AccScience Publishing / TD / Volume 1 / Issue 1 / DOI: 10.36922/td.v1i1.4
Cite this article
155
Download
1612
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
REVIEW

Monocytes in tumor: The perspectives of single-cell analysis

Xin Fu1 Mingzhu Yin1*
Show Less
1 Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
Tumor Discovery 2022, 1(1), 4 https://doi.org/10.36922/td.v1i1.4
Submitted: 2 December 2021 | Accepted: 14 February 2022 | Published: 10 March 2022
© 2022 by the Authors. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Infiltration of mononuclear phagocytes into the tumor microenvironment (TME) is known to orchestrate the tumor progression and is generally considered to interfere with the efficacy of immune checkpoint blockade therapies. For a more precise definition of monocytes, we review some recent advances in the functions of monocytes and macrophages in TME based on single-cell analysis. We also summarize the details of the different subpopulations of monocytes and macrophages involved in immunotherapy and their potential applications in clinical studies. In this review, we briefly introduce the developmental trajectory of mononuclear phagocytes, including monocytes, tumor-associated macrophages, dendritic cells and myeloid-derived suppressive cells, and their functions in TME. In this review, the potential of monocytes and their derived cells as diagnostic and therapeutic targets, with regard to the interaction between monocytes and immune checkpoint therapies, is also summarized.

Keywords
Monocytes
Single-cell RNA sequencing analysis
Tumor microenvironment
Tumor-associated macrophages
Dendritic cells
Funding
The National Natural Science Foundation of China
References
[1]

van Furth R, Cohn ZA, 1968, The origin and kinetics of mononuclear phagocytes. J Exp Med, 128: 415–435.

https://doi.org/10.1084/jem.128.3.415 

[2]

Engblom C, Pfirschke C, Pittet MJ, 2016, The role of myeloid cells in cancer therapies. Nat Rev Cancer, 16: 447–462.

https://doi.org/10.1038/nrc.2016.54

[3]

Pollard JW, 2004, Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer, 4: 71–78.

https://doi.org/10.1038/nrc1256

[4]

Sica A, Schioppa T, Mantovani A, et al., 2006, Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: Potential targets of anti-cancer therapy. Eur J Cancer, 42: 717–727.

https://doi.org/10.1016/j.ejca.2006.01.003

[5]

Yunna C, Mengru H, Lei W, et al., 2020. Macrophage M1/ M2 polarization. Eur J Pharmacol, 877: 173090.

https://doi.org/10.1016/j.ejphar.2020.173090

[6]

Krieg C, Nowicka M, Guglietta S, et al., 2018, High-dimensional Single-cell Analysis Predicts Response to Anti-PD-1 Immunotherapy. Nat Med, 24: 144–153.

https://doi.org/10.1038/nm.4466

[7]

Roberts SA, Waziri AE, Agrawal N, 2016, Development of a single-cell migration and extravasation platform through selective surface modification. Anal Chem, 88:2770–2776.

https://doi.org/10.1021/acs.analchem.5b04391

[8]

Chen YC, Allen SG, Ingram PN, et al., 2015, Single-cell migration chip for chemotaxis-based microfluidic selection of heterogeneous cell populations. Scientific reports, 5: 9980.

[9]

Zhuang J, Wu Y, Chen L, et al., 2018, Single-cell mobility analysis of metastatic breast cancer cells. Advanced Science (Weinheim, Baden-Wurttemberg, Germany), 5: 1801158.

https://doi.org/10.1002/advs.201801158

[10]

Trapnell C, 2015, Defining cell types and states with single-cell genomics. Genome Res, 25: 1491–1498.

[11]

Grün D, van Oudenaarden A, 2015, Design and analysis of single-cell sequencing experiments. Cell, 163: 799–810.

https://doi.org/10.1016/j.cell.2015.10.039

[12]

Topalian SL, Hodi FS, Brahmer JR, et al., 2012, Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med, 366: 2443–2454.

https://doi.org/10.1056/NEJMoa1200690

[13]

Powles T, Eder JP, Fine GD, et al., 2014, MPDL3280A (anti- PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature, 515: 558–562.

https://doi.org/10.1038/nature13904

[14]

Rizvi NA, Hellmann MD, Snyder A, et al., 2015, Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science (New York), 348: 124–128.

https://doi.org/10.1126/science.aaa1348 

[15]

Ansell SM, Lesokhin AM, Borrello I, et al., 2015, PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med, 372(4): 311–319.

