The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities
Abstract
:Simple Summary
Abstract
1. Introduction
2. Roles of Cellular TME Components in Regulating the Metastatic Cascade
2.1. Role of Immune Cells in Modulating Cancer Metastasis
2.2. Role of Mesenchymal Stem Cells in Regulating Metastasis
2.3. Cancer-Associated Fibroblasts in Promoting Metastasis
2.4. Endothelial Cells Attract Cancer Cells to the Metastatic Site
2.5. Components in the Blood Microenvironment That Facilitate Metastasis
3. Role of Extracellular TME Components in Tumor Progression
3.1. Role of the ECM in Cancer Metastasis
3.2. Hypoxic Microenvironment as a Major Driver of Cancer Metastasis
4. Targeting Cellular Constituents of the TME to Block Metastasis
4.1. TAMs as a Promising Target against Cancer Metastasis
4.2. Exploiting Cancer-Associated Mesenchymal Stroma/Stem-Like Cells to Control Metastasis
4.3. Targeting the Blood Microenvironment
5. Strategies to Improve Tumor Oxygenation and Therapeutic Efficacy
5.1. Remodeling the Tumor Vasculature
5.2. Stroma Normalization Strategy to Target Hypoxia
6. Discussion—Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
5′-FU | 5′-fluorouracil |
APCs | antigen presenting cells |
ATX | autotaxin |
BM | basement membrane |
BM-MSCs | bone marrow-derived mesenchymal stem cells |
CAFs | cancer associated fibroblasts |
CA-MSC | cancer-associated MSC |
CB2 | cannabinoid receptor 2 |
CAIX | carbonic anhydrase |
CCL2 | chemokine (C-C motif) ligand 2 |
CTCs | circulating tumor cells |
CTMs | circulating tumor microemboli |
CSF-1 | colony-stimulating factor-1 |
CRC | colorectal cancer |
PDAC | pancreatic ductal adenocarcinoma |
endMT | endothelial-to-mesenchymal transition |
EMT | epithelial-to-mesenchymal transition |
ECM | extracellular matrix |
FAP | fibroblast activation protein |
GC-MSCs | gastric cancer tissue-related mesenchymal stem cells |
AuNPs | gold nanoparticles |
GPCRs | G-protein coupled receptors |
GM-CSF | granulocyte-macrophage colony-stimulating factor |
HNSCC | head and neck squamous cell carcinoma |
HSC | hematopoietic stem cells |
HCC | hepatocellular carcinoma |
IMs | inflammatory monocytes |
IFN-γ | interferon-γ |
KCNN4 | intermediate-conductance Ca2+-activated potassium channels |
LECs | lymphatic endothelial cells |
LPA | lysophosphatidic acid |
LOX | lysyl oxidase |
MMPs | matrix metalloproteinases |
MSCs | mesenchymal stem cells |
MMT | mesenchymal-mesenchymal transition |
MAMs | metastasis-associated macrophages |
NPs | nanoparticles |
NK | natural killer cells |
NSCLC | non-small cell lung cancer cells |
PDX | patient-derived xenograft mouse |
PD-L1 | programmed death-ligand 1 |
PRL-3 | phosphatase of regenerating liver |
S-TRAIL | secretable tumor necrosis factor apoptosis inducing ligand |
sECM | synthetic extracellular matrix |
TSP | thrombospondin |
TLRs | toll-like Receptors |
TME | tumor microenvironment |
TMEM | tumor microenvironment of metastasis |
TNF-α | tumor necrosis factor-α |
TAMs | tumor-associated macrophages |
VCAM-1 | vascular cell adhesion molecule-1 |
α-SMA | α-smooth muscle actin |
References
- Fearon, E.R.; Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 1990, 61, 759–767. [Google Scholar] [CrossRef]
- Kurose, K.; Hoshaw-Woodard, S.; Adeyinka, A.; Lemeshow, S.; Watson, P.H.; Eng, C. Genetic model of multi-step breast carcino-genesis involving the epithelium and stroma: Clues to tumour-microenvironment interactions. Hum. Mol. Genet. 2001, 10, 1907–1913. [Google Scholar] [CrossRef] [Green Version]
- Kitamura, T.; Qian, B.-Z.; Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef] [PubMed]
- Dunn, G.P.; Old, L.J.; Schreiber, R.D. The Immunobiology of Cancer Immunosurveillance and Immunoediting. Immunity 2004, 21, 137–148. [Google Scholar] [CrossRef] [Green Version]
- Eyles, J.; Puaux, A.-L.; Wang, X.; Toh, B.; Prakash, C.; Hong, M.; Tan, T.G.; Zheng, L.; Ong, L.C.; Jin, Y.; et al. Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J. Clin. Investig. 2010, 120, 2030–2039. [Google Scholar] [CrossRef]
- Paolino, M.; Choidas, A.; Wallner, S.; Pranjic, B.; Uribesalgo, I.; Loeser, S.; Jamieson, A.M.; Langdon, W.Y.; Ikeda, F.; Fededa, J.P.; et al. The E3 ligase Cbl-b and TAM receptors regulate cancer metastasis via natural killer cells. Nat. Cell Biol. 2014, 507, 508–512. [Google Scholar] [CrossRef]
- Bidwell, B.N.; Slaney, C.Y.; Withana, N.P.; Forster, S.; Cao, Y.; Loi, S.; Andrews, D.; Mikeska, T.; Mangan, N.E.; Samarajiwa, S.A.; et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 2012, 18, 1224–1231. [Google Scholar] [CrossRef]
- McAllister, S.S.; Weinberg, R.A. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat. Cell Biol. 2014, 16, 717–727. [Google Scholar] [CrossRef]
- Neophytou, C.M.; Pierides, C.; Christodoulou, M.I.; Costeas, P.; Kyriakou, T.C.; Papageorgis, P. The Role of Tumor-Associated My-eloid Cells in Modulating Cancer Therapy. Front. Oncol. 2020, 10, 899. [Google Scholar] [CrossRef]
- Whipple, C.A. Tumor talk: Understanding the conversation between the tumor and its microenvironment. Cancer Cell Microenviron. 2015, 2, e773. [Google Scholar] [PubMed]
- Singh, S.; Mehta, N.; Lilan, J.; Budhthoki, M.B.; Chao, F.; Yong, L. Initiative action of tumor-associated macrophage during tumor metastasis. Biochim. Open 2017, 4, 8–18. [Google Scholar] [CrossRef]
- Smith, H.A.; Kang, Y. The metastasis-promoting roles of tumor-associated immune cells. J. Mol. Med. 2013, 91, 411–429. [Google Scholar] [CrossRef]
- Sanchez, L.R.; Borriello, L.; Entenberg, D.; Condeelis, J.S.; Oktay, M.H.; Karagiannis, G.S. The emerging roles of macrophages in cancer metastasis and response to chemotherapy. J. Leukoc. Biol. 2019, 106, 259–274. [Google Scholar] [CrossRef]
- Wang, R.; Zhang, J.; Chen, S.; Lu, M.; Luo, X.; Yao, S.; Liu, S.; Qin, Y.; Chen, H. Tumor-associated macrophages provide a suitable microenvironment for non-small lung cancer invasion and progression. Lung Cancer 2011, 74, 188–196. [Google Scholar] [CrossRef]
- Robinson, B.D.; Sica, G.L.; Liu, Y.-F.; Rohan, T.E.; Gertler, F.B.; Condeelis, J.S.; Jones, J.G. Tumor Microenvironment of Metastasis in Human Breast Carcinoma: A Potential Prognostic Marker Linked to Hematogenous Dissemination. Clin. Cancer Res. 2009, 15, 2433–2441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thiery, J.P.; Acloque, H.; Huang, R.Y.; Nieto, M.A. Epithelial-Mesenchymal Transitions in Development and Disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef]
- Papageorgis, P. TGFbeta Signaling in Tumor Initiation, Epithelial-to-Mesenchymal Transition, and Metastasis. J. Oncol. 2015, 2015, 587193. [Google Scholar] [CrossRef] [Green Version]
