Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression

Christopher Groth, Xiaoying Hu, Rebekka Weber, Viktor Fleming, Peter Altevogt, Jochen Utikal, Viktor Umansky, Christopher Groth, Xiaoying Hu, Rebekka Weber, Viktor Fleming, Peter Altevogt, Jochen Utikal, Viktor Umansky

Abstract

Under steady-state conditions, bone marrow-derived immature myeloid cells (IMC) differentiate into granulocytes, macrophages and dendritic cells (DCs). This differentiation is impaired under chronic inflammatory conditions, which are typical for tumour progression, leading to the accumulation of IMCs. These cells are capable of inducing strong immunosuppressive effects through the expression of various cytokines and immune regulatory molecules, inhibition of lymphocyte homing, stimulation of other immunosuppressive cells, depletion of metabolites critical for T cell functions, expression of ectoenzymes regulating adenosine metabolism, and the production of reactive species. IMCs are therefore designated as myeloid-derived suppressor cells (MDSCs), and have been shown to accumulate in tumour-bearing mice and cancer patients. MDSCs are considered to be a strong contributor to the immunosuppressive tumour microenvironment and thus an obstacle for many cancer immunotherapies. Consequently, numerous studies are focused on the characterisation of MDSC origin and their relationship to other myeloid cell populations, their immunosuppressive capacity, and possible ways to inhibit MDSC function with different approaches being evaluated in clinical trials. This review analyses the current state of knowledge on the origin and function of MDSCs in cancer, with a special emphasis on the immunosuppressive pathways pursued by MDSCs to inhibit T cell functions, resulting in tumour progression. In addition, we describe therapeutic strategies and clinical benefits of MDSC targeting in cancer.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Myelopoiesis is altered under chronic inflammation. Under physiological conditions, hematopoietic progenitor cells (HPC) differentiate via common myeloid progenitor cells (CMP) into granulocyte/macrophage progenitor cells (GMP). These immature myeloid cells (IMC) further differentiate into monocytic/dendritic progenitor cells (MDP) or myeloblasts (MB) from which these cells further develop into dendritic cells (DCs)/macrophages or neutrophils, respectively. Under cancerous conditions, the tumour alters myelopoiesis in general and impairs further differentiation of progenitor cells, leading to the accumulation of monocytic myeloid-derived suppressor cells (M-MDSCs) and polymorphonuclear MDSCs (PMN-MDSCs)
Fig. 2
Fig. 2
Myeloid-derived suppressor cells (MDSCs) are generated under chronic inflammatory conditions typical for cancer. Inflammatory factors that induce MDSC recruitment and expansion in the tumour microenvironment include interleukin (IL)-6, IL-10, IL-1β, granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (M-CSF), chemokine (C-C motif) ligand 2 (CCL)2, CCL5, CCL26, chemokine (C-X-C motif) ligand 8 (CXCL)8, CXL12, and prostaglandin E2 (PGE2), released as soluble mediators or via extracellular vesicles (EVs). Hypoxia in the tumour microenvironment facilitates the expression of hypoxia-inducible factors digoxin and Hypoxia-inducible factor 1-alpha (HIF-1α) that induce the expression of the chemokine CCL26 and adenosine-producing ectoenzymes by tumour cells, leading to MDSC recruitment and accumulation
Fig. 3
Fig. 3
