Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches
Abstract
:Simple Summary
Abstract
1. Introduction
2. Desmoplastic Reaction in the Pancreatic Tumor Microenvironment
2.1. Desmoplasia: The Impact on Tumor Development and Progression
2.2. The Contribution of Desmoplastic Components towards Chemoresistance
2.3. Targeting the Desmoplasia Improves Chemotherapy Outcomes
3. The Role of Extracellular Acidic pH and Hypoxia in the Resistance to Therapy
3.1. Extracellular Acidic pH Driving Tumor Progression
3.2. Multidrug Resistance and the Acidic Tumor Microenvironment
3.3. Hypoxia: A Promoting Factor in Cancer Survival and Proliferation
3.4. Hypoxia: Induction of Chemoresistance in Cancer Cells
3.5. Therapeutic Strategies Targeting the Acidic Extracellular pH and Hypoxia
4. The Tumor Metabolic Microenvironment Promotes Resistance against Chemotherapy
4.1. Metabolic Rewiring and Nutrient Scavenging in Cancer Cells
4.2. Fuel Source Plasticity towards Resistance to Therapy
4.2.1. Glucose
4.2.2. Glutamine and Other Amino Acids
4.2.3. Lipids and Fatty Acids
4.3. Targeting Metabolism to Overwhelm Chemoresistance
4.3.1. Glucose Transporters (GLUT Family)
4.3.2. Hexokinase (HK)
4.3.3. Fructose Biphosphate Aldolase
4.3.4. Lactate Dehydrogenase (LDH)
4.3.5. Pyruvate Kinase (PK) and Monocarboxylate Transporters (MCTs)
4.3.6. Fatty Acid Transporter CD36
4.3.7. Fatty Acid Synthase (FASN) and Carnitine Palmitoyl Transferase 1A (CPT1A)
4.3.8. Clinical Trials: First Evidence in PDAC Patients
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 2008–2020. [Google Scholar] [CrossRef]
- McGuigan, A.; Kelly, P.; Turkington, R.C.; Jones, C.; Coleman, H.G.; McCain, R.S. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J. Gastroenterol. 2018, 24, 4846–4861. [Google Scholar] [CrossRef]
- Binkley, C.E.; Zhang, L.; Greenson, J.K.; Giordano, T.J.; Kuick, R.; Misek, D.; Hanash, S.; Logsdon, C.D.; Simeone, D.M. The molecular basis of pancreatic fibrosis: Common stromal gene expression in chronic pancreatitis and pancreatic adenocarcinoma. Pancreas 2004, 29, 254–263. [Google Scholar] [CrossRef] [PubMed]
- Yu, S.; Zhang, C.; Xie, K.P. Therapeutic resistance of pancreatic cancer: Roadmap to its reversal. Biochim. Biophys. Acta. Rev. Cancer 2021, 1875, 188461. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Schober, M.; Jesenofsky, R.; Faissner, R.; Weidenauer, C.; Hagmann, W.; Michl, P.; Heuchel, R.L.; Haas, S.L.; Löhr, J.M. Desmoplasia and chemoresistance in pancreatic cancer. Cancers 2014, 6, 2137–2154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stylianopoulos, T. The Solid Mechanics of Cancer and Strategies for Improved Therapy. J. Biomech. Eng. 2017, 139, 021004. [Google Scholar] [CrossRef] [PubMed]
- Goetz, J.G.; Minguet, S.; Navarro-Lérida, I.; Lazcano, J.J.; Samaniego, R.; Calvo, E.; Tello, M.; Osteso-Ibáñez, T.; Pellinen, T.; Echarri, A.; et al. Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis. Cell 2011, 146, 148–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Erdogan, B.; Ao, M.; White, L.M.; Means, A.L.; Brewer, B.M.; Yang, L.; Washington, M.K.; Shi, C.; Franco, O.E.; Weaver, A.M.; et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J. Cell Biol. 2017, 216, 3799–3816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stanisavljevic, J.; Loubat-Casanovas, J.; Herrera, M.; Luque, T.; Peña, R.; Lluch, A.; Albanell, J.; Bonilla, F.; Rovira, A.; Peña, C.; et al. Snail1-expressing fibroblasts in the tumor microenvironment display mechanical properties that support metastasis. Cancer Res. 2015, 75, 284–295. [Google Scholar] [CrossRef] [Green Version]
- Provenzano, P.P.; Cuevas, C.; Chang, A.E.; Goel, V.K.; Von Hoff, D.D.; Hingorani, S.R. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012, 21, 418–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adamska, A.; Domenichini, A.; Falasca, M. Pancreatic Ductal Adenocarcinoma: Current and Evolving Therapies. Int. J. Mol. Sci. 2017, 18, 1338. [Google Scholar] [CrossRef]
- Ioannides, C.G.; Whiteside, T.L. T cell recognition of human tumors: Implications for molecular immunotherapy of cancer. Clin. Immunol. Immunopathol. 1993, 66, 91–106. [Google Scholar] [CrossRef]
- Apte, M.V.; Xu, Z.; Pothula, S.; Goldstein, D.; Pirola, R.C.; Wilson, J.S. Pancreatic cancer: The microenvironment needs attention too! Pancreatology 2015, 15 (Suppl. S4), S32–S38. [Google Scholar] [CrossRef]
- Chang, J.H.; Jiang, Y.; Pillarisetty, V.G. Role of immune cells in pancreatic cancer from bench to clinical application: An updated review. Medicine 2016, 95, e5541. [Google Scholar] [CrossRef]
- Alfarouk, K.O.; Stock, C.-M.; Taylor, S.; Walsh, M.; Muddathir, A.K.; Verduzco, D.; Bashir, A.H.; Mohammed, O.Y.; Elhassan, G.O.; Harguindey, S. Resistance to cancer chemotherapy: Failure in drug response from ADME to P-gp. Cancer Cell Int. 2015, 15, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Yeldag, G.; Rice, A.; del Río Hernández, A. Chemoresistance and the self-maintaining tumor microenvironment. Cancers 2018, 10, 471. [Google Scholar] [CrossRef] [Green Version]
- Zheng, H.-C. The molecular mechanisms of chemoresistance in cancers. Oncotarget 2017, 8, 59950. [Google Scholar] [CrossRef] [Green Version]
- Waghray, M.; Yalamanchili, M.; di Magliano, M.P.; Simeone, D.M. Deciphering the role of stroma in pancreatic cancer. Curr. Opin. Gastroenterol. 2013, 29, 537. [Google Scholar] [CrossRef] [Green Version]
- Hall, B.R.; Cannon, A.; Atri, P.; Wichman, C.S.; Smith, L.M.; Ganti, A.K.; Are, C.; Sasson, A.R.; Kumar, S.; Batra, S.K. Advanced pancreatic cancer: A meta-analysis of clinical trials over thirty years. Oncotarget 2018, 9, 19396. [Google Scholar] [CrossRef] [Green Version]
- Apte, M.; Park, S.; Phillips, P.; Santucci, N.; Goldstein, D.; Kumar, R.; Ramm, G.; Buchler, M.; Friess, H.; McCarroll, J. Desmoplastic reaction in pancreatic cancer: Role of pancreatic stellate cells. Pancreas 2004, 29, 179–187. [Google Scholar] [CrossRef] [Green Version]
- Lu, J.; Zhou, S.; Siech, M.; Habisch, H.; Seufferlein, T.; Bachem, M. Pancreatic stellate cells promote hapto-migration of cancer cells through collagen I-mediated signalling pathway. Br. J. Cancer 2014, 110, 409–420. [Google Scholar] [CrossRef] [Green Version]
- Hwang, R.F.; Moore, T.; Arumugam, T.; Ramachandran, V.; Amos, K.D.; Rivera, A.; Ji, B.; Evans, D.B.; Logsdon, C.D. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008, 68, 918–926. [Google Scholar] [CrossRef] [Green Version]
- Dunér, S.; Lindman, J.L.; Ansari, D.; Gundewar, C.; Andersson, R. Pancreatic cancer: The role of pancreatic stellate cells in tumor progression. Pancreatology 2010, 10, 673–681. [Google Scholar] [CrossRef] [Green Version]
- Sherman, M.H.; Ruth, T.Y.; Engle, D.D.; Ding, N.; Atkins, A.R.; Tiriac, H.; Collisson, E.A.; Connor, F.; Van Dyke, T.; Kozlov, S. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 2014, 159, 80–93. [Google Scholar] [CrossRef] [Green Version]
- Carapuça, E.F.; Gemenetzidis, E.; Feig, C.; Bapiro, T.E.; Williams, M.D.; Wilson, A.S.; Delvecchio, F.R.; Arumugam, P.; Grose, R.P.; Lemoine, N.R. Anti-stromal treatment together with chemotherapy targets multiple signalling pathways in pancreatic adenocarcinoma. J. Pathol. 2016, 239, 286–296. [Google Scholar] [CrossRef] [Green Version]
- Ene-Obong, A.; Clear, A.J.; Watt, J.; Wang, J.; Fatah, R.; Riches, J.C.; Marshall, J.F.; Chin-Aleong, J.; Chelala, C.; Gribben, J.G. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology 2013, 145, 1121–1132. [Google Scholar] [CrossRef] [Green Version]
- Masamune, A.; Hamada, S.; Kikuta, K.; Takikawa, T.; Miura, S.; Nakano, E.; Shimosegawa, T. The angiotensin II type I receptor blocker olmesartan inhibits the growth of pancreatic cancer by targeting stellate cell activities in mice. Scand. J. Gastroenterol. 2013, 48, 602–609. [Google Scholar] [CrossRef]
- Whatcott, C.J.; Posner, R.G.; Von Hoff, D.D.; Han, H. Desmoplasia and chemoresistance in pancreatic cancer. In Pancreatic Cancer and Tumor Microenvironment; Transworld Research Network: Trivandrum, India, 2012. [Google Scholar]
- Yen, T.W.; Aardal, N.P.; Bronner, M.P.; Thorning, D.R.; Savard, C.E.; Lee, S.P.; Bell, R.H., Jr. Myofibroblasts are responsible for the desmoplastic reaction surrounding human pancreatic carcinomas. Surgery 2002, 131, 129–134. [Google Scholar] [CrossRef]
- Bachem, M.G.; Schneider, E.; Groß, H.; Weidenbach, H.; Schmid, R.M.; Menke, A.; Siech, M.; Beger, H.; Grünert, A.; Adler, G. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology 1998, 115, 421–432. [Google Scholar] [CrossRef]
- Apte, M.; Haber, P.; Darby, S.; Rodgers, S.; McCaughan, G.; Korsten, M.; Pirola, R.; Wilson, J. Pancreatic stellate cells are activated by proinflammatory cytokines: Implications for pancreatic fibrogenesis. Gut 1999, 44, 534–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aoyagi, Y.; Oda, T.; Kinoshita, T.; Nakahashi, C.; Hasebe, T.; Ohkohchi, N.; Ochiai, A. Overexpression of TGF-β by infiltrated granulocytes correlates with the expression of collagen mRNA in pancreatic cancer. Br. J. Cancer 2004, 91, 1316–1326. [Google Scholar] [CrossRef] [PubMed]
- Löhr, M.; Schmidt, C.; Ringel, J.; Kluth, M.; Müller, P.; Nizze, H.; Jesnowski, R. Transforming growth factor-β1 induces desmoplasia in an experimental model of human pancreatic carcinoma. Cancer Res. 2001, 61, 550–555. [Google Scholar]
- Awaji, M.; Futakuchi, M.; Heavican, T.; Iqbal, J.; Singh, R.K. Cancer-associated fibroblasts enhance survival and progression of the aggressive pancreatic tumor Via FGF-2 and CXCL8. Cancer Microenviron. 2019, 12, 37–46. [Google Scholar] [CrossRef]
- Hartel, M.; di Mola, F.F.; Gardini, A.; Zimmermann, A.; Di Sebastiano, P.; Guweidhi, A.; Innocenti, P.; Giese, T.; Giese, N.; Büchler, M.W. Desmoplastic reaction influences pancreatic cancer growth behavior. World J. Surg. 2004, 28, 818–825. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Shapiro, B.; Vucic, E.A.; Vogt, S.; Bar-Sagi, D. Tumor Cell-Derived IL-1β Promotes Desmoplasia and Immune Suppression in Pancreatic Cancer. Cancer Res. 2020, 80, 1088–1101. [Google Scholar] [CrossRef] [Green Version]
- Stern, R. Hyaluronidases in cancer biology. Hyaluronan Cancer Biol. 2008, 207–220. [Google Scholar] [CrossRef]
- Marastoni, S.; Ligresti, G.; Lorenzon, E.; Colombatti, A.; Mongiat, M. Extracellular matrix: A matter of life and death. Connect. Tissue Res. 2008, 49, 203–206. [Google Scholar] [CrossRef]
- Kaspar, M.; Zardi, L.; Neri, D. Fibronectin as target for tumor therapy. Int. J. Cancer 2006, 118, 1331–1339. [Google Scholar] [CrossRef]
- Mahadevan, D.; Von Hoff, D.D. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol. Cancer Ther. 2007, 6, 1186–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. The extracellular matrix of animals. In Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002. [Google Scholar]
- Imamura, T.; Iguchi, H.; Manabe, T.; Ohshio, G.; Yoshimura, T.; Wang, Z.; Suwa, H.; Ishigami, S.; Imamura, M. Quantitative analysis of collagen and collagen subtypes I, III, and V in human pancreatic cancer, tumor-associated chronic pancreatitis, and alcoholic chronic pancreatitis. Pancreas 1995, 11, 357–364. [Google Scholar] [CrossRef] [PubMed]
- Mollenhauer, J.; Roether, I.; Kern, H. Distribution of extracellular matrix proteins in pancreatic ductal adenocarcinoma and its influence on tumor cell proliferation in vitro. Pancreas 1987, 2, 14–24. [Google Scholar] [CrossRef] [PubMed]