- Gupta, D.K.; Singh, N.; Sahu, D.K. TGF-beta Mediated Crosstalk Between Malignant Hepatocyte and Tumor Microenvironment in Hepatocellular Carcinoma. Cancer Growth Metastasis 2014, 7, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Zhu, X.-D.; Zhang, J.-B.; Zhuang, P.-Y.; Zhu, H.-G.; Zhang, W.; Xiong, Y.-Q.; Wu, W.-Z.; Wang, L.; Tang, Z.-Y.; Sun, H.-C. High Expression of Macrophage Colony-Stimulating Factor in Peritumoral Liver Tissue Is Associated with Poor Survival After Curative Resection of Hepatocellular Carcinoma. J. Clin. Oncol. 2008, 26, 2707–2716. [Google Scholar] [CrossRef]
- Lewis, C.E.; Pollard, J.W. Distinct Role of Macrophages in Different Tumor Microenvironments. Cancer Res. 2006, 66, 605–612. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.Y.; Xu, J.Y.; Shi, X.Y.; Huang, W.; Ruan, T.Y.; Xie, P.; Ding, J.L. M2-polarized tumor-associated macrophages promoted epitheli-al-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Lab. Investig. 2013, 93, 844–854. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Wang, X.; Li, X.; Fan, Y.; Li, G.; Guo, C.; Zhu, F.; Zhang, L.; Shi, Y. CD68(+)HLA-DR(+) M1-like macrophages promote motility of HCC cells via NF-kappaB/FAK pathway. Cancer Lett. 2014, 345, 91–99. [Google Scholar] [CrossRef]
- Chu, Z.; Lai, W.; Chen, S.; Wu, H.; Guan, Y.; Liu, L.; Zeng, Y.; Zhao, H.; Jiang, J. PRL-3 promotes the proliferation of LoVo cells via the upregulation of KCNN4 channels. Oncol. Rep. 2011, 26, 909–917. [Google Scholar] [CrossRef] [Green Version]
- Xu, H.; Lai, W.; Zhang, Y.; Liu, L.; Luo, X.; Zeng, Y.; Wu, H.; Lan, Q.; Chu, Z. Tumor-associated macrophage-derived IL-6 and IL-8 enhance invasive activity of LoVo cells induced by PRL-3 in a KCNN4 channel-dependent manner. BMC Cancer 2014, 14, 330. [Google Scholar] [CrossRef] [Green Version]
- Lin, L.; Chen, Y.S.; Yao, Y.D.; Chen, J.Q.; Chen, J.N.; Huang, S.Y.; Zeng, Y.J.; Yao, H.R.; Zeng, S.H.; Fu, Y.S.; et al. CCL18 from tumor-associated macrophages promotes angio-genesis in breast cancer. Oncotarget 2015, 6, 34758–34773. [Google Scholar] [CrossRef] [Green Version]
- Lin, Y.; Xu, J.; Lan, H. Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applica-tions. J. Hematol. Oncol. 2019, 12, 76. [Google Scholar] [CrossRef]
- Qian, B.; Deng, Y.; Im, J.H.; Muschel, R.J.; Zou, Y.; Li, J.; Lang, R.A.; Pollard, J.W. A Distinct Macrophage Population Mediates Metastatic Breast Cancer Cell Extravasation, Establishment and Growth. PLoS ONE 2009, 4, e6562. [Google Scholar] [CrossRef] [Green Version]
- Ueno, T.; Toi, M.; Saji, H.; Muta, M.; Bando, H.; Kuroi, K.; Koike, M.; Inadera, H.; Matsushima, K. Significance of macrophage chemoattractant protein-1 in macrophage recruitment, angiogenesis, and survival in human breast cancer. Clin. Cancer Res. 2000, 6, 3282–3289. [Google Scholar]
- Saji, H.; Koike, M.; Yamori, T.; Saji, S.; Seiki, M.; Matsushima, K.; Toi, M. Significant correlation of monocyte chemoattractant protein-1 expression with neovascularization and progression of breast carcinoma. Cancer 2001, 92, 1085–1091. [Google Scholar] [CrossRef]
- Qian, B.-Z.; Li, J.; Zhang, H.; Kitamura, T.; Zhang, J.; Campion, L.R.; Kaiser, E.A.; Snyder, L.A.; Pollard, J.W. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 2011, 475, 222–225. [Google Scholar] [CrossRef] [Green Version]
- Kitamura, T.; Qian, B.-Z.; Soong, D.; Cassetta, L.; Noy, R.; Sugano, G.; Kato, Y.; Li, J.; Pollard, J.W. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J. Exp. Med. 2015, 212, 1043–1059. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.; Zhang, X.H.-F.; Massagué, J. Macrophage Binding to Receptor VCAM-1 Transmits Survival Signals in Breast Cancer Cells that Invade the Lungs. Cancer Cell 2011, 20, 538–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, D.-C.; Shyu, W.-C.; Lin, S.-Z. Mesenchymal Stem Cells. Cell Transplant. 2011, 20, 5–14. [Google Scholar] [CrossRef] [Green Version]
- Zhou, J.; Tan, X.; Tan, Y.; Li, Q.; Ma, J.; Wang, G. Mesenchymal Stem Cell Derived Exosomes in Cancer Progression, Metastasis and Drug Delivery: A Comprehensive Review. J. Cancer 2018, 9, 3129–3137. [Google Scholar] [CrossRef] [Green Version]
- Atiya, H.; Frisbie, L.; Pressimone, C.; Coffman, L. Mesenchymal Stem Cells in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1234, 31–42. [Google Scholar] [CrossRef]
- Yeo, R.W.Y.; Lai, R.C.; Zhang, B.; Tan, S.S.; Yin, Y.; Teh, B.J.; Lim, S.K. Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Adv. Drug Deliv. Rev. 2013, 65, 336–341. [Google Scholar] [CrossRef] [PubMed]
- Simpson, R.J.; Lim, J.W.; Moritz, R.L.; Mathivanan, S. Exosomes: Proteomic insights and diagnostic potential. Expert Rev. Proteom. 2009, 6, 267–283. [Google Scholar] [CrossRef] [PubMed]
- Mathivanan, S.; Fahner, C.J.; Reid, G.E.; Simpson, R.J. ExoCarta 2012: Database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 2011, 40, D1241–D1244. [Google Scholar] [CrossRef] [Green Version]
- Lin, R.; Wang, S.; Zhao, R.C. Exosomes from human adipose-derived mesenchymal stem cells promote migration through Wnt signaling pathway in a breast cancer cell model. Mol. Cell. Biochem. 2013, 383, 13–20. [Google Scholar] [CrossRef]
- Wang, M.; Zhao, C.; Shi, H.; Zhang, B.; Zhang, L.; Zhang, X.; Wang, S.; Wu, X.; Yang, T.; Huang, F.; et al. Deregulated microRNAs in gastric cancer tissue-derived mesen-chymal stem cells: Novel biomarkers and a mechanism for gastric cancer. Br. J. Cancer. 2014, 110, 1199–1210. [Google Scholar] [CrossRef]
- Liu, K.; Li, G.; Fan, C.; Diao, Y.; Wu, B.; Li, J. Increased Expression of MicroRNA-221 in gastric cancer and its clinical significance. J. Int. Med. Res. 2012, 40, 467–474. [Google Scholar] [CrossRef] [PubMed]
- Gu, H.; Ji, R.; Zhang, X.; Wang, M.; Zhu, W.; Qian, H.; Chen, Y.; Jiang, P.; Xu, W. Exosomes derived from human mesenchymal stem cells promote gastric cancer cell growth and migration via the activation of the Akt pathway. Mol. Med. Rep. 2016, 14, 3452–3458. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Hendrix, A.; Hernot, S.; Lemaire, M.; De Bruyne, E.; Van Valckenborgh, E.; Lahoutte, T.; De Wever, O.; Vanderkerken, K.; Menu, E.; et al. Bone marrow stromal cell-derived ex-osomes as communicators in drug resistance in multiple myeloma cells. Blood 2014, 124, 555–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Casson, J.; Davies, O.G.; Smith, C.-A.; Dalby, M.J.; Berry, C.C. Mesenchymal stem cell-derived extracellular vesicles may promote breast cancer cell dormancy. J. Tissue Eng. 2018, 9. [Google Scholar] [CrossRef]
- Neophytou, C.M.; Kyriakou, T.-C.; Papageorgis, P. Mechanisms of Metastatic Tumor Dormancy and Implications for Cancer Therapy. Int. J. Mol. Sci. 2019, 20, 6158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ono, M.; Kosaka, N.; Tominaga, N.; Yoshioka, Y.; Takeshita, F.; Takahashi, R.-U.; Yoshida, M.; Tsuda, H.; Tamura, K.; Ochiya, T. Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells. Sci. Signal. 2014, 7, ra63. [Google Scholar] [CrossRef]
