Metabolic Pathways as a Novel Landscape in Pancreatic Ductal Adenocarcinoma
Abstract
:Simple Summary
Abstract
1. Introduction
2. Metabolic Reprogramming of the Main Energy Pathways in PDAC
2.1. Activation and Maintenance of Glycoltyc Flux
2.2. Amino Acids as an External Energy Resource
2.3. Fatty Acids Contribute to PDAC Progression
3. Immune Cells and Metabolic Response in PDAC Microenvironment
4. Clinical Perspectives
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
References
- 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] [PubMed]
- Rawla, P.; Sunkara, T.; Gaduputi, V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J. Oncol. 2019, 10, 10–27. [Google Scholar] [CrossRef]
- Kamisawa, T.; Wood, L.D.; Itoi, T.; Takaori, K. Pancreatic cancer. Lancet 2016, 388, 73–85. [Google Scholar] [CrossRef]
- Stathis, A.; Moore, M.J. Advanced pancreatic carcinoma: Current treatment and future challenges. Nat. Rev. Clin. Oncol. 2010, 7, 163–172. [Google Scholar] [CrossRef]
- Stokes, J.B.; Nolan, N.J.; Stelow, E.B.; Walters, D.M.; Weiss, G.R.; de Lange, E.E.; Rich, T.A.; Adams, R.B.; Bauer, T.W. Preoperative capecitabine and concurrent radiation for borderline resectable pancreatic cancer. Ann. Surg. Oncol. 2011, 18, 619–627. [Google Scholar] [CrossRef] [PubMed]
- Sugawara, T.; Ban, D.; Nishino, J.; Watanabe, S.; Maekawa, A.; Ishikawa, Y.; Akahoshi, K.; Ogawa, K.; Ono, H.; Kudo, A.; et al. Prediction of early recurrence of pancreatic ductal adenocarcinoma after resection. PLoS ONE 2021, 16, e0249885. [Google Scholar] [CrossRef] [PubMed]
- Shen, G.Q.; Aleassa, E.M.; Walsh, R.M.; Morris-Stiff, G. Next-Generation Sequencing in Pancreatic Cancer. Pancreas 2019, 48, 739–748. [Google Scholar] [CrossRef] [PubMed]
- Mueller, S.; Engleitner, T.; Maresch, R.; Zukowska, M.; Lange, S.; Kaltenbacher, T.; Konukiewitz, B.; Ollinger, R.; Zwiebel, M.; Strong, A.; et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature 2018, 554, 62–68. [Google Scholar] [CrossRef]
- Waters, A.M.; Der, C.J. KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb. Perspect Med. 2018, 8, a031435. [Google Scholar] [CrossRef]
- Scarpa, A.; Capelli, P.; Mukai, K.; Zamboni, G.; Oda, T.; Iacono, C.; Hirohashi, S. Pancreatic adenocarcinomas frequently show p53 gene mutations. Am. J. Pathol. 1993, 142, 1534–1543. [Google Scholar]
- Maddalena, M.; Mallel, G.; Nataraj, N.B.; Shreberk-Shaked, M.; Hassin, O.; Mukherjee, S.; Arandkar, S.; Rotkopf, R.; Kapsack, A.; Lambiase, G.; et al. TP53 missense mutations in PDAC are associated with enhanced fibrosis and an immunosuppressive microenvironment. Proc. Natl. Acad. Sci. USA 2021, 118, e2025631118. [Google Scholar] [CrossRef]
- Sikdar, N.; Saha, G.; Dutta, A.; Ghosh, S.; Shrikhande, S.V.; Banerjee, S. Genetic Alterations of Periampullary and Pancreatic Ductal Adenocarcinoma: An Overview. Curr. Genom. 2018, 19, 444–463. [Google Scholar] [CrossRef]
- Bardeesy, N.; Cheng, K.H.; Berger, J.H.; Chu, G.C.; Pahler, J.; Olson, P.; Hezel, A.F.; Horner, J.; Lauwers, G.Y.; Hanahan, D.; et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev. 2006, 20, 3130–3146. [Google Scholar] [CrossRef] [Green Version]
- Crane, C.H.; Varadhachary, G.R.; Yordy, J.S.; Staerkel, G.A.; Javle, M.M.; Safran, H.; Haque, W.; Hobbs, B.D.; Krishnan, S.; Fleming, J.B.; et al. Phase II trial of cetuximab, gemcitabine, and oxaliplatin followed by chemoradiation with cetuximab for locally advanced (T4) pancreatic adenocarcinoma: Correlation of Smad4(Dpc4) immunostaining with pattern of disease progression. J. Clin. Oncol. 2011, 29, 3037–3043. [Google Scholar] [CrossRef] [Green Version]
