Mechanically stressed cancer microenvironment: Role in pancreatic cancer progression

Matthew Hadden1, Anubhav Mittal2,3,4, Jaswinder Samra2,3,4, Hala Zreiqat1,5,6, Sumit Sahni2,3,4, Yogambha Ramaswamy1,6
1School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW 2006, Australia
2Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia
3Kolling Institute of Medical Research, University of Sydney, Australia
4Australian Pancreatic Centre, St Leonards, Sydney, Australia
5ARC Training Centre for Innovative BioEngineering, The University of Sydney, NSW 2006, Australia
6The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia

Tài liệu tham khảo

Bray, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA Cancer J. Clin., 68, 394, 10.3322/caac.21492 Rawla, 2019, Epidemiology of pancreatic cancer: global trends, etiology and risk factors, World J. Oncol., 10, 10, 10.14740/wjon1166 Rahib, 2014, Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States, Cancer Res., 74, 2913, 10.1158/0008-5472.CAN-14-0155 Garrido-Laguna, 2015, Pancreatic cancer: from state-of-the-art treatments to promising novel therapies, Nat. Rev. Clin. Oncol., 12, 319, 10.1038/nrclinonc.2015.53 Conroy, 2011, FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer, N. Engl. J. Med., 364, 1817, 10.1056/NEJMoa1011923 Von Hoff, 2013, Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine, N. Engl. J. Med., 369, 1691, 10.1056/NEJMoa1304369 Stopa, 2020, Pancreatic cancer and its microenvironment—recent advances and current controversies, Int. J. Mol. Sci., 21, 3218, 10.3390/ijms21093218 Xu, 2014, Pancreatic cancer and its stroma: a conspiracy theory, World J. Gastroenterol., 20, 11216, 10.3748/wjg.v20.i32.11216 Maitre, 2016, Asymmetric division of contractile domains couples cell positioning and fate specification, Nature, 536, 344, 10.1038/nature18958 Zeng, 2018, PIEZOs mediate neuronal sensing of blood pressure and the baroreceptor reflex, Science, 362, 464, 10.1126/science.aau6324 Geffeney, 2012, How we feel: ion channel partnerships that detect mechanical inputs and give rise to touch and pain perception, Neuron, 74, 609, 10.1016/j.neuron.2012.04.023 Engler, 2006, Matrix elasticity directs stem cell lineage specification, Cell, 126, 677, 10.1016/j.cell.2006.06.044 Reffay, 2014, Interplay of RhoA and mechanical forces in collective cell migration driven by leader cells, Nat. Cell Biol., 16, 217, 10.1038/ncb2917 Jansen, 2015, A guide to mechanobiology: Where biology and physics meet, Biochim. Biophys. Acta, 1853, 3043, 10.1016/j.bbamcr.2015.05.007 Fesinmeyer, 2005, Differences in survival by histologic type of pancreatic cancer, Cancer Epidemiol. Biomark. Prev., 14, 1766, 10.1158/1055-9965.EPI-05-0120 Jun, 2016, Nonductal pancreatic cancers, Surg. Pathol. Clin., 9, 581, 10.1016/j.path.2016.05.005 Klimstra, 2007, Nonductal neoplasms of the pancreas, Mod. Pathol., 20, S94, 10.1038/modpathol.3800686 Luchini, 2016, Pancreatic ductal adenocarcinoma and its variants, Surg. Pathol. Clin., 9, 547, 10.1016/j.path.2016.05.003 Papavramidis, 2005, Solid pseudopapillary tumors of the pancreas: review of 718 patients reported in English literature, J. Am. Coll. Surg., 200, 965, 10.1016/j.jamcollsurg.2005.02.011 Halfdanarson, 2008, Pancreatic endocrine neoplasms: epidemiology and prognosis of pancreatic endocrine tumors, Endocr. Relat. Cancer, 15, 409, 10.1677/ERC-07-0221 Peters, 2018, Progression to pancreatic ductal adenocarcinoma from pancreatic intraepithelial neoplasia: results of a simulation model, Pancreatology, 18, 928, 10.1016/j.pan.2018.07.009 Yachida, 2010, Distant metastasis occurs late during the genetic evolution of pancreatic cancer, Nature, 467, 1114, 10.1038/nature09515 Pittman, 2017, Classification, morphology, molecular pathogenesis, and outcome of premalignant lesions of the pancreas, Arch. Pathol. Lab. Med., 141, 1606, 10.5858/arpa.2016-0426-RA Kim, 2018, Precursor lesions of pancreatic cancer, Oncol. Res. Treat., 41, 603, 10.1159/000493554 Hruban, 2001, Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions, Am. J. Surg. Pathol., 25, 579, 10.1097/00000478-200105000-00003 Hingorani, 2003, Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse, Cancer Cell, 4, 437, 10.1016/S1535-6108(03)00309-X Downward, 2003, Targeting RAS signalling pathways in cancer therapy, Nat. Rev. Cancer, 3, 11, 10.1038/nrc969 Gillson, 2020, Small molecule KRAS inhibitors: the future for targeted pancreatic cancer therapy?, Cancers, 12, 1341, 10.3390/cancers12051341 Eser, 2014, Oncogenic KRAS signalling in pancreatic cancer, Br. J. Cancer, 111, 817, 10.1038/bjc.2014.215 Morris, 2010, KRAS, Hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma, Nat. Rev. Cancer, 10, 683, 10.1038/nrc2899 Biankin, 2012, Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes, Nature, 491, 399, 10.1038/nature11547 Bockman, 2003, Origin and development of the precursor lesions in experimental pancreatic cancer in rats, Lab. Investig., 83, 853, 10.1097/01.LAB.0000074918.31303.5A Friedlander, 2009, Context-dependent transformation of adult pancreatic cells by oncogenic K-Ras, Cancer Cell, 16, 379, 10.1016/j.ccr.2009.09.027 Pour, 2003, What is the origin of pancreatic adenocarcinoma?, Mol. Cancer, 2, 13, 10.1186/1476-4598-2-13 Perera, 2012, Ready, set, go: the EGF receptor at the pancreatic cancer starting line, Cancer Cell, 22, 281, 10.1016/j.ccr.2012.08.019 Xu, 2019, Ductal vs. acinar? Recent insights into identifying cell lineage of pancreatic ductal adenocarcinoma, Ann Pancreat. Cancer, 2, 11, 10.21037/apc.2019.06.03 Kopp, 2012, Identification of Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma, Cancer Cell, 22, 737, 10.1016/j.ccr.2012.10.025 Ray, 2011, Epithelial tissues have varying degrees of susceptibility to Kras(G12D)-initiated tumorigenesis in a mouse model, PLoS One, 6, 10.1371/journal.pone.0016786 Guerra, 2007, Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice, Cancer Cell, 11, 291, 10.1016/j.ccr.2007.01.012 De La, 