Targeting Myeloid-Derived Suppressor Cells to Bypass Tumor-Induced Immunosuppression

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

Abstract

The immune system has many sophisticated mechanisms to balance an extensive immune response. Distinct immunosuppressive cells could protect from excessive tissue damage and autoimmune disorders. Tumor cells take an advantage of those immunosuppressive mechanisms and establish a strongly immunosuppressive tumor microenvironment (TME), which inhibits antitumor immune responses, supporting the disease progression. Myeloid-derived suppressor cells (MDSC) play a crucial role in this immunosuppressive TME. Those cells represent a heterogeneous population of immature myeloid cells with a strong immunosuppressive potential. They inhibit an antitumor reactivity of T cells and NK cells. Furthermore, they promote angiogenesis, establish pre-metastatic niches, and recruit other immunosuppressive cells such as regulatory T cells. Accumulating evidences demonstrated that the enrichment and activation of MDSC correlated with tumor progression, recurrence, and negative clinical outcome. In the last few years, various preclinical studies and clinical trials targeting MDSC showed promising results. In this review, we discuss different therapeutic approaches on MDSC targeting to overcome immunosuppressive TME and enhance the efficiency of current tumor immunotherapies.

Keywords: cancer immunotherapy; immunosuppression; myeloid-derived suppressor cells; therapeutic targeting; tumor microenvironment.

Figures

Figure 1
Figure 1
Myeloid-derived suppressor cells (MDSC) recruitment and activation during tumor progression. Tumor and immune cells constantly release inflammatory mediators, leading to the dysregulation of normal myelopoiesis and to the conversion of immature myeloid cells (IMC) into MDSC in the bone marrow. The latter cells expand and migrate to the tumor site through the interaction between CCR and respective chemokines (CCL). In the tumor microenvironment, MDSC are activated and strongly inhibit an antitumor reactivity of T cells via various mechanisms.
Figure 2
Figure 2
Strategies for myeloid-derived suppressor cells (MDSC) targeting. The MDSC modulation could be achieved by the inhibition of their immunosuppressive activity (blue box), by the blockade of MDSC recruitment to the tumor site (green box), and by the regulation of myelopoiesis and/or depletion of MDSC (red box). Examples for each therapeutic approach are shown.

References

    1. Ostrand-Rosenberg S. Immune surveillance: a balance between protumor and antitumor immunity. Curr Opin Genet Dev (2008) 18:11–8.10.1016/j.gde.2007.12.007
    1. Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL. 2011: the immune hallmarks of cancer. Cancer Immunol Immunother (2011) 60:319–26.10.1007/s00262-010-0968-0
    1. Umansky V, Sevko A. Tumor microenvironment and myeloid-derived suppressor cells. Cancer Microenviron (2013) 6:169–77.10.1007/s12307-012-0126-7
    1. Hansen GL, Gaudernack G, Brunsvig PF, Cvancarova M, Kyte JA. Immunological factors influencing clinical outcome in lung cancer patients after telomerase peptide vaccination. Cancer Immunol Immunother (2015) 64:1609–21.10.1007/s00262-015-1766-5
    1. Limagne E, Euvrard R, Thibaudin M, Rébé C, Derangere V, Chevriaux A, et al. Accumulation of MDSC and Th17 cells in patients with metastatic colorectal cancer predict the efficacy of a FOLFOX-bevacizumab drug treatment regimen. Cancer Res (2016) 76:5241–52.10.1158/0008-5472.CAN-15-3164
    1. Gabrilovich D, Nagaraj S. Myeloid-derived-suppressor cells as regulators of the immune system. Nat Rev Immunol (2009) 9:162–74.10.1038/nri2506
    1. Gabrilovich D. Myeloid-derived suppressor cells. Cancer Immunol Res (2017) 5:3–8.10.1158/2326-6066