- Ilic, M.; Ilic, I. Epidemiology of pancreatic cancer. World J. Gastroenterol. 2016, 22, 9694–9705. [Google Scholar] [CrossRef]
- Ansari, D.; Tingstedt, B.; Andersson, B.; Holmquist, F.; Sturesson, C.; Williamsson, C.; Sasor, A.; Borg, D.; Bauden, M.; Andersson, R. Pancreatic cancer: Yesterday, today and tomorrow. Future Oncol 2016, 12, 1929–1946. [Google Scholar] [CrossRef] [Green Version]
- Collisson, E.A.; Bailey, P.; Chang, D.K.; Biankin, A.V. Molecular subtypes of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 207–220. [Google Scholar] [CrossRef]
- Huxley, R.; Ansary-Moghaddam, A.; Berrington de Gonzalez, A.; Barzi, F.; Woodward, M. Type-II diabetes and pancreatic cancer: A meta-analysis of 36 studies. Br. J. Cancer 2005, 92, 2076–2083. [Google Scholar] [CrossRef] [Green Version]
- Bosetti, C.; Rosato, V.; Li, D.; Silverman, D.; Petersen, G.M.; Bracci, P.M.; Neale, R.E.; Muscat, J.; Anderson, K.; Gallinger, S.; et al. Diabetes, antidiabetic medications, and pancreatic cancer risk: An analysis from the International Pancreatic Cancer Case-Control Consortium. Ann. Oncol. 2014, 25, 2065–2072. [Google Scholar] [CrossRef]
- Olson, S.H.; Xu, Y.; Herzog, K.; Saldia, A.; DeFilippis, E.M.; Li, P.; Allen, P.J.; O’Reilly, E.M.; Kurtz, R.C. Weight Loss, Diabetes, Fatigue, and Depression Preceding Pancreatic Cancer. Pancreas 2016, 45, 986–991. [Google Scholar] [CrossRef] [Green Version]
- Jeon, C.Y.; Kim, S.; Lin, Y.C.; Risch, H.A.; Goodarzi, M.O.; Nuckols, T.K.; Freedland, S.J.; Pandol, S.J.; Pisegna, J.R. Prediction of Pancreatic Cancer in Diabetes Patients with Worsening Glycemic Control. Cancer Epidemiol. Biomark. Prev. 2022, 31, 242–253. [Google Scholar] [CrossRef]
- Bodmer, M.; Becker, C.; Meier, C.; Jick, S.S.; Meier, C.R. Use of antidiabetic agents and the risk of pancreatic cancer: A case-control analysis. Am. J. Gastroenterol. 2012, 107, 620–626. [Google Scholar] [CrossRef]
- Colmers, I.N.; Bowker, S.L.; Tjosvold, L.A.; Johnson, J.A. Insulin use and cancer risk in patients with type 2 diabetes: A systematic review and meta-analysis of observational studies. Diabetes Metab. 2012, 38, 485–506. [Google Scholar] [CrossRef]
- Vaziri-Gohar, A.; Zarei, M.; Brody, J.R.; Winter, J.M. Metabolic Dependencies in Pancreatic Cancer. Front. Oncol. 2018, 8, 617. [Google Scholar] [CrossRef]
- Schiliro, C.; Firestein, B.L. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells 2021, 10, 1056. [Google Scholar] [CrossRef]
- Biancur, D.E.; Kimmelman, A.C. The plasticity of pancreatic cancer metabolism in tumor progression and therapeutic resistance. Biochim. Biophys Acta Rev. Cancer 2018, 1870, 67–75. [Google Scholar] [CrossRef]
- 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.; et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 2016, 536, 479–483. [Google Scholar] [CrossRef] [Green Version]
- Ahmad, R.S.; Eubank, T.D.; Lukomski, S.; Boone, B.A. Immune Cell Modulation of the Extracellular Matrix Contributes to the Pathogenesis of Pancreatic Cancer. Biomolecules 2021, 11, 901. [Google Scholar] [CrossRef]