- Linder, S.; Castaños-Velez, E.; Biberfeld, P. Immunohistochemical expression of extracellular matrix proteins and adhesion molecules in pancreatic carcinoma. Hepatogastroenterology 2001, 48, 1321–1327. [Google Scholar]
- Verrecchia, F.; Mauviel, A. TGF-β and TNF-α: Antagonistic cytokines controlling type I collagen gene expression. Cell. Signal. 2004, 16, 873–880. [Google Scholar] [CrossRef]
- Nieskoski, M.D.; Marra, K.; Gunn, J.R.; Hoopes, P.J.; Doyley, M.M.; Hasan, T.; Trembly, B.S.; Pogue, B.W. Collagen complexity spatially defines microregions of total tissue pressure in pancreatic cancer. Sci. Rep. 2017, 7, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Laurent, T.C.; Fraser, J. Hyaluronan. FASEB J. 1992, 6, 2397–2404. [Google Scholar] [CrossRef]
- Yu, M.; Tannock, I.F. Targeting tumor architecture to favor drug penetration: A new weapon to combat chemoresistance in pancreatic cancer? Cancer Cell 2012, 21, 327–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Provenzano, P.; Hingorani, S. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br. J. Cancer 2013, 108, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Johnsson, C.; Hällgren, R.; Tufveson, G. Role of hyaluronan in acute pancreatitis. Surgery 2000, 127, 650–658. [Google Scholar] [CrossRef]
- Voutouri, C.; Polydorou, C.; Papageorgis, P.; Gkretsi, V.; Stylianopoulos, T. Hyaluronan-derived swelling of solid tumors, the contribution of collagen and cancer cells, and implications for cancer therapy. Neoplasia 2016, 18, 732–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kultti, A.; Zhao, C.; Singha, N.C.; Zimmerman, S.; Osgood, R.J.; Symons, R.; Jiang, P.; Li, X.; Thompson, C.B.; Infante, J.R. Accumulation of extracellular hyaluronan by hyaluronan synthase 3 promotes tumor growth and modulates the pancreatic cancer microenvironment. BioMed Res. Int. 2014, 2014, 817613. [Google Scholar] [CrossRef] [Green Version]
- Jacobetz, M.A.; Chan, D.S.; Neesse, A.; Bapiro, T.E.; Cook, N.; Frese, K.K.; Feig, C.; Nakagawa, T.; Caldwell, M.E.; Zecchini, H.I. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013, 62, 112–120. [Google Scholar] [CrossRef]
- Akiyama, S.K.; Olden, K.; Yamada, K.M. Fibronectin and integrins in invasion and metastasis. Cancer Metastasis Rev. 1995, 14, 173–189. [Google Scholar] [CrossRef]
- Lenselink, E.A. Role of fibronectin in normal wound healing. Int. Wound J. 2015, 12, 313–316. [Google Scholar] [CrossRef] [PubMed]
- George, E.L.; Georges-Labouesse, E.N.; Patel-King, R.S.; Rayburn, H.; Hynes, R.O. Defects in mesoderm, neural tube and vascular development in mouse embryos lacking fibronectin. Development 1993, 119, 1079–1091. [Google Scholar] [CrossRef]
- Pankov, R.; Yamada, K.M. Fibronectin at a glance. J. Cell Sci. 2002, 115, 3861–3863. [Google Scholar] [CrossRef] [Green Version]
- To, W.S.; Midwood, K.S. Plasma and cellular fibronectin: Distinct and independent functions during tissue repair. Fibrogenes. Tissue Repair 2011, 4, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Midwood, K.S.; Williams, L.V.; Schwarzbauer, J.E. Tissue repair and the dynamics of the extracellular matrix. Int. J. Biochem. Cell Biol. 2004, 36, 1031–1037. [Google Scholar] [CrossRef]
- Ramaswamy, S.; Ross, K.N.; Lander, E.S.; Golub, T.R. A molecular signature of metastasis in primary solid tumors. Nat. Genet. 2003, 33, 49–54. [Google Scholar] [CrossRef]
- Forciniti, S.; Dalla Pozza, E.; Greco, M.R.; Amaral Carvalho, T.M.; Rolando, B.; Ambrosini, G.; Carmona-Carmona, C.A.; Pacchiana, R.; Di Molfetta, D.; Donadelli, M. Extracellular Matrix Composition Modulates the Responsiveness of Differentiated and Stem Pancreatic Cancer Cells to Lipophilic Derivate of Gemcitabine. Int. J. Mol. Sci. 2021, 22, 29. [Google Scholar] [CrossRef] [PubMed]
- Chand, S.N.; Zarei, M.; Schiewer, M.J.; Kamath, A.R.; Romeo, C.; Lal, S.; Cozzitorto, J.A.; Nevler, A.; Scolaro, L.; Londin, E. Posttranscriptional regulation of PARG mRNA by HuR facilitates DNA repair and resistance to PARP inhibitors. Cancer Res. 2017, 77, 5011–5025. [Google Scholar] [CrossRef] [Green Version]
- Perkhofer, L.; Gout, J.; Roger, E.; de Almeida, F.K.; Simões, C.B.; Wiesmüller, L.; Seufferlein, T.; Kleger, A. DNA damage repair as a target in pancreatic cancer: State-of-the-art and future perspectives. Gut 2021, 70, 606–617. [Google Scholar] [CrossRef] [PubMed]
- Hong, S.P.; Wen, J.; Bang, S.; Park, S.; Song, S.Y. CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int. J. Cancer 2009, 125, 2323–2331. [Google Scholar] [CrossRef]
- Fujita, Y.; Kitagawa, M.; Nakamura, S.; Azuma, K.; Ishii, G.; Higashi, M.; Kishi, H.; Hiwasa, T.; Koda, K.; Nakajima, N. CD44 signaling through focal adhesion kinase and its anti-apoptotic effect. FEBS Lett. 2002, 528, 101–108. [Google Scholar] [CrossRef] [Green Version]
- Mitsiades, C.S.; Mitsiades, N.; Koutsilieris, M. The Akt pathway: Molecular targets for anti-cancer drug development. Curr. Cancer Drug Targets 2004, 4, 235–256. [Google Scholar] [CrossRef]
- Henke, E.; Nandigama, R.; Ergün, S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front. Mol. Biosci. 2020, 6, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Diop-Frimpong, B.; Chauhan, V.P.; Krane, S.; Boucher, Y.; Jain, R.K. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc. Natl. Acad. Sci. USA 2011, 108, 2909–2914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Biondani, G.; Zeeberg, K.; Greco, M.R.; Cannone, S.; Dando, I.; Dalla Pozza, E.; Mastrodonato, M.; Forciniti, S.; Casavola, V.; Palmieri, M. Extracellular matrix composition modulates PDAC parenchymal and stem cell plasticity and behavior through the secretome. FEBS J. 2018, 285, 2104–2124. [Google Scholar] [CrossRef] [Green Version]
- Netti, P.A.; Berk, D.A.; Swartz, M.A.; Grodzinsky, A.J.; Jain, R.K. Role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer Res. 2000, 60, 2497–2503. [Google Scholar]
- Lieleg, O.; Baumgärtel, R.M.; Bausch, A.R. Selective filtering of particles by the extracellular matrix: An electrostatic bandpass. Biophys. J. 2009, 97, 1569–1577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thompson, C.B.; Shepard, H.M.; O’Connor, P.M.; Kadhim, S.; Jiang, P.; Osgood, R.J.; Bookbinder, L.H.; Li, X.; Sugarman, B.J.; Connor, R.J. Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models. Mol. Cancer Ther. 2010, 9, 3052–3064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Clark, D.A.; Coker, R. Transforming growth factor-beta (TGF-beta). Int. J. Biochem. Cell Biol. 1998, 30, 293–298. [Google Scholar] [CrossRef]
- Hilbig, A.; Oettle, H. Transforming growth factor beta in pancreatic cancer. Curr. Pharm. Biotechnol. 2011, 12, 2158–2164. [Google Scholar] [CrossRef] [PubMed]
- García-Montero, A.C.; Vasseur, S.; Giono, L.E.; Canepa, E.; Moreno, S.; Dagorn, J.C.; Iovanna, J.L. Transforming growth factor β-1 enhances Smad transcriptional activity through activation of p8 gene expression. Biochem. J. 2001, 357, 249–253. [Google Scholar] [CrossRef] [PubMed]
- Shields, M.A.; Dangi-Garimella, S.; Krantz, S.B.; Bentrem, D.J.; Munshi, H.G. Pancreatic cancer cells respond to type I collagen by inducing snail expression to promote membrane type 1 matrix metalloproteinase-dependent collagen invasion. J. Biol. Chem. 2011, 286, 10495–10504. [Google Scholar] [CrossRef] [Green Version]
- Kim, Y.J.; Hwang, J.S.; Hong, Y.B.; Bae, I.; Seong, Y.-S. Transforming growth factor beta receptor I inhibitor sensitizes drug-resistant pancreatic cancer cells to gemcitabine. Anticancer Res. 2012, 32, 799–806. [Google Scholar] [PubMed]
- Toole, B.P.; Slomiany, M.G. Hyaluronan, CD44 and Emmprin: Partners in cancer cell chemoresistance. Drug Resist. Updates 2008, 11, 110–121. [Google Scholar] [CrossRef] [Green Version]
- Edward, M.; Quinn, J.; Pasonen-Seppänen, S.; McCann, B.; Tammi, R. 4-Methylumbelliferone inhibits tumour cell growth and the activation of stromal hyaluronan synthesis by melanoma cell-derived factors. Br. J. Dermatol. 2010, 162, 1224–1232. [Google Scholar] [CrossRef]
- Kultti, A.; Pasonen-Seppänen, S.; Jauhiainen, M.; Rilla, K.J.; Kärnä, R.; Pyöriä, E.; Tammi, R.H.; Tammi, M.I. 4-Methylumbelliferone inhibits hyaluronan synthesis by depletion of cellular UDP-glucuronic acid and downregulation of hyaluronan synthase 2 and 3. Exp. Cell Res. 2009, 315, 1914–1923. [Google Scholar] [CrossRef]
- Urakawa, H.; Nishida, Y.; Wasa, J.; Arai, E.; Zhuo, L.; Kimata, K.; Kozawa, E.; Futamura, N.; Ishiguro, N. Inhibition of hyaluronan synthesis in breast cancer cells by 4-methylumbelliferone suppresses tumorigenicity in vitro and metastatic lesions of bone in vivo. Int. J. Cancer 2012, 130, 454–466. [Google Scholar] [CrossRef] [PubMed]
- Hajime, M.; Shuichi, Y.; Makoto, N.; Masanori, Y.; Ikuko, K.; Atsushi, K.; Mutsuo, S.; Keiichi, T. Inhibitory effect of 4-methylesculetin on hyaluronan synthesis slows the development of human pancreatic cancer in vitro and in nude mice. Int. J. Cancer 2007, 120, 2704–2709. [Google Scholar] [CrossRef]
- Morohashi, H.; Kon, A.; Nakai, M.; Yamaguchi, M.; Kakizaki, I.; Yoshihara, S.; Sasaki, M.; Takagaki, K. Study of hyaluronan synthase inhibitor, 4-methylumbelliferone derivatives on human pancreatic cancer cell (KP1-NL). Biochem. Biophys. Res. Commun. 2006, 345, 1454–1459. [Google Scholar] [CrossRef] [PubMed]
- Nakazawa, H.; Yoshihara, S.; Kudo, D.; Morohashi, H.; Kakizaki, I.; Kon, A.; Takagaki, K.; Sasaki, M. 4-methylumbelliferone, a hyaluronan synthase suppressor, enhances the anticancer activity of gemcitabine in human pancreatic cancer cells. Cancer Chemother. Pharmacol. 2006, 57, 165–170. [Google Scholar] [CrossRef]
- Michl, P.; Gress, T.M. Improving drug delivery to pancreatic cancer: Breaching the stromal fortress by targeting hyaluronic acid. Gut 2012, 61, 1377–1379. [Google Scholar] [CrossRef]
- Kohno, N.; Ohnuma, T.; Truog, P. Effects of hyaluronidase on doxorubicin penetration into squamous carcinoma multicellular tumor spheroids and its cell lethality. J. Cancer Res. Clin. Oncol. 1994, 120, 293–297. [Google Scholar] [CrossRef]
- Magzoub, M.; Jin, S.; Verkman, A. Enhanced macromolecule diffusion deep in tumors after enzymatic digestion of extracellular matrix collagen and its associated proteoglycan decorin. FASEB J. 2008, 22, 276–284. [Google Scholar] [CrossRef] [PubMed]
- Koltai, T.; Reshkin, S.J.; Carvalho, T.; Cardone, R.A. Targeting the Stromal Pro-Tumoral Hyaluronan-CD44 Pathway in Pancreatic Cancer. Int. J. Mol. Sci. 2021, 22, 3953. [Google Scholar] [CrossRef]
- Kelleher, F.C. Hedgehog signaling and therapeutics in pancreatic cancer. Carcinogenesis 2011, 32, 445–451. [Google Scholar] [CrossRef]
- Ji, Z.; Mei, F.C.; Xie, J.; Cheng, X. Oncogenic KRAS activates hedgehog signaling pathway in pancreatic cancer cells. J. Biol. Chem. 2007, 282, 14048–14055. [Google Scholar] [CrossRef] [Green Version]
- Bailey, J.M.; Swanson, B.J.; Hamada, T.; Eggers, J.P.; Singh, P.K.; Caffery, T.; Ouellette, M.M.; Hollingsworth, M.A. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin. Cancer Res. 2008, 14, 5995–6004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bai, Y.; Bai, Y.; Dong, J.; Li, Q.; Jin, Y.; Chen, B.; Zhou, M. Hedgehog signaling in pancreatic fibrosis and cancer. Medicine 2016, 95, e2996. [Google Scholar] [CrossRef]
