberria, I.; Cirella, A.; Egea, J.; Garasa, S.; Migueliz, I.; Eguren-Santamaria, I.; et al. A Therapeutically Actionable Protumoral Axis of Cytokines Involving IL-8, TNFα, and IL-1β. Cancer Discov. 2022, 12, 2140–2157. [Google Scholar] [CrossRef] [PubMed]
- Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef] [PubMed]
- Sandhu, J.K.; Privora, H.F.; Wenckebach, G.; Birnboim, H.C. Neutrophils, nitric oxide synthase, and mutations in the mutatect murine tumor model. Am. J. Pathol. 2000, 156, 509–518. [Google Scholar] [CrossRef]
- Wculek, S.K.; Bridgeman, V.L.; Peakman, F.; Malanchi, I. Early Neutrophil Responses to Chemical Carcinogenesis Shape Long-Term Lung Cancer Susceptibility. iScience 2020, 23, 101277. [Google Scholar] [CrossRef]
- Canli, O.; Nicolas, A.M.; Gupta, J.; Finkelmeier, F.; Goncharova, O.; Pesic, M.; Neumann, T.; Horst, D.; Lower, M.; Sahin, U.; et al. Myeloid Cell-Derived Reactive Oxygen Species Induce Epithelial Mutagenesis. Cancer Cell 2017, 32, 869–883. [Google Scholar] [CrossRef] [PubMed]
- Keane, M.P.; Belperio, J.A.; Xue, Y.Y.; Burdick, M.D.; Strieter, R.M. Depletion of CXCR2 inhibits tumor growth and angiogenesis in a murine model of lung cancer. J. Immunol. 2004, 172, 2853–2860. [Google Scholar] [CrossRef] [PubMed]
- Wallace, A.E.; Sales, K.J.; Catalano, R.D.; Anderson, R.A.; Williams, A.R.; Wilson, M.R.; Schwarze, J.; Wang, H.; Rossi, A.G.; Jabbour, H.N. Prostaglandin F2alpha-F-prostanoid receptor signaling promotes neutrophil chemotaxis via chemokine (C-X-C motif) ligand 1 in endometrial adenocarcinoma. Cancer Res. 2009, 69, 5726–5733. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.; Lu, X.; Dey, P.; Deng, P.; Wu, C.C.; Jiang, S.; Fang, Z.; Zhao, K.; Konaparthi, R.; Hua, S.; et al. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression. Cancer Discov. 2016, 6, 80–95. [Google Scholar] [CrossRef] [PubMed]
- Di Mitri, D.; Toso, A.; Chen, J.J.; Sarti, M.; Pinton, S.; Jost, T.R.; D’Antuono, R.; Montani, E.; Garcia-Escudero, R.; Guccini, I.; et al. Tumour-infiltrating Gr-1+ myeloid cells antagonize senescence in cancer. Nature 2014, 515, 134–137. [Google Scholar] [CrossRef] [PubMed]
- Jamieson, T.; Clarke, M.; Steele, C.W.; Samuel, M.S.; Neumann, J.; Jung, A.; Huels, D.; Olson, M.F.; Das, S.; Nibbs, R.J.; et al. Inhibition of CXCR2 profoundly suppresses inflammation-driven and spontaneous tumorigenesis. J. Clin. Investig. 2012, 122, 3127–3144. [Google Scholar] [CrossRef] [PubMed]
- Asfaha, S.; Dubeykovskiy, A.N.; Tomita, H.; Yang, X.; Stokes, S.; Shibata, W.; Friedman, R.A.; Ariyama, H.; Dubeykovskaya, Z.A.; Muthupalani, S.; et al. Mice that express human interleukin-8 have increased mobilization of immature myeloid cells, which exacerbates inflammation and accelerates colon carcinogenesis. Gastroenterology. 2013, 144, 155–166. [Google Scholar] [CrossRef] [PubMed]
- Katoh, H.; Wang, D.; Daikoku, T.; Sun, H.; Dey, S.K.; Dubois, R.N. CXCR2-expressing myeloid-derived suppressor cells are essential to promote colitis-associated tumorigenesis. Cancer Cell 2013, 24, 631–644. [Google Scholar] [CrossRef] [PubMed]
- Shamaladevi, N.; Lyn, D.A.; Escudero, D.O.; Lokeshwar, B.L. CXC receptor-1 silencing inhibits androgen-independent prostate cancer. Cancer Res. 2009, 69, 8265–8274. [Google Scholar] [CrossRef] [PubMed]
- Kitadai, Y.; Haruma, K.; Sumii, K.; Yamamoto, S.; Ue, T.; Yokozaki, H.; Yasui, W.; Ohmoto, Y.; Kajiyama, G.; Fidler, I.J.; et al. Expression of interleukin-8 correlates with vascularity in human gastric carcinomas. Am. J. Pathol. 1998, 152, 93–100. [Google Scholar] [PubMed]
- Arenberg, D.A.; Kunkel, S.L.; Polverini, P.J.; Glass, M.; Burdick, M.D.; Strieter, R.M. Inhibition of interleukin-8 reduces tumorigenesis of human non-small cell lung cancer in SCID mice. J. Clin. Investig. 1996, 97, 2792–2802. [Google Scholar] [CrossRef]
- Smith, D.R.; Polverini, P.J.; Kunkel, S.L.; Orringer, M.B.; Whyte, R.I.; Burdick, M.D.; Wilke, C.A.; Strieter, R.M. Inhibition of interleukin 8 attenuates angiogenesis in bronchogenic carcinoma. J. Exp. Med. 1994, 179, 1409–1415. [Google Scholar] [CrossRef]
- Jablonska, J.; Leschner, S.; Westphal, K.; Lienenklaus, S.; Weiss, S. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J. Clin. Investig. 2010, 120, 1151–1164. [Google Scholar] [CrossRef]
- Queen, M.M.; Ryan, R.E.; Holzer, R.G.; Keller-Peck, C.R.; Jorcyk, C.L. Breast cancer cells stimulate neutrophils to produce oncostatin M: Potential implications for tumor progression. Cancer Res. 2005, 65, 8896–8904. [Google Scholar] [CrossRef] [PubMed]
- Benelli, R.; Morini, M.; Carrozzino, F.; Ferrari, N.; Minghelli, S.; Santi, L.; Cassatella, M.; Noonan, D.M.; Albini, A. Neutrophils as a key cellular target for angiostatin: Implications for regulation of angiogenesis and inflammation. FASEB J. 2002, 16, 267–269. [Google Scholar] [CrossRef] [PubMed]
- Gong, L.; Cumpian, A.M.; Caetano, M.S.; Ochoa, C.E.; De la Garza, M.M.; Lapid, D.J.; Mirabolfathinejad, S.G.; Dickey, B.F.; Zhou, Q.; Moghaddam, S.J. Promoting effect of neutrophils on lung tumorigenesis is mediated by CXCR2 and neutrophil elastase. Mol. Cancer 2013, 12, 154. [Google Scholar] [CrossRef] [PubMed]
- Ai, S.; Cheng, X.W.; Inoue, A.; Nakamura, K.; Okumura, K.; Iguchi, A.; Murohara, T.; Kuzuya, M. Angiogenic activity of bFGF and VEGF suppressed by proteolytic cleavage by neutrophil elastase. Biochem. Biophys. Res. Commun. 2007, 364, 395–401. [Google Scholar] [CrossRef]
- Zhang, H.; Ye, Y.L.; Li, M.X.; Ye, S.B.; Huang, W.R.; Cai, T.T.; He, J.; Peng, J.Y.; Duan, T.H.; Cui, J.; et al. CXCL2/MIF-CXCR2 signaling promotes the recruitment of myeloid-derived suppressor cells and is correlated with prognosis in bladder cancer. Oncogene 2017, 36, 2095–2104. [Google Scholar] [CrossRef]
- Rotondo, R.; Barisione, G.; Mastracci, L.; Grossi, F.; Orengo, A.M.; Costa, R.; Truini, M.; Fabbi, M.; Ferrini, S.; Barbieri, O. IL-8 induces exocytosis of arginase 1 by neutrophil polymorphonuclears in nonsmall cell lung cancer. Int. J. Cancer 2009, 125, 887–893. [Google Scholar] [CrossRef]
- Bianchi, A.; De Castro Silva, I.; Deshpande, N.U.; Singh, S.; Mehra, S.; Garrido, V.T.; Guo, X.; Nivelo, L.A.; Kolonias, D.S.; Saigh, S.J.; et al. Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer. Cancer Discov. 2023, 13, 1428–1453. [Google Scholar] [CrossRef]
- Yang, J.; Yan, C.; Vilgelm, A.E.; Chen, S.C.; Ayers, G.D.; Johnson, C.A.; Richmond, A. Targeted Deletion of CXCR2 in Myeloid Cells Alters the Tumor Immune Environment to Improve Antitumor Immunity. Cancer Immunol. Res. 2021, 9, 200–213. [Google Scholar] [CrossRef] [PubMed]
- Teijeira, A.; Garasa, S.; Gato, M.; Alfaro, C.; Migueliz, I.; Cirella, A.; de Andrea, C.; Ochoa, M.C.; Otano, I.; Etxeberria, I.; et al. CXCR1 and CXCR2 Chemokine Receptor Agonists Produced by Tumors Induce Neutrophil Extracellular Traps that Interfere with Immune Cytotoxicity. Immunity 2020, 52, 856–871. [Google Scholar] [CrossRef]
- Molon, B.; Ugel, S.; Del Pozzo, F.; Soldani, C.; Zilio, S.; Avella, D.; De Palma, A.; Mauri, P.; Monegal, A.; Rescigno, M.; et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 2011, 208, 1949–1962. [Google Scholar] [CrossRef]
- Schouppe, E.; Mommer, C.; Movahedi, K.; Laoui, D.; Morias, Y.; Gysemans, C.; Luyckx, A.; De Baetselier, P.; Van Ginderachter, J.A. Tumor-induced myeloid-derived suppressor cell subsets exert either inhibitory or stimulatory effects on distinct CD8+ T-cell activation events. Eur. J. Immunol. 2013, 43, 2930–2942. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.L.; Ye, S.B.; OuYang, L.Y.; Zhang, H.; Chen, Y.S.; He, J.; Chen, Q.Y.; Qian, C.N.; Zhang, X.S.; Cui, J.; et al. COX-2 promotes metastasis in nasopharyngeal carcinoma by mediating interactions between cancer cells and myeloid-derived suppressor cells. Oncoimmunology 2015, 4, e1044712. [Google Scholar] [CrossRef]
- Peinado, H.; Olmeda, D.; Cano, A. Snail, Zeb and bHLH factors in tumour progression: An alliance against the epithelial phenotype? Nat. Rev. Cancer 2007, 7, 415–428. [Google Scholar] [CrossRef]
- Wang, D.; Sun, H.; Wei, J.; Cen, B.; DuBois, R.N. CXCL1 Is Critical for Premetastatic Niche Formation and Metastasis in Colorectal Cancer. Cancer Res. 2017, 77, 3655–3665. [Google Scholar] [CrossRef]