[16]

Robert C, Schachter J, Long GV, et al., 2015, Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med, 372(26): 2521–2532.

https://doi.org/10.1056/NEJMoa1503093

[17]

Ostrand-Rosenberg S, Sinha P, 2009, Myeloid-derived suppressor cells: Linking inflammation and cancer. J Immunol (Baltimore, Md. 1950), 182(8): 4499–4506.

https://doi.org/10.4049/jimmunol.0802740

[18]

Auffray C, Sieweke MH, Geissmann F, 2009, Blood monocytes: Development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol, 27: 669–692.

https://doi.org/10.1146/annurev.immunol.021908.132557

[19]

Shi C, Pamer EG, 2011, Monocyte recruitment during infection and inflammation. Nat Rev Immunol, 11(11): 762–774.

https://doi.org/10.1038/nri3070

[20]

Ziegler-Heitbrock L, Ancuta P, Crowe S, et al., 2010, Nomenclature of monocytes and dendritic cells in blood. Blood, 116(16): e74–e80.

https://doi.org/10.1182/blood-2010-02-258558

[21]

Auffray C, Fogg D, Garfa M, et al., 2007, Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science (New York), 317(5838): 666–670.

https://doi.org/10.1126/science.1142883

[22]

Movahedi K, Laoui D, Gysemans C, et al., 2010, Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res, 70(14): 5728–5739.

https://doi.org/10.1158/0008-5472.CAN-09-4672

[23]

Mildner A, Schönheit J, Giladi A, et al., 2017, Genomic characterization of murine monocytes reveals C/EBPβ transcription factor dependence of Ly6C cells. Immunity, 46(5): 849–862.e7.

https://doi.org/10.1016/j.immuni.2017.04.018

[24]

van Galen P, Hovestadt V, Wadsworth Ii MH, et al., 2019, Single-cell RNA-Seq reveals AML hierarchies relevant to disease progression and immunity. Cell, 176(6): 1265–1281.e1224.

https://doi.org/10.1016/j.cell.2019.01.031

[25]

Zilionis R, Engblom C, Pfirschke C, et al., 2019, Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity, 50(5): 1317–1334.e1310.

https://doi.org/10.1016/j.immuni.2019.03.009

[26]

Puram SV, Tirosh I, Parikh AS, et al., 2017, Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell, 171(7): 1611–1624.e24.

https://doi.org/10.1016/j.cell.2017.10.044

[27]

Passlick B, Flieger D, Ziegler-Heitbrock HW, 1989, Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood, 74(7): 2527–2534.

[28]

Olingy CE, Dinh HQ, Hedrick CC, 2019, Monocyte heterogeneity and functions in cancer. J Leukocyte Biol, 106(2): 309–322.

https://doi.org/10.1002/JLB.4RI0818-311R

[29]

Villani AC, Satija R, Reynolds G, et al., 2017, Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science (New York), 356(6335): eaah4573.

https://doi.org/10.1126/science.aah4573

[30]

Qian BZ, Li J, Zhang H, et al., 2011, CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature, 475(7355): 222–225.

https://doi.org/10.1038/nature10138

[31]

Schmall A, Al-Tamari HM, Herold S, et al., 2015, Macrophage and cancer cell cross-talk via CCR2 and CX3CR1 is a fundamental mechanism driving lung cancer. Am J Respir Crit Care Med., 191(4): 437–447.

https://doi.org/10.1164/rccm.201406-1137OC

[32]

Li X, Yao W, Yuan Y, et al., 2017, Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut, 66(1): 157–167.

https://doi.org/10.1136/gutjnl-2015-310514

[33]

Gordon IO, Freedman RS, 2006, Defective antitumor function of monocyte-derived macrophages from epithelial ovarian cancer patients. Clin Cancer Res, 12(5): 1515–1524.

https://doi.org/10.1158/1078-0432.CCR-05-2254

[34]

Sheng J, Chen Q, Soncin I, et al., 2017, A discrete subset of monocyte-derived cells among typical conventional Type 2 dendritic cells can efficiently cross-present. Cell Rep, 21(5): 1203–1214.

https://doi.org/10.1016/j.celrep.2017.10.024

[35]

Hanna RN, Cekic C, Sag D, et al., 2015, Patrolling monocytes control tumor metastasis to the lung. Science (New York), 350(6263): 985–990.