- Daemen, A.; Peterson, D.; Sahu, N.; McCord, R.; Du, X.; Liu, B.; Kowanetz, K.; Hong, R.; Moffat, J.; Gao, M.; et al. Metabolite profiling stratifies pancreatic ductal adenocarcinomas into subtypes with distinct sensitivities to metabolic inhibitors. Proc. Natl. Acad. Sci. USA 2015, 112, E4410–E4417. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; DeBerardinis, R.J. Mechanisms and Implications of Metabolic Heterogeneity in Cancer. Cell Metab. 2019, 30, 434–446. [Google Scholar] [CrossRef]
- Bernard, V.; Semaan, A.; Huang, J.; San Lucas, F.A.; Mulu, F.C.; Stephens, B.M.; Guerrero, P.A.; Huang, Y.; Zhao, J.; Kamyabi, N.; et al. Single-Cell Transcriptomics of Pancreatic Cancer Precursors Demonstrates Epithelial and Microenvironmental Heterogeneity as an Early Event in Neoplastic Progression. Clin. Cancer Res. 2019, 25, 2194–2205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, J.; Sun, B.F.; Chen, C.Y.; Zhou, J.Y.; Chen, Y.S.; Chen, H.; Liu, L.; Huang, D.; Jiang, J.; Cui, G.S.; et al. Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma. Cell Res. 2019, 29, 725–738. [Google Scholar] [CrossRef] [PubMed]
- Hausmann, S.; Kong, B.; Michalski, C.; Erkan, M.; Friess, H. The role of inflammation in pancreatic cancer. Adv. Exp. Med. Biol. 2014, 816, 129–151. [Google Scholar] [CrossRef] [PubMed]
- Palsson-McDermott, E.M.; O’Neill, L.A.J. Targeting immunometabolism as an anti-inflammatory strategy. Cell Res. 2020, 30, 300–314. [Google Scholar] [CrossRef] [Green Version]
- Cao, L.; Wu, J.; Qu, X.; Sheng, J.; Cui, M.; Liu, S.; Huang, X.; Xiang, Y.; Li, B.; Zhang, X.; et al. Glycometabolic rearrangements--aerobic glycolysis in pancreatic cancer: Causes, characteristics and clinical applications. J. Exp. Clin. Cancer Res. 2020, 39, 267. [Google Scholar] [CrossRef]
- Davidson, S.; Efremova, M.; Riedel, A.; Mahata, B.; Pramanik, J.; Huuhtanen, J.; Kar, G.; Vento-Tormo, R.; Hagai, T.; Chen, X.; et al. Single-Cell RNA Sequencing Reveals a Dynamic Stromal Niche That Supports Tumor Growth. Cell Rep. 2020, 31, 107628. [Google Scholar] [CrossRef]
- Assmann, N.; Finlay, D.K. Metabolic regulation of immune responses: Therapeutic opportunities. J. Clin. Investig. 2016, 126, 2031–2039. [Google Scholar] [CrossRef]
- Penny, H.L.; Sieow, J.L.; Gun, S.Y.; Lau, M.C.; Lee, B.; Tan, J.; Phua, C.; Toh, F.; Nga, Y.; Yeap, W.H.; et al. Targeting Glycolysis in Macrophages Confers Protection Against Pancreatic Ductal Adenocarcinoma. Int J. Mol. Sci 2021, 22, 6350. [Google Scholar] [CrossRef]
- Sharma, N.S.; Gupta, V.K.; Garrido, V.T.; Hadad, R.; Durden, B.C.; Kesh, K.; Giri, B.; Ferrantella, A.; Dudeja, V.; Saluja, A.; et al. Targeting tumor-intrinsic hexosamine biosynthesis sensitizes pancreatic cancer to anti-PD1 therapy. J. Clin. Investig. 2020, 130, 451–465. [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.; et al. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell 2012, 149, 656–670. [Google Scholar] [CrossRef] [Green Version]
- Yun, J.; Rago, C.; Cheong, I.; Pagliarini, R.; Angenendt, P.; Rajagopalan, H.; Schmidt, K.; Willson, J.K.; Markowitz, S.; Zhou, S.; et al. Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells. Science 2009, 325, 1555–1559. [Google Scholar] [CrossRef] [Green Version]
- Lau, A.N.; Li, Z.; Danai, L.V.; Westermark, A.M.; Darnell, A.M.; Ferreira, R.; Gocheva, V.; Sivanand, S.; Lien, E.C.; Sapp, K.M.; et al. Dissecting cell-type-specific metabolism in pancreatic ductal adenocarcinoma. Elife 2020, 9, e56782. [Google Scholar] [CrossRef]