- Sano, M.; Ijichi, H.; Takahashi, R.; Miyabayashi, K.; Fujiwara, H.; Yamada, T.; Kato, H.; Nakatsuka, T.; Tanaka, Y.; Tateishi, K.; et al. Blocking CXCLs-CXCR2 axis in tumor-stromal interactions contributes to survival in a mouse model of pancreatic ductal adenocarcinoma through reduced cell invasion/migration and a shift of immune-inflammatory microenvironment. Oncogenesis 2019, 8, 8. [Google Scholar] [CrossRef] [PubMed]
- Cui, D.; Zhao, Y.; Xu, J. Activated CXCL5-CXCR2 axis promotes the migration, invasion and EMT of papillary thyroid carcinoma cells via modulation of beta-catenin pathway. Biochimie 2018, 148, 1–11. [Google Scholar] [CrossRef]
- Xing, F.; Liu, Y.; Sharma, S.; Wu, K.; Chan, M.D.; Lo, H.W.; Carpenter, R.L.; Metheny-Barlow, L.J.; Zhou, X.; Qasem, S.A.; et al. Activation of the c-Met Pathway Mobilizes an Inflammatory Network in the Brain Microenvironment to Promote Brain Metastasis of Breast Cancer. Cancer Res. 2016, 76, 4970–4980. [Google Scholar] [CrossRef]
- Hasenberg, A.; Hasenberg, M.; Mann, L.; Neumann, F.; Borkenstein, L.; Stecher, M.; Kraus, A.; Engel, D.R.; Klingberg, A.; Seddigh, P.; et al. Catchup: A mouse model for imaging-based tracking and modulation of neutrophil granulocytes. Nat. Methods 2015, 12, 445–452. [Google Scholar] [CrossRef] [PubMed]
- Linde, N.; Casanova-Acebes, M.; Sosa, M.S.; Rahman, A.; Farias, E.; Harper, K.; Tardio, E.; Reyes Torres, I.; Jones, J.; Condeelis, J.; et al. Macrophages orchestrate breast cancer early dissemination and metastasis. Nat. Commun. 2018, 9, 21. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Cheng, S.; Wang, Y.; Wang, M.; Lu, Y.; Wen, Z.; Ge, Y.; Ma, Q.; Chen, Y.; Zhang, Y.; et al. Interrogation of the microenvironmental landscape in spinal ependymomas reveals dual functions of tumor-associated macrophages. Nat. Commun. 2021, 12, 6867. [Google Scholar] [CrossRef] [PubMed]
- Pombo Antunes, A.R.; Scheyltjens, I.; Lodi, F.; Messiaen, J.; Antoranz, A.; Duerinck, J.; Kancheva, D.; Martens, L.; De Vlaminck, K.; Van Hove, H.; et al. Single-cell profiling of myeloid cells in glioblastoma across species and disease stage reveals macrophage competition and specialization. Nat. Neurosci. 2021, 24, 595–610. [Google Scholar] [CrossRef] [PubMed]
- Pelka, K.; Hofree, M.; Chen, J.H.; Sarkizova, S.; Pirl, J.D.; Jorgji, V.; Bejnood, A.; Dionne, D.; Ge, W.H.; Xu, K.H.; et al. Spatially organized multicellular immune hubs in human colorectal cancer. Cell 2021, 184, 4734–4752. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.; He, H.; Liu, H.; Li, R.; Chen, Y.; Qi, Y.; Jiang, Q.; Chen, L.; Zhang, P.; Zhang, H.; et al. Tumour-associated macrophages-derived CXCL8 determines immune evasion through autonomous PD-L1 expression in gastric cancer. Gut 2019, 68, 1764–1773. [Google Scholar] [CrossRef] [PubMed]
- Langhans, B.; Kramer, B.; Louis, M.; Nischalke, H.D.; Huneburg, R.; Staratschek-Jox, A.; Odenthal, M.; Manekeller, S.; Schepke, M.; Kalff, J.; et al. Intrahepatic IL-8 producing Foxp3+ CD4+ regulatory T cells and fibrogenesis in chronic hepatitis C. J. Hepatol. 2013, 59, 229–235. [Google Scholar] [CrossRef] [PubMed]
- Morlacchi, S.; Dal Secco, V.; Soldani, C.; Glaichenhaus, N.; Viola, A.; Sarukhan, A. Regulatory T cells target chemokine secretion by dendritic cells independently of their capacity to regulate T cell proliferation. J. Immunol. 2011, 186, 6807–6814. [Google Scholar] [CrossRef] [PubMed]
- Otten, M.A.; Bakema, J.E.; Tuk, C.W.; Glennie, M.J.; Tutt, A.L.; Beelen, R.H.; van de Winkel, J.G.; van Egmond, M. Enhanced FcalphaRI-mediated neutrophil migration towards tumour colonies in the presence of endothelial cells. Eur. J. Immunol. 2012, 42, 1815–1821. [Google Scholar] [CrossRef]
- Sharma, A.; Seow, J.J.W.; Dutertre, C.A.; Pai, R.; Bleriot, C.; Mishra, A.; Wong, R.M.M.; Singh, G.S.N.; Sudhagar, S.; Khalilnezhad, S.; et al. Onco-fetal Reprogramming of Endothelial Cells Drives Immunosuppressive Macrophages in Hepatocellular Carcinoma. Cell 2020, 183, 377–394. [Google Scholar] [CrossRef]
- Jeong, H.Y.; Ham, I.H.; Lee, S.H.; Ryu, D.; Son, S.Y.; Han, S.U.; Kim, T.M.; Hur, H. Spatially Distinct Reprogramming of the Tumor Microenvironment Based On Tumor Invasion in Diffuse-Type Gastric Cancers. Clin. Cancer Res. 2021, 27, 6529–6542. [Google Scholar] [CrossRef] [PubMed]
- Xie, Y.; He, L.; Lugano, R.; Zhang, Y.; Cao, H.; He, Q.; Chao, M.; Liu, B.; Cao, Q.; Wang, J.; et al. Key molecular alterations in endothelial cells in human glioblastoma uncovered through single-cell RNA sequencing. JCI Insight 2021, 6, e150861. [Google Scholar] [CrossRef] [PubMed]
- Alsina-Sanchis, E.; Mulfarth, R.; Moll, I.; Bohn, S.; Wiedmann, L.; Jordana-Urriza, L.; Ziegelbauer, T.; Zimmer, E.; Taylor, J.; De Angelis Rigotti, F.; et al. Endothelial RBPJ Is Essential for the Education of Tumor-Associated Macrophages. Cancer Res. 2022, 82, 4414–4428. [Google Scholar] [CrossRef] [PubMed]
- Nagashima, K.; Sawa, S.; Nitta, T.; Tsutsumi, M.; Okamura, T.; Penninger, J.M.; Nakashima, T.; Takayanagi, H. Identification of subepithelial mesenchymal cells that induce IgA and diversify gut microbiota. Nat. Immunol. 2017, 18, 675–682. [Google Scholar] [CrossRef] [PubMed]
- Reboldi, A.; Coisne, C.; Baumjohann, D.; Benvenuto, F.; Bottinelli, D.; Lira, S.; Uccelli, A.; Lanzavecchia, A.; Engelhardt, B.; Sallusto, F. C-C chemokine receptor 6-regulated entry of Th-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat. Immunol. 2009, 10, 514–523. [Google Scholar] [CrossRef] [PubMed]
- Hippe, A.; Braun, S.A.; Olah, P.; Gerber, P.A.; Schorr, A.; Seeliger, S.; Holtz, S.; Jannasch, K.; Pivarcsi, A.; Buhren, B.; et al. EGFR/Ras-induced CCL20 production modulates the tumour microenvironment. Br. J. Cancer 2020, 123, 942–954. [Google Scholar] [CrossRef] [PubMed]
- Zhang, R.; Dong, M.; Tu, J.; Li, F.; Deng, Q.; Xu, J.; He, X.; Ding, J.; Xia, J.; Sheng, D.; et al. PMN-MDSCs modulated by CCL20 from cancer cells promoted breast cancer cell stemness through CXCL2-CXCR2 pathway. Signal Transduct Target Ther. 2023, 8, 97. [Google Scholar] [CrossRef]
- Martin-Garcia, D.; Silva-Vilches, C.; Will, R.; Enk, A.H.; Lonsdorf, A.S. Tumor-derived CCL20 affects B16 melanoma growth in mice. J. Dermatol. Sci. 2020, 97, 57–65. [Google Scholar] [CrossRef] [PubMed]
- Aspord, C.; Pedroza-Gonzalez, A.; Gallegos, M.; Tindle, S.; Burton, E.C.; Su, D.; Marches, F.; Banchereau, J.; Palucka, A.K. Breast cancer instructs dendritic cells to prime interleukin 13-secreting CD4+ T cells that facilitate tumor development. J. Exp. Med. 2007, 204, 1037–1047. [Google Scholar] [CrossRef]
- Bell, D.; Chomarat, P.; Broyles, D.; Netto, G.; Harb, G.M.; Lebecque, S.; Valladeau, J.; Davoust, J.; Palucka, K.A.; Banchereau, J. In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J. Exp. Med. 1999, 190, 1417–1426. [Google Scholar] [CrossRef]
- Barrio-Alonso, C.; Nieto-Valle, A.; Garcia-Martinez, E.; Gutierrez-Seijo, A.; Parra-Blanco, V.; Marquez-Rodas, I.; Aviles-Izquierdo, J.A.; Sanchez-Mateos, P.; Samaniego, R. Chemokine profiling of melanoma-macrophage crosstalk identifies CCL8 and CCL15 as prognostic factors in cutaneous melanoma. J. Pathol. 2024, 262, 495–504. [Google Scholar] [CrossRef] [PubMed]
- Bonnotte, B.; Crittenden, M.; Larmonier, N.; Gough, M.; Vile, R.G. MIP-3alpha transfection into a rodent tumor cell line increases intratumoral dendritic cell infiltration but enhances (facilitates) tumor growth and decreases immunogenicity. J. Immunol. 2004, 173, 4929–4935. [Google Scholar] [CrossRef] [PubMed]
- Jiao, X.; Velasco-Velazquez, M.A.; Wang, M.; Li, Z.; Rui, H.; Peck, A.R.; Korkola, J.E.; Chen, X.; Xu, S.; DuHadaway, J.B.; et al. CCR5 Governs DNA Damage Repair and Breast Cancer Stem Cell Expansion. Cancer Res. 2018, 78, 1657–1671. [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]
- Hawila, E.; Razon, H.; Wildbaum, G.; Blattner, C.; Sapir, Y.; Shaked, Y.; Umansky, V.; Karin, N. CCR5 Directs the Mobilization of CD11b(+)Gr1(+)Ly6C(low) Polymorphonuclear Myeloid Cells from the Bone Marrow to the Blood to Support Tumor Development. Cell Rep. 2017, 21, 2212–2222. [Google Scholar] [CrossRef] [PubMed]
- Ban, Y.; Mai, J.; Li, X.; Mitchell-Flack, M.; Zhang, T.; Zhang, L.; Chouchane, L.; Ferrari, M.; Shen, H.; Ma, X. Targeting Autocrine CCL5-CCR5 Axis Reprograms Immunosuppressive Myeloid Cells and Reinvigorates Antitumor Immunity. Cancer Res. 2017, 77, 2857–2868. [Google Scholar] [CrossRef] [PubMed]