https://doi.org/10.1126/science.aac9407

[36]

Romano E, Kusio-Kobialka M, Foukas PG, et al., 2015, Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A, 112(19): 6140–6145.

https://doi.org/10.1073/pnas.1417320112

[37]

De Palma M, Venneri MA, Galli R, et al., 2005, Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell, 8(3): 211–226.

https://doi.org/10.1016/j.ccr.2005.08.002

[38]

Venneri MA, De Palma M, Ponzoni M, et al., 2007, Identification of proangiogenic TIE2–expressing monocytes (TEMs) in human peripheral blood and cancer. Blood, 109(12): 5276–5285.

https://doi.org/10.1182/blood-2006-10-053504

[39]

Duan Z, Luo Y, 2021, Targeting macrophages in cancer immunotherapy. Signal Transduct Target Ther, 6(1): 127.

https://doi.org/10.1038/s41392-021-00506-6

[40]

Wynn TA, Chawla A, Pollard JW, 2013, Macrophage biology in development, homeostasis and disease. Nature, 496(7446): 445–455.

https://doi.org/10.1038/nature12034

[41]

Ginhoux F, Jung S, 2014, Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol, 14(6): 392–404.

https://doi.org/10.1038/nri3671

[42]

Fogg DK, Sibon C, Miled C, et al., 2006, A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science (New York), 311(5757): 83–87.

https://doi.org/10.1126/science.1117729

[43]

Lee J, Breton G, Oliveira TY, et al., 2015, Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow. J Exp Med, 212(3): 385–399.

https://doi.org/10.1084/jem.20141442

[44]

Menezes S, Melandri D, Anselmi G, et al., 2016, The heterogeneity of Ly6C monocytes controls their differentiation into iNOS macrophages or monocyte-derived dendritic cells. Immunity, 45(6): 1205–1218.

https://doi.org/10.1016/j.immuni.2016.12.001

[45]

Gabrilovich DI, Bronte V, Chen SH, et al., 2007, The terminology issue for myeloid–derived suppressor cells. Cancer Res, 67(1): 425; author reply 426.

https://doi.org/10.1158/0008-5472.CAN-06-3037

[46]

Veglia F, Perego M, Gabrilovich D, 2018, Myeloid-derived suppressor cells coming of age. Nat Immunol, 19(2): 108–119.

https://doi.org/10.1038/s41590-017-0022-x

[47]

Filipazzi P, Valenti R, Huber V, et al., 2007, Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol, 25(18): 2546–2553.

https://doi.org/10.1200/JCO.2006.08.5829

[48]

Zea AH, Rodriguez PC, Atkins MB, et al., 2005, Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: A mechanism of tumor evasion. Cancer Res, 65(8): 3044–3048.

https://doi.org/10.1158/0008-5472.CAN-04-4505

[49]

Srivastava MK, Bosch JJ, Thompson JA, et al., 2008, Lung cancer patients’ CD4(+) T cells are activated in vitro by MHC II cell-based vaccines despite the presence of myeloid-derived suppressor cells. Cancer Immunol Immunother, 57(10): 1493–1504.

https://doi.org/10.1007/s00262-008-0490-9

[50]

Hoechst B, Ormandy LA, Ballmaier M, et al., 2008, A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+) Foxp3(+) T cells. Gastroenterology, 135(1): 234–243.

https://doi.org/10.1053/j.gastro.2008.03.020

[51]

Canè S, Ugel S, Trovato R, et al., 2019, The endless saga of monocyte diversity. Front Immunol, 10: 1786.

https://doi.org/10.3389/fimmu.2019.01786

[52]

Cassetta L, Bruderek K, Skrzeczynska-Moncznik J, et al., 2020, Differential expansion of circulating human MDSC subsets in patients with cancer, infection and inflammation. J Immunother Cancer, 8(2): e001223.

https://doi.org/10.1136/jitc-2020-001223

[53]

Hartwig T, Montinaro A, von Karstedt S, et al., 2017, The TRAIL-induced cancer secretome promotes a tumor- supportive immune microenvironment via CCR2. Mol Cell, 65(4): 730–742.e5.

https://doi.org/10.1016/j.molcel.2017.01.021

[54]

Hettinger J, Richards DM, Hansson J, et al., 2013, Origin of monocytes and macrophages in a committed progenitor. Nat Immunol, 14(8): 821–830.