2008, Notch and Kras reprogram pancreatic acinar cells to ductal intraepithelial neoplasia, Proc. Natl. Acad. Sci., 105, 18907, 10.1073/pnas.0810111105 Habbe, 2008, Spontaneous induction of murine pancreatic intraepithelial neoplasia (mPanIN) by acinar cell targeting of oncogenic Kras in adult mice, Proc. Natl. Acad. Sci. U. S. A., 105, 18913, 10.1073/pnas.0810097105 Shi, 2009, Loss of the acinar-restricted transcription factor Mist1 accelerates Kras-induced pancreatic intraepithelial neoplasia, Gastroenterology, 136, 1368, 10.1053/j.gastro.2008.12.066 Ferreira, 2017, Duct- and acinar-derived pancreatic ductal adenocarcinomas show distinct tumor progression and marker expression, Cell Rep., 21, 966, 10.1016/j.celrep.2017.09.093 Storz, 2017, Acinar cell plasticity and development of pancreatic ductal adenocarcinoma, Nat. Rev. Gastroenterol. Hepatol., 14, 296, 10.1038/nrgastro.2017.12 Houbracken, 2011, Lineage tracing evidence for transdifferentiation of acinar to duct cells and plasticity of human pancreas, Gastroenterology, 141, 731, 10.1053/j.gastro.2011.04.050 Shi, 2013, Maintenance of acinar cell organization is critical to preventing Kras-induced acinar-ductal metaplasia, Oncogene, 32, 1950, 10.1038/onc.2012.210 Gao, 2019, PYK2 Is involved in premalignant acinar cell reprogramming and pancreatic ductal adenocarcinoma maintenance by phosphorylating beta-catenin (Y654), Cell Mol. Gastroenterol. Hepatol., 8, 561, 10.1016/j.jcmgh.2019.07.004 Wang, 2019, ATDC is required for the initiation of KRAS-induced pancreatic tumorigenesis, Genes Dev., 33, 641, 10.1101/gad.323303.118 Li, 2019, MSP-RON signaling is activated in the transition from pancreatic intraepithelial neoplasia (PanIN) to pancreatic ductal adenocarcinoma (PDAC), Front. Physiol., 10, 147, 10.3389/fphys.2019.00147 Babicky, 2019, MST1R kinase accelerates pancreatic cancer progression via effects on both epithelial cells and macrophages, Oncogene, 38, 5599, 10.1038/s41388-019-0811-9 Chen, 2015, NFATc1 links EGFR signaling to induction of Sox9 transcription and acinar-ductal transdifferentiation in the pancreas, Gastroenterology, 148, 1024, 10.1053/j.gastro.2015.01.033 Liu, 2016, TGF-beta1 promotes acinar to ductal metaplasia of human pancreatic acinar cells, Sci. Rep., 6, 30904, 10.1038/srep30904 Grabliauskaite, 2016, Inactivation of TGFbeta receptor II signalling in pancreatic epithelial cells promotes acinar cell proliferation, acinar-to-ductal metaplasia and fibrosis during pancreatitis, J. Pathol., 238, 434, 10.1002/path.4666 Wei, 2016, KLF4 Is essential for induction of cellular identity change and acinar-to-ductal reprogramming during early pancreatic carcinogenesis, Cancer Cell, 29, 324, 10.1016/j.ccell.2016.02.005 Nishikawa, 2019, Hes1 plays an essential role in Kras-driven pancreatic tumorigenesis, Oncogene, 38, 4283, 10.1038/s41388-019-0718-5 Feig, 2012, The pancreas cancer microenvironment, Clin. Cancer Res., 18, 4266, 10.1158/1078-0432.CCR-11-3114 Kibe, 2019, Cancer-associated acinar-to-ductal metaplasia within the invasive front of pancreatic cancer contributes to local invasion, Cancer Lett., 444, 70, 10.1016/j.canlet.2018.12.005 Liu, 2016, Genetic ablation of Smoothened in pancreatic fibroblasts increases acinar-ductal metaplasia, Genes Dev., 30, 1943, 10.1101/gad.283499.116 Sheetz, 1998, Cell migration: regulation of force on extracellular-matrix-integrin complexes, Trends Cell Biol., 8, 51, 10.1016/S0962-8924(98)80005-6 Panciera, 2017, Mechanobiology of YAP and TAZ in physiology and disease, Nat. Rev. Mol. Cell Biol., 18, 758, 10.1038/nrm.2017.87 Pan, 2010, The hippo signaling pathway in development and cancer, Dev. Cell, 19, 491, 10.1016/j.devcel.2010.09.011 Beyer, 2013, Switch enhancers interpret TGF-beta and Hippo signaling to control cell fate in human embryonic stem cells, Cell Rep., 5, 1611, 10.1016/j.celrep.2013.11.021 Zhong, 2013, Mesenchymal stem cell and chondrocyte fates in a multishear microdevice are regulated by Yes-associated protein, Stem Cells Dev., 22, 2083, 10.1089/scd.2012.0685 Wang, 2016, Flow-dependent YAP/TAZ activities regulate endothelial phenotypes and atherosclerosis, Proc. Natl. Acad. Sci. U. S. A., 113, 11525, 10.1073/pnas.1613121113 Elbediwy, 2016, Integrin signalling regulates YAP and TAZ to control skin homeostasis, Development, 143, 1674, 10.1242/dev.133728 Liu, 2015, Mechanosignaling through YAP and TAZ drives fibroblast activation and fibrosis, Am. J. Phys. Lung Cell. Mol. Phys., 308, L344 Dupont, 2011, Role of YAP/TAZ in mechanotransduction, Nature, 474, 179, 10.1038/nature10137 Zanconato, 2016, YAP/TAZ at the roots of cancer, Cancer Cell, 29, 783, 10.1016/j.ccell.2016.05.005 Kapoor, 2014, Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer, Cell, 158, 185, 10.1016/j.cell.2014.06.003 Xie, 2015, Hippo transducer TAZ promotes epithelial mesenchymal transition and supports pancreatic cancer progression, Oncotarget, 6, 35949, 10.18632/oncotarget.5772 Yang, 2015, Active YAP promotes pancreatic cancer cell motility, invasion and tumorigenesis in a mitotic phosphorylation-dependent manner through LPAR3, Oncotarget, 6, 36019, 10.18632/oncotarget.5935 Diep, 2012, Down-regulation of Yes Associated Protein 1 expression reduces cell proliferation and clonogenicity of pancreatic cancer cells, PLoS One, 7, 10.1371/journal.pone.0032783 Allende, 2017, Overexpression of Yes associated protein 1, an independent prognostic marker in patients with pancreatic ductal adenocarcinoma, correlated with liver metastasis and poor prognosis, Pancreas, 46, 913, 10.1097/MPA.0000000000000867 Morvaridi, 2015, Role of YAP and TAZ in pancreatic ductal adenocarcinoma and in stellate cells associated with cancer and chronic pancreatitis, Sci. Rep., 5, 16759, 10.1038/srep16759 Murakami, 2017, Yes-associated protein mediates immune reprogramming in pancreatic ductal adenocarcinoma, Oncogene, 36, 1232, 10.1038/onc.2016.288 Yuan, 2016, YAP overexpression promotes the epithelial-mesenchymal transition and