- Imai, T.; Hieshima, K.; Haskell, C.; Baba, M.; Nagira, M.; Nishimura, M.; Kakizaki, M.; Takagi, S.; Nomiyama, H.; Schall, T.J.; et al. Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion. Cell 1997, 91, 521–530. [Google Scholar] [CrossRef] [PubMed]
- Busada, J.T.; Ramamoorthy, S.; Cain, D.W.; Xu, X.; Cook, D.N.; Cidlowski, J.A. Endogenous glucocorticoids prevent gastric metaplasia by suppressing spontaneous inflammation. J. Clin. Investig. 2019, 129, 1345–1358. [Google Scholar] [CrossRef] [PubMed]
- Yamauchi, T.; Hoki, T.; Oba, T.; Saito, H.; Attwood, K.; Sabel, M.S.; Chang, A.E.; Odunsi, K.; Ito, F. CX3CR1-CD8+ T cells are critical in antitumor efficacy but functionally suppressed in the tumor microenvironment. JCI Insight 2020, 5, e133920. [Google Scholar] [CrossRef]
- Gurler Main, H.; Xie, J.; Muralidhar, G.G.; Elfituri, O.; Xu, H.; Kajdacsy-Balla, A.A.; Barbolina, M.V. Emergent role of the fractalkine axis in dissemination of peritoneal metastasis from epithelial ovarian carcinoma. Oncogene 2017, 36, 3025–3036. [Google Scholar] [CrossRef]
- Schmall, A.; Al-Tamari, H.M.; Herold, S.; Kampschulte, M.; Weigert, A.; Wietelmann, A.; Vipotnik, N.; Grimminger, F.; Seeger, W.; Pullamsetti, S.S.; et al. Macrophage and cancer cell cross-talk via CCR2 and CX3CR1 is a fundamental mechanism driving lung cancer. Am. J. Respir. Crit. Care Med. 2015, 191, 437–447. [Google Scholar] [CrossRef]
- Chaudhri, A.; Bu, X.; Wang, Y.; Gomez, M.; Torchia, J.A.; Hua, P.; Hung, S.H.; Davies, M.A.; Lizee, G.A.; von Andrian, U.; et al. The CX3CL1-CX3CR1 chemokine axis can contribute to tumor immune evasion and blockade with a novel CX3CR1 monoclonal antibody enhances response to anti-PD-1 immunotherapy. Front. Immunol. 2023, 14, 1237715. [Google Scholar] [CrossRef] [PubMed]
- Nukiwa, M.; Andarini, S.; Zaini, J.; Xin, H.; Kanehira, M.; Suzuki, T.; Fukuhara, T.; Mizuguchi, H.; Hayakawa, T.; Saijo, Y.; et al. Dendritic cells modified to express fractalkine/CX3CL1 in the treatment of preexisting tumors. Eur. J. Immunol. 2006, 36, 1019–1027. [Google Scholar] [CrossRef] [PubMed]
- DiNatale, A.; Kaur, R.; Qian, C.; Zhang, J.; Marchioli, M.; Ipe, D.; Castelli, M.; McNair, C.M.; Kumar, G.; Meucci, O.; et al. Subsets of cancer cells expressing CX3CR1 are endowed with metastasis-initiating properties and resistance to chemotherapy. Oncogene 2022, 41, 1337–1351. [Google Scholar] [CrossRef] [PubMed]
- Ehrengruber, M.U.; Geiser, T.; Deranleau, D.A. Activation of human neutrophils by C3a and C5A. Comparison of the effects on shape changes, chemotaxis, secretion, and respiratory burst. FEBS Lett. 1994, 346, 181–184. [Google Scholar] [PubMed]
- Birdsall, H.H.; Green, D.M.; Trial, J.; Youker, K.A.; Burns, A.R.; MacKay, C.R.; LaRosa, G.J.; Hawkins, H.K.; Smith, C.W.; Michael, L.H.; et al. Complement C5a, TGF-beta 1, and MCP-1, in sequence, induce migration of monocytes into ischemic canine myocardium within the first one to five hours after reperfusion. Circulation 1997, 95, 684–692. [Google Scholar] [CrossRef] [PubMed]
- Vandendriessche, S.; Cambier, S.; Proost, P.; Marques, P.E. Complement Receptors and Their Role in Leukocyte Recruitment and Phagocytosis. Front. Cell Dev. Biol. 2021, 9, 624025. [Google Scholar] [CrossRef] [PubMed]
- Cho, M.S.; Vasquez, H.G.; Rupaimoole, R.; Pradeep, S.; Wu, S.; Zand, B.; Han, H.D.; Rodriguez-Aguayo, C.; Bottsford-Miller, J.; Huang, J.; et al. Autocrine effects of tumor-derived complement. Cell Rep. 2014, 6, 1085–1095. [Google Scholar] [CrossRef] [PubMed]
- Zha, H.; Wang, X.; Zhu, Y.; Chen, D.; Han, X.; Yang, F.; Gao, J.; Hu, C.; Shu, C.; Feng, Y.; et al. Intracellular Activation of Complement C3 Leads to PD-L1 Antibody Treatment Resistance by Modulating Tumor-Associated Macrophages. Cancer Immunol. Res. 2019, 7, 193–207. [Google Scholar] [CrossRef]
- Hsu, B.E.; Roy, J.; Mouhanna, J.; Rayes, R.F.; Ramsay, L.; Tabaries, S.; Annis, M.G.; Watson, I.R.; Spicer, J.D.; Costantino, S.; et al. C3a elicits unique migratory responses in immature low-density neutrophils. Oncogene 2020, 39, 2612–2623. [Google Scholar] [CrossRef]
- Markiewski, M.M.; DeAngelis, R.A.; Benencia, F.; Ricklin-Lichtsteiner, S.K.; Koutoulaki, A.; Gerard, C.; Coukos, G.; Lambris, J.D. Modulation of the antitumor immune response by complement. Nat. Immunol. 2008, 9, 1225–1235. [Google Scholar] [CrossRef]
- Wang, Y.; Sun, S.N.; Liu, Q.; Yu, Y.Y.; Guo, J.; Wang, K.; Xing, B.C.; Zheng, Q.F.; Campa, M.J.; Patz, E.F., Jr.; et al. Autocrine Complement Inhibits IL10-Dependent T-cell-Mediated Antitumor Immunity to Promote Tumor Progression. Cancer Discov. 2016, 6, 1022–1035. [Google Scholar] [CrossRef]
- Kieran, M.W.; Kalluri, R.; Cho, Y.J. The VEGF pathyway in cancer and disease: Responses, resistance, and resistance, and the path forward. Cold Spring Harb. Perspect. Med. 2012, 2, a006593. [Google Scholar] [CrossRef] [PubMed]
- Cao, Y.; E, G.; Wang, E.; Pal, K.; Dutta, S.K.; Bar-Sagi, D.; Mukhopadhyay, D. VEGF exerts an angiogenesis-independent function in cancer cells to promote their malignant progression. Cancer Res. 2012, 72, 3912–3918. [Google Scholar] [CrossRef]
- Banerjee, K.; Kerzel, T.; Bekkhus, T.; de Souza Ferreira, S.; Wallmann, T.; Wallerius, M.; Landwehr, L.S.; Agardy, D.A.; Schauer, N.; Malmerfeldt, A.; et al. VEGF-C-expressing TAMs rewire the metastatic fate of breast cancer cells. Cell Rep. 2023, 42, 113507. [Google Scholar] [CrossRef]
- Sawano, A.; Iwai, S.; Sakurai, Y.; Ito, M.; Shitara, K.; Nakahata, T.; Shibuya, M. Flt-1, vascular endothelial growth factor receptor 1, is a novel cell surface marker for the lineage of monocyte-macrophages in humans. Blood 2001, 97, 785–791. [Google Scholar] [CrossRef]
- Barleon, B.; Sozzani, S.; Zhou, D.; Weich, H.A.; Mantovani, A.; Marme, D. Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood 1996, 87, 3336–3343. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Huang, H.; Coleman, M.; Ziemys, A.; Gopal, P.; Kazmi, S.M.; Brekken, R.A. VEGFR2 activity on myeloid cells mediates immune suppression in the tumor microenvironment. JCI Insight 2021, 6, e150735. [Google Scholar] [CrossRef]
- Skobe, M.; Hamberg, L.M.; Hawighorst, T.; Schirner, M.; Wolf, G.L.; Alitalo, K.; Detmar, M. Concurrent induction of lymphangiogenesis, angiogenesis, and macrophage recruitment by vascular endothelial growth factor-C in melanoma. Am. J. Pathol. 2001, 159, 893–903. [Google Scholar] [CrossRef] [PubMed]
- Massena, S.; Christoffersson, G.; Vagesjo, E.; Seignez, C.; Gustafsson, K.; Binet, F.; Herrera, H.C.; Giraud, A.; Lomei, J.; Westrom, S.; et al. Identification and characterization of VEGF-A-responsive neutrophils expressing CD49d, VEGFR1, and CXCR4 in mice and humans. Blood 2015, 126, 2016–2026. [Google Scholar] [CrossRef]
- Cursiefen, C.; Chen, L.; Borges, L.P.; Jackson, D.; Cao, J.; Radziejewski, C.; D’Amore, P.A.; Dana, M.R.; Wiegand, S.J.; Streilein, J.W. VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment. J. Clin. Investig. 2004, 113, 1040–1050. [Google Scholar] [CrossRef] [PubMed]
- Murdoch, C.; Giannoudis, A.; Lewis, C.E. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 2004, 104, 2224–2234. [Google Scholar] [CrossRef] [PubMed]
- Afonso, P.V.; Janka-Junttila, M.; Lee, Y.J.; McCann, C.P.; Oliver, C.M.; Aamer, K.A.; Losert, W.; Cicerone, M.T.; Parent, C.A. LTB4 is a signal-relay molecule during neutrophil chemotaxis. Dev. Cell 2012, 22, 1079–1091. [Google Scholar] [CrossRef] [PubMed]
- Li, P.; Oh, D.Y.; Bandyopadhyay, G.; Lagakos, W.S.; Talukdar, S.; Osborn, O.; Johnson, A.; Chung, H.; Maris, M.; Ofrecio, J.M.; et al. LTB4 promotes insulin resistance in obese mice by acting on macrophages, hepatocytes and myocytes. Nat. Med. 2015, 21, 239–247. [Google Scholar] [CrossRef] [PubMed]
- Lee, E.K.S.; Gillrie, M.R.; Li, L.; Arnason, J.W.; Kim, J.H.; Babes, L.; Lou, Y.; Sanati-Nezhad, A.; Kyei, S.K.; Kelly, M.M.; et al. Leukotriene B4-Mediated Neutrophil Recruitment Causes Pulmonary Capillaritis during Lethal Fungal Sepsis. Cell Host Microbe 2018, 23, 121–133. [Google Scholar] [CrossRef] [PubMed]
- Lammermann, T.; Afonso, P.V.; Angermann, B.R.; Wang, J.M.; Kastenmuller, W.; Parent, C.A.; Germain, R.N. Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature 2013, 498, 371–375. [Google Scholar] [CrossRef] [PubMed]