- Feldmann, G.; Fendrich, V.; McGovern, K.; Bedja, D.; Bisht, S.; Alvarez, H.; Koorstra, J.-B.M.; Habbe, N.; Karikari, C.; Mullendore, M. An orally bioavailable small-molecule inhibitor of Hedgehog signaling inhibits tumor initiation and metastasis in pancreatic cancer. Mol. Cancer Ther. 2008, 7, 2725–2735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kelleher, F.C.; McDermott, R. Aberrations and therapeutics involving the developmental pathway Hedgehog in pancreatic cancer. Vitam. Horm. 2012, 88, 355–378. [Google Scholar] [CrossRef]
- Bisht, S.; Brossart, P.; Maitra, A.; Feldmann, G. Agents targeting the Hedgehog pathway for pancreatic cancer treatment. Curr. Opin. Investig. Drugs 2010, 11, 1387–1398. [Google Scholar]
- Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Richards, D.A.; Stephenson, J.; Wolpin, B.M.; Becerra, C.; Hamm, J.T.; Messersmith, W.A.; Devens, S.; Cushing, J.; Schmalbach, T.; Fuchs, C.S. A phase Ib trial of IPI-926, a hedgehog pathway inhibitor, plus gemcitabine in patients with metastatic pancreatic cancer. J. Clin. Oncol. 2012, 30 (Suppl. S4), 213. [Google Scholar] [CrossRef]
- McCarroll, J.A.; Phillips, P.A.; Santucci, N.; Pirola, R.C.; Wilson, J.; Apte, M. Vitamin A inhibits pancreatic stellate cell activation: Implications for treatment of pancreatic fibrosis. Gut 2006, 55, 79–89. [Google Scholar] [CrossRef]
- Froeling, F.E.; Feig, C.; Chelala, C.; Dobson, R.; Mein, C.E.; Tuveson, D.A.; Clevers, H.; Hart, I.R.; Kocher, H.M. Retinoic acid–induced pancreatic stellate cell quiescence reduces paracrine Wnt–β-catenin signaling to slow tumor progression. Gastroenterology 2011, 141, 1486–1497.e14. [Google Scholar] [CrossRef]
- Chronopoulos, A.; Robinson, B.; Sarper, M.; Cortes, E.; Auernheimer, V.; Lachowski, D.; Attwood, S.; García, R.; Ghassemi, S.; Fabry, B. ATRA mechanically reprograms pancreatic stellate cells to suppress matrix remodelling and inhibit cancer cell invasion. Nat. Commun. 2016, 7, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Carter, N.J. Pirfenidone. Drugs 2011, 71, 1721–1732. [Google Scholar] [CrossRef]
- Taniguchi, H.; Ebina, M.; Kondoh, Y.; Ogura, T.; Azuma, A.; Suga, M.; Taguchi, Y.; Takahashi, H.; Nakata, K.; Sato, A. Pirfenidone in idiopathic pulmonary fibrosis. Eur. Respir. J. 2010, 35, 821–829. [Google Scholar] [CrossRef]
- Noble, P.W.; Albera, C.; Bradford, W.Z.; Costabel, U.; Glassberg, M.K.; Kardatzke, D.; King, T.E., Jr.; Lancaster, L.; Sahn, S.A.; Szwarcberg, J. Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): Two randomised trials. Lancet 2011, 377, 1760–1769. [Google Scholar] [CrossRef]
- Garcıa, L.; Hernández, I.; Sandoval, A.; Salazar, A.; Garcia, J.; Vera, J.; Grijalva, G.; Muriel, P.; Margolin, S.; Armendariz-Borunda, J. Pirfenidone effectively reverses experimental liver fibrosis. J. Hepatol. 2002, 37, 797–805. [Google Scholar] [CrossRef]
- Polydorou, C.; Mpekris, F.; Papageorgis, P.; Voutouri, C.; Stylianopoulos, T. Pirfenidone normalizes the tumor microenvironment to improve chemotherapy. Oncotarget 2017, 8, 24506. [Google Scholar] [CrossRef] [Green Version]
- Knüppel, L.; Ishikawa, Y.; Aichler, M.; Heinzelmann, K.; Hatz, R.; Behr, J.; Walch, A.; Bächinger, H.P.; Eickelberg, O.; Staab-Weijnitz, C.A. A novel antifibrotic mechanism of nintedanib and pirfenidone. Inhibition of collagen fibril assembly. Am. J. Respir. Cell Mol. Biol. 2017, 57, 77–90. [Google Scholar] [CrossRef] [PubMed]
- Kaneko, M.; Inoue, H.; Nakazawa, R.; Azuma, N.; Suzuki, M.; Yamauchi, S.; Margolin, S.; Tsubota, K.; Saito, I. Pirfenidone induces intercellular adhesion molecule-1 (ICAM-1) down-regulation on cultured human synovial fibroblasts. Clin. Exp. Immunol. 1998, 113, 72–76. [Google Scholar] [CrossRef]
- Shihab, F.S.; Bennett, W.M.; Yi, H.; Andoh, T.F. Pirfenidone treatment decreases transforming growth factor-β1 and matrix proteins and ameliorates fibrosis in chronic cyclosporine nephrotoxicity. Am. J. Transplant. 2002, 2, 111–119. [Google Scholar] [CrossRef] [PubMed]
- Nakazato, H.; Oku, H.; Yamane, S.; Tsuruta, Y.; Suzuki, R. A novel anti-fibrotic agent pirfenidone suppresses tumor necrosis factor-α at the translational level. Eur. J. Pharmacol. 2002, 446, 177–185. [Google Scholar] [CrossRef]
- Didiasova, M.; Singh, R.; Wilhelm, J.; Kwapiszewska, G.; Wujak, L.; Zakrzewicz, D.; Schaefer, L.; Markart, P.; Seeger, W.; Lauth, M. Pirfenidone exerts antifibrotic effects through inhibition of GLI transcription factors. FASEB J. 2017, 31, 1916–1928. [Google Scholar] [CrossRef] [Green Version]
- Oku, H.; Nakazato, H.; Horikawa, T.; Tsuruta, Y.; Suzuki, R. Pirfenidone suppresses tumor necrosis factor-α, enhances interleukin-10 and protects mice from endotoxic shock. Eur. J. Pharmacol. 2002, 446, 167–176. [Google Scholar] [CrossRef]
- Kozono, S.; Ohuchida, K.; Eguchi, D.; Ikenaga, N.; Fujiwara, K.; Cui, L.; Mizumoto, K.; Tanaka, M. Pirfenidone inhibits pancreatic cancer desmoplasia by regulating stellate cells. Cancer Res. 2013, 73, 2345–2356. [Google Scholar] [CrossRef] [Green Version]
- Usugi, E.; Ishii, K.; Hirokawa, Y.; Kanayama, K.; Matsuda, C.; Uchida, K.; Shiraishi, T.; Watanabe, M. Antifibrotic agent pirfenidone suppresses proliferation of human pancreatic cancer cells by inducing G0/G1 cell cycle arrest. Pharmacology 2019, 103, 250–256. [Google Scholar] [CrossRef] [PubMed]
- Yamada, T.; Kuno, A.; Masuda, K.; Ogawa, K.; Sogawa, M.; Nakamura, S.; Ando, T.; Sano, H.; Nakazawa, T.; Ohara, H. Candesartan, an angiotensin II receptor antagonist, suppresses pancreatic inflammation and fibrosis in rats. J. Pharmacol. Exp. Ther. 2003, 307, 17–23. [Google Scholar] [CrossRef] [PubMed]
- Stephenson, J.; Richards, D.; Wolpin, B.; Becerra, C.; Hamm, J.; Messersmith, W.; Devens, S.; Cushing, J.; Goddard, J.; Schmalbach, T. The safety of IPI-926, a novel hedgehog pathway inhibitor, in combination with gemcitabine in patients (pts) with metastatic pancreatic cancer. J. Clin. Oncol. 2011, 29 (Suppl. S15), 4114. [Google Scholar] [CrossRef]
- Ko, A.H.; LoConte, N.; Tempero, M.A.; Walker, E.J.; Kelley, R.K.; Lewis, S.; Chang, W.-C.; Kantoff, E.; Vannier, M.W.; Catenacci, D.V. A phase I study of FOLFIRINOX plus IPI-926, a hedgehog pathway inhibitor, for advanced pancreatic adenocarcinoma. Pancreas 2016, 45, 370. [Google Scholar] [CrossRef] [Green Version]
- Catenacci, D.V.; Junttila, M.R.; Karrison, T.; Bahary, N.; Horiba, M.N.; Nattam, S.R.; Marsh, R.; Wallace, J.; Kozloff, M.; Rajdev, L. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J. Clin. Oncol. 2015, 33, 4284. [Google Scholar] [CrossRef]
- Yoshida, E.; Kudo, D.; Nagase, H.; Shimoda, H.; Suto, S.; Negishi, M.; Kakizaki, I.; Endo, M.; Hakamada, K. Antitumor effects of the hyaluronan inhibitor 4-methylumbelliferone on pancreatic cancer. Oncol. Lett. 2016, 12, 2337–2344. [Google Scholar] [CrossRef] [PubMed]
- Lin, Z.; Zheng, L.-C.; Zhang, H.-J.; Tsang, S.W.; Bian, Z.-X. Anti-fibrotic effects of phenolic compounds on pancreatic stellate cells. BMC Complement. Altern. Med. 2015, 15, 259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kanai, M.; Yoshimura, K.; Asada, M.; Imaizumi, A.; Suzuki, C.; Matsumoto, S.; Nishimura, T.; Mori, Y.; Masui, T.; Kawaguchi, Y. A phase I/II study of gemcitabine-based chemotherapy plus curcumin for patients with gemcitabine-resistant pancreatic cancer. Cancer Chemother. Pharmacol. 2011, 68, 157–164. [Google Scholar] [CrossRef] [Green Version]
- Shehzad, A.; Qureshi, M.; Anwar, M.N.; Lee, Y.S. Multifunctional curcumin mediate multitherapeutic effects. J. Food Sci. 2017, 82, 2006–2015. [Google Scholar] [CrossRef] [Green Version]
- Goel, A.; Kunnumakkara, A.B.; Aggarwal, B.B. Curcumin as “Curecumin”: From kitchen to clinic. Biochem. Pharmacol. 2008, 75, 787–809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gundewar, C.; Ansari, D.; Tang, L.; Wang, Y.; Liang, G.; Rosendahl, A.H.; Saleem, M.A.; Andersson, R. Antiproliferative effects of curcumin analog L49H37 in pancreatic stellate cells: A comparative study. Ann. Gastroenterol. Q. Publ. Hell. Soc. Gastroenterol. 2015, 28, 391. [Google Scholar]
- Tsang, S.W.; Zhang, H.; Lin, C.; Xiao, H.; Wong, M.; Shang, H.; Yang, Z.-J.; Lu, A.; Yung, K.K.-L.; Bian, Z. Rhein, a natural anthraquinone derivative, attenuates the activation of pancreatic stellate cells and ameliorates pancreatic fibrosis in mice with experimental chronic pancreatitis. PLoS ONE 2013, 8, e82201. [Google Scholar] [CrossRef] [PubMed]
- Tsang, S.W.; Bian, Z.X. Anti-fibrotic and anti-tumorigenic effects of rhein, a natural anthraquinone derivative, in mammalian stellate and carcinoma cells. Phytother. Res. 2015, 29, 407–414. [Google Scholar] [CrossRef]
- Yan, B.; Cheng, L.; Jiang, Z.; Chen, K.; Zhou, C.; Sun, L.; Cao, J.; Qian, W.; Li, J.; Shan, T. Resveratrol inhibits ROS-promoted activation and glycolysis of pancreatic stellate cells via suppression of miR-21. Oxid. Med. Cell. Longev. 2018. [Google Scholar] [CrossRef]
- Wei, W.-T.; Lin, S.-Z.; Liu, D.-L.; Wang, Z.-H. The distinct mechanisms of the antitumor activity of emodin in different types of cancer. Oncol. Rep. 2013, 30, 2555–2562. [Google Scholar] [CrossRef] [Green Version]
- Cha, T.L.; Chuang, M.J.; Tang, S.H.; Wu, S.T.; Sun, K.H.; Chen, T.T.; Sun, G.H.; Chang, S.Y.; Yu, C.P.; Ho, J.Y. Emodin modulates epigenetic modifications and suppresses bladder carcinoma cell growth. Mol. Carcinog. 2015, 54, 167–177. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Wang, X.-B.; Yu, Q.-M.; Luo, Q.-Y.; Zhang, Z.-Z. Synergistic cancer growth-inhibitory effect of emodin and low-dose cisplatin on gastric cancer cells in vitro. Trop. J. Pharm. Res. 2015, 14, 1427–1434. [Google Scholar] [CrossRef] [Green Version]
- Jia, X.; Yu, F.; Wang, J.; Iwanowycz, S.; Saaoud, F.; Wang, Y.; Hu, J.; Wang, Q.; Fan, D. Emodin suppresses pulmonary metastasis of breast cancer accompanied with decreased macrophage recruitment and M2 polarization in the lungs. Breast Cancer Res. Treat. 2014, 148, 291–302. [Google Scholar] [CrossRef]
- Masamune, A.; Satoh, M.; Kikuta, K.; Suzuki, N.; Satoh, K.; Shimosegawa, T. Ellagic acid blocks activation of pancreatic stellate cells. Biochem. Pharmacol. 2005, 70, 869–878. [Google Scholar] [CrossRef]
- Aono, Y.; Nishioka, Y.; Inayama, M.; Ugai, M.; Kishi, J.; Uehara, H.; Izumi, K.; Sone, S. Imatinib as a novel antifibrotic agent in bleomycin-induced pulmonary fibrosis in mice. Am. J. Respir. Crit. Care Med. 2005, 171, 1279–1285. [Google Scholar] [CrossRef] [PubMed]
- Gioni, V.; Karampinas, T.; Voutsinas, G.; Roussidis, A.E.; Papadopoulos, S.; Karamanos, N.K.; Kletsas, D. Imatinib mesylate inhibits proliferation and exerts an antifibrotic effect in human breast stroma fibroblasts. Mol. Cancer Res. 2008, 6, 706–714. [Google Scholar] [CrossRef] [Green Version]
- Shiha, G.E.; Abu-Elsaad, N.M.; Zalata, K.R.; Ibrahim, T.M. Tracking anti-fibrotic pathways of nilotinib and imatinib in experimentally induced liver fibrosis: A n insight. Clin. Exp. Pharmacol. Physiol. 2014, 41, 788–797. [Google Scholar] [CrossRef] [PubMed]
- Yoshiji, H.; Noguchi, R.; Kuriyama, S.; Ikenaka, Y.; Yoshii, J.; Yanase, K.; Namisaki, T.; Kitade, M.; Masaki, T.; Fukui, H. Imatinib mesylate (STI-571) attenuates liver fibrosis development in rats. Am. J. Physiol.-Gastrointest. Liver Physiol. 2005, 288, G907–G913. [Google Scholar] [CrossRef]
