MORPHOLOGICAL AND FUNCTIONAL CHARACTERISTICS OF THE BREAST CANCER MICROENVIRONMENT: SCIENTIFIC REVIEW
https://doi.org/10.20340/mv-mn.2024.32(1).848
Abstract
Breast cancer is a heterogeneous malignant disease with a wide variety of morphology, molecular characteristics and clinical presentation. In the formation of the tumor behavioral phenotype and its response to treatment an important role in the formation of the tumor behavioral phenotype and its response to treatment plays the tumor microenvironment, which makes its assessment critical in choosing therapeutic tactics. However, the practical aspects of using breast cancer microenvironment data are insufficiently studied. The aim of the study is to systematize the morphological and functional characteristics of the main cell types in the breast cancer tumor microenvironment and analyze the possibility practical use of these data. The material was scientific databases, information and library resources for the relevant keywords, the analysis included literature from 2018 to 2023. It is shown that the components of the tumor stroma, both the cellular component and the extracellular matrix, play a special role in carcinogenesis, and this role is not always unambiguous. Analysis of cell polymorphism and tumor microenvironment types should not be limited to research interest only, but should also be an integral part of the histopathological characteristics of a specific clinical case for the possibility of its further practical application. The tumor microenvironment is not just a stroma that feeds the tumor tissue, but also an active participant in carcinogenesis. The breast cancer tumor microenvironment includes cellular and extracellular components, each of which has its own functional and morphological subtypes. Tumor microenvironment cells have functional polymorphism, which creates difficulties on the way to obtaining a complete picture of carcinogenesis and mutual influence in the «tumor cell – microenvironment» system. Analysis of the components of the tumor microenvironment, elucidation of its role and complex mechanisms of cellular interaction of the microenvironment with tumor tissue, including the use of artificial intelligence technologies, can significantly advance knowledge about the mechanisms of breast cancer development for the development of effective technologies for prevention and treatment.
About the Authors
Maksim V. MnikhovichRussian Federation
Candidate of Medical Sciences, Associate Professor, Leading Researcher of the Central Pathology Laboratory
Competing Interests:
The Author declares that he did have no conflicts of interest in planning, implementing, financing and using the results of this study
Polina A. Akhsanova
Russian Federation
Studentin
Competing Interests:
The Author declares that she did have no conflicts of interest in planning, implementing, financing and using the results of this study
Tat'yana V. Bezuglova
Russian Federation
Candidate of Biological Sciences, Senior Researcher of the Central Pathology Laboratory, Deputy Director at Science Activity
Competing Interests:
The Author declares that she did have no conflicts of interest in planning, implementing, financing and using the results of this study
Lyudmila M. Erofeeva
Russian Federation
Doctor of Biological Sciences, Professor, Leading Researcher of the Central Pathology Laboratory
Competing Interests:
The Author declares that she did have no conflicts of interest in planning, implementing, financing and using the results of this study
Ivan A. Shiripenko
Russian Federation
Student of the Pirogov Russian National Research Medical University; Laboratory Assistant-Researcher of the Central Pathology Laboratory of the Academician Petrovsky National Research Centre of Surgery
Competing Interests:
The Author declares that he did have no conflicts of interest in planning, implementing, financing and using the results of this study