Main mechanisms of immunosuppression mediated by myeloid-derived suppressor cells (MDSCs). Mechanisms include the generation of immunosuppressive M2 macrophages and regulatory T cells via interleukin (IL)-10 and interferon (IFN)-γ secretion (a); impairment of lymphocyte adhesion to endothelial cells (ECs) and extravasation through nitric oxide (NO)-mediated downregulation of adhesion molecules CD162 and CD44, and tumor necrosis factor-alpha-converting enzyme (TACE)-mediated cleavage of CD62L (L-Selectin) on T cells (b); the production of reactive oxygen (ROS) and nitrogen species (RNS) through NADPH oxidase 2 (NOX-2) and nitric oxide synthase 2 (NOS2), leading to increased cyclooxygenase 2 (Cox-2), Hypoxia-inducible factor 1-alpha (HIF-1α) and arginase 1 (ARG1) expression and reduced T cell receptor (TCR) expression (c); the depletion and intracellular degradation of the amino acids L-arginine and cystine through increased uptake via the CAT2B and SLC7A11 transporters, respectively (d); induction of the ectoenzymes CD39 and CD73 via HIF-1 through transforming growth factor beta (TGF-β and hypoxic conditions, leading to adenosine production and reduced phosphorylation of extracellular signal–regulated kinase (ERK), protein kinase B (Akt) and Zap70, and reduced expression of CD95L, perforin, IFN-γ and tumour necrosis factor alpha TNF-α in T cells (e); and the expression of immune regulatory molecules B7, programmed death-ligand 1 (PD-L1) and FasL, causing T cell anergy and apoptosis via binding to their respective receptors (f)

References

    1. Weiskopf Kipp, et al. Myeloid cell origins, differentiation, and clinical implications. Microbiol. Spectr. 2016;42:407–420.
    1. Velten L, et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 2017;19:271–281. doi: 10.1038/ncb3493.
    1. Ioannou M, et al. Crucial role of granulocytic myeloid-derived suppressor cells in the regulation of central nervous system autoimmune disease. J. Immunol. 2012;188:1136–1146. doi: 10.4049/jimmunol.1101816.
    1. Dorhoi, A. & Plessis, N. Du. Monocytic myeloid-derived suppressor cells in chronic infections. Front. Immunol. 8. 10.3389/fimmu.2017.01895 (2018).
    1. Meyer C, et al. Chronic inflammation promotes myeloid-derived suppressor cell activation blocking antitumor immunity in transgenic mouse melanoma model. Proc. . Natl Acad. Sci. USA. 2011;108:17111–17116. doi: 10.1073/pnas.1108121108.
    1. Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32:19–25. doi: 10.1016/j.it.2010.10.002.
    1. Ueda Y, Cain DW, Kuraoka M, Kondo M, Kelsoe G. IL-1R type I-dependent hemopoietic stem cell proliferation is necessary for inflammatory granulopoiesis and reactive neutrophilia. J. Immunol. 2009;182:6477–6484. doi: 10.4049/jimmunol.0803961.
    1. Ueda Y, Kondo M, Kelsoe G. Inflammation and the reciprocal production of granulocytes and lymphocytes in bone marrow. J. Exp. Med. 2005;201:1771–1780. doi: 10.1084/jem.20041419.
    1. Jordan KR, et al. Immunosuppressive myeloid-derived suppressor cells are increased in splenocytes from cancer patients. Cancer Immunol. Immunother. 2017;66:503–513. doi: 10.1007/s00262-016-1953-z.
    1. Gabrilovich DI, et al. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007;67:3986. doi: 10.1158/0008-5472.CAN-06-3037.
    1. Shi H, et al. Recruited monocytic myeloid-derived suppressor cells promote the arrest of tumor cells in the premetastatic niche through an IL-1β-mediated increase in E-selectin expression. Int. J. Cancer. 2017;140:1370–1383. doi: 10.1002/ijc.30538.
    1. Binsfeld M, et al. Granulocytic myeloid-derived suppressor cells promote angiogenesis in the context of multiple myeloma. Oncotarget. 2016;7:37931–37943. doi: 10.18632/oncotarget.9270.
    1. Kamran N, et al. Melanoma induced immunosuppression is mediated by hematopoietic dysregulation. Oncoimmunology. 2018;7:e1408750. doi: 10.1080/2162402X.2017.1408750.
    1. Sarvaria A, Madrigal JA, Saudemont A. B cell regulation in cancer and anti-tumor immunity. Cell Mol. Immunol. 2017;14:662–674. doi: 10.1038/cmi.2017.35.