- Bliss, S.A.; Sinha, G.; Sandiford, O.A.; Williams, L.M.; Engelberth, D.J.; Guiro, K.; Isenalumhe, L.L.; Greco, S.J.; Ayer, S.; Bryan, M.; et al. Mesenchymal stem cell–derived exosomes stimulate cycling quiescence and early breast cancer dormancy in bone marrow. Cancer Res. 2016, 76, 5832–5844. [Google Scholar] [CrossRef] [Green Version]
- Puré, E.; Blomberg, R. Pro-tumorigenic roles of fibroblast activation protein in cancer: Back to the basics. Oncogene 2018, 37, 4343–4357. [Google Scholar] [CrossRef]
- Costa, A.; Kieffer, Y.; Scholer-Dahirel, A.; Pelon, F.; Bourachot, B.; Cardon, M.; Sirven, P.; Magagna, I.; Fuhrmann, L.; Bernard, C.; et al. Fibroblast Heterogeneity and Immunosup-pressive Environment in Human Breast Cancer. Cancer Cell 2018, 33, 463–479.e10. [Google Scholar] [CrossRef] [Green Version]
- Shiga, K.; Hara, M.; Nagasaki, T.; Sato, T.; Takahashi, H.; Takeyama, H. Cancer-Associated Fibroblasts: Their Characteristics and Their Roles in Tumor Growth. Cancers 2015, 7, 2443–2458. [Google Scholar] [CrossRef] [PubMed]
- Heneberg, P. Paracrine tumor signaling induces transdifferentiation of surrounding fibroblasts. Crit. Rev. Oncol. 2016, 97, 303–311. [Google Scholar] [CrossRef]
- Räsänen, K.; Vaheri, A. Activation of fibroblasts in cancer stroma. Exp. Cell Res. 2010, 316, 2713–2722. [Google Scholar] [CrossRef]
- Gaggioli, C.; Hooper, S.; Hidalgo-Carcedo, C.; Grosse, R.; Marshall, J.F.; Harrington, K.; Sahai, E. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat. Cell Biol. 2007, 9, 1392–1400. [Google Scholar] [CrossRef] [PubMed]
- Eck, S.M.; Cote, A.L.; Winkelman, W.D.; Brinckerhoff, C.E. CXCR4 and matrix metalloproteinase-1 are elevated in breast carcino-ma-associated fibroblasts and in normal mammary fibroblasts exposed to factors secreted by breast cancer cells. Mol. Cancer Res. 2009, 7, 1033–1044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murphy, G.; Nagase, H. Progress in matrix metalloproteinase research. Mol. Asp. Med. 2008, 29, 290–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gonzalez-Avila, G.; Sommer, B.; Mendoza-Posada, D.A.; Ramos, C.; Garcia-Hernandez, A.A.; Falfan-Valencia, R. Matrix metallo-proteinases participation in the metastatic process and their diagnostic and therapeutic applications in cancer. Crit. Rev. Oncol. Hematol. 2019, 137, 57–83. [Google Scholar] [CrossRef]
- Erdogan, B.; Ao, M.; White, L.M.; Means, A.L.; Brewer, B.M.; Yang, L.; Washington, M.K.; Shi, C.; Franco, O.E.; Weaver, A.M.; et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J. Cell Biol. 2017, 216, 3799–3816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonneau, C.; Eliès, A.; Kieffer, Y.; Bourachot, B.; Ladoire, S.; Pelon, F.; Hequet, D.; Guinebretière, J.-M.; Blanchet, C.; Vincent-Salomon, A.; et al. A subset of activated fibroblasts is associated with distant relapse in early luminal breast cancer. Breast Cancer Res. 2020, 22, 1–22. [Google Scholar] [CrossRef]
- Rankin, E.B.; Giaccia, A.J. Hypoxic control of metastasis. Science 2016, 352, 175–180. [Google Scholar] [CrossRef] [Green Version]
- Smolarczyk, R.; Czapla, J.; Jarosz-Biej, M.; Czerwinski, K.; Cichoń, T. Vascular disrupting agents in cancer therapy. Eur. J. Pharmacol. 2021, 891, 173692. [Google Scholar] [CrossRef]
- Albrecht, I.; Christofori, G. Molecular mechanisms of lymphangiogenesis in development and cancer. Int. J. Dev. Biol. 2011, 55, 483–494. [Google Scholar] [CrossRef]
- Lee, E.; Fertig, E.J.; Jin, K.; Sukumar, S.; Pandey, N.B.; Popel, A.S. Breast cancer cells condition lymphatic endothelial cells within pre-metastatic niches to promote metastasis. Nat. Commun. 2014, 5, 1–16. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.-C.; Zhang, X.-F.; Peng, J.; Li, X.-F.; Wang, A.-L.; Bie, Y.-Q.; Shi, L.-H.; Lin, M.-B. Survival Mechanisms and Influence Factors of Circulating Tumor Cells. BioMed Res. Int. 2018, 2018, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Camerer, E.; Qazi, A.A.; Duong, D.N.; Cornelissen, I.; Advincula, R.; Coughlin, S.R. Platelets, protease-activated receptors, and fi-brinogen in hematogenous metastasis. Blood 2004, 104, 397–401. [Google Scholar] [CrossRef] [PubMed]
- Gay, L.J.; Felding-Habermann, B. Contribution of platelets to tumour metastasis. Nat. Rev. Cancer 2011, 11, 123–134. [Google Scholar] [CrossRef]
- Mutoh, T.; Rivera, R.; Chun, J. Insights into the pharmacological relevance of lysophospholipid receptors. Br. J. Pharmacol. 2012, 165, 829–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tseng, J.Y.; Yang, C.Y.; Liang, S.C.; Liu, R.S.; Yang, S.H.; Lin, J.K.; Chen, Y.M.; Wu, Y.C.; Jiang, J.K.; Lin, C.H. Interleukin-17A modulates circulating tumor cells in tumor draining vein of colorectal cancers and affects metastases. Clin. Cancer Res. 2014, 20, 2885–2897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leblanc, R.; Houssin, A.; Peyruchaud, O. Platelets, autotaxin and lysophosphatidic acid signalling: Win-win factors for cancer metastasis. Br. J. Pharmacol. 2018, 175, 3100–3110. [Google Scholar] [CrossRef] [PubMed]
- Frantz, C.; Stewart, K.M.; Weaver, V.M. The extracellular matrix at a glance. J. Cell Sci. 2010, 123 Pt 24, 4195–4200. [Google Scholar] [CrossRef] [Green Version]
- Khawar, I.A.; Kim, J.H.; Kuh, H.-J. Improving drug delivery to solid tumors: Priming the tumor microenvironment. J. Control. Release 2015, 201, 78–89. [Google Scholar] [CrossRef]
- Egeblad, M.; Rasch, M.G.; Weaver, V.M. Dynamic interplay between the collagen scaffold and tumor evolution. Curr. Opin. Cell Biol. 2010, 22, 697–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levental, K.R.; Yu, H.; Kass, L.; Lakins, J.N.; Egeblad, M.; Erler, J.T.; Fong, S.F.; Csiszar, K.; Giaccia, A.; Weninger, W.; et al. Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling. Cell 2009, 139, 891–906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Byrne, C.; Schairer, C.; Wolfe, J.; Parekh, N.; Salane, M.; Brinton, L.A.; Hoover, R.; Haile, R. Mammographic Features and Breast Cancer Risk: Effects with Time, Age, and Menopause Status. J. Natl. Cancer Inst. 1995, 87, 1622–1629. [Google Scholar] [CrossRef] [PubMed]
- Boyd, N.F.; Guo, H.; Martin, L.J.; Sun, L.; Stone, J.; Fishell, E.; Jong, R.A.; Hislop, G.; Chiarelli, A.; Minkin, S.; et al. Mammographic Density and the Risk and Detection of Breast Cancer. N. Engl. J. Med. 2007, 356, 227–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalli, M.; Stylianopoulos, T. Defining the Role of Solid Stress and Matrix Stiffness in Cancer Cell Proliferation and Metastasis. Front. Oncol. 2018, 8, 55. [Google Scholar] [CrossRef] [PubMed]
- Cheng, G.; Tse, J.; Jain, R.K.; Munn, L.L. Micro-Environmental Mechanical Stress Controls Tumor Spheroid Size and Morphology by Suppressing Proliferation and Inducing Apoptosis in Cancer Cells. PLoS ONE 2009, 4, e4632. [Google Scholar] [CrossRef] [Green Version]