- Li, M.; Abdollahi, A.; Gröne, H.-J.; Lipson, K.E.; Belka, C.; Huber, P.E. Late treatment with imatinib mesylate ameliorates radiation-induced lung fibrosis in a mouse model. Radiat. Oncol. 2009, 4, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Vittal, R.; Zhang, H.; Han, M.K.; Moore, B.B.; Horowitz, J.C.; Thannickal, V.J. Effects of the protein kinase inhibitor, imatinib mesylate, on epithelial/mesenchymal phenotypes: Implications for treatment of fibrotic diseases. J. Pharmacol. Exp. Ther. 2007, 321, 35–44. [Google Scholar] [CrossRef]
- Moss, R.A.; Moore, D.; Mulcahy, M.F.; Nahum, K.; Saraiya, B.; Eddy, S.; Kleber, M.; Poplin, E.A. A multi-institutional phase 2 study of imatinib mesylate and gemcitabine for first-line treatment of advanced pancreatic cancer. Gastrointest. Cancer Res. GCR 2012, 5, 77. [Google Scholar] [PubMed]
- Gharibo, M.; Patrick-Miller, L.; Zheng, L.; Guensch, L.; Juvidian, P.; Poplin, E. A phase II trial of imatinib mesylate in patients with metastatic pancreatic cancer. Pancreas 2008, 36, 341–345. [Google Scholar] [CrossRef]
- Duan, W.; Chen, K.; Jiang, Z.; Chen, X.; Sun, L.; Li, J.; Lei, J.; Xu, Q.; Ma, J.; Li, X. Desmoplasia suppression by metformin-mediated AMPK activation inhibits pancreatic cancer progression. Cancer Lett. 2017, 385, 225–233. [Google Scholar] [CrossRef]
- Qian, W.; Li, J.; Chen, K.; Jiang, Z.; Cheng, L.; Zhou, C.; Yan, B.; Cao, J.; Ma, Q.; Duan, W. Metformin suppresses tumor angiogenesis and enhances the chemosensitivity of gemcitabine in a genetically engineered mouse model of pancreatic cancer. Life Sci. 2018, 208, 253–261. [Google Scholar] [CrossRef] [PubMed]
- Elahi-Gedwillo, K.Y.; Carlson, M.; Zettervall, J.; Provenzano, P.P. Antifibrotic therapy disrupts stromal barriers and modulates the immune landscape in pancreatic ductal adenocarcinoma. Cancer Res. 2019, 79, 372–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wong, K.M.; Horton, K.J.; Coveler, A.L.; Hingorani, S.R.; Harris, W.P. Targeting the tumor stroma: The biology and clinical development of pegylated recombinant human hyaluronidase (PEGPH20). Curr. Oncol. Rep. 2017, 19, 1–9. [Google Scholar] [CrossRef]
- Vennin, C.; Chin, V.T.; Warren, S.C.; Lucas, M.C.; Herrmann, D.; Magenau, A.; Melenec, P.; Walters, S.N.; del Monte-Nieto, G.; Conway, J.R. Transient tissue priming via ROCK inhibition uncouples pancreatic cancer progression, sensitivity to chemotherapy, and metastasis. Sci. Transl. Med. 2017. [Google Scholar] [CrossRef] [Green Version]
- Whatcott, C.J.; Ng, S.; Barrett, M.T.; Hostetter, G.; Von Hoff, D.D.; Han, H. Inhibition of ROCK1 kinase modulates both tumor cells and stromal fibroblasts in pancreatic cancer. PLoS ONE 2017, 12, e0183871. [Google Scholar] [CrossRef] [Green Version]
- Vennin, C.; Rath, N.; Pajic, M.; Olson, M.F.; Timpson, P. Targeting ROCK activity to disrupt and prime pancreatic cancer for chemotherapy. Small GTPases 2020, 11, 45–52. [Google Scholar] [CrossRef] [PubMed]
- Takeda, H.; Okada, M.; Suzuki, S.; Kuramoto, K.; Sakaki, H.; Watarai, H.; Sanomachi, T.; Seino, S.; Yoshioka, T.; Kitanaka, C. Rho-associated protein kinase (ROCK) inhibitors inhibit survivin expression and sensitize pancreatic cancer stem cells to gemcitabine. Anticancer Res. 2016, 36, 6311–6318. [Google Scholar] [CrossRef] [Green Version]
- Rath, N.; Munro, J.; Cutiongco, M.F.; Jagiełło, A.; Gadegaard, N.; McGarry, L.; Unbekandt, M.; Michalopoulou, E.; Kamphorst, J.J.; Sumpton, D. Rho kinase inhibition by AT13148 blocks pancreatic ductal adenocarcinoma invasion and tumor growth. Cancer Res. 2018, 78, 3321–3336. [Google Scholar] [CrossRef] [Green Version]
- Wen, W.X.; Lee, S.Y.; Siang, R.; Koh, R.Y. Repurposing pentoxifylline for the treatment of fibrosis: An overview. Adv. Ther. 2017, 34, 1245–1269. [Google Scholar] [CrossRef] [PubMed]
- Bienvenu, J.; Doche, C.; Gutowski, M.-C.; Lenoble, M.; Lepape, A.; Perdrix, J.-P. Production of proinflammatory cytokines and cytokines involved in the TH1/TH2 balance is modulated by pentoxifylline. J. Cardiovasc. Pharmacol. 1995, 25, S80–S84. [Google Scholar] [CrossRef]
- Abdel-Salam, O.M.; Baiuomy, A.R.; El-Shenawy, S.M.; Arbid, M.S. The anti-inflammatory effects of the phosphodiesterase inhibitor pentoxifylline in the rat. Pharmacol. Res. 2003, 47, 331–340. [Google Scholar] [CrossRef]
- Zeng, X.-P.; Wang, L.-J.; Guo, H.-L.; He, L.; Bi, Y.-W.; Xu, Z.-L.; Li, Z.-S.; Hu, L.-H. Dasatinib ameliorates chronic pancreatitis induced by caerulein via anti-fibrotic and anti-inflammatory mechanism. Pharmacol. Res. 2019, 147, 104357. [Google Scholar] [CrossRef]
- Ho, W.J.; Jaffee, E.M.; Zheng, L. The tumour microenvironment in pancreatic cancer—Clinical challenges and opportunities. Nat. Rev. Clin. Oncol. 2020, 17, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Damaghi, M.; Wojtkowiak, J.W.; Gillies, R.J. pH sensing and regulation in cancer. Front. Physiol. 2013, 4, 370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Milito, A.; Fais, S. Tumor acidity, chemoresistance and proton pump inhibitors. Future Oncol. 2005, 1, 779–786. [Google Scholar] [CrossRef]
- Denko, N.C. Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat. Rev. Cancer 2008, 8, 705–713. [Google Scholar] [CrossRef]
- Warburg, O. The metabolism of carcinoma cells. J. Cancer Res. 1925, 9, 148–163. [Google Scholar] [CrossRef] [Green Version]
- Strapcova, S.; Takacova, M.; Csaderova, L.; Martinelli, P.; Lukacikova, L.; Gal, V.; Kopacek, J.; Svastova, E. Clinical and pre-clinical evidence of carbonic anhydrase ix in pancreatic cancer and its high expression in pre-cancerous lesions. Cancers 2020, 12, 2005. [Google Scholar] [CrossRef]
- Rong, Y.; Wu, W.; Ni, X.; Kuang, T.; Jin, D.; Wang, D.; Lou, W. Lactate dehydrogenase A is overexpressed in pancreatic cancer and promotes the growth of pancreatic cancer cells. Tumor Biol. 2013, 34, 1523–1530. [Google Scholar] [CrossRef]
- Kong, S.C.; Gianuzzo, A.; Novak, I.; Pedersen, S.F. Acid-base transport in pancreatic cancer: Molecular mechanisms and clinical potential. Biochem. Cell Biol. 2014, 92, 449–459. [Google Scholar] [CrossRef] [PubMed]
- Stock, C.; Pedersen, S.F. Roles of pH and the Na+/H+ exchanger NHE1 in cancer: From cell biology and animal models to an emerging translational perspective. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2017; pp. 5–16. [Google Scholar]
- Webb, B.A.; Chimenti, M.; Jacobson, M.P.; Barber, D.L. Dysregulated pH: A perfect storm for cancer progression. Nat. Rev. Cancer 2011, 11, 671–677. [Google Scholar] [CrossRef] [PubMed]
- Reshkin, S.J.; Bellizzi, A.; Caldeira, S.; Albarani, V.; Malanchi, I.; Poignee, M.; Alunni-Fabbroni, M.; Casavola, V.; Tommasino, M. Na+/H+ exchanger-dependent intracellular alkalinization is an early event in malignant transformation and plays an essential role in the development of subsequent transformation-associated phenotypes. FASEB J. 2000, 14, 2185–2197. [Google Scholar] [CrossRef] [Green Version]
- Boedtkjer, E.; Pedersen, S.F. The acidic tumor microenvironment as a driver of cancer. Annu. Rev. Physiol. 2020, 82, 103–126. [Google Scholar] [CrossRef] [Green Version]
- Duffy, M. The role of proteolytic enzymes in cancer invasion and metastasis. Clin. Exp. Metastasis 1992, 10, 145–155. [Google Scholar] [CrossRef] [PubMed]
- Palermo, C.; Joyce, J.A. Cysteine cathepsin proteases as pharmacological targets in cancer. Trends Pharmacol. Sci. 2008, 29, 22–28. [Google Scholar] [CrossRef]
- Harguindey, S.; Arranz, J.L.; Wahl, M.L.; Orive, G.; Reshkin, S.J. Proton transport inhibitors as potentially selective anticancer drugs. Anticancer Res. 2009, 29, 2127–2136. [Google Scholar]
- Cardone, R.A.; Casavola, V.; Reshkin, S.J. The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis. Nat. Rev. Cancer 2005, 5, 786–795. [Google Scholar] [CrossRef]
- Weniger, M.; Honselmann, K.C.; Liss, A.S. The extracellular matrix and pancreatic cancer: A complex relationship. Cancers 2018, 10, 316. [Google Scholar] [CrossRef] [Green Version]
- Shin, S.C.; Thomas, D.; Radhakrishnan, P.; Hollingsworth, M.A. Invasive phenotype induced by low extracellular pH requires mitochondria dependent metabolic flexibility. Biochem. Biophys. Res. Commun. 2020, 525, 162–168. [Google Scholar] [CrossRef]
- Dhup, S.; Kumar Dadhich, R.; Ettore Porporato, P.; Sonveaux, P. Multiple biological activities of lactic acid in cancer: Influences on tumor growth, angiogenesis and metastasis. Curr. Pharm. Des. 2012, 18, 1319–1330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, S.Y.C.; Collins, C.C.; Gout, P.W.; Wang, Y. Cancer-generated lactic acid: A regulatory, immunosuppressive metabolite? J. Pathol. 2013, 230, 350–355. [Google Scholar] [CrossRef] [Green Version]
- Gonen, N.; Assaraf, Y.G. Antifolates in cancer therapy: Structure, activity and mechanisms of drug resistance. Drug Resist. Updates 2012, 15, 183–210. [Google Scholar] [CrossRef] [PubMed]
- Liu, F.-S. Mechanisms of chemotherapeutic drug resistance in cancer therapy—A quick review. Taiwan. J. Obstet. Gynecol. 2009, 48, 239–244. [Google Scholar] [CrossRef] [Green Version]
- Gottesman, M.M. Mechanisms of cancer drug resistance. Annu. Rev. Med. 2002, 53, 615–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bailey, K.M.; Wojtkowiak, J.W.; Hashim, A.I.; Gillies, R.J. Targeting the metabolic microenvironment of tumors. Adv. Pharmacol. 2012, 65, 63–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Milito, A.; Fais, S. Proton pump inhibitors may reduce tumour resistance. Expert Opin. Pharmacother. 2005, 6, 1049–1054. [Google Scholar] [CrossRef]
- Roos, A. Weak acids, weak bases, and intracellular pH. Respir. Physiol. 1978, 33, 27–30. [Google Scholar] [CrossRef]
- Gillies, R.; Deamer, D. Intracellular pH changes during the cell cycle in Tetrahymena. J. Cell. Physiol. 1979, 100, 23–31. [Google Scholar] [CrossRef] [PubMed]
- Mahoney, B.P.; Raghunand, N.; Baggett, B.; Gillies, R.J. Tumor acidity, ion trapping and chemotherapeutics: I. Acid pH affects the distribution of chemotherapeutic agents in vitro. Biochem. Pharmacol. 2003, 66, 1207–1218. [Google Scholar] [CrossRef]
- Belhoussine, R.; Morjani, H.; Sharonov, S.; Ploton, D.; Manfait, M. Characterization of intracellular pH gradients in human multidrug-resistant tumor cells by means of scanning microspectrofluorometry and dual-emission-ratio probes. Int. J. Cancer 1999, 81, 81–89. [Google Scholar] [CrossRef]
- Newell, K.; Wood, P.; Stratford, I.; Tannock, I. Effects of agents which inhibit the regulation of intracellular pH on murine solid tumours. Br. J. Cancer 1992, 66, 311–317. [Google Scholar] [CrossRef] [Green Version]
- Ohtsubo, T.; Igawa, H.; Saito, T.; Matsumoto, H.; Park, H.J.; Song, C.W.; Kano, E.; Saito, H. Acidic environment modifies heat-or radiation-induced apoptosis in human maxillary cancer cells. Int. J. Radiat. Oncol. Biol. Phys. 2001, 49, 1391–1398. [Google Scholar] [CrossRef]
- Taylor, S.; Spugnini, E.P.; Assaraf, Y.G.; Azzarito, T.; Rauch, C.; Fais, S. Microenvironment acidity as a major determinant of tumor chemoresistance: Proton pump inhibitors (PPIs) as a novel therapeutic approach. Drug Resist. Updates 2015, 23, 69–78. [Google Scholar] [CrossRef]
- Luciani, F.; Molinari, A.; Lozupone, F.; Calcabrini, A.; Lugini, L.; Stringaro, A.; Puddu, P.; Arancia, G.; Cianfriglia, M.; Fais, S. P-glycoprotein–actin association through ERM family proteins: A role in P-glycoprotein function in human cells of lymphoid origin. Blood J. Am. Soc. Hematol. 2002, 99, 641–648. [Google Scholar] [CrossRef] [PubMed]