Ol'ga A. Sidorova
Russian Federation
Studentin
Competing Interests:
The Author declares that she did have no conflicts of interest in planning, implementing, financing and using the results of this study
Milena V. Lozina
Russian Federation
Research Engineer of the Museum and Collection Group
Competing Interests:
The Author declares that she did have no conflicts of interest in planning, implementing, financing and using the results of this study
Marina V. Dronova
Russian Federation
Forensic Medicine Expert, Senior Lecturer of the Department of Medical Disciplines and Life Safety
Competing Interests:
The Author declares that she did have no conflicts of interest in planning, implementing, financing and using the results of this study
References
1. Jai Prakash. The Tumor Stroma: Biology and Therapeutics. 1 ed. Stanford: Jenny Stanford Publishing Pte. Ltd., 2022. – 446pp
2. Lepucki A, Orlińska K, Mielczarek-Palacz A et al. The Role of Extracellular Matrix Proteins in Breast Cancer. JCM. 2022;11(5):1250. https://doi.org/10.3390/jcm11051250
3. Winkler J, Abisoye-Ogunniyan A, Metcalf KJ et al. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun. 2020;11(1):5120. https://doi.org/10.1038/s41467-020-18794-x
4. Harbeck N, Penault-Llorca F, Cortes J et al. Breast cancer. Nat Rev Dis Primers. 2019;5(1):66. https://doi.org/10.1038/s41572-019-0111-2
5. Ferlay J, Colombet M, Soerjomataram I et al. Cancer statistics for the year 2020: An overview. Int J Cancer. 2021;149(4):778-789. https://doi.org/10.1002/ijc.33588
6. Britt KL, Cuzick J, Phillips KA. Key steps for effective breast cancer prevention. Nat Rev Cancer. 2020;20(8):417–436. https://doi.org/10.1038/s41568-020-0266-x
7. Wild CP, Weiderpass E, Stewart BW. World Cancer Report: Cancer Research for Cancer Prevention. 2020. – 594pp. URL: https://www.iccp-portal.org/system/files/resources/IARC%20World%20Cancer%20Report%202020.pdf. Date of access 20.05.2024
8. Watkins EJ. Overview of breast cancer. JAAPA. 2019;32(10):13–17. https://doi.org/10.1097/01.JAA.0000580524.95733.3d
9. Gaudet MM, Carter BD, Brinton LA et al. Pooled analysis of active cigarette smoking and invasive breast cancer risk in 14 cohort studies. Int J Epidemiol. 2017;46(3):881-893. DOI: 10.1093/ije/dyw288
10. Anderson KN, Schwab RB, Martinez ME. Reproductive risk factors and breast cancer subtypes: a review of the literature. Breast Cancer Res Treat. 2014;144(1):1-10. https://doi.org/10.1007/s10549-014-2852-7
11. Loibl S, Poortmans P, Morrow M et al. Breast cancer. The Lancet.2021;397(10286):1750–1769. https://doi.org/10.1016/S0140-6736(20)32381-3
12. Shiovitz S, Korde LA. Genetics of breast cancer: a topic in evolution. Annals of Oncology. 2015;26(7):1291–1299. https://doi.org/10.1093/annonc/mdv022
13. Kuchenbaecker KB, Hopper JL, Barnes DR et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA. 2017;317(23):2402. https://doi.org/ 10.1001/jama.2017.7112
14. Easton DF, Pharoah PDP, Antoniou AC, et al. Gene-Panel Sequencing and the Prediction of Breast-Cancer Risk. N Engl J Med. 2015;372(23):2243–2257. https://doi.org/10.1056/NEJMsr1501341
15. Coughlin SS. Epidemiology of Breast Cancer in Women. Ed. by: A. Ahmad. Breast Cancer Metastasis and Drug Resistance. Cham: Springer International Publishing, 2019. - P. 9-29. https://doi.org/10.1007/978-3-030-20301-6_2
16. Aydiner A, Igci A, Soran A. Breast Cancer: a Guide to Clinical Practice. Cham, Switzerland: Springer, 2019. - 615pр
17. Tan PH, Ellis I, Allison K et al. Classification of Tumours Editorial Board. The 2019 World Health Organization classification of tumors of the breast. Histopathology. 2020;77(2):181-185. https://doi.org/10.1111/his.14091
18. Beatson GT. On the Treatment of Inoperable Cases of Carcinoma of the Mamma: Suggestions for a New Method of Treatment, with Illustrative Cases. Trans Med Chir Soc Edinb. 1896;15:153-179
