References:
1. Zhu, R., et al., Current progress in cancer treatment using nanomaterials. Frontiers in Oncology, 2022. 12 .
2. Cheng, Z., et al., Nanomaterials for cancer therapy: Current progress and perspectives. Journal of hematology & oncology, 2021.14 (1): p. 1-27.
3. Sun, G., et al., Role of small molecule targeted compounds in cancer: progress, opportunities, and challenges. Frontiers in cell and developmental biology, 2021. 9 : p. 694363.
4. Zhong, L., et al., Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal transduction and targeted therapy, 2021. 6 (1): p. 201.
5. Tan, S., et al., Exosomal miRNAs in tumor microenvironment.Journal of Experimental & Clinical Cancer Research, 2020. 39 : p. 1-15.
6. Zhong, S., et al., Targeting tumor microenvironment by small-molecule inhibitors. Translational oncology, 2020.13 (1): p. 57-69.
7. Lucero, R., et al., Glioma-derived miRNA-containing extracellular vesicles induce angiogenesis by reprogramming brain endothelial cells. Cell reports, 2020. 30 (7): p. 2065-2074. e4.
8. Varricchi, G., et al., Innate effector cells in angiogenesis and lymphangiogenesis. Current opinion in immunology, 2018.53 : p. 152-160.
9. Petrova, V., et al., The hypoxic tumour microenvironment.Oncogenesis, 2018. 7 (1): p. 10.
10. Zeltz, C., et al. Cancer-associated fibroblasts in desmoplastic tumors: emerging role of integrins . in Seminars in cancer biology . 2020. Elsevier.
11. Ma, L., et al., Tumor cell biodiversity drives microenvironmental reprogramming in liver cancer. Cancer cell, 2019.36 (4): p. 418-430. e6.
12. Zhang, X., et al., Inhibiting PI3 kinase-γ in both myeloid and plasma cells remodels the suppressive tumor microenvironment in desmoplastic tumors. Journal of Controlled Release, 2019. 309 : p. 173-180.
13. Clément-Colmou, K., et al., Influence of radiotherapy fractionation schedule on the tumor vascular microenvironment in prostate and lung cancer models. Cancers, 2020. 12 (1): p. 121.
14. Armignacco, R., et al., The adipose stem cell as a novel metabolic actor in adrenocortical carcinoma progression: Evidence from an in vitro tumor microenvironment crosstalk model. Cancers, 2019.11 (12): p. 1931.
15. Lei, X., et al., Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer letters, 2020. 470 : p. 126-133.
16. Zhang, H., et al., CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Molecular cancer, 2020. 19 : p. 1-17.
17. De Palma, M., D. Biziato, and T.V. Petrova, Microenvironmental regulation of tumour angiogenesis. Nature Reviews Cancer, 2017.17 (8): p. 457-474.
18. Moriwaki, K. and M. Asahi, Augmented TME O-GlcNAcylation Promotes Tumor Proliferation through the Inhibition of p38 MAPKEffect of O-GlcNAcylation in the TME. Molecular Cancer Research, 2017.15 (9): p. 1287-1298.
19. Khalaf, K., et al., Aspects of the tumor microenvironment involved in immune resistance and drug resistance. Frontiers in immunology, 2021. 12 : p. 656364.
20. Whiteside, T.L. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment . in Seminars in immunology . 2018. Elsevier.
21. Xie, F., et al., Extracellular vesicles in cancer immune microenvironment and cancer immunotherapy. Advanced science, 2019.6 (24): p. 1901779.
22. Daassi, D., K.M. Mahoney, and G.J. Freeman, The importance of exosomal PDL1 in tumour immune evasion. Nature Reviews Immunology, 2020. 20 (4): p. 209-215.
23. Jarosz-Biej, M., et al., Tumor microenvironment as a “game changer” in cancer radiotherapy. International journal of molecular sciences, 2019. 20 (13): p. 3212.
24. Peng, J., et al., Intratumoral fate of functional nanoparticles in response to microenvironment factor: Implications on cancer diagnosis and therapy. Advanced Drug Delivery Reviews, 2019.143 : p. 37-67.
25. Maacha, S., et al., Extracellular vesicles-mediated intercellular communication: roles in the tumor microenvironment and anti-cancer drug resistance. Molecular cancer, 2019. 18 : p. 1-16.
26. Sullivan, R., et al., The emerging roles of extracellular vesicles as communication vehicles within the tumor microenvironment and beyond. Frontiers in Endocrinology, 2017. 8 : p. 194.
27. Van Niel, G., G. d’Angelo, and G. Raposo, Shedding light on the cell biology of extracellular vesicles. Nature reviews Molecular cell biology, 2018. 19 (4): p. 213-228.
28. Minciacchi, V.R., M.R. Freeman, and D. Di Vizio. Extracellular vesicles in cancer: exosomes, microvesicles and the emerging role of large oncosomes . in Seminars in cell & developmental biology . 2015. Elsevier.
29. Maas, S.L., X.O. Breakefield, and A.M. Weaver, Extracellular vesicles: unique intercellular delivery vehicles. Trends in cell biology, 2017. 27 (3): p. 172-188.
30. Bebelman, M.P., et al., Biogenesis and function of extracellular vesicles in cancer. Pharmacology & therapeutics, 2018.188 : p. 1-11.
31. Tovar-Camargo, O.A., S. Toden, and A. Goel, Exosomal microRNA biomarkers: emerging frontiers in colorectal and other human cancers.Expert review of molecular diagnostics, 2016. 16 (5): p. 553-567.
32. Higginbotham, J.N., et al., Amphiregulin exosomes increase cancer cell invasion. Current Biology, 2011. 21 (9): p. 779-786.