- Serezani, C.H.; Divangahi, M.; Peters-Golden, M. Leukotrienes in Innate Immunity: Still Underappreciated after All These Years? J. Immunol. 2023, 210, 221–227. [Google Scholar] [CrossRef] [PubMed]
- Torres, R.M.; Cyster, J. Lipid mediators in the regulation of innate and adaptive immunity. Immunol. Rev. 2023, 317, 4–7. [Google Scholar] [CrossRef] [PubMed]
- Tian, W.; Jiang, X.; Kim, D.; Guan, T.; Nicolls, M.R.; Rockson, S.G. Leukotrienes in Tumor-Associated Inflammation. Front. Pharmacol. 2020, 11, 1289. [Google Scholar] [CrossRef] [PubMed]
- Higurashi, T.; Arimoto, J.; Ashikari, K.; Takatsu, T.; Misawa, N.; Yoshihara, T.; Matsuura, T.; Fuyuki, A.; Ohkubo, H.; Nakajima, A. The efficacy of a leukotriene receptor antagonist in the treatment of human rectal aberrant crypt foci: A nonrandomized, open-label, controlled trial. BMC Cancer 2020, 20, 770. [Google Scholar] [CrossRef] [PubMed]
- Maeda-Minami, A.; Hosokawa, M.; Ishikura, Y.; Onoda, A.; Kawano, Y.; Negishi, K.; Shimada, S.; Ihara, T.; Sugamata, M.; Takeda, K.; et al. Relationship Between Leukotriene Receptor Antagonists on Cancer Development in Patients With Bronchial Asthma: A Retrospective Analysis. Anticancer Res. 2022, 42, 3717–3724. [Google Scholar] [CrossRef] [PubMed]
- Jang, H.Y.; Kim, I.W.; Oh, J.M. Cysteinyl Leukotriene Receptor Antagonists Associated With a Decreased Incidence of Cancer: A Retrospective Cohort Study. Front. Oncol. 2022, 12, 858855. [Google Scholar] [CrossRef] [PubMed]
- Sutton, S.S.; Magagnoli, J.; Cummings, T.H.; Hardin, J.W. Leukotriene inhibition and the risk of lung cancer among U.S. veterans with asthma. Pulm. Pharmacol. Ther. 2021, 71, 102084. [Google Scholar] [CrossRef] [PubMed]
- Satapathy, S.R.; Ghatak, S.; Sjolander, A. The tumor promoter cysteinyl leukotriene receptor 1 regulates PD-L1 expression in colon cancer cells via the Wnt/β-catenin signaling axis. Cell Commun. Signal. 2023, 21, 138. [Google Scholar] [CrossRef] [PubMed]
- Tong, W.G.; Ding, X.Z.; Hennig, R.; Witt, R.C.; Standop, J.; Pour, P.M.; Adrian, T.E. Leukotriene B4 receptor antagonist LY293111 inhibits proliferation and induces apoptosis in human pancreatic cancer cells. Clin. Cancer Res. 2002, 8, 3232–3242. [Google Scholar] [PubMed]
- Ohd, J.F.; Nielsen, C.K.; Campbell, J.; Landberg, G.; Lofberg, H.; Sjolander, A. Expression of the leukotriene D4 receptor CysLT1, COX-2, and other cell survival factors in colorectal adenocarcinomas. Gastroenterology 2003, 124, 57–70. [Google Scholar] [CrossRef] [PubMed]
- Cheon, E.C.; Khazaie, K.; Khan, M.W.; Strouch, M.J.; Krantz, S.B.; Phillips, J.; Blatner, N.R.; Hix, L.M.; Zhang, M.; Dennis, K.L.; et al. Mast cell 5-lipoxygenase activity promotes intestinal polyposis in APCDelta468 mice. Cancer Res. 2011, 71, 1627–1636. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.; Cai, Q.; Chen, Y.; Shi, T.; Liu, W.; Mao, L.; Deng, B.; Ying, Z.; Gao, Y.; Luo, H.; et al. CAFs shape myeloid-derived suppressor cells to promote stemness of intrahepatic cholangiocarcinoma through 5-lipoxygenase. Hepatology 2022, 75, 28–42. [Google Scholar] [CrossRef] [PubMed]
- Yokota, Y.; Inoue, H.; Matsumura, Y.; Nabeta, H.; Narusawa, M.; Watanabe, A.; Sakamoto, C.; Hijikata, Y.; Iga-Murahashi, M.; Takayama, K.; et al. Absence of LTB4/BLT1 axis facilitates generation of mouse GM-CSF-induced long-lasting antitumor immunologic memory by enhancing innate and adaptive immune systems. Blood 2012, 120, 3444–3454. [Google Scholar] [CrossRef] [PubMed]
- Satpathy, S.R.; Jala, V.R.; Bodduluri, S.R.; Krishnan, E.; Hegde, B.; Hoyle, G.W.; Fraig, M.; Luster, A.D.; Haribabu, B. Crystalline silica-induced leukotriene B4-dependent inflammation promotes lung tumour growth. Nat. Commun. 2015, 6, 7064. [Google Scholar] [CrossRef] [PubMed]
- Allendorf, D.J.; Yan, J.; Ross, G.D.; Hansen, R.D.; Baran, J.T.; Subbarao, K.; Wang, L.; Haribabu, B. C5a-mediated leukotriene B4-amplified neutrophil chemotaxis is essential in tumor immunotherapy facilitated by anti-tumor monoclonal antibody and beta-glucan. J. Immunol. 2005, 174, 7050–7056. [Google Scholar] [CrossRef] [PubMed]
- Sharma, R.K.; Chheda, Z.; Jala, V.R.; Haribabu, B. Expression of leukotriene B(4) receptor-1 on CD8(+) T cells is required for their migration into tumors to elicit effective antitumor immunity. J. Immunol. 2013, 191, 3462–3470. [Google Scholar] [CrossRef] [PubMed]
- Hernandez, C.; Huebener, P.; Schwabe, R.F. Damage-associated molecular patterns in cancer: A double-edged sword. Oncogene 2016, 35, 5931–5941. [Google Scholar] [CrossRef] [PubMed]
- Hernandez, C.; Huebener, P.; Pradere, J.P.; Antoine, D.J.; Friedman, R.A.; Schwabe, R.F. HMGB1 links chronic liver injury to progenitor responses and hepatocarcinogenesis. J. Clin. Investig. 2018, 128, 2436–2451. [Google Scholar] [CrossRef] [PubMed]
- Huebener, P.; Pradere, J.P.; Hernandez, C.; Gwak, G.Y.; Caviglia, J.M.; Mu, X.; Loike, J.D.; Jenkins, R.E.; Antoine, D.J.; Schwabe, R.F. The HMGB1/RAGE axis triggers neutrophil-mediated injury amplification following necrosis. J. Clin. Investig. 2019, 130, 1802. [Google Scholar] [CrossRef] [PubMed]
- Tang, D.; Kang, R.; Zeh, H.J.; Lotze, M.T. The multifunctional protein HMGB1: 50 years of discovery. Nat. Rev. Immunol. 2023, 23, 824–841. [Google Scholar] [CrossRef] [PubMed]
- Rouhiainen, A.; Kuja-Panula, J.; Wilkman, E.; Pakkanen, J.; Stenfors, J.; Tuominen, R.K.; Lepantalo, M.; Carpen, O.; Parkkinen, J.; Rauvala, H. Regulation of monocyte migration by amphoterin (HMGB1). Blood 2004, 104, 1174–1182. [Google Scholar] [CrossRef] [PubMed]
- Bald, T.; Quast, T.; Landsberg, J.; Rogava, M.; Glodde, N.; Lopez-Ramos, D.; Kohlmeyer, J.; Riesenberg, S.; van den Boorn-Konijnenberg, D.; Homig-Holzel, C.; et al. Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature 2014, 507, 109–113. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Wu, K.; Zhao, E.; Shi, L.; Li, R.; Zhang, P.; Yin, Y.; Shuai, X.; Wang, G.; Tao, K. HMGB1 recruits myeloid derived suppressor cells to promote peritoneal dissemination of colon cancer after resection. Biochem. Biophys. Res. Commun. 2013, 436, 156–161. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.C.; Chen, K.C.; Chang, G.C.; Lin, H.; Wu, C.C.; Kao, W.H.; Teng, C.J.; Hsu, S.L.; Yang, T.Y. RAGE acts as an oncogenic role and promotes the metastasis of human lung cancer. Cell Death Dis. 2020, 11, 265. [Google Scholar] [CrossRef] [PubMed]
- Kuniyasu, H.; Oue, N.; Wakikawa, A.; Shigeishi, H.; Matsutani, N.; Kuraoka, K.; Ito, R.; Yokozaki, H.; Yasui, W. Expression of receptors for advanced glycation end-products (RAGE) is closely associated with the invasive and metastatic activity of gastric cancer. J. Pathol. 2002, 196, 163–170. [Google Scholar] [CrossRef] [PubMed]
- Gebhardt, C.; Riehl, A.; Durchdewald, M.; Nemeth, J.; Furstenberger, G.; Muller-Decker, K.; Enk, A.; Arnold, B.; Bierhaus, A.; Nawroth, P.P.; et al. RAGE signaling sustains inflammation and promotes tumor development. J. Exp. Med. 2008, 205, 275–285. [Google Scholar] [CrossRef] [PubMed]
- Vernon, P.J.; Loux, T.J.; Schapiro, N.E.; Kang, R.; Muthuswamy, R.; Kalinski, P.; Tang, D.; Lotze, M.T.; Zeh, H.J., 3rd. The receptor for advanced glycation end products promotes pancreatic carcinogenesis and accumulation of myeloid-derived suppressor cells. J. Immunol. 2013, 190, 1372–1379. [Google Scholar] [CrossRef] [PubMed]
- Vernon, P.J.; Zeh Iii, H.J.; Lotze, M.T. The myeloid response to pancreatic carcinogenesis is regulated by the receptor for advanced glycation end-products. Oncoimmunology 2013, 2, e24184. [Google Scholar] [CrossRef] [PubMed]
- Oshimi, Y.; Miyazaki, S.; Oda, S. ATP-induced Ca2+ response mediated by P2U and P2Y purinoceptors in human macrophages: Signalling from dying cells to macrophages. Immunology. 1999, 98, 220–227. [Google Scholar] [CrossRef] [PubMed]
- Blay, J.; White, T.D.; Hoskin, D.W. The extracellular fluid of solid carcinomas contains immunosuppressive concentrations of adenosine. Cancer Res. 1997, 57, 2602–2605. [Google Scholar] [PubMed]
- Di Virgilio, F.; Dal Ben, D.; Sarti, A.C.; Giuliani, A.L.; Falzoni, S. The P2X7 Receptor in Infection and Inflammation. Immunity 2017, 47, 15–31. [Google Scholar] [CrossRef] [PubMed]
- Mei, L.; Du, W.; Gao, W.; Mei, Q.B. Purinergic signaling: A novel mechanism in immune surveillance. Acta Pharmacol. Sin. 2010, 31, 1149–1153. [Google Scholar] [CrossRef] [PubMed]