chemoresistance in pancreatic cancer cells, Mol. Med. Rep., 13, 237, 10.3892/mmr.2015.4550 Gopal, 2019, Targeting cell surface-associated GRP78 enhances pancreatic cancer radiosensitivity by reducing YAP/TAZ protein signaling, J. Biol. Chem., 294, 13939, 10.1074/jbc.RA119.009091 Totaro, 2018, YAP/TAZ upstream signals and downstream responses, Nat. Cell Biol., 20, 888, 10.1038/s41556-018-0142-z Galluzzi, 2015, Autophagy in malignant transformation and cancer progression, EMBO J., 34, 856, 10.15252/embj.201490784 Perera, 2015, Transcriptional control of autophagy–lysosome function drives pancreatic cancer metabolism, Nature, 524, 361, 10.1038/nature14587 Zhou, 2019, YAP promotes multi-drug resistance and inhibits autophagy-related cell death in hepatocellular carcinoma via the RAC1-ROS-mTOR pathway, Cancer Cell Int., 19, 179, 10.1186/s12935-019-0898-7 Rozengurt, 2019, Central role of Yes-associated protein and WW-domain-containing transcriptional co-activator with PDZ-binding motif in pancreatic cancer development, World J. Gastroenterol., 25, 1797, 10.3748/wjg.v25.i15.1797 Ansari, 2019, The hippo signaling pathway in pancreatic cancer, Anticancer Res., 39, 3317, 10.21873/anticanres.13474 Zhang, 2016, Fbxw7 deletion accelerates KrasG12D-Driven pancreatic tumorigenesis via Yap accumulation, Neoplasia, 18, 666, 10.1016/j.neo.2016.08.009 Gruber, 2016, YAP1 and TAZ control pancreatic cancer initiation in mice by direct up-regulation of JAK-STAT3 signaling, Gastroenterology, 151, 526, 10.1053/j.gastro.2016.05.006 Albini, 2007, The tumour microenvironment as a target for chemoprevention, Nat. Rev. Cancer, 7, 139, 10.1038/nrc2067 Yu, 2011, Forcing form and function: biomechanical regulation of tumor evolution, Trends Cell Biol., 21, 47, 10.1016/j.tcb.2010.08.015 Dougan, 2017, The pancreatic cancer microenvironment, Cancer J., 23, 321, 10.1097/PPO.0000000000000288 Erkan, 2012, The role of stroma in pancreatic cancer: diagnostic and therapeutic implications, Nat. Rev. Gastroenterol. Hepatol., 9, 454, 10.1038/nrgastro.2012.115 Nguyen, 2016, Stiffness of pancreatic cancer cells is associated with increased invasive potential, Integr. Biol. (Camb.), 8, 1232, 10.1039/C6IB00135A Provenzano, 2013, Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer, Br. J. Cancer, 108, 1, 10.1038/bjc.2012.569 Chaudhuri, 2018, Mechanobiology of tumor growth, Chem. Rev., 118, 6499, 10.1021/acs.chemrev.8b00042 Ercan, 2017, Pancreatic cancer stem cells and therapeutic approaches, Anticancer Res., 37, 2761 Feig, 2012, The pancreas cancer microenvironment, Clin. Cancer Res., 18, 4266, 10.1158/1078-0432.CCR-11-3114 Bynigeri, 2017, Pancreatic stellate cell: Pandora's box for pancreatic disease biology, World J. Gastroenterol., 23, 382, 10.3748/wjg.v23.i3.382 Xiao, 2015, Retinoic acid ameliorates pancreatic fibrosis and inhibits the activation of pancreatic stellate cells in mice with experimental chronic pancreatitis via suppressing the Wnt/beta-catenin signaling pathway, PLoS One, 10, 10.1371/journal.pone.0141462 Lee, 2017, A role of pancreatic stellate cells in islet fibrosis and beta-cell dysfunction in type 2 diabetes mellitus, Biochem. Biophys. Res. Commun., 485, 328, 10.1016/j.bbrc.2017.02.082 Apte, 2004, Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells, Pancreas, 29, 179, 10.1097/00006676-200410000-00002 Phillips, 2003, Rat pancreatic stellate cells secrete matrix metalloproteinases: implications for extracellular matrix turnover, Gut, 52, 275, 10.1136/gut.52.2.275 Barnard, 2009, Synthetic retinoids: structure-activity relationships, Chemistry, 15, 11430, 10.1002/chem.200901952 Rosewicz, 1995, Retinoids: effects on growth, differentiation, and nuclear receptor expression in human pancreatic carcinoma cell lines, Gastroenterology, 109, 1646, 10.1016/0016-5085(95)90655-X Tulachan, 2003, All-trans retinoic acid induces differentiation of ducts and endocrine cells by mesenchymal/epithelial interactions in embryonic pancreas, Diabetes, 52, 76, 10.2337/diabetes.52.1.76 Ferdek, 2017, Biology of pancreatic stellate cells-more than just pancreatic cancer, Pflugers Arch., 469, 1039, 10.1007/s00424-017-1968-0 Apte, 2012, Pancreatic stellate cells: a starring role in normal and diseased pancreas, Front. Physiol., 3, 344, 10.3389/fphys.2012.00344 Jaster, 2004, Molecular regulation of pancreatic stellate cell function, Mol. Cancer, 3, 26, 10.1186/1476-4598-3-26 Bachem, 2005, Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells, Gastroenterology, 128, 907, 10.1053/j.gastro.2004.12.036 Vonlaufen, 2008, Pancreatic stellate cells: partners in crime with pancreatic cancer cells, Cancer Res., 68, 2085, 10.1158/0008-5472.CAN-07-2477 Sarper, 2016, ATRA modulates mechanical activation of TGF-beta by pancreatic stellate cells, Sci. Rep., 6, 27639, 10.1038/srep27639 Hinz, 2007, Formation and function of the myofibroblast during tissue repair, J. Investig. Dermatol., 127, 526, 10.1038/sj.jid.5700613 Tomasek, 2002, Myofibroblasts and mechano-regulation of connective tissue remodelling, Nat. Rev. Mol. Cell Biol., 3, 349, 10.1038/nrm809 Lachowski, 2017, Substrate rigidity controls activation and durotaxis in pancreatic stellate cells, Sci. Rep., 7, 2506, 10.1038/s41598-017-02689-x Chronopoulos, 2016, ATRA mechanically reprograms pancreatic stellate cells to suppress matrix remodelling and inhibit cancer cell invasion, Nat. Commun., 7, 12630, 10.1038/ncomms12630 Di Maggio, 2016, Pancreatic stellate cells regulate blood vessel density in the stroma of pancreatic ductal adenocarcinoma, Pancreatology, 16, 995, 10.1016/j.pan.2016.05.393 Cortes, 2019, GPER is a mechanoregulator of pancreatic stellate cells and the tumor microenvironment, EMBO Rep., 20, 10.15252/embr.201846556 Xiao, 2019, YAP1-mediated pancreatic stellate cell activation inhibits pancreatic cancer cell proliferation, Cancer Lett., 462, 51, 10.1016/j.canlet.2019.07.015 Masamune, 2008, Hypoxia stimulates pancreatic stellate cells to induce fibrosis and