Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer
Tóm tắt
The emergence of immunotherapy has been an astounding breakthrough in cancer treatments. In particular, immune checkpoint inhibitors, targeting PD-1 and CTLA-4, have shown remarkable therapeutic outcomes. However, response rates from immunotherapy have been reported to be varied, with some having pronounced success and others with minimal to no clinical benefit. An important aspect associated with this discrepancy in patient response is the immune-suppressive effects elicited by the tumour microenvironment (TME). Immune suppression plays a pivotal role in regulating cancer progression, metastasis, and reducing immunotherapy success. Most notably, myeloid-derived suppressor cells (MDSC), a heterogeneous population of immature myeloid cells, have potent mechanisms to inhibit T-cell and NK-cell activity to promote tumour growth, development of the pre-metastatic niche, and contribute to resistance to immunotherapy. Accumulating research indicates that MDSC can be a therapeutic target to alleviate their pro-tumourigenic functions and immunosuppressive activities to bolster the efficacy of checkpoint inhibitors. In this review, we provide an overview of the general immunotherapeutic approaches and discuss the characterisation, expansion, and activities of MDSCs with the current treatments used to target them either as a single therapeutic target or synergistically in combination with immunotherapy.
Từ khóa
Tài liệu tham khảo
Algarra, 2003, MHC class I antigens, immune surveillance, and tumor immune escape, J. Cell Physiol., 195, 346, 10.1002/jcp.10290
Burkholder, 2014, Tumor-induced perturbations of cytokines and immune cell networks, Biochim. Biophys. Acta, 1845, 182
Salgado, 2015, The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: Recommendations by an International TILs Working Group 2014, Ann. Oncol., 26, 259, 10.1093/annonc/mdu450
Chow, 2012, Inflammation and immune surveillance in cancer, Semin Cancer Biol., 22, 23, 10.1016/j.semcancer.2011.12.004
2018, Single-Cell Transcriptomics in Cancer Immunobiology: The Future of Precision Oncology, Front. Immunol., 9, 2582, 10.3389/fimmu.2018.02582
Lollini, 2015, The Promise of Preventive Cancer Vaccines, Vaccines (Basel), 3, 467, 10.3390/vaccines3020467
Zhang, 2018, Current status and future directions of cancer immunotherapy, J. Cancer, 9, 1773, 10.7150/jca.24577
Lim, 2009, Prevention of hepatocellular carcinoma in hepatitis B virus infection, J. Gastroenterol. Hepatol., 24, 1352, 10.1111/j.1440-1746.2009.05985.x
Mahdavi, 2005, Vaccines against human papillomavirus and cervical cancer: Promises and challenges, Oncologist, 10, 528, 10.1634/theoncologist.10-7-528
Vigneron, 2015, Human Tumor Antigens and Cancer Immunotherapy, Biomed. Res. Int., 2015, 948501, 10.1155/2015/948501
Hutchison, 2018, Identifying neoantigens for use in immunotherapy, Mamm. Genome, 29, 714, 10.1007/s00335-018-9771-6
Vermaelen, 2019, Vaccine Strategies to Improve Anti-cancer Cellular Immune Responses, Front. Immunol., 10, 8, 10.3389/fimmu.2019.00008
Mougel, 2019, Therapeutic Cancer Vaccine and Combinations With Antiangiogenic Therapies and Immune Checkpoint Blockade, Front. Immunol., 10, 467, 10.3389/fimmu.2019.00467
Hollingsworth, 2019, Turning the corner on therapeutic cancer vaccines, NPJ Vaccines, 4, 7, 10.1038/s41541-019-0103-y
Karlitepe, 2015, New approaches for cancer immunotherapy, Tumour Biol., 36, 4075, 10.1007/s13277-015-3491-2
Dahlen, 2018, Bispecific antibodies in cancer immunotherapy, Ther. Adv. Vaccines Immunother., 6, 3, 10.1177/2515135518763280
Vu, 2012, Trastuzumab: Updated mechanisms of action and resistance in breast cancer, Front. Oncol., 2, 62, 10.3389/fonc.2012.00062
McKeage, 2002, Trastuzumab: A review of its use in the treatment of metastatic breast cancer overexpressing HER2, Drugs, 62, 209, 10.2165/00003495-200262010-00008
Met, 2019, Principles of adoptive T cell therapy in cancer, Semin Immunopathol., 41, 49, 10.1007/s00281-018-0703-z
Dudley, 2002, Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes, Science, 298, 850, 10.1126/science.1076514
Rosenberg, 2011, Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy, Clin. Cancer Res., 17, 4550, 10.1158/1078-0432.CCR-11-0116
Besser, 2013, Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: Intent-to-treat analysis and efficacy after failure to prior immunotherapies, Clin. Cancer Res., 19, 4792, 10.1158/1078-0432.CCR-13-0380
Lee, 2017, Expansion of tumor-infiltrating lymphocytes and their potential for application as adoptive cell transfer therapy in human breast cancer, Oncotarget, 8, 113345, 10.18632/oncotarget.23007
Houot, 2015, T-cell-based Immunotherapy: Adoptive Cell Transfer and Checkpoint Inhibition, Cancer Immunol. Res., 3, 1115, 10.1158/2326-6066.CIR-15-0190
Rohaan, 2019, Adoptive cellular therapies: The current landscape, Virchows Arch., 474, 449, 10.1007/s00428-018-2484-0
June, 2015, Adoptive cellular therapy: A race to the finish line, Sci. Transl. Med., 7, 280ps7, 10.1126/scitranslmed.aaa3643
Maude, 2014, Chimeric antigen receptor T-cell therapy for ALL, Hematol. Am. Soc. Hematol. Educ. Program., 2014, 559, 10.1182/asheducation-2014.1.559
Pehlivan, 2018, CAR-T Cell Therapy for Acute Lymphoblastic Leukemia: Transforming the Treatment of Relapsed and Refractory Disease, Curr. Hematol. Malig. Rep., 13, 396, 10.1007/s11899-018-0470-x
Gattinoni, 2016, Adoptive T cell transfer: Imagining the next generation of cancer immunotherapies, Semin. Immunol., 28, 1, 10.1016/j.smim.2016.03.019
Li, 2018, Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: Lessons learned and strategies for moving forward, J. Hematol. Oncol., 11, 22, 10.1186/s13045-018-0568-6
Chu, 2018, Versatile CAR T-cells for cancer immunotherapy, Contemp. Oncol. (Pozn), 22, 73
2013, Breakthrough of the year 2013. Cancer immunotherapy, Science, 342, 1432
Guo, Z.S. (2018). The 2018 Nobel Prize in medicine goes to cancer immunotherapy (editorial for BMC cancer). BMC Cancer, 18.