33. Rak, J. and A. Guha, Extracellular vesicles–vehicles that spread cancer genes. Bioessays, 2012. 34 (6): p. 489-497.
34. Naito, Y., et al., How cancer cells dictate their microenvironment: present roles of extracellular vesicles. Cellular and Molecular Life Sciences, 2017. 74 : p. 697-713.
35. Fu, H., et al., The emerging roles of exosomes in tumor–stroma interaction. Journal of cancer research and clinical oncology, 2016. 142 : p. 1897-1907.
36. Santos, P. and F. Almeida, Role of exosomal miRNAs and the tumor microenvironment in drug resistance. Cells, 2020. 9 (6): p. 1450.
37. Xu, R., et al., Extracellular vesicles in cancer—implications for future improvements in cancer care. Nature reviews Clinical oncology, 2018. 15 (10): p. 617-638.
38. Li, I. and B.Y. Nabet, Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Molecular cancer, 2019.18 : p. 1-10.
39. Gangoda, L., et al., Extracellular vesicles including exosomes are mediators of signal transduction: are they protective or pathogenic? Proteomics, 2015. 15 (2-3): p. 260-271.
40. Steinbichler, T.B., et al., Therapy resistance mediated by exosomes. Molecular cancer, 2019. 18 (1): p. 1-11.
41. Mashouri, L., et al., Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Molecular cancer, 2019. 18 : p. 1-14.
42. Milman, N., L. Ginini, and Z. Gil, Exosomes and their role in tumorigenesis and anticancer drug resistance. Drug Resistance Updates, 2019. 45 : p. 1-12.
43. Anfossi, S., et al., MicroRNAs, regulatory messengers inside and outside cancer cells. Exosomes, Stem Cells and MicroRNA: Aging, Cancer and Age Related Disorders, 2018: p. 87-108.
44. Fattore, L., et al., Reprogramming miRNAs global expression orchestrates development of drug resistance in BRAF mutated melanoma.Cell Death & Differentiation, 2019. 26 (7): p. 1267-1282.
45. Malhotra, A., et al., Stabilization of miRNAs in esophageal cancer contributes to radioresistance and limits efficacy of therapy.Biochimie, 2019. 156 : p. 148-157.
46. Yan, W., et al., Cancer-cell-secreted exosomal miR-105 promotes tumour growth through the MYC-dependent metabolic reprogramming of stromal cells. Nature cell biology, 2018. 20 (5): p. 597-609.
47. Zhou, C.-F., et al., Cervical squamous cell carcinoma-secreted exosomal miR-221-3p promotes lymphangiogenesis and lymphatic metastasis by targeting VASH1. Oncogene, 2019. 38 (8): p. 1256-1268.
48. Cai, L., et al., Epstein–Barr virus-encoded microRNA BART1 induces tumour metastasis by regulating PTEN-dependent pathways in nasopharyngeal carcinoma. Nature communications, 2015. 6 (1): p. 7353.
49. Tian, R., et al., Differential expression of miR16 in glioblastoma and glioblastoma stem cells: their correlation with proliferation, differentiation, metastasis and prognosis. Oncogene, 2017. 36 (42): p. 5861-5873.
50. Uddin, M.N., M. Li, and X. Wang, Identification of Transcriptional Markers and microRNA–mRNA Regulatory Networks in Colon Cancer by Integrative Analysis of mRNA and microRNA Expression Profiles in Colon Tumor Stroma. Cells, 2019. 8 (9): p. 1054.
51. Slack, F.J. and A.M. Chinnaiyan, The role of non-coding RNAs in oncology. Cell, 2019. 179 (5): p. 1033-1055.
52. Kanchan, R.K., et al., microRNAs orchestrate pathophysiology of breast cancer brain metastasis: advances in therapy. Molecular cancer, 2020. 19 (1): p. 1-16.
53. Conti, I., et al., miRNAs as influencers of cell–cell communication in tumor microenvironment. Cells, 2020. 9 (1): p. 220.
54. Cheng, W.C., et al., RAB27B‐activated secretion of stem‐like tumor exosomes delivers the biomarker microRNA‐146a‐5p, which promotes tumorigenesis and associates with an immunosuppressive tumor microenvironment in colorectal cancer. International Journal of Cancer, 2019. 145 (8): p. 2209-2224.
55. Patel, H., et al., Modulating secreted components of tumor microenvironment: A masterstroke in tumor therapeutics. Cancer Biology & Therapy, 2018. 19 (1): p. 3-12.
56. Wang, M., et al., Emerging function and clinical values of exosomal microRNAs in cancer. Molecular therapy-Nucleic acids, 2019.16 : p. 791-804.
57. Sun, Z., et al., Effect of exosomal miRNA on cancer biology and clinical applications. Molecular cancer, 2018. 17 : p. 1-19.
58. Pontecorvi, G., et al., Tumor-derived extracellular vesicles and microRNAs: Functional roles, diagnostic, prognostic and therapeutic options. Cytokine & Growth Factor Reviews, 2020. 51 : p. 75-83.
59. Bach, D.H., et al., The role of exosomes and miRNAs in drug‐resistance of cancer cells. International journal of cancer, 2017.141 (2): p. 220-230.
60. Seo, H.A., et al., Microrna-based combinatorial cancer therapy: Effects of micrornas on the efficacy of anti-cancer therapies.Cells, 2019. 9 (1): p. 29.
61. Sharma, A., Chemoresistance in cancer cells: exosomes as potential regulators of therapeutic tumor heterogeneity. Nanomedicine, 2017. 12 (17): p. 2137-2148.
62. Au Yeung, C.L., et al., Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nature communications, 2016. 7 (1): p. 11150.
63. Chen, Y., et al., Tumor-associated macrophages: an accomplice in solid tumor progression. Journal of biomedical science, 2019.26 (1): p. 1-13.