19. Chlebowski RT, Anderson GL. Menopausal hormone therapy and cancer: changing clinical observations of target site specificity. Steroids. 2014;90:53-59. https://doi.org/10.1016/j.steroids.2014.06.001
20. Chen WY. Exogenous and endogenous hormones and breast cancer. Best Pract Res Clin Endocrinol Metab. 2008;22(4):573-585. https://doi.org/10.1016/j.beem.2008.08.001
21. Yue W, Yager JD, Wang JP et al. Estrogen receptor-dependent and independent mechanisms of breast cancer carcinogenesis. Steroids.2013;78(2):161-170. https://doi.org/10.1016/j.steroids.2012.11.001
22. Joshi H, Press MF. Molecular Oncology of Breast Cancer. В: The Breast. Elsevier. 2018;282-307.e5. https://doi.org/10.1016/B978-0-323-35955-9.00022-2
23. Creighton CJ, Kent Osborne C, van de Vijver MJ et al. Molecular profiles of progesterone receptor loss in human breast tumors. Breast Cancer Res Treat. 2009;114(2):287-299. https://doi.org/10.1007/s10549-008-0017-2
24. Gross GE, Clark GM, Chamness GC et al. Multiple progesterone receptor assays in human breast cancer. Cancer Res. 1984;44(2):836-840
25. Rugo HS, Rumble RB, Macrae E et al. Endocrine Therapy for Hormone Receptor-Positive Metastatic Breast Cancer: American Society of Clinical Oncology Guideline. J Clin Oncol. 2016;34(25):3069-3103
26. Slamon DJ, Clark GM, Wong SG et al. Human Breast Cancer: Correlation of Relapse and Survival with Amplification of the HER-2/ neu Oncogene. Science. 1987;235(4785):177-182. https://doi.org/10.1126/science.3798106
27. Eroglu Z, Tagawa T, Somlo G. Human epidermal growth factor receptor family-targeted therapies in the treatment of HER2-overexpressing breast cancer. Oncologist. 2014;19(2):135-150. https://doi.org/10.1634/theoncologist.2013-0283
28. Puchinskaya MV. Epitelialno-mesenkhimal’ny perekhod v norme i patologii. Arkh patol. 2015;77(1):75-83. In Russian
29. Yang J, Antin P, Berx G et al. On behalf of the EMT International Association (TEMTIA). Guidelines and definitions for research on epithelial–mesenchymal transition. Nat Rev Mol Cell Biol. 2020;21(6):341-352. https://doi.org/10.1038/s41580-021-00428-9
30. Russo J. The Pathobiology of Breast Cancer. Cham: Springer International Publishing, 2016. - https://doi.org/10.1007/978-3-319-40815-6
31. Liu F, Gu LN, Shan BE et al. Biomarkers for EMT and MET in breast cancer: An update. Oncology Letters. 2016;12(6):4869-4876. https://doi.org/10.3892/ol.2016.5369
32. Yamashita N, Tokunaga E, Iimori M et al. Epithelial Paradox: Clinical Significance of Coexpression of E-cadherin and Vimentin With Regard to Invasion and Metastasis of Breast Cancer. Clin Breast Cancer. 2018;18(5):e1003-1009. https://doi.org/10.1016/j.clbc.2018.02.002
33. Jorgensen CLT, Forsare C, Bendahl PO et al. Expression of epithelial-mesenchymal transition-related markers and phenotypes during breast cancer progression. Breast Cancer Res Treat. 2020;181(2):369-381. https://doi.org/10.1007/s10549-020-05627-0
34. Klenova N.A. Biokhimiya patologicheskikh sostoyany: uchebnoe posobie. Samara: Izd-vo «Samarsky universitet». 2006. - 216s. In Russian
35. Dzobo K, Senthebane DA, Dandara C. The Tumor Microenvironment in Tumorigenesis and Therapy Resistance Revisited. Cancer. 2023;15(2):376. https://doi.org/10.3390/cancers15020376
36. Hanley CJ, Mellone M, Ford K et al. Targeting the Myofibroblastic Cancer-Associated Fibroblast Phenotype Through Inhibition of NOX4. JNCI: Journal of the National Cancer Institute. 2018;110(1):109-120. https://doi.org/10.1093/jnci/djx121
37. Arina A, Idel C, Hyjek EM et al. Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc Natl Acad Sci USA. 2016;113(27):7551-7556. https://doi.org/10.1073/pnas.1600363113
38. Bochet L, Lehuédé C, Dauvillier S et al. Adipocyte-Derived Fibroblasts Promote Tumor Progression and Contribute to the Desmoplastic Reaction in Breast Cancer. Cancer Research. 2013;73(18):5657-5668. https://doi.org/10.1158/0008-5472.CAN-13-0530
39. Bartoschek M, Oskolkov N, Bocci M et al. Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat Commun. 2018;9(1):5150.