    1. Barreda DR, Hanington PC, Belosevic M. Regulation of myeloid development and function by colony stimulating factors. Dev. Comp. Immunol. 2004;28:509–554. doi: 10.1016/j.dci.2003.09.010.
    1. Dolcetti L, et al. Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF. Eur. J. Immunol. 2010;40:22–35. doi: 10.1002/eji.200939903.
    1. Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells. J. Immunol. 2010;185:2273–2284. doi: 10.4049/jimmunol.1000901.
    1. Jiang M, et al. Interleukin-6 trans-signaling pathway promotes immunosuppressive myeloid-derived suppressor cells via suppression of suppressor of cytokine signaling 3 in breast cancer. Front. Immunol. 2017;8:1840. doi: 10.3389/fimmu.2017.01840.
    1. Bronte V, et al. Unopposed production of granulocyte-macrophage colony-stimulating factor by tumors inhibits CD8+T cell responses by dysregulating antigen-presenting cell maturation. J. Immunol. 1999;162:5728–5737.
    1. Morales JK, Kmieciak M, Knutson KL, Bear HD, Manjili MH. GM-CSF is one of the main breast tumor-derived soluble factors involved in the differentiation of CD11b-Gr1- bone marrow progenitor cells into myeloid-derived suppressor cells. Breast Cancer Res. Treat. 2010;123:39–49. doi: 10.1007/s10549-009-0622-8.
    1. Gargett T, et al. GM-CSF signalling blockade and chemotherapeutic agents act in concert to inhibit the function of myeloid-derived suppressor cells in vitro. Clin. Transl. Immunol. 2016;5:e119. doi: 10.1038/cti.2016.80.
    1. Xiang X, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int. J. Cancer. 2009;124:2621–2633. doi: 10.1002/ijc.24249.
    1. Liu Y, et al. Contribution of MyD88 to the tumor exosome-mediated induction of myeloid derived suppressor cells. Am. J. Pathol. 2010;176:2490–2499. doi: 10.2353/ajpath.2010.090777.
    1. Holmgaard RB, et al. Tumor-expressed ido recruits and activates MDSCs in a treg-dependent manner. Cell Rep. 2015;13:412–424. doi: 10.1016/j.celrep.2015.08.077.
    1. Munn DH. Blocking IDO activity to enhance anti-tumor immunity. Front. Biosci. 2012;E4:734–745. doi: 10.2741/e414.
    1. Chiu DKC, et al. Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26. Hepatology. 2016;64:797–813. doi: 10.1002/hep.28655.
    1. Chiu DKC, et al. Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nat. Commun. 2017;8:517. doi: 10.1038/s41467-017-00530-7.
    1. Noman MZ, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014;211:781–790. doi: 10.1084/jem.20131916.
    1. Umansky V, Sevko A. Tumor microenvironment and myeloid-derived suppressor cells. Cancer Microenviron. 2013;6:169–177. doi: 10.1007/s12307-012-0126-7.
    1. Chun E, et al. CCL2 Promotes colorectal carcinogenesis by enhancing polymorphonuclear myeloid-derived suppressor cell population and function. Cell Rep. 2015;12:244–257. doi: 10.1016/j.celrep.2015.06.024.
    1. Chang AL, 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. doi: 10.1158/0008-5472.CAN-16-0144.
    1. Obermajer N, Muthuswamy R, Odunsi K, Edwards RP, Kalinski P. PGE2-induced CXCL12 production and CXCR4 expression controls the accumulation of human mdscs in ovarian cancer environment. Cancer Res. 2011;71:7463–7470. doi: 10.1158/0008-5472.CAN-11-2449.
    1. Alfaro C, et al. Tumor-produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs) Clin. Cancer Res. 2016;22:3924–3936. doi: 10.1158/1078-0432.CCR-15-2463.