    1. Parker KH, Beury DW, Ostrand-Rosenberg S. Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv Cancer Res (2015) 128:95–139.10.1016/bs.acr.2015.04.002
    1. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol (2016) 37:208–20.10.1016/j.it.2016.01.004
    1. Meirow Y, Kanterman J, Baniyash M. Paving the road to tumor development and spreading: myeloid-derived suppressor cells are ruling the fate. Front Immunol (2015) 6:523.10.3389/fimmu.2015.00523
    1. Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J Immunol (2009) 182:4499–506.10.4049/jimmunol.0802740
    1. Ray A, Chakraborty K, Ray P. Immunosuppressive MDSCs induced by TLR signaling during infection and role in resolution of inflammation. Front Cell Infect Microbiol (2013) 3:52.10.3389/fcimb.2013.00052
    1. Loftus TJ, Mohr AM, Moldawer LL. Dysregulated myelopoiesis and hematopoietic function following acute physiologic insult. Curr Opin Hematol (2018) 25(1):37–43.10.1097/MOH.0000000000000395
    1. Ueha S, Shand FH, Matsushima K. Myeloid cell population dynamics in healthy and tumor-bearing mice. Int Immunopharmacol (2011) 11:783–8.10.1016/j.intimp.2011.03.003
    1. Yin B, Ma G, Yen CY, Zhou Z, Wang GX, Divino CM, et al. Myeloid-derived suppressor cells prevent type 1 diabetes in murine models. J Immunol (2010) 185:5828–34.10.4049/jimmunol.0903636
    1. Gabrilovich D, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol (2012) 12:253–68.10.1038/nri3175
    1. Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Mandruzzato S, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun (2016) 7:12150.10.1038/ncomms12150
    1. Solito S, Marigo I, Pinton L, Damuzzo V, Mandruzzato S, Bronte V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann N Y Acad Sci (2014) 1319:47–65.10.1111/nyas.12469
    1. Condamine T, Dominguez GA, Youn JI, Kossenkov AV, Mony S, Alicea-Torres K, et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol (2016) 1(2):aaf8943.10.1126/sciimmunol.aaf8943
    1. Umansky V, Blattner C, Gebhardt C, Utikal J. The role of myeloid-derived suppressor cells (MDSC) in cancer progression. Vaccines (Basel) (2016) 4:36.10.3390/vaccines4040036
    1. Wesolowski R, Markowitz J, Carson WE. Myeloid derived suppressor cells – a new therapeutic target in the treatment of cancer. J Immunother Cancer (2013) 1:10.10.1186/2051-1426-1-10
    1. Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest (2015) 125:3356–64.10.1172/JCI80005
    1. Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest (2015) 125:3365–76.10.1172/JCI80006
    1. Isola AL, Chen S. Exosomes: the messengers of health and disease. Curr Neuropharmacol (2017) 15:157–65.10.2174/1570159X14666160825160421
    1. Ruivo CF, Adem B, Silva M, Melo SA. The biology of cancer exosomes: insights and new perspectives. Cancer Res (2017) 77(23):6480–8.10.1158/0008-5472.CAN-17-0994
    1. Ridder K, Sevko A, Heide J, Rupp AK, Macas J, Starmann J, et al. Extracellular vesicle-mediated transfer of functional RNA in the tumor microenvironment. Oncoimmunology (2015) 4:e1008371.10.1080/2162402X.2015.1008371
    1. Filipazzi P, Bürdek M, Villa A, Rivoltini L, Huber V. Recent advances on the role of tumor exosomes in immunosuppression and disease progression. Semin Cancer Biol (2012) 22:342–9.10.1016/j.semcancer.2012.02.005
    1. Bretz NP, Ridinger J, Rupp AK, Rimbach K, Keller S, Rupp C, et al. Body fluid exosomes promote secretion of inflammatory cytokines in monocytic cells via toll-like receptor signaling. J Biol Chem (2013) 288:36691–702.10.1074/jbc.M113.512806
    1. Chow A, Zhou W, Liu L, Fong MY, Champer J, Van Haute D, et al. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-κ B. Sci Rep (2014) 4:5750.10.1038/srep05750
    1. Chalmin F, Ladoire S, Mignot G, Vincent J, Bruchard M, Remy-Martin JP, et al. Membrane associated Hsp72 from tumor derived exosomes mediates STAT3 dependent immunosuppressive function of mouse and human myeloid derived suppressor cells. J Clin Invest (2010) 120:457–71.10.1172/JCI40483
