Skip to main content
Top
Published in: Cancer and Metastasis Reviews 1/2024

15-07-2023 | Metastasis | Review

Molecular profile of metastasis, cell plasticity and EMT in pancreatic cancer: a pre-clinical connection to aggressiveness and drug resistance

Authors: Zhenli Guo, Milad Ashrafizadeh, Wei Zhang, Rongjun Zou, Gautam Sethi, Xianbin Zhang

Published in: Cancer and Metastasis Reviews | Issue 1/2024

Login to get access

Abstract

The metastasis is a multistep process in which a small proportion of cancer cells are detached from the colony to enter into blood cells for obtaining a new place for metastasis and proliferation. The metastasis and cell plasticity are considered major causes of cancer-related deaths since they improve the malignancy of cancer cells and provide poor prognosis for patients. Furthermore, enhancement in the aggressiveness of cancer cells has been related to the development of drug resistance. Metastasis of pancreatic cancer (PC) cells has been considered one of the major causes of death in patients and their undesirable prognosis. PC is among the most malignant tumors of the gastrointestinal tract and in addition to lifestyle, smoking, and other factors, genomic changes play a key role in its progression. The stimulation of EMT in PC cells occurs as a result of changes in molecular interaction, and in addition to increasing metastasis, EMT participates in the development of chemoresistance. The epithelial, mesenchymal, and acinar cell plasticity can occur and determines the progression of PC. The major molecular pathways including STAT3, PTEN, PI3K/Akt, and Wnt participate in regulating the metastasis of PC cells. The communication in tumor microenvironment can provide by exosomes in determining PC metastasis. The components of tumor microenvironment including macrophages, neutrophils, and cancer-associated fibroblasts can modulate PC progression and the response of cancer cells to chemotherapy.

Graphical Abstract

Literature
1.
go back to reference Zeng, S., et al. (2019). Chemoresistance in Pancreatic Cancer. International Journal of Molecular Sciences, 20(18), 4504.PubMedPubMedCentral Zeng, S., et al. (2019). Chemoresistance in Pancreatic Cancer. International Journal of Molecular Sciences, 20(18), 4504.PubMedPubMedCentral
2.
go back to reference Rahib, L., et al. (2014). Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Research, 74(11), 2913–2921.PubMed Rahib, L., et al. (2014). Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Research, 74(11), 2913–2921.PubMed
3.
go back to reference Kamisawa, T., et al. (2016). Pancreatic cancer. Lancet, 388(10039), 73–85.PubMed Kamisawa, T., et al. (2016). Pancreatic cancer. Lancet, 388(10039), 73–85.PubMed
4.
go back to reference Rawla, P., Sunkara, T., & Gaduputi, V. (2019). Epidemiology of pancreatic cancer: Global trends, etiology and risk factors. World journal of oncology, 10(1), 10–27.PubMedPubMedCentral Rawla, P., Sunkara, T., & Gaduputi, V. (2019). Epidemiology of pancreatic cancer: Global trends, etiology and risk factors. World journal of oncology, 10(1), 10–27.PubMedPubMedCentral
5.
go back to reference Siegel, R. L., Miller, K. D., & Jemal, A. (2018). Cancer statistics, 2018. CA: a Cancer Journal for Clinicians, 68(1), 7–30.PubMed Siegel, R. L., Miller, K. D., & Jemal, A. (2018). Cancer statistics, 2018. CA: a Cancer Journal for Clinicians, 68(1), 7–30.PubMed
6.
go back to reference Kleeff, J., et al. (2016). Pancreatic cancer. Nature reviews Disease primers, 2, 16022.PubMed Kleeff, J., et al. (2016). Pancreatic cancer. Nature reviews Disease primers, 2, 16022.PubMed
7.
go back to reference Kaur, S., et al. (2017). A combination of MUC5AC and CA19-9 improves the diagnosis of pancreatic cancer: A multicenter study. The American Journal of Gastroenterology, 112(1), 172–183.PubMed Kaur, S., et al. (2017). A combination of MUC5AC and CA19-9 improves the diagnosis of pancreatic cancer: A multicenter study. The American Journal of Gastroenterology, 112(1), 172–183.PubMed
8.
go back to reference Kaur, S., et al. (2012). Early diagnosis of pancreatic cancer: challenges and new developments. Biomarkers in Medicine, 6(5), 597–612.PubMed Kaur, S., et al. (2012). Early diagnosis of pancreatic cancer: challenges and new developments. Biomarkers in Medicine, 6(5), 597–612.PubMed
9.
go back to reference Papaefthymiou, A., Doukatas, A., & Galanopoulos, M. (2022). Pancreatic cancer and oligonucleotide therapy: Exploring novel therapeutic options and targeting chemoresistance. Clinics and Research in Hepatology and Gastroenterology, 46(5), 101911.PubMed Papaefthymiou, A., Doukatas, A., & Galanopoulos, M. (2022). Pancreatic cancer and oligonucleotide therapy: Exploring novel therapeutic options and targeting chemoresistance. Clinics and Research in Hepatology and Gastroenterology, 46(5), 101911.PubMed
10.
go back to reference Kamimura, K., Yokoo, T., & Terai, S. (2018). Gene therapy for pancreatic diseases: current status. International journal of molecular sciences, 19(11), 3415.PubMedPubMedCentral Kamimura, K., Yokoo, T., & Terai, S. (2018). Gene therapy for pancreatic diseases: current status. International journal of molecular sciences, 19(11), 3415.PubMedPubMedCentral
11.
go back to reference Hu, C., et al. (2018). Association between inherited germline mutations in cancer predisposition genes and risk of pancreatic cancer. Jama, 319(23), 2401–2409.PubMedPubMedCentral Hu, C., et al. (2018). Association between inherited germline mutations in cancer predisposition genes and risk of pancreatic cancer. Jama, 319(23), 2401–2409.PubMedPubMedCentral
12.
go back to reference Di Magliano, M. P., & Logsdon, C. D. J. G. (2013). Roles for KRAS in pancreatic tumor development and progression. Gastroenterology, 144(6), 1220–1229.PubMed Di Magliano, M. P., & Logsdon, C. D. J. G. (2013). Roles for KRAS in pancreatic tumor development and progression. Gastroenterology, 144(6), 1220–1229.PubMed
13.
go back to reference Gleeson, F. C., et al. (2016). Targeted next generation sequencing of endoscopic ultrasound acquired cytology from ampullary and pancreatic adenocarcinoma has the potential to aid patient stratification for optimal therapy selection. Oncotarget, 7(34), 54526.PubMedPubMedCentral Gleeson, F. C., et al. (2016). Targeted next generation sequencing of endoscopic ultrasound acquired cytology from ampullary and pancreatic adenocarcinoma has the potential to aid patient stratification for optimal therapy selection. Oncotarget, 7(34), 54526.PubMedPubMedCentral
14.
go back to reference Wang, S., et al. (2020). Tumor microenvironment in chemoresistance, metastasis and immunotherapy of pancreatic cancer. American Journal of Cancer Research, 10(7), 1937–1953.PubMedPubMedCentral Wang, S., et al. (2020). Tumor microenvironment in chemoresistance, metastasis and immunotherapy of pancreatic cancer. American Journal of Cancer Research, 10(7), 1937–1953.PubMedPubMedCentral
15.
go back to reference Jones, S., et al. (2008). Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science, 321(5897), 1801–1806.PubMedPubMedCentral Jones, S., et al. (2008). Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science, 321(5897), 1801–1806.PubMedPubMedCentral
16.
go back to reference Zhang, D., et al. (2022). SEMA3C supports pancreatic cancer progression by regulating the autophagy process and tumor immune microenvironment. Frontiers in Oncology, 12, 890154.PubMedPubMedCentral Zhang, D., et al. (2022). SEMA3C supports pancreatic cancer progression by regulating the autophagy process and tumor immune microenvironment. Frontiers in Oncology, 12, 890154.PubMedPubMedCentral
17.
go back to reference Li, M., et al. (2022). IPO7 promotes pancreatic cancer progression via regulating ERBB pathway. Clinics (São Paulo, Brazil), 77, 100044.PubMed Li, M., et al. (2022). IPO7 promotes pancreatic cancer progression via regulating ERBB pathway. Clinics (São Paulo, Brazil), 77, 100044.PubMed
18.
go back to reference Cao, J. Z., et al. (2022). UBE2C promotes the progression of pancreatic cancer and glycolytic activity via EGFR stabilization-mediated PI3K-Akt pathway activation. Journal of gastrointestinal oncology, 13(3), 1444–1453.PubMedPubMedCentral Cao, J. Z., et al. (2022). UBE2C promotes the progression of pancreatic cancer and glycolytic activity via EGFR stabilization-mediated PI3K-Akt pathway activation. Journal of gastrointestinal oncology, 13(3), 1444–1453.PubMedPubMedCentral
19.
go back to reference Wen, Z., et al. (2022). COL10A1-DDR2 axis promotes the progression of pancreatic cancer by regulating MEK/ERK signal transduction. Frontiers in Oncology, 12, 1049345.PubMedPubMedCentral Wen, Z., et al. (2022). COL10A1-DDR2 axis promotes the progression of pancreatic cancer by regulating MEK/ERK signal transduction. Frontiers in Oncology, 12, 1049345.PubMedPubMedCentral
20.
go back to reference Li, J., et al. (2022). HOXA10 promote pancreatic cancer progression via directly activating canonical NF-κB signaling pathway. Carcinogenesis, 43(8), 787–796.PubMed Li, J., et al. (2022). HOXA10 promote pancreatic cancer progression via directly activating canonical NF-κB signaling pathway. Carcinogenesis, 43(8), 787–796.PubMed
21.
go back to reference Song, Y., Gao, Z., & Zheng, C. (2022). Silencing LINC01234 represses pancreatic cancer progression by inhibiting the malignant phenotypes of pancreatic cancer cells. Immunobiology, 227(6), 152295.PubMed Song, Y., Gao, Z., & Zheng, C. (2022). Silencing LINC01234 represses pancreatic cancer progression by inhibiting the malignant phenotypes of pancreatic cancer cells. Immunobiology, 227(6), 152295.PubMed
22.
go back to reference Shi, J., et al. (2022). JNK inhibitor IX restrains pancreatic cancer through p53 and p21. Frontiers in Oncology, 12, 1006131.PubMedPubMedCentral Shi, J., et al. (2022). JNK inhibitor IX restrains pancreatic cancer through p53 and p21. Frontiers in Oncology, 12, 1006131.PubMedPubMedCentral
23.
go back to reference Nimmakayala, R. K., et al. (2022). PAF1 cooperates with YAP1 in metaplastic ducts to promote pancreatic cancer. Cell Death & Disease, 13(10), 839. Nimmakayala, R. K., et al. (2022). PAF1 cooperates with YAP1 in metaplastic ducts to promote pancreatic cancer. Cell Death & Disease, 13(10), 839.
24.
go back to reference Park, S. J., et al. (2023). Enhanced glutaminolysis drives hypoxia-induced chemoresistance in pancreatic cancer. Cancer Research, 83(5), 735–752.PubMed Park, S. J., et al. (2023). Enhanced glutaminolysis drives hypoxia-induced chemoresistance in pancreatic cancer. Cancer Research, 83(5), 735–752.PubMed
25.
go back to reference Liu, H., et al. (2023). Pancreatic stellate cells exploit Wnt/β-catenin/TCF7-mediated glutamine metabolism to promote pancreatic cancer cells growth. Cancer Letters, 555, 216040.PubMed Liu, H., et al. (2023). Pancreatic stellate cells exploit Wnt/β-catenin/TCF7-mediated glutamine metabolism to promote pancreatic cancer cells growth. Cancer Letters, 555, 216040.PubMed
26.
go back to reference Huang, C. K., et al. (2023). ENO1 promotes immunosuppression and tumor growth in pancreatic cancer. Clinical & Translational Oncology, 25(7), 2250–2264. Huang, C. K., et al. (2023). ENO1 promotes immunosuppression and tumor growth in pancreatic cancer. Clinical & Translational Oncology, 25(7), 2250–2264.
27.
go back to reference Wu, J. (2022). Pancreatic cancer-derived exosomes promote the proliferation, invasion, and metastasis of pancreatic cancer by the miR-3960/TFAP2A Axis. Journal of Oncology, 2022, 3590326.PubMedPubMedCentral Wu, J. (2022). Pancreatic cancer-derived exosomes promote the proliferation, invasion, and metastasis of pancreatic cancer by the miR-3960/TFAP2A Axis. Journal of Oncology, 2022, 3590326.PubMedPubMedCentral
28.
go back to reference Zhang, W., et al. (2022). Thiostrepton induces ferroptosis in pancreatic cancer cells through STAT3/GPX4 signalling. Cell Death & Disease, 13(7), 630. Zhang, W., et al. (2022). Thiostrepton induces ferroptosis in pancreatic cancer cells through STAT3/GPX4 signalling. Cell Death & Disease, 13(7), 630.
29.
go back to reference Jacoberger-Foissac, C., et al. (2023). CD73 Inhibits cGAS-STING and cooperates with CD39 to promote pancreatic cancer. Cancer Immunology Research, 11(1), 56–71.PubMedPubMedCentral Jacoberger-Foissac, C., et al. (2023). CD73 Inhibits cGAS-STING and cooperates with CD39 to promote pancreatic cancer. Cancer Immunology Research, 11(1), 56–71.PubMedPubMedCentral
30.
go back to reference Simmler, P., et al. (2022). SF3B1 facilitates HIF1-signaling and promotes malignancy in pancreatic cancer. Cell Reports, 40(8), 111266.PubMed Simmler, P., et al. (2022). SF3B1 facilitates HIF1-signaling and promotes malignancy in pancreatic cancer. Cell Reports, 40(8), 111266.PubMed
31.
go back to reference Lee, M. S., et al. (2023). Ornithine aminotransferase supports polyamine synthesis in pancreatic cancer. Nature, 616(7956), 339–347.PubMedPubMedCentral Lee, M. S., et al. (2023). Ornithine aminotransferase supports polyamine synthesis in pancreatic cancer. Nature, 616(7956), 339–347.PubMedPubMedCentral
32.
go back to reference Lefler, J. E., et al. (2022). STAT3 in tumor fibroblasts promotes an immunosuppressive microenvironment in pancreatic cancer. Life Science Alliance, 5(11), 1. Lefler, J. E., et al. (2022). STAT3 in tumor fibroblasts promotes an immunosuppressive microenvironment in pancreatic cancer. Life Science Alliance, 5(11), 1.
33.
go back to reference Su, H., et al. (2022). Collagenolysis-dependent DDR1 signalling dictates pancreatic cancer outcome. Nature, 610(7931), 366–372.PubMedPubMedCentral Su, H., et al. (2022). Collagenolysis-dependent DDR1 signalling dictates pancreatic cancer outcome. Nature, 610(7931), 366–372.PubMedPubMedCentral
34.
go back to reference Rademaker, G., et al. (2022). Myoferlin targeting triggers mitophagy and primes ferroptosis in pancreatic cancer cells. Redox Biology, 53, 102324.PubMedPubMedCentral Rademaker, G., et al. (2022). Myoferlin targeting triggers mitophagy and primes ferroptosis in pancreatic cancer cells. Redox Biology, 53, 102324.PubMedPubMedCentral
35.
go back to reference Lan, L., et al. (2022). GREM1 is required to maintain cellular heterogeneity in pancreatic cancer. Nature, 607(7917), 163–168.PubMed Lan, L., et al. (2022). GREM1 is required to maintain cellular heterogeneity in pancreatic cancer. Nature, 607(7917), 163–168.PubMed
36.
go back to reference Wang, S., et al. (2022). RNA-binding proteins and cancer metastasis. In Seminars in Cancer Biology. Elsevier. Wang, S., et al. (2022). RNA-binding proteins and cancer metastasis. In Seminars in Cancer Biology. Elsevier.
37.
go back to reference Fidler, I. J. (2003). The pathogenesis of cancer metastasis: The'seed and soil'hypothesis revisited. Nature reviews cancer, 3(6), 453–458.PubMed Fidler, I. J. (2003). The pathogenesis of cancer metastasis: The'seed and soil'hypothesis revisited. Nature reviews cancer, 3(6), 453–458.PubMed
38.
go back to reference Chambers, A. F., Groom, A. C., & MacDonald, I. C. J. N. R. C. (2002). Dissemination and growth of cancer cells in metastatic sites. Nature Reviews Cancer, 2(8), 563–572.PubMed Chambers, A. F., Groom, A. C., & MacDonald, I. C. J. N. R. C. (2002). Dissemination and growth of cancer cells in metastatic sites. Nature Reviews Cancer, 2(8), 563–572.PubMed
39.
go back to reference Polyak, K., & Weinberg, R. A. J. N. R. C. (2009). Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nature Reviews Cancer, 9(4), 265–273.PubMed Polyak, K., & Weinberg, R. A. J. N. R. C. (2009). Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nature Reviews Cancer, 9(4), 265–273.PubMed
40.
41.
go back to reference Yang, Y., & Cao, Y. (2022). The impact of VEGF on cancer metastasis and systemic disease. In Seminars in Cancer Biology. Elsevier. Yang, Y., & Cao, Y. (2022). The impact of VEGF on cancer metastasis and systemic disease. In Seminars in Cancer Biology. Elsevier.