    1. Park YJ, et al. Tumor microenvironmental conversion of natural killer cells into myeloid-derived suppressor cells. Cancer Res. 2013;73:5669–5681. doi: 10.1158/0008-5472.CAN-13-0545.
    1. Huang B, et al. 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. doi: 10.1158/0008-5472.CAN-05-1299.
    1. Zhao F, Korangy F, Greten TF. Cellular immune suppressor mechanisms in patients with hepatocellular carcinoma. Dig. Dis. 2012;30:477–482. doi: 10.1159/000341695.
    1. Fujimura T, Ring S, Umansky V, Mahnke K, Enk AH. Regulatory T cells stimulate B7-H1 expression in myeloid-derived suppressor cells in ret melanomas. J. Invest. Dermatol. 2012;132:1239–1246. doi: 10.1038/jid.2011.416.
    1. Schlecker E, et al. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J. Immunol. 2012;189:5602–5611. doi: 10.4049/jimmunol.1201018.
    1. Lee Cho-Rong, Lee Wongeun, Cho Steve, Park Sung-Gyoo. Characterization of Multiple Cytokine Combinations and TGF-β on Differentiation and Functions of Myeloid-Derived Suppressor Cells. International Journal of Molecular Sciences. 2018;19(3):869. doi: 10.3390/ijms19030869.
    1. Beury DW, et al. Cross-talk among myeloid-derived suppressor cells, macrophages, and tumor cells impacts the inflammatory milieu of solid tumors. J. Leukoc. Biol. 2014;96:1109–1118. doi: 10.1189/jlb.3A0414-210R.
    1. Ku AW, et al. Tumor-induced MDSC act via remote control to inhibit L-selectin-dependent adaptive immunity in lymph nodes. eLife. 2016;5:e17375. doi: 10.7554/eLife.17375.
    1. Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J. Immunol. 2009;182:4499–4506. doi: 10.4049/jimmunol.0802740.
    1. Hanson EM, Clements VK, 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. doi: 10.4049/jimmunol.0804253.
    1. Schouppe E, et al. 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. doi: 10.1002/eji.201343349.
    1. Liu Y, Wei J, Guo G, Zhou J. Norepinephrine-induced myeloid-derived suppressor cells block T cell responses via generation of reactive oxygen species. Immunopharmacol. Immunotoxicol. 2015;37:359–365. doi: 10.3109/08923973.2015.1059442.
    1. Corzo CA, et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J. Immunol. 2009;182:5693–5701. doi: 10.4049/jimmunol.0900092.
    1. Kusmartsev S, et al. Oxidative stress regulates expression of VEGFR1 in myeloid cells: link to tumor-induced immune suppression in renal cell carcinoma. J. Immunol. 2008;181:346–353. doi: 10.4049/jimmunol.181.1.346.
    1. Jayaraman P, et al. Tumor-expressed inducible nitric oxide synthase controls induction of functional myeloid-derived suppressor cells through modulation of vascular endothelial growth factor release. J. Immunol. 2012;188:5365–5376. doi: 10.4049/jimmunol.1103553.
    1. Horikawa N, et al. Expression of vascular endothelial growth factor in ovarian cancer inhibits tumor immunity through the accumulation of myeloid-derived suppressor cells. Clin. Cancer Res. 2017;23:587–599. doi: 10.1158/1078-0432.CCR-16-0387.
    1. Wang T, et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat. Med. 2004;10:48–54. doi: 10.1038/nm976.
    1. Zhao D, et al. VEGF drives cancer-initiating stem cells through VEGFR-2/Stat3 signaling to upregulate Myc and Sox2. Oncogene. 2015;34:3107–3119. doi: 10.1038/onc.2014.257.
    1. Bartoli M, et al. VEGF differentially activates STAT3 in microvascular endothelial cells. Faseb. J. 2003;17:1562–1564. doi: 10.1096/fj.02-1084fje.
    1. Niu G, et al. Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene. 2002;21:2000–2008. doi: 10.1038/sj.onc.1205260.