Sambi, 2019, Current Challenges in Cancer Immunotherapy: Multimodal Approaches to Improve Efficacy and Patient Response Rates, J. Oncol., 2019, 4508794, 10.1155/2019/4508794
Escors, D. (2014). Tumour immunogenicity, antigen presentation and immunological barriers in cancer immunotherapy. New J. Sci., 2014.
Lipson, 2015, Antagonists of PD-1 and PD-L1 in Cancer Treatment, Semin. Oncol., 42, 587, 10.1053/j.seminoncol.2015.05.013
Nanda, 2016, Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study, J. Clin. Oncol., 34, 2460, 10.1200/JCO.2015.64.8931
Ventola, 2017, Cancer Immunotherapy, Part 3: Challenges and Future Trends, Pharm. Ther., 42, 514
Rugo, 2018, Safety and Antitumor Activity of Pembrolizumab in Patients with Estrogen Receptor-Positive/Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer, Clin. Cancer Res., 24, 2804, 10.1158/1078-0432.CCR-17-3452
Yang, 2017, Recognizing and managing on toxicities in cancer immunotherapy, Tumour Biol., 39, 1010428317694542, 10.1177/1010428317694542
Nikanjam, 2017, Dosing immunotherapy combinations: Analysis of 3,526 patients for toxicity and response patterns, Oncoimmunology, 6, e1338997, 10.1080/2162402X.2017.1338997
Hendry, 2016, The Role of the Tumor Vasculature in the Host Immune Response: Implications for Therapeutic Strategies Targeting the Tumor Microenvironment, Front. Immunol., 7, 621, 10.3389/fimmu.2016.00621
Tuccitto, 2019, Immunosuppressive circuits in tumor microenvironment and their influence on cancer treatment efficacy, Virchows Arch., 474, 407, 10.1007/s00428-018-2477-z
Yang, 2004, Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis, Cancer Cell, 6, 409, 10.1016/j.ccr.2004.08.031
Motz, 2014, Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors, Nat. Med., 20, 607, 10.1038/nm.3541
Gabrilovich, 2009, Myeloid-derived suppressor cells as regulators of the immune system, Nat. Rev. Immunol., 9, 162, 10.1038/nri2506
Frydrychowicz, 2017, The Dual Role of Treg in Cancer, Scand. J. Immunol., 86, 436, 10.1111/sji.12615
Takeuchi, 2016, Roles of regulatory T cells in cancer immunity, Int. Immunol., 28, 401, 10.1093/intimm/dxw025
Weber, 2018, Myeloid-Derived Suppressor Cells Hinder the Anti-Cancer Activity of Immune Checkpoint Inhibitors, Front. Immunol., 9, 1310, 10.3389/fimmu.2018.01310
Gabrilovich, 2017, Myeloid-Derived Suppressor Cells, Cancer Immunol. Res., 5, 3, 10.1158/2326-6066.CIR-16-0297
Takizawa, 2012, Demand-adapted regulation of early hematopoiesis in infection and inflammation, Blood, 119, 2991, 10.1182/blood-2011-12-380113
2016, Improving cancer immunotherapy by targeting the STATe of MDSCs, Oncoimmunology, 5, e1196312, 10.1080/2162402X.2016.1196312
Gabrilovich, 2012, Coordinated regulation of myeloid cells by tumours, Nat. Rev. Immunol., 12, 253, 10.1038/nri3175
Umansky, V. (2016). The Role of Myeloid-Derived Suppressor Cells (MDSC) in Cancer Progression. Vaccines (Basel), 4.
Meirow, 2015, Paving the Road to Tumor Development and Spreading: Myeloid-Derived Suppressor Cells are Ruling the Fate, Front. Immunol., 6, 523, 10.3389/fimmu.2015.00523
Sawanobori, 2008, Chemokine-mediated rapid turnover of myeloid-derived suppressor cells in tumor-bearing mice, Blood, 111, 5457, 10.1182/blood-2008-01-136895
Sinha, 2009, Myeloid-derived suppressor cells: Linking inflammation and cancer, J. Immunol., 182, 4499, 10.4049/jimmunol.0802740
Sica, 2007, Altered macrophage differentiation and immune dysfunction in tumor development, J. Clin. Invest., 117, 1155, 10.1172/JCI31422
Condamine, 2015, Transcriptional regulation of myeloid-derived suppressor cells, J. Leukoc. Biol., 98, 913, 10.1189/jlb.4RI0515-204R
Yan, 2010, Gr-1+CD11b+ myeloid cells tip the balance of immune protection to tumor promotion in the premetastatic lung, Cancer Res., 70, 6139, 10.1158/0008-5472.CAN-10-0706
Gao, 2012, Myeloid progenitor cells in the premetastatic lung promote metastases by inducing mesenchymal to epithelial transition, Cancer Res., 72, 1384, 10.1158/0008-5472.CAN-11-2905
Erler, 2009, Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche, Cancer Cell, 15, 35, 10.1016/j.ccr.2008.11.012
Sceneay, 2012, Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche, Cancer Res., 72, 3906, 10.1158/0008-5472.CAN-11-3873
Zhang, S. (2016). The Role of Myeloid-Derived Suppressor Cells in Patients with Solid Tumors: A Meta-Analysis. PLoS ONE, 11.