42.
go back to reference Friedl, P., & Alexander, S. J. C. (2011). Cancer invasion and the microenvironment: Plasticity and reciprocity. Cell, 147(5), 992–1009.PubMed Friedl, P., & Alexander, S. J. C. (2011). Cancer invasion and the microenvironment: Plasticity and reciprocity. Cell, 147(5), 992–1009.PubMed
43.
go back to reference Sun, X., et al. (2022). Inflammatory cell-derived CXCL3 promotes pancreatic cancer metastasis through a novel myofibroblast-hijacked cancer escape mechanism. Gut, 71(1), 129–147.PubMed Sun, X., et al. (2022). Inflammatory cell-derived CXCL3 promotes pancreatic cancer metastasis through a novel myofibroblast-hijacked cancer escape mechanism. Gut, 71(1), 129–147.PubMed
44.
go back to reference Yang, Y., et al. (2016). The PDGF-BB-SOX7 axis-modulated IL-33 in pericytes and stromal cells promotes metastasis through tumour-associated macrophages. Nature communications, 7(1), 11385.PubMedPubMedCentral Yang, Y., et al. (2016). The PDGF-BB-SOX7 axis-modulated IL-33 in pericytes and stromal cells promotes metastasis through tumour-associated macrophages. Nature communications, 7(1), 11385.PubMedPubMedCentral
45.
go back to reference Li, Y., et al. (2022). METTL3 acetylation impedes cancer metastasis via fine-tuning its nuclear and cytosolic functions. Nature Communications, 13(1), 6350.PubMedPubMedCentral Li, Y., et al. (2022). METTL3 acetylation impedes cancer metastasis via fine-tuning its nuclear and cytosolic functions. Nature Communications, 13(1), 6350.PubMedPubMedCentral
46.
go back to reference Delaunay, S., et al. (2022). Mitochondrial RNA modifications shape metabolic plasticity in metastasis. Nature, 607(7919), 593–603.PubMedPubMedCentral Delaunay, S., et al. (2022). Mitochondrial RNA modifications shape metabolic plasticity in metastasis. Nature, 607(7919), 593–603.PubMedPubMedCentral
47.
go back to reference Du, Q., et al. (2022). FASN promotes lymph node metastasis in cervical cancer via cholesterol reprogramming and lymphangiogenesis. Cell Death & Disease, 13(5), 488. Du, Q., et al. (2022). FASN promotes lymph node metastasis in cervical cancer via cholesterol reprogramming and lymphangiogenesis. Cell Death & Disease, 13(5), 488.
48.
go back to reference Liu, W., et al. (2022). MYBL2 promotes proliferation and metastasis of bladder cancer through transactivation of CDCA3. Oncogene, 41(41), 4606–4617.PubMed Liu, W., et al. (2022). MYBL2 promotes proliferation and metastasis of bladder cancer through transactivation of CDCA3. Oncogene, 41(41), 4606–4617.PubMed
49.
go back to reference Meng, Q., et al. (2022). Arginine methylation of MTHFD1 by PRMT5 enhances anoikis resistance and cancer metastasis. Oncogene, 41(32), 3912–3924.PubMed Meng, Q., et al. (2022). Arginine methylation of MTHFD1 by PRMT5 enhances anoikis resistance and cancer metastasis. Oncogene, 41(32), 3912–3924.PubMed
50.
go back to reference Li, F., et al. (2022). Targeting HECTD3-IKKα axis inhibits inflammation-related metastasis. Signal Transduction and Targeted Therapy, 7(1), 264.PubMedPubMedCentral Li, F., et al. (2022). Targeting HECTD3-IKKα axis inhibits inflammation-related metastasis. Signal Transduction and Targeted Therapy, 7(1), 264.PubMedPubMedCentral
51.
go back to reference Verma, A., et al. (2022). EZH2-H3K27me3 mediated KRT14 upregulation promotes TNBC peritoneal metastasis. Nature Communications, 13(1), 7344.PubMedPubMedCentral Verma, A., et al. (2022). EZH2-H3K27me3 mediated KRT14 upregulation promotes TNBC peritoneal metastasis. Nature Communications, 13(1), 7344.PubMedPubMedCentral
52.
go back to reference Mirzaei, S., et al. (2023). Exosome-mediated miR-200a delivery into TGF-β-treated AGS cells abolished epithelial-mesenchymal transition with normalization of ZEB1, vimentin and Snail1 expression. Environmental Research, 231(Part 1), 116115. Mirzaei, S., et al. (2023). Exosome-mediated miR-200a delivery into TGF-β-treated AGS cells abolished epithelial-mesenchymal transition with normalization of ZEB1, vimentin and Snail1 expression. Environmental Research231(Part 1), 116115.
53.
go back to reference Ashrafizadeh, M., et al. (2020). Association of the epithelial–mesenchymal transition (EMT) with cisplatin resistance. International journal of molecular sciences, 21(11), 4002.PubMedPubMedCentral Ashrafizadeh, M., et al. (2020). Association of the epithelial–mesenchymal transition (EMT) with cisplatin resistance. International journal of molecular sciences, 21(11), 4002.PubMedPubMedCentral
54.
go back to reference Ashrafizadeh, M., et al. (2021). New insight towards development of paclitaxel and docetaxel resistance in cancer cells: EMT as a novel molecular mechanism and therapeutic possibilities. Biomedicine & Pharmacotherapy, 141, 111824. Ashrafizadeh, M., et al. (2021). New insight towards development of paclitaxel and docetaxel resistance in cancer cells: EMT as a novel molecular mechanism and therapeutic possibilities. Biomedicine & Pharmacotherapy, 141, 111824.
55.
go back to reference Recouvreux, M. V., et al. (2020). Glutamine depletion regulates Slug to promote EMT and metastasis in pancreatic cancer. The Journal of Experimental Medicine, 217(9), 1. Recouvreux, M. V., et al. (2020). Glutamine depletion regulates Slug to promote EMT and metastasis in pancreatic cancer. The Journal of Experimental Medicine, 217(9), 1.
56.
go back to reference Sheng, W., et al. (2020). Musashi2 promotes EGF-induced EMT in pancreatic cancer via ZEB1-ERK/MAPK signaling. Journal of Experimental & Clinical Cancer Research, 39(1), 16. Sheng, W., et al. (2020). Musashi2 promotes EGF-induced EMT in pancreatic cancer via ZEB1-ERK/MAPK signaling. Journal of Experimental & Clinical Cancer Research, 39(1), 16.
57.
go back to reference Sheng, W., et al. (2017). Calreticulin promotes EGF-induced EMT in pancreatic cancer cells via Integrin/EGFR-ERK/MAPK signaling pathway. Cell Death & Disease, 8(10), e3147. Sheng, W., et al. (2017). Calreticulin promotes EGF-induced EMT in pancreatic cancer cells via Integrin/EGFR-ERK/MAPK signaling pathway. Cell Death & Disease, 8(10), e3147.
58.
go back to reference Chen, Y., et al. (2020). FAM172A inhibits EMT in pancreatic cancer via ERK-MAPK signaling. Biology open, 9(2), bio048462.PubMedPubMedCentral Chen, Y., et al. (2020). FAM172A inhibits EMT in pancreatic cancer via ERK-MAPK signaling. Biology open, 9(2), bio048462.PubMedPubMedCentral
59.
go back to reference Fu, X., et al. (2021). Downregulation of NEAT1 sensitizes gemcitabine-resistant pancreatic cancer cells to gemcitabine through modulation of the miR-506-3p/ZEB2/EMT axis. American Journal of Cancer Research, 11(8), 3841–3856.PubMedPubMedCentral Fu, X., et al. (2021). Downregulation of NEAT1 sensitizes gemcitabine-resistant pancreatic cancer cells to gemcitabine through modulation of the miR-506-3p/ZEB2/EMT axis. American Journal of Cancer Research, 11(8), 3841–3856.PubMedPubMedCentral
60.
go back to reference Simeonov, K. P., et al. (2021). Single-cell lineage tracing of metastatic cancer reveals selection of hybrid EMT states. Cancer Cell, 39(8), 1150–1162.e9.PubMedPubMedCentral Simeonov, K. P., et al. (2021). Single-cell lineage tracing of metastatic cancer reveals selection of hybrid EMT states. Cancer Cell, 39(8), 1150–1162.e9.PubMedPubMedCentral
61.
go back to reference Zhang, X., et al. (2022). MACC1 promotes pancreatic cancer metastasis by interacting with the EMT regulator SNAI1. Cell Death & Disease, 13(11), 923. Zhang, X., et al. (2022). MACC1 promotes pancreatic cancer metastasis by interacting with the EMT regulator SNAI1. Cell Death & Disease, 13(11), 923.
62.
go back to reference Li, N., et al. (2018). Emodin inhibits pancreatic cancer EMT and invasion by up-regulating microRNA-1271. Molecular Medicine Reports, 18(3), 3366–3374.PubMedPubMedCentral Li, N., et al. (2018). Emodin inhibits pancreatic cancer EMT and invasion by up-regulating microRNA-1271. Molecular Medicine Reports, 18(3), 3366–3374.PubMedPubMedCentral
63.
go back to reference Sun, L., et al. (2019). Betulinic acid inhibits stemness and EMT of pancreatic cancer cells via activation of AMPK signaling. International Journal of Oncology, 54(1), 98–110.PubMed Sun, L., et al. (2019). Betulinic acid inhibits stemness and EMT of pancreatic cancer cells via activation of AMPK signaling. International Journal of Oncology, 54(1), 98–110.PubMed
64.
go back to reference Liu, F., et al. (2019). SMARCAD1 promotes pancreatic cancer cell growth and metastasis through Wnt/β-catenin-mediated EMT. International Journal of Biological Sciences, 15(3), 636–646.PubMedPubMedCentral Liu, F., et al. (2019). SMARCAD1 promotes pancreatic cancer cell growth and metastasis through Wnt/β-catenin-mediated EMT. International Journal of Biological Sciences, 15(3), 636–646.PubMedPubMedCentral
65.
go back to reference Peng, X., et al. (2022). Calpain2 upregulation regulates EMT-mediated pancreatic cancer metastasis via the Wnt/β-catenin Signaling Pathway. Frontiers of medicine (Lausanne), 9, 783592. Peng, X., et al. (2022). Calpain2 upregulation regulates EMT-mediated pancreatic cancer metastasis via the Wnt/β-catenin Signaling Pathway. Frontiers of medicine (Lausanne), 9, 783592.
66.
go back to reference Tan, J., et al. (2014). Par-4 downregulation confers cisplatin resistance in pancreatic cancer cells via PI3K/Akt pathway-dependent EMT. Toxicology Letters, 224(1), 7–15.PubMed Tan, J., et al. (2014). Par-4 downregulation confers cisplatin resistance in pancreatic cancer cells via PI3K/Akt pathway-dependent EMT. Toxicology Letters, 224(1), 7–15.PubMed
67.
go back to reference Yang, R. M., et al. (2017). miR-3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. Cell Death & Disease, 8(10), e3129. Yang, R. M., et al. (2017). miR-3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. Cell Death & Disease, 8(10), e3129.
68.
go back to reference Li, Y., et al. (2019). Oncolytic Ad co-expressing decorin and Wnt decoy receptor overcomes chemoresistance of desmoplastic tumor through degradation of ECM and inhibition of EMT. Cancer Letters, 459, 15–29.PubMed Li, Y., et al. (2019). Oncolytic Ad co-expressing decorin and Wnt decoy receptor overcomes chemoresistance of desmoplastic tumor through degradation of ECM and inhibition of EMT. Cancer Letters, 459, 15–29.PubMed
69.
go back to reference Okada, Y., et al. (2021). LAMC2 promotes cancer progression and gemcitabine resistance through modulation of EMT and ATP-binding cassette transporters in pancreatic ductal adenocarcinoma. Carcinogenesis, 42(4), 546–556.PubMedPubMedCentral Okada, Y., et al. (2021). LAMC2 promotes cancer progression and gemcitabine resistance through modulation of EMT and ATP-binding cassette transporters in pancreatic ductal adenocarcinoma. Carcinogenesis, 42(4), 546–556.PubMedPubMedCentral
70.
go back to reference An, N., & Zheng, B. (2020). MiR-203a-3p inhibits pancreatic cancer cell proliferation, EMT, and apoptosis by regulating SLUG. Technology in Cancer Research & Treatment, 19, 1533033819898729. An, N., & Zheng, B. (2020). MiR-203a-3p inhibits pancreatic cancer cell proliferation, EMT, and apoptosis by regulating SLUG. Technology in Cancer Research & Treatment, 19, 1533033819898729.
71.
72.
go back to reference Yu, S., et al. (2021). Circ_0092367 Inhibits EMT and gemcitabine resistance in pancreatic cancer via regulating the miR-1206/ESRP1 axis. Genes (Basel), 12(11), 1701.PubMed Yu, S., et al. (2021). Circ_0092367 Inhibits EMT and gemcitabine resistance in pancreatic cancer via regulating the miR-1206/ESRP1 axis. Genes (Basel), 12(11), 1701.PubMed
73.
go back to reference Zhao, Y., et al. (2021). Targeted intervention of eIF4A1 inhibits EMT and metastasis of pancreatic cancer cells via c-MYC/miR-9 signaling. Cancer Cell International, 21(1), 670.PubMedPubMedCentral Zhao, Y., et al. (2021). Targeted intervention of eIF4A1 inhibits EMT and metastasis of pancreatic cancer cells via c-MYC/miR-9 signaling. Cancer Cell International, 21(1), 670.PubMedPubMedCentral
74.
go back to reference Hu, W., et al. (2019). IQGAP1 promotes pancreatic cancer progression and epithelial-mesenchymal transition (EMT) through Wnt/β-catenin signaling. Scientific Reports, 9(1), 7539.PubMedPubMedCentral Hu, W., et al. (2019). IQGAP1 promotes pancreatic cancer progression and epithelial-mesenchymal transition (EMT) through Wnt/β-catenin signaling. Scientific Reports, 9(1), 7539.PubMedPubMedCentral
75.
go back to reference Wang, W., et al. (2018). E-cadherin is downregulated by microenvironmental changes in pancreatic cancer and induces EMT. Oncology Reports, 40(3), 1641–1649.PubMed Wang, W., et al. (2018). E-cadherin is downregulated by microenvironmental changes in pancreatic cancer and induces EMT. Oncology Reports, 40(3), 1641–1649.PubMed
76.
go back to reference Garg, R., et al. (2022). Targeting FTO suppresses pancreatic carcinogenesis via regulating stem cell maintenance and EMT pathway. Cancers (Basel), 14(23), 5919.PubMed Garg, R., et al. (2022). Targeting FTO suppresses pancreatic carcinogenesis via regulating stem cell maintenance and EMT pathway. Cancers (Basel), 14(23), 5919.PubMed
77.
go back to reference Santoro, R., et al. (2018). MEKK3 Sustains EMT and Stemness in Pancreatic Cancer by Regulating YAP and TAZ Transcriptional Activity. Anticancer Research, 38(4), 1937–1946.PubMed Santoro, R., et al. (2018). MEKK3 Sustains EMT and Stemness in Pancreatic Cancer by Regulating YAP and TAZ Transcriptional Activity. Anticancer Research, 38(4), 1937–1946.PubMed
78.
go back to reference Li, W., et al. (2020). Curcumin inhibits pancreatic cancer cell invasion and EMT by interfering with tumor-stromal crosstalk under hypoxic conditions via the IL-6/ERK/NF-κB axis. Oncology Reports, 44(1), 382–392.PubMed Li, W., et al. (2020). Curcumin inhibits pancreatic cancer cell invasion and EMT by interfering with tumor-stromal crosstalk under hypoxic conditions via the IL-6/ERK/NF-κB axis. Oncology Reports, 44(1), 382–392.PubMed
79.
go back to reference Sheng, W., et al. (2020). Calreticulin promotes EMT in pancreatic cancer via mediating Ca(2+) dependent acute and chronic endoplasmic reticulum stress. Journal of Experimental & Clinical Cancer Research, 39(1), 209. Sheng, W., et al. (2020). Calreticulin promotes EMT in pancreatic cancer via mediating Ca(2+) dependent acute and chronic endoplasmic reticulum stress. Journal of Experimental & Clinical Cancer Research, 39(1), 209.
80.
go back to reference Yan, T., et al. (2017). Astaxanthin inhibits gemcitabine-resistant human pancreatic cancer progression through EMT inhibition and gemcitabine resensitization. Oncology Letters, 14(5), 5400–5408.PubMedPubMedCentral Yan, T., et al. (2017). Astaxanthin inhibits gemcitabine-resistant human pancreatic cancer progression through EMT inhibition and gemcitabine resensitization. Oncology Letters, 14(5), 5400–5408.PubMedPubMedCentral
81.
go back to reference Weadick, B., et al. (2021). EMT-induced gemcitabine resistance in pancreatic cancer involves the functional loss of equilibrative nucleoside transporter 1. Molecular Cancer Therapeutics, 20(2), 410–422.PubMed Weadick, B., et al. (2021). EMT-induced gemcitabine resistance in pancreatic cancer involves the functional loss of equilibrative nucleoside transporter 1. Molecular Cancer Therapeutics, 20(2), 410–422.PubMed
82.
go back to reference Zhu, S., et al. (2017). ASIC1 and ASIC3 contribute to acidity-induced EMT of pancreatic cancer through activating Ca(2+)/RhoA pathway. Cell Death & Disease, 8(5), e2806. Zhu, S., et al. (2017). ASIC1 and ASIC3 contribute to acidity-induced EMT of pancreatic cancer through activating Ca(2+)/RhoA pathway. Cell Death & Disease, 8(5), e2806.
83.
go back to reference Shi, C., et al. (2021). NUDCD1 knockdown inhibits the proliferation, migration, and invasion of pancreatic cancer via the EMT process. Aging (Albany NY), 13(14), 18298–18309.PubMed Shi, C., et al. (2021). NUDCD1 knockdown inhibits the proliferation, migration, and invasion of pancreatic cancer via the EMT process. Aging (Albany NY), 13(14), 18298–18309.PubMed
84.
go back to reference Zhang, Z., et al. (2017). miR-135b-5p Promotes migration, invasion and EMT of pancreatic cancer cells by targeting NR3C2. Biomedicine & Pharmacotherapy, 96, 1341–1348. Zhang, Z., et al. (2017). miR-135b-5p Promotes migration, invasion and EMT of pancreatic cancer cells by targeting NR3C2. Biomedicine & Pharmacotherapy, 96, 1341–1348.
85.
go back to reference Rumman, M., et al. (2016). HS-173, a novel PI3K inhibitor suppresses EMT and metastasis in pancreatic cancer. Oncotarget, 7(47), 78029–78047.PubMedPubMedCentral Rumman, M., et al. (2016). HS-173, a novel PI3K inhibitor suppresses EMT and metastasis in pancreatic cancer. Oncotarget, 7(47), 78029–78047.PubMedPubMedCentral
86.
go back to reference Shen, J., et al. (2019). LncRNA XIST promotes pancreatic cancer migration, invasion and EMT by sponging miR-429 to modulate ZEB1 expression. The International Journal of Biochemistry & Cell Biology, 113, 17–26. Shen, J., et al. (2019). LncRNA XIST promotes pancreatic cancer migration, invasion and EMT by sponging miR-429 to modulate ZEB1 expression. The International Journal of Biochemistry & Cell Biology, 113, 17–26.