- Brambilla, D.; Zamboni, S.; Federici, C.; Lugini, L.; Lozupone, F.; Milito, A.D.; Cecchetti, S.; Cianfriglia, M.; Fais, S. P-glycoprotein binds to ezrin at amino acid residues 149–242 in the FERM domain and plays a key role in the multidrug resistance of human osteosarcoma. Int. J. Cancer 2012, 130, 2824–2834. [Google Scholar] [CrossRef] [PubMed]
- Luciani, F.; Spada, M.; De Milito, A.; Molinari, A.; Rivoltini, L.; Montinaro, A.; Marra, M.; Lugini, L.; Logozzi, M.; Lozupone, F. Effect of proton pump inhibitor pretreatment on resistance of solid tumors to cytotoxic drugs. J. Natl. Cancer Inst. 2004, 96, 1702–1713. [Google Scholar] [CrossRef]
- Koong, A.C.; Mehta, V.K.; Le, Q.T.; Fisher, G.A.; Terris, D.J.; Brown, J.M.; Bastidas, A.J.; Vierra, M. Pancreatic tumors show high levels of hypoxia. Int. J. Radiat. Oncol. Biol. Phys. 2000, 48, 919–922. [Google Scholar] [CrossRef]
- Colbert, L.E.; Fisher, S.B.; Balci, S.; Saka, B.; Chen, Z.; Kim, S.; El-Rayes, B.F.; Adsay, N.V.; Maithel, S.K.; Landry, J.C. High nuclear hypoxia-inducible factor 1 alpha expression is a predictor of distant recurrence in patients with resected pancreatic adenocarcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2015, 91, 631–639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Semenza, G.L.; Wang, G.L. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol. Cell. Biol. 1992, 12, 5447–5454. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.L.; Semenza, G.L. Purification and Characterization of Hypoxia-inducible Factor 1 (∗). J. Biol. Chem. 1995, 270, 1230–1237. [Google Scholar] [CrossRef] [Green Version]
- Semenza, G.L. Regulation of oxygen homeostasis by hypoxia-inducible factor 1. Physiology 2009, 24, 97–106. [Google Scholar] [CrossRef] [Green Version]
- Ke, Q.; Costa, M. Hypoxia-inducible factor-1 (HIF-1). Mol. Pharmacol. 2006, 70, 1469–1480. [Google Scholar] [CrossRef]
- Tan, Z.; Xu, J.; Zhang, B.; Shi, S.; Yu, X.; Liang, C. Hypoxia: A barricade to conquer the pancreatic cancer. Cell. Mol. Life Sci. 2020, 77, 1–7. [Google Scholar] [CrossRef]
- Huang, L.E.; Arany, Z.; Livingston, D.M.; Bunn, H.F. Activation of hypoxia-inducible transcription factor depends primarily upon redox-sensitive stabilization of its α subunit. J. Biol. Chem. 1996, 271, 32253–32259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wenger, R.H.; Gassmann, M. Oxygen (es) and the hypoxia-inducible factor-1. Biol. Chem. 1997, 378, 609–616. [Google Scholar]
- Semenza, G.L. HIF-1: Mediator of physiological and pathophysiological responses to hypoxia. J. Appl. Physiol. 2000. [Google Scholar] [CrossRef] [Green Version]
- Itakura, J.; Ishiwata, T.; Friess, H.; Fujii, H.; Matsumoto, Y.; Büchler, M.W.; Korc, M. Enhanced expression of vascular endothelial growth factor in human pancreatic cancer correlates with local disease progression. Clin. Cancer Res. 1997, 3, 1309–1316. [Google Scholar] [PubMed]
- Erkan, M.; Reiser-Erkan, C.; Michalski, C.W.; Deucker, S.; Sauliunaite, D.; Streit, S.; Esposito, I.; Friess, H.; Kleeff, J. Cancer-stellate cell interactions perpetuate the hypoxia-fibrosis cycle in pancreatic ductal adenocarcinoma. Neoplasia 2009, 11, 497–508. [Google Scholar] [CrossRef] [Green Version]
- Folkman, J. Angiogenesis inhibitors generated by tumors. Mol. Med. 1995, 1, 120–122. [Google Scholar] [CrossRef] [Green Version]
- Giatromanolaki, A.; Koukourakis, M.; Sivridis, E.; Turley, H.; Talks, K.; Pezzella, F.; Gatter, K.; Harris, A. Relation of hypoxia inducible factor 1 α and 2 α in operable non-small cell lung cancer to angiogenic/molecular profile of tumours and survival. Br. J. Cancer 2001, 85, 881–890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pouysségur, J.; Dayan, F.; Mazure, N.M. Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 2006, 441, 437–443. [Google Scholar] [CrossRef]
- Qiu, G.Z.; Jin, M.Z.; Dai, J.X.; Sun, W.; Feng, J.H.; Jin, W.L. Reprogramming of the Tumor in the Hypoxic Niche: The Emerging Concept and Associated Therapeutic Strategies. Trends Pharmacol. Sci. 2017, 38, 669–686. [Google Scholar] [CrossRef] [Green Version]
- Shibaji, T.; Nagao, M.; Ikeda, N.; Kanehiro, H.; Hisanaga, M.; Ko, S.; Fukumoto, A.; Nakajima, Y. Prognostic significance of HIF-1 alpha overexpression in human pancreatic cancer. Anticancer Res. 2003, 23, 4721–4727. [Google Scholar] [PubMed]
- Couvelard, A.; O’Toole, D.; Leek, R.; Turley, H.; Sauvanet, A.; Degott, C.; Ruszniewski, P.; Belghiti, J.; Harris, A.; Gatter, K. Expression of hypoxia-inducible factors is correlated with the presence of a fibrotic focus and angiogenesis in pancreatic ductal adenocarcinomas. Histopathology 2005, 46, 668–676. [Google Scholar] [CrossRef]
- Díaz, B.; Yuen, A.; Iizuka, S.; Higashiyama, S.; Courtneidge, S.A. Notch increases the shedding of HB-EGF by ADAM12 to potentiate invadopodia formation in hypoxia. J. Cell Biol. 2013, 201, 279–292. [Google Scholar] [CrossRef] [PubMed]
- Lucien, F.; Brochu-Gaudreau, K.; Arsenault, D.; Harper, K.; Dubois, C.M. Hypoxia-induced invadopodia formation involves activation of NHE-1 by the p90 ribosomal S6 kinase (p90RSK). PLoS ONE 2011, 6, e28851. [Google Scholar] [CrossRef] [Green Version]
- Hanna, S.C.; Krishnan, B.; Bailey, S.T.; Moschos, S.J.; Kuan, P.-F.; Shimamura, T.; Osborne, L.D.; Siegel, M.B.; Duncan, L.M.; O’Brien, E.T. HIF1α and HIF2α independently activate SRC to promote melanoma metastases. J. Clin. Investig. 2013, 123, 2078–2093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, S.; Huang, S.; Sun, Y.L. Epithelial-mesenchymal transition in pancreatic cancer: A review. BioMed Res. Int. 2017, 2017, 2646148. [Google Scholar] [CrossRef] [Green Version]
- Rhim, A.D.; Mirek, E.T.; Aiello, N.M.; Maitra, A.; Bailey, J.M.; McAllister, F.; Reichert, M.; Beatty, G.L.; Rustgi, A.K.; Vonderheide, R.H. EMT and dissemination precede pancreatic tumor formation. Cell 2012, 148, 349–361. [Google Scholar] [CrossRef] [Green Version]
- Li, W.; Kang, Y. Probing the fifty shades of EMT in metastasis. Trends Cancer 2016, 2, 65–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, W.-T. Membrane proteases: Roles in tissue remodeling and tumour invasion. Curr. Opin. Cell Biol. 1992, 4, 802–809. [Google Scholar] [CrossRef]
- Arumugam, T.; Ramachandran, V.; Fournier, K.F.; Wang, H.; Marquis, L.; Abbruzzese, J.L.; Gallick, G.E.; Logsdon, C.D.; McConkey, D.J.; Choi, W. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009, 69, 5820–5828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ide, T.; Kitajima, Y.; Miyoshi, A.; Ohtsuka, T.; Mitsuno, M.; Ohtaka, K.; Miyazaki, K. The hypoxic environment in tumor-stromal cells accelerates pancreatic cancer progression via the activation of paracrine hepatocyte growth factor/c-Met signaling. Ann. Surg. Oncol. 2007, 14, 2600–2607. [Google Scholar] [CrossRef]
- Spivak-Kroizman, T.R.; Hostetter, G.; Posner, R.; Aziz, M.; Hu, C.; Demeure, M.J.; Von Hoff, D.; Hingorani, S.R.; Palculict, T.B.; Izzo, J. Hypoxia triggers hedgehog-mediated tumor–stromal interactions in pancreatic cancer. Cancer Res. 2013, 73, 3235–3247. [Google Scholar] [CrossRef] [Green Version]
- Masamune, A.; Kikuta, K.; Watanabe, T.; Satoh, K.; Hirota, M.; Shimosegawa, T. Hypoxia stimulates pancreatic stellate cells to induce fibrosis and angiogenesis in pancreatic cancer. Am. J. Physiol.—Gastrointest. Liver Physiol. 2008, 295, G709–G717. [Google Scholar] [CrossRef] [Green Version]
- Greco, O.; Marples, B.; Joiner, M.C.; Scott, S.D. How to overcome (and exploit) tumor hypoxia for targeted gene therapy. J. Cell. Physiol. 2003, 197, 312–325. [Google Scholar] [CrossRef]
- Brown, J.M. Exploiting the hypoxic cancer cell: Mechanisms and therapeutic strategies. Mol. Med. Today 2000, 6, 157–162. [Google Scholar] [CrossRef]
- Papandreou, I.; Krishna, C.; Kaper, F.; Cai, D.; Giaccia, A.J.; Denko, N.C. Anoxia is necessary for tumor cell toxicity caused by a low-oxygen environment. Cancer Res. 2005, 65, 3171–3178. [Google Scholar] [CrossRef] [Green Version]
- Kinoshita, M.; Johnson, D.L.; Shatney, C.H.; Lee, Y.L.; Mochizuki, H. Cancer cells surviving hypoxia obtain hypoxia resistance and maintain anti-apoptotic potential under reoxygenation. Int. J. Cancer 2001, 91, 322–326. [Google Scholar] [CrossRef]
- Erler, J.T.; Cawthorne, C.J.; Williams, K.J.; Koritzinsky, M.; Wouters, B.G.; Wilson, C.; Miller, C.; Demonacos, C.; Stratford, I.J.; Dive, C. Hypoxia-mediated down-regulation of Bid and Bax in tumors occurs via hypoxia-inducible factor 1-dependent and-independent mechanisms and contributes to drug resistance. Mol. Cell. Biol. 2004, 24, 2875–2889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bacon, A.; Harris, A. Hypoxia-inducible factors and hypoxic cell death in tumour physiology. Ann. Med. 2004, 36, 530–539. [Google Scholar] [CrossRef]
- Yokoi, K.; Fidler, I.J. Hypoxia increases resistance of human pancreatic cancer cells to apoptosis induced by gemcitabine. Clin. Cancer Res. 2004, 10, 2299–2306. [Google Scholar] [CrossRef] [Green Version]
- Fardel, O.; Lecureur, V.; Guillouzo, A. The P-glycoprotein multidrug transporter. Gen. Pharmacol. Vasc. Syst. 1996, 27, 1283–1291. [Google Scholar] [CrossRef]
- Comerford, K.M.; Colgan, S.P. Assessing oxygen sensitivity of the multidrug resistance (MDR) gene. Methods Enzymol. 2004, 381, 376–387. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Bi, R.; Yin, H.; Liu, H.; Li, L. ENO1 silencing impaires hypoxia-induced gemcitabine chemoresistance associated with redox modulation in pancreatic cancer cells. Am. J. Transl. Res. 2019, 11, 4470. [Google Scholar] [PubMed]
- Abdalla, M.Y.; Britigan, B.E.; Wen, F.; Icardi, M.; McCormick, M.L.; LaBrecque, D.R.; Voigt, M.; Brown, K.E.; Schmidt, W.N. Down-regulation of heme oxygenase-1 by hepatitis C virus infection in vivo and by the in vitro expression of hepatitis C core protein. J. Infect. Dis. 2004, 190, 1109–1118. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Qu, M.; Huang, S.; Fu, Y.; Yang, L.; He, S.; Li, L.; Zhang, Z.; Lin, Q.; Zhang, L. A novel α-enolase-targeted drug delivery system for high efficacy prostate cancer therapy. Nanoscale 2018, 10, 13673–13683. [Google Scholar] [CrossRef]
- Pandol, S.; Edderkaoui, M.; Gukovsky, I.; Lugea, A.; Gukovskaya, A. Desmoplasia of pancreatic ductal adenocarcinoma. Clin. Gastroenterol. Hepatol. 2009, 7, S44–S47. [Google Scholar] [CrossRef] [Green Version]
- Hingorani, S.R.; Harris, W.P.; Beck, J.T.; Berdov, B.A.; Wagner, S.A.; Pshevlotsky, E.M.; Tjulandin, S.; Gladkov, O.; Holcombe, R.F.; Jiang, P. Final results of a phase Ib study of gemcitabine plus PEGPH20 in patients with stage IV previously untreated pancreatic cancer. J. Clin. Oncol. 2015, 33, 359. [Google Scholar] [CrossRef]
- Ding, X.; Zhou, X.; Jiang, B.; Zhao, Q.; Zhou, G. Triptolide suppresses proliferation, hypoxia-inducible factor-1α and c-Myc expression in pancreatic cancer cells. Mol. Med. Rep. 2015, 12, 4508–4513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, L.; Salnikov, A.V.; Bauer, N.; Aleksandrowicz, E.; Labsch, S.; Nwaeburu, C.; Mattern, J.; Gladkich, J.; Schemmer, P.; Werner, J. Triptolide reverses hypoxia-induced epithelial–mesenchymal transition and stem-like features in pancreatic cancer by NF-κB downregulation. Int. J. Cancer 2014, 134, 2489–2503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- ClinicalTrials.gov Identifier: NCT01927965. Study of Minnelide™ in Patients With Advanced GI Tumors. Available online: https://clinicaltrials.gov/ct2/show/NCT01927965 (accessed on 23 September 2021).