angiogenesis in pancreatic cancer, Am. J. Physiol. Gastrointest. Liver Physiol., 295, G709, 10.1152/ajpgi.90356.2008 Erkan, 2009, Cancer-stellate cell interactions perpetuate the hypoxia-fibrosis cycle in pancreatic ductal adenocarcinoma, Neoplasia (New York, N.Y.), 11, 497, 10.1593/neo.81618 Spivak-Kroizman, 2013, Hypoxia triggers hedgehog-mediated tumor-stromal interactions in pancreatic cancer, Cancer Res., 73, 3235, 10.1158/0008-5472.CAN-11-1433 Eguchi, 2013, Hypoxia enhances the interaction between pancreatic stellate cells and cancer cells via increased secretion of connective tissue growth factor, J. Surg. Res., 181, 225, 10.1016/j.jss.2012.06.051 Bennewith, 2009, The role of tumor cell-derived connective tissue growth factor (CTGF/CCN2) in pancreatic tumor growth, Cancer Res., 69, 775, 10.1158/0008-5472.CAN-08-0987 Lee, 2014, Stromal response to Hedgehog signaling restrains pancreatic cancer progression, Proc. Natl. Acad. Sci., 111, E3091, 10.1073/pnas.1411679111 Özdemir, 2014, Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival, Cancer Cell, 25, 719, 10.1016/j.ccr.2014.04.005 Rhim, 2014, Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma, Cancer Cell, 25, 735, 10.1016/j.ccr.2014.04.021 Özdemir, 2014, Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival, Cancer Cell, 25, 719, 10.1016/j.ccr.2014.04.005 Rhim, 2014, Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma, Cancer Cell, 25, 735, 10.1016/j.ccr.2014.04.021 Miyai, 2020, Cancer-associated fibroblasts that restrain cancer progression: hypotheses and perspectives, Cancer Sci., 111, 1047, 10.1111/cas.14346 Neuzillet, 2019, Inter- and intra-tumoural heterogeneity in cancer-associated fibroblasts of human pancreatic ductal adenocarcinoma, J. Pathol., 248, 51, 10.1002/path.5224 Thomas, 2020, Pancreatic stellate cells: the key orchestrator of the pancreatic tumor microenvironment, 57 Mizutani, 2019, Meflin-positive cancer-associated fibroblasts inhibit pancreatic carcinogenesis, Cancer Res., 79, 5367, 10.1158/0008-5472.CAN-19-0454 Qiu, 2015, Cancer stem cells: a potential target for cancer therapy, Cell. Mol. Life Sci., 72, 3411, 10.1007/s00018-015-1920-4 Li, 2007, Identification of pancreatic cancer stem cells, Cancer Res., 67, 1030, 10.1158/0008-5472.CAN-06-2030 Hermann, 2007, Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer, Cell Stem Cell, 1, 313, 10.1016/j.stem.2007.06.002 Rao, 2015, New insights into pancreatic cancer stem cells, World J. Stem Cells, 7, 547, 10.4252/wjsc.v7.i3.547 Lonardo, 2012, Pancreatic stellate cells form a niche for cancer stem cells and promote their self-renewal and invasiveness, Cell Cycle, 11, 1282, 10.4161/cc.19679 Hamada, 2012, Pancreatic stellate cells enhance stem cell-like phenotypes in pancreatic cancer cells, Biochem. Biophys. Res. Commun., 421, 349, 10.1016/j.bbrc.2012.04.014 Glumac, 2018, The role of CD133 in cancer: a concise review, Clin. Transl. Med., 7, 18, 10.1186/s40169-018-0198-1 Wang, 2018, The role of CD44 and cancer stem cells, Methods Mol. Biol., 1692, 31, 10.1007/978-1-4939-7401-6_3 Biondani, 2018, Extracellular matrix composition modulates PDAC parenchymal and stem cell plasticity and behavior through the secretome, FEBS J., 285, 2104, 10.1111/febs.14471 Kalluri, 2003, Basement membranes: structure, assembly and role in tumour angiogenesis, Nat. Rev. Cancer, 3, 422, 10.1038/nrc1094 Theocharis, 2016, Extracellular matrix structure, Adv. Drug Deliv. Rev., 97, 4, 10.1016/j.addr.2015.11.001 Clause, 2013, Extracellular matrix signaling in morphogenesis and repair, Curr. Opin. Biotechnol., 24, 830, 10.1016/j.copbio.2013.04.011 Frantz, 2010, The extracellular matrix at a glance, J. Cell Sci., 123, 4195, 10.1242/jcs.023820 Theocharis, 2012, 3 De Paepe, 2012, The Ehlers-Danlos syndrome, a disorder with many faces, Clin. Genet., 82, 1, 10.1111/j.1399-0004.2012.01858.x Perrucci, 2017, Vascular smooth muscle cells in Marfan syndrome aneurysm: the broken bricks in the aortic wall, Cell. Mol. Life Sci., 74, 267, 10.1007/s00018-016-2324-9 Kwak, 2014, Lung cancer risk among patients with combined pulmonary fibrosis and emphysema, Respir. Med., 108, 524, 10.1016/j.rmed.2013.11.013 Sim, 2018, Hepatocellular carcinoma in the era of immunotherapy, Curr. Probl. Cancer, 42, 40, 10.1016/j.currproblcancer.2017.10.007 Acerbi, 2015, Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration, Integr. Biol. (Camb.), 7, 1120, 10.1039/c5ib00040h Monboisse, 2014, Matrikines from basement membrane collagens: a new anti-cancer strategy, Biochim. Biophys. Acta, 1840, 2589, 10.1016/j.bbagen.2013.12.029 Laklai, 2016, Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression, Nat. Med., 22, 497, 10.1038/nm.4082 Imamura, 1995, Quantitative analysis of collagen and collagen subtypes I, III, and V in human pancreatic cancer, tumor-associated chronic pancreatitis, and alcoholic chronic pancreatitis, Pancreas, 11, 357, 10.1097/00006676-199511000-00007 Frantz, 2010, The extracellular matrix at a glance, J. Cell Sci., 123, 4195, 10.1242/jcs.023820 Bonnans, 2014, Remodelling the extracellular matrix in development and disease, Nat. Rev. Mol. Cell Biol., 15, 786, 10.1038/nrm3904 Morla, 2019, Glycosaminoglycans and glycosaminoglycan mimetics in cancer and inflammation, Int. J. Mol. Sci., 20, 10.3390/ijms20081963 Melstrom, 2017, The pancreatic cancer microenvironment: a true double agent, J. Surg. Oncol., 116, 7, 10.1002/jso.24643 Karsdal, 2017, The good and the bad collagens of fibrosis - their role in signaling and organ function, Adv. Drug Deliv. Rev., 121, 43, 10.1016/j.addr.2017.07.014 Olivares, 2017, Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions, Nat. Commun., 8, 16031, 10.1038/ncomms16031 Linder, 2001, Immunohistochemical expression of extracellular matrix proteins and adhesion molecules in pancreatic carcinoma, Hepatogastroenterology, 48, 1321 Whatcott, 2015, Desmoplasia in primary tumors and metastatic lesions of pancreatic cancer, Clin. Cancer Res., 21, 3561, 