87.
go back to reference Zhang, R., et al. (2021). Silencing of circRNA circ_0001666 represses EMT in pancreatic cancer through upregulating miR-1251 and downregulating SOX4. Frontiers in Molecular Biosciences, 8, 684866.PubMedPubMedCentral Zhang, R., et al. (2021). Silencing of circRNA circ_0001666 represses EMT in pancreatic cancer through upregulating miR-1251 and downregulating SOX4. Frontiers in Molecular Biosciences, 8, 684866.PubMedPubMedCentral
88.
go back to reference Zhang, Y., et al. (2019). Upregulation of LASP2 inhibits pancreatic cancer cell migration and invasion through suppressing TGF-β-induced EMT. Journal of Cellular Biochemistry, 120(8), 13651–13657.PubMed Zhang, Y., et al. (2019). Upregulation of LASP2 inhibits pancreatic cancer cell migration and invasion through suppressing TGF-β-induced EMT. Journal of Cellular Biochemistry, 120(8), 13651–13657.PubMed
89.
go back to reference Bhutia, Y. D., et al. (2020). Chronic exposure to excess iron promotes EMT and cancer via p53 loss in pancreatic cancer. Asian Journal of Pharmaceutical Sciences, 15(2), 237–251.PubMedPubMedCentral Bhutia, Y. D., et al. (2020). Chronic exposure to excess iron promotes EMT and cancer via p53 loss in pancreatic cancer. Asian Journal of Pharmaceutical Sciences, 15(2), 237–251.PubMedPubMedCentral
90.
go back to reference Li, P., et al. (2020). FHL3 promotes pancreatic cancer invasion and metastasis through preventing the ubiquitination degradation of EMT associated transcription factors. Aging (Albany NY), 12(1), 53–69.PubMed Li, P., et al. (2020). FHL3 promotes pancreatic cancer invasion and metastasis through preventing the ubiquitination degradation of EMT associated transcription factors. Aging (Albany NY), 12(1), 53–69.PubMed
91.
go back to reference Zhang, L., et al. (2016). CCL21/CCR7 axis contributed to CD133+ pancreatic cancer stem-like cell metastasis via EMT and Erk/NF-κB pathway. PLoS One, 11(8), e0158529.PubMedPubMedCentral Zhang, L., et al. (2016). CCL21/CCR7 axis contributed to CD133+ pancreatic cancer stem-like cell metastasis via EMT and Erk/NF-κB pathway. PLoS One, 11(8), e0158529.PubMedPubMedCentral
92.
go back to reference Zhao, H., et al. (2017). Up-regulation of glycolysis promotes the stemness and EMT phenotypes in gemcitabine-resistant pancreatic cancer cells. Journal of Cellular and Molecular Medicine, 21(9), 2055–2067.PubMedPubMedCentral Zhao, H., et al. (2017). Up-regulation of glycolysis promotes the stemness and EMT phenotypes in gemcitabine-resistant pancreatic cancer cells. Journal of Cellular and Molecular Medicine, 21(9), 2055–2067.PubMedPubMedCentral
93.
go back to reference Martinelli, P., et al. (2017). GATA6 regulates EMT and tumour dissemination, and is a marker of response to adjuvant chemotherapy in pancreatic cancer. Gut, 66(9), 1665–1676.PubMed Martinelli, P., et al. (2017). GATA6 regulates EMT and tumour dissemination, and is a marker of response to adjuvant chemotherapy in pancreatic cancer. Gut, 66(9), 1665–1676.PubMed
94.
go back to reference Gao, C., et al. (2021). Yap1-2 Isoform Is the Primary Mediator in TGF-β1 Induced EMT in Pancreatic Cancer. Frontiers in Oncology, 11, 649290.PubMedPubMedCentral Gao, C., et al. (2021). Yap1-2 Isoform Is the Primary Mediator in TGF-β1 Induced EMT in Pancreatic Cancer. Frontiers in Oncology, 11, 649290.PubMedPubMedCentral
95.
go back to reference Baer, R., et al. (2014). Pancreatic cell plasticity and cancer initiation induced by oncogenic Kras is completely dependent on wild-type PI 3-kinase p110α. Genes & Development, 28(23), 2621–2635. Baer, R., et al. (2014). Pancreatic cell plasticity and cancer initiation induced by oncogenic Kras is completely dependent on wild-type PI 3-kinase p110α. Genes & Development, 28(23), 2621–2635.
96.
go back to reference Sancho, P., et al. (2015). MYC/PGC-1α balance determines the metabolic phenotype and plasticity of pancreatic cancer stem cells. Cell Metabolism, 22(4), 590–605.PubMed Sancho, P., et al. (2015). MYC/PGC-1α balance determines the metabolic phenotype and plasticity of pancreatic cancer stem cells. Cell Metabolism, 22(4), 590–605.PubMed
97.
go back to reference Zhao, S., et al. (2016). CD44 expression level and isoform contributes to pancreatic cancer cell plasticity, invasiveness, and response to therapy. Clinical Cancer Research, 22(22), 5592–5604.PubMedPubMedCentral Zhao, S., et al. (2016). CD44 expression level and isoform contributes to pancreatic cancer cell plasticity, invasiveness, and response to therapy. Clinical Cancer Research, 22(22), 5592–5604.PubMedPubMedCentral
98.
go back to reference Suzuki, K., et al. (2017). Metadherin promotes metastasis by supporting putative cancer stem cell properties and epithelial plasticity in pancreatic cancer. Oncotarget, 8(39), 66098–66111.PubMedPubMedCentral Suzuki, K., et al. (2017). Metadherin promotes metastasis by supporting putative cancer stem cell properties and epithelial plasticity in pancreatic cancer. Oncotarget, 8(39), 66098–66111.PubMedPubMedCentral
99.
go back to reference Cruz, V. H., et al. (2019). Axl-mediated activation of TBK1 drives epithelial plasticity in pancreatic cancer. JCI Insight, 5(9), e126117.PubMed Cruz, V. H., et al. (2019). Axl-mediated activation of TBK1 drives epithelial plasticity in pancreatic cancer. JCI Insight, 5(9), e126117.PubMed
100.
go back to reference Kirane, A., et al. (2015). Warfarin blocks Gas6-mediated Axl activation required for pancreatic cancer epithelial plasticity and metastasis. Cancer Research, 75(18), 3699–3705.PubMedPubMedCentral Kirane, A., et al. (2015). Warfarin blocks Gas6-mediated Axl activation required for pancreatic cancer epithelial plasticity and metastasis. Cancer Research, 75(18), 3699–3705.PubMedPubMedCentral
101.
go back to reference Feldmann, K., et al. (2021). Mesenchymal Plasticity Regulated by Prrx1 Drives Aggressive Pancreatic Cancer Biology. Gastroenterology, 160(1), 346–361.e24.PubMed Feldmann, K., et al. (2021). Mesenchymal Plasticity Regulated by Prrx1 Drives Aggressive Pancreatic Cancer Biology. Gastroenterology, 160(1), 346–361.e24.PubMed
102.
go back to reference Perusina Lanfranca, M., et al. (2020). Interleukin 22 signaling regulates acinar cell plasticity to promote pancreatic tumor development in mice. Gastroenterology, 158(5), 1417–1432.e11.PubMed Perusina Lanfranca, M., et al. (2020). Interleukin 22 signaling regulates acinar cell plasticity to promote pancreatic tumor development in mice. Gastroenterology, 158(5), 1417–1432.e11.PubMed
103.
go back to reference Sharma, N., et al. (2020). Metabolic plasticity imparts erlotinib-resistance in pancreatic cancer by upregulating glucose-6-phosphate dehydrogenase. Cancer and Metabolism, 8, 19.PubMedPubMedCentral Sharma, N., et al. (2020). Metabolic plasticity imparts erlotinib-resistance in pancreatic cancer by upregulating glucose-6-phosphate dehydrogenase. Cancer and Metabolism, 8, 19.PubMedPubMedCentral
104.
go back to reference Yi, Z., et al. (2022). KDM6A Regulates Cell Plasticity and Pancreatic Cancer Progression by Noncanonical Activin Pathway. Cellular and Molecular Gastroenterology and Hepatology, 13(2), 643–667.PubMed Yi, Z., et al. (2022). KDM6A Regulates Cell Plasticity and Pancreatic Cancer Progression by Noncanonical Activin Pathway. Cellular and Molecular Gastroenterology and Hepatology, 13(2), 643–667.PubMed
105.
go back to reference Singh, S. K., et al. (2015). Antithetical NFATc1-Sox2 and p53-miR200 signaling networks govern pancreatic cancer cell plasticity. The EMBO Journal, 34(4), 517–530.PubMedPubMedCentral Singh, S. K., et al. (2015). Antithetical NFATc1-Sox2 and p53-miR200 signaling networks govern pancreatic cancer cell plasticity. The EMBO Journal, 34(4), 517–530.PubMedPubMedCentral
106.
go back to reference Liu, M., et al. (2021). Zinc-Dependent regulation of ZEB1 and YAP1 coactivation promotes epithelial-mesenchymal transition plasticity and metastasis in pancreatic cancer. Gastroenterology, 160(5), 1771–1783.e1.PubMed Liu, M., et al. (2021). Zinc-Dependent regulation of ZEB1 and YAP1 coactivation promotes epithelial-mesenchymal transition plasticity and metastasis in pancreatic cancer. Gastroenterology, 160(5), 1771–1783.e1.PubMed
107.
go back to reference Krebs, A. M., et al. (2017). The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nature Cell Biology, 19(5), 518–529.PubMed Krebs, A. M., et al. (2017). The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nature Cell Biology, 19(5), 518–529.PubMed
108.
go back to reference Kloesch, B., et al. (2022). A GATA6-centred gene regulatory network involving HNFs and ΔNp63 controls plasticity and immune escape in pancreatic cancer. Gut, 71(4), 766–777.PubMed Kloesch, B., et al. (2022). A GATA6-centred gene regulatory network involving HNFs and ΔNp63 controls plasticity and immune escape in pancreatic cancer. Gut, 71(4), 766–777.PubMed
109.
go back to reference Nishino, H., et al. (2017). Grainyhead-like 2 (GRHL2) regulates epithelial plasticity in pancreatic cancer progression. Cancer Medicine, 6(11), 2686–2696.PubMedPubMedCentral Nishino, H., et al. (2017). Grainyhead-like 2 (GRHL2) regulates epithelial plasticity in pancreatic cancer progression. Cancer Medicine, 6(11), 2686–2696.PubMedPubMedCentral
110.
go back to reference Raghavan, S., et al. (2021). Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer. Cell, 184(25), 6119–6137.e26.PubMedPubMedCentral Raghavan, S., et al. (2021). Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer. Cell, 184(25), 6119–6137.e26.PubMedPubMedCentral
111.
go back to reference Ireland, L., et al. (2020). Blockade of Stromal Gas6 Alters Cancer Cell Plasticity, Activates NK Cells, and Inhibits Pancreatic Cancer Metastasis. Frontiers in Immunology, 11, 297.PubMedPubMedCentral Ireland, L., et al. (2020). Blockade of Stromal Gas6 Alters Cancer Cell Plasticity, Activates NK Cells, and Inhibits Pancreatic Cancer Metastasis. Frontiers in Immunology, 11, 297.PubMedPubMedCentral
112.
go back to reference Datta, J., et al. (2022). Combined MEK and STAT3 inhibition uncovers stromal plasticity by enriching for cancer-associated fibroblasts with mesenchymal stem cell-like features to overcome immunotherapy resistance in pancreatic cancer. Gastroenterology, 163(6), 1593–1612.PubMed Datta, J., et al. (2022). Combined MEK and STAT3 inhibition uncovers stromal plasticity by enriching for cancer-associated fibroblasts with mesenchymal stem cell-like features to overcome immunotherapy resistance in pancreatic cancer. Gastroenterology, 163(6), 1593–1612.PubMed
113.
go back to reference Cao, R., et al. (2020). miR-3613-5p enhances the metastasis of pancreatic cancer by targeting CDK6. Cell Cycle, 19(22), 3086–3095.PubMedPubMedCentral Cao, R., et al. (2020). miR-3613-5p enhances the metastasis of pancreatic cancer by targeting CDK6. Cell Cycle, 19(22), 3086–3095.PubMedPubMedCentral
114.
go back to reference Chen, Z., et al. (2018). MiR-1297 suppresses pancreatic cancer cell proliferation and metastasis by targeting MTDH. Molecular and Cellular Probes, 40, 19–26.PubMed Chen, Z., et al. (2018). MiR-1297 suppresses pancreatic cancer cell proliferation and metastasis by targeting MTDH. Molecular and Cellular Probes, 40, 19–26.PubMed
115.
go back to reference Wu, X., et al. (2018). MiR-216b inhibits pancreatic cancer cell progression and promotes apoptosis by down-regulating KRAS. Archives of Medical Science, 14(6), 1321–1332.PubMed Wu, X., et al. (2018). MiR-216b inhibits pancreatic cancer cell progression and promotes apoptosis by down-regulating KRAS. Archives of Medical Science, 14(6), 1321–1332.PubMed
116.
go back to reference Zhang, D., et al. (2021). MiR-489-3p reduced pancreatic cancer proliferation and metastasis by targeting PKM2 and LDHA involving glycolysis. Frontiers in Oncology, 11, 651535.PubMedPubMedCentral Zhang, D., et al. (2021). MiR-489-3p reduced pancreatic cancer proliferation and metastasis by targeting PKM2 and LDHA involving glycolysis. Frontiers in Oncology, 11, 651535.PubMedPubMedCentral
118.
go back to reference Mirzaei, S., et al. (2022). Molecular landscape of LncRNAs in prostate cancer: A focus on pathways and therapeutic targets for intervention. Journal of Experimental & Clinical Cancer Research, 41(1), 214. Mirzaei, S., et al. (2022). Molecular landscape of LncRNAs in prostate cancer: A focus on pathways and therapeutic targets for intervention. Journal of Experimental & Clinical Cancer Research, 41(1), 214.
119.
go back to reference Luo, Y., et al. (2020). LncRNA DANCR promotes proliferation and metastasis in pancreatic cancer by regulating miRNA-33b. FEBS Open Bio, 10(1), 18–27.PubMed Luo, Y., et al. (2020). LncRNA DANCR promotes proliferation and metastasis in pancreatic cancer by regulating miRNA-33b. FEBS Open Bio, 10(1), 18–27.PubMed
120.
go back to reference Hui, B., et al. (2019). RREB1-induced upregulation of the lncRNA AGAP2-AS1 regulates the proliferation and migration of pancreatic cancer partly through suppressing ANKRD1 and ANGPTL4. Cell Death & Disease, 10(3), 207. Hui, B., et al. (2019). RREB1-induced upregulation of the lncRNA AGAP2-AS1 regulates the proliferation and migration of pancreatic cancer partly through suppressing ANKRD1 and ANGPTL4. Cell Death & Disease, 10(3), 207.
121.
go back to reference Feng, Y., et al. (2020). LncRNA NEAT1 facilitates pancreatic cancer growth and metastasis through stabilizing ELF3 mRNA. American Journal of Cancer Research, 10(1), 237–248.PubMedPubMedCentral Feng, Y., et al. (2020). LncRNA NEAT1 facilitates pancreatic cancer growth and metastasis through stabilizing ELF3 mRNA. American Journal of Cancer Research, 10(1), 237–248.PubMedPubMedCentral
122.
go back to reference Ren, X., et al. (2020). lncRNA-PLACT1 sustains activation of NF-κB pathway through a positive feedback loop with IκBα/E2F1 axis in pancreatic cancer. Molecular Cancer, 19(1), 35.PubMedPubMedCentral Ren, X., et al. (2020). lncRNA-PLACT1 sustains activation of NF-κB pathway through a positive feedback loop with IκBα/E2F1 axis in pancreatic cancer. Molecular Cancer, 19(1), 35.PubMedPubMedCentral
123.
go back to reference Shen, X., et al. (2021). Identification of Circ_001569 as a Potential biomarker in the diagnosis and prognosis of pancreatic cancer. Technology in Cancer Research & Treatment, 20, 1533033820983302. Shen, X., et al. (2021). Identification of Circ_001569 as a Potential biomarker in the diagnosis and prognosis of pancreatic cancer. Technology in Cancer Research & Treatment, 20, 1533033820983302.