    1. Beury DW, et al. Myeloid-derived suppressor cell survival and function are regulated by the transcription Factor Nrf2. J. Immunol. 2016;196:3470–3478. doi: 10.4049/jimmunol.1501785.
    1. Jian Shiou-Ling, Chen Wei-Wei, Su Yu-Chia, Su Yu-Wen, Chuang Tsung-Hsien, Hsu Shu-Ching, Huang Li-Rung. Glycolysis regulates the expansion of myeloid-derived suppressor cells in tumor-bearing hosts through prevention of ROS-mediated apoptosis. Cell Death & Disease. 2017;8(5):e2779–e2779. doi: 10.1038/cddis.2017.192.
    1. Kusmartsev S, Gabrilovich DI. Inhibition of myeloid cell differentiation in cancer: the role of reactive oxygen species. J. Leukoc. Biol. 2003;74:186–196. doi: 10.1189/jlb.0103010.
    1. Raber PL, et al. Subpopulations of myeloid-derived suppressor cells impair T cell responses through independent nitric oxide-related pathways. Int. J. Cancer. 2014;134:2853–2864. doi: 10.1002/ijc.28622.
    1. Chun KS, et al. Nitric oxide induces expression of cyclooxygenase-2 in mouse skin through activation of NF-kappaB. Carcinogenesis. 2004;25:445–454. doi: 10.1093/carcin/bgh021.
    1. Olson N, van der Vliet A. Interactions between nitric oxide and hypoxia-inducible factor signaling pathways in inflammatory disease. Nitric Oxide. 2012;25:125–137. doi: 10.1016/j.niox.2010.12.010.
    1. Park JY, Pillinger MH, Abramson SB. Prostaglandin E2 synthesis and secretion: The role of PGE2 synthases. Clin. Immunol. 2006;119:229–240. doi: 10.1016/j.clim.2006.01.016.
    1. Obermajer N, Kalinski P. Generation of myeloid-derived suppressor cells using prostaglandin E2. Transplant. Res. 2012;1:15. doi: 10.1186/2047-1440-1-15.
    1. Rodríguez-Ubreva J, et al. Prostaglandin E2 Leads to the Acquisition of DNMT3A-Dependent Tolerogenic Functions in Human Myeloid-Derived Suppressor Cells. Cell Rep. 2017;21:154–167. doi: 10.1016/j.celrep.2017.09.018.
    1. Li YL, Zhao H, Ren XB. Relationship of VEGF/VEGFR with immune and cancer cells: staggering or forward? Cancer Biol. Med. 2016;13:206–214. doi: 10.20892/j.issn.2095-3941.2015.0070.
    1. Nagaraj S, et al. Altered recognition of antigen is a mechanism of CD8+T cell tolerance in cancer. Nat. Med. 2007;13:828–835. doi: 10.1038/nm1609.
    1. Molon B, et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 2011;208:1949–1962. doi: 10.1084/jem.20101956.
    1. Stiff A, et al. Nitric oxide production by myeloid derived suppressor cells plays a role in impairing fc receptor-mediated natural killer cell function. Clin. Cancer Res. 2018;24:1891–1904. doi: 10.1158/1078-0432.CCR-17-0691.
    1. Youn JI, Collazo M, Shalova IN, Biswas SK, Gabrilovich DI. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J. Leukoc. Biol. 2012;91:167–181. doi: 10.1189/jlb.0311177.
    1. Rodriguez PC, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 2009;69:1553–1560. doi: 10.1158/0008-5472.CAN-08-1921.
    1. Rodríguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol. Rev. 2008;222:180–191. doi: 10.1111/j.1600-065X.2008.00608.x.
    1. Rodriguez PC, et al. Arginase I Production in the Tumor Microenvironment by Mature Myeloid Cells Inhibits T-Cell Receptor Expression and Antigen-Specific T-Cell Responses. Cancer Res. 2004;64:5839–5849. doi: 10.1158/0008-5472.CAN-04-0465.