- Butcher, D.T.; Alliston, T.; Weaver, V.M. A tense situation: Forcing tumour progression. Nat. Rev. Cancer 2009, 9, 108–122. [Google Scholar] [CrossRef] [PubMed]
- Northey, J.J.; Przybyla, L.; Weaver, V.M. Tissue Force Programs Cell Fate and Tumor Aggression. Cancer Discov. 2017, 7, 1224–1237. [Google Scholar] [CrossRef] [Green Version]
- Samuel, M.S.; Lopez, J.I.; McGhee, E.J.; Croft, D.R.; Strachan, D.; Timpson, P.; Munro, J.; Schröder, E.; Zhou, J.; Brunton, V.G.; et al. Actomyosin-mediated cellular tension drives increased tissue stiffness and beta-catenin activation to induce epidermal hyperplasia and tumor growth. Cancer Cell 2011, 19, 776–791. [Google Scholar] [CrossRef] [Green Version]
- Lawson, C.D.; Burridge, K. The on-off relationship of Rho and Rac during integrin-mediated adhesion and cell migration. Small GTPases 2014, 5, e27958. [Google Scholar] [CrossRef] [Green Version]
- Polacheck, W.J.; Zervantonakis, I.K.; Kamm, R.D. Tumor cell migration in complex microenvironments. Cell. Mol. Life Sci. 2012, 70, 1335–1356. [Google Scholar] [CrossRef] [Green Version]
- Katsumi, A.; Orr, A.W.; Tzima, E.; Schwartz, M.A. Integrins in Mechanotransduction. J. Biol. Chem. 2004, 279, 12001–12004. [Google Scholar] [CrossRef] [Green Version]
- Shi, Q.; Boettiger, D. A Novel Mode for Integrin-mediated Signaling: Tethering Is Required for Phosphorylation of FAK Y397. Mol. Biol. Cell 2003, 14, 4306–4315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalli, M.; Papageorgis, P.; Gkretsi, V.; Stylianopoulos, T. Solid Stress Facilitates Fibroblasts Activation to Promote Pancreatic Cancer Cell Migration. Ann. Biomed. Eng. 2018, 46, 657–669. [Google Scholar] [CrossRef]
- Kalli, M.; Minia, A.; Pliaka, V.; Fotis, C.; Alexopoulos, L.G.; Stylianopoulos, T. Solid stress-induced migration is mediated by GDF15 through Akt pathway activation in pancreatic cancer cells. Sci. Rep. 2019, 9, 1–12. [Google Scholar] [CrossRef]
- Kalli, M.; Voutouri, C.; Minia, A.; Pliaka, V.; Fotis, C.; Alexopoulos, L.G.; Stylianopoulos, T. Mechanical Compression Regulates Brain Cancer Cell Migration Through MEK1/Erk1 Pathway Activation and GDF15 Expression. Front. Oncol. 2019, 9, 992. [Google Scholar] [CrossRef] [Green Version]
- Naba, A.; Clauser, K.R.; Lamar, J.M.; Carr, S.A.; Hynes, R.O. Extracellular matrix signatures of human mammary carcinoma identify novel metastasis promoters. eLife 2014, 3, e01308. [Google Scholar] [CrossRef] [Green Version]
- Laklai, H.; Miroshnikova, Y.A.; Pickup, M.W.; Collisson, E.A.; Kim, G.E.; Barrett, A.S.; Hill, R.C.; Lakins, J.N.; Schlaepfer, D.D.; Mouw, J.K.; et al. Genotype tunes pancreatic ductal adeno-carcinoma tissue tension to induce matricellular fibrosis and tumor progression. Nat. Med. 2016, 22, 497–505. [Google Scholar] [CrossRef] [Green Version]
- Jiang, H.; Hegde, S.; Knolhoff, B.L.; Zhu, Y.; Herndon, J.M.; Meyer, M.A.; Nywening, T.M.; Hawkins, T.M.N.W.G.; Shapiro, I.M.; Weaver, D.T.; et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med. 2016, 22, 851–860. [Google Scholar] [CrossRef] [PubMed]
- Stylianopoulos, T.; Martin, J.D.; Snuderl, M.; Mpekris, F.; Jain, S.R.; Jain, R.K. Coevolution of Solid Stress and Interstitial Fluid Pressure in Tumors during Progression: Implications for Vascular Collapse. Cancer Res. 2013, 73, 3833–3841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, X.; Kang, Y. Hypoxia and Hypoxia-Inducible Factors: Master Regulators of Metastasis. Clin. Cancer Res. 2010, 16, 5928–5935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsai, Y.-P.; Wu, K.-J. Hypoxia-regulated target genes implicated in tumor metastasis. J. Biomed. Sci. 2012, 19, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stubbs, M.; McSheehy, P.M.J.; Griffiths, J.R.; Bashford, C.L. Causes and consequences of tumour acidity and implications for treat-ment. Mol. Med. Today 2000, 6, 15–19. [Google Scholar] [CrossRef]
- Yang, S.; Gao, H. Nanoparticles for modulating tumor microenvironment to improve drug delivery and tumor therapy. Pharmacol. Res. 2017, 126, 97–108. [Google Scholar] [CrossRef]
- Romero-Garcia, S.; Lopez-Gonzalez, J.S.; B´ez-Viveros, J.L.; Aguilar-Cazares, D.; Prado-Garcia, H. Tumor cell metabolism: An integral view. Cancer Biol. Ther. 2011, 12, 939–948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schito, L.; Semenza, G.L. Hypoxia-inducible factors: Master regulators of cancer progression. Trends Cancer 2016, 2, 758–770. [Google Scholar] [CrossRef] [Green Version]
- Dhani, N.; Fyles, A.; Hedley, D.; Milosevic, M. The Clinical Significance of Hypoxia in Human Cancers. Semin. Nucl. Med. 2015, 45, 110–121. [Google Scholar] [CrossRef]
- Sullivan, R.; Graham, C.H. Hypoxia-driven selection of the metastatic phenotype. Cancer Metastasis Rev. 2007, 26, 319–331. [Google Scholar] [CrossRef]
- Vaupel, P.; Mayer, A. Hypoxia in cancer: Significance and impact on clinical outcome. Cancer Metastasis Rev. 2007, 26, 225–239. [Google Scholar] [CrossRef]
- Preet, A.; Qamri, Z.; Nasser, M.W.; Prasad, A.; Shilo, K.; Zou, X.; Groopman, J.E.; Ganju, R.K. Cannabinoid Receptors, CB1 and CB2, as Novel Targets for Inhibition of Non–Small Cell Lung Cancer Growth and Metastasis. Cancer Prev. Res. 2011, 4, 65–75. [Google Scholar] [CrossRef] [Green Version]
- Ravi, J.; Elbaz, M.; Wani, N.A.; Nasser, M.W.; Ganju, R.K. Cannabinoid receptor-2 agonist inhibits macrophage induced EMT in non-small cell lung cancer by downregulation of EGFR pathway. Mol. Carcinog. 2016, 55, 2063–2076. [Google Scholar] [CrossRef]
- Pixley, F.J.; Stanley, E.R. CSF-1 regulation of the wandering macrophage: Complexity in action. Trends Cell Biol. 2004, 14, 628–638. [Google Scholar] [CrossRef] [PubMed]
- Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a para-digm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef]
- Ries, C.H.; Hoves, S.; Cannarile, M.A.; Rüttinger, D. CSF-1/CSF-1R targeting agents in clinical development for cancer therapy. Curr. Opin. Pharmacol. 2015, 23, 45–51. [Google Scholar] [CrossRef] [PubMed]
- Green, J.R.; Clezardin, P. Mechanisms of bisphosphonate effects on osteoclasts, tumor cell growth, and metastasis. Am. J. Clin. Oncol. 2002, 25 (Suppl. 1), S3–S9. [Google Scholar] [CrossRef] [PubMed]
- Green, J.R. Bisphosphonates: Preclinical review. Oncologist 2004, 9 (Suppl. 4), 3–13. [Google Scholar] [CrossRef] [PubMed]
- Zeisberger, S.M.; Odermatt, B.; Marty, C.; Zehnder-Fjallman, A.H.; Ballmer-Hofer, K.; Schwendener, R.A. Clodro-nate-liposome-mediated depletion of tumour-associated macrophages: A new and highly effective antiangiogenic therapy approach. Br. J. Cancer 2006, 95, 272–281. [Google Scholar] [CrossRef]