10.1158/1078-0432.CCR-14-1051 Lu, 2014, Pancreatic stellate cells promote hapto-migration of cancer cells through collagen I-mediated signalling pathway, Br. J. Cancer, 110, 409, 10.1038/bjc.2013.706 Begum, 2017, The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma, PLoS One, 12, 10.1371/journal.pone.0180181 Duan, 2014, The activation of beta1-integrin by type i collagen coupling with the hedgehog pathway promotes the epithelial-mesenchymal transition in pancreatic cancer, Curr. Cancer Drug Targets, 14, 446, 10.2174/1568009614666140402105101 Weniger, 2018, The extracellular matrix and pancreatic cancer: a complex relationship, Cancers (Basel), 10, 10.3390/cancers10090316 Grzesiak, 2005, Type I collagen and divalent cation shifts disrupt cell-cell adhesion, increase migration, and decrease PTHrP, IL-6, and IL-8 expression in pancreatic cancer cells, Int. J. Gastrointest. Cancer, 36, 131, 10.1385/IJGC:36:3:131 Clementz, 2013, Collagen XV inhibits epithelial to mesenchymal transition in pancreatic adenocarcinoma cells, PLoS One, 8, 10.1371/journal.pone.0072250 Amenta, 2000, Type XV collagen in human colonic adenocarcinomas has a different distribution than other basement membrane zone proteins, Hum. Pathol., 31, 359, 10.1016/S0046-8177(00)80251-8 Amenta, 2003, Loss of types XV and XIX collagen precedes basement membrane invasion in ductal carcinoma of the female breast, J. Pathol., 199, 298, 10.1002/path.1303 van der Zee, 2012, Tumour basement membrane laminin expression predicts outcome following curative resection of pancreatic head cancer, Br. J. Cancer, 107, 1153, 10.1038/bjc.2012.373 Procacci, 2018, Tumor(-)stroma cross-talk in human pancreatic ductal adenocarcinoma: a focus on the effect of the extracellular matrix on tumor cell phenotype and invasive potential, Cells, 7, 158, 10.3390/cells7100158 Ohlund, 2009, Type IV collagen is a tumour stroma-derived biomarker for pancreas cancer, Br. J. Cancer, 101, 91, 10.1038/sj.bjc.6605107 Ohlund, 2013, Type IV collagen stimulates pancreatic cancer cell proliferation, migration, and inhibits apoptosis through an autocrine loop, BMC Cancer, 13, 154, 10.1186/1471-2407-13-154 Paszek, 2005, Tensional homeostasis and the malignant phenotype, Cancer Cell, 8, 241, 10.1016/j.ccr.2005.08.010 Rice, 2017, Matrix stiffness induces epithelial-mesenchymal transition and promotes chemoresistance in pancreatic cancer cells, Oncogenesis, 6, 10.1038/oncsis.2017.54 Haage, 2014, Cellular contractility and extracellular matrix stiffness regulate matrix metalloproteinase activity in pancreatic cancer cells, FASEB J., 28, 3589, 10.1096/fj.13-245613 Cox, 2013, LOX-mediated collagen crosslinking is responsible for fibrosis-enhanced metastasis, Cancer Res., 73, 1721, 10.1158/0008-5472.CAN-12-2233 Lee, 2015, Tissue transglutaminase mediated tumor-stroma interaction promotes pancreatic cancer progression, Clin. Cancer Res., 21, 4482, 10.1158/1078-0432.CCR-15-0226 Miller, 2015, Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: inhibition of LOX abrogates metastasis and enhances drug efficacy, EMBO Mol. Med., 7, 1063, 10.15252/emmm.201404827 Ohtsubo, 2006, Glycosylation in cellular mechanisms of health and disease, Cell, 126, 855, 10.1016/j.cell.2006.08.019 Pan, 2014, Quantitative glycoproteomics analysis reveals changes in N-glycosylation level associated with pancreatic ductal adenocarcinoma, J. Proteome Res., 13, 1293, 10.1021/pr4010184 Pan, 2009, Quantitative proteomics investigation of pancreatic intraepithelial neoplasia, Electrophoresis, 30, 1132, 10.1002/elps.200800752 Chen, 2012, Stromal galectin-1 expression is associated with long-term survival in resectable pancreatic ductal adenocarcinoma, Cancer Biol. Ther., 13, 899, 10.4161/cbt.20842 Chen, 2015, Proteins associated with pancreatic cancer survival in patients with resectable pancreatic ductal adenocarcinoma, Lab. Investig., 95, 43, 10.1038/labinvest.2014.128 Liu, 2015, Role of pancreatic stellate cells and periostin in pancreatic cancer progression, Tumour Biol., 36, 3171, 10.1007/s13277-015-3386-2 Baril, 2007, Periostin promotes invasiveness and resistance of pancreatic cancer cells to hypoxia-induced cell death: role of the beta4 integrin and the PI3k pathway, Oncogene, 26, 2082, 10.1038/sj.onc.1210009 Kanno, 2008, Periostin, secreted from stromal cells, has biphasic effect on cell migration and correlates with the epithelial to mesenchymal transition of human pancreatic cancer cells, Int. J. Cancer, 122, 2707, 10.1002/ijc.23332 Liu, 2017, Role of microenvironmental periostin in pancreatic cancer progression, Oncotarget, 8, 89552, 10.18632/oncotarget.11533 Sahni, 2020, Identification of novel biomarkers in pancreatic tumor tissue to predict response to neoadjuvant chemotherapy, Front. Oncol., 10, 237, 10.3389/fonc.2020.00237 Martinez-Bosch, 2014, Galectin-1 drives pancreatic carcinogenesis through stroma remodeling and Hedgehog signaling activation, Cancer Res., 74, 3512, 10.1158/0008-5472.CAN-13-3013 Orozco, 2018, Targeting galectin-1 inhibits pancreatic cancer progression by modulating tumor-stroma crosstalk, Proc. Natl. Acad. Sci. U. S. A., 115, E3769, 10.1073/pnas.1722434115 Zeltz, 2014, Molecular composition and function of integrin-based collagen glues-introducing COLINBRIs, Biochim. Biophys. Acta, 1840, 2533, 10.1016/j.bbagen.2013.12.022 Qazi, 2011, Recent advances in underlying pathologies provide insight into interleukin-8 expression-mediated inflammation and angiogenesis, Int. J. Inflamm., 2011, 908468, 10.4061/2011/908468 Shi, 1999, Constitutive and inducible interleukin 8 expression by hypoxia and acidosis renders human pancreatic cancer cells more tumorigenic and metastatic, Clin. Cancer Res., 5, 3711 Yao, 2011, Role of alpha(5)beta(1) integrin up-regulation in radiation-induced invasion by human pancreatic cancer cells, Transl. Oncol., 4, 282, 10.1593/tlo.11133 Miyamoto, 2004, Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins, Pancreas, 28, 38, 10.1097/00006676-200401000-00006 