    1. Rodriguez PC, et al. Regulation of T cell receptor CD3zeta chain expression by L-arginine. J. Biol. Chem. 2002;277:21123–21129. doi: 10.1074/jbc.M110675200.
    1. Bian Z, et al. Arginase-1 is neither constitutively expressed in nor required for myeloid-derived suppressor cell (MDSC)-mediated inhibition of T cell proliferation. Eur. J. Immunol. 2018;48:1046–1058. doi: 10.1002/eji.201747355.
    1. Bannai S. Transport of cystine and cysteine in mammalian cells. Biochim. Biophys. Acta. 1984;779:289–306. doi: 10.1016/0304-4157(84)90014-5.
    1. Gmünder H, Eck HP, Dröge W. Low membrane transport activity for cystine in resting and mitogenically stimulated human lymphocyte preparations and human T cell clones. Eur. J. Biochem. 1991;201:113–117. doi: 10.1111/j.1432-1033.1991.tb16263.x.
    1. Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-derived suppressor cells inhibit T cell activation by depleting cystine and cysteine. Cancer Res. 2010;70:68–77. doi: 10.1158/0008-5472.CAN-09-2587.
    1. Ostrand-Rosenberg S. Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity. Cancer Immunol. Immunother. 2010;59:1593–1600. doi: 10.1007/s00262-010-0855-8.
    1. Li J, et al. CD39/CD73 upregulation on myeloid-derived suppressor cells via TGF-β-mTOR-HIF-1 signaling in patients with non-small cell lung cancer. Oncoimmunology. 2017;6:e1320011. doi: 10.1080/2162402X.2017.1320011.
    1. Linnemann Carsten, Schildberg Frank A., Schurich Anna, Diehl Linda, Hegenbarth Silke I., Endl Elmar, Lacher Svenja, Müller Christa E., Frey Jürgen, Simeoni Luca, Schraven Burkhart, Stabenow Dirk, Knolle Percy A. Adenosine regulates CD8 T-cell priming by inhibition of membrane-proximal T-cell receptor signalling. Immunology. 2009;128(1pt2):e728–e737. doi: 10.1111/j.1365-2567.2009.03075.x.
    1. Hoskin DW, Mader JS, Furlong SJ, Conrad DM, Blay J. Inhibition of T cell and natural killer cell function by adenosine and its contribution to immune evasion by tumor cells (review) Int. J. Oncol. 2008;32:527–535.
    1. Zhang Y, et al. Metformin-induced reduction of CD39 and CD73 blocks myeloid-derived suppressor cell activity in patients with ovarian cancer. Cancer Res. 2018;78:1779–1791. doi: 10.1158/0008-5472.CAN-17-2460.
    1. Juneja VR, et al. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 2017;214:895–904. doi: 10.1084/jem.20160801.
    1. Teng MWL, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T cell infiltration and PD-L1. Cancer Res. 2015;75:2139–2145. doi: 10.1158/0008-5472.CAN-15-0255.
    1. Wargo JA, Reddy SM, Reuben A, Sharma P. Monitoring immune responses in the tumor microenvironment. Curr. Opin. Immunol. 2016;41:23–31. doi: 10.1016/j.coi.2016.05.006.
    1. Yamauchi Y, et al. Circulating and tumor myeloid-derived suppressor cells in resectable non-small-cell lung cancer. Am. J. Respir. Crit. Care. Med. 2018 doi: 10.1164/rccm.201708-1707OC.
    1. Lu C, Redd PS, Lee JR, Savage N, Liu K. The expression profiles and regulation of PD-L1 in tumor-induced myeloid-derived suppressor cells. Oncoimmunology. 2016;5:e1247135. doi: 10.1080/2162402X.2016.1247135.
    1. Gebhardt C, et al. Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin. Cancer Res. 2015;21:5453–5459. doi: 10.1158/1078-0432.CCR-15-0676.