40. Raz Y, Cohen N, Shani O et al. Bone marrow–derived fibroblasts are a functionally distinct stromal cell population in breast cancer. Journal of Experimental Medicine. 2012;215(12):3075-3093. https://doi.org/10.1084/jem.20180818
41. Karnoub AE, Dash AB, Vo AP et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449(7162):557-563. https://doi.org/10.1038/nature06188
42. Zeisberg EM, Potenta S, Xie L et al. Discovery of Endothelial to Mesenchymal Transition as a Source for Carcinoma-Associated Fibroblasts. Cancer Research. 2007;67(21):10123-10128. https://doi.org/10.1158/0008-5472.CAN-07-3127
43. Iwano M, Plieth D, Danoff TM et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110(3):341-350. https://doi.org/10.1172/JCI15518
44. Kojima Y, Acar A, Eaton EN et al. Autocrine TGF-β and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc Natl Acad Sci USA. 2010;107(46):20009-200014. https://doi.org/10.1073/pnas.1013805107
45. Arcucci A, Ruocco MR, Granato G et al. Cancer: An Oxidative Crosstalk between Solid Tumor Cells and Cancer Associated Fibroblasts. BioMed Research International. 2016;2016:1–7. https://doi.org/10.1155/2016/4502846
46. De Wever O, Demetter P, Mareel M et al. Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer. 2008;123(10):2229-2238. https://doi.org/10.1002/ijc.23925
47. Zeltz C, Primac I, Erusappan P et al. Cancer-associated fibroblasts in desmoplastic tumors: emerging role of integrins. Seminars in Cancer Biology. 2020;62:166-181. https://doi.org/10.1016/j.semcancer.2019.08.004
48. Gaggioli C, Hooper S, Hidalgo-Carcedo C et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9(12):1392-1400. https://doi.org/10.1038/ncb1658
49. Cui Q, Wang B, Li K et al. Upregulating MMP 1 in carcinoma associated fibroblasts reduces the efficacy of Taxotere on breast cancer synergized by Collagen IV. Oncol Lett. 2018. https://doi.org/10.3892/ol.2018.9092
50. Chen W, Tang T, Eastham-Anderson J et al. Canonical hedgehog signaling augments tumor angiogenesis by induction of VEGF-A in stromal perivascular cells. Proc Natl Acad Sci USA. 2011;108(23):9589-9594. https://doi.org/10.1073/pnas.1017945108
51. Zhao H, Yang L, Baddour J et al. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. eLife;5:e10250. https://doi.org/10.7554/eLife.10250
52. Labernadie A, Kato T, Brugués A et al. A mechanically active heterotypic E-cadherin/N-cadherin adhesion enables fibroblasts to drive cancer cell invasion. Nat Cell Biol. 2017;19(3):224-237. https://doi.org/10.1038/ncb3478
53. Roberts EW, Deonarine A, Jones JO et al. Depletion of stromal cells expressing fibroblast activation protein-α from skeletal muscle and bone marrow results in cachexia and anemia. Journal of Experimental Medicine. 2013;210(6):1137-11351. https://doi.org/ 10.1084/jem.20122344
54. Kraman M, Bambrough PJ, Arnold JN et al. Suppression of Antitumor Immunity by Stromal Cells Expressing Fibroblast Activation Protein–α. Science. 2010;330(6005):827-830. https://doi.org/ 10.1126/science.1195300