- Elliott, M.R.; Ravichandran, K.S. The Dynamics of Apoptotic Cell Clearance. Dev. Cell 2016, 38, 147–160. [Google Scholar] [CrossRef] [PubMed]
- Rose, F.R.; Hirschhorn, R.; Weissmann, G.; Cronstein, B.N. Adenosine promotes neutrophil chemotaxis. J. Exp. Med. 1988, 167, 1186–1194. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Corriden, R.; Inoue, Y.; Yip, L.; Hashiguchi, N.; Zinkernagel, A.; Nizet, V.; Insel, P.A.; Junger, W.G. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science 2006, 314, 1792–1795. [Google Scholar] [CrossRef] [PubMed]
- Schnurr, M.; Toy, T.; Shin, A.; Hartmann, G.; Rothenfusser, S.; Soellner, J.; Davis, I.D.; Cebon, J.; Maraskovsky, E. Role of adenosine receptors in regulating chemotaxis and cytokine production of plasmacytoid dendritic cells. Blood 2004, 103, 1391–1397. [Google Scholar] [CrossRef] [PubMed]
- Elliott, M.R.; Chekeni, F.B.; Trampont, P.C.; Lazarowski, E.R.; Kadl, A.; Walk, S.F.; Park, D.; Woodson, R.I.; Ostankovich, M.; Sharma, P.; et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 2009, 461, 282–286. [Google Scholar] [CrossRef] [PubMed]
- Huang, Z.; Xie, N.; Illes, P.; Di Virgilio, F.; Ulrich, H.; Semyanov, A.; Verkhratsky, A.; Sperlagh, B.; Yu, S.G.; Huang, C.; et al. From purines to purinergic signalling: Molecular functions and human diseases. Signal Transduct Target Ther. 2021, 6, 162. [Google Scholar] [CrossRef] [PubMed]
- Ohta, A.; Gorelik, E.; Prasad, S.J.; Ronchese, F.; Lukashev, D.; Wong, M.K.; Huang, X.; Caldwell, S.; Liu, K.; Smith, P.; et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc. Natl. Acad. Sci. USA 2006, 103, 13132–13137. [Google Scholar] [CrossRef] [PubMed]
- Beavis, P.A.; Divisekera, U.; Paget, C.; Chow, M.T.; John, L.B.; Devaud, C.; Dwyer, K.; Stagg, J.; Smyth, M.J.; Darcy, P.K. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc. Natl. Acad. Sci. USA 2013, 110, 14711–14716. [Google Scholar] [CrossRef] [PubMed]
- Young, A.; Ngiow, S.F.; Barkauskas, D.S.; Sult, E.; Hay, C.; Blake, S.J.; Huang, Q.; Liu, J.; Takeda, K.; Teng, M.W.L.; et al. Co-inhibition of CD73 and A2AR Adenosine Signaling Improves Anti-tumor Immune Responses. Cancer Cell 2016, 30, 391–403. [Google Scholar] [CrossRef] [PubMed]
- Perrot, I.; Michaud, H.A.; Giraudon-Paoli, M.; Augier, S.; Docquier, A.; Gros, L.; Courtois, R.; Dejou, C.; Jecko, D.; Becquart, O.; et al. Blocking Antibodies Targeting the CD39/CD73 Immunosuppressive Pathway Unleash Immune Responses in Combination Cancer Therapies. Cell Rep. 2019, 27, 2411–2425. [Google Scholar] [CrossRef] [PubMed]
- Montalban Del Barrio, I.; Penski, C.; Schlahsa, L.; Stein, R.G.; Diessner, J.; Wockel, A.; Dietl, J.; Lutz, M.B.; Mittelbronn, M.; Wischhusen, J.; et al. Adenosine-generating ovarian cancer cells attract myeloid cells which differentiate into adenosine-generating tumor associated macrophages—A self-amplifying, CD39- and CD73-dependent mechanism for tumor immune escape. J. Immunother. Cancer 2016, 4, 49. [Google Scholar] [CrossRef] [PubMed]
- Li, X.Y.; Moesta, A.K.; Xiao, C.; Nakamura, K.; Casey, M.; Zhang, H.; Madore, J.; Lepletier, A.; Aguilera, A.R.; Sundarrajan, A.; et al. Targeting CD39 in Cancer Reveals an Extracellular ATP- and Inflammasome-Driven Tumor Immunity. Cancer Discov. 2019, 9, 1754–1773. [Google Scholar] [CrossRef] [PubMed]
- Lai, C.P.; Bechberger, J.F.; Thompson, R.J.; MacVicar, B.A.; Bruzzone, R.; Naus, C.C. Tumor-suppressive effects of pannexin 1 in C6 glioma cells. Cancer Res. 2007, 67, 1545–1554. [Google Scholar] [CrossRef] [PubMed]
- Penuela, S.; Gyenis, L.; Ablack, A.; Churko, J.M.; Berger, A.C.; Litchfield, D.W.; Lewis, J.D.; Laird, D.W. Loss of pannexin 1 attenuates melanoma progression by reversion to a melanocytic phenotype. J. Biol. Chem. 2012, 287, 29184–29193. [Google Scholar] [CrossRef] [PubMed]
- Furlow, P.W.; Zhang, S.; Soong, T.D.; Halberg, N.; Goodarzi, H.; Mangrum, C.; Wu, Y.G.; Elemento, O.; Tavazoie, S.F. Mechanosensitive pannexin-1 channels mediate microvascular metastatic cell survival. Nat. Cell Biol. 2015, 17, 943–952. [Google Scholar] [CrossRef] [PubMed]
- Pelegrin, P.; Surprenant, A. Pannexin-1 mediates large pore formation and interleukin-1beta release by the ATP-gated P2X7 receptor. EMBO J. 2006, 25, 5071–5082. [Google Scholar] [CrossRef] [PubMed]
- Caronni, N.; La Terza, F.; Vittoria, F.M.; Barbiera, G.; Mezzanzanica, L.; Cuzzola, V.; Barresi, S.; Pellegatta, M.; Canevazzi, P.; Dunsmore, G.; et al. IL-1beta(+) macrophages fuel pathogenic inflammation in pancreatic cancer. Nature 2023, 623, 415–422. [Google Scholar] [CrossRef] [PubMed]
- Wu, L.J.; Vadakkan, K.I.; Zhuo, M. ATP-induced chemotaxis of microglial processes requires P2Y receptor-activated initiation of outward potassium currents. Glia 2007, 55, 810–821. [Google Scholar] [CrossRef] [PubMed]
- Sáez, P.J.; Vargas, P.; Shoji, K.F.; Harcha, P.A.; Lennon-Duménil, A.M.; Sáez, J.C. ATP promotes the fast migration of dendritic cells through the activity of pannexin 1 channels and P2X7 receptors. Sci. Signal. 2017, 10, eaah7107. [Google Scholar] [CrossRef] [PubMed]
- Trabanelli, S.; Ocadlíková, D.; Gulinelli, S.; Curti, A.; Salvestrini, V.; Vieira, R.P.; Idzko, M.; Di Virgilio, F.; Ferrari, D.; Lemoli, R.M. Extracellular ATP exerts opposite effects on activated and regulatory CD4+ T cells via purinergic P2 receptor activation. J. Immunol. 2012, 189, 1303–1310. [Google Scholar] [CrossRef] [PubMed]
- Di Virgilio, F.; Adinolfi, E. Extracellular purines, purinergic receptors and tumor growth. Oncogene 2017, 36, 293–303. [Google Scholar] [CrossRef]
- Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef]
- Ferrari, D.; Chiozzi, P.; Falzoni, S.; Dal Susino, M.; Melchiorri, L.; Baricordi, O.R.; Di Virgilio, F. Extracellular ATP triggers IL-1 beta release by activating the purinergic P2Z receptor of human macrophages. J. Immunol. 1997, 159, 1451–1458. [Google Scholar] [CrossRef] [PubMed]
- Kurashima, Y.; Amiya, T.; Nochi, T.; Fujisawa, K.; Haraguchi, T.; Iba, H.; Tsutsui, H.; Sato, S.; Nakajima, S.; Iijima, H.; et al. Extracellular ATP mediates mast cell-dependent intestinal inflammation through P2X7 purinoceptors. Nat. Commun. 2012, 3, 1034. [Google Scholar] [CrossRef] [PubMed]
- Adinolfi, E.; Capece, M.; Franceschini, A.; Falzoni, S.; Giuliani, A.L.; Rotondo, A.; Sarti, A.C.; Bonora, M.; Syberg, S.; Corigliano, D.; et al. Accelerated tumor progression in mice lacking the ATP receptor P2X7. Cancer Res. 2015, 75, 635–644. [Google Scholar] [CrossRef] [PubMed]
- Qin, J.; Zhang, X.; Tan, B.; Zhang, S.; Yin, C.; Xue, Q.; Zhang, Z.; Ren, H.; Chen, J. Blocking P2X7-Mediated Macrophage Polarization Overcomes Treatment Resistance in Lung Cancer. Cancer Immunol. Res. 2020, 8, 1426–1439. [Google Scholar] [CrossRef] [PubMed]
- Romagnani, A.; Rottoli, E.; Mazza, E.M.C.; Rezzonico-Jost, T.; De Ponte Conti, B.; Proietti, M.; Perotti, M.; Civanelli, E.; Perruzza, L.; Catapano, A.L.; et al. P2X7 Receptor Activity Limits Accumulation of T Cells within Tumors. Cancer Res. 2020, 80, 3906–3919. [Google Scholar] [CrossRef] [PubMed]
- Tatsumi, K. The pathogenesis of cancer-associated thrombosis. Int. J. Hematol. 2024, 119, 495–504. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.R.; Yousef, G.M.; Ni, H. Cancer and platelet crosstalk: Opportunities and challenges for aspirin and other antiplatelet agents. Blood 2018, 131, 1777–1789. [Google Scholar] [CrossRef] [PubMed]
- Camerer, E.; Qazi, A.A.; Duong, D.N.; Cornelissen, I.; Advincula, R.; Coughlin, S.R. Platelets, protease-activated receptors, and fibrinogen in hematogenous metastasis. Blood 2004, 104, 397–401. [Google Scholar] [CrossRef]
- Ho-Tin-Noe, B.; Goerge, T.; Cifuni, S.M.; Duerschmied, D.; Wagner, D.D. Platelet granule secretion continuously prevents intratumor hemorrhage. Cancer Res. 2008, 68, 6851–6858. [Google Scholar] [CrossRef] [PubMed]
- Mueller, B.M.; Reisfeld, R.A.; Edgington, T.S.; Ruf, W. Expression of tissue factor by melanoma cells promotes efficient hematogenous metastasis. Proc. Natl. Acad. Sci. USA 1992, 89, 11832–11836. [Google Scholar] [CrossRef] [PubMed]
- Nierodzik, M.L.; Plotkin, A.; Kajumo, F.; Karpatkin, S. Thrombin stimulates tumor-platelet adhesion in vitro and metastasis in vivo. J. Clin. Investig. 1991, 87, 229–236. [Google Scholar] [CrossRef] [PubMed]