    1. Sade-Feldman M, et al. Clinical significance of circulating CD33+CD11b+HLA-DR- myeloid cells in patients with stage iv melanoma treated with ipilimumab. Clin. Cancer Res. 2016;22:5661–5672. doi: 10.1158/1078-0432.CCR-15-3104.
    1. Iwata T, et al. PD-L1+MDSCs are increased in HCC patients and induced by soluble factor in the tumor microenvironment. Sci. Rep. 2016;6:39296. doi: 10.1038/srep39296.
    1. Villarino AV, Kanno Y, O’Shea JJ. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat. Immunol. 2017;18:374–384. doi: 10.1038/ni.3691.
    1. Pinton L, et al. Activated T cells sustain myeloid-derived suppressor cell-mediated immune suppression. Oncotarget. 2016;7:1168–1184.
    1. Trikha P, Carson WE., III Signaling pathways involved in MDSC regulation. Biochim. Biophys. Acta. 2014;1846:55–65.
    1. Cheng P, et al. Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein. J. Exp. Med. 2008;205:2235–2249. doi: 10.1084/jem.20080132.
    1. Sinha P, et al. Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells. J. Immunol. 2008;181:4666–4675. doi: 10.4049/jimmunol.181.7.4666.
    1. Lim JW, Kim H, Kim KH. Nuclear factor-kappaB regulates cyclooxygenase-2 expression and cell proliferation in human gastric cancer cells. Lab. Invest. 2001;81:349–360. doi: 10.1038/labinvest.3780243.
    1. Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007;67:4507–4513. doi: 10.1158/0008-5472.CAN-06-4174.
    1. Medina-Echeverz J, et al. IFN-γ regulates survival and function of tumor-induced CD11b + Gr-1high myeloid derived suppressor cells by modulating the anti-apoptotic molecule Bcl2a1. Eur. J. Immunol. 2014;44:2457–2467. doi: 10.1002/eji.201444497.
    1. Gabrilovich, D. I. & Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9,162–174 (2009).
    1. Liang W, Ferrara N. The Complex role of neutrophils in tumor angiogenesis and metastasis. Cancer Immunol. Res. 2016;4:83–91. doi: 10.1158/2326-6066.CIR-15-0313.
    1. Elliott LA, Doherty GA, Sheahan K, Ryan EJ. Human tumor-infiltrating myeloid cells: phenotypic and functional diversity. Front. Immunol. 2017;8:86. doi: 10.3389/fimmu.2017.00086.
    1. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J. Immunol. 2008;181:5791–5802. doi: 10.4049/jimmunol.181.8.5791.
    1. Zhou J, Nefedova Y, Lei A, Gabrilovich D. Neutrophils and PMN-MDSC: their biological role and interaction with stromal cells. Semin. Immunol. 2018;35:19–28. doi: 10.1016/j.smim.2017.12.004.
    1. Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat. Rev. Cancer. 2016;16:431–446. doi: 10.1038/nrc.2016.52.
    1. Fridlender ZG, et al. Transcriptomic analysis comparing tumor-associated neutrophils with granulocytic myeloid-derived suppressor cells and normal neutrophils. PLoS ONE. 2012;7:e31524. doi: 10.1371/journal.pone.0031524.
    1. Condamine T, et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci. Immunol. 2016;1:8943–8943. doi: 10.1126/sciimmunol.aaf8943.
    1. Sagiv JY, et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 2015;10:562–573. doi: 10.1016/j.celrep.2014.12.039.
    1. Fleming V, et al. Targeting myeloid-derived suppressor cells to bypass tumor-induced immunosuppression. Front. Immunol. 2018;9:398. doi: 10.3389/fimmu.2018.00398.
    1. Eriksson E., Wenthe J., Irenaeus S., Loskog A. & Ullenhag G. Gemcitabine reduces MDSCs, tregs and TGFβ-1 while restoring the teff/treg ratio in patients with pancreatic cancer. J. Transl. Med. 14. 10.1186/s12967-016-1037-z (2016).