64. Netea-Maier, R.T., J.W. Smit, and M.G. Netea, Metabolic changes in tumor cells and tumor-associated macrophages: a mutual relationship. Cancer letters, 2018. 413 : p. 102-109.
65. Sawa-Wejksza, K. and M. Kandefer-Szerszeń, Tumor-associated macrophages as target for antitumor therapy. Archivum immunologiae et therapiae experimentalis, 2018. 66 : p. 97-111.
66. Park, J.E., et al., Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene, 2019. 38 (26): p. 5158-5173.
67. Cooks, T., et al., Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nature communications, 2018. 9 (1): p. 771.
68. Hsieh, C.-H., S.-K. Tai, and M.-H. Yang, Snail-overexpressing cancer cells promote M2-like polarization of tumor-associated macrophages by delivering MiR-21-abundant exosomes. Neoplasia, 2018.20 (8): p. 775-788.
69. Lin, F., et al., Bladder cancer cell‑secreted exosomal miR‑21 activates the PI3K/AKT pathway in macrophages to promote cancer progression. International journal of oncology, 2020. 56 (1): p. 151-164.
70. Chen, X., et al., Exosomes derived from hypoxic epithelial ovarian cancer deliver microRNA-940 to induce macrophage M2 polarization. Oncology Reports, 2017. 38 (1): p. 522-528.
71. Chen, X., et al., Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer letters, 2018. 435 : p. 80-91.
72. Nieto, M.A., Epithelial plasticity: a common theme in embryonic and cancer cells. Science, 2013. 342 (6159): p. 1234850.
73. Gavert, N. and A. Ben-Ze’ev, Epithelial–mesenchymal transition and the invasive potential of tumors. Trends in molecular medicine, 2008. 14 (5): p. 199-209.
74. Du, B. and J.S. Shim, Targeting epithelial–mesenchymal transition (EMT) to overcome drug resistance in cancer. Molecules, 2016. 21 (7): p. 965.
75. Bigagli, E., et al., Transcriptomic characterization, chemosensitivity and regulatory effects of exosomes in spontaneous EMT/MET transitions of breast cancer cells. Cancer Genomics & Proteomics, 2019. 16 (3): p. 163-173.
76. Hardin, H., et al., Thyroid cancer stem-like cell exosomes: regulation of EMT via transfer of lncRNAs. Laboratory Investigation, 2018. 98 (9): p. 1133-1142.
77. Xiao, D., et al., Melanoma cell–derived exosomes promote epithelial–mesenchymal transition in primary melanocytes through paracrine/autocrine signaling in the tumor microenvironment. Cancer letters, 2016. 376 (2): p. 318-327.
78. Mizushima, N. and M. Komatsu, Autophagy: renovation of cells and tissues. Cell, 2011. 147 (4): p. 728-741.
79. Huang, F., B.-R. Wang, and Y.-G. Wang, Role of autophagy in tumorigenesis, metastasis, targeted therapy and drug resistance of hepatocellular carcinoma. World journal of gastroenterology, 2018.24 (41): p. 4643.
80. Folkerts, H., et al., The multifaceted role of autophagy in cancer and the microenvironment. Medicinal research reviews, 2019.39 (2): p. 517-560.
81. Janji, B., G. Berchem, and S. Chouaib, Targeting autophagy in the tumor microenvironment: new challenges and opportunities for regulating tumor immunity. Frontiers in Immunology, 2018. 9 : p. 887.
82. Gewirtz, D.A., Cytoprotective and nonprotective autophagy in cancer therapy . 2013, Taylor & Francis. p. 1263-1265.
83. Dutta, S., et al., Interactions between exosomes from breast cancer cells and primary mammary epithelial cells leads to generation of reactive oxygen species which induce DNA damage response, stabilization of p53 and autophagy in epithelial cells. PloS one, 2014.9 (5): p. e97580.
84. Liu, D.X., et al., Exosomes derived from HBV‑associated liver cancer promote chemoresistance by upregulating chaperone‑mediated autophagy. Oncology Letters, 2019. 17 (1): p. 323-331.
85. Qu, Y., et al., Exosomes derived from miR‐181‐5p‐modified adipose‐derived mesenchymal stem cells prevent liver fibrosis via autophagy activation. Journal of cellular and molecular medicine, 2017.21 (10): p. 2491-2502.
86. Chen, J., et al., Micro RNA‐30a ameliorates hepatic fibrosis by inhibiting Beclin1‐mediated autophagy. Journal of Cellular and Molecular Medicine, 2017. 21 (12): p. 3679-3692.
87. Yuwen, D., et al., Prognostic role of circulating exosomal miR-425-3p for the response of NSCLC to platinum-based chemotherapy.Cancer Epidemiology, Biomarkers & Prevention, 2019. 28 (1): p. 163-173.
88. Pathania, A.S. and K.B. Challagundla, Exosomal long non-coding RNAs: emerging players in the tumor microenvironment. Molecular Therapy-Nucleic Acids, 2021. 23 : p. 1371-1383.
89. Zhang, Z., et al., Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma.Journal of Experimental & Clinical Cancer Research, 2019.38 (1): p. 1-16.
90. Masoud, G.N. and W. Li, HIF-1α pathway: role, regulation and intervention for cancer therapy. Acta Pharmaceutica Sinica B, 2015.5 (5): p. 378-389.
91. Takahashi, K., et al., Modulation of hypoxia-signaling pathways by extracellular linc-RoR. Journal of cell science, 2014.127 (7): p. 1585-1594.
92. Xue, M., et al., Hypoxic exosomes facilitate bladder tumor growth and development through transferring long non-coding RNA-UCA1.Molecular cancer, 2017. 16 : p. 1-13.
93. Sun, K., et al., Paradoxical roles of autophagy in different stages of tumorigenesis: protector for normal or cancer cells. Cell & bioscience, 2013. 3 (1): p. 1-8.