55. Neri S, Ishii G, Hashimoto H et al. Podoplanin-expressing cancer-associated fibroblasts lead and enhance the local invasion of cancer cells in lung adenocarcinoma: PDPN-CAFs lead and enhance cancer cell invasion. Int J Cancer. 2015;137(4):784-796. https://doi.org/ 10.1002/ijc.29464
56. Yamamoto Y, Kasashima H, Fukui Y et al. The heterogeneity of cancer‐associated fibroblast subpopulations: Their origins, biomarkers, and roles in the tumor microenvironment. Cancer Science. 2023;114(1):16-24. https://doi.org/10.1111/cas.15609
57. Miyai Y, Esaki N, Takahashi M et al. Cancer‐associated fibroblasts that restrain cancer progression: Hypotheses and perspectives. Cancer Sci. 2020;111(4):1047-1057. https://doi.org/10.1111/cas.14346
58. Mizutani Y, Kobayashi H, Iida T et al. Meflin-Positive Cancer-Associated Fibroblasts Inhibit Pancreatic Carcinogenesis. Cancer Research. 2019;79(20):5367-5381. https://doi.org/10.1158/0008-5472.CAN-19-0454
59. Paulsson J, Micke P. Prognostic relevance of cancer-associated fibroblasts in human cancer. Seminars in Cancer Biology. 2014;25:61-68. https://doi.org/10.1016/j.semcancer.2014.02.006
60. Richardson AM, Havel LS, Koyen AE et al. Vimentin Is Required for Lung Adenocarcinoma Metastasis via Heterotypic Tumor Cell–Cancer-Associated Fibroblast Interactions during Collective Invasion. Clinical Cancer Research. 2018;24(2):420-432. https://doi.org/10.1158/1078-0432.CCR-17-1776
61. O’Connell JT, Sugimoto H, Cooke VG et al. VEGF-A and Tenascin-C produced by S100A4 + stromal cells are important for metastatic colonization. Proc Natl Acad Sci USA. 2011;108(38):16002-16007. https://doi.org/10.1073/pnas.1109493108
62. Yang X, Lin Y, Shi Y et al. FAP Promotes Immunosuppression by Cancer-Associated Fibroblasts in the Tumor Microenvironment via STAT3–CCL2 Signaling. Cancer Research. 2016;76(14):4124-4135. https://doi.org/10.1158/0008-5472.CAN-15-2973
63. Hu G, Wang S, Xu F et al. Tumor-Infiltrating Podoplanin+ Fibroblasts Predict Worse Outcome in Solid Tumors. Cell Physiol Biochem. 2018;51(3):1041–1050. https://doi.org/10.1159/000495484
64. Tamma R, Guidolin D, Annese T et al. Spatial distribution of mast cells and macrophages around tumor glands in human breast ductal carcinoma. Experimental Cell Research. 2017;359(1):179-184. https://doi.org/10.1016/j.yexcr.2017.07.033
65. Meyer N, Zenclussen AC. Mast cells-Good guys with a bad image? Am J Reprod Immunol. 2018;80(4):e13002. https://doi.org/10.1111/aji.13002
66. Komi DEA, Redegeld FA. Role of Mast Cells in Shaping the Tumor Microenvironment. Clinic Rev Allerg Immunol. 2020;58(3):313-325. https://doi.org/10.1007/s12016-019-08753-w
67. Hempel HA, Cuka NS, Kulac I et al. Low Intratumoral Mast Cells Are Associated with a Higher Risk of Prostate Cancer Recurrence: Mast Cells and Prostate Cancer Recurrence. Prostate. 2017;77(4):412-424. https://doi.org/10.1002/pros.23280
68. De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 2017;17(8):457–474. https://doi.org/10.1038/nrc.2017.51
69. Mnihovich MV, Ternov MM, Miglyas VG. Predrak i rak molochnoy zhelezy: svetovaya i elektronnomikroskopicheskaya otsenka ekstratsellyulyarnogo matriksa, angiogeneza i kletochnogo mikrookruzheniya. Patologiya. 2011;8(1):36-41. In Russian