Law, 2017, The innate and adaptive infiltrating immune systems as targets for breast cancer immunotherapy, Endocr. Relat. Cancer, 24, R123, 10.1530/ERC-16-0404
Solito, 2014, Myeloid-derived suppressor cell heterogeneity in human cancers, Ann. N Y Acad. Sci., 1319, 47, 10.1111/nyas.12469
Vetsika, 2014, A circulating subpopulation of monocytic myeloid-derived suppressor cells as an independent prognostic/predictive factor in untreated non-small lung cancer patients, J. Immunol. Res., 2014, 659294, 10.1155/2014/659294
Hoechst, 2008, A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells, Gastroenterology, 135, 234, 10.1053/j.gastro.2008.03.020
Obermajer, 2011, Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells toward stable myeloid-derived suppressor cells, Blood, 118, 5498, 10.1182/blood-2011-07-365825
2009, Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy, Cancer Immunol. Immunother., 58, 49, 10.1007/s00262-008-0523-4
Zhao, 2012, S100A9 a new marker for monocytic human myeloid-derived suppressor cells, Immunology, 136, 176, 10.1111/j.1365-2567.2012.03566.x
Trikha, 2014, Signaling pathways involved in MDSC regulation, Biochim. Biophys. Acta, 1846, 55
Lesokhin, 2012, Monocytic CCR2(+) myeloid-derived suppressor cells promote immune escape by limiting activated CD8 T-cell infiltration into the tumor microenvironment, Cancer Res., 72, 876, 10.1158/0008-5472.CAN-11-1792
Liang, 2017, Host STING-dependent MDSC mobilization drives extrinsic radiation resistance, Nat. Commun., 8, 1736, 10.1038/s41467-017-01566-5
Blattner, 2018, CCR5(+) Myeloid-Derived Suppressor Cells Are Enriched and Activated in Melanoma Lesions, Cancer Res., 78, 157, 10.1158/0008-5472.CAN-17-0348
Umansky, 2017, CCR5 in recruitment and activation of myeloid-derived suppressor cells in melanoma, Cancer Immunol. Immunother., 66, 1015, 10.1007/s00262-017-1988-9
Metelitsa, 2011, Anti-tumor potential of type-I NKT cells against CD1d-positive and CD1d-negative tumors in humans, Clin. Immunol., 140, 119, 10.1016/j.clim.2010.10.005
Youn, 2010, The biology of myeloid-derived suppressor cells: The blessing and the curse of morphological and functional heterogeneity, Eur. J. Immunol., 40, 2969, 10.1002/eji.201040895
Youn, 2008, Subsets of myeloid-derived suppressor cells in tumor-bearing mice, J. Immunol., 181, 5791, 10.4049/jimmunol.181.8.5791
Karakasheva, 2015, CD38-Expressing Myeloid-Derived Suppressor Cells Promote Tumor Growth in a Murine Model of Esophageal Cancer, Cancer Res., 75, 4074, 10.1158/0008-5472.CAN-14-3639
Ryzhov, 2011, Adenosinergic regulation of the expansion and immunosuppressive activity of CD11b+Gr1+ cells, J. Immunol., 187, 6120, 10.4049/jimmunol.1101225
Umansky, 2014, Extracellular adenosine metabolism in immune cells in melanoma, Cancer Immunol. Immunother., 63, 1073, 10.1007/s00262-014-1553-8
Wang, 2015, Functional characterization of myeloid-derived suppressor cell subpopulations during the development of experimental arthritis, Eur. J. Immunol., 45, 464, 10.1002/eji.201444799
Ding, 2015, CD40 controls CXCR5-induced recruitment of myeloid-derived suppressor cells to gastric cancer, Oncotarget, 6, 38901, 10.18632/oncotarget.5644
Pan, 2010, Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer, Cancer Res., 70, 99, 10.1158/0008-5472.CAN-09-1882
Shen, 2014, Downregulation of CD40 expression contributes to the accumulation of myeloid-derived suppressor cells in gastric tumors, Oncol Lett., 8, 775, 10.3892/ol.2014.2174
Cho, 2016, Immunomodulatory effect of captopril and local irradiation on myeloid-derived suppressor cells, Radiat. Oncol. J., 34, 223, 10.3857/roj.2016.01816
Gallego-Ortega, D. (2015). ELF5 Drives Lung Metastasis in Luminal Breast Cancer through Recruitment of Gr1+ CD11b+ Myeloid-Derived Suppressor Cells. PLoS Biol., 13.
