Principles of premetastatic niche formation

封面

如何引用文章

全文:

详细

The article is devoted to premetastatic niche as a complex term, including stromal cells, vessels, extracellular matrix and their changes during interaction with the primary tumor. On example of different malignant tumors authors describe as primary tumor through tumor exosomes prepares certain organs-recipients to metastatic clone implantation. In the area of premetastatic niche under the influence of tumorous exosomes polarization of macrophages towards M2 type takes place. The cells are the main agents, providing survival as well as migration of tumorous cells. Affecting extracellular matrix, macrophages change the microcirculatory bed permeability. This mechanism is directed towards increase of its permeability to entrance of metastatic clone cells form vessels into premetastatic niche. Besides macrophages fibroblasts and polypotent bone marrow stem cells are also reprogrammed, that results in metabolism and local immunity changes at the place of future implantation. As a result, only when tissue of recipient-organ is prepared for contact with metastatic clone, their interaction take place with consequent formation of secondary tumor – metastatic niche. Thus, this review describes pathogenesis of metastasis, different from its early understanding as spread of metastatic clone with lymph and blood.

These peculiarities may in future have significant impact in practical medicine, Blockage of signal spread from primary tumor through exosomes is one of the promising directions in pathogenetic therapy of malignant tumors. Investigation of principles of premetastatic niche formation may become a theoretical substantiation for prophylaxis of metastatic disease and inhibition of micrometastasis to macrometastasis transformation.

作者简介

Yulia Korneva

Smolensk State Medical University; Smolensk Regional Institute of Pathology

编辑信件的主要联系方式.
Email: ksu1546@yandex.ru
ORCID iD: 0000-0002-8080-904X

Cand. Sci. (Med.), Smolensk State Medical University, Smolensk Regional Institute of Pathology

俄罗斯联邦, Smolensk

Roman Ukrainets

Smolensk State Medical University; Smolensk Regional Institute of Pathology

Email: ukrainets.roman@yandex.ru
ORCID iD: 0000-0002-0590-1399

Аssistant, Smolensk State Medical University, Smolensk Regional Institute of Pathology