- Zhang, H.; Bosch-Marce, M.; Shimoda, L.A.; Tan, Y.S.; Baek, J.H.; Wesley, J.B.; Gonzalez, F.J.; Semenza, G.L. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J. Biol. Chem. 2008, 283, 10892–10903. [Google Scholar] [CrossRef] [Green Version]
- Lang, M.; Wang, X.; Wang, H.; Dong, J.; Lan, C.; Hao, J.; Huang, C.; Li, X.; Yu, M.; Yang, Y. Arsenic trioxide plus PX-478 achieves effective treatment in pancreatic ductal adenocarcinoma. Cancer Lett. 2016, 378, 87–96. [Google Scholar] [CrossRef] [PubMed]
- Abraham, R. mTOR as a positive regulator of tumor cell responses to hypoxia. In TOR; Springer: Berlin/Heidelberg, Germany, 2004; pp. 299–319. [Google Scholar] [CrossRef]
- Wolpin, B.M.; Hezel, A.F.; Abrams, T.; Blaszkowsky, L.S.; Meyerhardt, J.A.; Chan, J.A.; Enzinger, P.C.; Allen, B.; Clark, J.W.; Ryan, D.P. Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J. Clin. Oncol. 2009, 27, 193. [Google Scholar] [CrossRef] [PubMed]
- Kordes, S.; Klümpen, H.-J.; Weterman, M.J.; Schellens, J.H.; Richel, D.J.; Wilmink, J.W. Phase II study of capecitabine and the oral mTOR inhibitor everolimus in patients with advanced pancreatic cancer. Cancer Chemother. Pharmacol. 2015, 75, 1135–1141. [Google Scholar] [CrossRef] [Green Version]
- Shimojo, Y.; Akimoto, M.; Hisanaga, T.; Tanaka, T.; Tajima, Y.; Honma, Y.; Takenaga, K. Attenuation of reactive oxygen species by antioxidants suppresses hypoxia-induced epithelial-mesenchymal transition and metastasis of pancreatic cancer cells. Clin. Exp. Metastasis 2013, 30, 143–154. [Google Scholar] [CrossRef] [PubMed]
- Sibenaller, Z.A.; Welsh, J.L.; Du, C.; Witmer, J.R.; Schrock, H.E.; Du, J.; Buettner, G.R.; Goswami, P.C.; Cieslak III, J.A.; Cullen, J.J. Extracellular superoxide dismutase suppresses hypoxia-inducible factor-1α in pancreatic cancer. Free Radic. Biol. Med. 2014, 69, 357–366. [Google Scholar] [CrossRef] [Green Version]
- Yang, H.; Villani, R.M.; Wang, H.; Simpson, M.J.; Roberts, M.S.; Tang, M.; Liang, X. The role of cellular reactive oxygen species in cancer chemotherapy. J. Exp. Clin. Cancer Res. 2018, 37, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Jang, M.; Kim, S.S.; Lee, J. Cancer cell metabolism: Implications for therapeutic targets. Exp. Mol. Med. 2013, 45, e45. [Google Scholar] [CrossRef] [Green Version]
- Hsu, P.P.; Sabatini, D.M. Cancer cell metabolism: Warburg and beyond. Cell 2008, 134, 703–707. [Google Scholar] [CrossRef] [Green Version]
- Anastasiou, D. Tumour microenvironment factors shaping the cancer metabolism landscape. Br. J. Cancer 2017, 116, 277–286. [Google Scholar] [CrossRef]
- Liberti, M.V.; Locasale, J.W. The Warburg effect: How does it benefit cancer cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [Green Version]
- Patra, K.C.; Hay, N. The pentose phosphate pathway and cancer. Trends Biochem. Sci. 2014, 39, 347–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- DeNicola, G.M.; Cantley, L.C. Cancer’s fuel choice: New flavors for a picky eater. Mol. Cell 2015, 60, 514–523. [Google Scholar] [CrossRef] [Green Version]
- Kamphorst, J.J.; Nofal, M.; Commisso, C.; Hackett, S.R.; Lu, W.; Grabocka, E.; Vander Heiden, M.G.; Miller, G.; Drebin, J.A.; Bar-Sagi, D. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res. 2015, 75, 544–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Finicle, B.T.; Jayashankar, V.; Edinger, A.L. Nutrient scavenging in cancer. Nat. Rev. Cancer 2018, 18, 619–633. [Google Scholar] [CrossRef]
- Biancur, D.E.; Kimmelman, A.C. The plasticity of pancreatic cancer metabolism in tumor progression and therapeutic resistance. Biochim. Biophys. Acta (BBA)—Rev. Cancer 2018, 1870, 67–75. [Google Scholar] [CrossRef]
- Glick, D.; Barth, S.; Macleod, K.F. Autophagy: Cellular and molecular mechanisms. J. Pathol. 2010, 221, 3–12. [Google Scholar] [CrossRef] [Green Version]
- Kaur, J.; Debnath, J. Autophagy at the crossroads of catabolism and anabolism. Nat. Rev. Mol. Cell Biol. 2015, 16, 461–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell 2008, 132, 27–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, S.; Wang, X.; Contino, G.; Liesa, M.; Sahin, E.; Ying, H.; Bause, A.; Li, Y.; Stommel, J.M.; Dell’Antonio, G. Pancreatic cancers require autophagy for tumor growth. Genes Dev. 2011, 25, 717–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, A.; Rajeshkumar, N.; Wang, X.; Yabuuchi, S.; Alexander, B.M.; Chu, G.C.; Von Hoff, D.D.; Maitra, A.; Kimmelman, A.C. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov. 2014, 4, 905–913. [Google Scholar] [CrossRef] [Green Version]
- Kerr, M.C.; Teasdale, R.D. Defining macropinocytosis. Traffic 2009, 10, 364–371. [Google Scholar] [CrossRef]
- Commisso, C.; Davidson, S.M.; Soydaner-Azeloglu, R.G.; Parker, S.J.; Kamphorst, J.J.; Hackett, S.; Grabocka, E.; Nofal, M.; Drebin, J.A.; Thompson, C.B. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature 2013, 497, 633–637. [Google Scholar] [CrossRef] [Green Version]
- Davidson, S.M.; Jonas, O.; Keibler, M.A.; Hou, H.W.; Luengo, A.; Mayers, J.R.; Wyckoff, J.; Del Rosario, A.M.; Whitman, M.; Chin, C.R. Direct evidence for cancer-cell-autonomous extracellular protein catabolism in pancreatic tumors. Nat. Med. 2017, 23, 235–241. [Google Scholar] [CrossRef] [PubMed]
- Olivares, O.; Mayers, J.R.; Gouirand, V.; Torrence, M.E.; Gicquel, T.; Borge, L.; Lac, S.; Roques, J.; Lavaut, M.-N.; Berthezène, P. Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions. Nat. Commun. 2017, 8, 1–14. [Google Scholar] [CrossRef]
- DeBerardinis, R.J.; Chandel, N.S. Fundamentals of cancer metabolism. Sci. Adv. 2016, 2, e1600200. [Google Scholar] [CrossRef] [Green Version]
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
- Ferreira, L.M. Cancer metabolism: The Warburg effect today. Exp. Mol. Pathol. 2010, 89, 372–380. [Google Scholar] [CrossRef] [PubMed]
- Pavlides, S.; Whitaker-Menezes, D.; Castello-Cros, R.; Flomenberg, N.; Witkiewicz, A.K.; Frank, P.G.; Casimiro, M.C.; Wang, C.; Fortina, P.; Addya, S. The reverse Warburg effect: Aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle 2009, 8, 3984–4001. [Google Scholar] [CrossRef] [Green Version]
- Sancho, P.; Burgos-Ramos, E.; Tavera, A.; Kheir, T.B.; Jagust, P.; Schoenhals, M.; Barneda, D.; Sellers, K.; Campos-Olivas, R.; Graña, O. MYC/PGC-1α balance determines the metabolic phenotype and plasticity of pancreatic cancer stem cells. Cell Metab. 2015, 22, 590–605. [Google Scholar] [CrossRef] [Green Version]
- Derle, A.; De Santis, M.C.; Gozzelino, L.; Ratto, E.; Martini, M. The role of metabolic adaptation to nutrient stress in pancreatic cancer. Cell Stress 2018, 2, 332. [Google Scholar] [CrossRef]
- Hezel, A.F.; Kimmelman, A.C.; Stanger, B.Z.; Bardeesy, N.; DePinho, R.A. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2006, 20, 1218–1249. [Google Scholar] [CrossRef] [Green Version]
- DeBerardinis, R.J.; Lum, J.J.; Hatzivassiliou, G.; Thompson, C.B. The biology of cancer: Metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008, 7, 11–20. [Google Scholar] [CrossRef] [Green Version]
- Ying, H.; Kimmelman, A.C.; Lyssiotis, C.A.; Hua, S.; Chu, G.C.; Fletcher-Sananikone, E.; Locasale, J.W.; Son, J.; Zhang, H.; Coloff, J.L. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell 2012, 149, 656–670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, C.-W.; Purkayastha, A.; Jones, K.T.; Thaker, S.K.; Banerjee, U. In vivo genetic dissection of tumor growth and the Warburg effect. eLife 2016, 5, e18126. [Google Scholar] [CrossRef] [PubMed]
- Raho, S.; Capobianco, L.; Malivindi, R.; Vozza, A.; Piazzolla, C.; De Leonardis, F.; Gorgoglione, R.; Scarcia, P.; Pezzuto, F.; Agrimi, G. KRAS-regulated glutamine metabolism requires UCP2-mediated aspartate transport to support pancreatic cancer growth. Nat. Metab. 2020, 2, 1373–1381. [Google Scholar] [CrossRef] [PubMed]
- Vousden, K.H.; Ryan, K.M. p53 and metabolism. Nat. Rev. Cancer 2009, 9, 691–700. [Google Scholar] [CrossRef]
- Canto, C.; Menzies, K.J.; Auwerx, J. NAD+ metabolism and the control of energy homeostasis: A balancing act between mitochondria and the nucleus. Cell Metab. 2015, 22, 31–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martínez-Reyes, I.; Chandel, N.S. Waste not, want not: Lactate oxidation fuels the TCA cycle. Cell Metab. 2017, 26, 803–804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guillaumond, F.; Leca, J.; Olivares, O.; Lavaut, M.-N.; Vidal, N.; Berthezène, P.; Dusetti, N.J.; Loncle, C.; Calvo, E.; Turrini, O. Strengthened glycolysis under hypoxia supports tumor symbiosis and hexosamine biosynthesis in pancreatic adenocarcinoma. Proc. Natl. Acad. Sci. USA 2013, 110, 3919–3924. [Google Scholar] [CrossRef] [Green Version]
- Feron, O. Pyruvate into lactate and back: From the Warburg effect to symbiotic energy fuel exchange in cancer cells. Radiother. Oncol. 2009, 92, 329–333. [Google Scholar] [CrossRef] [PubMed]
- Alfarouk, K.O.; Verduzco, D.; Rauch, C.; Muddathir, A.K.; Adil, H.B.; Elhassan, G.O.; Ibrahim, M.E.; Orozco, J.D.P.; Cardone, R.A.; Reshkin, S.J. Glycolysis, tumor metabolism, cancer growth and dissemination. A new pH-based etiopathogenic perspective and therapeutic approach to an old cancer question. Oncoscience 2014, 1, 777. [Google Scholar] [CrossRef] [PubMed]
- Wise, D.R.; Thompson, C.B. Glutamine addiction: A new therapeutic target in cancer. Trends Biochem. Sci. 2010, 35, 427–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.; Pavlova, N.N.; Thompson, C.B. Cancer cell metabolism: The essential role of the nonessential amino acid, glutamine. EMBO J. 2017, 36, 1302–1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grasso, C.; Jansen, G.; Giovannetti, E. Drug resistance in pancreatic cancer: Impact of altered energy metabolism. Crit. Rev. Oncol. Hematol. 2017, 114, 139–152. [Google Scholar] [CrossRef]
- Guo, Y.; Deng, Y.; Li, X.; Ning, Y.; Lin, X.; Guo, S.; Chen, M.; Han, M. Glutaminolysis was induced by TGF-β1 through PP2Ac regulated Raf-MEK-ERK signaling in endothelial cells. PLoS ONE 2016, 11, e0162658. [Google Scholar] [CrossRef] [PubMed]
- Son, J.; Lyssiotis, C.A.; Ying, H.; Wang, X.; Hua, S.; Ligorio, M.; Perera, R.M.; Ferrone, C.R.; Mullarky, E.; Shyh-Chang, N. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 2013, 496, 101–105. [Google Scholar] [CrossRef] [PubMed]
- Abrego, J.; Gunda, V.; Vernucci, E.; Shukla, S.K.; King, R.J.; Dasgupta, A.; Goode, G.; Murthy, D.; Yu, F.; Singh, P.K. GOT1-mediated anaplerotic glutamine metabolism regulates chronic acidosis stress in pancreatic cancer cells. Cancer Lett. 2017, 400, 37–46. [Google Scholar] [CrossRef] [PubMed]
- Mayers, J.R.; Wu, C.; Clish, C.B.; Kraft, P.; Torrence, M.E.; Fiske, B.P.; Yuan, C.; Bao, Y.; Townsend, M.K.; Tworoger, S.S. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat. Med. 2014, 20, 1193–1198. [Google Scholar] [CrossRef] [PubMed]
- Tumas, J.; Baskirova, I.; Petrenas, T.; Norkuniene, J.; Strupas, K.; Sileikis, A. Towards a personalized approach in pancreatic cancer diagnostics through plasma amino acid analysis. Anticancer Res. 2019, 39, 2035–2042. [Google Scholar] [CrossRef] [PubMed]
- Sousa, C.M.; Biancur, D.E.; Wang, X.; Halbrook, C.J.; Sherman, M.H.; Zhang, L.; Kremer, D.; Hwang, R.F.; Witkiewicz, A.K.; Ying, H. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 2016, 536, 479–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parker, S.J.; Amendola, C.R.; Hollinshead, K.E.; Yu, Q.; Yamamoto, K.; Encarnación-Rosado, J.; Rose, R.E.; LaRue, M.M.; Sohn, A.S.; Biancur, D.E. Selective alanine transporter utilization creates a targetable metabolic niche in pancreatic cancer. Cancer Discov. 2020, 10, 1018–1037. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Z.; Achreja, A.; Meurs, N.; Animasahun, O.; Owen, S.; Mittal, A.; Parikh, P.; Lo, T.-W.; Franco-Barraza, J.; Shi, J. Tumour-reprogrammed stromal BCAT1 fuels branched-chain ketoacid dependency in stromal-rich PDAC tumours. Nat. Metab. 2020, 2, 775–792. [Google Scholar] [CrossRef] [PubMed]
- Altan, B.; Kaira, K.; Watanabe, A.; Kubo, N.; Bao, P.; Dolgormaa, G.; Bilguun, E.-O.; Araki, K.; Kanai, Y.; Yokobori, T. Relationship between LAT1 expression and resistance to chemotherapy in pancreatic ductal adenocarcinoma. Cancer Chemother. Pharmacol. 2018, 81, 141–153. [Google Scholar] [CrossRef]