124.
go back to reference Zhang, J., et al. (2023). Circ_0000284 upregulates RHPN2 to facilitate pancreatic cancer proliferation, metastasis, and angiogenesis through sponging miR-1179. Journal of Biochemical and Molecular Toxicology, 37(3), e23274.PubMed Zhang, J., et al. (2023). Circ_0000284 upregulates RHPN2 to facilitate pancreatic cancer proliferation, metastasis, and angiogenesis through sponging miR-1179. Journal of Biochemical and Molecular Toxicology, 37(3), e23274.PubMed
125.
go back to reference Li, C., et al. (2023). Downregulation of circ-STK39 suppresses pancreatic cancer progression by sponging mir-140-3p and regulating TRAM2-mediated epithelial-mesenchymal transition. Apoptosis, 28, 1024–1034.PubMed Li, C., et al. (2023). Downregulation of circ-STK39 suppresses pancreatic cancer progression by sponging mir-140-3p and regulating TRAM2-mediated epithelial-mesenchymal transition. Apoptosis, 28, 1024–1034.PubMed
126.
go back to reference Li, M., et al. (2020). miR-193a-5p promotes pancreatic cancer cell metastasis through SRSF6-mediated alternative splicing of OGDHL and ECM1. American Journal of Cancer Research, 10(1), 38–59.PubMedPubMedCentral Li, M., et al. (2020). miR-193a-5p promotes pancreatic cancer cell metastasis through SRSF6-mediated alternative splicing of OGDHL and ECM1. American Journal of Cancer Research, 10(1), 38–59.PubMedPubMedCentral
127.
go back to reference Mody, H. R., et al. (2017). miR-202 Diminishes TGFβ Receptors and Attenuates TGFβ1-Induced EMT in Pancreatic Cancer. Molecular Cancer Research, 15(8), 1029–1039.PubMed Mody, H. R., et al. (2017). miR-202 Diminishes TGFβ Receptors and Attenuates TGFβ1-Induced EMT in Pancreatic Cancer. Molecular Cancer Research, 15(8), 1029–1039.PubMed
128.
go back to reference Jiang, J., et al. (2015). Reduction of miR-29c enhances pancreatic cancer cell migration and stem cell-like phenotype. Oncotarget, 6(5), 2767–2778.PubMed Jiang, J., et al. (2015). Reduction of miR-29c enhances pancreatic cancer cell migration and stem cell-like phenotype. Oncotarget, 6(5), 2767–2778.PubMed
129.
go back to reference Wu, X., et al. (2016). MiR-200a suppresses the proliferation and metastasis in pancreatic ductal adenocarcinoma through downregulation of DEK Gene. Translational Oncology, 9(1), 25–31.PubMedPubMedCentral Wu, X., et al. (2016). MiR-200a suppresses the proliferation and metastasis in pancreatic ductal adenocarcinoma through downregulation of DEK Gene. Translational Oncology, 9(1), 25–31.PubMedPubMedCentral
130.
go back to reference Hu, Y., et al. (2012). miR-143 inhibits the metastasis of pancreatic cancer and an associated signaling pathway. Tumour Biology, 33(6), 1863–1870.PubMed Hu, Y., et al. (2012). miR-143 inhibits the metastasis of pancreatic cancer and an associated signaling pathway. Tumour Biology, 33(6), 1863–1870.PubMed
131.
go back to reference Chen, Y., et al. (2019). MicroRNA hsa-miR-623 directly suppresses MMP1 and attenuates IL-8-induced metastasis in pancreatic cancer. International Journal of Oncology, 55(1), 142–156.PubMedPubMedCentral Chen, Y., et al. (2019). MicroRNA hsa-miR-623 directly suppresses MMP1 and attenuates IL-8-induced metastasis in pancreatic cancer. International Journal of Oncology, 55(1), 142–156.PubMedPubMedCentral
132.
go back to reference Ma, L., et al. (2019). The miR-141/neuropilin-1 axis is associated with the clinicopathology and contributes to the growth and metastasis of pancreatic cancer. Cancer Cell International, 19, 248.PubMedPubMedCentral Ma, L., et al. (2019). The miR-141/neuropilin-1 axis is associated with the clinicopathology and contributes to the growth and metastasis of pancreatic cancer. Cancer Cell International, 19, 248.PubMedPubMedCentral
133.
go back to reference Wang, L., Mu, N., & Qu, N. (2021). Methylation of the miR-29b-3p promoter contributes to angiogenesis, invasion, and migration in pancreatic cancer. Oncology Reports, 45(1), 65–72.PubMedPubMedCentral Wang, L., Mu, N., & Qu, N. (2021). Methylation of the miR-29b-3p promoter contributes to angiogenesis, invasion, and migration in pancreatic cancer. Oncology Reports, 45(1), 65–72.PubMedPubMedCentral
134.
go back to reference Ma, C., et al. (2014). miR-212 promotes pancreatic cancer cell growth and invasion by targeting the hedgehog signaling pathway receptor patched-1. Journal of Experimental & Clinical Cancer Research, 33(1), 54. Ma, C., et al. (2014). miR-212 promotes pancreatic cancer cell growth and invasion by targeting the hedgehog signaling pathway receptor patched-1. Journal of Experimental & Clinical Cancer Research, 33(1), 54.
135.
go back to reference Xue, L., et al. (2020). miR-539 suppresses the proliferation, migration, invasion and epithelial mesenchymal transition of pancreatic cancer cells through targeting SP1. International Journal of Molecular Medicine, 45(6), 1771–1782.PubMedPubMedCentral Xue, L., et al. (2020). miR-539 suppresses the proliferation, migration, invasion and epithelial mesenchymal transition of pancreatic cancer cells through targeting SP1. International Journal of Molecular Medicine, 45(6), 1771–1782.PubMedPubMedCentral
136.
go back to reference Hiramoto, H., et al. (2017). miR-509-5p and miR-1243 increase the sensitivity to gemcitabine by inhibiting epithelial-mesenchymal transition in pancreatic cancer. Scientific Reports, 7(1), 4002.PubMedPubMedCentral Hiramoto, H., et al. (2017). miR-509-5p and miR-1243 increase the sensitivity to gemcitabine by inhibiting epithelial-mesenchymal transition in pancreatic cancer. Scientific Reports, 7(1), 4002.PubMedPubMedCentral
137.
go back to reference Deng, S. J., et al. (2018). Hypoxia-induced LncRNA-BX111 promotes metastasis and progression of pancreatic cancer through regulating ZEB1 transcription. Oncogene, 37(44), 5811–5828.PubMed Deng, S. J., et al. (2018). Hypoxia-induced LncRNA-BX111 promotes metastasis and progression of pancreatic cancer through regulating ZEB1 transcription. Oncogene, 37(44), 5811–5828.PubMed
138.
go back to reference Cheng, C., et al. (2022). Positive feedback regulation of lncRNA TPT1-AS1 and ITGB3 promotes cell growth and metastasis in pancreatic cancer. Cancer Science, 113(9), 2986–3001.PubMedPubMedCentral Cheng, C., et al. (2022). Positive feedback regulation of lncRNA TPT1-AS1 and ITGB3 promotes cell growth and metastasis in pancreatic cancer. Cancer Science, 113(9), 2986–3001.PubMedPubMedCentral
139.
go back to reference Lou, S., et al. (2019). Downregulation of lncRNA AFAP1-AS1 by oridonin inhibits the epithelial-to-mesenchymal transition and proliferation of pancreatic cancer cells. Acta Biochimica et Biophysica Sinica Shanghai, 51(8), 814–825. Lou, S., et al. (2019). Downregulation of lncRNA AFAP1-AS1 by oridonin inhibits the epithelial-to-mesenchymal transition and proliferation of pancreatic cancer cells. Acta Biochimica et Biophysica Sinica Shanghai, 51(8), 814–825.
140.
go back to reference Cui, X. P., et al. (2019). LncRNA TP73-AS1 sponges miR-141-3p to promote the migration and invasion of pancreatic cancer cells through the up-regulation of BDH2. Bioscience Reports, 39(3), BSR20181937.PubMedPubMedCentral Cui, X. P., et al. (2019). LncRNA TP73-AS1 sponges miR-141-3p to promote the migration and invasion of pancreatic cancer cells through the up-regulation of BDH2. Bioscience Reports, 39(3), BSR20181937.PubMedPubMedCentral
141.
go back to reference Liu, W., et al. (2020). A novel lncRNA PTTG3P/miR-132/212-3p/FoxM1 feedback loop facilitates tumorigenesis and metastasis of pancreatic cancer. Cell death discovery, 6(1), 136.PubMedPubMedCentral Liu, W., et al. (2020). A novel lncRNA PTTG3P/miR-132/212-3p/FoxM1 feedback loop facilitates tumorigenesis and metastasis of pancreatic cancer. Cell death discovery, 6(1), 136.PubMedPubMedCentral
142.
go back to reference An, Y., et al. (2018). LncRNA DLX6-AS1 promoted cancer cell proliferation and invasion by attenuating the endogenous function of miR-181b in pancreatic cancer. Cancer Cell International, 18, 143.PubMedPubMedCentral An, Y., et al. (2018). LncRNA DLX6-AS1 promoted cancer cell proliferation and invasion by attenuating the endogenous function of miR-181b in pancreatic cancer. Cancer Cell International, 18, 143.PubMedPubMedCentral
143.
go back to reference Liu, B., et al. (2018). lncRNA GAS5 reverses EMT and tumor stem cell-mediated gemcitabine resistance and metastasis by targeting miR-221/SOCS3 in pancreatic cancer. Molecular Therapy--Nucleic Acids, 13, 472–482.PubMedPubMedCentral Liu, B., et al. (2018). lncRNA GAS5 reverses EMT and tumor stem cell-mediated gemcitabine resistance and metastasis by targeting miR-221/SOCS3 in pancreatic cancer. Molecular Therapy--Nucleic Acids, 13, 472–482.PubMedPubMedCentral
144.
go back to reference Gu, L., et al. (2017). lncRNA MEG3 had anti-cancer effects to suppress pancreatic cancer activity. Biomedicine & Pharmacotherapy, 89, 1269–1276. Gu, L., et al. (2017). lncRNA MEG3 had anti-cancer effects to suppress pancreatic cancer activity. Biomedicine & Pharmacotherapy, 89, 1269–1276.
145.
go back to reference Liu, S., et al. (2019). LncRNA SNHG16 promotes tumor growth of pancreatic cancer by targeting miR-218-5p. Biomedicine & Pharmacotherapy, 114, 108862. Liu, S., et al. (2019). LncRNA SNHG16 promotes tumor growth of pancreatic cancer by targeting miR-218-5p. Biomedicine & Pharmacotherapy, 114, 108862.
146.
go back to reference Deng, P. C., et al. (2019). LncRNA SNHG14 potentiates pancreatic cancer progression via modulation of annexin A2 expression by acting as a competing endogenous RNA for miR-613. Journal of Cellular and Molecular Medicine, 23(11), 7222–7232.PubMedPubMedCentral Deng, P. C., et al. (2019). LncRNA SNHG14 potentiates pancreatic cancer progression via modulation of annexin A2 expression by acting as a competing endogenous RNA for miR-613. Journal of Cellular and Molecular Medicine, 23(11), 7222–7232.PubMedPubMedCentral
147.
go back to reference Huang, S., et al. (2021). LncRNA PWAR6 regulates proliferation and migration by epigenetically silencing YAP1 in tumorigenesis of pancreatic ductal adenocarcinoma. Journal of Cellular and Molecular Medicine, 25(9), 4275–4286.PubMedPubMedCentral Huang, S., et al. (2021). LncRNA PWAR6 regulates proliferation and migration by epigenetically silencing YAP1 in tumorigenesis of pancreatic ductal adenocarcinoma. Journal of Cellular and Molecular Medicine, 25(9), 4275–4286.PubMedPubMedCentral
148.
go back to reference Chen, S., et al. (2020). LncRNA STXBP5-AS1 suppresses stem cell-like properties of pancreatic cancer by epigenetically inhibiting neighboring androglobin gene expression. Clinical Epigenetics, 12(1), 168.PubMedPubMedCentral Chen, S., et al. (2020). LncRNA STXBP5-AS1 suppresses stem cell-like properties of pancreatic cancer by epigenetically inhibiting neighboring androglobin gene expression. Clinical Epigenetics, 12(1), 168.PubMedPubMedCentral
149.
go back to reference Cai, J., et al. (2021). Curcumin attenuates lncRNA H19-induced epithelial-mesenchymal transition in tamoxifen-resistant breast cancer cells. Molecular Medicine Reports, 23(1), 1. Cai, J., et al. (2021). Curcumin attenuates lncRNA H19-induced epithelial-mesenchymal transition in tamoxifen-resistant breast cancer cells. Molecular Medicine Reports, 23(1), 1.
150.
go back to reference Zhou, M., et al. (2022). LncRNA FAM83H-AS1 promotes the malignant progression of pancreatic ductal adenocarcinoma by stabilizing FAM83H mRNA to protect β-catenin from degradation. Journal of Experimental & Clinical Cancer Research, 41(1), 288. Zhou, M., et al. (2022). LncRNA FAM83H-AS1 promotes the malignant progression of pancreatic ductal adenocarcinoma by stabilizing FAM83H mRNA to protect β-catenin from degradation. Journal of Experimental & Clinical Cancer Research, 41(1), 288.
151.
go back to reference Cai, H., et al. (2017). LncRNA HOTAIR acts a competing endogenous RNA to control the expression of notch3 via sponging miR-613 in pancreatic cancer. Oncotarget, 8(20), 32905–32917.PubMedPubMedCentral Cai, H., et al. (2017). LncRNA HOTAIR acts a competing endogenous RNA to control the expression of notch3 via sponging miR-613 in pancreatic cancer. Oncotarget, 8(20), 32905–32917.PubMedPubMedCentral
152.
go back to reference Miao, H., et al. (2021). LncRNA TP73-AS1 enhances the malignant properties of pancreatic ductal adenocarcinoma by increasing MMP14 expression through miRNA -200a sponging. Journal of Cellular and Molecular Medicine, 25(7), 3654–3664.PubMedPubMedCentral Miao, H., et al. (2021). LncRNA TP73-AS1 enhances the malignant properties of pancreatic ductal adenocarcinoma by increasing MMP14 expression through miRNA -200a sponging. Journal of Cellular and Molecular Medicine, 25(7), 3654–3664.PubMedPubMedCentral
153.
go back to reference Zhang, X., et al. (2020). Circ_0075829 facilitates the progression of pancreatic carcinoma by sponging miR-1287-5p and activating LAMTOR3 signalling. Journal of Cellular and Molecular Medicine, 24(24), 14596–14607.PubMedPubMedCentral Zhang, X., et al. (2020). Circ_0075829 facilitates the progression of pancreatic carcinoma by sponging miR-1287-5p and activating LAMTOR3 signalling. Journal of Cellular and Molecular Medicine, 24(24), 14596–14607.PubMedPubMedCentral
154.
go back to reference Xing, C., et al. (2019). Circular RNA ADAM9 facilitates the malignant behaviours of pancreatic cancer by sponging miR-217 and upregulating PRSS3 expression. Artificial Cells, Nanomedicine and Biotechnology, 47(1), 3920–3928.PubMed Xing, C., et al. (2019). Circular RNA ADAM9 facilitates the malignant behaviours of pancreatic cancer by sponging miR-217 and upregulating PRSS3 expression. Artificial Cells, Nanomedicine and Biotechnology, 47(1), 3920–3928.PubMed
155.
go back to reference Wang, K. Q., et al. (2022). Circular RNA fibroblast growth factor receptor 1 promotes pancreatic cancer progression by targeting microRNA-532-3p/PIK3CB Axis. Pancreas, 51(8), 930–942.PubMed Wang, K. Q., et al. (2022). Circular RNA fibroblast growth factor receptor 1 promotes pancreatic cancer progression by targeting microRNA-532-3p/PIK3CB Axis. Pancreas, 51(8), 930–942.PubMed
156.
go back to reference Messex, J. K., et al. (2022). Oncogenic Kras-mediated cytokine CCL15 regulates pancreatic cancer cell migration and invasion through ROS. Cancers (Basel), 14(9), 2153.PubMed Messex, J. K., et al. (2022). Oncogenic Kras-mediated cytokine CCL15 regulates pancreatic cancer cell migration and invasion through ROS. Cancers (Basel), 14(9), 2153.PubMed
157.
go back to reference Deng, J., et al. (2022). High Glucose promotes pancreatic ductal adenocarcinoma gemcitabine resistance and invasion through modulating ROS/MMP-3 signaling pathway. Oxidative Medicine and Cellular Longevity, 2022, 3243647.PubMedPubMedCentral Deng, J., et al. (2022). High Glucose promotes pancreatic ductal adenocarcinoma gemcitabine resistance and invasion through modulating ROS/MMP-3 signaling pathway. Oxidative Medicine and Cellular Longevity, 2022, 3243647.PubMedPubMedCentral
158.
go back to reference Binker-Cosen, M. J., et al. (2017). Palmitic acid increases invasiveness of pancreatic cancer cells AsPC-1 through TLR4/ROS/NF-κB/MMP-9 signaling pathway. Biochemical and Biophysical Research Communications, 484(1), 152–158.PubMed Binker-Cosen, M. J., et al. (2017). Palmitic acid increases invasiveness of pancreatic cancer cells AsPC-1 through TLR4/ROS/NF-κB/MMP-9 signaling pathway. Biochemical and Biophysical Research Communications, 484(1), 152–158.PubMed
159.
go back to reference Shimojo, Y., et al. (2013). Attenuation of reactive oxygen species by antioxidants suppresses hypoxia-induced epithelial-mesenchymal transition and metastasis of pancreatic cancer cells. Clinical & Experimental Metastasis, 30(2), 143–154. Shimojo, Y., et al. (2013). Attenuation of reactive oxygen species by antioxidants suppresses hypoxia-induced epithelial-mesenchymal transition and metastasis of pancreatic cancer cells. Clinical & Experimental Metastasis, 30(2), 143–154.
160.
go back to reference Wörmann, S. M., et al. (2021). APOBEC3A drives deaminase domain-independent chromosomal instability to promote pancreatic cancer metastasis. Nature Cancer, 2(12), 1338–1356.PubMed Wörmann, S. M., et al. (2021). APOBEC3A drives deaminase domain-independent chromosomal instability to promote pancreatic cancer metastasis. Nature Cancer, 2(12), 1338–1356.PubMed
161.
go back to reference Zhao, T., et al. (2020). IL-37/ STAT3/ HIF-1α negative feedback signaling drives gemcitabine resistance in pancreatic cancer. Theranostics, 10(9), 4088–4100.PubMedPubMedCentral Zhao, T., et al. (2020). IL-37/ STAT3/ HIF-1α negative feedback signaling drives gemcitabine resistance in pancreatic cancer. Theranostics, 10(9), 4088–4100.PubMedPubMedCentral
162.
go back to reference Hu, H., et al. (2020). MicroRNA-301a promotes pancreatic cancer invasion and metastasis through the JAK/STAT3 signaling pathway by targeting SOCS5. Carcinogenesis, 41(4), 502–514.PubMed Hu, H., et al. (2020). MicroRNA-301a promotes pancreatic cancer invasion and metastasis through the JAK/STAT3 signaling pathway by targeting SOCS5. Carcinogenesis, 41(4), 502–514.PubMed
163.
go back to reference Huang, C., et al. (2011). STAT3-targeting RNA interference inhibits pancreatic cancer angiogenesis in vitro and in vivo. International Journal of Oncology, 38(6), 1637–1644.PubMed Huang, C., et al. (2011). STAT3-targeting RNA interference inhibits pancreatic cancer angiogenesis in vitro and in vivo. International Journal of Oncology, 38(6), 1637–1644.PubMed
164.
go back to reference Fofaria, N. M., & Srivastava, S. K. (2015). STAT3 induces anoikis resistance, promotes cell invasion and metastatic potential in pancreatic cancer cells. Carcinogenesis, 36(1), 142–150.PubMed Fofaria, N. M., & Srivastava, S. K. (2015). STAT3 induces anoikis resistance, promotes cell invasion and metastatic potential in pancreatic cancer cells. Carcinogenesis, 36(1), 142–150.PubMed
165.
go back to reference Chen, H., et al. (2023). Selectively Targeting STAT3 Using a Small Molecule Inhibitor is a Potential Therapeutic Strategy for Pancreatic Cancer. Clinical Cancer Research, 29(4), 815–830.PubMed Chen, H., et al. (2023). Selectively Targeting STAT3 Using a Small Molecule Inhibitor is a Potential Therapeutic Strategy for Pancreatic Cancer. Clinical Cancer Research, 29(4), 815–830.PubMed
166.