    1. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells. Nat Med (2012) 18:883–91.10.1038/nm.2753
    1. Nagaraj S, Gabrilovich DI. Tumor escape mechanism governed by myeloid-derived suppressor cells. Cancer Res (2008) 68:2561–3.10.1158/0008-5472.CAN-07-6229
    1. Umansky V, Blattner C, Fleming V, Hu X, Gebhardt C, Altevogt P, et al. Myeloid-derived suppressor cells and tumor escape from immune surveillance. Semin Immunopathol (2017) 39:295–305.10.1007/s00281-016-0597-6
    1. Bronte V, Serafini P, De Stanto C, Marigo I, Tosello V, Mazzoni A, et al. IL-4-induced arginase 1 suppresses alloreactive T cells in tumor-bearing mice. J Immunol (2003) 170:270–8.10.4049/jimmunol.170.1.270
    1. Bronte V, Zanovello P. Regulation of immune responses by l-arginine metabolism. Nat Rev Immunol (2005) 5:641–54.10.1038/nri1668
    1. Rodriguez PC, Quiceno DG, Ochoa AC. l-arginine availability regulates T-lymphocyte cell-cycle progression. Blood (2007) 109:1568–74.10.1182/blood-2006-06-031856
    1. Baniyash M. TCR ζ-chain downregulation: curtailing an excessive inflammatory immune response. Nat Rev Immunol (2004) 4:675–87.10.1038/nri1434
    1. Mazzoni A, Bronte V, Visintin A, Spitzer JH, Apolloni E, Serafini P, et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol (2002) 168:689–95.10.4049/jimmunol.168.2.689
    1. Platten M, Wick W, Van den Eynde BJ. Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion. Cancer Res (2012) 72:5435–40.10.1158/0008-5472.CAN-12-0569
    1. Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron D, et al. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity (2005) 22:633–42.10.1016/j.immuni.2005.03.013
    1. Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J Exp Med (2002) 196:459–68.10.1084/jem.20020121
    1. Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol (2010) 185:3190–8.10.4049/jimmunol.0903670
    1. Fallarino F, Grohmann U, Vacca C, Orabona C, Spreca A, Fioretti MC, et al. T cell apoptosis by kynurenines. Adv Exp Med Biol (2003) 527:183–90.10.1007/978-1-4615-0135-0_21
    1. Della CM, Carlomagno S, Frumento G, Balsamo M, Cantoni C, Conte R, et al. The tryptophan catabolite l-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood (2006) 108:4118–25.10.1182/blood-2006-03-006700
    1. Hardy LL, Wick DA, Webb JR. Conversion of tyrosine to the inflammation-associated analog 3′-nitrotyrosine at either TCR- or MHC-contact positions can profoundly affect recognition of the MHC class i-restricted epitope of lymphocytic choriomeningitis virus glycoprotein 33 by CD8 T cells. J Immunol (2008) 180:5956–62.10.4049/jimmunol.180.9.5956
    1. Umemura N, Saio M, Suwa T, Kitoh Y, Bai J, Nonaka K, et al. Tumor-infiltrating myeloid-derived suppressor cells are pleiotropic-inflamed monocytes/macrophages that bear M1- and M2-type characteristics. J Leukoc Biol (2008) 83:1136–44.10.1189/jlb.090761
    1. Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand-Rosenberg S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J Immunol (2007) 179:977–83.10.4049/jimmunol.179.2.977
    1. Berger KN, Pu JJ. PD-1 pathway and its clinical application: a 20 year journey after discovery of the complete human PD-1 gene. Gene (2017) 638:20–5.10.1016/j.gene.2017.09.050
    1. Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, 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) 211:781–90.10.1084/jem.20131916
    1. Sinha P, Chornoguz O, Clements VK, Artemenko KA, Zubarev RA, Ostrand-Rosenberg S. Myeloid-derived suppressor cells express the death receptor Fas and apoptose in response to T cell-expressed FasL. Blood (2011) 117:5381–90.10.1182/blood-2010-11-321752
    1. Safarzadeh E, Orangi M, Mohammadi H, Babai F, Baradaran B. Myeloid-derived suppressor cells: important contributors to tumor progression and metastasis. J Cell Physiol (2018) 233(4):3024–36.10.1002/jcp.26075