- Hiraoka, K.; Zenmyo, M.; Watari, K.; Iguchi, H.; Fotovati, A.; Kimura, Y.N.; Hosoi, F.; Shoda, T.; Nagata, K.; Osada, H.; et al. Inhibition of bone and muscle metastases of lung cancer cells by a decrease in the number of monocytes/macrophages. Cancer Sci. 2008, 99, 1595–1602. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Gao, L.; Cai, Y.; Liu, H.; Gao, D.; Lai, J.; Jia, B.; Wang, F.; Liu, Z. Inhibition of tumor growth and metastasis by photoimmunotherapy targeting tumor-associated macrophage in a sorafenib-resistant tumor model. Biomaterials 2016, 84, 1–12. [Google Scholar] [CrossRef]
- Timaner, M.; Bril, R.; Kaidar-Person, O.; Rachman-Tzemah, C.; Alishekevitz, D.; Kotsofruk, R.; Miller, V.; Nevelsky, A.; Daniel, S.; Raviv, Z.; et al. Dequalinium blocks mac-rophage-induced metastasis following local radiation. Oncotarget 2015, 6, 27537–27554. [Google Scholar] [CrossRef] [Green Version]
- Deryugina, E.I.; Zajac, E.; Juncker-Jensen, A.; Kupriyanova, T.A.; Welter, L.; Quigley, J.P. Tissue-Infiltrating Neutrophils Constitute the Major In Vivo Source of Angiogenesis-Inducing MMP-9 in the Tumor Microenvironment. Neoplasia 2014, 16, 771–788. [Google Scholar] [CrossRef] [Green Version]
- Deryugina, E.I.; Quigley, J.P. Pleiotropic roles of matrix metalloproteinases in tumor angiogenesis: Contrasting, overlapping and compensatory functions. Biochim. Biophys. Acta 2010, 1803, 103–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.-Y.; Lin, Y.-C.; Huang, W.-L.; Hong, C.-Q.; Chen, J.-Y.; You, Y.-J.; Li, W.-B. Zoledronic acid inhibits proliferation and impairs migration and invasion through downregulating VEGF and MMPs expression in human nasopharyngeal carcinoma cells. Med. Oncol. 2011, 29, 714–720. [Google Scholar] [CrossRef]
- Dedes, P.; Kanakis, I.; Gialeli, C.; Theocharis, A.; Tsegenidis, T.; Kletsas, D.; Tzanakakis, G.; Karamanos, N. Preclinical evaluation of zoledronate using an in vitro mimetic cellular model for breast cancer metastatic bone disease. Biochim. Biophys. Acta (BBA) Gen. Subj. 2013, 1830, 3625–3634. [Google Scholar] [CrossRef]
- Coleman, R.; Cook, R.; Hirsh, V.; Major, P.; Lipton, A. Zoledronic acid use in cancer patients: More than just supportive care? Cancer 2011, 117, 11–23. [Google Scholar] [CrossRef] [PubMed]
- Sun, T.; Yang, Y.; Luo, X.; Cheng, Y.; Zhang, M.; Wang, K.; Ge, C. Inhibition of tumor angiogenesis by interferon-gamma by suppression of tumor-associated macrophage differentiation. Oncol. Res. 2014, 21, 227–235. [Google Scholar] [CrossRef] [PubMed]
- Choi, H.-J.; Chung, T.-W.; Ha, K.-T. Luteolin inhibits recruitment of monocytes and migration of Lewis lung carcinoma cells by suppressing chemokine (C–C motif) ligand 2 expression in tumor-associated macrophage. Biochem. Biophys. Res. Commun. 2016, 470, 101–106. [Google Scholar] [CrossRef]
- Guo, Z.; Xing, Z.; Cheng, X.; Fang, Z.; Jiang, C.; Su, J.; Zhou, Z.; Xu, Z.; Holmberg, A.; Nilsson, S.; et al. Somatostatin Derivate (smsDX) Attenuates the TAM-Stimulated Prolif-eration, Migration and Invasion of Prostate Cancer via NF-kappaB Regulation. PLoS ONE 2015, 10, e0124292. [Google Scholar]
- Babajani, A.; Soltani, P.; Jamshidi, E.; Farjoo, M.H.; Niknejad, H. Recent Advances on Drug-Loaded Mesenchymal Stem Cells with Anti-neoplastic Agents for Targeted Treatment of Cancer. Front. Bioeng. Biotechnol. 2020, 8, 748. [Google Scholar] [CrossRef] [PubMed]
- Melzer, C.; Rehn, V.; Yang, Y.; Bähre, H.; Von Der Ohe, J.; Hass, R. Taxol-Loaded MSC-Derived Exosomes Provide a Therapeutic Vehicle to Target Metastatic Breast Cancer and Other Carcinoma Cells. Cancers 2019, 11, 798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nicolay, N.H.; Rühle, A.; Perez, R.L.; Trinh, T.; Sisombath, S.; Weber, K.J.; Ho, A.D.; Debus, J.; Saffrich, R.; Huber, P.E. Mesenchymal stem cells are sensitive to bleomycin treatment. Sci. Rep. 2016, 6, 26645. [Google Scholar] [CrossRef]
- Lee, H.K.; Finniss, S.; Cazacu, S.; Bucris, E.; Ziv-Av, A.; Xiang, C.; Bobbitt, K.; Rempel, S.A.; Hasselbach, L.; Mikkelsen, T.; et al. Mesenchymal stem cells deliver synthetic microRNA mimics to glioma cells and glioma stem cells and inhibit their cell migration and self-renewal. Oncotarget 2013, 4, 346–361. [Google Scholar] [CrossRef] [Green Version]
- Shimbo, K.; Miyaki, S.; Ishitobi, H.; Kato, Y.; Kubo, T.; Shimose, S.; Ochi, M. Exosome-formed synthetic microRNA-143 is transferred to osteosarcoma cells and inhibits their migration. Biochem. Biophys. Res. Commun. 2014, 445, 381–387. [Google Scholar] [CrossRef] [PubMed]
- Kauer, T.M.; Figueiredo, J.-L.; Hingtgen, S.; Shah, K. Encapsulated therapeutic stem cells implanted in the tumor resection cavity induce cell death in gliomas. Nat. Neurosci. 2011, 15, 197–204. [Google Scholar] [CrossRef] [Green Version]
- Uluçkan, Ö.; Eagleton, M.C.; Floyd, D.H.; Morgan, E.A.; Hirbe, A.C.; Kramer, M.; Dowland, N.; Prior, J.L.; Piwnica-Worms, D.; Jeong, S.S.; et al. APT102, a novel adpase, cooperates with aspirin to disrupt bone metastasis in mice. J. Cell. Biochem. 2008, 104, 1311–1323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leblanc, R.; Lee, S.C.; David, M.; Bordet, J.C.; Norman, D.D.; Patil, R.; Miller, D.; Sahay, D.; Ribeiro, J.; Ribeiro, P.; et al. Interaction of platelet-derived autotaxin with tumor integrin alphaVbeta3 controls metastasis of breast cancer cells to bone. Blood 2014, 124, 3141–3150. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Sharkey, C.C.; Wun, B.; Liesveld, J.L.; King, M.R. Genetic engineering of platelets to neutralize circulating tumor cells. J. Control. Release 2016, 228, 38–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roy, L.D.; Sahraei, M.; Schettini, J.L.; Gruber, H.E.; Besmer, D.M.; Mukherjee, P. Systemic neutralization of IL-17A significantly reduces breast cancer associated metastasis in arthritic mice by reducing CXCL12/SDF-1 expression in the metastatic niches. BMC Cancer 2014, 14, 225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muller, A.; Homey, B.; Soto, H.; Ge, N.; Catron, D.; Buchanan, M.E.; McClanahan, T.; Murphy, E.; Yuan, W.; Wagner, S.N.; et al. Involvement of chemokine receptors in breast cancer me-tastasis. Nature 2001, 410, 50–56. [Google Scholar] [CrossRef] [PubMed]
- Hobbs, S.K.; Monsky, W.L.; Yuan, F.; Roberts, W.G.; Griffith, L.; Torchilin, V.P.; Jain, R.K. Regulation of transport pathways in tumor vessels: Role of tumor type and microenvironment. Proc. Natl. Acad. Sci. USA 1998, 95, 4607–4612. [Google Scholar] [CrossRef] [Green Version]
- Stylianopoulos, T.; Jain, R.K. Design considerations for nanotherapeutics in oncology. Nanomedicine 2015, 11, 1893–1907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, R.K.; Martin, J.D.; Stylianopoulos, T. The Role of Mechanical Forces in Tumor Growth and Therapy. Annu. Rev. Biomed. Eng. 2014, 16, 321–346. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.; Goel, S.; Duda, D.G.; Fukumura, D.; Jain, R.K. Vascular normalization as an emerging strategy to enhance cancer im-munotherapy. Cancer Res. 2013, 73, 2943–2948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salgaller, M.L. Technology evaluation: Bevacizumab, Genentech/Roche. Curr. Opin. Mol. Ther. 2003, 5, 657–667. [Google Scholar] [PubMed]