俄罗斯联邦, Smolensk

参考

  1. Doglioni G, Parik S, Fendt SM. Interactions in the (Pre)metastatic Niche Support Metastasis Formation. Front Oncol 2019; 9: 219. doi: 10.3389/fonc.2019.00219
  2. Medeiros B, Allan AL. Molecular Mechanisms of Breast Cancer Metastasis to the Lung: Clinical and Experimental Perspectives. Int J Mol Sci 2019; 20 (9): pii: E2272. doi: 10.3390/ijms20092272
  3. Wortzel I, Dror S, Kenific CM, Lyden D. Exosome-Mediated Metastasis: Communication from a Distance. Dev Cell 2019; 49 (3): 347–60. doi: 10.1016/j.devcel.2019.04.011
  4. Cheung KJ, Ewald AJ. A collective route to metastasis: seeding by tumor cell clusters. Science 2016; 352: 167. doi: 10.1126/science.aaf6546
  5. Peinado H, Zhang H, Matei IR et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat Rev Cancer 2017; 17: 302–17.
  6. Yousefi M, Nosrati R, Salmaninejad A et al. Organ-specific metastasis of breast cancer: molecular and cellular mechanisms underlying lung metastasis. Cell Oncol (Dordr) 2018; 41 (2): 123–40. doi: 10.1007/s13402-018-0376-6
  7. Ewing J. Neoplastic Diseases: A Textbook on Tumors. Philadelphia, W.B.Saunders, London, 1919.
  8. Paget S. The distribution of secondary growths in cancer of the breast. Lancet 1889; 1: 571–3.
  9. Akhtar M, Haider A, Rashid S, Al-Nabet ADMH. Paget’s «Seed and Soil» Theory of Cancer Metastasis: An Idea Whose Time has Come. Adv Anat Pathol 2019; 26 (1): 69–74. doi: 10.1097/PAP. 0000000000000219
  10. Elia I, Broekaert D, Christen S et al. Proline metabolism supports metastasis formation and could be inhibited to selectively target metastasizing cancer cells. Nat Commun 2017; 8: 15267. doi: 10.1038/ncomms15267
  11. Elia I, Rossi M, Stegen S et al. Breast cancer cells rely on environmental pyruvate to shape the metastatic niche. Nature 2019; 568 (7750): 117–21. doi: 10.1038/s41586-019-0977-x
  12. Fong MY, Zhou W, Liu L et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol 2015; 17: 183–94. doi: 10.1038/ncb3094
  13. Nieman KM, Kenny HA, Penicka CV et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat Med 2011; 17: 1498–503. doi: 10.1038/nm.2492
  14. Perelmuter VM, Manskikh VN. Preniche as missing link of the metastatic niche concept explaining organ-preferential metastasis of malignant tumors and the type of metastatic disease. Biochemistry (Mosc) 2012; 77 (1): 111–8. doi: 10.1134/S000 6297912010142
  15. Tung KH, Ernstoff MS, Allen C, Shu S. A Review of Exosomes and their Role in The Tumor Microenvironment and Host-Tumor «Macroenvironment». J Immunol Sci 2019; 3 (1): 4–8. doi: 10.29245/2578-3009/2019/1.1165
  16. Wang HX, Gires O. Tumor-derived extracellular vesicles in breast cancer: From bench to bedside. Cancer Lett 2019; 460: 54–64. doi: 10.1016/j.canlet.2019.06.012
  17. Santos JC, Lima NDS, Sarian LO et al. Exosome-mediated breast cancer chemoresistance via miR-155 transfer. Sci Rep 2018; 8: 829.
  18. Peng J, Wang W, Hua S, Liu L. Roles of Extracellular Vesicles in Metastatic Breast Cancer. Breast Cancer (Auckl) 2018; 12: 1178223418767666. doi: 10.1177/1178223418767666
  19. Wu J, Li H, Xie H et al. The malignant role of exosomes in the communication among colorectal cancer cell, macrophage and microbiome. Carcinogenesis 2019; 40 (5): 601–10. doi: 10.1093/carcin/bgy138
  20. Shao C, Yang F, Miao S et al. Role of hypoxia-induced exosomes in tumor biology. Mol Cancer Res 2018; 17: 120.
  21. Zhou W, Fong MY, Min Y et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014; 25: 501–15.
  22. Paiva AE, Lousado L, Guerra DAP et al. Pericytes in the Premetastatic Niche. Cancer Res 2018; 78 (11): 2779–86. doi: 10.1158/0008-5472.CAN-17-3883
  23. Kai F, Drain AP, Weaver VM. The Extracellular Matrix Modulates the Metastatic Journey. Dev Cell 2019; 49 (3): 332–46. DOI: 10.1016/ j.devcel.2019.03.026
  24. Kaplan RN, Riba RD, Zacharoulis S et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nat Cell Biol 2005; 438: 820–7.
  25. Chow A, Zhou W, Liu L et al. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-B. Sci Rep 2014; 4: 5750. DOI: 10.1038/ srep05750
  26. Wang D, Dubois RN. Immunosuppression associated with chronic inflammation in the tumor microenvironment. CARCIN 2015; 36: 1085–93.