go back to reference Li, H., et al. (2011). STAT3 knockdown reduces pancreatic cancer cell invasiveness and matrix metalloproteinase-7 expression in nude mice. PLoS One, 6(10), e25941.PubMedPubMedCentral Li, H., et al. (2011). STAT3 knockdown reduces pancreatic cancer cell invasiveness and matrix metalloproteinase-7 expression in nude mice. PLoS One, 6(10), e25941.PubMedPubMedCentral
167.
go back to reference Li, Y., Zhang, Y., & Liu, J. (2019). NETO2 promotes pancreatic cancer cell proliferation, invasion and migration via activation of the STAT3 signaling pathway. Cancer Management and Research, 11, 5147–5156.PubMedPubMedCentral Li, Y., Zhang, Y., & Liu, J. (2019). NETO2 promotes pancreatic cancer cell proliferation, invasion and migration via activation of the STAT3 signaling pathway. Cancer Management and Research, 11, 5147–5156.PubMedPubMedCentral
168.
go back to reference Hu, B., et al. (2016). HIC1 attenuates invasion and metastasis by inhibiting the IL-6/STAT3 signalling pathway in human pancreatic cancer. Cancer Letters, 376(2), 387–398.PubMed Hu, B., et al. (2016). HIC1 attenuates invasion and metastasis by inhibiting the IL-6/STAT3 signalling pathway in human pancreatic cancer. Cancer Letters, 376(2), 387–398.PubMed
169.
go back to reference Zuo, C., et al. (2018). Celecoxib suppresses proliferation and metastasis of pancreatic cancer cells by down-regulating STAT3 / NF-kB and L1CAM activities. Pancreatology, 18(3), 328–333.PubMed Zuo, C., et al. (2018). Celecoxib suppresses proliferation and metastasis of pancreatic cancer cells by down-regulating STAT3 / NF-kB and L1CAM activities. Pancreatology, 18(3), 328–333.PubMed
170.
go back to reference Lian, J., et al. (2020). Ubiquitin specific peptidase 5 enhances STAT3 signaling and promotes migration and invasion in Pancreatic Cancer. Journal of Cancer, 11(23), 6802–6811.PubMedPubMedCentral Lian, J., et al. (2020). Ubiquitin specific peptidase 5 enhances STAT3 signaling and promotes migration and invasion in Pancreatic Cancer. Journal of Cancer, 11(23), 6802–6811.PubMedPubMedCentral
171.
go back to reference Chen, J., et al. (2016). Interleukin-32α inactivates JAK2/STAT3 signaling and reverses interleukin-6-induced epithelial-mesenchymal transition, invasion, and metastasis in pancreatic cancer cells. Oncotargets and Therapy, 9, 4225–4237.PubMedPubMedCentral Chen, J., et al. (2016). Interleukin-32α inactivates JAK2/STAT3 signaling and reverses interleukin-6-induced epithelial-mesenchymal transition, invasion, and metastasis in pancreatic cancer cells. Oncotargets and Therapy, 9, 4225–4237.PubMedPubMedCentral
172.
go back to reference Zhang, Z., et al. (2019). BRM transcriptionally regulates miR-302a-3p to target SOCS5/STAT3 signaling axis to potentiate pancreatic cancer metastasis. Cancer Letters, 449, 215–225.PubMed Zhang, Z., et al. (2019). BRM transcriptionally regulates miR-302a-3p to target SOCS5/STAT3 signaling axis to potentiate pancreatic cancer metastasis. Cancer Letters, 449, 215–225.PubMed
174.
go back to reference Liu, L., et al. (2019). Circular RNA ciRS-7 promotes the proliferation and metastasis of pancreatic cancer by regulating miR-7-mediated EGFR/STAT3 signaling pathway. Hepatobiliary & Pancreatic Diseases International, 18(6), 580–586. Liu, L., et al. (2019). Circular RNA ciRS-7 promotes the proliferation and metastasis of pancreatic cancer by regulating miR-7-mediated EGFR/STAT3 signaling pathway. Hepatobiliary & Pancreatic Diseases International, 18(6), 580–586.
175.
go back to reference Guo, Y., et al. (2021). The anti-dysenteric drug fraxetin enhances anti-tumor efficacy of gemcitabine and suppresses pancreatic cancer development by antagonizing STAT3 activation. Aging (Albany NY), 13(14), 18545–18563.PubMed Guo, Y., et al. (2021). The anti-dysenteric drug fraxetin enhances anti-tumor efficacy of gemcitabine and suppresses pancreatic cancer development by antagonizing STAT3 activation. Aging (Albany NY), 13(14), 18545–18563.PubMed
176.
go back to reference Al-Ismaeel, Q., et al. (2019). ZEB1 and IL-6/11-STAT3 signalling cooperate to define invasive potential of pancreatic cancer cells via differential regulation of the expression of S100 proteins. British Journal of Cancer, 121(1), 65–75.PubMedPubMedCentral Al-Ismaeel, Q., et al. (2019). ZEB1 and IL-6/11-STAT3 signalling cooperate to define invasive potential of pancreatic cancer cells via differential regulation of the expression of S100 proteins. British Journal of Cancer, 121(1), 65–75.PubMedPubMedCentral
177.
go back to reference Su, K., et al. (2023). Solasodine suppresses the metastasis of gastric cancer through claudin-2 via the AMPK/STAT3/NF-κB pathway. Chemico-Biological Interactions, 379, 110520.PubMed Su, K., et al. (2023). Solasodine suppresses the metastasis of gastric cancer through claudin-2 via the AMPK/STAT3/NF-κB pathway. Chemico-Biological Interactions, 379, 110520.PubMed
178.
go back to reference Huang, C., et al. (2013). Regulation of miR-155 affects pancreatic cancer cell invasiveness and migration by modulating the STAT3 signaling pathway through SOCS1. Oncology Reports, 30(3), 1223–1230.PubMed Huang, C., et al. (2013). Regulation of miR-155 affects pancreatic cancer cell invasiveness and migration by modulating the STAT3 signaling pathway through SOCS1. Oncology Reports, 30(3), 1223–1230.PubMed
179.
go back to reference Zhao, G., et al. (2013). MiR-130b is a prognostic marker and inhibits cell proliferation and invasion in pancreatic cancer through targeting STAT3. PLoS One, 8(9), e73803.PubMedPubMedCentral Zhao, G., et al. (2013). MiR-130b is a prognostic marker and inhibits cell proliferation and invasion in pancreatic cancer through targeting STAT3. PLoS One, 8(9), e73803.PubMedPubMedCentral
180.
go back to reference Wei, D., et al. (2003). Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene, 22(3), 319–329.PubMed Wei, D., et al. (2003). Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene, 22(3), 319–329.PubMed
181.
go back to reference Zhao, S., et al. (2008). Inhibition of STAT3 Tyr705 phosphorylation by Smad4 suppresses transforming growth factor beta-mediated invasion and metastasis in pancreatic cancer cells. Cancer Research, 68(11), 4221–4228.PubMed Zhao, S., et al. (2008). Inhibition of STAT3 Tyr705 phosphorylation by Smad4 suppresses transforming growth factor beta-mediated invasion and metastasis in pancreatic cancer cells. Cancer Research, 68(11), 4221–4228.PubMed
182.
go back to reference Sasaki, N., et al. (2022). Gp130-Mediated STAT3 Activation contributes to the aggressiveness of pancreatic cancer through H19 long non-coding RNA expression. Cancers (Basel), 14(9), 2055.PubMed Sasaki, N., et al. (2022). Gp130-Mediated STAT3 Activation contributes to the aggressiveness of pancreatic cancer through H19 long non-coding RNA expression. Cancers (Basel), 14(9), 2055.PubMed
183.
go back to reference Wu, Y. S., et al. (2017). Paracrine IL-6 signaling mediates the effects of pancreatic stellate cells on epithelial-mesenchymal transition via Stat3/Nrf2 pathway in pancreatic cancer cells. Biochimica et Biophysica Acta - General Subjects, 1861(2), 296–306.PubMed Wu, Y. S., et al. (2017). Paracrine IL-6 signaling mediates the effects of pancreatic stellate cells on epithelial-mesenchymal transition via Stat3/Nrf2 pathway in pancreatic cancer cells. Biochimica et Biophysica Acta - General Subjects, 1861(2), 296–306.PubMed
184.
go back to reference Liu, M., et al. (2019). Hypoxia-induced feedback of HIF-1α and lncRNA-CF129 contributes to pancreatic cancer progression through stabilization of p53 protein. Theranostics, 9(16), 4795–4810.PubMedPubMedCentral Liu, M., et al. (2019). Hypoxia-induced feedback of HIF-1α and lncRNA-CF129 contributes to pancreatic cancer progression through stabilization of p53 protein. Theranostics, 9(16), 4795–4810.PubMedPubMedCentral
185.
go back to reference Jin, Y., et al. (2021). Positive Reciprocal Feedback of lncRNA ZEB1-AS1 and HIF-1α contributes to hypoxia-promoted tumorigenesis and metastasis of pancreatic cancer. Frontiers in Oncology, 11, 761979.PubMedPubMedCentral Jin, Y., et al. (2021). Positive Reciprocal Feedback of lncRNA ZEB1-AS1 and HIF-1α contributes to hypoxia-promoted tumorigenesis and metastasis of pancreatic cancer. Frontiers in Oncology, 11, 761979.PubMedPubMedCentral
186.
go back to reference Niu, Y., et al. (2018). MiRNA-646-mediated reciprocal repression between HIF-1α and MIIP contributes to tumorigenesis of pancreatic cancer. Oncogene, 37(13), 1743–1758.PubMed Niu, Y., et al. (2018). MiRNA-646-mediated reciprocal repression between HIF-1α and MIIP contributes to tumorigenesis of pancreatic cancer. Oncogene, 37(13), 1743–1758.PubMed
187.
go back to reference Yue, H., Liu, L., & Song, Z. (2019). miR-212 regulated by HIF-1α promotes the progression of pancreatic cancer. Experimental and Therapeutic Medicine, 17(3), 2359–2365.PubMedPubMedCentral Yue, H., Liu, L., & Song, Z. (2019). miR-212 regulated by HIF-1α promotes the progression of pancreatic cancer. Experimental and Therapeutic Medicine, 17(3), 2359–2365.PubMedPubMedCentral
188.
go back to reference Wang, L., et al. (2021). N2E4, a Monoclonal antibody targeting neuropilin-2, inhibits tumor growth and metastasis in pancreatic ductal adenocarcinoma via suppressing FAK/Erk/HIF-1α Signaling. Frontiers in Oncology, 11, 657008.PubMedPubMedCentral Wang, L., et al. (2021). N2E4, a Monoclonal antibody targeting neuropilin-2, inhibits tumor growth and metastasis in pancreatic ductal adenocarcinoma via suppressing FAK/Erk/HIF-1α Signaling. Frontiers in Oncology, 11, 657008.PubMedPubMedCentral
189.
go back to reference Zhang, D., et al. (2016). β2-adrenogenic signaling regulates NNK-induced pancreatic cancer progression via upregulation of HIF-1α. Oncotarget, 7(14), 17760–17772.PubMed Zhang, D., et al. (2016). β2-adrenogenic signaling regulates NNK-induced pancreatic cancer progression via upregulation of HIF-1α. Oncotarget, 7(14), 17760–17772.PubMed
190.
go back to reference Zeng, Z., et al. (2019). LncRNA-MTA2TR functions as a promoter in pancreatic cancer via driving deacetylation-dependent accumulation of HIF-1α. Theranostics, 9(18), 5298–5314.PubMedPubMedCentral Zeng, Z., et al. (2019). LncRNA-MTA2TR functions as a promoter in pancreatic cancer via driving deacetylation-dependent accumulation of HIF-1α. Theranostics, 9(18), 5298–5314.PubMedPubMedCentral
191.
go back to reference Zhu, G. H., et al. (2013). Hypoxia-induced snail expression through transcriptional regulation by HIF-1α in pancreatic cancer cells. Digestive Diseases and Sciences, 58(12), 3503–3515.PubMed Zhu, G. H., et al. (2013). Hypoxia-induced snail expression through transcriptional regulation by HIF-1α in pancreatic cancer cells. Digestive Diseases and Sciences, 58(12), 3503–3515.PubMed
192.
go back to reference Zhang, Q., et al. (2020). Expression of the PTEN/FOXO3a/PLZF signalling pathway in pancreatic cancer and its significance in tumourigenesis and progression. Investigational New Drugs, 38(2), 321–328.PubMed Zhang, Q., et al. (2020). Expression of the PTEN/FOXO3a/PLZF signalling pathway in pancreatic cancer and its significance in tumourigenesis and progression. Investigational New Drugs, 38(2), 321–328.PubMed
193.
go back to reference Gao, Z. Q., et al. (2017). Long non-coding RNA GAS5 suppresses pancreatic cancer metastasis through modulating miR-32-5p/PTEN axis. Cell & Bioscience, 7, 66. Gao, Z. Q., et al. (2017). Long non-coding RNA GAS5 suppresses pancreatic cancer metastasis through modulating miR-32-5p/PTEN axis. Cell & Bioscience, 7, 66.
194.
go back to reference Liu, Y., et al. (2020). Role of miR-92a-3p/PTEN axis in regulation of pancreatic cancer cell proliferation and metastasis. Zhong Nan Da Xue Xue Bao. Yi Xue Ban, 45(3), 280–289.PubMed Liu, Y., et al. (2020). Role of miR-92a-3p/PTEN axis in regulation of pancreatic cancer cell proliferation and metastasis. Zhong Nan Da Xue Xue Bao. Yi Xue Ban, 45(3), 280–289.PubMed
195.
go back to reference Gu, J., et al. (2016). GFRα2 prompts cell growth and chemoresistance through down-regulating tumor suppressor gene PTEN via Mir-17-5p in pancreatic cancer. Cancer Letters, 380(2), 434–441.PubMed Gu, J., et al. (2016). GFRα2 prompts cell growth and chemoresistance through down-regulating tumor suppressor gene PTEN via Mir-17-5p in pancreatic cancer. Cancer Letters, 380(2), 434–441.PubMed
196.
go back to reference Wang, M. C., et al. (2016). Polycomb complex protein BMI-1 promotes invasion and metastasis of pancreatic cancer stem cells by activating PI3K/AKT signaling, an ex vivo, in vitro, and in vivo study. Oncotarget, 7(8), 9586–9599.PubMedPubMedCentral Wang, M. C., et al. (2016). Polycomb complex protein BMI-1 promotes invasion and metastasis of pancreatic cancer stem cells by activating PI3K/AKT signaling, an ex vivo, in vitro, and in vivo study. Oncotarget, 7(8), 9586–9599.PubMedPubMedCentral
197.
go back to reference Zhao, H., et al. (2014). Activation of glucagon-like peptide-1 receptor inhibits tumourigenicity and metastasis of human pancreatic cancer cells via PI3K/Akt pathway. Diabetes, Obesity and Metabolism, 16(9), 850–860.PubMed Zhao, H., et al. (2014). Activation of glucagon-like peptide-1 receptor inhibits tumourigenicity and metastasis of human pancreatic cancer cells via PI3K/Akt pathway. Diabetes, Obesity and Metabolism, 16(9), 850–860.PubMed
198.
go back to reference Fujita, M., et al. (2014). Nitric oxide increases the invasion of pancreatic cancer cells via activation of the PI3K-AKT and RhoA pathways after carbon ion irradiation. FEBS Letters, 588(17), 3240–3250.PubMed Fujita, M., et al. (2014). Nitric oxide increases the invasion of pancreatic cancer cells via activation of the PI3K-AKT and RhoA pathways after carbon ion irradiation. FEBS Letters, 588(17), 3240–3250.PubMed
199.
go back to reference Li, H., et al. (2014). Effect of PTEN and KAI1 gene overexpression on the proliferation, metastasis and radiosensitivity of ASPC-1 pancreatic cancer cells under hypoxic conditions. Molecular Medicine Reports, 10(4), 1973–1977.PubMed Li, H., et al. (2014). Effect of PTEN and KAI1 gene overexpression on the proliferation, metastasis and radiosensitivity of ASPC-1 pancreatic cancer cells under hypoxic conditions. Molecular Medicine Reports, 10(4), 1973–1977.PubMed
200.
go back to reference Ma, J., et al. (2010). IGF-1 mediates PTEN suppression and enhances cell invasion and proliferation via activation of the IGF-1/PI3K/Akt signaling pathway in pancreatic cancer cells. The Journal of Surgical Research, 160(1), 90–101.PubMed Ma, J., et al. (2010). IGF-1 mediates PTEN suppression and enhances cell invasion and proliferation via activation of the IGF-1/PI3K/Akt signaling pathway in pancreatic cancer cells. The Journal of Surgical Research, 160(1), 90–101.PubMed
201.
go back to reference Zhang, H., et al. (2019). MiR-132 promotes the proliferation, invasion and migration of human pancreatic carcinoma by inhibition of the tumor suppressor gene PTEN. Progress in Biophysics and Molecular Biology, 148, 65–72.PubMed Zhang, H., et al. (2019). MiR-132 promotes the proliferation, invasion and migration of human pancreatic carcinoma by inhibition of the tumor suppressor gene PTEN. Progress in Biophysics and Molecular Biology, 148, 65–72.PubMed
202.
go back to reference Zhang, H., et al. (2019). Long non-coding RNA CASC2 upregulates PTEN to suppress pancreatic carcinoma cell metastasis by downregulating miR-21. Cancer Cell International, 19, 18.PubMedPubMedCentral Zhang, H., et al. (2019). Long non-coding RNA CASC2 upregulates PTEN to suppress pancreatic carcinoma cell metastasis by downregulating miR-21. Cancer Cell International, 19, 18.PubMedPubMedCentral
203.
go back to reference Wu, Y., et al. (2019). ITGA6 and RPSA synergistically promote pancreatic cancer invasion and metastasis via PI3K and MAPK signaling pathways. Experimental Cell Research, 379(1), 30–47.PubMed Wu, Y., et al. (2019). ITGA6 and RPSA synergistically promote pancreatic cancer invasion and metastasis via PI3K and MAPK signaling pathways. Experimental Cell Research, 379(1), 30–47.PubMed
204.
go back to reference Zhu, J. H., et al. (2020). MiR-139-5p/SLC7A11 inhibits the proliferation, invasion and metastasis of pancreatic carcinoma via PI3K/Akt signaling pathway. Biochimica et Biophysica Acta - Molecular Basis of Disease, 1866(6), 165747.PubMed Zhu, J. H., et al. (2020). MiR-139-5p/SLC7A11 inhibits the proliferation, invasion and metastasis of pancreatic carcinoma via PI3K/Akt signaling pathway. Biochimica et Biophysica Acta - Molecular Basis of Disease, 1866(6), 165747.PubMed
205.