- Chauhan, V.P.; Stylianopoulos, T.; Martin, J.D.; Popović, Z.; Chen, O.; Kamoun, W.S.; Bawendi, M.G.; Fukumura, D.; Jain, R.K. Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat. Nanotechnol. 2012, 7, 383–388. [Google Scholar] [CrossRef] [Green Version]
- Stylianopoulos, T.; Jain, R.K. Combining two strategies to improve perfusion and drug delivery in solid tumors. Proc. Natl. Acad. Sci. USA 2013, 110, 18632–18637. [Google Scholar] [CrossRef] [Green Version]
- Heist, R.S.; Duda, D.G.; Sahani, D.V.; Ancukiewicz, M.; Fidias, P.; Sequist, L.V.; Temel, J.S.; Shaw, A.T.; Pennell, N.A.; Neal, J.W.; et al. Improved tumor vascularization after anti-VEGF therapy with carboplatin and nab-paclitaxel associates with survival in lung cancer. Proc. Natl. Acad. Sci. USA 2015, 112, 1547–1552. [Google Scholar] [CrossRef] [Green Version]
- Izumi, Y.; Xu, L.; Di Tomaso, E.; Fukumura, D.; Jain, R.K. Herceptin acts as an anti-angiogenic cocktail. Nat. Cell Biol. 2002, 416, 279–280. [Google Scholar] [CrossRef]
- Rosca, E.V.; Koskimaki, J.E.; Rivera, C.G.; Pandey, N.B.; Tamiz, A.P.; Popel, A.S. Anti-angiogenic peptides for cancer therapeutics. Curr. Pharm. Biotechnol. 2011, 12, 1101–1116. [Google Scholar] [CrossRef]
- Zhang, B.; Shi, W.; Jiang, T.; Wang, L.; Mei, H.; Lu, H.; Hu, Y.; Pang, Z. Optimization of the tumor microenvironment and nanomedicine properties simultaneously to improve tumor therapy. Oncotarget 2016, 7, 62607–62618. [Google Scholar] [CrossRef] [Green Version]
- Jiang, W.; Huang, Y.; An, Y.; Kim, B.Y.S. Remodeling Tumor Vasculature to Enhance Delivery of Intermediate-Sized Nanoparticles. ACS Nano 2015, 9, 8689–8696. [Google Scholar] [CrossRef]
- Sakurai, Y.; Hada, T.; Yamamoto, S.; Kato, A.; Mizumura, W.; Harashima, H. Remodeling of the Extracellular Matrix by Endothelial Cell-Targeting siRNA Improves the EPR-Based Delivery of 100 nm Particles. Mol. Ther. 2016, 24, 2090–2099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smith, N.R.; Baker, D.; Farren, M.; Pommier, A.; Swann, R.; Wang, X.; Mistry, S.; McDaid, K.; Kendrew, J.; Womack, C.; et al. Tumor Stromal Architecture Can Define the Intrinsic Tumor Response to VEGF-Targeted Therapy. Clin. Cancer Res. 2013, 19, 6943–6956. [Google Scholar] [CrossRef] [Green Version]
- Tolaney, S.M.; Boucher, Y.; Duda, D.G.; Martin, J.D.; Seano, G.; Ancukiewicz, M.; Barry, W.T.; Goel, S.; Lahdenrata, J.; Isakoff, S.J.; et al. Role of vascular density and normalization in response to neoadjuvant bevacizumab and chemotherapy in breast cancer patients. Proc. Natl. Acad. Sci. USA 2015, 112, 14325–14330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, W.; Liu, R.; Zhou, Y.; Gao, H. Size-Tunable Strategies for a Tumor Targeted Drug Delivery System. ACS Central Sci. 2020, 6, 100–116. [Google Scholar] [CrossRef] [Green Version]
- Stylianopoulos, T.; Martin, J.D.; Chauhan, V.P.; Jain, S.R.; Diop-Frimpong, B.; Bardeesy, N.; Smith, B.L.; Ferrone, C.R.; Hornicek, F.J.; Boucher, Y.; et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 15101–15108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stylianopoulos, T. The Solid Mechanics of Cancer and Strategies for Improved Therapy. J. Biomech. Eng. 2017, 139. [Google Scholar] [CrossRef] [PubMed]
- Stylianopoulos, T.; Munn, L.L.; Jain, R.K. Reengineering the Tumor Vasculature: Improving Drug Delivery and Efficacy. Trends Cancer 2018, 4, 258–259. [Google Scholar] [CrossRef] [Green Version]
- Mpekris, F.; Panagi, M.; Voutouri, C.; Martin, J.D.; Samuel, R.; Takahashi, S.; Gotohda, N.; Suzuki, T.; Papageorgis, P.; Demetriou, P.; et al. Normalizing the Microenvironment Overcomes Vessel Compression and Resistance to Nano-immunotherapy in Breast Cancer Lung Metastasis. Adv. Sci. 2021, 8. [Google Scholar] [CrossRef]
- Chen, I.X.; Chauhan, V.P.; Posada, J.; Ng, M.R.; Wu, M.W.; Adstamongkonkul, P.; Huang, P.; Lindeman, N.; Langer, R.; Jain, R.K. Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 4558–4566. [Google Scholar] [CrossRef] [Green Version]
- Mpekris, F.; Voutouri, C.; Baish, J.W.; Duda, D.G.; Munn, L.L.; Stylianopoulos, T.; Jain, R.K. Combining microenvironment normalization strategies to improve cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2020, 117, 3728–3737. [Google Scholar] [CrossRef] [Green Version]
- Tsai, M.; Lu, Z.; Wientjes, M.G.; Au, J.L.-S. Paclitaxel-loaded polymeric microparticles: Quantitative relationships between in vitro drug release rate and in vivo pharmacodynamics. J. Control. Release 2013, 172, 737–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tong, R.; Chiang, H.H.; Kohane, D.S. Photoswitchable nanoparticles for in vivo cancer chemotherapy. Proc. Natl. Acad. Sci. USA 2013, 110, 19048–19053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pietras, K.; Rubin, K.; Sjöblom, T.; Buchdunger, E.; Sjöquist, M.; Heldin, C.-H.; Ostman, A. Inhibition of PDGF receptor signaling in tumor stroma enhances antitumor effect of chemotherapy. Cancer Res. 2002, 62, 5476–5484. [Google Scholar] [PubMed]
- Mpekris, F.; Papageorgis, P.; Polydorou, C.; Voutouri, C.; Kalli, M.; Pirentis, A.P.; Stylianopoulos, T. Sonic-hedgehog pathway inhibition nor-malizes desmoplastic tumor microenvironment to improve chemo-and nanotherapy. J. Control. Release 2017, 261, 105–112. [Google Scholar] [CrossRef] [Green Version]
- Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of Hedgehog Signaling Enhances Delivery of Chemotherapy in a Mouse Model of Pancreatic Cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef] [Green Version]
- Rhim, A.D.; Oberstein, P.E.; Thomas, D.H.; Mirek, E.T.; Palermo, C.F.; Sastra, S.A.; Dekleva, E.N.; Saunders, T.; Becerra, C.P.; Tattersall, I.W.; et al. Stromal Elements Act to Restrain, Rather Than Support, Pancreatic Ductal Adenocarcinoma. Cancer Cell 2014, 25, 735–747. [Google Scholar] [CrossRef] [Green Version]
- Ozdemir, B.C.; Pentcheva-Hoang, T.; Carstens, J.L.; Zheng, X.; Wu, C.C.; Simpson, T.R.; Laklai, H.; Sugimoto, H.; Kahlert, C.; Novitskiy, S.V.; et al. Depletion of carcinoma-associated fi-broblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014, 25, 719–734. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Kim, J.; Yang, S.; Wang, H.; Wu, C.-J.; Sugimoto, H.; LeBleu, V.S.; Kalluri, R. Type I collagen deletion in αSMA+ myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell 2021, 39, 548–565.e6. [Google Scholar] [CrossRef]
- Monteran, L.; Erez, N. The Dark Side of Fibroblasts: Cancer-Associated Fibroblasts as Mediators of Immunosuppression in the Tumor Microenvironment. Front. Immunol. 2019, 10, 1835. [Google Scholar] [CrossRef] [Green Version]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holmgaard, R.B.; Schaer, D.A.; Li, Y.; Castaneda, S.P.; Murphy, M.Y.; Xu, X.; Inigo, I.; Dobkin, J.; Manro, J.R.; Iversen, P.W.; et al. Targeting the TGFβ pathway with galunisertib, a TGFβRI small molecule inhibitor, promotes anti-tumor immunity leading to durable, complete responses, as monotherapy and in combination with checkpoint blockade. J. Immunother. Cancer 2018, 6, 47. [Google Scholar] [CrossRef] [PubMed]
- Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel III, E.E.; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Liao, S.; Diop-Frimpong, B.; Chen, W.; Goel, S.; Naxerova, K.; Huang, Y.; Boucher, Y.; Xu, L. TGF-beta blockade improves the distribution and efficacy of therapeutics in breast carcinoma by normalizing the tumor stroma. Proc. Natl. Acad. Sci. USA 2012, 109, 16618–16623. [Google Scholar] [CrossRef] [Green Version]
- De Caestecker, M.P.; Piek, E.; Roberts, A.B. Role of transforming growth factor-beta signaling in cancer. J. Nat. Cancer Inst. 2000, 92, 1388–1402. [Google Scholar] [CrossRef] [Green Version]
- Chauhan, V.P.; Martin, J.D.; Liu, H.; Lacorre, D.A.; Jain, S.R.; Kozin, S.V.; Stylianopoulos, T.; Mousa, A.S.; Han, X.; Adstamongkonkul, P.; et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat. Commun. 2013, 4, 2516. [Google Scholar] [CrossRef] [Green Version]
- Diop-Frimpong, B.; Chauhan, V.P.; Krane, S.; Boucher, Y.; Jain, R.K. Losartan inhibits collagen I synthesis and improves the dis-tribution and efficacy of nanotherapeutics in tumors. Proc. Natl. Acad. Sci. USA 2011, 108, 2909–2914. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Cao, J.; Melamed, A.; Worley, M.; Gockley, A.; Jones, D.; Nia, H.T.; Zhang, Y.; Stylianopoulos, T.; Kumar, A.S.; et al. Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma. Proc. Natl. Acad. Sci. USA 2019, 116, 2210–2219. [Google Scholar] [CrossRef] [Green Version]
- Li, W.; Li, S.; Chen, I.X.; Liu, Y.; Ramjiawan, R.R.; Leung, C.H.; Gerweck, L.E.; Fukumura, D.; Loeffler, J.S.; Jain, R.K.; et al. Combining losartan with radiotherapy increases tumor control and inhibits lung metastases from a HER2/neu-positive orthotopic breast cancer model. Radiat. Oncol. 2021, 16, 48. [Google Scholar] [CrossRef]
- Wilop, S.; von Hobe, S.; Crysandt, M.; Esser, A.; Osieka, R.; Jost, E. Impact of angiotensin I converting enzyme inhibitors and an-giotensin II type 1 receptor blockers on survival in patients with advanced non-small-cell lung cancer undergoing first-line platinum-based chemotherapy. J. Cancer Res. Clin. Oncol. 2009, 135, 1429–1435. [Google Scholar] [CrossRef]
- Keizman, D.; Huang, P.; Eisenberger, M.A.; Pili, R.; Kim, J.J.; Antonarakis, E.S.; Hammers, H.; Carducci, M.A. Angiotensin system inhibitors and outcome of sunitinib treatment in patients with metastatic renal cell carcinoma: A retrospective examination. Eur. J. Cancer 2011, 47, 1955–1961. [Google Scholar] [CrossRef] [Green Version]
- Nakai, Y.; Isayama, H.; Ijichi, H.; Sasaki, T.; Takahara, N.; Ito, Y.; Matsubara, S.; Uchino, R.; Yagioka, H.; Arizumi, T.; et al. A multicenter phase II trial of gemcitabine and candesartan combination therapy in patients with advanced pancreatic cancer: GECA2. Investig. New Drugs 2013, 31, 1294–1299. [Google Scholar] [CrossRef] [PubMed]
- Murphy, J.E.; Wo, J.Y.; Ryan, D.P.; Clark, J.W.; Jiang, W.; Yeap, B.Y.; Drapek, L.C.; Ly, L.; Baglini, C.V.; Blaszkowsky, L.S.; et al. Total neoadjuvant therapy with FOLFIRINOX in combination with losartan followed by chemoradiotherapy for locally advanced pancreatic cancer: A phase 2 clinical trial. JAMA Oncol. 2019, 5, 1020–1027. [Google Scholar] [CrossRef]
- Papageorgis, P.; Polydorou, C.; Mpekris, F.; Voutouri, C.; Agathokleous, E.; Kapnissi-Christodoulou, C.P.; Stylianopoulos, T. Tranilast-induced stress alleviation in solid tumors improves the efficacy of chemo- and nanotherapeutics in a size-independent manner. Sci. Rep. 2017, 7, 46140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panagi, M.; Voutouri, C.; Mpekris, F.; Papageorgis, P.; Martin, M.R.; Martin, J.D.; Demetriou, P.; Pierides, C.; Polydorou, C.; Stylianou, A.; et al. TGF-β inhibition combined with cytotoxic nanomedicine normalizes triple negative breast cancer microenvironment towards anti-tumor immunity. Theranostics 2020, 10, 1910–1922. [Google Scholar] [CrossRef] [PubMed]
- Martin, J.D.; Panagi, M.; Wang, C.; Khan, T.T.; Martin, M.R.; Voutouri, C.; Toh, K.; Papageorgis, P.; Mpekris, F.; Polydorou, C.; et al. Dexamethasone Increases Cisplatin-Loaded Nanocarrier Delivery and Efficacy in Metastatic Breast Cancer by Normalizing the Tumor Microenvironment. ACS Nano 2019, 13, 6396–6408. [Google Scholar] [CrossRef] [PubMed]
- Polydorou, C.; Mpekris, F.; Papageorgis, P.; Voutouri, C.; Stylianopoulos, T. Pirfenidone normalizes the tumor microenvironment to improve chemotherapy. Oncotarget 2017, 8, 24506–24517. [Google Scholar] [CrossRef] [Green Version]
- Incio, J.; Suboj, P.; Chin, S.M.; Vardam-Kaur, T.; Liu, H.; Hato, T.; Babykutty, S.; Chen, I.; Deshpande, V.; Jain, R.K.; et al. Metformin reduces desmoplasia in pancreatic cancer by re-programming stellate cells and tumor-associated macrophages. PLoS ONE 2015, 10, e0141392. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.H.; Shin, B.C.; Park, W.S.; Lee, J.; Kuh, H. Antifibrotic effects of pentoxifylline improve the efficacy of gemcitabine in human pancreatic tumor xenografts. Cancer Sci. 2017, 108, 2470–2477. [Google Scholar] [CrossRef] [PubMed]
- Sherman, M.H.; Yu, R.T.; Engle, D.D.; Ding, N.; Atkins, A.R.; Tiriac, H.; Collisson, E.A.; Connor, F.; Van Dyke, T.; Kozlov, S.; et al. Vitamin D Receptor-Mediated Stromal Reprogramming Suppresses Pancreatitis and Enhances Pancreatic Cancer Therapy. Cell 2014, 159, 80–93. [Google Scholar] [CrossRef] [Green Version]
- Masterson, R.; Hewitson, T.D.; Kelynack, K.; Martic, M.; Parry, L.; Bathgate, R.; Darby, I.; Becker, G. Relaxin down-regulates renal fibroblast function and promotes matrix remodelling in vitro. Nephrol. Dial. Transplant. 2004, 19, 544–552. [Google Scholar] [CrossRef] [Green Version]
- Brown, E.; McKee, T.; diTomaso, E.; Pluen, A.; Seed, B.; Boucher, Y.; Jain, R.K. Dynamic imaging of collagen and its modulation in tumors in vivo using second-harmonic generation. Nat. Med. 2003, 9, 796–800. [Google Scholar] [CrossRef] [PubMed]
- Unemori, E.N.; Amento, E.P. Relaxin modulates synthesis and secretion of procollagenase and collagen by human dermal fi-broblasts. J. Biol. Chem. 1990, 265, 10681–10685. [Google Scholar] [CrossRef]
- Feng, S.; Agoulnik, I.U.; Bogatcheva, N.V.; Kamat, A.A.; Kwabi-Addo, B.; Li, R.; Ayala, G.; Ittmann, M.M.; Agoulnik, A.I. Relaxin Promotes Prostate Cancer Progression. Clin. Cancer Res. 2007, 13, 1695–1702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eltahir, M.; Persson, H.; Mangsbo, S. Tumor localized agonistic anti-CD40 therapy and beyond. Expert Opin. Biol. Ther. 2020, 20, 215–217. [Google Scholar] [CrossRef]
- Khong, A.; Brown, M.D.; Vivian, J.B.; Robinson, B.W.; Currie, A.J. Agonistic anti-CD40 antibody therapy is effective against post-operative cancer recurrence and metastasis in a murine tumor model. J. Immunother. 2013, 36, 365–372. [Google Scholar] [CrossRef]
- Gajewski, T.F.; Schreiber, H.; Fu, Y.-X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [Green Version]
- O’Hara, M.H.; O’Reilly, E.M.; Varadhachary, G.; Wolff, R.A.; Wainberg, Z.A.; Ko, A.H.; Fisher, G.; Rahma, O.; Lyman, J.P.; Cabanski, C.R.; et al. CD40 agonistic monoclonal antibody APX005M (sotigalimab) and chemotherapy, with or without nivolumab, for the treatment of metastatic pancreatic adenocarcinoma: An open-label, multicentre, phase 1b study. Lancet Oncol. 2021, 22, 118–131. [Google Scholar] [CrossRef]
- Sato-Kaneko, F.; Yao, S.; Ahmadi, A.; Zhang, S.S.; Hosoya, T.; Kaneda, M.M.; Varner, J.A.; Pu, M.; Messer, K.S.; Guiducci, C.; et al. Combination immunotherapy with TLR agonists and checkpoint inhibitors suppresses head and neck cancer. JCI Insight 2017, 2. [Google Scholar] [CrossRef]
- Roma-Rodrigues, C.; Pombo, I.; Raposo, L.; Pedrosa, P.; Fernandes, A.R.; Baptista, P.V. Nanotheranostics Targeting the Tumor Mi-croenvironment. Front. Bioeng. Biotechnol. 2019, 7, 197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ali, A.; Kataoka, N.; Ranneh, A.-H.; Iwao, Y.; Noguchi, S.; Oka, T.; Itai, S. Enhancing the Solubility and Oral Bioavailability of Poorly Water-Soluble Drugs Using Monoolein Cubosomes. Chem. Pharm. Bull. 2017, 65, 42–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pettersen, E.O.; Ebbesen, P.; Gieling, R.G.; Williams, K.J.; Dubois, L.; Lambin, P.; Ward, C.; Meehan, J.; Kunkler, I.H.; Langdon, S.P.; et al. Targeting tumour hypoxia to prevent cancer metastasis. From biology, biosensing and technology to drug development: The METOXIA consortium. J. Enzym. Inhib. Med. Chem. 2015, 30, 689–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Potter, C.P.S.; Harris, A.L. Diagnostic, prognostic and therapeutic implications of carbonic anhydrases in cancer. Br. J. Cancer 2003, 89, 2–7. [Google Scholar] [CrossRef] [Green Version]
- Neri, D.; Supuran, C.T. Interfering with pH regulation in tumours as a therapeutic strategy. Nat. Rev. Drug Discov. 2011, 10, 767–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Williams, K.J.; Gieling, R.G. Preclinical Evaluation of Ureidosulfamate Carbonic Anhydrase IX/XII Inhibitors in the Treatment of Cancers. Int. J. Mol. Sci. 2019, 20, 6080. [Google Scholar] [CrossRef] [Green Version]
- Ward, C.; Meehan, J.; Mullen, P.; Supuran, C.; Dixon, J.M.; Thomas, J.S.; Winum, J.-Y.; Lambin, P.; Dubois, L.; Pavathaneni, N.-K.; et al. Evaluation of carbonic anhydrase IX as a therapeutic target for inhibition of breast cancer invasion and metastasis using a series of in vitro breast cancer models. Oncotarget 2015, 6, 24856–24870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lou, Y.; McDonald, P.C.; Oloumi, A.; Chia, S.; Ostlund, C.; Ahmadi, A.; Kyle, A.; Keller, U.A.D.; Leung, S.; Huntsman, D.; et al. Targeting Tumor Hypoxia: Suppression of Breast Tumor Growth and Metastasis by Novel Carbonic Anhydrase IX Inhibitors. Cancer Res. 2011, 71, 3364–3376. [Google Scholar] [CrossRef] [Green Version]
- Wichert, M.; Krall, N. Targeting carbonic anhydrase IX with small organic ligands. Curr. Opin. Chem. Biol. 2015, 26, 48–54. [Google Scholar] [CrossRef]
- Aleman, J.; Skardal, A. A multi-site metastasis-on-a-chip microphysiological system for assessing metastatic preference of cancer cells. Biotechnol. Bioeng. 2019, 116, 936–944. [Google Scholar] [CrossRef]
- Wang, Y.; Wu, D.; Wu, G.; Wu, J.; Lu, S.; Lo, J.; He, Y.; Zhao, C.; Zhao, X.; Zhang, H.; et al. Metastasis-on-a-chip mimicking the progression of kidney cancer in the liver for predicting treatment efficacy. Theranostics 2020, 10, 300–311. [Google Scholar] [CrossRef]
- Walsh, N.C.; Kenney, L.L.; Jangalwe, S.; Aryee, K.-E.; Greiner, D.L.; Brehm, M.A.; Shultz, L.D. Humanized Mouse Models of Clinical Disease. Annu. Rev. Pathol. Mech. Dis. 2017, 12, 187–215. [Google Scholar] [CrossRef] [Green Version]
- Shi, C.; Chen, X.; Tan, D. Development of patient-derived xenograft models of prostate cancer for maintaining tumor het-erogeneity. Trans. Androl. Urol. 2019, 8, 519–528. [Google Scholar] [CrossRef]
- Morton, J.J.; Bird, G.; Refaeli, Y.; Jimeno, A. Humanized Mouse Xenograft Models: Narrowing the Tumor–Microenvironment Gap. Cancer Res. 2016, 76, 6153–6158. [Google Scholar] [CrossRef] [Green Version]
- Pearson, T.; Greiner, D.L.; Shultz, L.D. Creation of “humanized” mice to study human immunity. Curr. Protocols Immunol. 2008, 15, 15–21. [Google Scholar] [CrossRef]
- Lefley, D.; Howard, F.; Arshad, F.; Bradbury, S.; Brown, H.; Tulotta, C.; Eyre, R.; Alférez, D.; Wilkinson, J.M.; Holen, I.; et al. Development of clinically relevant in vivo metastasis models using human bone discs and breast cancer patient-derived xenografts. Breast Cancer Res. 2019, 21, 130. [Google Scholar] [CrossRef] [PubMed]
- Cacho-Diaz, B.; Garcia-Botello, D.R.; Wegman-Ostrosky, T.; Reyes-Soto, G.; Ortiz-Sanchez, E.; Herrera-Montalvo, L.A. Tumor mi-croenvironment differences between primary tumor and brain metastases. J. Trans. Med. 2020, 18, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Connell, J.T.; Sugimoto, H.; Cooke, V.G.; Macdonald, B.A.; Mehta, A.I.; LeBleu, V.S.; Dewar, R.; Rocha, R.M.; Brentani, R.R.; Resnick, M.B.; et al. VEGF-A and Tenascin-C produced by S100A4+ stromal cells are important for metastatic colonization. Proc. Natl. Acad. Sci. USA 2011, 108, 16002–16007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; Macdonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef]
- Duda, D.G.; Duyverman, A.M.M.J.; Kohno, M.; Snuderl, M.; Steller, E.J.A.; Fukumura, D.; Jain, R.K. Malignant cells facilitate lung metastasis by bringing their own soil. Proc. Natl. Acad. Sci. USA 2010, 107, 21677–21682. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Neophytou, C.M.; Panagi, M.; Stylianopoulos, T.; Papageorgis, P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers 2021, 13, 2053. https://doi.org/10.3390/cancers13092053
Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers. 2021; 13(9):2053. https://doi.org/10.3390/cancers13092053
Chicago/Turabian StyleNeophytou, Christiana M., Myrofora Panagi, Triantafyllos Stylianopoulos, and Panagiotis Papageorgis. 2021. "The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities" Cancers 13, no. 9: 2053. https://doi.org/10.3390/cancers13092053
APA StyleNeophytou, C. M., Panagi, M., Stylianopoulos, T., & Papageorgis, P. (2021). The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers, 13(9), 2053. https://doi.org/10.3390/cancers13092053