  27. Perelmuter VM, Manskikh VN. The Concept of a Preniche for Localization of Future Metastases. In Tumors of the Central Nervous System; Hayat M., Ed.; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2013; 13, Chapter 11; p. 93–106. https://doi.org/10.1007/978-94-007-7602-9_11
  28. Doglioni G, Parik S, Fendt SM. Interactions in the (Pre)metastatic Niche Support Metastasis Formation. Front Oncol 2019; 9: 219. doi: 10.3389/fonc.2019.00219
  29. Nielsen SR, Schmid MC. Macrophages as key drivers of cancer progression and metastasis. Mediators Inflamm 2017: 9624760. doi: 10.1155/2017/9624760
  30. Wu S, Zheng Q, Xing X et al. Matrix stiffness-upregulated LOXL2 promotes fibronectin production, MMP9 and CXCL12 expression and BMDCs recruitment to assist pre-metastatic niche formation. J Exp Clin Cancer Res 2018; 37: 99. doi: 10.1186/s13046-018-0761-z
  31. Reynaud C, Ferreras L, Di Mauro P et al. Lysyl oxidase is a strong determinant of tumor cell colonization in bone. Cancer Res 2017; 77: 268. doi: 10.1158/0008-5472.CAN-15-2621
  32. Sun B, Zhou Y, Fang Y et al. Colorectal cancer exosomes induce lymphatic network remodeling in lymph nodes. Int J Cancer 2019; 145 (6): 1648–59. doi: 10.1002/ijc.32196
  33. Costa-Silva B, Aiello NM, Ocean AJ et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 2015; 17: 816–26. doi: 10.1038/ncb3169
  34. Kim H, Chung H, Kim J et al. Macrophages-Triggered Sequential Remodeling of Endothelium-Interstitial Matrix to Form Pre-Metastatic Niche in Microfluidic Tumor Microenvironment. Adv Sci (Weinh) 2019; 6 (11): 1900195. doi: 10.1002/advs.201900195
  35. Ogiya R, Niikura N, Kumaki N et al. Comparison of tumor-infiltrating lymphocytes between primary and metastatic tumors in breast cancer patients. Cancer Sci 2016; 107: 1730–5. doi: 10.1111/cas.13101
  36. Domenis R, Cesselli D, Toffoletto B et al. Systemic T Cells Immunosuppression of Glioma Stem Cell-Derived Exosomes Is Mediated by Monocytic Myeloid-Derived Suppressor Cells. PLoS ONE 2017; 12 (1): e0169932.
  37. Xiang X, Poliakov A, Liu C et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer 2009; 124: 2621–33.
  38. Wculek SK, Malanchi I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 2015; 528: 413–7. doi: 10.1038/nature16140
  39. Lunt SY, Fendt S-M. Metabolism – A cornerstone of cancer initiation, progression, immune evasion and treatment response. Curr Opin Syst Biol 2018; 8: 67–72. doi: 10.1016/j.coisb.2017.12.006
  40. Shu S, Yang Y, Allen CL et al. Metabolic reprogramming of stromal fibroblasts by melanoma exosome microRNA favours a pre-metastatic microenvironment. Sci Rep 2018; 8 (1): 12905. doi: 10.1038/s41598-018-31323-7
  41. Morrot A, da Fonseca LM, Salustiano EJ et al. Metabolic symbiosis and immunomodulation: how tumor cell-derived lactate may disturb innate and adaptive immune responses. Front Oncol 2018; 8: 81. doi: 10.3389/fonc.2018.00081
  42. Peinado H, Alečković M, Lavotshkin S et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nature Medicine 2012; 18 (6): 883–91.
  43. Yang Y, Li CW, Chan LC et al. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res 2018; 28: 862–4.
  44. Nan X, Wang J, Liu HN et al. Epithelial-Mesenchymal Plasticity in Organotropism Metastasis and Tumor Immune Escape. J Clin Med 2019; 8 (5). pii: E747. doi: 10.3390/jcm8050747
  45. Bu P, Chen KY, Xiang K et al. Aldolase B-mediated fructose metabolism drives metabolic reprogramming of colon cancer liver metastasis. Cell Metab 2018; 27: 1249–1262.e4. doi: 10.1016/j.cmet.2018.04.003
  46. Chen G, Huang AC, Zhang W et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018; 560 (7718): 382–6.
  47. Lee JHJ, Lyle M, Menzies AM et al. Metastasis specific patterns of response and progression with anti-PD-1 treatment in metastatic melanoma. Pigment Cell Melanoma Res 2018; 31: 404–10. doi: 10.1111/pcmr.12675
  48. Chen G, Huang AC, Zhang W et al. Exosomal PD-L1 Contributes to Immunosuppression and is Associated with anti-PD-1 Response. Nat Cell Biol 2018; 560: 382–6.
  49. Chen C, Yao X, Xu Y et al. Dahuang Zhechong Pill suppresses colorectal cancer liver metastasis via ameliorating exosomal CCL2 primed pre-metastatic niche. J Ethnopharmacol 2019; 238: 111878. doi: 10.1016/j.jep.2019.111878

版权所有 © Consilium Medicum, 2019

Creative Commons License
此作品已接受知识共享署名-非商业性使用 4.0国际许可协议的许可。
 


##common.cookie##