    1. Baniyash M. Myeloid-derived suppressor cells as intruders and targets: clinical implications in cancer therapy. Cancer Immunol Immunother (2016) 65:857–67.10.1007/s00262-016-1849-y
    1. Peinado H, Lavotshkin S, Lyden D. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol (2011) 21:139–46.10.1016/j.semcancer.2011.01.002
    1. Kitamura T, Qian B, Pollard JW. Immune cell promotion of metastasis. Nat Rev Immunol (2015) 15:73–86.10.1038/nri3789
    1. Sceneay J, Parker BS, Smyth MJ, Möller A. Hypoxia-driven immunosuppression contributes to the pre-metastatic niche. Oncoimmunology (2013) 2:e22355.10.4161/onci.22355
    1. Jacob A, Prekeris R. The regulation of MMP targeting to invadopodia during cancer metastasis. Front Cell Dev Biol (2015) 3:4.10.3389/fcell.2015.00004
    1. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med (1971) 285:1182–6.10.1056/NEJM197111182852108
    1. Shen P, Wang A, He M, Wang Q, Zheng S. Increased circulating Lin−/low CD33+ HLA-DR− myeloid-derived suppressor cells in hepatocellular carcinoma patients. Hepatol Res (2014) 44:639–50.10.1111/hepr.12167
    1. Pan W, Sun Q, Wang Y, Wang J, Cao S, Ren X. Highlights on mechanisms of drugs targeting MDSCs: providing a novel perspective on cancer treatment. Tumor Biol (2015) 36:3159–69.10.1007/s13277-015-3363-9
    1. Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol (2001) 166:678–89.10.4049/jimmunol.166.1.678
    1. Gabitass RF, Annels NE, Stocken DD, Pandha HA, Middleton GW. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol Immunother (2011) 60:1419–30.10.1007/s00262-011-1028-0
    1. Eruslanov E, Neuberger M, Daurkin I, Perrin GQ, Algood C, Dahm P, et al. Circulating and tumor-infiltrating myeloid cell subsets in patients with bladder cancer. Int J Cancer (2012) 130:1109–19.10.1002/ijc.26123
    1. Jiang H, Gebhardt C, Umansky L, Beckhove P, Schulze TJ, Utikal J, et al. Elevated chronic inflammatory factors and myeloid-derived suppressor cells indicate poor prognosis in advanced melanoma patients. Int J Cancer (2015) 136:2352–60.10.1002/ijc.29297
    1. Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother (2009) 58:49–59.10.1007/s00262-008-0523-4
    1. Pico de Coaña Y, Poschke I, Gentilcore G, Mao Y, Nyström M, Hansson J, et al. Ipilimumab treatment results in an early decrease in the frequency of circulating granulocytic myeloid-derived suppressor cells as well as their arginase1 production. Cancer Immunol Res (2013) 1:158–62.10.1158/2326-6066.CIR-13-0016
    1. Gebhardt C, Sevko A, Jiang H, Lichtenberger R, Reith M, Tarnanidis K, et al. Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin Cancer Res (2015) 21:5453–9.10.1158/1078-0432.CCR-15-0676
    1. Santegoets SJ, Stam AG, Lougheed SM, Gall H, Jooss K, Sacks N, et al. Myeloid derived suppressor and dendritic cell subsets are related to clinical outcome in prostate cancer patients treated with prostate GVAX and ipilimumab. J Immunother Cancer (2014) 2:31.10.1186/s40425-014-0031-3
    1. Sade-Feldman M, Kanterman J, Klieger Y, Ish-Shalom E, Olga M, Saragovi A, 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) 22:5661–72.10.1158/1078-0432.CCR-15-3104
    1. Serafini P, Meckel K, Kelso M, Noonan K, Califano J, Koch W, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med (2006) 203:2691–702.10.1084/jem.20061104
    1. Meyer C, Sevko A, Ramacher M, Bazhin AV, Falk CS, Osen W, et al. Chronic inflammation promotes myeloid-derived suppressor cell activation blocking antitumor immunity in transgenic mouse melanoma model. Proc Natl Acad Sci U S A (2011) 108:17111–6.10.1073/pnas.1108121108
    1. Lin S, Wang J, Wang L, Wen J, Guo Y, Qiao W, et al. Phosphodiesterase-5 inhibition suppresses colonic inflammation-induced tumorigenesis via blocking the recruitment of MDSC. Am J Cancer Res (2017) 7:41–52.