- Daher, B.; Parks, S.K.; Durivault, J.; Cormerais, Y.; Baidarjad, H.; Tambutte, E.; Pouysségur, J.; Vučetić, M. Genetic ablation of the cystine transporter xCT in PDAC cells inhibits mTORC1, growth, survival, and tumor formation via nutrient and oxidative stresses. Cancer Res. 2019, 79, 3877–3890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tao, P.; Wang, H.; Gao, G. CYP2J2-produced epoxyeicosatrienoic acids contribute to the ferroptosis resistance of pancreatic ductal adenocarcinoma in a PPAR γ-dependent manner. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2021, 46, 932–941. [Google Scholar] [CrossRef]
- Dai, E.; Han, L.; Liu, J.; Xie, Y.; Kroemer, G.; Klionsky, D.J.; Zeh, H.J.; Kang, R.; Wang, J.; Tang, D. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy 2020, 16, 2069–2083. [Google Scholar] [CrossRef] [PubMed]
- Dai, E.; Han, L.; Liu, J.; Xie, Y.; Zeh, H.J.; Kang, R.; Bai, L.; Tang, D. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat. Commun. 2020, 11, 1–11. [Google Scholar] [CrossRef]
- Zhu, S.; Zhang, Q.; Sun, X.; Zeh, H.J.; Lotze, M.T.; Kang, R.; Tang, D. HSPA5 regulates ferroptotic cell death in cancer cells. Cancer Res. 2017, 77, 2064–2077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, Y.; Zhu, S.; Zhong, M.; Yang, M.; Sun, X.; Liu, J.; Kroemer, G.; Lotze, M.; Zeh III, H.J.; Kang, R. Inhibition of Aurora kinase A induces necroptosis in pancreatic carcinoma. Gastroenterology 2017, 153, 1429–1443.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eling, N.; Reuter, L.; Hazin, J.; Hamacher-Brady, A.; Brady, N.R. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience 2015, 2, 517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Zhang, Y.; Liu, J.; Kang, R.; Klionsky, D.J.; Tang, D. Mitochondrial DNA stress triggers autophagy-dependent ferroptotic death. Autophagy 2021, 17, 948–960. [Google Scholar] [CrossRef]
- Tang, R.; Wu, Z.; Rong, Z.; Xu, J.; Wang, W.; Zhang, B.; Yu, X.; Shi, S. Ferroptosis-related lncRNA pairs to predict the clinical outcome and molecular characteristics of pancreatic ductal adenocarcinoma. Brief. Bioinform. 2021. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.; Santofimia-Castaño, P.; Liu, X.; Xia, Y.; Peng, L.; Gotorbe, C.; Neira, J.L.; Tang, D.; Pouyssegur, J.; Iovanna, J. NUPR1 inhibitor ZZW-115 induces ferroptosis in a mitochondria-dependent manner. Cell Death Discov. 2021, 7, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Kuang, F.; Liu, J.; Xie, Y.; Tang, D.; Kang, R. MGST1 is a redox-sensitive repressor of ferroptosis in pancreatic cancer cells. Cell Chem. Biol. 2021, 28, 765–775.e5. [Google Scholar] [CrossRef]
- Liu, J.; Dai, E.; Kang, R.; Kroemer, G.; Tang, D. The dark side of ferroptosis in pancreatic cancer. Oncoimmunology 2021, 10, 1868691. [Google Scholar] [CrossRef]
- Chen, X.; Kang, R.; Kroemer, G.; Tang, D. Targeting ferroptosis in pancreatic cancer: A double-edged sword. Trends Cancer 2021. [Google Scholar] [CrossRef]
- Zaidi, N.; Lupien, L.; Kuemmerle, N.B.; Kinlaw, W.B.; Swinnen, J.V.; Smans, K. Lipogenesis and lipolysis: The pathways exploited by the cancer cells to acquire fatty acids. Prog. Lipid Res. 2013, 52, 585–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beloribi-Djefaflia, S.; Vasseur, S.; Guillaumond, F. Lipid metabolic reprogramming in cancer cells. Oncogenesis 2016, 5, e189. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y. Fatty acid oxidation is a dominant bioenergetic pathway in prostate cancer. Prostate Cancer Prostatic Dis. 2006, 9, 230–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, Y.; Temkin, S.M.; Hawkridge, A.M.; Guo, C.; Wang, W.; Wang, X.-Y.; Fang, X. Fatty acid oxidation: An emerging facet of metabolic transformation in cancer. Cancer Lett. 2018, 435, 92–100. [Google Scholar] [CrossRef] [PubMed]
- Reyes-Castellanos, G.; Masoud, R.; Carrier, A. Mitochondrial metabolism in PDAC: From better knowledge to new targeting strategies. Biomedicines 2020, 8, 270. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Zhao, X.; Ouyang, H.; Sun, F.; Zhang, H.; Zhou, C.; Shen, H. The metabolic features of normal pancreas and pancreatic adenocarcinoma: Preliminary result of in vivo proton magnetic resonance spectroscopy at 3.0 T. J. Comput. Assist. Tomogr. 2011, 35, 539–543. [Google Scholar] [CrossRef]
- Guillaumond, F.; Bidaut, G.; Ouaissi, M.; Servais, S.; Gouirand, V.; Olivares, O.; Lac, S.; Borge, L.; Roques, J.; Gayet, O. Cholesterol uptake disruption, in association with chemotherapy, is a promising combined metabolic therapy for pancreatic adenocarcinoma. Proc. Natl. Acad. Sci. USA 2015, 112, 2473–2478. [Google Scholar] [CrossRef] [Green Version]
- Philip, B.; Roland, C.L.; Daniluk, J.; Liu, Y.; Chatterjee, D.; Gomez, S.B.; Ji, B.; Huang, H.; Wang, H.; Fleming, J.B. A high-fat diet activates oncogenic Kras and COX2 to induce development of pancreatic ductal adenocarcinoma in mice. Gastroenterology 2013, 145, 1449–1458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, F.; Kumagai-Braesch, M.; Herrington, M.K.; Larsson, J.; Permert, J. Increased lipid metabolism and cell turnover of MiaPaCa2 cells induced by high-fat diet in an orthotopic system. Metabolism 2009, 58, 1131–1136. [Google Scholar] [CrossRef] [PubMed]
- Kamphorst, J.J.; Cross, J.R.; Fan, J.; De Stanchina, E.; Mathew, R.; White, E.P.; Thompson, C.B.; Rabinowitz, J.D. Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. Proc. Natl. Acad. Sci. USA 2013, 110, 8882–8887. [Google Scholar] [CrossRef] [Green Version]
- Bian, Y.; Yu, Y.; Wang, S.; Li, L. Up-regulation of fatty acid synthase induced by EGFR/ERK activation promotes tumor growth in pancreatic cancer. Biochem. Biophys. Res. Commun. 2015, 463, 612–617. [Google Scholar] [CrossRef]
- Sunami, Y.; Rebelo, A.; Kleeff, J. Lipid metabolism and lipid droplets in pancreatic cancer and stellate cells. Cancers 2018, 10, 3. [Google Scholar] [CrossRef] [Green Version]
- Schug, Z.T.; Peck, B.; Jones, D.T.; Zhang, Q.; Grosskurth, S.; Alam, I.S.; Goodwin, L.M.; Smethurst, E.; Mason, S.; Blyth, K. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell 2015, 27, 57–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Icard, P.; Poulain, L.; Lincet, H. Understanding the central role of citrate in the metabolism of cancer cells. Biochim. Biophys. Acta (BBA)—Rev. Cancer 2012, 1825, 111–116. [Google Scholar] [CrossRef] [PubMed]
- Schug, Z.T.; Voorde, J.V.; Gottlieb, E. The metabolic fate of acetate in cancer. Nat. Rev. Cancer 2016, 16, 708–717. [Google Scholar] [CrossRef] [PubMed]
- Jeon, J.Y.; Lee, M.; Whang, S.H.; Kim, J.-W.; Cho, A.; Yun, M. Regulation of acetate utilization by monocarboxylate transporter 1 (MCT1) in hepatocellular carcinoma (HCC). Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2018, 26, 71–81. [Google Scholar] [CrossRef] [PubMed]
- Parks, S.K.; Chiche, J.; Pouysségur, J. Disrupting proton dynamics and energy metabolism for cancer therapy. Nat. Rev. Cancer 2013, 13, 611. [Google Scholar] [CrossRef]
- Bulusu, V.; Tumanov, S.; Michalopoulou, E.; van den Broek, N.J.; MacKay, G.; Nixon, C.; Dhayade, S.; Schug, Z.T.; Voorde, J.V.; Blyth, K. Acetate recapturing by nuclear acetyl-CoA synthetase 2 prevents loss of histone acetylation during oxygen and serum limitation. Cell Rep. 2017, 18, 647–658. [Google Scholar] [CrossRef] [Green Version]
- Nishi, K.; Suzuki, K.; Sawamoto, J.; Tokizawa, Y.; Iwase, Y.; Yumita, N.; Ikeda, T. Inhibition of fatty acid synthesis induces apoptosis of human pancreatic cancer cells. Anticancer Res. 2016, 36, 4655–4660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bracci, P.M. Obesity and pancreatic cancer: Overview of epidemiologic evidence and biologic mechanisms. Mol. Carcinog. 2012, 51, 53–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohammed, A.; Janakiram, N.B.; Brewer, M.; Duff, A.; Lightfoot, S.; Brush, R.S.; Anderson, R.E.; Rao, C.V. Endogenous n-3 polyunsaturated fatty acids delay progression of pancreatic ductal adenocarcinoma in Fat-1-p48Cre/+-LSL-KrasG12D/+ mice. Neoplasia 2012, 14, 1249–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duckwall, C.S.; Murphy, T.A.; Young, J.D. Mapping cancer cell metabolism with13C flux analysis: Recent progress and future challenges. J. Carcinog. 2013, 12, 13. [Google Scholar] [CrossRef] [PubMed]
- Kinnaird, A.; Zhao, S.; Wellen, K.E.; Michelakis, E.D. Metabolic control of epigenetics in cancer. Nat. Rev. Cancer 2016, 16, 694–707. [Google Scholar] [CrossRef]
- Wang, J.; Ye, C.; Chen, C.; Xiong, H.; Xie, B.; Zhou, J.; Chen, Y.; Zheng, S.; Wang, L. Glucose transporter GLUT1 expression and clinical outcome in solid tumors: A systematic review and meta-analysis. Oncotarget 2017, 8, 16875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharen, G.; Peng, Y.; Cheng, H.; Liu, Y.; Shi, Y.; Zhao, J. Prognostic value of GLUT-1 expression in pancreatic cancer: Results from 538 patients. Oncotarget 2017, 8, 19760. [Google Scholar] [CrossRef] [PubMed]
- Lu, K.; Yang, J.; Li, D.C.; He, S.B.; Zhu, D.M.; Zhang, L.F.; Zhang, X.; Chen, X.C.; Zhang, B.; Zhou, J. Expression and clinical significance of glucose transporter-1 in pancreatic cancer. Oncol. Lett. 2016, 12, 243–249. [Google Scholar] [CrossRef] [Green Version]
- Davis-Yadley, A.H.; Abbott, A.M.; Pimiento, J.M.; Chen, D.-T.; Malafa, M.P. Increased Expression of the GLUT-1 Gene is Associated With Worse Overall Survival in Resected Pancreatic Adenocarcinoma. Pancreas 2016, 45, 974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, X.; Lu, P.; Zhou, S.; Zhang, L.; Zhao, J.-h.; Tang, J.-h. Predictive value of glucose transporter-1 and glucose transporter-3 for survival of cancer patients: A meta-analysis. Oncotarget 2017, 8, 13206. [Google Scholar] [CrossRef] [PubMed]
- Madunić, J.; Madunić, I.V.; Gajski, G.; Popić, J.; Garaj-Vrhovac, V. Apigenin: A dietary flavonoid with diverse anticancer properties. Cancer Lett. 2018, 413, 11–22. [Google Scholar] [CrossRef]
- Melstrom, L.G.; Salabat, M.R.; Ding, X.-Z.; Strouch, M.J.; Grippo, P.J.; Mirzoeva, S.; Pelling, J.C.; Bentrem, D.J. Apigenin down-regulates the hypoxia response genes: HIF-1α, GLUT-1, and VEGF in human pancreatic cancer cells. J. Surg. Res. 2011, 167, 173–181. [Google Scholar] [CrossRef] [PubMed]
- Melstrom, L.G.; Salabat, M.R.; Ding, X.-Z.; Milam, B.M.; Strouch, M.; Pelling, J.C.; Bentrem, D.J. Apigenin inhibits the GLUT-1 glucose transporter and the phosphoinositide 3-kinase/Akt pathway in human pancreatic cancer cells. Pancreas 2008, 37, 426–431. [Google Scholar] [CrossRef] [PubMed]
- Ashrafizadeh, M.; Bakhoda, M.R.; Bahmanpour, Z.; Ilkhani, K.; Zarrabi, A.; Makvandi, P.; Khan, H.; Mazaheri, S.; Darvish, M.; Mirzaei, H. Apigenin as Tumor Suppressor in Cancers: Biotherapeutic Activity, Nanodelivery, and Mechanisms with Emphasis on Pancreatic Cancer. Front. Chem. 2020, 8. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Ren, B.; Yang, G.; Wang, H.; Chen, G.; You, L.; Zhang, T.; Zhao, Y. The enhancement of glycolysis regulates pancreatic cancer metastasis. Cell. Mol. Life Sci. 2020, 77, 305–321. [Google Scholar] [CrossRef]
- Anderson, M.; Marayati, R.; Moffitt, R.; Yeh, J.J. Hexokinase 2 promotes tumor growth and metastasis by regulating lactate production in pancreatic cancer. Oncotarget 2017, 8, 56081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Min, J.W.; Kim, K.I.; Kim, H.-A.; Kim, E.-K.; Noh, W.C.; Jeon, H.B.; Cho, D.-H.; Oh, J.S.; Park, I.-C.; Hwang, S.-G. INPP4B-mediated tumor resistance is associated with modulation of glucose metabolism via hexokinase 2 regulation in laryngeal cancer cells. Biochem. Biophys. Res. Commun. 2013, 440, 137–142. [Google Scholar] [CrossRef] [PubMed]
- Gatenby, R.A.; Gillies, R.J. Glycolysis in cancer: A potential target for therapy. Int. J. Biochem. Cell Biol. 2007, 39, 1358–1366. [Google Scholar] [CrossRef] [PubMed]
- Morten, K.J.; Badder, L.; Knowles, H.J. Differential regulation of HIF-mediated pathways increases mitochondrial metabolism and ATP production in hypoxic osteoclasts. J. Pathol. 2013, 229, 755–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Isayev, O.; Rausch, V.; Bauer, N.; Liu, L.; Fan, P.; Zhang, Y.; Gladkich, J.; Nwaeburu, C.C.; Mattern, J.; Mollenhauer, M. Inhibition of glucose turnover by 3-bromopyruvate counteracts pancreatic cancer stem cell features and sensitizes cells to gemcitabine. Oncotarget 2014, 5, 5177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ota, S.; Geschwind, J.-F.H.; Buijs, M.; Wijlemans, J.W.; Kwak, B.K.; Ganapathy-Kanniappan, S. Ultrasound-guided direct delivery of 3-bromopyruvate blocks tumor progression in an orthotopic mouse model of human pancreatic cancer. Target. Oncol. 2013, 8, 145–151. [Google Scholar] [CrossRef] [PubMed]