Haile, 2010, CD49d is a new marker for distinct myeloid-derived suppressor cell subpopulations in mice, J. Immunol., 185, 203, 10.4049/jimmunol.0903573
2010, Myeloid-derived suppressor cells: More mechanisms for inhibiting antitumor immunity, Cancer Immunol. Immunother., 59, 1593, 10.1007/s00262-010-0855-8
Lacotte, 2016, Impact of myeloid-derived suppressor cell on Kupffer cells from mouse livers with hepatocellular carcinoma, Oncoimmunology, 5, e1234565, 10.1080/2162402X.2016.1234565
Marigo, 2010, Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor, Immunity, 32, 790, 10.1016/j.immuni.2010.05.010
Liechtenstein, 2014, A highly efficient tumor-infiltrating MDSC differentiation system for discovery of anti-neoplastic targets, which circumvents the need for tumor establishment in mice, Oncotarget, 5, 7843, 10.18632/oncotarget.2279
Dong, 2011, CD71 is selectively and ubiquitously expressed at high levels in erythroid precursors of all maturation stages: A comparative immunochemical study with glycophorin A and hemoglobin A, Am. J. Surg. Pathol., 35, 723, 10.1097/PAS.0b013e31821247a8
Movahedi, 2008, Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity, Blood, 111, 4233, 10.1182/blood-2007-07-099226
Bunt, 2009, Inflammation enhances myeloid-derived suppressor cell cross-talk by signaling through Toll-like receptor 4, J. Leukoc. Biol., 85, 996, 10.1189/jlb.0708446
Sansom, 2000, CD28, CTLA-4 and their ligands: Who does what and to whom?, Immunology, 101, 169, 10.1046/j.1365-2567.2000.00121.x
Yang, 2006, CD80 in immune suppression by mouse ovarian carcinoma-associated Gr-1+CD11b+ myeloid cells, Cancer Res., 66, 6807, 10.1158/0008-5472.CAN-05-3755
Yang, 2013, LPS converts Gr-1(+)CD115(+) myeloid-derived suppressor cells from M2 to M1 via P38 MAPK, Exp. Cell Res., 319, 1774, 10.1016/j.yexcr.2013.05.007
Youn, 2012, Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice, J. Leukoc. Biol., 91, 167, 10.1189/jlb.0311177
Albeituni, 2013, Hampering immune suppressors: Therapeutic targeting of myeloid-derived suppressor cells in cancer, Cancer J., 19, 490, 10.1097/PPO.0000000000000006
Tang, 1992, M-CSF (monocyte colony stimulating factor) and M-CSF receptor expression by breast tumour cells: M-CSF mediated recruitment of tumour infiltrating monocytes?, J. Cell Biochem., 50, 350, 10.1002/jcb.240500403
Hu, 2014, Transmembrane TNF-alpha promotes suppressive activities of myeloid-derived suppressor cells via TNFR2, J. Immunol., 192, 1320, 10.4049/jimmunol.1203195
Mandruzzato, 2009, IL4Ralpha+ myeloid-derived suppressor cell expansion in cancer patients, J. Immunol., 182, 6562, 10.4049/jimmunol.0803831
Sinha, 2012, Tumor-induced myeloid-derived suppressor cell function is independent of IFN-gamma and IL-4Ralpha, Eur. J. Immunol., 42, 2052, 10.1002/eji.201142230
Gallina, 2006, Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells, J. Clin. Invest., 116, 2777, 10.1172/JCI28828
Roth, 2012, Aptamer-mediated blockade of IL4Ralpha triggers apoptosis of MDSCs and limits tumor progression, Cancer Res., 72, 1373, 10.1158/0008-5472.CAN-11-2772
Schouppe, 2013, Tumor-induced myeloid-derived suppressor cell subsets exert either inhibitory or stimulatory effects on distinct CD8+ T-cell activation events, Eur. J. Immunol., 43, 2930, 10.1002/eji.201343349
Goldmann, 2017, Identification of a Novel Subset of Myeloid-Derived Suppressor Cells During Chronic Staphylococcal Infection That Resembles Immature Eosinophils, J. Infect. Dis., 216, 1444, 10.1093/infdis/jix494
Georgoudaki, 2015, CD244 is expressed on dendritic cells and regulates their functions, Immunol. Cell Biol., 93, 581, 10.1038/icb.2014.124
Lu, 2016, The expression profiles and regulation of PD-L1 in tumor-induced myeloid-derived suppressor cells, Oncoimmunology, 5, e1247135, 10.1080/2162402X.2016.1247135
Lee, 2016, Myeloid-Derived Suppressor Cells Are Controlled by Regulatory T Cells via TGF-beta during Murine Colitis, Cell Rep., 17, 3219, 10.1016/j.celrep.2016.11.062
Chiu, 2016, Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26, Hepatology, 64, 797, 10.1002/hep.28655
Hart, 2009, Phenotypic and functional delineation of murine CX(3)CR1 monocyte-derived cells in ovarian cancer, Neoplasia, 11, 564, 10.1593/neo.09228
Zhao, 2015, Subsets of myeloid-derived suppressor cells in hepatocellular carcinoma express chemokines and chemokine receptors differentially, Int. Immunopharmacol., 26, 314, 10.1016/j.intimp.2015.04.010
Katoh, 2013, CXCR2-expressing myeloid-derived suppressor cells are essential to promote colitis-associated tumorigenesis, Cancer Cell, 24, 631, 10.1016/j.ccr.2013.10.009