70. Cheng S, Li Z, Gao R et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell. 2021;184(3):792-809.e23. https://doi.org/10.1016/j.cell.2021.01.010
71. Pittet MJ, Michielin O, Migliorini D. Clinical relevance of tumour-associated macrophages. Nat Rev Clin Oncol. 2022;19(6):402-421. https://doi.org/10.3389/fimmu.2023.1078705
72. Jayasingam SD, Citartan M, Thang TH et al. Evaluating the Polarization of Tumor-Associated Macrophages Into M1 and M2 Phenotypes in Human Cancer Tissue: Technicalities and Challenges in Routine Clinical Practice. Front Oncol. 2020;9:1512. https://doi.org/10.3389/fonc.2019.01512
73. Yang L, Zhang Y. Tumor-associated macrophages: from basic research to clinical application. J Hematol Oncol. 2017;10(1):58. https://doi.org/10.1186/s13045-017-0430-2
74. Chen Y, Tan W, Wang C. Tumor-associated macrophage-derived cytokines enhance cancer stem-like characteristics through epithelial-mesenchymal transition. OTT. 2018;11:3817–2386. https://doi.org/10.2147/OTT.S168317
75. Erin N, Grahovac J, Brozovic A et al. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resistance Updates. 2020;53:100715. https://doi.org/10.1016/j.drup.2020.100715
76. Ireland L, Santos A, Campbell F et al. Blockade of insulin-like growth factors increases efficacy of paclitaxel in metastatic breast cancer. Oncogene. 2018;37(15):2022-2036. https://doi.org/10.1038/s41388-017-0115-x
77. Lee YS, Radford KJ. The role of dendritic cells in cancer. В: International Review of Cell and Molecular Biology. Amsterdam: Elsevier, 2019.- P. 123–178. https://doi.org/10.1016/bs.ircmb.2019.07.006
78. Wu, Saxena, Awaji, Singh. Tumor-Associated Neutrophils in Cancer: Going Pro. Cancers. 2019;11(4):564. https://doi.org/10.3390/cancers11040564
79. Galdiero MR, Bonavita E, Barajon I et al. Tumor associated macrophages and neutrophils in cancer. Immunobiology. 2013;218(11):1402-1410. https://doi.org/10.1016/j.imbio.2013.06.003
80. Jabłońska-Trypuć A, Matejczyk M, Rosochacki S. Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. Journal of Enzyme Inhibition and Medicinal Chemistry. 2016;31(suppl1):177-183. https://doi.org/10.3109/14756366.2016.1161620
81. Ku HC, Cheng CF. Role of adipocyte browning in prostate and breast tumor microenvironment. Tzu Chi Med J. 2022;34(4):359. https://doi.org/10.4103/tcmj.tcmj_62_22
82. Camarda R, Williams J, Malkov S et al. Tumor cell-adipocyte gap junctions activate lipolysis in breast cancer. Cancer Biology. BioRXiv.2018. https://doi.org/10.1101/277939
83. Wu Q, Li B, Li Z et al. Cancer-associated adipocytes: key players in breast cancer progression. J Hematol Oncol. 2019;12(1):95. https://doi.org/10.1186/s13045-019-0778-6
84. Schaaf MB, Garg AD, Agostinis P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 2018;9(2):115. https://doi.org/10.1038/s41419-017-0061-0
85. López-Soto A, Gonzalez S, Smyth MJ et al. Control of Metastasis by NK Cells. Cancer Cell. 2017;32(2):135-154. https://doi.org/10.1016/j.ccell.2017.06.009
86. Rastogi I, Jeon D, Moseman JE et al. Role of B cells as antigen presenting cells. Front Immunol. 2022;13:954936. https://doi.org/10.3389/fimmu.2022.954936