94. Ding, L., et al., A novel stromal lncRNA signature reprograms fibroblasts to promote the growth of oral squamous cell carcinoma via LncRNA-CAF/interleukin-33. Carcinogenesis, 2018. 39 (3): p. 397-406.
95. Warburg, O., F. Wind, and E. Negelein, The metabolism of tumors in the body. The Journal of general physiology, 1927.8 (6): p. 519.
96. Liberti, M.V. and J.W. Locasale, The Warburg effect: how does it benefit cancer cells? Trends in biochemical sciences, 2016.41 (3): p. 211-218.
97. Papa, S., P.M. Choy, and C. Bubici, The ERK and JNK pathways in the regulation of metabolic reprogramming. Oncogene, 2019.38 (13): p. 2223-2240.
98. Baghban, R., et al., Tumor microenvironment complexity and therapeutic implications at a glance. Cell Communication and Signaling, 2020. 18 : p. 1-19.
99. Liu, T., et al., Cancer-associated fibroblasts build and secure the tumor microenvironment. Frontiers in cell and developmental biology, 2019. 7 : p. 60.
100. Walker, C., E. Mojares, and A. del Río Hernández, Role of extracellular matrix in development and cancer progression.International journal of molecular sciences, 2018. 19 (10): p. 3028.
101. Lu, P., V.M. Weaver, and Z. Werb, The extracellular matrix: a dynamic niche in cancer progression. Journal of cell biology, 2012.196 (4): p. 395-406.
102. Senthebane, D.A., et al., The role of tumor microenvironment in chemoresistance: to survive, keep your enemies closer. International journal of molecular sciences, 2017. 18 (7): p. 1586.
103. Page-McCaw, A., A.J. Ewald, and Z. Werb, Matrix metalloproteinases and the regulation of tissue remodelling. Nature reviews Molecular cell biology, 2007. 8 (3): p. 221-233.
104. Cox, T.R. and J.T. Erler, Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer.Disease models & mechanisms, 2011. 4 (2): p. 165-178.
105. Theocharis, A.D., et al., Extracellular matrix structure.Advanced drug delivery reviews, 2016. 97 : p. 4-27.
106. Wei, R., et al., Cellular and extracellular components in tumor microenvironment and their application in early diagnosis of cancers. Analytical Cellular Pathology, 2020. 2020 .
107. Clause, K.C. and T.H. Barker, Extracellular matrix signaling in morphogenesis and repair. Current opinion in biotechnology, 2013.24 (5): p. 830-833.
108. Tan, T.-T. and L.M. Coussens, Humoral immunity, inflammation and cancer. Current opinion in immunology, 2007. 19 (2): p. 209-216.
109. Liu, H., H. Zhao, and Y. Sun. Tumor microenvironment and cellular senescence: Understanding therapeutic resistance and harnessing strategies . in Seminars in Cancer Biology . 2022. Elsevier.
110. Schuster, R., et al., The inflammatory speech of fibroblasts. Immunological reviews, 2021. 302 (1): p. 126-146.
111. Wu, P., et al., Adaptive mechanisms of tumor therapy resistance driven by tumor microenvironment. Frontiers in cell and developmental biology, 2021. 9 : p. 641469.
112. Ni, Y., et al., The role of tumor-stroma interactions in drug resistance within tumor microenvironment. Frontiers in Cell and Developmental Biology, 2021. 9 : p. 637675.
113. Morin, P.J., Drug resistance and the microenvironment: nature and nurture. Drug Resistance Updates, 2003. 6 (4): p. 169-172.
114. Chen, Y., et al., Therapeutic remodeling of the tumor microenvironment enhances nanoparticle delivery. Advanced Science, 2019. 6 (5): p. 1802070.
115. Jo, Y., et al., Chemoresistance of cancer cells: requirements of tumor microenvironment-mimicking in vitro models in anti-cancer drug development. Theranostics, 2018. 8 (19): p. 5259.
116. Xu, S., et al., The role of collagen in cancer: from bench to bedside. Journal of translational medicine, 2019. 17 : p. 1-22.
117. Natarajan, S., et al., Collagen remodeling in the hypoxic tumor-mesothelial niche promotes ovarian cancer metastasis. Cancer research, 2019. 79 (9): p. 2271-2284.
118. Naito, Y., et al., Micro RNA‐143 regulates collagen type III expression in stromal fibroblasts of scirrhous type gastric cancer.Cancer science, 2014. 105 (2): p. 228-235.
119. Mu, W., S. Rana, and M. Zöller, Host matrix modulation by tumor exosomes promotes motility and invasiveness. Neoplasia, 2013.15 (8): p. 875-IN4.
120. Mathot, P., et al., DNA methylation signal has a major role in the response of human breast cancer cells to the microenvironment.Oncogenesis, 2017. 6 (10): p. e390-e390.
121. Roodhart, J.M., et al., Mesenchymal stem cells induce resistance to chemotherapy through the release of platinum-induced fatty acids. Cancer cell, 2011. 20 (3): p. 370-383.
122. Han, Z., et al., Mesenchymal stem cells contribute to the chemoresistance of hepatocellular carcinoma cells in inflammatory environment by inducing autophagy. Cell & bioscience, 2014.4 (1): p. 1-11.
123. Ji, R., et al., Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell cycle, 2015.14 (15): p. 2473-2483.
124. Xu, H., et al., Tumor‑derived mesenchymal‑stem‑cell‑secreted IL‑6 enhances resistance to cisplatin via the STAT3 pathway in breast cancer. Oncology Letters, 2018. 15 (6): p. 9142-9150.