go back to reference Li, W., et al. (2018). Curcumin inhibits superoxide dismutase-induced epithelial-to-mesenchymal transition via the PI3K/Akt/NF-κB pathway in pancreatic cancer cells. International Journal of Oncology, 52(5), 1593–1602.PubMed Li, W., et al. (2018). Curcumin inhibits superoxide dismutase-induced epithelial-to-mesenchymal transition via the PI3K/Akt/NF-κB pathway in pancreatic cancer cells. International Journal of Oncology, 52(5), 1593–1602.PubMed
206.
go back to reference Liu, X., et al. (2018). Phosphoglycerate Mutase 1 (PGAM1) Promotes pancreatic ductal adenocarcinoma (PDAC) Metastasis by acting as a novel downstream target of the PI3K/Akt/mTOR Pathway. Oncology Research, 26(7), 1123–1131.PubMedPubMedCentral Liu, X., et al. (2018). Phosphoglycerate Mutase 1 (PGAM1) Promotes pancreatic ductal adenocarcinoma (PDAC) Metastasis by acting as a novel downstream target of the PI3K/Akt/mTOR Pathway. Oncology Research, 26(7), 1123–1131.PubMedPubMedCentral
207.
go back to reference Deng, J., et al. (2021). N(6) -methyladenosine-mediated upregulation of WTAPP1 promotes WTAP translation and Wnt signaling to facilitate pancreatic cancer progression. Cancer Research, 81(20), 5268–5283.PubMedPubMedCentral Deng, J., et al. (2021). N(6) -methyladenosine-mediated upregulation of WTAPP1 promotes WTAP translation and Wnt signaling to facilitate pancreatic cancer progression. Cancer Research, 81(20), 5268–5283.PubMedPubMedCentral
208.
go back to reference Gokturk, F., Erkoc-Kaya, D., & Arikoglu, H. (2021). Juglone can inhibit angiogenesis and metastasis in pancreatic cancer cells by targeting Wnt/β-catenin signaling. Bratislavské Lekárske Listy, 122(2), 132–137.PubMed Gokturk, F., Erkoc-Kaya, D., & Arikoglu, H. (2021). Juglone can inhibit angiogenesis and metastasis in pancreatic cancer cells by targeting Wnt/β-catenin signaling. Bratislavské Lekárske Listy, 122(2), 132–137.PubMed
209.
go back to reference Chen, T., et al. (2020). Linc00261 inhibits metastasis and the WNT signaling pathway of pancreatic cancer by regulating a miR-552-5p/FOXO3 axis. Oncology Reports, 43(3), 930–942.PubMedPubMedCentral Chen, T., et al. (2020). Linc00261 inhibits metastasis and the WNT signaling pathway of pancreatic cancer by regulating a miR-552-5p/FOXO3 axis. Oncology Reports, 43(3), 930–942.PubMedPubMedCentral
210.
go back to reference Xu, D., et al. (2020). Cadherin 13 Inhibits Pancreatic Cancer Progression and Epithelial-mesenchymal Transition by Wnt/β-Catenin Signaling. Journal of Cancer, 11(8), 2101–2112.PubMedPubMedCentral Xu, D., et al. (2020). Cadherin 13 Inhibits Pancreatic Cancer Progression and Epithelial-mesenchymal Transition by Wnt/β-Catenin Signaling. Journal of Cancer, 11(8), 2101–2112.PubMedPubMedCentral
211.
go back to reference Nielsen, M. F., Mortensen, M. B., & Detlefsen, S. (2016). Key players in pancreatic cancer-stroma interaction: Cancer-associated fibroblasts, endothelial and inflammatory cells. World Journal of Gastroenterology, 22(9), 2678–2700.PubMedPubMedCentral Nielsen, M. F., Mortensen, M. B., & Detlefsen, S. (2016). Key players in pancreatic cancer-stroma interaction: Cancer-associated fibroblasts, endothelial and inflammatory cells. World Journal of Gastroenterology, 22(9), 2678–2700.PubMedPubMedCentral
212.
go back to reference Hosein, A. N., Brekken, R. A., & Maitra, A. (2020). Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nature Reviews. Gastroenterology & Hepatology, 17(8), 487–505. Hosein, A. N., Brekken, R. A., & Maitra, A. (2020). Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nature Reviews. Gastroenterology & Hepatology, 17(8), 487–505.
213.
go back to reference Veenstra, V. L., et al. (2018). ADAM12 is a circulating marker for stromal activation in pancreatic cancer and predicts response to chemotherapy. Oncogenesis, 7(11), 87.PubMedPubMedCentral Veenstra, V. L., et al. (2018). ADAM12 is a circulating marker for stromal activation in pancreatic cancer and predicts response to chemotherapy. Oncogenesis, 7(11), 87.PubMedPubMedCentral
214.
go back to reference Cannon, A., et al. (2018). Desmoplasia in pancreatic ductal adenocarcinoma: insight into pathological function and therapeutic potential. Genes & Cancer, 9(3-4), 78–86. Cannon, A., et al. (2018). Desmoplasia in pancreatic ductal adenocarcinoma: insight into pathological function and therapeutic potential. Genes & Cancer, 9(3-4), 78–86.
215.
go back to reference Schnittert, J., et al. (2019). Integrin α11 in pancreatic stellate cells regulates tumor stroma interaction in pancreatic cancer. The FASEB Journal, 33(5), 6609–6621.PubMed Schnittert, J., et al. (2019). Integrin α11 in pancreatic stellate cells regulates tumor stroma interaction in pancreatic cancer. The FASEB Journal, 33(5), 6609–6621.PubMed
216.
go back to reference Goehrig, D., et al. (2019). Stromal protein βig-h3 reprogrammes tumour microenvironment in pancreatic cancer. Gut, 68(4), 693–707.PubMed Goehrig, D., et al. (2019). Stromal protein βig-h3 reprogrammes tumour microenvironment in pancreatic cancer. Gut, 68(4), 693–707.PubMed
217.
go back to reference Awaji, M., & Singh, R. K. (2019). Cancer-associated fibroblasts' functional heterogeneity in pancreatic ductal adenocarcinoma. Cancers (Basel), 11(3), 290.PubMed Awaji, M., & Singh, R. K. (2019). Cancer-associated fibroblasts' functional heterogeneity in pancreatic ductal adenocarcinoma. Cancers (Basel), 11(3), 290.PubMed
218.
go back to reference Sun, Q., et al. (2018). The impact of cancer-associated fibroblasts on major hallmarks of pancreatic cancer. Theranostics, 8(18), 5072–5087.PubMedPubMedCentral Sun, Q., et al. (2018). The impact of cancer-associated fibroblasts on major hallmarks of pancreatic cancer. Theranostics, 8(18), 5072–5087.PubMedPubMedCentral
219.
go back to reference Kobayashi, H., et al. (2019). Cancer-associated fibroblasts in gastrointestinal cancer. Nature Reviews. Gastroenterology & Hepatology, 16(5), 282–295. Kobayashi, H., et al. (2019). Cancer-associated fibroblasts in gastrointestinal cancer. Nature Reviews. Gastroenterology & Hepatology, 16(5), 282–295.
220.
go back to reference Geleta, B., et al. (2022). Targeting Wnt/tenascin C-mediated cross talk between pancreatic cancer cells and stellate cells via activation of the metastasis suppressor NDRG1. The Journal of Biological Chemistry, 298(3), 101608.PubMedPubMedCentral Geleta, B., et al. (2022). Targeting Wnt/tenascin C-mediated cross talk between pancreatic cancer cells and stellate cells via activation of the metastasis suppressor NDRG1. The Journal of Biological Chemistry, 298(3), 101608.PubMedPubMedCentral
221.
go back to reference Liu, Q. Q., et al. (2016). Oridonin inhibits pancreatic cancer cell migration and epithelial-mesenchymal transition by suppressing Wnt/β-catenin signaling pathway. Cancer Cell International, 16, 57.PubMedPubMedCentral Liu, Q. Q., et al. (2016). Oridonin inhibits pancreatic cancer cell migration and epithelial-mesenchymal transition by suppressing Wnt/β-catenin signaling pathway. Cancer Cell International, 16, 57.PubMedPubMedCentral
222.
go back to reference Quan, M., et al. (2015). Merlin/NF2 suppresses pancreatic tumor growth and metastasis by attenuating the FOXM1-mediated Wnt/β-Catenin signaling. Cancer Research, 75(22), 4778–4789.PubMedPubMedCentral Quan, M., et al. (2015). Merlin/NF2 suppresses pancreatic tumor growth and metastasis by attenuating the FOXM1-mediated Wnt/β-Catenin signaling. Cancer Research, 75(22), 4778–4789.PubMedPubMedCentral
223.
go back to reference Li, F., Dai, L., & Niu, J. (2020). GPX2 silencing relieves epithelial-mesenchymal transition, invasion, and metastasis in pancreatic cancer by downregulating Wnt pathway. Journal of Cellular Physiology, 235(11), 7780–7790.PubMed Li, F., Dai, L., & Niu, J. (2020). GPX2 silencing relieves epithelial-mesenchymal transition, invasion, and metastasis in pancreatic cancer by downregulating Wnt pathway. Journal of Cellular Physiology, 235(11), 7780–7790.PubMed
224.
go back to reference Garg, B., et al. (2017). Modulation of post-translational modifications in β-catenin and LRP6 inhibits Wnt signaling pathway in pancreatic cancer. Cancer Letters, 388, 64–72.PubMed Garg, B., et al. (2017). Modulation of post-translational modifications in β-catenin and LRP6 inhibits Wnt signaling pathway in pancreatic cancer. Cancer Letters, 388, 64–72.PubMed
225.
go back to reference Jiang, H., et al. (2014). Activation of the Wnt pathway through Wnt2 promotes metastasis in pancreatic cancer. American Journal of Cancer Research, 4(5), 537–544.PubMedPubMedCentral Jiang, H., et al. (2014). Activation of the Wnt pathway through Wnt2 promotes metastasis in pancreatic cancer. American Journal of Cancer Research, 4(5), 537–544.PubMedPubMedCentral
226.
go back to reference Zhou, P., et al. (2019). NMIIA promotes tumor growth and metastasis by activating the Wnt/β-catenin signaling pathway and EMT in pancreatic cancer. Oncogene, 38(27), 5500–5515.PubMed Zhou, P., et al. (2019). NMIIA promotes tumor growth and metastasis by activating the Wnt/β-catenin signaling pathway and EMT in pancreatic cancer. Oncogene, 38(27), 5500–5515.PubMed
227.
go back to reference Wang, L., et al. (2009). Oncogenic function of ATDC in pancreatic cancer through Wnt pathway activation and beta-catenin stabilization. Cancer Cell, 15(3), 207–219.PubMedPubMedCentral Wang, L., et al. (2009). Oncogenic function of ATDC in pancreatic cancer through Wnt pathway activation and beta-catenin stabilization. Cancer Cell, 15(3), 207–219.PubMedPubMedCentral
228.
go back to reference Sun, Y., et al. (2019). Restoration of miRNA-148a in pancreatic cancer reduces invasion and metastasis by inhibiting the Wnt/β-catenin signaling pathway via downregulating maternally expressed gene-3. Experimental and Therapeutic Medicine, 17(1), 639–648.PubMed Sun, Y., et al. (2019). Restoration of miRNA-148a in pancreatic cancer reduces invasion and metastasis by inhibiting the Wnt/β-catenin signaling pathway via downregulating maternally expressed gene-3. Experimental and Therapeutic Medicine, 17(1), 639–648.PubMed
229.
go back to reference Zhang, Q., et al. (2017). Hypoxia-inducible factor-2α promotes tumor progression and has crosstalk with Wnt/β-catenin signaling in pancreatic cancer. Molecular Cancer, 16(1), 119.PubMedPubMedCentral Zhang, Q., et al. (2017). Hypoxia-inducible factor-2α promotes tumor progression and has crosstalk with Wnt/β-catenin signaling in pancreatic cancer. Molecular Cancer, 16(1), 119.PubMedPubMedCentral
230.
go back to reference Weng, Y. C., et al. (2019). Long non-coding RNA LINC01133 silencing exerts antioncogenic effect in pancreatic cancer through the methylation of DKK1 promoter and the activation of Wnt signaling pathway. Cancer Biology & Therapy, 20(3), 368–380. Weng, Y. C., et al. (2019). Long non-coding RNA LINC01133 silencing exerts antioncogenic effect in pancreatic cancer through the methylation of DKK1 promoter and the activation of Wnt signaling pathway. Cancer Biology & Therapy, 20(3), 368–380.
231.
go back to reference Wu, X., et al. (2019). LncRNA BANCR promotes pancreatic cancer tumorigenesis via modulating MiR-195-5p/Wnt/β-catenin signaling pathway. Technology in Cancer Research & Treatment, 18, 1533033819887962. Wu, X., et al. (2019). LncRNA BANCR promotes pancreatic cancer tumorigenesis via modulating MiR-195-5p/Wnt/β-catenin signaling pathway. Technology in Cancer Research & Treatment, 18, 1533033819887962.
232.
go back to reference Yang, Y., Bai, Y. S., & Wang, Q. (2017). CDGSH iron sulfur domain 2 activates proliferation and EMT of pancreatic cancer cells via Wnt/β-catenin pathway and has prognostic value in human pancreatic cancer. Oncology Research, 25(4), 605–615.PubMedPubMedCentral Yang, Y., Bai, Y. S., & Wang, Q. (2017). CDGSH iron sulfur domain 2 activates proliferation and EMT of pancreatic cancer cells via Wnt/β-catenin pathway and has prognostic value in human pancreatic cancer. Oncology Research, 25(4), 605–615.PubMedPubMedCentral
233.
go back to reference Kim, J. H., et al. (2022). Vitamin C suppresses pancreatic carcinogenesis through the inhibition of both glucose metabolism and Wnt signaling. International Journal of Molecular Sciences, 23(20), 12249.PubMedPubMedCentral Kim, J. H., et al. (2022). Vitamin C suppresses pancreatic carcinogenesis through the inhibition of both glucose metabolism and Wnt signaling. International Journal of Molecular Sciences, 23(20), 12249.PubMedPubMedCentral
234.
go back to reference Zhan, T., et al. (2020). miR-455-3p functions as a tumor suppressor by restraining Wnt/β-catenin signaling via TAZ in pancreatic cancer. Cancer Management and Research, 12, 1483–1492.PubMedPubMedCentral Zhan, T., et al. (2020). miR-455-3p functions as a tumor suppressor by restraining Wnt/β-catenin signaling via TAZ in pancreatic cancer. Cancer Management and Research, 12, 1483–1492.PubMedPubMedCentral
235.
go back to reference Yang, J., et al. (2019). Long noncoding RNA DLX6-AS1 promotes tumorigenesis by modulating miR-497-5p/FZD4/FZD6/Wnt/β-catenin pathway in pancreatic cancer. Cancer Management and Research, 11, 4209–4221.PubMedPubMedCentral Yang, J., et al. (2019). Long noncoding RNA DLX6-AS1 promotes tumorigenesis by modulating miR-497-5p/FZD4/FZD6/Wnt/β-catenin pathway in pancreatic cancer. Cancer Management and Research, 11, 4209–4221.PubMedPubMedCentral
236.
go back to reference Wang, Z., et al. (2008). Blockade of SDF-1/CXCR4 signalling inhibits pancreatic cancer progression in vitro via inactivation of canonical Wnt pathway. British Journal of Cancer, 99(10), 1695–1703.PubMedPubMedCentral Wang, Z., et al. (2008). Blockade of SDF-1/CXCR4 signalling inhibits pancreatic cancer progression in vitro via inactivation of canonical Wnt pathway. British Journal of Cancer, 99(10), 1695–1703.PubMedPubMedCentral
237.
go back to reference Douchi, D., et al. (2015). Silencing of LRRFIP1 reverses the epithelial-mesenchymal transition via inhibition of the Wnt/β-catenin signaling pathway. Cancer Letters, 365(1), 132–140.PubMed Douchi, D., et al. (2015). Silencing of LRRFIP1 reverses the epithelial-mesenchymal transition via inhibition of the Wnt/β-catenin signaling pathway. Cancer Letters, 365(1), 132–140.PubMed
238.
go back to reference Ashrafizadeh, M., et al. (2022). Exosomes as promising nanostructures in diabetes mellitus: From insulin sensitivity to ameliorating diabetic complications. International Journal of Nanomedicine, 17, 1229–1253.PubMedPubMedCentral Ashrafizadeh, M., et al. (2022). Exosomes as promising nanostructures in diabetes mellitus: From insulin sensitivity to ameliorating diabetic complications. International Journal of Nanomedicine, 17, 1229–1253.PubMedPubMedCentral
239.
go back to reference Paskeh, M. D. A., et al. (2022). Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. Journal of Hematology & Oncology, 15(1), 1–39. Paskeh, M. D. A., et al. (2022). Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. Journal of Hematology & Oncology, 15(1), 1–39.
240.
go back to reference Li, J., et al. (2018). Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. Journal of Experimental & Clinical Cancer Research, 37(1), 177. Li, J., et al. (2018). Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. Journal of Experimental & Clinical Cancer Research, 37(1), 177.