Edderkaoui, 2005, Extracellular matrix stimulates reactive oxygen species production and increases pancreatic cancer cell survival through 5-lipoxygenase and NADPH oxidase, Am. J. Physiol. Gastrointest. Liver Physiol., 289, G1137, 10.1152/ajpgi.00197.2005 Vaquero, 2004, Reactive oxygen species produced by NAD(P)H oxidase inhibit apoptosis in pancreatic cancer cells, J. Biol. Chem., 279, 34643, 10.1074/jbc.M400078200 Donadelli, 2011, Gemcitabine/cannabinoid combination triggers autophagy in pancreatic cancer cells through a ROS-mediated mechanism, Cell Death Dis., 2, 10.1038/cddis.2011.36 Oskarsson, 2011, Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs, Nat. Med., 17, 867, 10.1038/nm.2379 Li, 2016, Proteomic analysis of stromal proteins in different stages of colorectal cancer establishes Tenascin-C as a stromal biomarker for colorectal cancer metastasis, Oncotarget, 7, 37226, 10.18632/oncotarget.9362 Giblin, 2015, Tenascin-C: form versus function, Cell Adhes. Migr., 9, 48, 10.4161/19336918.2014.987587 Esposito, 2006, Tenascin C and annexin II expression in the process of pancreatic carcinogenesis, J. Pathol., 208, 673, 10.1002/path.1935 Cai, 2018, Tenascin-C modulates cell cycle progression to enhance tumour cell proliferation through AKT/FOXO1 signalling in pancreatic cancer, J. Cancer, 9, 4449, 10.7150/jca.25926 Xu, 2015, The co-expression of MMP-9 and Tenascin-C is significantly associated with the progression and prognosis of pancreatic cancer, Diagn. Pathol., 10, 211, 10.1186/s13000-015-0445-3 Cai, 2017, Tenascin-C induces migration and invasion through JNK/c-Jun signalling in pancreatic cancer, Oncotarget, 8, 74406, 10.18632/oncotarget.20160 Shi, 2015, Tenascin-C induces resistance to apoptosis in pancreatic cancer cell through activation of ERK/NF-kappaB pathway, Apoptosis, 20, 843, 10.1007/s10495-015-1106-4 Leppanen, 2019, Tenascin C, fibronectin, and tumor-stroma ratio in pancreatic ductal adenocarcinoma, Pancreas, 48, 43, 10.1097/MPA.0000000000001195 Qian, 2019, Exosomal Tenascin-c induces proliferation and invasion of pancreatic cancer cells by WNT signaling, Onco Targets Ther., 12, 3197, 10.2147/OTT.S192218 Yoneura, 2018, Expression of annexin II and stromal tenascin C promotes epithelial to mesenchymal transition and correlates with distant metastasis in pancreatic cancer, Int. J. Mol. Med., 42, 821 Koninger, 2004, Overexpressed decorin in pancreatic cancer: potential tumor growth inhibition and attenuation of chemotherapeutic action, Clin. Cancer Res., 10, 4776, 10.1158/1078-0432.CCR-1190-03 Weber, 2001, Biglycan is overexpressed in pancreatic cancer and induces G1-arrest in pancreatic cancer cell lines, Gastroenterology, 121, 657, 10.1053/gast.2001.27222 Wang, 2017, Asporin promotes pancreatic cancer cell invasion and migration by regulating the epithelial-to-mesenchymal transition (EMT) through both autocrine and paracrine mechanisms, Cancer Lett., 398, 24, 10.1016/j.canlet.2017.04.001 Asano, 2017, Stromal versican regulates tumor growth by promoting angiogenesis, Sci. Rep., 7, 17225, 10.1038/s41598-017-17613-6 Ping Lu, 2002, Lumican expression in alpha cells of islets in pancreas and pancreatic cancer cells, J. Pathol., 196, 324, 10.1002/path.1037 Thakur, 2016, TAp73 loss favors Smad-independent TGF-beta signaling that drives EMT in pancreatic ductal adenocarcinoma, Cell Death Differ., 23, 1358, 10.1038/cdd.2016.18 Chen, 2002, Smad4/DPC4-dependent regulation of biglycan gene expression by transforming growth factor-beta in pancreatic tumor cells, J. Biol. Chem., 277, 36118, 10.1074/jbc.M203709200 Groth, 2005, Adhesion and Rac1-dependent regulation of biglycan gene expression by transforming growth factor-beta. Evidence for oxidative signaling through NADPH oxidase, J. Biol. Chem., 280, 33190, 10.1074/jbc.M504249200 Li, 2014, Extracellular lumican inhibits pancreatic cancer cell growth and is associated with prolonged survival after surgery, Clin. Cancer Res., 20, 6529, 10.1158/1078-0432.CCR-14-0970 Sarcar, 2019, Hypoxia-induced autophagy degrades stromal lumican into tumor microenvironment of pancreatic ductal adenocarcinoma: a mini-review, J. Cancer Treatment Diagn., 3, 22, 10.29245/2578-2967/2019/1.1165 Li, 2019, Hypoxia-induced autophagy of stellate cells inhibits expression and secretion of lumican into microenvironment of pancreatic ductal adenocarcinoma, Cell Death Differ., 26, 382, 10.1038/s41418-018-0207-3 Li, 2017, Prolonged exposure to extracellular lumican restrains pancreatic adenocarcinoma growth, Oncogene, 36, 5432, 10.1038/onc.2017.125 Theocharis, 2000, Pancreatic carcinoma is characterized by elevated content of hyaluronan and chondroitin sulfate with altered disaccharide composition, Biochim. Biophys. Acta, 1502, 201, 10.1016/S0925-4439(00)00051-X Cheng, 2015, Receptor for hyaluronic acid-mediated motility is associated with poor survival in pancreatic ductal adenocarcinoma, J. Cancer, 6, 1093, 10.7150/jca.12990 Cheng, 2016, Hyaluronan stimulates pancreatic cancer cell motility, Oncotarget, 7, 4829, 10.18632/oncotarget.6617 Kohi, 2016, Hyaluromycin, a novel hyaluronidase inhibitor, attenuates pancreatic cancer cell migration and proliferation, J. Oncol., 2016, 9063087, 10.1155/2016/9063087 DuFort, 2016, Interstitial pressure in pancreatic ductal adenocarcinoma is dominated by a gel-fluid phase, Biophys. J., 110, 2106, 10.1016/j.bpj.2016.03.040 Jacobetz, 2013, Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer, Gut, 62, 112, 10.1136/gutjnl-2012-302529 Arcangeli, 2014, Interaction of tumour cells with their microenvironment: ion channels and cell adhesion molecules. A focus on pancreatic cancer, Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci., 369, 20130101, 10.1098/rstb.2013.0101 Desgrosellier, 2010, Integrins in cancer: biological implications and therapeutic opportunities, Nat. Rev. Cancer, 10, 9, 10.1038/nrc2748 Gleason, 2005, Immunohistochemical detection of the β4 integrin subunit in pancreatic