- Zhang, P.; Ozdemir, T.; Chung, C.Y.; Robertson, G.P.; Dong, C. Sequential binding of αVβ3 and ICAM-1 determines fibrin-mediated melanoma capture and stable adhesion to CD11b/CD18 on neutrophils. J. Immunol. 2011, 186, 242–254. [Google Scholar] [CrossRef] [PubMed]
- Borsig, L.; Wong, R.; Feramisco, J.; Nadeau, D.R.; Varki, N.M.; Varki, A. Heparin and cancer revisited: Mechanistic connections involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. Proc. Natl. Acad. Sci. USA 2001, 98, 3352–3357. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.J.; Borsig, L.; Han, H.L.; Varki, N.M.; Varki, A. Distinct selectin ligands on colon carcinoma mucins can mediate pathological interactions among platelets, leukocytes, and endothelium. Am. J. Pathol. 1999, 155, 461–472. [Google Scholar] [CrossRef] [PubMed]
- Lecot, P.; Ardin, M.; Dussurgey, S.; Alcazer, V.; Moudombi, L.; Pereira Abrantes, M.; Hubert, M.; Swalduz, A.; Hernandez-Vargas, H.; Viari, A.; et al. Gene signature of circulating platelet-bound neutrophils is associated with poor prognosis in cancer patients. Int. J. Cancer 2022, 151, 138–152. [Google Scholar] [CrossRef] [PubMed]
- Labelle, M.; Begum, S.; Hynes, R.O. Platelets guide the formation of early metastatic niches. Proc. Natl. Acad. Sci. USA 2014, 111, E3053–E3061. [Google Scholar] [CrossRef] [PubMed]
- Gil-Bernabe, A.M.; Ferjancic, S.; Tlalka, M.; Zhao, L.; Allen, P.D.; Im, J.H.; Watson, K.; Hill, S.A.; Amirkhosravi, A.; Francis, J.L.; et al. Recruitment of monocytes/macrophages by tissue factor-mediated coagulation is essential for metastatic cell survival and premetastatic niche establishment in mice. Blood 2012, 119, 3164–3175. [Google Scholar] [CrossRef] [PubMed]
- Feinauer, M.J.; Schneider, S.W.; Berghoff, A.S.; Robador, J.R.; Tehranian, C.; Karreman, M.A.; Venkataramani, V.; Solecki, G.; Grosch, J.K.; Gunkel, K.; et al. Local blood coagulation drives cancer cell arrest and brain metastasis in a mouse model. Blood 2021, 137, 1219–1232. [Google Scholar] [CrossRef] [PubMed]
- Im, J.H.; Fu, W.; Wang, H.; Bhatia, S.K.; Hammer, D.A.; Kowalska, M.A.; Muschel, R.J. Coagulation facilitates tumor cell spreading in the pulmonary vasculature during early metastatic colony formation. Cancer Res. 2004, 64, 8613–8619. [Google Scholar] [CrossRef] [PubMed]
- Karpatkin, S.; Pearlstein, E.; Ambrogio, C.; Coller, B.S. Role of adhesive proteins in platelet tumor interaction in vitro and metastasis formation in vivo. J. Clin. Investig. 1988, 81, 1012–1019. [Google Scholar] [CrossRef] [PubMed]
- Bauer, A.T.; Gorzelanny, C.; Gebhardt, C.; Pantel, K.; Schneider, S.W. Interplay between coagulation and inflammation in cancer: Limitations and therapeutic opportunities. Cancer Treat. Rev. 2022, 102, 102322. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Lu, Z.; Wu, S.; Chu, T.; Li, B.; Qi, F.; Zhao, Y.; Nie, G. The dynamic role of platelets in cancer progression and their therapeutic implications. Nat. Rev. Cancer 2024, 24, 72–87. [Google Scholar] [CrossRef] [PubMed]
- Serrano, C.V., Jr.; Fraticelli, A.; Paniccia, R.; Teti, A.; Noble, B.; Corda, S.; Faraggiana, T.; Ziegelstein, R.C.; Zweier, J.L.; Capogrossi, M.C. pH dependence of neutrophil-endothelial cell adhesion and adhesion molecule expression. Am. J. Physiol. 1996, 271, C962–C970. [Google Scholar] [CrossRef] [PubMed]
- Shi, Q.; Le, X.; Wang, B.; Xiong, Q.; Abbruzzese, J.L.; Xie, K. Regulation of interleukin-8 expression by cellular pH in human pancreatic adenocarcinoma cells. J. Interferon Cytokine Res. 2000, 20, 1023–1028. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Xie, K.; Mukaida, N.; Matsushima, K.; Fidler, I.J. Hypoxia-induced elevation in interleukin-8 expression by human ovarian carcinoma cells. Cancer Res. 1999, 59, 5822–5829. [Google Scholar] [PubMed]
- Taylor, C.T.; Scholz, C.C. The effect of HIF on metabolism and immunity. Nat. Rev. Nephrol. 2022, 18, 573–587. [Google Scholar] [CrossRef] [PubMed]
- McGettrick, A.F.; O’Neill, L.A.J. The Role of HIF in Immunity and Inflammation. Cell Metab. 2020, 32, 524–536. [Google Scholar] [CrossRef] [PubMed]
- Petrova, V.; Annicchiarico-Petruzzelli, M.; Melino, G.; Amelio, I. The hypoxic tumour microenvironment. Oncogenesis 2018, 7, 10. [Google Scholar] [CrossRef] [PubMed]
- Chiu, D.K.; Xu, I.M.; Lai, R.K.; Tse, A.P.; Wei, L.L.; Koh, H.Y.; Li, L.L.; Lee, D.; Lo, R.C.; Wong, C.M.; et al. Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26. Hepatology 2016, 64, 797–813. [Google Scholar] [CrossRef] [PubMed]
- Nambiar, D.K.; Viswanathan, V.; Cao, H.; Zhang, W.; Guan, L.; Chamoli, M.; Holmes, B.; Kong, C.; Hildebrand, R.; Koong, A.J.; et al. Galectin-1 Mediates Chronic STING Activation in Tumors to Promote Metastasis through MDSC Recruitment. Cancer Res. 2023, 83, 3205–3219. [Google Scholar] [CrossRef] [PubMed]
- Cools-Lartigue, J.; Spicer, J.; McDonald, B.; Gowing, S.; Chow, S.; Giannias, B.; Bourdeau, F.; Kubes, P.; Ferri, L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Investig. 2013, 123, 3446–3458. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Liu, Q.; Zhang, X.; Liu, X.; Zhou, B.; Chen, J.; Huang, D.; Li, J.; Li, H.; Chen, F.; et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature 2020, 583, 133–138. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Li, N.; Yu, X.; Xiao, X.; Cheng, K.; Hu, J.; Wang, J.; Zhang, D.; Cheng, S.; Liu, S. Expression of intercellular adhesion molecule 1 by hepatocellular carcinoma stem cells and circulating tumor cells. Gastroenterology 2013, 144, 1031–1041. [Google Scholar] [CrossRef]
- Spicer, J.D.; McDonald, B.; Cools-Lartigue, J.J.; Chow, S.C.; Giannias, B.; Kubes, P.; Ferri, L.E. Neutrophils promote liver metastasis via Mac-1-mediated interactions with circulating tumor cells. Cancer Res. 2012, 72, 3919–3927. [Google Scholar] [CrossRef] [PubMed]
- Wu, Q.D.; Wang, J.H.; Condron, C.; Bouchier-Hayes, D.; Redmond, H.P. Human neutrophils facilitate tumor cell transendothelial migration. Am. J. Physiol. Cell Physiol. 2001, 280, C814–C822. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Chen, Q.; Zhang, X.H.; Massague, J. Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs. Cancer Cell 2011, 20, 538–549. [Google Scholar] [CrossRef] [PubMed]
- Evani, S.J.; Prabhu, R.G.; Gnanaruban, V.; Finol, E.A.; Ramasubramanian, A.K. Monocytes mediate metastatic breast tumor cell adhesion to endothelium under flow. FASEB J. 2013, 27, 3017–3029. [Google Scholar] [CrossRef] [PubMed]
- Lu, X.; Mu, E.; Wei, Y.; Riethdorf, S.; Yang, Q.; Yuan, M.; Yan, J.; Hua, Y.; Tiede, B.J.; Lu, X.; et al. VCAM-1 promotes osteolytic expansion of indolent bone micrometastasis of breast cancer by engaging alpha4beta1-positive osteoclast progenitors. Cancer Cell 2011, 20, 701–714. [Google Scholar] [CrossRef]
- Granot, Z.; Henke, E.; Comen, E.A.; King, T.A.; Norton, L.; Benezra, R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell 2011, 20, 300–314. [Google Scholar] [CrossRef]
- Hagerling, C.; Gonzalez, H.; Salari, K.; Wang, C.Y.; Lin, C.; Robles, I.; van Gogh, M.; Dejmek, A.; Jirstrom, K.; Werb, Z. Immune effector monocyte-neutrophil cooperation induced by the primary tumor prevents metastatic progression of breast cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 21704–21714. [Google Scholar] [CrossRef] [PubMed]
- Hanna, R.N.; Cekic, C.; Sag, D.; Tacke, R.; Thomas, G.D.; Nowyhed, H.; Herrley, E.; Rasquinha, N.; McArdle, S.; Wu, R.; et al. Patrolling monocytes control tumor metastasis to the lung. Science 2015, 350, 985–990. [Google Scholar] [CrossRef] [PubMed]
- Grisaru-Tal, S.; Dulberg, S.; Beck, L.; Zhang, C.; Itan, M.; Hediyeh-Zadeh, S.; Caldwell, J.; Rozenberg, P.; Dolitzky, A.; Avlas, S.; et al. Metastasis-Entrained Eosinophils Enhance Lymphocyte-Mediated Antitumor Immunity. Cancer Res. 2021, 81, 5555–5571. [Google Scholar] [CrossRef] [PubMed]
- Matlung, H.L.; Babes, L.; Zhao, X.W.; van Houdt, M.; Treffers, L.W.; van Rees, D.J.; Franke, K.; Schornagel, K.; Verkuijlen, P.; Janssen, H.; et al. Neutrophils Kill Antibody-Opsonized Cancer Cells by Trogoptosis. Cell Rep. 2018, 23, 3946–3959. [Google Scholar] [CrossRef] [PubMed]
- Vermi, W.; Micheletti, A.; Finotti, G.; Tecchio, C.; Calzetti, F.; Costa, S.; Bugatti, M.; Calza, S.; Agostinelli, C.; Pileri, S.; et al. slan(+) Monocytes and Macrophages Mediate CD20-Dependent B-cell Lymphoma Elimination via ADCC and ADCP. Cancer Res. 2018, 78, 3544–3559. [Google Scholar] [CrossRef] [PubMed]