    1. Weiss JM, et al. Regulatory T cells and myeloid-derived suppressor cells in the tumor microenvironment undergo fas-dependent cell death during IL-2/ CD40 therapy. J. Immunol. 2014;192:5821–5829. doi: 10.4049/jimmunol.1400404.
    1. Dang Y, et al. TLR8 ligation induces apoptosis of monocytic myeloid-derived suppressor cells. J. Leukoc. Biol. 2018;103:157–164. doi: 10.1002/JLB.5AB0217-070R.
    1. Kaneda MM, et al. PI3Kγ 3 is a molecular switch that controls immune suppression. Nature. 2016;539:437–442. doi: 10.1038/nature19834.
    1. Ali K, et al. Inactivation of PI(3)K p110δ breaks regulatory T cell-mediated immune tolerance to cancer. Nature. 2014;510:407–411. doi: 10.1038/nature13444.
    1. Davis RJ, et al. Anti-PD-L1 efficacy can be enhanced by inhibition of myeloid-derived suppressor cells with a selective inhibitor of PI3Kδ/γ. Cancer Res. 2017;77:2607–2619. doi: 10.1158/0008-5472.CAN-16-2534.
    1. Kwon ED, et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014;15:700–712. doi: 10.1016/S1470-2045(14)70189-5.
    1. Lu X, et al. Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature. 2017;543:728–732. doi: 10.1038/nature21676.
    1. Bill MA, et al. The small molecule curcumin analog FLLL32 induces apoptosis in melanoma cells via STAT3 inhibition and retains the cellular response to cytokines with anti-tumor activity. Mol. Cancer. 2010;9:165. doi: 10.1186/1476-4598-9-165.
    1. Ratner M. Setback for JAK2 inhibitors. Nat. Biotechnol. 2014;32:119. doi: 10.1038/nbt0214-119a.
    1. Melani C, Sangaletti S, Barazzetta FM, Werb Z, Colombo MP. Amino-biphosphonate-mediated MMP-9 inhibition breaks the tumor-bone marrow axis responsible for myeloid-derived suppressor cell expansion and macrophage infiltration in tumor stroma. Cancer Res. 2007;67:11438–11446. doi: 10.1158/0008-5472.CAN-07-1882.
    1. Overall C, Kleifeld O. Tumour microenvironment - opinion: validating matrix metalloproteinases as drug targets and anti-targets for cancer therapy. Nat. Rev. Cancer. 2006;6:227–239. doi: 10.1038/nrc1821.
    1. Santini D, et al. Pamidronate induces modifications of circulating angiogenetic factors in cancer patients. Clin. Cancer Res. 2002;8:1080–1084.
    1. Santini D, et al. Zoledronic acid induces significant and long-lasting modifications of circulating angiogenic factors in cancer patients. Clin. Cancer Res. 2003;9:2893–2897.
    1. Ferretti G, et al. Zoledronic-acid-induced circulating level modifications of angiogenic factors, metalloproteinases and proinflammatory cytokines in metastatic breast cancer patients. Oncology. 2005;69:35–43. doi: 10.1159/000087286.
    1. Nefedova Y, et al. Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells. Cancer Res. 2007;67:11021–11028. doi: 10.1158/0008-5472.CAN-07-2593.
    1. Iclozan C, Antonia S, Chiappori A, Chen DT, Gabrilovich D. Therapeutic regulation of myeloid-derived suppressor cells and immune response to cancer vaccine in patients with extensive stage small cell lung cancer. Cancer Immunol. Immunother. 2013;62:909–918. doi: 10.1007/s00262-013-1396-8.
    1. Movahedi K, et al. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood. 2008;111:4233–4244. doi: 10.1182/blood-2007-07-099226.
    1. Bronte V, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016;7:12150. doi: 10.1038/ncomms12150.

Source: PubMed

3
Sottoscrivi