2015, Subpopulations of M-MDSCs from mice infected by an immunodeficiency-causing retrovirus and their differential suppression of T- vs B-cell responses, Virology, 485, 263, 10.1016/j.virol.2015.07.020
Ortiz, 2014, Myeloid-derived suppressor cells in the development of lung cancer, Cancer Immunol. Res., 2, 50, 10.1158/2326-6066.CIR-13-0129
Wang, 2014, Galectin-3 contributes to cisplatin-induced myeloid derived suppressor cells (MDSCs) recruitment in Lewis lung cancer-bearing mice, Mol. Biol. Rep., 41, 4069, 10.1007/s11033-014-3276-5
Pyzer, 2016, Myeloid-derived suppressor cells as effectors of immune suppression in cancer, Int. J. Cancer, 139, 1915, 10.1002/ijc.30232
Talmadge, 2013, History of myeloid-derived suppressor cells, Nat. Rev. Cancer, 13, 739, 10.1038/nrc3581
Nagaraj, 2012, Antigen-specific CD4(+) T cells regulate function of myeloid-derived suppressor cells in cancer via retrograde MHC class II signaling, Cancer Res., 72, 928, 10.1158/0008-5472.CAN-11-2863
Sato, 2015, Characterization of the myeloid-derived suppressor cell subset regulated by NK cells in malignant lymphoma, Oncoimmunology, 4, e995541, 10.1080/2162402X.2014.995541
Zoglmeier, 2011, CpG blocks immunosuppression by myeloid-derived suppressor cells in tumor-bearing mice, Clin. Cancer Res., 17, 1765, 10.1158/1078-0432.CCR-10-2672
Horikawa, 2017, Expression of Vascular Endothelial Growth Factor in Ovarian Cancer Inhibits Tumor Immunity through the Accumulation of Myeloid-Derived Suppressor Cells, Clin. Cancer Res., 23, 587, 10.1158/1078-0432.CCR-16-0387
Kumar, 2016, The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment, Trends Immunol., 37, 208, 10.1016/j.it.2016.01.004
Huang, 2007, CCL2/CCR2 pathway mediates recruitment of myeloid suppressor cells to cancers, Cancer Lett., 252, 86, 10.1016/j.canlet.2006.12.012
Obermajer, 2011, PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment, Cancer Res., 71, 7463, 10.1158/0008-5472.CAN-11-2449
Bronte, 2016, Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards, Nat. Commun., 7, 12150, 10.1038/ncomms12150
Zhang, 2017, CD13(hi) Neutrophil-like myeloid-derived suppressor cells exert immune suppression through Arginase 1 expression in pancreatic ductal adenocarcinoma, Oncoimmunology, 6, e1258504, 10.1080/2162402X.2016.1258504
Resheq, 2017, Impaired Transmigration of Myeloid-Derived Suppressor Cells across Human Sinusoidal Endothelium Is Associated with Decreased Expression of CD13, J. Immunol., 199, 1672, 10.4049/jimmunol.1600466
Haziot, 1993, Neutrophil CD14: Biochemical properties and role in the secretion of tumor necrosis factor-alpha in response to lipopolysaccharide, J. Immunol., 150, 5556, 10.4049/jimmunol.150.12.5556
Damuzzo, 2015, Complexity and challenges in defining myeloid-derived suppressor cells, Cytometry B Clin. Cytom., 88, 77, 10.1002/cytob.21206
Romano, 2015, Circulating myeloid-derived suppressor cells correlate with clinical outcome in Hodgkin Lymphoma patients treated up-front with a risk-adapted strategy, Br. J. Haematol., 168, 689, 10.1111/bjh.13198
Karakasheva, T.A. (2018). CD38+ M-MDSC expansion characterizes a subset of advanced colorectal cancer patients. JCI Insight, 3.
Li, 2018, Metformin-Induced Reduction of CD39 and CD73 Blocks Myeloid-Derived Suppressor Cell Activity in Patients with Ovarian Cancer, Cancer Res., 78, 1779, 10.1158/0008-5472.CAN-17-2460
Li, 2017, CD39/CD73 upregulation on myeloid-derived suppressor cells via TGF-beta-mTOR-HIF-1 signaling in patients with non-small cell lung cancer, Oncoimmunology, 6, e1320011, 10.1080/2162402X.2017.1320011
Rodriguez, 2009, Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes, Cancer Res., 69, 1553, 10.1158/0008-5472.CAN-08-1921
Shao, 2018, Distinct alterations of CD68(+)CD163(+) M2-like macrophages and myeloid-derived suppressor cells in newly diagnosed primary immune thrombocytopenia with or without CR after high-dose dexamethasone treatment, J. Transl. Med., 16, 48, 10.1186/s12967-018-1424-8
Bergenfelz, C. (2015). Systemic Monocytic-MDSCs Are Generated from Monocytes and Correlate with Disease Progression in Breast Cancer Patients. PLoS ONE, 10.
Greten, 2011, Myeloid derived suppressor cells in human diseases, Int. Immunopharmacol., 11, 802, 10.1016/j.intimp.2011.01.003
Poschke, 2010, Immature immunosuppressive CD14+HLA-DR-/low cells in melanoma patients are Stat3hi and overexpress CD80, CD83, and DC-sign, Cancer Res., 70, 4335, 10.1158/0008-5472.CAN-09-3767
Ju, 2016, The Analysis of CD83 Expression on Human Immune Cells Identifies a Unique CD83+-Activated T Cell Population, J. Immunol., 197, 4613, 10.4049/jimmunol.1600339
Zhang, B. (2013). Circulating and tumor-infiltrating myeloid-derived suppressor cells in patients with colorectal carcinoma. PLoS ONE, 8.