- Szczerba, B.M.; Castro-Giner, F.; Vetter, M.; Krol, I.; Gkountela, S.; Landin, J.; Scheidmann, M.C.; Donato, C.; Scherrer, R.; Singer, J.; et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature 2019, 566, 553–557. [Google Scholar] [CrossRef] [PubMed]
- Simons, J.W.; Jaffee, E.M.; Weber, C.E.; Levitsky, H.I.; Nelson, W.G.; Carducci, M.A.; Lazenby, A.J.; Cohen, L.K.; Finn, C.C.; Clift, S.M.; et al. Bioactivity of autologous irradiated renal cell carcinoma vaccines generated by ex vivo granulocyte-macrophage colony-stimulating factor gene transfer. Cancer Res. 1997, 57, 1537–1546. [Google Scholar] [PubMed]
- Huang, Y.; Kim, B.Y.S.; Chan, C.K.; Hahn, S.M.; Weissman, I.L.; Jiang, W. Improving immune-vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 195–203. [Google Scholar] [CrossRef] [PubMed]
- Hillinger, S.; Yang, S.C.; Zhu, L.; Huang, M.; Duckett, R.; Atianzar, K.; Batra, R.K.; Strieter, R.M.; Dubinett, S.M.; Sharma, S. EBV-induced molecule 1 ligand chemokine (ELC/CCL19) promotes IFN-gamma-dependent antitumor responses in a lung cancer model. J. Immunol. 2003, 171, 6457–6465. [Google Scholar] [CrossRef] [PubMed]
- Foeng, J.; Comerford, I.; McColl, S.R. Harnessing the chemokine system to home CAR-T cells into solid tumors. Cell Rep. Med. 2022, 3, 100543. [Google Scholar] [CrossRef] [PubMed]
- Luo, H.; Su, J.; Sun, R.; Sun, Y.; Wang, Y.; Dong, Y.; Shi, B.; Jiang, H.; Li, Z. Coexpression of IL7 and CCL21 Increases Efficacy of CAR-T Cells in Solid Tumors without Requiring Preconditioned Lymphodepletion. Clin. Cancer Res. 2020, 26, 5494–5505. [Google Scholar] [CrossRef] [PubMed]
- Adachi, K.; Kano, Y.; Nagai, T.; Okuyama, N.; Sakoda, Y.; Tamada, K. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat. Biotechnol. 2018, 36, 346–351. [Google Scholar] [CrossRef] [PubMed]
- De Marco, R.C.; Monzo, H.J.; Ojala, P.M. CAR T Cell Therapy: A Versatile Living Drug. Int. J. Mol. Sci. 2023, 24, 6300. [Google Scholar] [CrossRef] [PubMed]
- Xu, Z.; Zhu, C.; Chen, C.; Zong, Y.; Feng, H.; Liu, D.; Feng, W.; Zhao, J.; Lu, A. CCL19 suppresses angiogenesis through promoting miR-206 and inhibiting Met/ERK/Elk-1/HIF-1alpha/VEGF-A pathway in colorectal cancer. Cell Death Dis. 2018, 9, 974. [Google Scholar] [CrossRef] [PubMed]
- Dangaj, D.; Bruand, M.; Grimm, A.J.; Ronet, C.; Barras, D.; Duttagupta, P.A.; Lanitis, E.; Duraiswamy, J.; Tanyi, J.L.; Benencia, F.; et al. Cooperation between Constitutive and Inducible Chemokines Enables T Cell Engraftment and Immune Attack in Solid Tumors. Cancer Cell 2019, 35, 885–900. [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]
- Wang, C.L.; Ho, A.S.; Chang, C.C.; Sie, Z.L.; Peng, C.L.; Chang, J.; Cheng, C.C. Radiotherapy enhances CXCR3(high)CD8(+) T cell activation through inducing IFNgamma-mediated CXCL10 and ICAM-1 expression in lung cancer cells. Cancer Immunol. Immunother. 2023, 72, 1865–1880. [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. [Google Scholar] [CrossRef] [PubMed]
- Qu, Y.; Wen, J.; Thomas, G.; Yang, W.; Prior, W.; He, W.; Sundar, P.; Wang, X.; Potluri, S.; Salek-Ardakani, S. Baseline Frequency of Inflammatory Cxcl9-Expressing Tumor-Associated Macrophages Predicts Response to Avelumab Treatment. Cell Rep. 2020, 32, 108115. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Pitt, L.A.; Tikhonova, A.N.; Hu, H.; Trimarchi, T.; King, B.; Gong, Y.; Sanchez-Martin, M.; Tsirigos, A.; Littman, D.R.; Ferrando, A.A.; et al. CXCL12-Producing Vascular Endothelial Niches Control Acute T Cell Leukemia Maintenance. Cancer Cell 2015, 27, 755–768. [Google Scholar] [CrossRef] [PubMed]
- Zeng, Y.; Li, B.; Liang, Y.; Reeves, P.M.; Qu, X.; Ran, C.; Liu, Q.; Callahan, M.V.; Sluder, A.E.; Gelfand, J.A.; et al. Dual blockade of CXCL12-CXCR4 and PD-1-PD-L1 pathways prolongs survival of ovarian tumor-bearing mice by prevention of immunosuppression in the tumor microenvironment. FASEB J. 2019, 33, 6596–6608. [Google Scholar] [CrossRef] [PubMed]
- Liu, B.; Zhang, Y.; Wang, D.; Hu, X.; Zhang, Z. Single-cell meta-analyses reveal responses of tumor-reactive CXCL13(+) T cells to immune-checkpoint blockade. Nat. Cancer 2022, 3, 1123–1136. [Google Scholar] [CrossRef] [PubMed]
- An, M.; Mehta, A.; Min, B.H.; Heo, Y.J.; Wright, S.J.; Parikh, M.; Bi, L.; Lee, H.; Kim, T.J.; Lee, S.Y.; et al. Early immune remodeling steers clinical response to frontline chemoimmunotherapy in advanced gastric cancer. Cancer Discov. 2024, 14, 766–785. [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]
- Dai, S.; Zeng, H.; Liu, Z.; Jin, K.; Jiang, W.; Wang, Z.; Lin, Z.; Xiong, Y.; Wang, J.; Chang, Y.; et al. Intratumoral CXCL13(+)CD8(+)T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J. Immunother. Cancer 2021, 9, e001823. [Google Scholar] [CrossRef] [PubMed]
- Korman, A.J.; Garrett-Thomson, S.C.; Lonberg, N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat. Rev. Drug Discov. 2022, 21, 509–528. [Google Scholar] [CrossRef] [PubMed]
- Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [PubMed]
- Sailer, C.J.; Hong, Y.; Dahal, A.; Ryan, A.T.; Mir, S.; Gerber, S.A.; Reagan, P.M.; Kim, M. PD-1(Hi) CAR-T cells provide superior protection against solid tumors. Front. Immunol. 2023, 14, 1187850. [Google Scholar] [CrossRef] [PubMed]
- Bakst, R.L.; Xiong, H.; Chen, C.H.; Deborde, S.; Lyubchik, A.; Zhou, Y.; He, S.; McNamara, W.; Lee, S.Y.; Olson, O.C.; et al. Inflammatory Monocytes Promote Perineural Invasion via CCL2-Mediated Recruitment and Cathepsin B Expression. Cancer Res. 2017, 77, 6400–6414. [Google Scholar] [CrossRef] [PubMed]
- Fan, Q.; Gu, D.; Liu, H.; Yang, L.; Zhang, X.; Yoder, M.C.; Kaplan, M.H.; Xie, J. Defective TGF-beta signaling in bone marrow-derived cells prevents hedgehog-induced skin tumors. Cancer Res. 2014, 74, 471–483. [Google Scholar] [CrossRef] [PubMed]
- Popivanova, B.K.; Kostadinova, F.I.; Furuichi, K.; Shamekh, M.M.; Kondo, T.; Wada, T.; Egashira, K.; Mukaida, N. Blockade of a chemokine, CCL2, reduces chronic colitis-associated carcinogenesis in mice. Cancer Res. 2009, 69, 7884–7892. [Google Scholar] [CrossRef] [PubMed]
- Flores-Toro, J.A.; Luo, D.; Gopinath, A.; Sarkisian, M.R.; Campbell, J.J.; Charo, I.F.; Singh, R.; Schall, T.J.; Datta, M.; Jain, R.K.; et al. CCR2 inhibition reduces tumor myeloid cells and unmasks a checkpoint inhibitor effect to slow progression of resistant murine gliomas. Proc. Natl. Acad. Sci. USA 2020, 117, 1129–1138. [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]
- Shi, L.; Wang, J.; Ding, N.; Zhang, Y.; Zhu, Y.; Dong, S.; Wang, X.; Peng, C.; Zhou, C.; Zhou, L.; et al. Inflammation induced by incomplete radiofrequency ablation accelerates tumor progression and hinders PD-1 immunotherapy. Nat. Commun. 2019, 10, 5421. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Singh, R.; Hsu, D.K.; Zhou, Y.; Yu, S.; Han, D.; Shi, Z.; Huynh, M.; Campbell, J.J.; Hwang, S.T. A Small Molecule CCR2 Antagonist Depletes Tumor Macrophages and Synergizes with Anti-PD-1 in a Murine Model of Cutaneous T-Cell Lymphoma (CTCL). J. Investig. Dermatol. 2020, 140, 1390–1400. [Google Scholar] [CrossRef] [PubMed]
- Fan, F.; Gao, J.; Zhao, Y.; Wang, J.; Meng, L.; Ma, J.; Li, T.; Han, H.; Lai, J.; Gao, Z.; et al. Elevated Mast Cell Abundance Is Associated with Enrichment of CCR2+ Cytotoxic T Cells and Favorable Prognosis in Lung Adenocarcinoma. Cancer Res. 2023, 83, 2690–2703. [Google Scholar] [CrossRef] [PubMed]
- Ma, Y.; Mattarollo, S.R.; Adjemian, S.; Yang, H.; Aymeric, L.; Hannani, D.; Portela Catani, J.P.; Duret, H.; Teng, M.W.; Kepp, O.; et al. CCL2/CCR2-dependent recruitment of functional antigen-presenting cells into tumors upon chemotherapy. Cancer Res. 2014, 74, 436–445. [Google Scholar] [CrossRef] [PubMed]
- Weber, K.S.; Nelson, P.J.; Grone, H.J.; Weber, C. Expression of CCR2 by endothelial cells: Implications for MCP-1 mediated wound injury repair and In vivo inflammatory activation of endothelium. Arterioscler. Thromb. Vasc. Biol. 1999, 19, 2085–2093. [Google Scholar] [CrossRef] [PubMed]
- Nolan, E.; Bridgeman, V.L.; Ombrato, L.; Karoutas, A.; Rabas, N.; Sewnath, C.A.N.; Vasquez, M.; Rodrigues, F.S.; Horswell, S.; Faull, P.; et al. Radiation exposure elicits a neutrophil-driven response in healthy lung tissue that enhances metastatic colonization. Nat. Cancer 2022, 3, 173–187. [Google Scholar] [CrossRef] [PubMed]