125. Lu, M., et al., Notoginsenoside R1 counteracts mesenchymal stem cell-evoked oncogenesis and doxorubicin resistance in osteosarcoma cells by blocking IL-6 secretion-induced JAK2/STAT3 signaling.Investigational New Drugs, 2021. 39 : p. 416-425.
126. Raghavan, S., et al., Carcinoma-associated mesenchymal stem cells promote chemoresistance in ovarian cancer stem cells via PDGF signaling. Cancers, 2020. 12 (8): p. 2063.
127. Wang, S., et al., The CXCR4 antagonist, AMD3100, reverses mesenchymal stem cell-mediated drug resistance in relapsed/refractory acute lymphoblastic leukemia. OncoTargets and therapy, 2020.13 : p. 6583.
128. Ridge, S.M., F.J. Sullivan, and S.A. Glynn, Mesenchymal stem cells: key players in cancer progression. Molecular cancer, 2017.16 (1): p. 1-10.
129. Wang, J., et al., Cell adhesion-mediated mitochondria transfer contributes to mesenchymal stem cell-induced chemoresistance on T cell acute lymphoblastic leukemia cells. Journal of Hematology & Oncology, 2018. 11 (1): p. 1-13.
130. Shi, Y., et al., Tumour-associated mesenchymal stem/stromal cells: emerging therapeutic targets. Nature reviews Drug discovery, 2017. 16 (1): p. 35-52.
131. Scherzed, A., et al., BMSC enhance the survival of paclitaxel treated squamous cell carcinoma cells in vitro. Cancer Biology & Therapy, 2011. 11 (3): p. 349-357.
132. Luo, J., et al., Infiltrating bone marrow mesenchymal stem cells increase prostate cancer stem cell population and metastatic ability via secreting cytokines to suppress androgen receptor signaling. Oncogene, 2014. 33 (21): p. 2768-2778.
133. Adamo, A., et al., Role of mesenchymal stromal cell-derived extracellular vesicles in tumour microenvironment. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer, 2019. 1871 (1): p. 192-198.
134. Wu, Q., et al., Unraveling adipocytes and Cancer links: is there a role for senescence? Frontiers in cell and developmental biology, 2020. 8 : p. 282.
135. Cao, Y., Adipocyte and lipid metabolism in cancer drug resistance. The Journal of clinical investigation, 2019.129 (8): p. 3006-3017.
136. Nieman, K.M., et al., Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids, 2013. 1831 (10): p. 1533-1541.
137. Deng, T., et al., Obesity, inflammation, and cancer. Annual Review of Pathology: Mechanisms of Disease, 2016. 11 : p. 421-449.
138. Elaraby, E., et al., Natural Killer Cell Dysfunction in Obese Patients with Breast Cancer: A Review of a Triad and Its Implications.Journal of Immunology Research, 2021. 2021 .
139. Duong, M.N., et al., The fat and the bad: Mature adipocytes, key actors in tumor progression and resistance. Oncotarget, 2017.8 (34): p. 57622.
140. Yu, W., et al., Adipocytes secreted leptin is a pro-tumor factor for survival of multiple myeloma under chemotherapy. Oncotarget, 2016. 7 (52): p. 86075.
141. Satoh, M., et al., Modulation of resistance to anticancer drugs by inhibition of metallothionein synthesis. Cancer research, 1994. 54 (20): p. 5255-5257.
142. Choi, J., Y.J. Cha, and J.S. Koo, Adipocyte biology in breast cancer: From silent bystander to active facilitator. Progress in lipid research, 2018. 69 : p. 11-20.
143. Iyengar, P., et al., Adipocyte-derived collagen VI affects early mammary tumor progression in vivo, demonstrating a critical interaction in the tumor/stroma microenvironment. The Journal of clinical investigation, 2005. 115 (5): p. 1163-1176.
144. Sherman-Baust, C.A., et al., Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells. Cancer cell, 2003. 3 (4): p. 377-386.
145. Pérez de Heredia, F., I.S. Wood, and P. Trayhurn, Hypoxia stimulates lactate release and modulates monocarboxylate transporter (MCT1, MCT2, and MCT4) expression in human adipocytes. Pflügers Archiv-European Journal of Physiology, 2010. 459 : p. 509-518.
146. Yang, E., et al., Exosome-mediated metabolic reprogramming: the emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal transduction and targeted therapy, 2020.5 (1): p. 242.
147. Sheng, X., et al., Adipocytes sequester and metabolize the chemotherapeutic daunorubicin. Molecular Cancer Research, 2017.15 (12): p. 1704-1713.
148. Arneth, B., Tumor microenvironment. Medicina, 2019.56 (1): p. 15.
149. Hulikova, A. and P. Swietach, Rapid CO2 permeation across biological membranes: implications for CO2 venting from tissue. The FASEB Journal, 2014. 28 (7): p. 2762-2774.
150. Vaupel, P. and A. Mayer, Hypoxia in tumors: pathogenesis-related classification, characterization of hypoxia subtypes, and associated biological and clinical implications. Oxygen transport to tissue XXXVI, 2014: p. 19-24.
151. Rohani, N., et al., Acidification of tumor at stromal boundaries drives transcriptome alterations associated with aggressive phenotypes. Cancer research, 2019. 79 (8): p. 1952-1966.
152. Peppicelli, S., et al., The acidic microenvironment as a possible niche of dormant tumor cells. Cellular and molecular life sciences, 2017. 74 : p. 2761-2771.
153. Hanahan, D. and R.A. Weinberg, Hallmarks of cancer: the next generation. cell, 2011. 144 (5): p. 646-674.
154. Siska, P.J., et al., The immunological Warburg effect: Can a metabolic‐tumor‐stroma score (MeTS) guide cancer immunotherapy?Immunological reviews, 2020. 295 (1): p. 187-202.