OuYang, 2015, Tumor-induced myeloid-derived suppressor cells promote tumor progression through oxidative metabolism in human colorectal cancer, J. Transl. Med., 13, 47, 10.1186/s12967-015-0410-7
Kohanbash, 2013, GM-CSF promotes the immunosuppressive activity of glioma-infiltrating myeloid cells through interleukin-4 receptor-alpha, Cancer Res., 73, 6413, 10.1158/0008-5472.CAN-12-4124
Seo, 2018, Association of Chemokines and Chemokine Receptor Expression with Monocytic-Myeloid-Derived Suppressor Cells during Tumor Progression, Immune Netw, 18, e23, 10.4110/in.2018.18.e23
Highfill, 2014, Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy, Sci. Transl Med., 6, 237ra67, 10.1126/scitranslmed.3007974
Kusmartsev, 2008, Oxidative stress regulates expression of VEGFR1 in myeloid cells: Link to tumor-induced immune suppression in renal cell carcinoma, J. Immunol., 181, 346, 10.4049/jimmunol.181.1.346
Kitamura, 2015, CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages, J. Exp. Med., 212, 1043, 10.1084/jem.20141836
Yu, 2013, Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer, J. Immunol., 190, 3783, 10.4049/jimmunol.1201449
Qian, 2011, CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis, Nature, 475, 222, 10.1038/nature10138
Chun, 2015, CCL2 Promotes Colorectal Carcinogenesis by Enhancing Polymorphonuclear Myeloid-Derived Suppressor Cell Population and Function, Cell Rep., 12, 244, 10.1016/j.celrep.2015.06.024
Reichel, 2012, C-C motif chemokine CCL3 and canonical neutrophil attractants promote neutrophil extravasation through common and distinct mechanisms, Blood, 120, 880, 10.1182/blood-2012-01-402164
Condamine, 2015, Regulation of tumor metastasis by myeloid-derived suppressor cells, Annu. Rev. Med., 66, 97, 10.1146/annurev-med-051013-052304
Hiratsuka, 2006, Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis, Nat. Cell Biol., 8, 1369, 10.1038/ncb1507
Steele, 2016, CXCR2 Inhibition Profoundly Suppresses Metastases and Augments Immunotherapy in Pancreatic Ductal Adenocarcinoma, Cancer Cell, 29, 832, 10.1016/j.ccell.2016.04.014
Nefedova, 2004, Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer, J. Immunol., 172, 464, 10.4049/jimmunol.172.1.464
Arai, 2008, S100A8 and S100A9 overexpression is associated with poor pathological parameters in invasive ductal carcinoma of the breast, Curr. Cancer Drug Targets, 8, 243, 10.2174/156800908784533445
Sinha, 2008, Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells, J. Immunol., 181, 4666, 10.4049/jimmunol.181.7.4666
Wang, 2013, Increased myeloid-derived suppressor cells in gastric cancer correlate with cancer stage and plasma S100A8/A9 proinflammatory proteins, J. Immunol., 190, 794, 10.4049/jimmunol.1202088
Kumar, 2016, CD45 Phosphatase Inhibits STAT3 Transcription Factor Activity in Myeloid Cells and Promotes Tumor-Associated Macrophage Differentiation, Immunity, 44, 303, 10.1016/j.immuni.2016.01.014
Corzo, 2010, HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment, J. Exp. Med., 207, 2439, 10.1084/jem.20100587
Liu, 2014, SIRT1 limits the function and fate of myeloid-derived suppressor cells in tumors by orchestrating HIF-1alpha-dependent glycolysis, Cancer Res., 74, 727, 10.1158/0008-5472.CAN-13-2584
Noman, 2014, PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation, J. Exp. Med., 211, 781, 10.1084/jem.20131916
Huang, 2006, 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., 66, 1123, 10.1158/0008-5472.CAN-05-1299
Srivastava, 2010, Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine, Cancer Res., 70, 68, 10.1158/0008-5472.CAN-09-2587
Fletcher, 2015, l-Arginine depletion blunts antitumor T-cell responses by inducing myeloid-derived suppressor cells, Cancer Res., 75, 275, 10.1158/0008-5472.CAN-14-1491
Bronte, 2005, Regulation of immune responses by L-arginine metabolism, Nat. Rev. Immunol., 5, 641, 10.1038/nri1668
Bingisser, 1998, Macrophage-derived nitric oxide regulates T cell activation via reversible disruption of the Jak3/STAT5 signaling pathway, J. Immunol., 160, 5729, 10.4049/jimmunol.160.12.5729
Rivoltini, 2002, Immunity to cancer: Attack and escape in T lymphocyte-tumor cell interaction, Immunol. Rev., 188, 97, 10.1034/j.1600-065X.2002.18809.x
Rodriguez, 2007, L-arginine availability regulates T-lymphocyte cell-cycle progression, Blood, 109, 1568, 10.1182/blood-2006-06-031856
Harari, 2004, Inhibition of MHC II gene transcription by nitric oxide and antioxidants, Curr. Pharm. Des., 10, 893, 10.2174/1381612043452893
Nagaraj, 2007, Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer, Nat. Med., 13, 828, 10.1038/nm1609
Lu, 2011, Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice, J. Clin. Invest., 121, 4015, 10.1172/JCI45862
Kusmartsev, 2003, Inhibition of myeloid cell differentiation in cancer: The role of reactive oxygen species, J. Leukoc. Biol., 74, 186, 10.1189/jlb.0103010
Kusmartsev, 2004, Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species, J. Immunol., 172, 989, 10.4049/jimmunol.172.2.989