87. Ghosh D, Jiang W, Mukhopadhyay D et al. New insights into B cells as antigen presenting cells. Current Opinion in Immunology. 2021;70:129-137. https://doi.org/10.1016/j.coi.2021.06.003
88. Kuroda H, Jamiyan T, Yamaguchi R et al. Tumor microenvironment in triple-negative breast cancer: the correlation of tumor-associated macrophages and tumor-infiltrating lymphocytes. Clin Transl Oncol. 2021;23(12):2513-2525. https://doi.org/10.1007/s12094-021-02652-3
89. Catalán D, Mansilla MA, Ferrier A et al. Immunosuppressive Mechanisms of Regulatory B Cells. Front Immunol. 2021;12:611795. https://doi.org/10.3389/fimmu.2021.611795
90. Dees S, Ganesan R, Singh S et al. Regulatory T cell targeting in cancer: Emerging strategies in immunotherapy. Eur J Immunol. 2021;51(2):280-291. https://doi.org/10.1002/eji.202048992
91. Hsu YL, Yen MC, Chang WA et al. CXCL17-derived CD11b+Gr-1+ myeloid-derived suppressor cells contribute to lung metastasis of breast cancer through platelet-derived growth factor-BB. Breast Cancer Res. 2019;21(1):23. https://doi.org/10.1186/s13058-019-1114-3
92. Sasidharan Nair V, Saleh R, Toor SM et al. Transcriptomic profiling disclosed the role of DNA methylation and histone modifications in tumor-infiltrating myeloid-derived suppressor cell subsets in colorectal cancer. Clin Epigenet. 2020;12(1):13. https://doi.org/10.1186/s13148-020-0808-9
93. Bonowicz K, Mikołajczyk K, Faisal I et al. Mechanism of Extracellular Vesicle Secretion Associated with TGF-β-Dependent Inflammatory Response in the Tumor Microenvironment. IJMS. 2022;23(23):15335. https://doi.org/10.3390/ijms232315335
94. Ansell SM, Vonderheide RH. Cellular Composition of the Tumor Microenvironment. American Society of Clinical Oncology Educational Book. 2013;(33):e91-7. https://org/doi/10.14694/EdBook_AM.2013.33.e91
95. Hu Y, Qi W, Sun L et al. Effect of TGF β1 on blood CD4+CD25high regulatory T cell proliferation and Foxp3 expression during non small cell lung cancer blood metastasis. Exp Ther Med. 2018;16(2):1403-1410. https://doi.org/10.3892/etm.2018.6306
96. Draganov D, Han Z, Rana A et al. Ivermectin converts cold tumors hot and synergizes with immune checkpoint blockade for treatment of breast cancer. NPJ Breast Cancer. 2021;7(1):22. https://doi.org/10.1038/s41523-021-00229-5
97. Savas P, Salgado R, Denkert C et al. Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat Rev Clin Oncol. 2016;13(4):228-241. https://doi.org/10.1038/nrclinonc.2015.215
98. Ding JH, Xiao Y, Zhao S et al. Integrated analysis reveals the molecular features of fibrosis in triple-negative breast cancer. Molecular Therapy - Oncolytics. 2022;24:624–635. https://doi.org/10.1016/j.omto.2022.02.003
99. Salgado R, Denkert C, Demaria S et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Annals of Oncology. 2015;26(2):259-271. https://doi.org/10.1093/annonc/mdu450
100. Li JJ, Tsang JY, Tse GM. Tumor Microenvironment in Breast Cancer - Updates on Therapeutic Implications and Pathologic Assessment. Cancers. 2021;13(16):4233. https://doi.org/10.3390/cancers13164233