adenocarcinoma, J. Histochem. Cytochem., 53, 799, 10.1369/jhc.4B6522.2005 Schnittert, 2019, Integrin α11 in pancreatic stellate cells regulates tumor stroma interaction in pancreatic cancer, FASEB J., 33, 6609, 10.1096/fj.201802336R Sipos, 2004, Immunohistochemical screening for beta6-integrin subunit expression in adenocarcinomas using a novel monoclonal antibody reveals strong up-regulation in pancreatic ductal adenocarcinomas in vivo and in vitro, Histopathology, 45, 226, 10.1111/j.1365-2559.2004.01919.x Del Rio, 2009, Stretching single talin rod molecules activates vinculin binding, Science, 323, 638, 10.1126/science.1162912 Cabodi, 2010, Integrins and signal transduction, Adv. Exp. Med. Biol., 674, 43, 10.1007/978-1-4419-6066-5_5 Hynes, 2002, Integrins: bidirectional, allosteric signaling machines, Cell, 110, 673, 10.1016/S0092-8674(02)00971-6 Takada, 2007, The integrins, Genome Biol., 8, 215, 10.1186/gb-2007-8-5-215 Keizer, 2011, Evaluation of α2-integrin expression as a biomarker for tumor growth inhibition for the investigational integrin inhibitor E7820 in preclinical and clinical studies, AAPS J., 13, 230, 10.1208/s12248-011-9260-2 Semba, 2004, An angiogenesis inhibitor E7820 shows broad-spectrum tumor growth inhibition in a xenograft model: possible value of integrin alpha2 on platelets as a biological marker, Clin. Cancer Res., 10, 1430, 10.1158/1078-0432.CCR-0109-03 Ramirez, 2011, The alpha(2)beta(1) integrin is a metastasis suppressor in mouse models and human cancer, J. Clin. Invest., 121, 226, 10.1172/JCI42328 Lee, 2009, Core3 O-glycan synthase suppresses tumor formation and metastasis of prostate carcinoma PC3 and LNCaP cells through down-regulation of alpha2beta1 integrin complex, J. Biol. Chem., 284, 17157, 10.1074/jbc.M109.010934 Langsenlehner, 2006, Integrin alpha-2 and beta-3 gene polymorphisms and breast cancer risk, Breast Cancer Res. Treat., 97, 67, 10.1007/s10549-005-9089-4 Avraamides, 2008, Integrins in angiogenesis and lymphangiogenesis, Nat. Rev. Cancer, 8, 604, 10.1038/nrc2353 Erdogan, 2017, Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin, J. Cell Biol., 216, 3799, 10.1083/jcb.201704053 Reader, 2019, The integrin αvβ6 drives pancreatic cancer through diverse mechanisms and represents an effective target for therapy, J. Pathol., 249, 332, 10.1002/path.5320 Moore, 2020, Integrin αvβ6-specific therapy for pancreatic cancer developed from foot-and-mouth-disease virus, Theranostics, 10, 2930, 10.7150/thno.38702 Murphy, 2015, Tumor angiogenesis in the absence of fibronectin or its cognate integrin receptors, PLoS One, 10, 10.1371/journal.pone.0120872 Topalovski, 2016, Hypoxia and transforming growth factor β cooperate to induce fibulin-5 expression in pancreatic cancer, J. Biol. Chem., 291, 22244, 10.1074/jbc.M116.730945 Cruz-Monserrate, 2007, Upregulation and redistribution of integrin alpha6beta4 expression occurs at an early stage in pancreatic adenocarcinoma progression, Mod. Pathol., 20, 656, 10.1038/modpathol.3800782 Franco-Barraza, 2017, Matrix-regulated integrin α(v)β(5) maintains α(5)β(1)-dependent desmoplastic traits prognostic of neoplastic recurrence, Elife, 6, 10.7554/eLife.20600 Grzesiak, 2007, The integrin-extracellular matrix axis in pancreatic cancer, Pancreas, 35, 293, 10.1097/mpa.0b013e31811f4526 Broders-Bondon, 2018, Mechanotransduction in tumor progression: The dark side of the force, J. Cell Biol., 217, 1571, 10.1083/jcb.201701039 Liotta, 2001, The microenvironment of the tumour-host interface, Nature, 411, 375, 10.1038/35077241 Butcher, 2009, A tense situation: forcing tumour progression, Nat. Rev. Cancer, 9, 108, 10.1038/nrc2544 Stylianopoulos, 2012, Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors, Proc. Natl. Acad. Sci. U. S. A., 109, 15101, 10.1073/pnas.1213353109 Jain, 2014, The role of mechanical forces in tumor growth and therapy, Annu. Rev. Biomed. Eng., 16, 321, 10.1146/annurev-bioeng-071813-105259 Nia, 2016, Solid stress and elastic energy as measures of tumour mechanopathology, Nat. Biomed. Eng., 1, 10.1038/s41551-016-0004 Cheng, 2009, Micro-environmental mechanical stress controls tumor spheroid size and morphology by suppressing proliferation and inducing apoptosis in cancer cells, PLoS One, 4, 10.1371/journal.pone.0004632 Alessandri, 2013, Cellular capsules as a tool for multicellular spheroid production and for investigating the mechanics of tumor progression in vitro, Proc. Natl. Acad. Sci. U. S. A., 110, 14843, 10.1073/pnas.1309482110 Kalli, 2018, Solid stress facilitates fibroblasts activation to promote pancreatic cancer cell migration, Ann. Biomed. Eng., 46, 657, 10.1007/s10439-018-1997-7 Cheng, 2009, Micro-environmental mechanical stress controls tumor spheroid size and morphology by suppressing proliferation and inducing apoptosis in cancer cells, PLoS One, 4, 10.1371/journal.pone.0004632 Helmlinger, 1997, Solid stress inhibits the growth of multicellular tumor spheroids, Nat. Biotechnol., 15, 778, 10.1038/nbt0897-778 Kalli, 2019, Solid stress-induced migration is mediated by GDF15 through Akt pathway activation in pancreatic cancer cells, Sci. Rep., 9, 978, 10.1038/s41598-018-37425-6 Levental, 2009, Matrix crosslinking forces tumor progression by enhancing integrin signaling, Cell, 139, 891, 10.1016/j.cell.2009.10.027 Cavaco, 2018, The interaction between laminin-332 and alpha3beta1 integrin determines differentiation and maintenance of CAFs, and supports invasion of pancreatic duct adenocarcinoma cells, Cancers (Basel), 11, e4632, 10.3390/cancers11010014 Qi, 2016, P-selectin-mediated LOX expression promotes insulinoma growth in Rip1-Tag2 mice by increasing tissue stiffness, Int. J. Biol. Sci., 12, 1289, 10.7150/ijbs.16405 Zhao, 2018, Simultaneous inhibition of hedgehog signaling and tumor proliferation remodels stroma and enhances pancreatic cancer therapy, Biomaterials, 159, 215, 10.1016/j.biomaterials.2018.01.014 Stylianou, 2019, Collagen content and extracellular matrix cause cytoskeletal remodelling