241.
go back to reference Yu, Z., et al. (2017). Pancreatic cancer-derived exosomes promote tumor metastasis and liver pre-metastatic niche formation. Oncotarget, 8(38), 63461–63483.PubMedPubMedCentral Yu, Z., et al. (2017). Pancreatic cancer-derived exosomes promote tumor metastasis and liver pre-metastatic niche formation. Oncotarget, 8(38), 63461–63483.PubMedPubMedCentral
242.
go back to reference Tang, P., et al. (2019). Serum derived exosomes from pancreatic cancer patients promoted metastasis: An iTRAQ-based proteomic analysis. Oncotargets and Therapy, 12, 9329–9339.PubMedPubMedCentral Tang, P., et al. (2019). Serum derived exosomes from pancreatic cancer patients promoted metastasis: An iTRAQ-based proteomic analysis. Oncotargets and Therapy, 12, 9329–9339.PubMedPubMedCentral
243.
go back to reference Ogawa, K., et al. (2020). Prometastatic secretome trafficking via exosomes initiates pancreatic cancer pulmonary metastasis. Cancer Letters, 481, 63–75.PubMedPubMedCentral Ogawa, K., et al. (2020). Prometastatic secretome trafficking via exosomes initiates pancreatic cancer pulmonary metastasis. Cancer Letters, 481, 63–75.PubMedPubMedCentral
244.
go back to reference Kimoto, A., et al. (2023). Exosomes in ascites from patients with human pancreatic cancer enhance remote metastasis partially through endothelial-mesenchymal transition. Pancreatology, 23(4), 377–388.PubMed Kimoto, A., et al. (2023). Exosomes in ascites from patients with human pancreatic cancer enhance remote metastasis partially through endothelial-mesenchymal transition. Pancreatology, 23(4), 377–388.PubMed
245.
go back to reference Zhou, X., et al. (2022). Pancreatic cancer cell-derived exosomes promote lymphangiogenesis by downregulating ABHD11-AS1 expression. Cancers (Basel), 14(19), 4612.PubMed Zhou, X., et al. (2022). Pancreatic cancer cell-derived exosomes promote lymphangiogenesis by downregulating ABHD11-AS1 expression. Cancers (Basel), 14(19), 4612.PubMed
246.
go back to reference Xu, Y., et al. (2020). Anticancer effects of miR-124 delivered by BM-MSC derived exosomes on cell proliferation, epithelial mesenchymal transition, and chemotherapy sensitivity of pancreatic cancer cells. Aging (Albany NY), 12(19), 19660–19676.PubMed Xu, Y., et al. (2020). Anticancer effects of miR-124 delivered by BM-MSC derived exosomes on cell proliferation, epithelial mesenchymal transition, and chemotherapy sensitivity of pancreatic cancer cells. Aging (Albany NY), 12(19), 19660–19676.PubMed
247.
go back to reference Wei, Q., et al. (2020). EphA2-enriched exosomes promote cell migration and are a potential diagnostic serum marker in pancreatic cancer. Molecular Medicine Reports, 22(4), 2941–2947.PubMedPubMedCentral Wei, Q., et al. (2020). EphA2-enriched exosomes promote cell migration and are a potential diagnostic serum marker in pancreatic cancer. Molecular Medicine Reports, 22(4), 2941–2947.PubMedPubMedCentral
248.
go back to reference Zhang, Y. F., et al. (2019). Pancreatic cancer-derived exosomes promoted pancreatic stellate cells recruitment by pancreatic cancer. Journal of Cancer, 10(18), 4397–4407.PubMedPubMedCentral Zhang, Y. F., et al. (2019). Pancreatic cancer-derived exosomes promoted pancreatic stellate cells recruitment by pancreatic cancer. Journal of Cancer, 10(18), 4397–4407.PubMedPubMedCentral
249.
go back to reference Nakayama, F., et al. (2022). Pancreatic cancer cell-derived exosomes induce epithelial-mesenchymal transition in human pancreatic cancer cells themselves partially via transforming growth factor β1. Medical Molecular Morphology, 55(3), 227–235.PubMedPubMedCentral Nakayama, F., et al. (2022). Pancreatic cancer cell-derived exosomes induce epithelial-mesenchymal transition in human pancreatic cancer cells themselves partially via transforming growth factor β1. Medical Molecular Morphology, 55(3), 227–235.PubMedPubMedCentral
250.
go back to reference Wang, X., et al. (2018). Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kγ to promote pancreatic cancer metastasis. Cancer Research, 78(16), 4586–4598.PubMed Wang, X., et al. (2018). Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kγ to promote pancreatic cancer metastasis. Cancer Research, 78(16), 4586–4598.PubMed
251.
go back to reference Chen, K., et al. (2022). Hypoxic pancreatic cancer derived exosomal miR-30b-5p promotes tumor angiogenesis by inhibiting GJA1 expression. International Journal of Biological Sciences, 18(3), 1220–1237.PubMedPubMedCentral Chen, K., et al. (2022). Hypoxic pancreatic cancer derived exosomal miR-30b-5p promotes tumor angiogenesis by inhibiting GJA1 expression. International Journal of Biological Sciences, 18(3), 1220–1237.PubMedPubMedCentral
252.
go back to reference Li, M., et al. (2022). Exosomal miR-485-3p derived from pancreatic ductal epithelial cells inhibits pancreatic cancer metastasis through targeting PAK1. Chinese Medical Journal, 135(19), 2326–2337.PubMedPubMedCentral Li, M., et al. (2022). Exosomal miR-485-3p derived from pancreatic ductal epithelial cells inhibits pancreatic cancer metastasis through targeting PAK1. Chinese Medical Journal, 135(19), 2326–2337.PubMedPubMedCentral
253.
go back to reference Cao, W., et al. (2021). Hypoxic pancreatic stellate cell-derived exosomal mirnas promote proliferation and invasion of pancreatic cancer through the PTEN/AKT pathway. Aging (Albany NY), 13(5), 7120–7132.PubMed Cao, W., et al. (2021). Hypoxic pancreatic stellate cell-derived exosomal mirnas promote proliferation and invasion of pancreatic cancer through the PTEN/AKT pathway. Aging (Albany NY), 13(5), 7120–7132.PubMed
254.
go back to reference Li, Z., et al. (2018). Tumor-secreted exosomal miR-222 promotes tumor progression via regulating P27 expression and re-localization in pancreatic cancer. Cellular Physiology and Biochemistry, 51(2), 610–629.PubMed Li, Z., et al. (2018). Tumor-secreted exosomal miR-222 promotes tumor progression via regulating P27 expression and re-localization in pancreatic cancer. Cellular Physiology and Biochemistry, 51(2), 610–629.PubMed
255.
go back to reference Sun, Z., et al. (2021). Exosomal linc-ROR mediates crosstalk between cancer cells and adipocytes to promote tumor growth in pancreatic cancer. Molecular Therapy--Nucleic Acids, 26, 253–268.PubMedPubMedCentral Sun, Z., et al. (2021). Exosomal linc-ROR mediates crosstalk between cancer cells and adipocytes to promote tumor growth in pancreatic cancer. Molecular Therapy--Nucleic Acids, 26, 253–268.PubMedPubMedCentral
256.
go back to reference Wu, M., et al. (2020). Role of exosomal microRNA-125b-5p in conferring the metastatic phenotype among pancreatic cancer cells with different potential of metastasis. Life Sciences, 255, 117857.PubMed Wu, M., et al. (2020). Role of exosomal microRNA-125b-5p in conferring the metastatic phenotype among pancreatic cancer cells with different potential of metastasis. Life Sciences, 255, 117857.PubMed
257.
go back to reference Xie, Z., et al. (2022). Exosome-delivered CD44v6/C1QBP complex drives pancreatic cancer liver metastasis by promoting fibrotic liver microenvironment. Gut, 71(3), 568–579.PubMed Xie, Z., et al. (2022). Exosome-delivered CD44v6/C1QBP complex drives pancreatic cancer liver metastasis by promoting fibrotic liver microenvironment. Gut, 71(3), 568–579.PubMed
258.
go back to reference He, Z., et al. (2022). Exosome-derived FGD5-AS1 promotes tumor-associated macrophage M2 polarization-mediated pancreatic cancer cell proliferation and metastasis. Cancer Letters, 548, 215751.PubMed He, Z., et al. (2022). Exosome-derived FGD5-AS1 promotes tumor-associated macrophage M2 polarization-mediated pancreatic cancer cell proliferation and metastasis. Cancer Letters, 548, 215751.PubMed
259.
go back to reference Costa-Silva, B., et al. (2015). Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nature Cell Biology, 17(6), 816–826.PubMedPubMedCentral Costa-Silva, B., et al. (2015). Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nature Cell Biology, 17(6), 816–826.PubMedPubMedCentral
260.
go back to reference Williams, C. B., Yeh, E. S., & Soloff, A. C. (2016). Tumor-associated macrophages: unwitting accomplices in breast cancer malignancy. NPJ Breast Cancer, 2, 15025.PubMedPubMedCentral Williams, C. B., Yeh, E. S., & Soloff, A. C. (2016). Tumor-associated macrophages: unwitting accomplices in breast cancer malignancy. NPJ Breast Cancer, 2, 15025.PubMedPubMedCentral
261.
go back to reference Ye, H., et al. (2018). Tumor-associated macrophages promote progression and the Warburg effect via CCL18/NF-kB/VCAM-1 pathway in pancreatic ductal adenocarcinoma. Cell Death & Disease, 9(5), 453. Ye, H., et al. (2018). Tumor-associated macrophages promote progression and the Warburg effect via CCL18/NF-kB/VCAM-1 pathway in pancreatic ductal adenocarcinoma. Cell Death & Disease, 9(5), 453.
262.
go back to reference Meng, F., et al. (2015). CCL18 promotes epithelial-mesenchymal transition, invasion and migration of pancreatic cancer cells in pancreatic ductal adenocarcinoma. International Journal of Oncology, 46(3), 1109–1120.PubMed Meng, F., et al. (2015). CCL18 promotes epithelial-mesenchymal transition, invasion and migration of pancreatic cancer cells in pancreatic ductal adenocarcinoma. International Journal of Oncology, 46(3), 1109–1120.PubMed
263.
go back to reference Quail, D. F., & Joyce, J. A. (2013). Microenvironmental regulation of tumor progression and metastasis. Nature Medicine, 19(11), 1423–1437.PubMedPubMedCentral Quail, D. F., & Joyce, J. A. (2013). Microenvironmental regulation of tumor progression and metastasis. Nature Medicine, 19(11), 1423–1437.PubMedPubMedCentral
264.
go back to reference Chávez-Galán, L., et al. (2015). Much More than M1 and M2 Macrophages, There are also CD169(+) and TCR(+) Macrophages. Frontiers in Immunology, 6, 263.PubMedPubMedCentral Chávez-Galán, L., et al. (2015). Much More than M1 and M2 Macrophages, There are also CD169(+) and TCR(+) Macrophages. Frontiers in Immunology, 6, 263.PubMedPubMedCentral
265.
go back to reference Tarique, A. A., et al. (2015). Phenotypic, functional, and plasticity features of classical and alternatively activated human macrophages. American Journal of Respiratory Cell and Molecular Biology, 53(5), 676–688.PubMed Tarique, A. A., et al. (2015). Phenotypic, functional, and plasticity features of classical and alternatively activated human macrophages. American Journal of Respiratory Cell and Molecular Biology, 53(5), 676–688.PubMed
266.
go back to reference Helm, O., et al. (2014). M1 and M2: there is no "good" and "bad"-How macrophages promote malignancy-associated features in tumorigenesis. Oncoimmunology, 3(7), e946818.PubMedPubMedCentral Helm, O., et al. (2014). M1 and M2: there is no "good" and "bad"-How macrophages promote malignancy-associated features in tumorigenesis. Oncoimmunology, 3(7), e946818.PubMedPubMedCentral
267.
go back to reference Stöger, J. L., et al. (2012). Distribution of macrophage polarization markers in human atherosclerosis. Atherosclerosis, 225(2), 461–468.PubMed Stöger, J. L., et al. (2012). Distribution of macrophage polarization markers in human atherosclerosis. Atherosclerosis, 225(2), 461–468.PubMed
268.
go back to reference Genard, G., Lucas, S., & Michiels, C. (2017). reprogramming of tumor-associated macrophages with anticancer therapies: Radiotherapy versus chemo- and immunotherapies. Frontiers in Immunology, 8, 828.PubMedPubMedCentral Genard, G., Lucas, S., & Michiels, C. (2017). reprogramming of tumor-associated macrophages with anticancer therapies: Radiotherapy versus chemo- and immunotherapies. Frontiers in Immunology, 8, 828.PubMedPubMedCentral
269.
go back to reference Chang, Y. T., et al. (2020). Pancreatic cancer-derived small extracellular vesical Ezrin regulates macrophage polarization and promotes metastasis. American Journal of Cancer Research, 10(1), 12–37.PubMedPubMedCentral Chang, Y. T., et al. (2020). Pancreatic cancer-derived small extracellular vesical Ezrin regulates macrophage polarization and promotes metastasis. American Journal of Cancer Research, 10(1), 12–37.PubMedPubMedCentral
270.
go back to reference Nielsen, S. R., et al. (2016). Macrophage-secreted granulin supports pancreatic cancer metastasis by inducing liver fibrosis. Nature Cell Biology, 18(5), 549–560.PubMedPubMedCentral Nielsen, S. R., et al. (2016). Macrophage-secreted granulin supports pancreatic cancer metastasis by inducing liver fibrosis. Nature Cell Biology, 18(5), 549–560.PubMedPubMedCentral
271.
go back to reference Liu, B., et al. (2016). Tumor-associated macrophage-derived CCL20 enhances the growth and metastasis of pancreatic cancer. Acta Biochimica et Biophysica Sinica Shanghai, 48(12), 1067–1074. Liu, B., et al. (2016). Tumor-associated macrophage-derived CCL20 enhances the growth and metastasis of pancreatic cancer. Acta Biochimica et Biophysica Sinica Shanghai, 48(12), 1067–1074.
272.
go back to reference Thibault, B., et al. (2021). Pancreatic cancer intrinsic PI3Kα activity accelerates metastasis and rewires macrophage component. EMBO Molecular Medicine, 13(7), e13502.PubMedPubMedCentral Thibault, B., et al. (2021). Pancreatic cancer intrinsic PI3Kα activity accelerates metastasis and rewires macrophage component. EMBO Molecular Medicine, 13(7), e13502.PubMedPubMedCentral
273.
go back to reference Griesmann, H., et al. (2017). Pharmacological macrophage inhibition decreases metastasis formation in a genetic model of pancreatic cancer. Gut, 66(7), 1278–1285.PubMed Griesmann, H., et al. (2017). Pharmacological macrophage inhibition decreases metastasis formation in a genetic model of pancreatic cancer. Gut, 66(7), 1278–1285.PubMed
274.
go back to reference Lin, L., et al. (2023). UTMD inhibits pancreatic cancer growth and metastasis by inducing macrophage polarization and vessel normalization. Biomedicine & Pharmacotherapy, 160, 114322. Lin, L., et al. (2023). UTMD inhibits pancreatic cancer growth and metastasis by inducing macrophage polarization and vessel normalization. Biomedicine & Pharmacotherapy, 160, 114322.
275.
go back to reference Novizio, N., et al. (2021). ANXA1 contained in EVs Regulates macrophage polarization in tumor microenvironment and promotes pancreatic cancer progression and metastasis. International Journal of Molecular Sciences, 22(20), 11018.PubMedPubMedCentral Novizio, N., et al. (2021). ANXA1 contained in EVs Regulates macrophage polarization in tumor microenvironment and promotes pancreatic cancer progression and metastasis. International Journal of Molecular Sciences, 22(20), 11018.PubMedPubMedCentral
276.
go back to reference Song, Y., et al. (2023). Circ_0018909 knockdown inhibits the development of pancreatic cancer via the miR-545-3p/FASN axis and reduces macrophage polarization to M2. Journal of Biochemical and Molecular Toxicology, 37(4), e23293.PubMed Song, Y., et al. (2023). Circ_0018909 knockdown inhibits the development of pancreatic cancer via the miR-545-3p/FASN axis and reduces macrophage polarization to M2. Journal of Biochemical and Molecular Toxicology, 37(4), e23293.PubMed
277.
go back to reference Kurahara, H., et al. (2013). M2-polarized tumor-associated macrophage infiltration of regional lymph nodes is associated with nodal lymphangiogenesis and occult nodal involvement in pN0 pancreatic cancer. Pancreas, 42(1), 155–159.PubMed Kurahara, H., et al. (2013). M2-polarized tumor-associated macrophage infiltration of regional lymph nodes is associated with nodal lymphangiogenesis and occult nodal involvement in pN0 pancreatic cancer. Pancreas, 42(1), 155–159.PubMed
278.
go back to reference Ma, X., et al. (2016). The pancreatic cancer secreted REG4 promotes macrophage polarization to M2 through EGFR/AKT/CREB pathway. Oncology Reports, 35(1), 189–196.PubMed Ma, X., et al. (2016). The pancreatic cancer secreted REG4 promotes macrophage polarization to M2 through EGFR/AKT/CREB pathway. Oncology Reports, 35(1), 189–196.PubMed
279.
go back to reference Geng, Y., et al. (2021). A notch-dependent inflammatory feedback circuit between macrophages and cancer cells regulates pancreatic cancer metastasis. Cancer Research, 81(1), 64–76.PubMed Geng, Y., et al. (2021). A notch-dependent inflammatory feedback circuit between macrophages and cancer cells regulates pancreatic cancer metastasis. Cancer Research, 81(1), 64–76.PubMed
280.
go back to reference Wu, J., et al. (2022). TNFSF9 promotes metastasis of pancreatic cancer by regulating M2 polarization of macrophages through Src/FAK/p-Akt/IL-1β signaling. International Immunopharmacology, 102, 108429.PubMed Wu, J., et al. (2022). TNFSF9 promotes metastasis of pancreatic cancer by regulating M2 polarization of macrophages through Src/FAK/p-Akt/IL-1β signaling. International Immunopharmacology, 102, 108429.PubMed
281.
go back to reference Chen, Q., et al. (2019). Tumour cell-derived debris and IgG synergistically promote metastasis of pancreatic cancer by inducing inflammation via tumour-associated macrophages. British Journal of Cancer, 121(9), 786–795.PubMedPubMedCentral Chen, Q., et al. (2019). Tumour cell-derived debris and IgG synergistically promote metastasis of pancreatic cancer by inducing inflammation via tumour-associated macrophages. British Journal of Cancer, 121(9), 786–795.PubMedPubMedCentral
282.
go back to reference Gu, H., et al. (2022). NLRP3 activation in tumor-associated macrophages enhances lung metastasis of pancreatic ductal adenocarcinoma. Translational Lung Cancer Research, 11(5), 858–868.PubMedPubMedCentral Gu, H., et al. (2022). NLRP3 activation in tumor-associated macrophages enhances lung metastasis of pancreatic ductal adenocarcinoma. Translational Lung Cancer Research, 11(5), 858–868.PubMedPubMedCentral
283.
go back to reference Menen, R. S., et al. (2012). Tumor-educated macrophages promote tumor growth and peritoneal metastasis in an orthotopic nude mouse model of human pancreatic cancer. In Vivo, 26(4), 565–569.PubMed Menen, R. S., et al. (2012). Tumor-educated macrophages promote tumor growth and peritoneal metastasis in an orthotopic nude mouse model of human pancreatic cancer. In Vivo, 26(4), 565–569.PubMed
284.
go back to reference Penny, H. L., et al. (2016). Warburg metabolism in tumor-conditioned macrophages promotes metastasis in human pancreatic ductal adenocarcinoma. Oncoimmunology, 5(8), e1191731.PubMedPubMedCentral Penny, H. L., et al. (2016). Warburg metabolism in tumor-conditioned macrophages promotes metastasis in human pancreatic ductal adenocarcinoma. Oncoimmunology, 5(8), e1191731.PubMedPubMedCentral
285.
go back to reference Liu, C. Y., et al. (2013). M2-polarized tumor-associated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Laboratory Investigation, 93(7), 844–854.PubMed Liu, C. Y., et al. (2013). M2-polarized tumor-associated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Laboratory Investigation, 93(7), 844–854.PubMed
286.