https://doi.org/10.1038/ni.2638

[55]

Kawamura S, Onai N, Miya F, et al., 2017, Identification of a human clonogenic progenitor with strict monocyte differentiation potential: A counterpart of mouse cMoPs. Immunity, 46(5): 835–848.e4.

https://doi.org/10.1016/j.immuni.2017.04.019

[56]

Drissen R, Buza-Vidas N, Woll P, et al., 2016, Distinct myeloid progenitor-differentiation pathways identified through single-cell RNA sequencing. Nat Immunol, 17(6): 666–676.

https://doi.org/10.1038/ni.3412

[57]

Sasmono RT, Oceandy D, Pollard JW, et al., 2003, A macrophage colony-stimulating factor receptor-green fluorescent protein transgene is expressed throughout the mononuclear phagocyte system of the mouse. Blood, 101(3): 1155–1163.

https://doi.org/10.1182/blood-2002-02-0569

[58]

Van Dyken SJ, Locksley RM, 2013, Interleukin-4- and interleukin-13-mediated alternatively activated macrophages: Roles in homeostasis and disease. Annu Rev Immunol, 31: 317–343.

https://doi.org/10.1146/annurev-immunol-032712-095906

[59]

Nirschl CJ, Suárez-Fariñas M, Izar B, et al., 2017, IFNγ- dependent tissue-immune homeostasis is co-opted in the tumor microenvironment. Cell, 170(1): 127–141.e115.

https://doi.org/10.1016/j.cell.2017.06.016

[60]

Long AH, Highfill SL, Cui Y, et al., 2016, Reduction of MDSCs with all-trans retinoic acid improves CAR therapy efficacy for sarcomas. Cancer Immunol Res, 4(10): 869–880.

https://doi.org/10.1158/2326-6066.CIR-15-0230

[61]

Michielon E, González ML, Burm JL, et al., Micro-environmental cross-talk in an organotypic human melanoma-in-skin model directs M2-like monocyte differentiation via IL-10. Cancer Immunol Immunother, 69(11): 2319–2331.

https://doi.org/10.1007/s00262-020-02626-4

[62]

Sander J, Schmidt SV, Cirovic B, et al., 2017, Cellular differentiation of human monocytes is regulated by time-dependent interleukin-4 signaling and the transcriptional regulator NCOR2. Immunity, 47(6): 1051–1066.e12.

https://doi.org/10.1016/j.immuni.2017.11.024 

[63]

Shen CK, Huang BR, Yeh WL, et al., 2021, Regulatory effects of IL-1β in the interaction of GBM and tumor-associated monocyte through VCAM-1 and ICAM-1. Eur J Pharmacol, 905: 174216.

https://doi.org/10.1016/j.ejphar.2021.174216

[64]

Franklin RA, Liao W, Sarkar A, et al., 2014, The cellular and molecular origin of tumor-associated macrophages. Science (New York), 344(6186): 921–925.

https://doi.org/10.1126/science.1252510

[65]

Jenkins SJ, Ruckerl D, Cook PC, et al., 2011, Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science (New York), 332(6035): 1284–1288.

https://doi.org/10.1126/science.1204351

[66]

Orecchioni M, Ghosheh Y, Pramod AB, et al., 2019, Macrophage polarization: Different gene signatures in M1(LPS+) vs. classically and M2(LPS–) vs. alternatively activated macrophages. Front Immunol, 10: 1084.

https://doi.org/10.3389/fimmu.2019.01084

[67]

Okabe Y, Medzhitov R, 2014, Tissue-specific signals control reversible program of localization and functional polarization of macrophages. Cell, 157(4): 832–844.

https://doi.org/10.1016/j.cell.2014.04.016 

[68]

Devalaraja S, To TK, Folkert IW, et al., 2020, Tumor-derived retinoic acid regulates intratumoral monocyte differentiation to promote immune suppression. Cell, 180(6): 1098–1114.e16.

https://doi.org/10.1016/j.cell.2020.02.042

[69]

Chun E, Lavoie S, Michaud M, et al., 2015, CCL2 promotes colorectal carcinogenesis by enhancing polymorphonuclear myeloid-derived suppressor cell population and function. Cell Rep, 12(2): 244–257.

https://doi.org/10.1016/j.celrep.2015.06.024

[70]