- Chan, A.K.; Bruce, J.I.; Siriwardena, A.K. Glucose metabolic phenotype of pancreatic cancer. World J. Gastroenterol. 2016, 22, 3471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jang, M.; Kang, H.J.; Lee, S.Y.; Chung, S.J.; Kang, S.; Chi, S.W.; Cho, S.; Lee, S.C.; Lee, C.-K.; Park, B.C. Glyceraldehyde-3-phosphate, a glycolytic intermediate, plays a key role in controlling cell fate via inhibition of caspase activity. Mol. Cells 2009, 28, 559–563. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Jiang, F.; Ge, Z.; Chen, B.; Yu, J.; Xin, M.; Wang, J.; An, L.; Wei, J.; Wu, L. Fructose-bisphosphate aldolase a regulates hypoxic adaptation in hepatocellular carcinoma and involved with tumor malignancy. Dig. Dis. Sci. 2019, 64, 3215–3227. [Google Scholar] [CrossRef] [PubMed]
- Yu, S.-L.; Xu, L.-T.; Qi, Q.; Geng, Y.-W.; Chen, H.; Meng, Z.-Q.; Wang, P.; Chen, Z. Serum lactate dehydrogenase predicts prognosis and correlates with systemic inflammatory response in patients with advanced pancreatic cancer after gemcitabine-based chemotherapy. Sci. Rep. 2017, 7, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Augoff, K.; Hryniewicz-Jankowska, A.; Tabola, R. Lactate dehydrogenase 5: An old friend and a new hope in the war on cancer. Cancer Lett. 2015, 358, 1–7. [Google Scholar] [CrossRef]
- Kolev, Y.; Uetake, H.; Takagi, Y.; Sugihara, K. Lactate dehydrogenase-5 (LDH-5) expression in human gastric cancer: Association with hypoxia-inducible factor (HIF-1α) pathway, angiogenic factors production and poor prognosis. Ann. Surg. Oncol. 2008, 15, 2336–2344. [Google Scholar] [CrossRef]
- Maftouh, M.; Avan, A.; Sciarrillo, R.; Granchi, C.; Leon, L.; Rani, R.; Funel, N.; Smid, K.; Honeywell, R.; Boggi, U. Synergistic interaction of novel lactate dehydrogenase inhibitors with gemcitabine against pancreatic cancer cells in hypoxia. Br. J. Cancer 2014, 110, 172–182. [Google Scholar] [CrossRef] [PubMed]
- Le, A.; Cooper, C.R.; Gouw, A.M.; Dinavahi, R.; Maitra, A.; Deck, L.M.; Royer, R.E.; Vander Jagt, D.L.; Semenza, G.L.; Dang, C.V. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc. Natl. Acad. Sci. USA 2010, 107, 2037–2042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohammad, G.H.; Vassileva, V.; Acedo, P.; Olde Damink, S.W.; Malago, M.; Dhar, D.K.; Pereira, S.P. Targeting pyruvate kinase M2 and lactate dehydrogenase a is an effective combination strategy for the treatment of pancreatic cancer. Cancers 2019, 11, 1372. [Google Scholar] [CrossRef] [Green Version]
- Israelsen, W.J.; Vander Heiden, M.G. Pyruvate kinase: Function, regulation and role in cancer. Semin. Cell Dev. Biol. 2015, 43, 43–51. [Google Scholar] [CrossRef] [Green Version]
- Gupta, V.; Bamezai, R.N. Human pyruvate kinase M2: A multifunctional protein. Protein Sci. 2010, 19, 2031–2044. [Google Scholar] [CrossRef] [Green Version]
- Li, C.; Zhao, Z.; Zhou, Z.; Liu, R. PKM2 promotes cell survival and invasion under metabolic stress by enhancing Warburg effect in pancreatic ductal adenocarcinoma. Dig. Dis. Sci. 2016, 61, 767–773. [Google Scholar] [CrossRef]
- Yokoyama, M.; Tanuma, N.; Shibuya, R.; Shiroki, T.; Abue, M.; Yamamoto, K.; Miura, K.; Yamaguchi, K.; Sato, I.; Tamai, K. Pyruvate kinase type M2 contributes to the development of pancreatic ductal adenocarcinoma by regulating the production of metabolites and reactive oxygen species. Int. J. Oncol. 2018, 52, 881–891. [Google Scholar] [CrossRef] [Green Version]
- Tian, S.; Li, P.; Sheng, S.; Jin, X. Upregulation of pyruvate kinase M2 expression by fatty acid synthase contributes to gemcitabine resistance in pancreatic cancer. Oncol. Lett. 2018, 15, 2211–2217. [Google Scholar] [CrossRef] [Green Version]
- Van Veelen, C.W.; Verbiest, H.; Vlug, A.M.; Rijksen, G.; Staal, G.E. Isozymes of pyruvate kinase from human brain, meningiomas, and malignant gliomas. Cancer Res. 1978, 38, 4681–4687. [Google Scholar]
- Kim, D.J.; Park, Y.S.; Kang, M.G.; You, Y.-M.; Jung, Y.; Koo, H.; Kim, J.-A.; Kim, M.-J.; Hong, S.-M.; Lee, K.B. Pyruvate kinase isoenzyme M2 is a therapeutic target of gemcitabine-resistant pancreatic cancer cells. Exp. Cell Res. 2015, 336, 119–129. [Google Scholar] [CrossRef] [PubMed]
- Lee, D.C.; Sohn, H.A.; Park, Z.-Y.; Oh, S.; Kang, Y.K.; Lee, K.-m.; Kang, M.; Jang, Y.J.; Yang, S.-J.; Hong, Y.K. A lactate-induced response to hypoxia. Cell 2015, 161, 595–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S. Inhibitory effect of tumor cell–derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef]
- Baek, G.; Yan, F.T.; Hu, Z.; Cox, D.; Buboltz, N.; McCue, P.; Yeo, C.J.; White, M.A.; DeBerardinis, R.J.; Knudsen, E.S. MCT4 defines a glycolytic subtype of pancreatic cancer with poor prognosis and unique metabolic dependencies. Cell Rep. 2014, 9, 2233–2249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Porporato, P.E.; Dadhich, R.K.; Dhup, S.; Copetti, T.; Sonveaux, P. Anticancer targets in the glycolytic metabolism of tumors: A comprehensive review. Front. Pharmacol. 2011, 2, 49. [Google Scholar] [CrossRef] [Green Version]
- Lai, I.-L.; Chou, C.-C.; Lai, P.-T.; Fang, C.-S.; Shirley, L.A.; Yan, R.; Mo, X.; Bloomston, M.; Kulp, S.K.; Bekaii-Saab, T. Targeting the Warburg effect with a novel glucose transporter inhibitor to overcome gemcitabine resistance in pancreatic cancer cells. Carcinogenesis 2014, 35, 2203. [Google Scholar] [CrossRef] [Green Version]
- Noushmehr, H.; D’Amico, E.; Farilla, L.; Hui, H.; Wawrowsky, K.A.; Mlynarski, W.; Doria, A.; Abumrad, N.A.; Perfetti, R. Fatty acid translocase (FAT/CD36) is localized on insulin-containing granules in human pancreatic β-cells and mediates fatty acid effects on insulin secretion. Diabetes 2005, 54, 472–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kubo, M.; Gotoh, K.; Eguchi, H.; Kobayashi, S.; Iwagami, Y.; Tomimaru, Y.; Akita, H.; Asaoka, T.; Noda, T.; Takeda, Y. Impact of CD36 on chemoresistance in pancreatic ductal adenocarcinoma. Ann. Surg. Oncol. 2020, 27, 610–619. [Google Scholar] [CrossRef] [PubMed]
- Alo, P.L.; Amini, M.; Piro, F.; Pizzuti, L.; Sebastiani, V.; Botti, C.; Murari, R.; Zotti, G.; Di Tondo, U. Immnunohistochemical expression and prognostic significance of fatty acid synthase in pancreatic carcinoma. Anticancer Res. 2007, 27, 2523–2527. [Google Scholar] [PubMed]
- Walter, K.; Hong, S.-M.; Nyhan, S.; Canto, M.; Fedarko, N.; Klein, A.; Griffith, M.; Omura, N.; Medghalchi, S.; Kuhajda, F. Serum fatty acid synthase as a marker of pancreatic neoplasia. Cancer Epidemiol. Prev. Biomark. 2009, 18, 2380–2385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Y.; Li, H.; Li, Z.; Zhao, Z.; Yip-Schneider, M.; Fan, Q.; Schmidt, C.M.; Chiorean, E.G.; Xie, J.; Cheng, L. Role of fatty acid synthase in gemcitabine and radiation resistance of pancreatic cancers. Int. J. Biochem. Mol. Biol. 2011, 2, 89. [Google Scholar]
- Zhao, Y.; Butler, E.B.; Tan, M. Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis. 2013, 4, e532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, J.; Hong, Y.; Tao, X.; Wei, X.; Zhang, L.; Li, Q. An indispensable role of CPT-1a to survive cancer cells during energy stress through rewiring cancer metabolism. Tumor Biol. 2016, 37, 15795–15804. [Google Scholar] [CrossRef]
- Butler, E.B.; Zhao, Y.; Muñoz-Pinedo, C.; Lu, J.; Tan, M. Stalling the engine of resistance: Targeting cancer metabolism to overcome therapeutic resistance. Cancer Res. 2013, 73, 2709–2717. [Google Scholar] [CrossRef] [Green Version]
- Whatcott, C.; Han, H.; Posner, R.G.; Von Hoff, D.D. Tumor-stromal interactions in pancreatic cancer. Crit. Rev. Oncog. 2013, 18, 135–151. [Google Scholar] [CrossRef]
- Banerjee, S.; Modi, S.; McGinn, O.; Zhao, X.; Dudeja, V.; Ramakrishnan, S.; Saluja, A.K. Impaired synthesis of stromal components in response to minnelide improves vascular function, drug delivery, and survival in pancreatic cancer. Clin. Cancer Res. 2016, 22, 415–425. [Google Scholar] [CrossRef] [Green Version]
- Kordes, S.; Pollak, M.N.; Zwinderman, A.H.; Mathôt, R.A.; Weterman, M.J.; Beeker, A.; Punt, C.J.; Richel, D.J.; Wilmink, J.W. Metformin in patients with advanced pancreatic cancer: A double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol. 2015, 16, 839–847. [Google Scholar] [CrossRef]
- Alistar, A.; Morris, B.B.; Desnoyer, R.; Klepin, H.D.; Hosseinzadeh, K.; Clark, C.; Cameron, A.; Leyendecker, J.; D’Agostino, R., Jr.; Topaloglu, U. Safety and tolerability of the first-in-class agent CPI-613 in combination with modified FOLFIRINOX in patients with metastatic pancreatic cancer: A single-centre, open-label, dose-escalation, phase 1 trial. Lancet Oncol. 2017, 18, 770–778. [Google Scholar] [CrossRef]
Drug | Refs | Effects |
---|---|---|
All trans-retinoic acid (ATRA) | [100,101,102] | ATRA inhibits the activation of stellate cells. |
Pirfenidone | [103,104,105,106,107,108,109,110,111,112,113,114,115] | Inhibits collagen fibrils assembly; downregulates the intercellular adhesion molecule-1 (ICAM1); decreases the transformation grow factor beta (TGFβ) at the translational level; down-regulates the pro-fibrotic hedgehog signaling pathway; decreases fibroblast proliferation; blocks myofibroblast differentiation; suppresses tumor necrosis factor alpha (TNFα); decreases cell migration-inducing and hyaluronan-binding proteins. |
Candesartan | [116] | Angiotensin II receptor inhibitor, which consequently leads to the reduction in stellate cell proliferation. |
Olmesartan | [29] | Angiotensin II receptor inhibitor, which consequently leads to the reduction in stellate cell proliferation. |
Saridegib (IPI-926) | [98,117,118] | Hedgehog signaling inhibition. |
Vismodegib | [119] | Hedgehog signaling inhibition. |
4-methyl umbelliferone (4MU) | [85,86,120] | Inhibition of hyaluronan synthase, decreases hyaluronan synthesis; Synergistic activity with gemcitabine. |
Curcumin | [121,122,123,124] | Inhibits activation of stellate cells. |
L49H37 a curcumin synthetic analog | [125] | Stellate cell inhibitor. |
Rhein (natural anthraquinone derivative) | [126,127] | Anti-fibrotic action in PDAC. Reduces collagen I and fibronectin. |
Resveratrol | [128] | Impedes stellate cell activation by downregulating miRNA 21. This miRNA is also a participant in gemcitabine resistance. |
Emodin | [129,130,131,132] | Emodin has a wide spectrum of activities related with anti-cancer effects and anti-fibrotic actions. |
Ellagic acid | [133] | Inhibits the activation and proliferation of stellate cells. |
Imatinib | [134,135,136,137,138,139,140,141] | Imatinib is anti-fibrotic in pulmonary-induced fibrosis by bleomycin. It is also anti-fibrotic in breast cancer and the liver. However, in a clinical trial of imatinib associated with gemcitabine it did not show any benefits. |
Metformin | [142,143] | Suppresses desmoplasia by activating AMPK and enhances gemcitabine chemosensitivity. |
Halofuginone | [144] | Halofuginone is an analog of quinazolinone that shows strong anti-fibrotic properties in an experimental PDAC model. It inhibits the activation of stellate cells. |
Pegylated recombinant human hyaluronidase | [145] | Acts by enzymatic degradation of hyaluronate. This device can incorporate chemo drugs including checkpoint inhibitors. Research is ongoing. |
Fasudil priming before chemotherapy | [146,147,148,149,150] | Fasudil is a Rho kinase inhibitor. Administered before chemotherapy it decreased stromal density allowing a better level of drug at the tumor. |
Pentoxiphyllin | [151,152,153] | Pentoxiphyllin is a reducer of blood viscosity and cytokine production, including TNFα, IL-6, and IL-8 with anti-inflammatory effects and with clear anti-fibrotic effects. |
Dasatinib | [154] | Dasatinib decreased pancreatic fibrosis in an experimental model of pancreatitis. |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Carvalho, T.M.A.; Di Molfetta, D.; Greco, M.R.; Koltai, T.; Alfarouk, K.O.; Reshkin, S.J.; Cardone, R.A. Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches. Cancers 2021, 13, 6135. https://doi.org/10.3390/cancers13236135
Carvalho TMA, Di Molfetta D, Greco MR, Koltai T, Alfarouk KO, Reshkin SJ, Cardone RA. Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches. Cancers. 2021; 13(23):6135. https://doi.org/10.3390/cancers13236135
Chicago/Turabian StyleCarvalho, Tiago M. A., Daria Di Molfetta, Maria Raffaella Greco, Tomas Koltai, Khalid O. Alfarouk, Stephan J. Reshkin, and Rosa A. Cardone. 2021. "Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches" Cancers 13, no. 23: 6135. https://doi.org/10.3390/cancers13236135
APA StyleCarvalho, T. M. A., Di Molfetta, D., Greco, M. R., Koltai, T., Alfarouk, K. O., Reshkin, S. J., & Cardone, R. A. (2021). Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches. Cancers, 13(23), 6135. https://doi.org/10.3390/cancers13236135