101. Mir MA. Role of Tumor Microenvironment in Breast Cancer and Targeted Therapies. 1 ed. Amsterdam: Academic Press, 2022.- 275pp. https://doi.org/10.1016/C2022-0-00074-X
102. Beguinot M, Dauplat MM, Kwiatkowski F et al. Analysis of tumour-infiltrating lymphocytes reveals two new biologically different subgroups of breast ductal carcinoma in situ. BMC Cancer. 2018;18(1):129. https://doi.org/10.1186/s12885-018-4013-6
103. Bhatia JK, Chaudhary T, Boruah D et al. Study of angiogenesis in invasive breast carcinoma by morphometry and immunohistochemistry. Medical Journal Armed Forces India. 2022;78(3):345–354. https://doi.org/10.1016/j.mjafi.2021.10.013
104. Monneur A, Gonçalves A, Bertucci F. Expression de PD-L1 et inhibiteurs de la voie PD-1/PD-L1 dans le cancer du sein. Bulletin du Cancer. 2018;105(3):263-274. https://doi.org/10.1016/j.bulcan.2017.11.012
105. Wang B, Liu J, Han Y et al. The Presence of Tertiary Lymphoid Structures Provides New Insight Into the Clinicopathological Features and Prognosis of Patients With Breast Cancer. Front Immunol. 2022;13:868155. https://doi.org/10.3389/fimmu.2022.868155
106. Mohammed ZMA, Going JJ, Edwards J et al. The relationship between lymphocyte subsets and clinico-pathological determinants of survival in patients with primary operable invasive ductal breast cancer. Br J Cancer. 2013;109(6):1676-1684. https://doi.org/10.1038/bjc.2013.493
107. Karancsi Z, Hagenaars SC, Németh K et al. Tumour-stroma ratio (TSR) in breast cancer: comparison of scoring core biopsies versus resection specimens. Virchows Arch. 2023. https://doi.org/10.1007/s00428-023-03555-0
108. Hagenaars SC, Vangangelt KMH, Van Pelt GW, et al. Standardization of the tumor-stroma ratio scoring method for breast cancer research. Breast Cancer Res Treat. 2022;193(3):545-553. https://doi.org/10.1007/s10549-022-06587-3
109. Mesker WE, Junggeburt JMC, Szuhai K et al. The Carcinoma–Stromal Ratio of Colon Carcinoma Is an Independent Factor for Survival Compared to Lymph Node Status and Tumor Stage. Analytical Cellular Pathology. 2007;29(5):387–398. https://doi.org/10.1155/2007/175276
110. Öztürk Ç, Okcu O, Şen B et al. An easy and practical prognostic parameter: tumor–stroma ratio in Luminal, Her2, and triple-negative breast cancers. Rev Assoc Med Bras. 2022;68(2):227-233. https://doi.org/10.1590/1806-9282.20210979
111. Baxi V, Edwards R, Montalto M et al. Digital pathology and artificial intelligence in translational medicine and clinical practice. Modern Pathology. 2022;35(1):23-32. https://doi.org/10.1038/s41379-021-00919-2
Supplementary files
The scientific review systematizes and analyzes morphological and functional data about of the breast cancer microenvironment main cell types and the possibilities of their practical use
Review
For citations:
Mnikhovich M.V., Akhsanova P.A., Bezuglova T.V., Erofeeva L.M., Shiripenko I.A., Sidorova O.A., Lozina M.V., Dronova M.V. MORPHOLOGICAL AND FUNCTIONAL CHARACTERISTICS OF THE BREAST CANCER MICROENVIRONMENT: SCIENTIFIC REVIEW. Morphological newsletter. 2024;32(1):ID-848. (In Russ.) https://doi.org/10.20340/mv-mn.2024.32(1).848