155. Apicella, M., et al., Increased lactate secretion by cancer cells sustains non-cell-autonomous adaptive resistance to MET and EGFR targeted therapies. Cell metabolism, 2018. 28 (6): p. 848-865. e6.
156. Yoshida, G.J., Metabolic reprogramming: the emerging concept and associated therapeutic strategies. Journal of experimental & clinical cancer research, 2015. 34 : p. 1-10.
157. Martinez-Outschoorn, U.E., et al., Cancer metabolism: a therapeutic perspective. Nature reviews Clinical oncology, 2017.14 (1): p. 11-31.
158. Gupta, S., A. Roy, and B.S. Dwarakanath, Metabolic cooperation and competition in the tumor microenvironment: implications for therapy. Frontiers in oncology, 2017. 7 : p. 68.
159. Wang, Q., et al., Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Medicine, 2023.
160. Libutti, S.K., L. Tamarkin, and N. Nilubol, Targeting the invincible barrier for drug delivery in solid cancers: interstitial fluid pressure. Oncotarget, 2018. 9 (87): p. 35723.
161. Yu, T., et al., High interstitial fluid pressure promotes tumor progression through inducing lymphatic metastasis-related protein expressions in oral squamous cell carcinoma. Clinical and Translational Oncology, 2014. 16 : p. 539-547.
162. Haider, T., et al., Recent advances in tumor microenvironment associated therapeutic strategies and evaluation models. Materials Science and Engineering: C, 2020. 116 : p. 111229.
163. Trédan, O., et al., Drug resistance and the solid tumor microenvironment. Journal of the National Cancer Institute, 2007.99 (19): p. 1441-1454.
164. Chen, F., et al., New horizons in tumor microenvironment biology: challenges and opportunities. BMC medicine, 2015.13 (1): p. 1-14.
165. Wang, S., et al., Tumor microenvironment in chemoresistance, metastasis and immunotherapy of pancreatic cancer. American journal of cancer research, 2020. 10 (7): p. 1937.
166. Romano, A., et al., PMN-MDSC and arginase are increased in myeloma and may contribute to resistance to therapy. Expert review of molecular diagnostics, 2018. 18 (7): p. 675-683.
167. Calcinotto, A., et al., IL-23 secreted by myeloid cells drives castration-resistant prostate cancer. Nature, 2018.559 (7714): p. 363-369.
168. Dzobo, K., D.A. Senthebane, and C. Dandara, The tumor microenvironment in tumorigenesis and therapy resistance revisited.Cancers, 2023. 15 (2): p. 376.
169. Maimela, N.R., S. Liu, and Y. Zhang, Fates of CD8+ T cells in tumor microenvironment. Computational and structural biotechnology journal, 2019. 17 : p. 1-13.
170. Bamias, A., et al., Correlation of NK T-like CD3+ CD56+ cells and CD4+ CD25+ (hi) regulatory T cells with VEGF and TNFα in ascites from advanced ovarian cancer: Association with platinum resistance and prognosis in patients receiving first-line, platinum-based chemotherapy. Gynecologic oncology, 2008. 108 (2): p. 421-427.
171. Long, Y., et al., Dysregulation of glutamate transport enhances treg function that promotes VEGF blockade resistance in glioblastoma. Cancer research, 2020. 80 (3): p. 499-509.
172. Imbert, C., et al., Resistance of melanoma to immune checkpoint inhibitors is overcome by targeting the sphingosine kinase-1. Nature communications, 2020. 11 (1): p. 437.
173. Li, C., et al., Foxp3 overexpression decreases sensitivity to chemotherapy in mouse Lewis lung cancer cells. Molecular medicine reports, 2012. 6 (5): p. 977-982.
174. Wang, D., et al., Colorectal cancer cell-derived CCL20 recruits regulatory T cells to promote chemoresistance via FOXO1/CEBPB/NF-κB signaling. Journal for immunotherapy of cancer, 2019.7 (1): p. 1-15.
175. Schuler, P.J., et al., Effects of Adjuvant Chemoradiotherapy on the Frequency and Function of Regulatory T Cells in Patients with Head and Neck CancerCD4+ CD39+ Treg and CRT. Clinical cancer research, 2013. 19 (23): p. 6585-6596.
176. Liu, X.-D., et al., Resistance to Antiangiogenic Therapy Is Associated with an Immunosuppressive Tumor Microenvironment in Metastatic Renal Cell CarcinomaAntiangiogenic Therapy Increases PD-L1 Expression. Cancer immunology research, 2015. 3 (9): p. 1017-1029.
177. Aspord, C., et al., Breast cancer instructs dendritic cells to prime interleukin 13–secreting CD4+ T cells that facilitate tumor development. The Journal of experimental medicine, 2007.204 (5): p. 1037-1047.
178. Vicari, A.P., C. Caux, and G. Trinchieri. Tumour escape from immune surveillance through dendritic cell inactivation . inSeminars in cancer biology . 2002. Elsevier.
179. Incio, J., et al., Obesity-induced inflammation and desmoplasia promote pancreatic cancer progression and resistance to chemotherapy. Cancer discovery, 2016. 6 (8): p. 852-869.
180. Shaul, M.E. and Z.G. Fridlender, Tumour-associated neutrophils in patients with cancer. Nature reviews Clinical oncology, 2019. 16 (10): p. 601-620.
181. Bui, T.M., L.K. Yalom, and R. Sumagin, Tumor-associated neutrophils: orchestrating cancer pathobiology and therapeutic resistance. Expert opinion on therapeutic targets, 2021.25 (7): p. 573-583.
182. Chong, A.S.-F., et al., Diverse multidrug-resistance-modification agents inhibit cytolytic activity of natural killer cells. Cancer Immunology, Immunotherapy, 1993.36 : p. 133-139.