Zhang J, Patel L, Pienta KJ. CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis. Cytokine Growth Factor Rev, 21(1): 41–48.

https://doi.org/10.1016/j.cytogfr.2009.11.009

[71]

Ma RY, Zhang H, Li XF, et al., 2020, Monocyte-derived macrophages promote breast cancer bone metastasis outgrowth. J Exp Med, 217(11): e20191820.

https://doi.org/10.1084/jem.20191820

[72]

Cortez-Retamozo V, Etzrodt M, Newton A, et al., 2012, Origins of tumor-associated macrophages and neutrophils. Proc Natl Acad Sci U S A, 109(7): 2491–2496.

https://doi.org/10.1073/pnas.1113744109

[73]

Yeap WH, Wong KL, Shimasaki N, et al., 2016, CD16 is indispensable for antibody-dependent cellular cytotoxicity by human monocytes. Sci Rep, 6: 34310.

https://doi.org/10.1038/srep34310

[74]

Kusmartsev S, Nefedova Y, Yoder D, et al., 2004, Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol (Baltimore, Md. 1950), 172(2): 989–999.

https://doi.org/10.4049/jimmunol.172.2.989

[75]

Bronte V, Serafini P, Mazzoni A, et al., 2003, L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol, 24(6): 302–306.

https://doi.org/10.1016/s1471-4906(03)00132-7

[76]

Baniyash M, 2004, TCR zeta-chain downregulation: Curtailing an excessive inflammatory immune response. Nat Rev Immunol, 4(9): 675–687.

https://doi.org/10.1038/nri1434

[77]

Jakubzick CV, Randolph GJ, Henson PM, 2017, Monocyte differentiation and antigen-presenting functions. Nat Rev Immunol, 17(6): 349–362.

https://doi.org/10.1038/nri.2017.28

[78]

Ma Y, Adjemian S, Mattarollo SR, et al., 2013, Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity, 38(4): 729–741.

https://doi.org/10.1038/nri.2017.28

[79]

Kuhn S, Yang J, Ronchese F, 2015, Monocyte-derived dendritic cells are essential for CD8(+) T cell activation and antitumor responses after local immunotherapy. Front Immunol, 6: 584.

https://doi.org/10.3389/fimmu.2015.00584

[80]

Zhu C, Kros JM, Cheng C, et al., 2017, The contribution of tumor-associated macrophages in glioma neo-angiogenesis and implications for anti-angiogenic strategies. Neuro Oncol, 19(11): 1435–1446.

https://doi.org/10.1093/neuonc/nox081

[81]

Chittezhath M, Dhillon MK, Lim JY, et al., 2014, Molecular profiling reveals a tumor-promoting phenotype of monocytes and macrophages in human cancer progression. Immunity, 41(5): 815–829.

https://doi.org/10.1016/j.immuni.2014.09.014

[82]

De Palma M, Venneri MA, Roca C, et al, 2003, Targeting exogenous genes to tumor angiogenesis by transplantation of genetically modified hematopoietic stem cells. Nat Med, 9(6): 789–795.

https://doi.org/10.1038/nm871

[83]

Shojaei F, Wu X, Malik AK, et al., 2007, Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol, 25(8): 911–920.

https://doi.org/10.1038/nbt1323

[84]

Kaplan RN, Riba RD, Zacharoulis S, et al., 2005, VEGFR1- positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature, 438(7069): 820–827.

https://doi.org/10.1038/nature04186

[85]

Matsubara T, Kanto T, Kuroda S, et al., 2013, TIE2-expressing monocytes as a diagnostic marker for hepatocellular carcinoma correlates with angiogenesis. Hepatology, 57(4): 1416–1425.

https://doi.org/10.1002/hep.25965

[86]

Harney AS, Arwert EN, Entenberg D, et al., 2015, Real-time imaging reveals local, transient vascular permeability, and tumor cell intravasation stimulated by TIE2hi macrophage-derived VEGFA. Cancer Discov, 5(9): 932–943.

https://doi.org/10.1158/2159-8290.CD-15-0012

[87]

Lu P, Weaver VM, Werb Z, 2012, The extracellular matrix: A dynamic niche in cancer progression. J Cell Biol, 196(4): 395–406.

https://doi.org/10.1083/jcb.201102147

[88]

Naba A, Clauser KR, Hoersch S, et al., 2012, The matrisome: In silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices. Mol Cell Proteomics, 11(4): M111.014647.