Molon, 2011, Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells, J. Exp. Med., 208, 1949, 10.1084/jem.20101956
Beury, 2016, Myeloid-Derived Suppressor Cell Survival and Function Are Regulated by the Transcription Factor Nrf2, J. Immunol., 196, 3470, 10.4049/jimmunol.1501785
2006, The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function, Blood, 108, 4118, 10.1182/blood-2006-03-006700
Song, 2011, L-kynurenine-induced apoptosis in human NK cells is mediated by reactive oxygen species, Int. Immunopharmacol., 11, 932, 10.1016/j.intimp.2011.02.005
Fleming, 2018, Targeting Myeloid-Derived Suppressor Cells to Bypass Tumor-Induced Immunosuppression, Front. Immunol., 9, 398, 10.3389/fimmu.2018.00398
Mezrich, 2010, An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells, J. Immunol., 185, 3190, 10.4049/jimmunol.0903670
Fallarino, 2003, T cell apoptosis by kynurenines, Adv. Exp. Med. Biol., 527, 183, 10.1007/978-1-4615-0135-0_21
Curti, 2007, Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells, Blood, 109, 2871, 10.1182/blood-2006-07-036863
Cheng, 2019, S100A9-induced overexpression of PD-1/PD-L1 contributes to ineffective hematopoiesis in myelodysplastic syndromes, Leukemia, 33, 2034, 10.1038/s41375-019-0397-9
Hart, 2011, IL-10 immunomodulation of myeloid cells regulates a murine model of ovarian cancer, Front. Immunol., 2, 29, 10.3389/fimmu.2011.00029
Marvel, 2015, Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected, J. Clin. Invest., 125, 3356, 10.1172/JCI80005
Messmer, 2015, Tumor-induced myeloid dysfunction and its implications for cancer immunotherapy, Cancer Immunol. Immunother., 64, 1, 10.1007/s00262-014-1639-3
Dolcetti, 2010, Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF, Eur. J. Immunol., 40, 22, 10.1002/eji.200939903
Cuenca, 2011, A paradoxical role for myeloid-derived suppressor cells in sepsis and trauma, Mol. Med., 17, 281, 10.2119/molmed.2010.00178
Maenhout, 2014, Enhanced suppressive capacity of tumor-infiltrating myeloid-derived suppressor cells compared with their peripheral counterparts, Int. J. Cancer, 134, 1077, 10.1002/ijc.28449
Mantovani, 2010, The growing diversity and spectrum of action of myeloid-derived suppressor cells, Eur. J. Immunol., 40, 3317, 10.1002/eji.201041170
Meyer, 2014, Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab, Cancer Immunol. Immunother., 63, 247, 10.1007/s00262-013-1508-5
Tarhini, A.A. (2014). Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS ONE, 9.
Weber, 2016, Phase I/II Study of Metastatic Melanoma Patients Treated with Nivolumab Who Had Progressed after Ipilimumab, Cancer Immunol. Res., 4, 345, 10.1158/2326-6066.CIR-15-0193
Groth, 2019, Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression, Br. J. Cancer, 120, 16, 10.1038/s41416-018-0333-1
Suzuki, 2005, Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity, Clin. Cancer Res., 11, 6713, 10.1158/1078-0432.CCR-05-0883
Sevko, 2013, Antitumor effect of paclitaxel is mediated by inhibition of myeloid-derived suppressor cells and chronic inflammation in the spontaneous melanoma model, J. Immunol., 190, 2464, 10.4049/jimmunol.1202781
Eriksson, 2016, Gemcitabine reduces MDSCs, tregs and TGFbeta-1 while restoring the teff/treg ratio in patients with pancreatic cancer, J. Transl. Med., 14, 282, 10.1186/s12967-016-1037-z
Vincent, 2010, 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity, Cancer Res., 70, 3052, 10.1158/0008-5472.CAN-09-3690
Mikyskova, 2012, Cyclophosphamide-induced myeloid-derived suppressor cell population is immunosuppressive but not identical to myeloid-derived suppressor cells induced by growing TC-1 tumors, J. Immunother., 35, 374, 10.1097/CJI.0b013e318255585a
Iida, 2017, Contrasting effects of cyclophosphamide on anti-CTL-associated protein 4 blockade therapy in two mouse tumor models, Cancer Sci., 108, 1974, 10.1111/cas.13337
Ko, 2009, Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients, Clin. Cancer Res., 15, 2148, 10.1158/1078-0432.CCR-08-1332
Kodera, 2011, 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., 13, R66, 10.1186/bcr2903
Qin, 2014, Generation of a new therapeutic peptide that depletes myeloid-derived suppressor cells in tumor-bearing mice, Nat. Med., 20, 676, 10.1038/nm.3560
Robinson, 2003, A chemokine receptor antagonist inhibits experimental breast tumor growth, Cancer Res., 63, 8360
2012, CCR5 antagonist blocks metastasis of basal breast cancer cells, Cancer Res., 72, 3839, 10.1158/0008-5472.CAN-11-3917
Toh, B. (2011). Mesenchymal transition and dissemination of cancer cells is driven by myeloid-derived suppressor cells infiltrating the primary tumor. PLoS Biol., 9.
Zhu, 2017, CXCR2(+) MDSCs promote breast cancer progression by inducing EMT and activated T cell exhaustion, Oncotarget, 8, 114554, 10.18632/oncotarget.23020
2014, Tumour-infiltrating Gr-1+ myeloid cells antagonize senescence in cancer, Nature, 515, 134, 10.1038/nature13638
Sun, L. (2019). Inhibiting myeloid-derived suppressor cell trafficking enhances T cell immunotherapy. JCI Insight, 4.