- Wisdom, A.J.; Hong, C.S.; Lin, A.J.; Xiang, Y.; Cooper, D.E.; Zhang, J.; Xu, E.S.; Kuo, H.C.; Mowery, Y.M.; Carpenter, D.J.; et al. Neutrophils promote tumor resistance to radiation therapy. Proc. Natl. Acad. Sci. USA 2019, 116, 18584–18589. [Google Scholar] [CrossRef] [PubMed]
- Gong, Z.; Li, Q.; Shi, J.; Li, P.; Hua, L.; Shultz, L.D.; Ren, G. Immunosuppressive reprogramming of neutrophils by lung mesenchymal cells promotes breast cancer metastasis. Sci. Immunol. 2023, 8, eadd5204. [Google Scholar] [CrossRef] [PubMed]
- Liao, W.; Overman, M.J.; Boutin, A.T.; Shang, X.; Zhao, D.; Dey, P.; Li, J.; Wang, G.; Lan, Z.; Li, J.; et al. KRAS-IRF2 Axis Drives Immune Suppression and Immune Therapy Resistance in Colorectal Cancer. Cancer Cell 2019, 35, 559–572. [Google Scholar] [CrossRef] [PubMed]
- Montemagno, C.; Jacquel, A.; Pandiani, C.; Rastoin, O.; Dawaliby, R.; Schmitt, T.; Bourgoin, M.; Palenzuela, H.; Rossi, A.L.; Ambrosetti, D.; et al. Unveiling CXCR2 as a promising therapeutic target in renal cell carcinoma: Exploring the immunotherapeutic paradigm shift through its inhibition by RCT001. J. Exp. Clin. Cancer Res. 2024, 43, 86. [Google Scholar] [CrossRef] [PubMed]
- Greene, S.; Robbins, Y.; Mydlarz, W.K.; Huynh, A.P.; Schmitt, N.C.; Friedman, J.; Horn, L.A.; Palena, C.; Schlom, J.; Maeda, D.Y.; et al. Inhibition of MDSC Trafficking with SX-682, a CXCR1/2 Inhibitor, Enhances NK-Cell Immunotherapy in Head and Neck Cancer Models. Clin. Cancer Res. 2020, 26, 1420–1431. [Google Scholar] [CrossRef] [PubMed]
- Horn, L.A.; Riskin, J.; Hempel, H.A.; Fousek, K.; Lind, H.; Hamilton, D.H.; McCampbell, K.K.; Maeda, D.Y.; Zebala, J.A.; Su, Z.; et al. Simultaneous inhibition of CXCR1/2, TGF-beta, and PD-L1 remodels the tumor and its microenvironment to drive antitumor immunity. J. Immunother. Cancer 2020, 8, e000326. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.; Sadanandam, A.; Nannuru, K.C.; Varney, M.L.; Mayer-Ezell, R.; Bond, R.; Singh, R.K. Small-molecule antagonists for CXCR2 and CXCR1 inhibit human melanoma growth by decreasing tumor cell proliferation, survival, and angiogenesis. Clin. Cancer Res. 2009, 15, 2380–2386. [Google Scholar] [CrossRef] [PubMed]
- Robert, C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat. Commun. 2020, 11, 3801. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Jin, J.; Guo, D.; Tao, Z.; Hu, X. Immune Checkpoint Inhibitors Combined with Targeted Therapy: The Recent Advances and Future Potentials. Cancers 2023, 15, 2858. [Google Scholar] [CrossRef] [PubMed]
- Han, D.; Xu, Z.; Zhuang, Y.; Ye, Z.; Qian, Q. Current Progress in CAR-T Cell Therapy for Hematological Malignancies. J. Cancer 2021, 12, 326–334. [Google Scholar] [CrossRef] [PubMed]
- Hong, Y.; Walling, B.L.; Kim, H.R.; Serratelli, W.S.; Lozada, J.R.; Sailer, C.J.; Amitrano, A.M.; Lim, K.; Mongre, R.K.; Kim, K.D.; et al. ST3GAL1 and betaII-spectrin pathways control CAR T cell migration to target tumors. Nat. Immunol. 2023, 24, 1007–1019. [Google Scholar] [CrossRef] [PubMed]
- Schmidts, A.; Maus, M.V. Making CAR T Cells a Solid Option for Solid Tumors. Front. Immunol. 2018, 9, 2593. [Google Scholar] [CrossRef] [PubMed]
- Xie, N.; Shen, G.; Gao, W.; Huang, Z.; Huang, C.; Fu, L. Neoantigens: Promising targets for cancer therapy. Signal Transduct Target Ther. 2023, 8, 9. [Google Scholar] [CrossRef] [PubMed]
- Orr, B.; Mahdi, H.; Fang, Y.; Strange, M.; Uygun, I.; Rana, M.; Zhang, L.; Suarez Mora, A.; Pusateri, A.; Elishaev, E.; et al. Phase I Trial Combining Chemokine-Targeting with Loco-Regional Chemoimmunotherapy for Recurrent, Platinum-Sensitive Ovarian Cancer Shows Induction of CXCR3 Ligands and Markers of Type 1 Immunity. Clin. Cancer Res. 2022, 28, 2038–2049. [Google Scholar] [CrossRef] [PubMed]
- Guo, C.; Sharp, A.; Crespo, M.; Figuueiredo, I.; Jain, S.; Cogl, U.; Rekowski, J.; Rouhifard, M.; Gallagher, L.; Yuan, W.; et al. Targeting myeloid cell chemotaxis to reverse prostate cancer therapy resistance. Nature 2023, 623, 1053–1601. [Google Scholar] [CrossRef] [PubMed]
- Pernas, S.; Martin, M.; Kaufman, P.A.; Gil-Martin, M.; Gomez Pardo, P.; Lopez-Tarruella, S.; Manso, L.; Ciruelos, E.; Perez-Fidalgo, J.A.; Hernando, C.; et al. Balixafortide plus eribulin in HER2-negative metastatic breast cancer: A phase 1, single-arm, dose-escalation trial. Lancet Oncol. 2018, 19, 812–824. [Google Scholar] [CrossRef] [PubMed]
- Martin, M.; Mayer, I.A.; Walenkamp, A.M.E.; Lapa, C.; Andreeff, M.; Bobirca, A. At the Bedside: Profiling and treating patients with CXCR4-expressing cancers. J. Leukoc. Biol. 2021, 109, 953–967. [Google Scholar] [CrossRef] [PubMed]
- Leslie, J.; Mackey, J.B.G.; Jamieson, T.; Ramon-Gil, E.; Drake, T.M.; Fercoq, F.; Clark, W.; Gilroy, K.; Hedley, A.; Nixon, C.; et al. CXCR2 inhibition enables NASH-HCC immunotherapy. Gut 2022, 71, 2093–2106. [Google Scholar] [CrossRef] [PubMed]
- Goldstein, L.J.; Mansuttis, M.; Levy, C.; Chang, J.C.; Henry, S.; Fernandez-perez, I.; Prausova, J.; Staroslawska, E.; Viale, G.; Butler, B.; et al. A randomized placebo-controlled phase 2 study of paclitaxel in combination with reparixin compared to paclitaxel alone as front-line therapy for metastatic triple-negative breast cancer (fRida). Breat Cancer Res. Treat. 2021, 190, 265–275. [Google Scholar] [CrossRef] [PubMed]
- Walle, T.; Kraske, J.A.; Liao, B.; Lenoir, B.; Timke, C.; von Bohlen Und Halbach, E.; Tran, F.; Griebel, P.; Albrecht, D.; Suarez-Carmona, N.; et al. Radiotherapy orchestrates natural killer cell dependent antitumor immune responses through CXCL8. Sci. Adv. 2022, 8, eabh4050. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, A.L.; Geva, R.; Chung, H.C.; Lemech, C.; Miller, W.H.; Hansen, A.R.; Lee, J.S.; Tsai, F.; Solomon, B.J.; Kim, T.M.; et al. CXCR2 antagoinst navarixin in combination with pembrolizumab in select advanced solid tumors: A phase 2 randomized trial. Investig. New Drugs 2024, 42, 145–159. [Google Scholar] [CrossRef] [PubMed]
- Noel, M.; O’Reilly, E.M.; Wolpin, B.M.; Ryan, D.P.; Bullock, A.J.; Britten, C.D.; Linehan, D.C.; Belt, B.A.; Gamelin, E.C.; Ganguly, B.; et al. Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nabpacilitaxel/gemcitabline in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Investig. New Drugs 2020, 38, 800–811. [Google Scholar] [CrossRef] [PubMed]
- Nywening, T.M.; Belt, B.A.; Cullinan, D.R.; Panni, R.Z.; Han, B.J.; Sanford, D.E.; Jacobs, R.C.; Ye, J.; Paten, A.A.; Gillanders, W.E.; et al. Targeting both tumor-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] [PubMed]
- Fujikawa, K.; Saito, T.; Kurose, K.; Kojima, T.; Funakoshi, T.; Sata, E.; Kakimi, K.; Lida, S.; Doki, Y.; Oka, M.; et al. Integrated analysis of phase 1a and 1b randomized controlled trials; treg-targeted cancer immunotherapy with the humanized anti-CCR4 antibody, KW-0761, for advanced solid tumors. PLoS ONE 2023, 18, e0291772. [Google Scholar] [CrossRef] [PubMed]
- Haag, G.M.; Spring, C.; Grun, B.; Apostolidis, L.; Zschabitz, S.; Dietrich, M.; Berger, A.K.; Weber, T.F.; Zoernig, I.; Schaaf, M.; et al. Pembrolizumab and maraviroc in refractory mismatch repar proficient/microsatellite-stable metastatic colorectal cancer—The PICCASSO phase I trial. Eur. J. Cancer 2022, 167, 112–122. [Google Scholar] [CrossRef] [PubMed]
- Ajona, D.; Ortiz-Espinosa, S.; Moreno, H.; Lozano, T.; Parares, M.J.; Agorreta, J.; Bertolo, C.; Lasarte, J.J.; Vicent, S.; Hoehlig, K.; et al. A combined PD-1/C5a blockade synergistically protects against lung cancer growth and metastasis. Cancer Discov. 2017, 7, 694–703. [Google Scholar] [CrossRef] [PubMed]
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
Ryan, A.T.; Kim, M.; Lim, K. Immune Cell Migration to Cancer. Cells 2024, 13, 844. https://doi.org/10.3390/cells13100844
Ryan AT, Kim M, Lim K. Immune Cell Migration to Cancer. Cells. 2024; 13(10):844. https://doi.org/10.3390/cells13100844
Chicago/Turabian StyleRyan, Allison T., Minsoo Kim, and Kihong Lim. 2024. "Immune Cell Migration to Cancer" Cells 13, no. 10: 844. https://doi.org/10.3390/cells13100844
APA StyleRyan, A. T., Kim, M., & Lim, K. (2024). Immune Cell Migration to Cancer. Cells, 13(10), 844. https://doi.org/10.3390/cells13100844