in pancreatic fibroblasts, J. R. Soc. Interface, 16, 20190226, 10.1098/rsif.2019.0226 Park, 2020, Mechanical regulation of glycolysis via cytoskeleton architecture, Nature, 1 Boucher, 1992, Microvascular pressure is the principal driving force for interstitial hypertension in solid tumors: implications for vascular collapse, Cancer Res., 52, 5110 Park, 2009, Perfusion CT: noninvasive surrogate marker for stratification of pancreatic cancer response to concurrent chemo- and radiation therapy, Radiology, 250, 110, 10.1148/radiol.2493080226 Olive, 2009, Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer, Science, 324, 1457, 10.1126/science.1171362 Provenzano, 2012, Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma, Cancer Cell, 21, 418, 10.1016/j.ccr.2012.01.007 Grzesiak, 2007, Determination of the ligand-binding specificities of the alpha2beta1 and alpha1beta1 integrins in a novel 3-dimensional in vitro model of pancreatic cancer, Pancreas, 34, 220, 10.1097/01.mpa.0000250129.64650.f6 Shintani, 2008, Collagen I-mediated up-regulation of N-cadherin requires cooperative signals from integrins and discoidin domain receptor 1, J. Cell Biol., 180, 1277, 10.1083/jcb.200708137 Bassagañas, 2014, Pancreatic cancer cell glycosylation regulates cell adhesion and invasion through the modulation of α2β1 integrin and E-cadherin function, PLoS One, 9, 10.1371/journal.pone.0098595 Shields, 2012, Interplay between β1-integrin and Rho signaling regulates differential scattering and motility of pancreatic cancer cells by snail and Slug proteins, J. Biol. Chem., 287, 6218, 10.1074/jbc.M111.308940 Grzesiak, 2010, Divalent cations modulate alpha2beta1 integrin-mediated malignancy in a novel 3-dimensional in vitro model of pancreatic cancer, Pancreas, 39, 904, 10.1097/MPA.0b013e3181ce60a3 Grzesiak, 2006, The alpha2beta1 integrin mediates the malignant phenotype on type I collagen in pancreatic cancer cell lines, Br. J. Cancer, 94, 1311, 10.1038/sj.bjc.6603088 Cavaco, 2018, The interaction between laminin-332 and α3β1 integrin determines differentiation and maintenance of CAFs, and supports invasion of pancreatic duct adenocarcinoma cells, Cancers (Basel), 11, 14, 10.3390/cancers11010014 Liu, 2020, ZIP4 increases expression of transcription factor zeb1 to promote integrin α3β1 signaling and inhibit expression of the gemcitabine transporter ENT1 in pancreatic cancer cells, Gastroenterology, 158, 679, 10.1053/j.gastro.2019.10.038 Grzesiak, 2007, Integrin-mediated laminin-1 adhesion upregulates CXCR4 and IL-8 expression in pancreatic cancer cells, Surgery, 141, 804, 10.1016/j.surg.2006.12.016 Schmid, 2011, Combined blockade of integrin-α4β1 plus cytokines SDF-1α or IL-1β potently inhibits tumor inflammation and growth, Cancer Res., 71, 6965, 10.1158/0008-5472.CAN-11-0588 Lowrie, 2004, Latent effects of fibronectin, alpha5beta1 integrin, alphaVbeta5 integrin and the cytoskeleton regulate pancreatic carcinoma cell IL-8 secretion, Br. J. Cancer, 91, 1327, 10.1038/sj.bjc.6602132 Primo, 2010, Increased expression of alpha6 integrin in endothelial cells unveils a proangiogenic role for basement membrane, Cancer Res., 70, 5759, 10.1158/0008-5472.CAN-10-0507 Chen, 2009, Role of fibrillar Tenascin-C in metastatic pancreatic cancer, Int. J. Oncol., 34, 1029 Sawai, 2006, Interleukin-1alpha enhances the aggressive behavior of pancreatic cancer cells by regulating the alpha6beta1-integrin and urokinase plasminogen activator receptor expression, BMC Cell Biol., 7, 8, 10.1186/1471-2121-7-8 Zeltz, 2019, α11β1 integrin is induced in a subset of cancer-associated fibroblasts in desmoplastic tumor stroma and mediates in vitro cell migration, Cancers (Basel), 11, 10.3390/cancers11060765 Seguin, 2014, An integrin β₃-KRAS-RalB complex drives tumour stemness and resistance to EGFR inhibition, Nat. Cell Biol., 16, 457, 10.1038/ncb2953 Desgrosellier, 2009, An integrin alpha(v)beta(3)-c-Src oncogenic unit promotes anchorage-independence and tumor progression, Nat. Med., 15, 1163, 10.1038/nm.2009 Hosotani, 2002, Expression of integrin alphaVbeta3 in pancreatic carcinoma: relation to MMP-2 activation and lymph node metastasis, Pancreas, 25, e30, 10.1097/00006676-200208000-00021 Masugi, 2015, Upregulation of integrin β4 promotes epithelial-mesenchymal transition and is a novel prognostic marker in pancreatic ductal adenocarcinoma, Lab. Investig., 95, 308, 10.1038/labinvest.2014.166 Giancotti, 2007, Targeting integrin β4 for cancer and anti-angiogenic therapy, Trends Pharmacol. Sci., 28, 506, 10.1016/j.tips.2007.08.004 Costanza, 2019, Transforming growth factor beta-induced, an extracellular matrix interacting protein, enhances glycolysis and promotes pancreatic cancer cell migration, Int. J. Cancer, 145, 1570, 10.1002/ijc.32247 Mijatovic, 2007, Neurotensin is a versatile modulator of in vitro human pancreatic ductal adenocarcinoma cell (PDAC) migration, Cell. Oncol., 29, 315 Singh, 2009, A gene expression signature associated with "K-Ras addiction" reveals regulators of EMT and tumor cell survival, Cancer Cell, 15, 489, 10.1016/j.ccr.2009.03.022 Lee, 2012, RNA interference characterization of proteins discovered by proteomic analysis of pancreatic cancer reveals function in cell growth and survival, Pancreas, 41, 84, 10.1097/MPA.0b013e3182236385 Li, 2016, Integrin β6 acts as an unfavorable prognostic indicator and promotes cellular malignant behaviors via ERK-ETS1 pathway in pancreatic ductal adenocarcinoma (PDAC), Tumour Biol., 37, 5117, 10.1007/s13277-015-4353-7 Jin, 2019, β8 integrin mediates pancreatic cancer cell radiochemoresistance, Mol. Cancer Res., 17, 2126, 10.1158/1541-7786.MCR-18-1352 Parkin, 2001, Estimating the world cancer burden: Globocan 2000, Int. J. Cancer, 94, 153, 10.1002/ijc.1440 Ferlay, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008, Int. J. Cancer, 127, 2893, 10.1002/ijc.25516 Ferlay, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012, Int. J. Cancer, 136, E359, 10.1002/ijc.29210