183. Savas, B., et al., P-glycoprotein-mediated multidrug resistance and cytotoxic effector cells. Nat Immun, 1992.11 (4): p. 177-92.
184. Takahashi, M., et al., Role of P-glycoprotein in human natural killer-like cell line-mediated cytotoxicity. Experimental Cell Research, 1999. 253 (2): p. 396-402.
185. Sugimura, K., et al., High infiltration of tumor‐associated macrophages is associated with a poor response to chemotherapy and poor prognosis of patients undergoing neoadjuvant chemotherapy for esophageal cancer. Journal of surgical oncology, 2015. 111 (6): p. 752-759.
186. Li, X., et al., Harnessing tumor-associated macrophages as aids for cancer immunotherapy. Molecular Cancer, 2019. 18 (1): p. 1-16.
187. Liu, Y. and X. Cao, The origin and function of tumor-associated macrophages. Cellular & molecular immunology, 2015.12 (1): p. 1-4.
188. Moradi-Chaleshtori, M., et al., In vitro and in vivo evaluation of anti-tumoral effect of M1 phenotype induction in macrophages by miR-130 and miR-33 containing exosomes. Cancer Immunology, Immunotherapy, 2021. 70 : p. 1323-1339.
189. Huang, W.-C., et al., Cisplatin resistant lung cancer cells promoted M2 polarization of tumor-associated macrophages via the Src/CD155/MIF functional pathway. Journal of Experimental & Clinical Cancer Research, 2019. 38 (1): p. 1-17.
190. Liu, H., et al., Jagged1 promotes aromatase inhibitor resistance by modulating tumor-associated macrophage differentiation in breast cancer patients. Breast Cancer Research and Treatment, 2017.166 : p. 95-107.
191. Yu, S., et al., Activated HIF1α of tumor cells promotes chemoresistance development via recruiting GDF15-producing tumor-associated macrophages in gastric cancer. Cancer Immunology, Immunotherapy, 2020. 69 : p. 1973-1987.
192. Amit, M. and Z. Gil, Macrophages increase the resistance of pancreatic adenocarcinoma cells to gemcitabine by upregulating cytidine deaminase. Oncoimmunology, 2013. 2 (12): p. e27231.
193. Weizman, N., et al., Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase. Oncogene, 2014. 33 (29): p. 3812-3819.
194. Binenbaum, Y., et al., Transfer of miRNA in Macrophage-Derived Exosomes Induces Drug Resistance in Pancreatic AdenocarcinomaExosomes Induce Gemcitabine Resistance in Pancreatic Cancer. Cancer research, 2018. 78 (18): p. 5287-5299.
195. Xian, G., et al., Simvastatin attenuates macrophage-mediated gemcitabine resistance of pancreatic ductal adenocarcinoma by regulating the TGF-β1/Gfi-1 axis. Cancer letters, 2017. 385 : p. 65-74.
196. Ireland, L., et al., Chemoresistance in Pancreatic Cancer Is Driven by Stroma-Derived Insulin-Like Growth FactorsStroma-Derived IGFs Enhance Chemoresistance in PDAC. Cancer research, 2016.76 (23): p. 6851-6863.
197. Kuwada, K., et al., The epithelial-to-mesenchymal transition induced by tumor-associated macrophages confers chemoresistance in peritoneally disseminated pancreatic cancer. Journal of Experimental & Clinical Cancer Research, 2018. 37 : p. 1-10.
198. Huang, H., et al., Reciprocal Network between Cancer Stem-Like Cells and Macrophages Facilitates the Progression and Androgen Deprivation Therapy Resistance of Prostate CancerCSC-Mø Facilitates the Progression and ADT Resistance of PCa. Clinical cancer research, 2018.24 (18): p. 4612-4626.
199. Torgovnick, A. and B. Schumacher, DNA repair mechanisms in cancer development and therapy. Frontiers in genetics, 2015.6 : p. 157.
200. Wang, M., S. Chen, and D. Ao, Targeting DNA repair pathway in cancer: Mechanisms and clinical application. MedComm, 2021.2 (4): p. 654-691.
201. Kiwerska, K. and K. Szyfter, DNA repair in cancer initiation, progression, and therapy—a double-edged sword. Journal of applied genetics, 2019. 60 (3-4): p. 329-334.
202. Goldstein, M. and M.B. Kastan, The DNA damage response: implications for tumor responses to radiation and chemotherapy. Annual review of medicine, 2015. 66 : p. 129-143.
203. Abbotts, R., N. Thompson, and S. Madhusudan, DNA repair in cancer: emerging targets for personalized therapy. Cancer management and research, 2014: p. 77-92.
204. Huang, R. and P.-K. Zhou, DNA damage repair: Historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduction and Targeted Therapy, 2021.6 (1): p. 254.
205. Blasiak, J., Single-strand annealing in cancer.International Journal of Molecular Sciences, 2021. 22 (4): p. 2167.
206. Jackson, S.P. and J. Bartek, The DNA-damage response in human biology and disease. Nature, 2009. 461 (7267): p. 1071-1078.
207. Jurkovicova, D., et al., DNA Damage Response in Cancer Therapy and Resistance: Challenges and Opportunities. International Journal of Molecular Sciences, 2022. 23 (23): p. 14672.
208. Czerninski, R., et al., Promoter hypermethylation of mismatch repair genes, hMLH1 and hMSH2 in oral squamous cell carcinoma. Oral diseases, 2009. 15 (3): p. 206-213.
209. Guan, H., et al., Hypermethylation of the DNA mismatch repair gene hMLH1 and its association with lymph node metastasis and T1799A BRAF mutation in patients with papillary thyroid cancer. Cancer, 2008.113 (2): p. 247-255.
210. Wang, Y.-C., et al., Inactivation of hMLH1 and hMSH2 by promoter methylation in primary non-small cell lung tumors and matched sputum samples. The Journal of clinical investigation, 2003.111 (6): p. 887-895.