    1. Califano J, Khan Z, Noonan K, Rudraraju L, Zhang Z, Wang H, et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res (2015) 21:30–8.10.1158/1078-0432.CCR-14-1716
    1. Weed DT, Vella JL, Reis IM, De la Fuente AC, Gomez C, Sargi Z, et al. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res (2015) 21:39–48.10.1158/1078-0432.CCR-14-1711
    1. Hassel JC, Jiang H, Bender C, Winkler J, Sevko A, Shevchenko I, et al. Tadalafil has biologic activity in human melanoma. Results of a pilot trial with Tadalafil in patients with metastatic melanoma (TaMe). Oncoimmunology (2017) 6:e1326440.10.1080/2162402X.2017.1326440
    1. Orillion A, Hashimoto A, Damayanti N, Shen L, Adelaiye-Ogala R, Arisa S, et al. Entinostat neutralizes myeloid derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma. Clin Cancer Res (2017) 23:5187–201.10.1158/1078-0432.CCR-17-0741
    1. Kim K, Skora AD, Li Z, Liu Q, Tam AJ, Blosser RL, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A (2014) 111:11774–9.10.1073/pnas.1410626111
    1. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer (2009) 9:798–809.10.1038/nrc2734
    1. Kortylewski M, Moreira D. Myeloid cells as a target for oligonucleotide therapeutics: turning obstacles into opportunities. Cancer Immunol Immunother (2017) 66:979–88.10.1007/s00262-017-1966-2
    1. Spinetti T, Spagnuolo L, Mottas I, Secondini C, Treinies M, Rüegg C, et al. TLR7-based cancer immunotherapy decreases intratumoral myeloid-derived suppressor cells and blocks their immunosuppressive function. Oncoimmunology (2016) 5:e1230578.10.1080/2162402X.2016.1230578
    1. Zhang Q, Hossain DM, Duttagupta P, Moreira D, Zhao X, Won H, et al. Serum-resistant CpG-STAT3 decoy for targeting survival and immune checkpoint signaling in acute myeloid leukemia. Blood (2016) 127:1687–700.10.1182/blood-2015-08-665604
    1. Hossain F, Al-Khami AA, Wyczechowska D, Hernandez C, Zheng L, Reiss K, et al. Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances cancer therapies. Cancer Immunol Res (2015) 3:1236–47.10.1158/2326-6066.CIR-15-0036
    1. Al-Khami AA, Rodriguez PC, Ochoa AC. Energy metabolic pathways control the fate and function of myeloid immune cells. J Leukoc Biol (2017) 102:369–80.10.1189/jlb.1VMR1216-535R
    1. Al-Khami AA, Zheng L, Del Valle L, Hossain F, Wyczechowska D, Zabaleta J, et al. Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells. Oncoimmunology (2017) 30:e1344804.10.1080/2162402X.2017.1344804
    1. Homey B, Müller A, Zlotnik A. Chemokines: agents for the immunotherapy of cancer? Nat Rev Immunol (2002) 2:175–84.10.1038/nri748
    1. Lesokhin AM, Hohl TM, Kitano S, Cortez C, Hirschhorn-Cymerman D, Avogadri F, 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) 72:876–86.10.1158/0008-5472.CAN-11-1792
    1. Izhak L, Wildbaum G, Weinberg U, Shaked Y, Alami J, Dumont D, et al. Predominant expression of CCL2 at the tumor site of prostate cancer patients directs a selective loss of immunological tolerance to CCL2 that could be amplified in a beneficial manner. J Immunol (2010) 184:1092–101.10.4049/jimmunol.0902725
    1. Blattner C, Fleming V, Weber R, Himmelhan BS, Altevogt P, Gebhardt C, et al. CCR5+ myeloid-derived suppressor cells are enriched and activated in melanoma lesions. Cancer Res (2018) 78:157–67.10.1158/0008-5472.CAN-17-0348
    1. Izhak L, Wildbaum G, Zohar Y, Anunu R, Klapper L, Elkeles A, et al. A novel recombinant fusion protein encoding a 20-amino acid residue of the third extracellular (E3) domain of CCR2 neutralizes the biological activity of CCL2. J Immunol (2009) 183:732–9.10.4049/jimmunol.0802746
    1. Combadiere C, Ahuja SK, Tiffany HL, Murphy PM. Cloning and functional expression of CC CKR5, a human monocyte CC chemokine receptor selective for MIP-1(alpha), MIP-1(beta), and RANTES. J Leukoc Biol (1996) 60:147–52.10.1002/jlb.60.1.147
    1. Balistreri CR, Carruba G, Calabrò M, Campisi I, Di Carlo D, Lio D, et al. CCR5 proinflammatory allele in prostate cancer risk: a pilot study in patients and centenarians from Sicily. Ann N Y Acad Sci (2009) 1155:289–92.10.1111/j.1749-6632.2008.03691.x
    1. Umansky V, Blattner C, Gebhardt C, Utikal J. CCR5 in recruitment and activation of myeloid-derived suppressor cells in melanoma. Cancer Immunol Immunother (2017) 66:1015–23.10.1007/s00262-017-1988-9
    1. Robinson SC, Scott KA, Wilson JL, Thompson RG, Proudfoot AE, Balkwill FR. A chemokine receptor antagonist inhibits experimental breast tumor growth. Cancer Res (2003) 63:8360–5.