https://doi.org/10.1074/mcp.M111.014647

[89]

Kessenbrock K, Plaks V, Werb Z, 2010, Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell, 141(1): 52–67.

https://doi.org/10.1016/j.cell.2010.03.015

[90]

Mason SD, Joyce JA, 2011, Proteolytic networks in cancer. Trends Cell Biol, 21(4): 228–237.

https://doi.org/10.1016/j.tcb.2010.12.002

[91]

Afik R, Zigmond E, Vugman M, et al., 2016, Tumor macrophages are pivotal constructors of tumor collagenous matrix. J Exp Med, 213(11): 2315–2331.

https://doi.org/10.1084/jem.20151193

[92]

Madsen DH, Jürgensen HJ, Siersbæk MS, et al., 2017, Tumor-associated macrophages derived from circulating inflammatory monocytes degrade collagen through cellular uptake. Cell Rep, 21(13): 3662–3671.

https://doi.org/10.1016/j.celrep.2017.12.011

[93]

Madsen DH, Leonard D, Masedunskas A, et al., 2013, M2-like macrophages are responsible for collagen degradation through a mannose receptor-mediated pathway. J Cell Biol, 202(6): 951–966.

https://doi.org/10.1083/jcb.201301081

[94]

Guilliams M, Ginhoux F, Jakubzick C, et al., 2014, Dendritic cells, monocytes and macrophages: A unified nomenclature based on ontogeny. Nat Rev Immunol, 14(8): 571–578.

https://doi.org/10.1038/nri3712

[95]

DeNardo DG, Ruffell B, 2019, Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol, 19(6): 369–382.

https://doi.org/10.1038/s41577-019-0127-6

[96]

Zhu Y, Herndon JM, Sojka DK, et al., 2017, Tissue-resident macrophages in pancreatic ductal adenocarcinoma originate from embryonic hematopoiesis and promote tumor progression. Immunity, 47(2): 323–338.e6.

https://doi.org/10.1016/j.immuni.2017.07.014

[97]

Sica A, Bronte V, 2007, Altered macrophage differentiation and immune dysfunction in tumor development. J Clin Investig, 117(5): 1155–1166.

https://doi.org/10.1172/JCI31422

[98]

Yin M, Li X, Tan S, et al., 2016, Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. J Clin Investig, 126(11): 4157–4173.

https://doi.org/10.1172/JCI87252

[99]

Richards DM, Hettinger J, Feuerer M, 2013, Monocytes and macrophages in cancer: development and functions. Cancer Microenviron, 6(2): 179–191.

https://doi.org/10.1007/s12307-012-0123-x

[100]

Martinez FO, Helming L, Gordon S, 2009, Alternative activation of macrophages: An immunologic functional perspective. Annu Rev Immunol, 27: 451–483.

https://doi.org/10.1146/annurev.immunol.021908.132532

[101]

Mantovani A, Sozzani S, Locati M, et al., 2002, Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol, 23(11): 549–555.

https://doi.org/10.1016/s1471-4906(02)02302-5

[102]

Minopoli M, Sarno S, Di Carluccio G, et al., 2020, Inhibiting monocyte recruitment to prevent the pro-tumoral activity of tumor-associated macrophages in chondrosarcoma. Cells, 9(4): 1062.

https://doi.org/10.3390/cells9041062 

[103]

Murray PJ, Allen JE, Biswas SK, et al., 2014, Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity, 41(1): 14–20.

https://doi.org/10.1016/j.immuni.2014.06.008

[104]

Azizi E, Carr AJ, Plitas G, et al., 2018, Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell, 174()5: 1293–1308.e36.

https://doi.org/10.1016/j.cell.2018.05.060

[105]

Vitale I, Shema E, Loi S, et al., 2021, Intratumoral heterogeneity in cancer progression and response to immunotherapy. Nat Med, 27(2): 212–224.

https://doi.org/10.1038/s41591-021-01233-9

[106]

Singhal S, Stadanlick J, Annunziata MJ, et al., 2019, Human tumor-associated monocytes/macrophages and their regulation of T cell responses in early-stage lung cancer. Sci Transl Med, 11(479): eaat1500.

https://doi.org/10.1126/scitranslmed.aat1500

[107]

Laviron M, Boissonnas A, 2019, Ontogeny of tumor-associated macrophages. Front Immunol, 10: 1799.

[108]

Müller S, Kohanbash G, Liu SJ, et al., 2017, Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome Biol, 18: 234.

[109]

Bowman RL, Klemm F, Akkari L, et al., 2016, Macrophage ontogeny underlies differences in tumor-specific education in brain malignancies. Cell Rep, 17(9): 2445–2459.

https://doi.org/10.1016/j.celrep.2016.10.052

[110]

Balkwill F, Mantovani A, 2001, Inflammation and cancer: Back to virchow? Lancet, 357(9255): 539–545.

https://doi.org/10.1016/S0140-6736(00)04046-0

[111]

Kumar V, Patel S, Tcyganov E, et al., 2016, The nature of Myeloid-Derived suppressor cells in the tumor microenvironment. Trends Immunol, 37(3): 208–220.

https://doi.org/10.1016/j.it.2016.01.004

[112]

Kumar V, Cheng P, Condamine T, et al., 2016, CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity, 44(2): 303–315.

https://doi.org/10.1016/j.immuni.2016.01.014

[113]

Laoui D, Keirsse J, Morias Y, et al., 2016, The tumour microenvironment harbours ontogenically distinct dendritic cell populations with opposing effects on tumour immunity. Nat Commun, 7: 13720.

https://doi.org/10.1038/ncomms13720

[114]

Veglia F, Gabrilovich DI, 2017, Dendritic cells in cancer: The role revisited. Curr Opin Immunol, 45: 43–51.

https://doi.org/10.1016/j.coi.2017.01.002

[115]

Broz ML, Binnewies M, Boldajipour B, et al., 2014, Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell, 26(5): 638–652.

https://doi.org/10.1016/j.ccell.2014.09.007

[116]

He YF, Wang CQ, Yu Y, et al., 2015, Tie2-expressing monocytes are associated with identification and prognoses of hepatitis B virus related hepatocellular carcinoma after resection. PLoS One, 10(11): e0143657.

https://doi.org/10.1371/journal.pone.0143657

[117]

Awad RM, De Vlaeminck Y, Maebe J, et al., 2018, Turn back the TIMe: Targeting tumor infiltrating myeloid cells to revert cancer progression. Front Immunol, 9: 1977.

https://doi.org/10.3389/fimmu.2018.01977

[118]

Nywening TM, Wang-Gillam A, Sanford DE, et al., 2016, Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: A single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol, 17(5): 651–662.

https://doi.org/10.1016/S1470-2045(16)00078-4

[119]

Bonapace L, Coissieux MM, Wyckoff J, et al., 2014, Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature, 515(7525): 130–133.

https://doi.org/10.1038/nature13862

[120]

Steggerda SM, Bennett MK, Chen J, et al., 2017, Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J Immunother. Cancer, 5(1): 101.

https://doi.org/10.1186/s40425-017-0308-4

[121]

Nefedova Y, Fishman M, Sherman S, et al., 2007, Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells. Cancer Res, 67(22): 11021–11028.

https://doi.org/10.1158/0008-5472.CAN-07-2593

[122]

Vincent J, Mignot G, Chalmin F, et al, 2010, 5-fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res, 70(8): 3052–3061.

https://doi.org/10.1158/0008-5472.CAN-09-3690

[123]

Dempke WC, Fenchel K, Uciechowski P, et al., 2017, Second-and third-generation drugs for immuno-oncology treatment-the more the better? Eur J Cancer, 74: 55–72.

https://doi.org/10.1016/j.ejca.2017.01.001

[124]

Mellman I, Coukos G, Dranoff G, 2011, Cancer immunotherapy comes of age. Nature, 480(7378): 480–489.

https://doi.org/10.1038/nature10673

[125]

Drake CG, Jaffee E, Pardoll DM, 2006, Mechanisms of immune evasion by tumors. Adv Immunol, 90: 51–81.

https://doi.org/10.1016/S0065-2776(06)90002-9

[126]

van der Leun AM, Thommen DS, Schumacher TN, 2020, CD8 T cell states in human cancer: Insights from single-cell analysis. Nat Rev Cancer, 20(4): 218–232.

https://doi.org/10.1038/s41568-019-0235-4

[127]

Ribas A, Wolchok JD, 2018, Cancer immunotherapy using checkpoint blockade. Science (New York), 359(6382): 1350– 1355.

https://doi.org/10.1126/science.aar4060

[128]

Keir ME, Butte MJ, Freeman GJ, et al., 2008, PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol, 26: 677–704.

https://doi.org/10.1146/annurev.immunol.26.021607.090331

[129]

Topalian SL, Drake CG, Pardoll DM, 2015, Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell, 27(4): 450–461.

https://doi.org/10.1016/j.ccell.2015.03.001

[130]

Schetters ST, Rodriguez E, Kruijssen LJ, et al., 2020, Monocyte-derived APCs are central to the response of PD1 checkpoint blockade and provide a therapeutic target for combination therapy. J Immunother Cancer, 8(2): e000588.

https://doi.org/10.1136/jitc-2020-000588

[131]

Hugo W, Zaretsky JM, Sun L, et al., 2016, Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell, 165(1): 35–44.

https://doi.org/10.1016/j.cell.2016.02.065

[132]

Hamid O, Robert C, Daud A, et al., 2013, Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med, 369(2): 134–144.

https://doi.org/10.1056/NEJMoa1305133

[133]

Das S, Camphausen K, Shankavaram U, 2020, Cancer-specific immune prognostic signature in solid tumors and its relation to immune checkpoint therapies. Cancers, 12(9): 2476.

https://doi.org/10.3390/cancers12092476

[134]

Marcovecchio PM, Thomas G, Salek-Ardakani S, 2021, CXCL9-expressing tumor-associated macrophages: New players in the fight against cancer. J Immunother Cancer, 9(2): e002045.

https://doi.org/10.1136/jitc-2020-002045

[135]

Huber V, Di Guardo L, Lalli L, et al., 2021, Back to simplicity: A four-marker blood cell score to quantify prognostically relevant myeloid cells in melanoma patients. J Immunother Cancer, 9(2): e001167.

https://doi.org/10.1136/jitc-2020-001167

[136]

Gordon SR, Maute RL, Dulken BW, et al., 2017, PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature, 545(7655): 495–499.

https://doi.org/10.1038/nature22396

[137]

Zou W, Wolchok JD, Chen L, 2016, PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med, 8(328): 328–324.

https://doi.org/10.1126/scitranslmed.aad7118

[138]

Liu M, Zhou J, Liu X, et al., 2020, Targeting monocyte-intrinsic enhancer reprogramming improves immunotherapy efficacy in hepatocellular carcinoma. Gut, 69(2): 365–379.

https://doi.org/10.1136/gutjnl-2018-317257

[139]

Escobar G, Barbarossa L, Barbiera G, et al., 2018, Interferon gene therapy reprograms the leukemia microenvironment inducing protective immunity to multiple tumor antigens. Nat Commun, 9: 2896.

[140]

Gubin MM, Esaulova E, Ward JP, et al., 2018, High-dimensional analysis delineates myeloid and lymphoid compartment remodeling during successful immune-checkpoint cancer therapy. Cell, 175(4): 1443.

https://doi.org/10.1016/j.cell.2018.09.030

[141]

Yin M, Guo Y, Hu R, et al., 2020, Potent BRD4 inhibitor suppresses cancer cell-macrophage interaction. Nat Commun, 11(1): 1833.

https://doi.org/10.1038/s41467-020-15290-0

[142]

June CH, Sadelain M, 2018, Chimeric antigen receptor therapy. N Engl J Med, 379(1): 64–73.

https://doi.org/10.1056/NEJMra1706169

[143]

Green DS, Nunes AT, David-Ocampo V, et al., 2018, A phase 1 trial of autologous monocytes stimulated ex vivo with sylatron (peginterferon alfa-2b) and actimmune (interferon gamma-1b) for intra-peritoneal administration in recurrent ovarian cancer. J Transl Med, 16(1): 196.

https://doi.org/10.1186/s12967-018-1569-5

[144]

Liu Y, Cao X, 2015, The origin and function of tumor-associated macrophages. Cell Mol Immunol, 12: 1–4.

https://doi.org/10.1038/cmi.2014.83

[145]

Noy R, Pollard JW, 2014, Tumor-associated macrophages: From mechanisms to therapy. Immunity, 41(1): 49–61.

https://doi.org/10.1016/j.immuni.2014.06.010

Conflict of interest
The authors declare no conflict of interest.
Share
Back to top
Tumor Discovery, Electronic ISSN: 2810-9775 Published by AccScience Publishing