211. Lee, M.-N., et al., Epigenetic inactivation of the chromosomal stability control genes BRCA1, BRCA2, and XRCC5 in non–small cell lung cancer. Clinical cancer research, 2007.13 (3): p. 832-838.
212. Liu, K., et al., Promoter hypermethylation: an important epigenetic mechanism for hMLH1 gene inactivation in head and neck squamous cell carcinoma. Otolaryngology-Head and Neck Surgery, 2002.126 (5): p. 548-553.
213. Fleisher, A.S., et al., Hypermethylation of the hMLH1 gene promoter is associated with microsatellite instability in early human gastric neoplasia. Oncogene, 2001. 20 (3): p. 329-335.
214. Bernal, C., et al., DNA methylation profile in diffuse type gastric cancer: evidence for hypermethylation of the BRCA1 promoter region in early-onset gastric carcinogenesis. Biological research, 2008. 41 (3): p. 303-315.
215. Seedhouse, C., E. Das-Gupta, and N. Russell, Methylation of the hMLH1 promoter and its association with microsatellite instability in acute myeloid leukemia. Leukemia, 2003. 17 (1): p. 83-88.
216. Zhang, L. and X. Long, Association of BRCA1 promoter methylation with sporadic breast cancers: Evidence from 40 studies.Scientific reports, 2015. 5 (1): p. 17869.
217. Gras, E., et al., Loss of heterozygosity on chromosome 13q12–q14, BRCA‐2 mutations and lack of BRCA‐2 promoter hypermethylation in sporadic epithelial ovarian tumors. Cancer, 2001.92 (4): p. 787-795.
218. Yu, J., et al., A novel set of DNA methylation markers in urine sediments for sensitive/specific detection of bladder cancer.Clinical cancer research, 2007. 13 (24): p. 7296-7304.
219. Fojo, T., Cancer, DNA repair mechanisms, and resistance to chemotherapy . 2001, Oxford University Press. p. 1434-1436.
220. Phi, L.T.H., et al., Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem cells international, 2018. 2018 .
221. Papaccio, F., et al., Concise review: cancer cells, cancer stem cells, and mesenchymal stem cells: influence in cancer development. Stem cells translational medicine, 2017. 6 (12): p. 2115-2125.
222. Aramini, B., et al., Dissecting tumor growth: the role of cancer stem cells in drug resistance and recurrence. Cancers, 2022.14 (4): p. 976.
223. Pattabiraman, D.R. and R.A. Weinberg, Tackling the cancer stem cells—what challenges do they pose? Nature reviews Drug discovery, 2014. 13 (7): p. 497-512.
224. Yang, Z.-J. and R.J. Wechsler-Reya, Hit’em where they live: targeting the cancer stem cell niche. Cancer cell, 2007.11 (1): p. 3-5.
225. Gaggianesi, M., et al., Messing up the cancer stem cell chemoresistance mechanisms supported by tumor microenvironment.Frontiers in Oncology, 2021: p. 2847.
226. Li, Y., et al., Drug resistance and Cancer stem cells. Cell Communication and Signaling, 2021. 19 (1): p. 1-11.
227. Prieto-Vila, M., et al., Drug resistance driven by cancer stem cells and their niche. International journal of molecular sciences, 2017. 18 (12): p. 2574.
228. Barbato, L., et al., Cancer stem cells and targeting strategies. Cells, 2019. 8 (8): p. 926.
229. Liang, L. and A.M. Kaufmann, The Significance of Cancer Stem Cells and Epithelial–Mesenchymal Transition in Metastasis and Anti-Cancer Therapy. International Journal of Molecular Sciences, 2023.24 (3): p. 2555.
230. Agliano, A., A. Calvo, and C. Box. The challenge of targeting cancer stem cells to halt metastasis . in Seminars in Cancer Biology . 2017. Elsevier.
231. Knoll, S., S. Emmrich, and B.M. Pützer, The E2F1-miRNA cancer progression network. MicroRNA Cancer Regulation: Advanced Concepts, Bioinformatics and Systems Biology Tools, 2013: p. 135-147.
232. Dar, A.A., et al., miRNA-205 suppresses melanoma cell proliferation and induces senescence via regulation of E2F1 protein.Journal of Biological Chemistry, 2011. 286 (19): p. 16606-16614.
233. Gregory, P.A., et al., The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nature cell biology, 2008. 10 (5): p. 593-601.
234. Godlewski, J., et al., Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal. Cancer research, 2008. 68 (22): p. 9125-9130.
235. Deng, J., et al., Long non-coding RNA HOTAIR regulates the proliferation, self-renewal capacity, tumor formation and migration of the cancer stem-like cell (CSC) subpopulation enriched from breast cancer cells. PLoS One, 2017. 12 (1): p. e0170860.
236. O’Brien, C.A., et al., ID1 and ID3 regulate the self-renewal capacity of human colon cancer-initiating cells through p21. Cancer cell, 2012. 21 (6): p. 777-792.
237. Nahand, J.S., et al., microRNAs: new prognostic, diagnostic, and therapeutic biomarkers in cervical cancer. Journal of cellular physiology, 2019. 234 (10): p. 17064-17099.
238. Naeli, P., et al., Circular RNAs and gastrointestinal cancers: epigenetic regulators with a prognostic and therapeutic role.Critical reviews in oncology/hematology, 2020. 145 : p. 102854.
239. Yan, H. and P. Bu, Non-coding RNAs in cancer stem cells.Cancer letters, 2018. 421 : p. 121-126.
240. Davis, B.N. and A. Hata, Regulation of MicroRNA Biogenesis: A miRiad of mechanisms. Cell Communication and Signaling, 2009.7 : p. 1-22.