    1. Tan MC, Goedegebuure PS, Belt BA, Flaherty B, Sankpal N, Gillanders WE, et al. Disruption of CCR5-dependent homing of regulatory T cells inhibits tumor growth in a murine model of pancreatic cancer. J Immunol (2009) 182:1746–55.10.4049/jimmunol.182.3.1746
    1. Velasco-Velázquez M, Jiao X, De La Fuente M, Pestell TG, Ertel A, Lisanti MP, et al. CCR5 antagonist blocks metastasis of basal breast cancer cells. Cancer Res (2012) 72:3839–50.10.1158/0008-5472.CAN-11-3917
    1. Nefedova Y, Fishman M, Sherman S, Wang X, Beg AA, Gabrilovich DI. Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells. Cancer Res (2007) 67:11021–8.10.1158/0008-5472.CAN-07-2593
    1. Li Y, Wongsiriroj N, Blaner WS. The multifaceted nature of retinoid transport and metabolism. Hepatobiliary Surg Nutr (2014) 3:126–39.10.3978/j.issn.2304-3881.2014.05.04
    1. Mirza N, Fishman M, Fricke I, Dunn M, Neuger AM, Frost TJ, et al. All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients. Cancer Res (2006) 66:9299–307.10.1158/0008-5472.CAN-06-1690
    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) 6:909–18.10.1007/s00262-013-1396-8
    1. Kodera Y, Katanasaka Y, Kitamura Y, Tsuda H, Nishio K, Tamura T, et al. Sunitinib inhibits lymphatic endothelial cell functions and lymph node metastasis in a breast cancer model through inhibition of vascular endothelial growth factor receptor 3. Breast Cancer Res (2011) 13(3):R66.10.1186/bcr2903
    1. Ko JS, Zea AH, Rini BI, Ireland JL, Elson P, Cohen P, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res (2009) 15:2148–57.10.1158/1078-0432.CCR-08-1332
    1. Nayak D, Roth TL, Mcgavern DB. Myeloid-derived suppressor cells as an immune parameter in patients with concurrent sunitinib and stereotactic body radiotherapy. Clin Cancer Res (2010) 21:4073–85.10.1158/1078-0432.CCR-14-2742
    1. Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res (2010) 70:3052–61.10.1158/0008-5472.CAN-09-3690
    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 (2016) 14:282.10.1186/s12967-016-1037-z
    1. Kanterman J, Sade-Feldman M, Biton M, Ish-Shalom E, Lasry A, Goldshtein A, et al. Adverse immunoregulatory effects of 5FU and CPT11 chemotherapy on myeloid-derived suppressor cells and colorectal cancer outcomes. Cancer Res (2014) 74:6022–35.10.1158/0008-5472.CAN-14-0657
    1. Kodumudi KN, Woan K, Gilvary DL, Sahakian E, Wei S, Djeu JY. A novel chemoimmunomodulating property of docetaxel: suppression of myeloid-derived suppressor cells in tumor bearers. Clin Cancer Res (2010) 16:4583–94.10.1158/1078-0432.CCR-10-0733
    1. Alizadeh D, Trad M, Hanke NT, Larmonier CB, Janikashvili N, Bonnotte B, et al. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res (2014) 74:104–18.10.1158/0008-5472.CAN-13-1545
    1. Sevko A, Michels T, Vrohlings M, Umansky L, Beckhove P, Kato M, et al. Antitumor effect of paclitaxel is mediated by inhibition of myeloid-derived suppressor cells and chronic inflammation in the spontaneous melanoma model. J Immunol (2013) 190:2464–71.10.4049/jimmunol.1202781
    1. Michels T, Shurin GV, Naiditch H, Sevko A, Umansky V, Shurin MR. Paclitaxel promotes differentiation of myeloid-derived suppressor cells into dendritic cells in vitro in a TLR4-independent manner. J Immunotoxicol (2012) 9:292–300.10.3109/1547691X.2011.642418
    1. Danishmalik SN, Sin JI. Therapeutic tumor control of HER2 DNA vaccines is achieved by an alteration of tumor cells and tumor microenvironment by gemcitabine and anti-Gr-1 Ab treatment in a HER2-expressing tumor model. DNA Cell Biol (2017) 6:801–11.10.1089/dna.2017.3810
    1. Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells – an overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology (2015) 4:e954829.10.4161/21624011.2014.954829
    1. Tobin RP, Davis D, Jordan KR, McCarter MD. The clinical evidence for targeting human myeloid-derived suppressor cells in cancer patients. J Leukoc Biol (2017) 102:381–91.10.1189/jlb.5VMR1016-449R

Source: PubMed

3
Abonner