Holmgaard, 2016, Targeting myeloid-derived suppressor cells with colony stimulating factor-1 receptor blockade can reverse immune resistance to immunotherapy in indoleamine 2,3-dioxygenase-expressing tumors, EBioMedicine, 6, 50, 10.1016/j.ebiom.2016.02.024
Zhu, 2014, CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models, Cancer Res., 74, 5057, 10.1158/0008-5472.CAN-13-3723
Richardsen, 2015, Macrophage-colony stimulating factor (CSF1) predicts breast cancer progression and mortality, AntiCancer Res., 35, 865
Mok, 2014, Inhibition of CSF-1 receptor improves the antitumor efficacy of adoptive cell transfer immunotherapy, Cancer Res., 74, 153, 10.1158/0008-5472.CAN-13-1816
Kumar, 2017, Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors, Cancer Cell, 32, 654, 10.1016/j.ccell.2017.10.005
Elliott, 2017, Human Tumor-Infiltrating Myeloid Cells: Phenotypic and Functional Diversity, Front. Immunol., 8, 86, 10.3389/fimmu.2017.00086
Sinha, 2007, Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells, Cancer Res., 67, 4507, 10.1158/0008-5472.CAN-06-4174
Eruslanov, 2010, Pivotal Advance: Tumor-mediated induction of myeloid-derived suppressor cells and M2-polarized macrophages by altering intracellular PGE(2) catabolism in myeloid cells, J. Leukoc. Biol., 88, 839, 10.1189/jlb.1209821
Ochoa, 2007, Arginase, prostaglandins, and myeloid-derived suppressor cells in renal cell carcinoma, Clin. Cancer Res., 13, 721s, 10.1158/1078-0432.CCR-06-2197
Veltman, 2010, COX-2 inhibition improves immunotherapy and is associated with decreased numbers of myeloid-derived suppressor cells in mesothelioma. Celecoxib influences MDSC function, EBioMedicine, 10, 464
Zelenay, 2015, Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity, Cell, 162, 1257, 10.1016/j.cell.2015.08.015
Serafini, 2006, Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function, J. Exp. Med., 203, 2691, 10.1084/jem.20061104
Lin, 2017, Phosphodiesterase-5 inhibition suppresses colonic inflammation-induced tumorigenesis via blocking the recruitment of MDSC, Am. J. Cancer Res., 7, 41
Tai, 2018, Phosphodiesterase-5 inhibition reduces postoperative metastatic disease by targeting surgery-induced myeloid derived suppressor cell-dependent inhibition of Natural Killer cell cytotoxicity, Oncoimmunology, 7, e1431082, 10.1080/2162402X.2018.1431082
Califano, 2015, Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma, Clin. Cancer Res., 21, 30, 10.1158/1078-0432.CCR-14-1716
Hassel, 2017, Tadalafil has biologic activity in human melanoma. Results of a pilot trial with Tadalafil in patients with metastatic Melanoma (TaMe), Oncoimmunology, 6, e1326440, 10.1080/2162402X.2017.1326440
Weed, 2015, 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., 21, 39, 10.1158/1078-0432.CCR-14-1711
Ohl, 2018, Reactive Oxygen Species as Regulators of MDSC-Mediated Immune Suppression, Front. Immunol., 9, 2499, 10.3389/fimmu.2018.02499
Wang, 2014, Bardoxolone methyl (CDDO-Me) as a therapeutic agent: An update on its pharmacokinetic and pharmacodynamic properties, Drug Des. Devel. Ther., 8, 2075
Hiramoto, 2014, Myeloid lineage-specific deletion of antioxidant system enhances tumor metastasis, Cancer Prev. Res. (Phila), 7, 835, 10.1158/1940-6207.CAPR-14-0094
Nagaraj, 2010, Anti-inflammatory triterpenoid blocks immune suppressive function of MDSCs and improves immune response in cancer, Clin. Cancer Res., 16, 1812, 10.1158/1078-0432.CCR-09-3272
2005, Nitroaspirin corrects immune dysfunction in tumor-bearing hosts and promotes tumor eradication by cancer vaccination, Proc. Natl. Acad. Sci. USA, 102, 4185, 10.1073/pnas.0409783102
Reilley, 2018, STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: Results of a phase 1b trial, J. Immunother. Cancer, 6, 119, 10.1186/s40425-018-0436-5
Kusmartsev, 2008, Reversal of myeloid cell-mediated immunosuppression in patients with metastatic renal cell carcinoma, Clin. Cancer Res., 14, 8270, 10.1158/1078-0432.CCR-08-0165
Mirza, 2006, All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients, Cancer Res., 66, 9299, 10.1158/0008-5472.CAN-06-1690
Iclozan, 2013, 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., 62, 909, 10.1007/s00262-013-1396-8
Nefedova, 2007, Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells, Cancer Res., 67, 11021, 10.1158/0008-5472.CAN-07-2593
Cheng, 2014, Effects of notch signaling on regulation of myeloid cell differentiation in cancer, Cancer Res., 74, 141, 10.1158/0008-5472.CAN-13-1686
Orillion, 2017, 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., 23, 5187, 10.1158/1078-0432.CCR-17-0741
Christmas, 2018, Entinostat Converts Immune-Resistant Breast and Pancreatic Cancers into Checkpoint-Responsive Tumors by Reprogramming Tumor-Infiltrating MDSCs, Cancer Immunol. Res., 6, 1561, 10.1158/2326-6066.CIR-18-0070
Mikyskova, 2014, DNA demethylating agent 5-azacytidine inhibits myeloid-derived suppressor cells induced by tumor growth and cyclophosphamide treatment, J. Leukoc. Biol., 95, 743, 10.1189/jlb.0813435
Kodumudi, 2010, A novel chemoimmunomodulating property of docetaxel: Suppression of myeloid-derived suppressor cells in tumor bearers, Clin. Cancer Res., 16, 4583, 10.1158/1078-0432.CCR-10-0733
Michels, 2012, Paclitaxel promotes differentiation of myeloid-derived suppressor cells into dendritic cells in vitro in a TLR4-independent manner, J. Immunotoxicol., 9, 292, 10.3109/1547691X.2011.642418