287.
go back to reference Niu, N., et al. (2023). Tumor Cell-Intrinsic SETD2 Deficiency reprograms neutrophils to foster immune escape in pancreatic tumorigenesis. Advanced Science(Weinh), 10(2), e2202937. Niu, N., et al. (2023). Tumor Cell-Intrinsic SETD2 Deficiency reprograms neutrophils to foster immune escape in pancreatic tumorigenesis. Advanced Science(Weinh), 10(2), e2202937.
288.
go back to reference Papayannopoulos, V. (2018). Neutrophil extracellular traps in immunity and disease. Nature Reviews. Immunology, 18(2), 134–147.PubMed Papayannopoulos, V. (2018). Neutrophil extracellular traps in immunity and disease. Nature Reviews. Immunology, 18(2), 134–147.PubMed
289.
go back to reference Cedervall, J., & Olsson, A. K. (2016). Immunity gone astray - NETs in cancer. Trends Cancer, 2(11), 633–634.PubMed Cedervall, J., & Olsson, A. K. (2016). Immunity gone astray - NETs in cancer. Trends Cancer, 2(11), 633–634.PubMed
290.
go back to reference Deng, J., et al. (2021). DDR1-induced neutrophil extracellular traps drive pancreatic cancer metastasis. JCI Insight, 6(17), e146133.PubMedPubMedCentral Deng, J., et al. (2021). DDR1-induced neutrophil extracellular traps drive pancreatic cancer metastasis. JCI Insight, 6(17), e146133.PubMedPubMedCentral
291.
go back to reference Kajioka, H., et al. (2021). Targeting neutrophil extracellular traps with thrombomodulin prevents pancreatic cancer metastasis. Cancer Letters, 497, 1–13.PubMed Kajioka, H., et al. (2021). Targeting neutrophil extracellular traps with thrombomodulin prevents pancreatic cancer metastasis. Cancer Letters, 497, 1–13.PubMed
292.
go back to reference Miller-Ocuin, J. L., et al. (2019). DNA released from neutrophil extracellular traps (NETs) activates pancreatic stellate cells and enhances pancreatic tumor growth. Oncoimmunology, 8(9), e1605822.PubMedPubMedCentral Miller-Ocuin, J. L., et al. (2019). DNA released from neutrophil extracellular traps (NETs) activates pancreatic stellate cells and enhances pancreatic tumor growth. Oncoimmunology, 8(9), e1605822.PubMedPubMedCentral
293.
go back to reference Takesue, S., et al. (2020). Neutrophil extracellular traps promote liver micrometastasis in pancreatic ductal adenocarcinoma via the activation of cancer-associated fibroblasts. International Journal of Oncology, 56(2), 596–605.PubMed Takesue, S., et al. (2020). Neutrophil extracellular traps promote liver micrometastasis in pancreatic ductal adenocarcinoma via the activation of cancer-associated fibroblasts. International Journal of Oncology, 56(2), 596–605.PubMed
294.
go back to reference Bellomo, G., et al. (2022). Chemotherapy-induced infiltration of neutrophils promotes pancreatic cancer metastasis via Gas6/AXL signalling axis. Gut, 71(11), 2284–2299.PubMed Bellomo, G., et al. (2022). Chemotherapy-induced infiltration of neutrophils promotes pancreatic cancer metastasis via Gas6/AXL signalling axis. Gut, 71(11), 2284–2299.PubMed
295.
go back to reference Lianyuan, T., et al. (2020). Tumor associated neutrophils promote the metastasis of pancreatic ductal adenocarcinoma. Cancer Biology & Therapy, 21(10), 937–945. Lianyuan, T., et al. (2020). Tumor associated neutrophils promote the metastasis of pancreatic ductal adenocarcinoma. Cancer Biology & Therapy, 21(10), 937–945.
296.
go back to reference Li, Y. X., et al. (2022). ACLP promotes activation of cancer-associated fibroblasts and tumor metastasis via ACLP-PPARγ-ACLP feedback loop in pancreatic cancer. Cancer Letters, 544, 215802.PubMed Li, Y. X., et al. (2022). ACLP promotes activation of cancer-associated fibroblasts and tumor metastasis via ACLP-PPARγ-ACLP feedback loop in pancreatic cancer. Cancer Letters, 544, 215802.PubMed
297.
go back to reference Cai, W., et al. (2021). PERK-eIF2α-ERK1/2 axis drives mesenchymal-endothelial transition of cancer-associated fibroblasts in pancreatic cancer. Cancer Letters, 515, 86–95.PubMed Cai, W., et al. (2021). PERK-eIF2α-ERK1/2 axis drives mesenchymal-endothelial transition of cancer-associated fibroblasts in pancreatic cancer. Cancer Letters, 515, 86–95.PubMed
298.
go back to reference Zaghdoudi, S., et al. (2020). FAK activity in cancer-associated fibroblasts is a prognostic marker and a druggable key metastatic player in pancreatic cancer. EMBO Molecular Medicine, 12(11), e12010.PubMedPubMedCentral Zaghdoudi, S., et al. (2020). FAK activity in cancer-associated fibroblasts is a prognostic marker and a druggable key metastatic player in pancreatic cancer. EMBO Molecular Medicine, 12(11), e12010.PubMedPubMedCentral
299.
go back to reference Chu, X., Yang, Y., & Tian, X. (2022). Crosstalk between Pancreatic Cancer Cells and Cancer-Associated Fibroblasts in the Tumor Microenvironment Mediated by Exosomal MicroRNAs. International Journal of Molecular Sciences, 23(17), 9512.PubMedPubMedCentral Chu, X., Yang, Y., & Tian, X. (2022). Crosstalk between Pancreatic Cancer Cells and Cancer-Associated Fibroblasts in the Tumor Microenvironment Mediated by Exosomal MicroRNAs. International Journal of Molecular Sciences, 23(17), 9512.PubMedPubMedCentral
300.
go back to reference Stylianou, A., Gkretsi, V., & Stylianopoulos, T. (2018). Transforming growth factor-β modulates pancreatic cancer associated fibroblasts cell shape, stiffness and invasion. Biochimica et Biophysica Acta - General Subjects, 1862(7), 1537–1546.PubMed Stylianou, A., Gkretsi, V., & Stylianopoulos, T. (2018). Transforming growth factor-β modulates pancreatic cancer associated fibroblasts cell shape, stiffness and invasion. Biochimica et Biophysica Acta - General Subjects, 1862(7), 1537–1546.PubMed
301.
go back to reference Goicoechea, S. M., et al. (2014). Palladin promotes invasion of pancreatic cancer cells by enhancing invadopodia formation in cancer-associated fibroblasts. Oncogene, 33(10), 1265–1273.PubMed Goicoechea, S. M., et al. (2014). Palladin promotes invasion of pancreatic cancer cells by enhancing invadopodia formation in cancer-associated fibroblasts. Oncogene, 33(10), 1265–1273.PubMed
302.
go back to reference Shan, T., et al. (2017). Cancer-associated fibroblasts enhance pancreatic cancer cell invasion by remodeling the metabolic conversion mechanism. Oncology Reports, 37(4), 1971–1979.PubMedPubMedCentral Shan, T., et al. (2017). Cancer-associated fibroblasts enhance pancreatic cancer cell invasion by remodeling the metabolic conversion mechanism. Oncology Reports, 37(4), 1971–1979.PubMedPubMedCentral
303.
go back to reference Wang, Q., et al. (2017). Curcumin suppresses epithelial-to-mesenchymal transition and metastasis of pancreatic cancer cells by inhibiting cancer-associated fibroblasts. American Journal of Cancer Research, 7(1), 125–133.PubMedPubMedCentral Wang, Q., et al. (2017). Curcumin suppresses epithelial-to-mesenchymal transition and metastasis of pancreatic cancer cells by inhibiting cancer-associated fibroblasts. American Journal of Cancer Research, 7(1), 125–133.PubMedPubMedCentral
304.
go back to reference Chen, X., et al. (2021). CCL26 is upregulated by nab-paclitaxel in pancreatic cancer-associated fibroblasts and promotes PDAC invasiveness through activation of the PI3K/AKT/mTOR pathway. Acta Biochimica et Biophysica Sinica Shanghai, 53(5), 612–619. Chen, X., et al. (2021). CCL26 is upregulated by nab-paclitaxel in pancreatic cancer-associated fibroblasts and promotes PDAC invasiveness through activation of the PI3K/AKT/mTOR pathway. Acta Biochimica et Biophysica Sinica Shanghai, 53(5), 612–619.
305.
go back to reference Liu, J., et al. (2020). Cell metabolomics reveals berberine-inhibited pancreatic cancer cell viability and metastasis by regulating citrate metabolism. Journal of Proteome Research, 19(9), 3825–3836.PubMed Liu, J., et al. (2020). Cell metabolomics reveals berberine-inhibited pancreatic cancer cell viability and metastasis by regulating citrate metabolism. Journal of Proteome Research, 19(9), 3825–3836.PubMed
306.
go back to reference Tian, W., et al. (2022). Berberine suppresses lung metastasis of cancer via inhibiting endothelial transforming growth factor beta receptor 1. Frontiers in Pharmacology, 13, 917827.PubMedPubMedCentral Tian, W., et al. (2022). Berberine suppresses lung metastasis of cancer via inhibiting endothelial transforming growth factor beta receptor 1. Frontiers in Pharmacology, 13, 917827.PubMedPubMedCentral
307.
go back to reference Kim, S. O., & Kim, M. R. (2013). (-)-Epigallocatechin 3-gallate inhibits invasion by inducing the expression of Raf kinase inhibitor protein in AsPC-1 human pancreatic adenocarcinoma cells through the modulation of histone deacetylase activity. International Journal of Oncology, 42(1), 349–358.PubMed Kim, S. O., & Kim, M. R. (2013). (-)-Epigallocatechin 3-gallate inhibits invasion by inducing the expression of Raf kinase inhibitor protein in AsPC-1 human pancreatic adenocarcinoma cells through the modulation of histone deacetylase activity. International Journal of Oncology, 42(1), 349–358.PubMed
308.
go back to reference Arya, G., Das, M., & Sahoo, S. K. (2018). Evaluation of curcumin loaded chitosan/PEG blended PLGA nanoparticles for effective treatment of pancreatic cancer. Biomedicine & Pharmacotherapy, 102, 555–566. Arya, G., Das, M., & Sahoo, S. K. (2018). Evaluation of curcumin loaded chitosan/PEG blended PLGA nanoparticles for effective treatment of pancreatic cancer. Biomedicine & Pharmacotherapy, 102, 555–566.
309.
go back to reference Hoca, M., et al. (2020). The effect of resveratrol and quercetin on epithelial-mesenchymal transition in pancreatic cancer stem cell. Nutrition and Cancer, 72(7), 1231–1242.PubMed Hoca, M., et al. (2020). The effect of resveratrol and quercetin on epithelial-mesenchymal transition in pancreatic cancer stem cell. Nutrition and Cancer, 72(7), 1231–1242.PubMed
310.
go back to reference Li, W., et al. (2013). Resveratrol inhibits the epithelial-mesenchymal transition of pancreatic cancer cells via suppression of the PI-3K/Akt/NF-κB pathway. Current Medicinal Chemistry, 20(33), 4185–4194.PubMedPubMedCentral Li, W., et al. (2013). Resveratrol inhibits the epithelial-mesenchymal transition of pancreatic cancer cells via suppression of the PI-3K/Akt/NF-κB pathway. Current Medicinal Chemistry, 20(33), 4185–4194.PubMedPubMedCentral
311.
go back to reference Yu, D., et al. (2017). Quercetin inhibits epithelial-mesenchymal transition, decreases invasiveness and metastasis, and reverses IL-6 induced epithelial-mesenchymal transition, expression of MMP by inhibiting STAT3 signaling in pancreatic cancer cells. Oncotargets and Therapy, 10, 4719–4729.PubMedPubMedCentral Yu, D., et al. (2017). Quercetin inhibits epithelial-mesenchymal transition, decreases invasiveness and metastasis, and reverses IL-6 induced epithelial-mesenchymal transition, expression of MMP by inhibiting STAT3 signaling in pancreatic cancer cells. Oncotargets and Therapy, 10, 4719–4729.PubMedPubMedCentral
312.
go back to reference Guo, Y., et al. (2021). Quercetin suppresses pancreatic ductal adenocarcinoma progression via inhibition of SHH and TGF-β/Smad signaling pathways. Cell Biology and Toxicology, 37(3), 479–496.PubMed Guo, Y., et al. (2021). Quercetin suppresses pancreatic ductal adenocarcinoma progression via inhibition of SHH and TGF-β/Smad signaling pathways. Cell Biology and Toxicology, 37(3), 479–496.PubMed
313.
go back to reference Srivastava, R. K., et al. (2011). Sulforaphane synergizes with quercetin to inhibit self-renewal capacity of pancreatic cancer stem cells. Frontiers in Bioscience (Elite Edition), 3(2), 515–528.PubMed Srivastava, R. K., et al. (2011). Sulforaphane synergizes with quercetin to inhibit self-renewal capacity of pancreatic cancer stem cells. Frontiers in Bioscience (Elite Edition), 3(2), 515–528.PubMed
314.
go back to reference Lee, J., et al. (2016). Quercetin-3-O-glucoside suppresses pancreatic cancer cell migration induced by tumor-deteriorated growth factors in vitro. Oncology Reports, 35(4), 2473–2479.PubMed Lee, J., et al. (2016). Quercetin-3-O-glucoside suppresses pancreatic cancer cell migration induced by tumor-deteriorated growth factors in vitro. Oncology Reports, 35(4), 2473–2479.PubMed
315.
go back to reference Wang, C., et al. (2021). Metformin inhibits pancreatic cancer metastasis caused by SMAD4 deficiency and consequent HNF4G upregulation. Protein & Cell, 12(2), 128–144. Wang, C., et al. (2021). Metformin inhibits pancreatic cancer metastasis caused by SMAD4 deficiency and consequent HNF4G upregulation. Protein & Cell, 12(2), 128–144.
316.
go back to reference Yu, X., et al. (2020). Propofol affects the growth and metastasis of pancreatic cancer via ADAM8. Pharmacological Reports, 72(2), 418–426.PubMed Yu, X., et al. (2020). Propofol affects the growth and metastasis of pancreatic cancer via ADAM8. Pharmacological Reports, 72(2), 418–426.PubMed
317.
go back to reference Polireddy, K., et al. (2017). High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study. Scientific Reports, 7(1), 17188.PubMedPubMedCentral Polireddy, K., et al. (2017). High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study. Scientific Reports, 7(1), 17188.PubMedPubMedCentral
318.
go back to reference Yamada, S., et al. (2020). Phase I/II study of adding intraperitoneal paclitaxel in patients with pancreatic cancer and peritoneal metastasis. The British Journal of Surgery, 107(13), 1811–1817.PubMed Yamada, S., et al. (2020). Phase I/II study of adding intraperitoneal paclitaxel in patients with pancreatic cancer and peritoneal metastasis. The British Journal of Surgery, 107(13), 1811–1817.PubMed
319.
go back to reference Takahara, N., et al. (2021). A phase I study of intraperitoneal paclitaxel combined with gemcitabine plus nab-paclitaxel for pancreatic cancer with peritoneal metastasis. Investigational New Drugs, 39(1), 175–181.PubMed Takahara, N., et al. (2021). A phase I study of intraperitoneal paclitaxel combined with gemcitabine plus nab-paclitaxel for pancreatic cancer with peritoneal metastasis. Investigational New Drugs, 39(1), 175–181.PubMed
320.
go back to reference Nishiyama, Y., et al. (2005). Contribution of whole body FDG-PET to the detection of distant metastasis in pancreatic cancer. Annals of Nuclear Medicine, 19(6), 491–497.PubMed Nishiyama, Y., et al. (2005). Contribution of whole body FDG-PET to the detection of distant metastasis in pancreatic cancer. Annals of Nuclear Medicine, 19(6), 491–497.PubMed
321.
go back to reference Ishikawa, T., et al. (2007). Angiotensin-II administration is useful for the detection of liver metastasis from pancreatic cancer during pharmacoangiographic computed tomography. World Journal of Gastroenterology, 13(22), 3080–3083.PubMedPubMedCentral Ishikawa, T., et al. (2007). Angiotensin-II administration is useful for the detection of liver metastasis from pancreatic cancer during pharmacoangiographic computed tomography. World Journal of Gastroenterology, 13(22), 3080–3083.PubMedPubMedCentral
322.
go back to reference Hayashibe, A., et al. (2007). Clinical results on intra-arterial adjuvant chemotherapy for prevention of liver metastasis following curative resection of pancreatic cancer. Annals of Surgical Oncology, 14(1), 190–194.PubMed Hayashibe, A., et al. (2007). Clinical results on intra-arterial adjuvant chemotherapy for prevention of liver metastasis following curative resection of pancreatic cancer. Annals of Surgical Oncology, 14(1), 190–194.PubMed
323.
go back to reference Wobser, M., et al. (2006). Complete remission of liver metastasis of pancreatic cancer under vaccination with a HLA-A2 restricted peptide derived from the universal tumor antigen survivin. Cancer Immunology, Immunotherapy, 55(10), 1294–1298.PubMed Wobser, M., et al. (2006). Complete remission of liver metastasis of pancreatic cancer under vaccination with a HLA-A2 restricted peptide derived from the universal tumor antigen survivin. Cancer Immunology, Immunotherapy, 55(10), 1294–1298.PubMed
Metadata
Title
Molecular profile of metastasis, cell plasticity and EMT in pancreatic cancer: a pre-clinical connection to aggressiveness and drug resistance
Authors
Zhenli Guo
Milad Ashrafizadeh
Wei Zhang
Rongjun Zou
Gautam Sethi
Xianbin Zhang
Publication date
15-07-2023
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue 1/2024
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-023-10125-y

Other articles of this Issue 1/2024

Cancer and Metastasis Reviews 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine