- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, P. R. China;
Lung cancer has a high morbidity and mortality, and invasion is one of the major factors that cause recurrence and death in lung cancer patients. Tumor-associated macrophages (TAMs) are cells that have the potential to secrete cytokines, growth hormones, inflammatory substrates, and protein hydrolases, which are associated with the growth, invasion and metastasis of tumors. In this article, we will explore the various chemicals that are manufactured to promote the invasion of lung cancer, as well as the numerous clinical therapeutic features that TAMs possess in the treatment of lung cancer. In addition, we look at the possibility that TAMs might be beneficial in the treatment of lung cancer. We have an innovative investigation of the huge variety of complex substances generated by TAMs, with the goal of determining whether or not the molecules under investigation have the potential to serve as new therapeutic targets. Throughout the whole of the presentation, a significant focus is placed on doing in-depth research to ascertain whether TAMs have the capability to reinforce as viable carriers for unique and creative medications. This not only provides novel concepts for the creation of new targeted therapies but also leads to the development of brand-new, cutting-edge methods for the manufacture of individualized medicines and drug carriers.
Citation: REN Qinglin, WU Jun, CHEN Yong, SHU Yusheng. Research advances of tumor-associated macrophages in lung cancer invasion and treatment. Chinese Journal of Clinical Thoracic and Cardiovascular Surgery, 2023, 30(12): 1773-1778. doi: 10.7507/1007-4848.202205048 Copy
1. | Xia C, Dong X, Li H, et al. Cancer statistics in China and United States, 2022: Profiles, trends, and determinants. Chin Med J, 2022, 135(5): 584-590. |
2. | Pan Y, Yu Y, Wang X, et al. Tumor-associated macrophages in tumor immunity. Front Immunol, 2020, 11: 583084. |
3. | Li H, Huang N, Zhu W, et al. Modulation the crosstalk between tumor-associated macrophages and non-small cell lung cancer to inhibit tumor migration and invasion by ginsenoside Rh2. BMC Cancer, 2018, 18(1): 579. |
4. | Sarode P, Zheng X, Giotopoulou GA, et al. Reprogramming of tumor-associated macrophages by targeting β-catenin/FOSL2/ARID5A signaling: A potential treatment of lung cancer. Sci Adv, 2020, 6(23): eaaz6105. |
5. | Chávez-Galán L, Olleros ML, Vesin D, et al. Much more than M1 and M2 macrophages, there are also CD169(+) and TCR(+) macrophages. Front Immunol, 2015, 6: 263. |
6. | Casanova-Acebes M, Dalla E, Leader AM, et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature, 2021, 595(7868): 578-584. |
7. | Tzetzo SL, Abrams SI. Redirecting macrophage function to sustain their "defender" antitumor activity. Cancer Cell, 2021, 39(6): 734-737. |
8. | Han Y, Guo W, Ren T, et al. Tumor-associated macrophages promote lung metastasis and induce epithelial-mesenchymal transition in osteosarcoma by activating the COX-2/STAT3 axis. Cancer Lett, 2019, 440-441: 116-125. |
9. | Cassetta L, Pollard JW. Tumor-associated macrophages. Curr Biol, 2020, 30(6): R246-R248. |
10. | Chen Y, Song Y, Du W, et al. Tumor-associated macrophages: An accomplice in solid tumor progression. J Biomed Sci, 2019, 26(1): 78. |
11. | Xue J, Schmidt SV, Sander J, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity, 2014, 40(2): 274-288. |
12. | Xu F, Wei Y, Tang Z, et al. Tumor-associated macrophages in lung cancer: Friend or foe? Mol Med Rep, 2020, 22(5): 4107-4115. |
13. | Cotechini T, Atallah A, Grossman A. Tissue-resident and recruited macrophages in primary tumor and metastatic microenvironments: Potential targets in cancer therapy. Cells, 2021, 10(4): 960. |
14. | Zou G, Zhang X, Wang L, et al. Herb-sourced emodin inhibits angiogenesis of breast cancer by targeting VEGFA transcription. Theranostics, 2020, 10(15): 6839-6853. |
15. | Fu LQ, Du WL, Cai MH, et al. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol, 2020, 353: 104119. |
16. | Martin JD, Seano G, Jain RK. Normalizing function of tumor vessels: Progress, opportunities, and challenges. Annu Rev Physiol, 2019, 81: 505-534. |
17. | Dallavalasa S, Beeraka NM, Basavaraju CG, et al. The role of tumor associated macrophages (TAMs) in cancer progression, chemoresistance, angiogenesis and metastasis—Current status. Curr Med Chem, 2021, 28(39): 8203-8236. |
18. | Jeon SH, Chae BC, Kim HA, et al. Mechanisms underlying TGF-beta1-induced expression of VEGF and Flk-1 in mouse macrophages and their implications for angiogenesis. J Leukoc Biol, 2007, 81(2): 557-566. |
19. | Frezzetti D, Gallo M, Maiello MR, et al. VEGF as a potential target in lung cancer. Expert Opin Ther Targets, 2017, 21(10): 959-966. |
20. | Nilsson MB, Robichaux J, Herynk MH, et al. Altered regulation of HIF-1α in naive- and drug-resistant EGFR-mutant NSCLC: Implications for a vascular endothelial growth factor-dependent phenotype. J Thorac Oncol, 2021, 16(3): 439-451. |
21. | Spagnuolo A, Palazzolo G, Sementa C, et al. Vascular endothelial growth factor receptor tyrosine kinase inhibitors for the treatment of advanced non-small cell lung cancer. Expert Opin Pharmacother, 2020, 21(4): 491-506. |
22. | Shen G, Zheng F, Ren D, et al. Anlotinib: A novel multi-targeting tyrosine kinase inhibitor in clinical development. J Hematol Oncol, 2018, 11(1): 120. |
23. | Winer A, Adams S, Mignatti P. Matrix metalloproteinase inhibitors in cancer therapy: Turning past failures into future successes. Mol Cancer Ther, 2018, 17(6): 1147-1155. |
24. | Alaseem A, Alhazzani K, Dondapati P, et al. Matrix metalloproteinases: A challenging paradigm of cancer management. Semin Cancer Biol, 2019, 56: 100-115. |
25. | Gonzalez-Avila G, Sommer B, García-Hernández AA, et al. Matrix metalloproteinases' role in tumor microenvironment. Adv Exp Med Biol, 2020, 1245: 97-131. |
26. | Pai FC, Huang HW, Tsai YL, et al. Inhibition of FABP6 reduces tumor cell invasion and angiogenesis through the decrease in MMP-2 and VEGF in human glioblastoma cells. Cells, 2021, 10(10): 2782. |
27. | Quintero-Fabián S, Arreola R, Becerril-Villanueva E, et al. Role of matrix metalloproteinases in angiogenesis and cancer. Front Oncol, 2019, 9: 1370. |
28. | Song L, Li XX, Liu XY, et al. EFEMP2 suppresses the invasion of lung cancer cells by inhibiting epithelial-mesenchymal transition (EMT) and down-regulating MMPs. Onco Targets Ther, 2020, 13: 1375-1396. |
29. | Shen KH, Hung JH, Liao YC, et al. Sinomenine inhibits migration and invasion of human lung cancer cell through downregulating expression of miR-21 and MMPs. Int J Mol Sci, 2020, 21(9): 3080. |
30. | Hwang KE, Kim HJ, Song IS, et al. Salinomycin suppresses TGF-β1-induced EMT by down-regulating MMP-2 and MMP-9 via the AMPK/SIRT1 pathway in non-small cell lung cancer. Int J Med Sci, 2021, 18(3): 715-726. |
31. | Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer, 2019, 1871(2): 455-468. |
32. | Lan J, Sun L, Xu F, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res, 2019, 79(1): 146-158. |
33. | Kwon Y, Kim M, Kim Y, et al. Exosomal microRNAs as mediators of cellular interactions between cancer cells and macrophages. Front Immunol, 2020, 11: 1167. |
34. | Chen J, Zhang K, Zhi Y, et al. Tumor-derived exosomal miR-19b-3p facilitates M2 macrophage polarization and exosomal LINC00273 secretion to promote lung adenocarcinoma metastasis via Hippo pathway. Clin Transl Med, 2021, 11(9): e478. |
35. | Lei J, Chen P, Zhang F, et al. M2 macrophages-derived exosomal microRNA-501-3p promotes the progression of lung cancer via targeting WD repeat domain 82. Cancer Cell Int, 2021, 21(1): 91. |
36. | Bunggulawa EJ, Wang W, Yin T, et al. Recent advancements in the use of exosomes as drug delivery systems. J Nanobiotechnology, 2018, 16(1): 81. |
37. | Kim MS, Haney MJ, Zhao Y, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: In vitro and in vivo evaluations. Nanomedicine, 2018, 14(1): 195-204. |
38. | Choo YW, Kang M, Kim HY, et al. M1 Macrophage-derived nanovesicles potentiate the anticancer efficacy of immune checkpoint inhibitors. ACS Nano, 2018, 12(9): 8977-8993. |
39. | Rayamajhi S, Nguyen TDT, Marasini R, et al. Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery. Acta Biomater, 2019, 94: 482-494. |
40. | Maimon A, Levi-Yahid V, Ben-Meir K, et al. Myeloid cell-derived PROS1 inhibits tumor metastasis by regulating inflammatory and immune responses via IL-10. J Clin Invest, 2021, 131(10): e126089. |
41. | Yang L, Dong Y, Li Y, et al. IL-10 derived from M2 macrophage promotes cancer stemness via JAK1/STAT1/NF-κB/Notch1 pathway in non-small cell lung cancer. Int J Cancer, 2019, 145(4): 1099-1110. |
42. | Che D, Zhang S, Jing Z, et al. Macrophages induce EMT to promote invasion of lung cancer cells through the IL-6-mediated COX-2/PGE 2/β-catenin signalling pathway. Mol Immunol, 2017, 90: 197-210. |
43. | Wang X, Yang X, Tsai Y, et al. IL-6 mediates macrophage infiltration after irradiation via up-regulation of CCL2/CCL5 in non-small cell lung cancer. Radiat Res, 2017, 187(1): 50-59. |
44. | Hao Y, Baker D, Ten Dijke P. TGF-β-mediated epithelial-mesenchymal transition and cancer metastasis. Int J Mol Sci, 2019, 20(11): 2767. |
45. | Batlle E, Massagué J. Transforming growth factor-β signaling in immunity and cancer. Immunity, 2019, 50(4): 924-940. |
46. | Wanna-Udom S, Terashima M, Suphakhong K, et al. KDM2B is involved in the epigenetic regulation of TGF-β-induced epithelial-mesenchymal transition in lung and pancreatic cancer cell lines. J Biol Chem, 2021, 296: 100213. |
47. | Shima T, Shimoda M, Shigenobu T, et al. Infiltration of tumor-associated macrophages is involved in tumor programmed death-ligand 1 expression in early lung adenocarcinoma. Cancer Sci, 2020, 111(2): 727-738. |
48. | Huang Q, Liu J, Wu S, et al. Spi-B promotes the recruitment of tumor-associated macrophages via enhancing CCL4 expression in lung cancer. Front Oncol, 2021, 11: 659131. |
49. | Li Y, Zhai P, Zheng Y, et al. CSF2 attenuated sepsis-induced acute kidney injury by promoting alternative macrophage transition. Front Immunol, 2020, 11: 1415. |
50. | Lu X, Yang R, Zhang L, et al. Macrophage colony-stimulating factor mediates the recruitment of macrophages in triple negative breast cancer. Int J Biol Sci, 2019, 15(13): 2859-2871. |
51. | Lu CS, Shiau AL, Su BH, et al. Oct4 promotes M2 macrophage polarization through upregulation of macrophage colony-stimulating factor in lung cancer. J Hematol Oncol, 2020, 13(1): 62. |
52. | Thawani R, McLane M, Beig N, et al. Radiomics and radiogenomics in lung cancer: A review for the clinician. Lung Cancer, 2018, 115: 34-41. |
53. | Tu MM, Abdel-Hafiz HA, Jones RT, et al. Inhibition of the CCL2 receptor, CCR2, enhances tumor response to immune checkpoint therapy. Commun Biol, 2020, 3(1): 720. |
54. | Peranzoni E, Lemoine J, Vimeux L, et al. Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc Natl Acad Sci U S A, 2018, 115(17): E4041-E4050. |
55. | Fei L, Ren X, Yu H, et al. Targeting the CCL2/CCR2 axis in cancer immunotherapy: One stone, three birds? Front Immunol, 2021, 12: 771210. |
56. | Fritz JM, Tennis MA, Orlicky DJ, et al. Depletion of tumor-associated macrophages slows the growth of chemically induced mouse lung adenocarcinomas. Front Immunol, 2014, 5: 587. |
57. | Pienta KJ, Machiels JP, Schrijvers D, et al. Phase 2 study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 (CCL2), in metastatic castration-resistant prostate cancer. Invest New Drugs, 2013, 31(3): 760-768. |
58. | Zhang L, Zhang K, Zhang J, et al. Loss of fragile site-associated tumor suppressor promotes antitumor immunity via macrophage polarization. Nat Commun, 2021, 12(1): 4300. |
59. | Rodell CB, Arlauckas SP, Cuccarese MF, et al. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat Biomed Eng, 2018, 2(8): 578-588. |
60. | Cuccarese MF, Dubach JM, Pfirschke C, et al. Heterogeneity of macrophage infiltration and therapeutic response in lung carcinoma revealed by 3D organ imaging. Nat Commun, 2017, 8: 14293. |
61. | Weissleder R, Nahrendorf M, Pittet MJ. Imaging macrophages with nanoparticles. Nat Mater, 2014, 13(2): 125-138. |
62. | Bolli E, Scherger M, Arnouk SM, et al. Targeted repolarization of tumor-associated macrophages via imidazoquinoline-linked nanobodies. Adv Sci (Weinh), 2021, 8(10): 2004574. |
63. | O'Brien SA, Orf J, Skrzypczynska KM, et al. Activity of tumor-associated macrophage depletion by CSF1R blockade is highly dependent on the tumor model and timing of treatment. Cancer Immunol Immunother, 2021, 70(8): 2401-2410. |
64. | Cieslewicz M, Tang J, Yu JL, et al. Targeted delivery of proapoptotic peptides to tumor-associated macrophages improves survival. Proc Natl Acad Sci U S A, 2013, 110(40): 15919-15924. |
65. | Yan D, Kowal J, Akkari L, et al. Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene, 2017, 36(43): 6049-6058. |
66. | Bak SP, Walters JJ, Takeya M, et al. Scavenger receptor-A-targeted leukocyte depletion inhibits peritoneal ovarian tumor progression. Cancer Res, 2007, 67(10): 4783-4789. |
67. | Zhou J, Kroll AV, Holay M, et al. Biomimetic nanotechnology toward personalized vaccines. Adv Mater, 2020, 32(13): e1901255. |
68. | Yu H, Yang Z, Li F, et al. Cell-mediated targeting drugs delivery systems. Drug Deliv, 2020, 27(1): 1425-1437. |
69. | Guo L, Zhang Y, Yang Z, et al. Tunneling nanotubular expressways for ultrafast and accurate M1 macrophage delivery of anticancer drugs to metastatic ovarian carcinoma. ACS Nano, 2019, 13(2): 1078-1096. |
70. | Sakai H, Kokura S, Ishikawa T, et al. Effects of anticancer agents on cell viability, proliferative activity and cytokine production of peripheral blood mononuclear cells. J Clin Biochem Nutr, 2013, 52(1): 64-71. |
71. | Li S, Feng S, Ding L, et al. Nanomedicine engulfed by macrophages for targeted tumor therapy. Int J Nanomedicine, 2016, 11: 4107-4124. |
72. | Doshi N, Swiston AJ, Gilbert JB, et al. Cell-based drug delivery devices using phagocytosis-resistant backpacks. Adv Mater, 2011, 23(12): H105-H109. |
73. | Estelrich J, Busquets MA. Iron oxide nanoparticles in photothermal therapy. Molecules, 2018, 23(7): 1567. |
74. | Qiang L, Cai Z, Jiang W, et al. A novel macrophage-mediated biomimetic delivery system with NIR-triggered release for prostate cancer therapy. J Nanobiotechnology, 2019, 17(1): 83. |
- 1. Xia C, Dong X, Li H, et al. Cancer statistics in China and United States, 2022: Profiles, trends, and determinants. Chin Med J, 2022, 135(5): 584-590.
- 2. Pan Y, Yu Y, Wang X, et al. Tumor-associated macrophages in tumor immunity. Front Immunol, 2020, 11: 583084.
- 3. Li H, Huang N, Zhu W, et al. Modulation the crosstalk between tumor-associated macrophages and non-small cell lung cancer to inhibit tumor migration and invasion by ginsenoside Rh2. BMC Cancer, 2018, 18(1): 579.
- 4. Sarode P, Zheng X, Giotopoulou GA, et al. Reprogramming of tumor-associated macrophages by targeting β-catenin/FOSL2/ARID5A signaling: A potential treatment of lung cancer. Sci Adv, 2020, 6(23): eaaz6105.
- 5. Chávez-Galán L, Olleros ML, Vesin D, et al. Much more than M1 and M2 macrophages, there are also CD169(+) and TCR(+) macrophages. Front Immunol, 2015, 6: 263.
- 6. Casanova-Acebes M, Dalla E, Leader AM, et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature, 2021, 595(7868): 578-584.
- 7. Tzetzo SL, Abrams SI. Redirecting macrophage function to sustain their "defender" antitumor activity. Cancer Cell, 2021, 39(6): 734-737.
- 8. Han Y, Guo W, Ren T, et al. Tumor-associated macrophages promote lung metastasis and induce epithelial-mesenchymal transition in osteosarcoma by activating the COX-2/STAT3 axis. Cancer Lett, 2019, 440-441: 116-125.
- 9. Cassetta L, Pollard JW. Tumor-associated macrophages. Curr Biol, 2020, 30(6): R246-R248.
- 10. Chen Y, Song Y, Du W, et al. Tumor-associated macrophages: An accomplice in solid tumor progression. J Biomed Sci, 2019, 26(1): 78.
- 11. Xue J, Schmidt SV, Sander J, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity, 2014, 40(2): 274-288.
- 12. Xu F, Wei Y, Tang Z, et al. Tumor-associated macrophages in lung cancer: Friend or foe? Mol Med Rep, 2020, 22(5): 4107-4115.
- 13. Cotechini T, Atallah A, Grossman A. Tissue-resident and recruited macrophages in primary tumor and metastatic microenvironments: Potential targets in cancer therapy. Cells, 2021, 10(4): 960.
- 14. Zou G, Zhang X, Wang L, et al. Herb-sourced emodin inhibits angiogenesis of breast cancer by targeting VEGFA transcription. Theranostics, 2020, 10(15): 6839-6853.
- 15. Fu LQ, Du WL, Cai MH, et al. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol, 2020, 353: 104119.
- 16. Martin JD, Seano G, Jain RK. Normalizing function of tumor vessels: Progress, opportunities, and challenges. Annu Rev Physiol, 2019, 81: 505-534.
- 17. Dallavalasa S, Beeraka NM, Basavaraju CG, et al. The role of tumor associated macrophages (TAMs) in cancer progression, chemoresistance, angiogenesis and metastasis—Current status. Curr Med Chem, 2021, 28(39): 8203-8236.
- 18. Jeon SH, Chae BC, Kim HA, et al. Mechanisms underlying TGF-beta1-induced expression of VEGF and Flk-1 in mouse macrophages and their implications for angiogenesis. J Leukoc Biol, 2007, 81(2): 557-566.
- 19. Frezzetti D, Gallo M, Maiello MR, et al. VEGF as a potential target in lung cancer. Expert Opin Ther Targets, 2017, 21(10): 959-966.
- 20. Nilsson MB, Robichaux J, Herynk MH, et al. Altered regulation of HIF-1α in naive- and drug-resistant EGFR-mutant NSCLC: Implications for a vascular endothelial growth factor-dependent phenotype. J Thorac Oncol, 2021, 16(3): 439-451.
- 21. Spagnuolo A, Palazzolo G, Sementa C, et al. Vascular endothelial growth factor receptor tyrosine kinase inhibitors for the treatment of advanced non-small cell lung cancer. Expert Opin Pharmacother, 2020, 21(4): 491-506.
- 22. Shen G, Zheng F, Ren D, et al. Anlotinib: A novel multi-targeting tyrosine kinase inhibitor in clinical development. J Hematol Oncol, 2018, 11(1): 120.
- 23. Winer A, Adams S, Mignatti P. Matrix metalloproteinase inhibitors in cancer therapy: Turning past failures into future successes. Mol Cancer Ther, 2018, 17(6): 1147-1155.
- 24. Alaseem A, Alhazzani K, Dondapati P, et al. Matrix metalloproteinases: A challenging paradigm of cancer management. Semin Cancer Biol, 2019, 56: 100-115.
- 25. Gonzalez-Avila G, Sommer B, García-Hernández AA, et al. Matrix metalloproteinases' role in tumor microenvironment. Adv Exp Med Biol, 2020, 1245: 97-131.
- 26. Pai FC, Huang HW, Tsai YL, et al. Inhibition of FABP6 reduces tumor cell invasion and angiogenesis through the decrease in MMP-2 and VEGF in human glioblastoma cells. Cells, 2021, 10(10): 2782.
- 27. Quintero-Fabián S, Arreola R, Becerril-Villanueva E, et al. Role of matrix metalloproteinases in angiogenesis and cancer. Front Oncol, 2019, 9: 1370.
- 28. Song L, Li XX, Liu XY, et al. EFEMP2 suppresses the invasion of lung cancer cells by inhibiting epithelial-mesenchymal transition (EMT) and down-regulating MMPs. Onco Targets Ther, 2020, 13: 1375-1396.
- 29. Shen KH, Hung JH, Liao YC, et al. Sinomenine inhibits migration and invasion of human lung cancer cell through downregulating expression of miR-21 and MMPs. Int J Mol Sci, 2020, 21(9): 3080.
- 30. Hwang KE, Kim HJ, Song IS, et al. Salinomycin suppresses TGF-β1-induced EMT by down-regulating MMP-2 and MMP-9 via the AMPK/SIRT1 pathway in non-small cell lung cancer. Int J Med Sci, 2021, 18(3): 715-726.
- 31. Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer, 2019, 1871(2): 455-468.
- 32. Lan J, Sun L, Xu F, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res, 2019, 79(1): 146-158.
- 33. Kwon Y, Kim M, Kim Y, et al. Exosomal microRNAs as mediators of cellular interactions between cancer cells and macrophages. Front Immunol, 2020, 11: 1167.
- 34. Chen J, Zhang K, Zhi Y, et al. Tumor-derived exosomal miR-19b-3p facilitates M2 macrophage polarization and exosomal LINC00273 secretion to promote lung adenocarcinoma metastasis via Hippo pathway. Clin Transl Med, 2021, 11(9): e478.
- 35. Lei J, Chen P, Zhang F, et al. M2 macrophages-derived exosomal microRNA-501-3p promotes the progression of lung cancer via targeting WD repeat domain 82. Cancer Cell Int, 2021, 21(1): 91.
- 36. Bunggulawa EJ, Wang W, Yin T, et al. Recent advancements in the use of exosomes as drug delivery systems. J Nanobiotechnology, 2018, 16(1): 81.
- 37. Kim MS, Haney MJ, Zhao Y, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: In vitro and in vivo evaluations. Nanomedicine, 2018, 14(1): 195-204.
- 38. Choo YW, Kang M, Kim HY, et al. M1 Macrophage-derived nanovesicles potentiate the anticancer efficacy of immune checkpoint inhibitors. ACS Nano, 2018, 12(9): 8977-8993.
- 39. Rayamajhi S, Nguyen TDT, Marasini R, et al. Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery. Acta Biomater, 2019, 94: 482-494.
- 40. Maimon A, Levi-Yahid V, Ben-Meir K, et al. Myeloid cell-derived PROS1 inhibits tumor metastasis by regulating inflammatory and immune responses via IL-10. J Clin Invest, 2021, 131(10): e126089.
- 41. Yang L, Dong Y, Li Y, et al. IL-10 derived from M2 macrophage promotes cancer stemness via JAK1/STAT1/NF-κB/Notch1 pathway in non-small cell lung cancer. Int J Cancer, 2019, 145(4): 1099-1110.
- 42. Che D, Zhang S, Jing Z, et al. Macrophages induce EMT to promote invasion of lung cancer cells through the IL-6-mediated COX-2/PGE 2/β-catenin signalling pathway. Mol Immunol, 2017, 90: 197-210.
- 43. Wang X, Yang X, Tsai Y, et al. IL-6 mediates macrophage infiltration after irradiation via up-regulation of CCL2/CCL5 in non-small cell lung cancer. Radiat Res, 2017, 187(1): 50-59.
- 44. Hao Y, Baker D, Ten Dijke P. TGF-β-mediated epithelial-mesenchymal transition and cancer metastasis. Int J Mol Sci, 2019, 20(11): 2767.
- 45. Batlle E, Massagué J. Transforming growth factor-β signaling in immunity and cancer. Immunity, 2019, 50(4): 924-940.
- 46. Wanna-Udom S, Terashima M, Suphakhong K, et al. KDM2B is involved in the epigenetic regulation of TGF-β-induced epithelial-mesenchymal transition in lung and pancreatic cancer cell lines. J Biol Chem, 2021, 296: 100213.
- 47. Shima T, Shimoda M, Shigenobu T, et al. Infiltration of tumor-associated macrophages is involved in tumor programmed death-ligand 1 expression in early lung adenocarcinoma. Cancer Sci, 2020, 111(2): 727-738.
- 48. Huang Q, Liu J, Wu S, et al. Spi-B promotes the recruitment of tumor-associated macrophages via enhancing CCL4 expression in lung cancer. Front Oncol, 2021, 11: 659131.
- 49. Li Y, Zhai P, Zheng Y, et al. CSF2 attenuated sepsis-induced acute kidney injury by promoting alternative macrophage transition. Front Immunol, 2020, 11: 1415.
- 50. Lu X, Yang R, Zhang L, et al. Macrophage colony-stimulating factor mediates the recruitment of macrophages in triple negative breast cancer. Int J Biol Sci, 2019, 15(13): 2859-2871.
- 51. Lu CS, Shiau AL, Su BH, et al. Oct4 promotes M2 macrophage polarization through upregulation of macrophage colony-stimulating factor in lung cancer. J Hematol Oncol, 2020, 13(1): 62.
- 52. Thawani R, McLane M, Beig N, et al. Radiomics and radiogenomics in lung cancer: A review for the clinician. Lung Cancer, 2018, 115: 34-41.
- 53. Tu MM, Abdel-Hafiz HA, Jones RT, et al. Inhibition of the CCL2 receptor, CCR2, enhances tumor response to immune checkpoint therapy. Commun Biol, 2020, 3(1): 720.
- 54. Peranzoni E, Lemoine J, Vimeux L, et al. Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc Natl Acad Sci U S A, 2018, 115(17): E4041-E4050.
- 55. Fei L, Ren X, Yu H, et al. Targeting the CCL2/CCR2 axis in cancer immunotherapy: One stone, three birds? Front Immunol, 2021, 12: 771210.
- 56. Fritz JM, Tennis MA, Orlicky DJ, et al. Depletion of tumor-associated macrophages slows the growth of chemically induced mouse lung adenocarcinomas. Front Immunol, 2014, 5: 587.
- 57. Pienta KJ, Machiels JP, Schrijvers D, et al. Phase 2 study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 (CCL2), in metastatic castration-resistant prostate cancer. Invest New Drugs, 2013, 31(3): 760-768.
- 58. Zhang L, Zhang K, Zhang J, et al. Loss of fragile site-associated tumor suppressor promotes antitumor immunity via macrophage polarization. Nat Commun, 2021, 12(1): 4300.
- 59. Rodell CB, Arlauckas SP, Cuccarese MF, et al. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat Biomed Eng, 2018, 2(8): 578-588.
- 60. Cuccarese MF, Dubach JM, Pfirschke C, et al. Heterogeneity of macrophage infiltration and therapeutic response in lung carcinoma revealed by 3D organ imaging. Nat Commun, 2017, 8: 14293.
- 61. Weissleder R, Nahrendorf M, Pittet MJ. Imaging macrophages with nanoparticles. Nat Mater, 2014, 13(2): 125-138.
- 62. Bolli E, Scherger M, Arnouk SM, et al. Targeted repolarization of tumor-associated macrophages via imidazoquinoline-linked nanobodies. Adv Sci (Weinh), 2021, 8(10): 2004574.
- 63. O'Brien SA, Orf J, Skrzypczynska KM, et al. Activity of tumor-associated macrophage depletion by CSF1R blockade is highly dependent on the tumor model and timing of treatment. Cancer Immunol Immunother, 2021, 70(8): 2401-2410.
- 64. Cieslewicz M, Tang J, Yu JL, et al. Targeted delivery of proapoptotic peptides to tumor-associated macrophages improves survival. Proc Natl Acad Sci U S A, 2013, 110(40): 15919-15924.
- 65. Yan D, Kowal J, Akkari L, et al. Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene, 2017, 36(43): 6049-6058.
- 66. Bak SP, Walters JJ, Takeya M, et al. Scavenger receptor-A-targeted leukocyte depletion inhibits peritoneal ovarian tumor progression. Cancer Res, 2007, 67(10): 4783-4789.
- 67. Zhou J, Kroll AV, Holay M, et al. Biomimetic nanotechnology toward personalized vaccines. Adv Mater, 2020, 32(13): e1901255.
- 68. Yu H, Yang Z, Li F, et al. Cell-mediated targeting drugs delivery systems. Drug Deliv, 2020, 27(1): 1425-1437.
- 69. Guo L, Zhang Y, Yang Z, et al. Tunneling nanotubular expressways for ultrafast and accurate M1 macrophage delivery of anticancer drugs to metastatic ovarian carcinoma. ACS Nano, 2019, 13(2): 1078-1096.
- 70. Sakai H, Kokura S, Ishikawa T, et al. Effects of anticancer agents on cell viability, proliferative activity and cytokine production of peripheral blood mononuclear cells. J Clin Biochem Nutr, 2013, 52(1): 64-71.
- 71. Li S, Feng S, Ding L, et al. Nanomedicine engulfed by macrophages for targeted tumor therapy. Int J Nanomedicine, 2016, 11: 4107-4124.
- 72. Doshi N, Swiston AJ, Gilbert JB, et al. Cell-based drug delivery devices using phagocytosis-resistant backpacks. Adv Mater, 2011, 23(12): H105-H109.
- 73. Estelrich J, Busquets MA. Iron oxide nanoparticles in photothermal therapy. Molecules, 2018, 23(7): 1567.
- 74. Qiang L, Cai Z, Jiang W, et al. A novel macrophage-mediated biomimetic delivery system with NIR-triggered release for prostate cancer therapy. J Nanobiotechnology, 2019, 17(1): 83.