- Kurahara, H.; Maemura, K.; Mataki, Y.; Sakoda, M.; Iino, S.; Kawasaki, Y.; Arigami, T.; Mori, S.; Kijima, Y.; Ueno, S.; et al. Significance of Glucose Transporter Type 1 (GLUT-1) Expression in the Therapeutic Strategy for Pancreatic Ductal Adenocarcinoma. Ann. Surg. Oncol. 2018, 25, 1432–1439. [Google Scholar] [CrossRef]
- Stopa, K.B.; Kusiak, A.A.; Szopa, M.D.; Ferdek, P.E.; Jakubowska, M.A. Pancreatic Cancer and Its Microenvironment-Recent Advances and Current Controversies. Int. J. Mol. Sci. 2020, 21, 3218. [Google Scholar] [CrossRef]
- Cox, T.R. The matrix in cancer. Nat. Rev. Cancer 2021, 21, 217–238. [Google Scholar] [CrossRef]
- Apte, M.V.; Wilson, J.S.; Lugea, A.; Pandol, S.J. A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology 2013, 144, 1210–1219. [Google Scholar] [CrossRef] [Green Version]
- Sinn, M.; Denkert, C.; Striefler, J.K.; Pelzer, U.; Stieler, J.M.; Bahra, M.; Lohneis, P.; Dorken, B.; Oettle, H.; Riess, H.; et al. alpha-Smooth muscle actin expression and desmoplastic stromal reaction in pancreatic cancer: Results from the CONKO-001 study. Br. J. Cancer 2014, 111, 1917–1923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tjomsland, V.; Spangeus, A.; Valila, J.; Sandstrom, P.; Borch, K.; Druid, H.; Falkmer, S.; Falkmer, U.; Messmer, D.; Larsson, M. Interleukin 1alpha sustains the expression of inflammatory factors in human pancreatic cancer microenvironment by targeting cancer-associated fibroblasts. Neoplasia 2011, 13, 664–675. [Google Scholar] [CrossRef] [Green Version]
- Zhuang, Z.; Ju, H.Q.; Aguilar, M.; Gocho, T.; Li, H.; Iida, T.; Lee, H.; Fan, X.; Zhou, H.; Ling, J.; et al. IL1 Receptor Antagonist Inhibits Pancreatic Cancer Growth by Abrogating NF-kappaB Activation. Clin. Cancer Res. 2016, 22, 1432–1444. [Google Scholar] [CrossRef] [Green Version]
- Biffi, G.; Oni, T.E.; Spielman, B.; Hao, Y.; Elyada, E.; Park, Y.; Preall, J.; Tuveson, D.A. IL1-Induced JAK/STAT Signaling Is Antagonized by TGFbeta to Shape CAF Heterogeneity in Pancreatic Ductal Adenocarcinoma. Cancer Discov. 2019, 9, 282–301. [Google Scholar] [CrossRef] [Green Version]
- Leppanen, V.M.; Saharinen, P.; Alitalo, K. Structural basis of Tie2 activation and Tie2/Tie1 heterodimerization. Proc. Natl. Acad. Sci. USA 2017, 114, 4376–4381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Korhonen, E.A.; Lampinen, A.; Giri, H.; Anisimov, A.; Kim, M.; Allen, B.; Fang, S.; D’Amico, G.; Sipila, T.J.; Lohela, M.; et al. Tie1 controls angiopoietin function in vascular remodeling and inflammation. J. Clin. Investig. 2016, 126, 3495–3510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Inoue, M.; Hager, J.H.; Ferrara, N.; Gerber, H.P.; Hanahan, D. VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell 2002, 1, 193–202. [Google Scholar] [CrossRef] [Green Version]
- Longo, V.; Brunetti, O.; Gnoni, A.; Cascinu, S.; Gasparini, G.; Lorusso, V.; Ribatti, D.; Silvestris, N. Angiogenesis in pancreatic ductal adenocarcinoma: A controversial issue. Oncotarget 2016, 7, 58649–58658. [Google Scholar] [CrossRef] [Green Version]
- Sarantis, P.; Koustas, E.; Papadimitropoulou, A.; Papavassiliou, A.G.; Karamouzis, M.V. Pancreatic ductal adenocarcinoma: Treatment hurdles, tumor microenvironment and immunotherapy. World J. Gastrointest. Oncol. 2020, 12, 173–181. [Google Scholar] [CrossRef]
- Jiang, B.; Zhou, L.; Lu, J.; Wang, Y.; Liu, C.; You, L.; Guo, J. Stroma-Targeting Therapy in Pancreatic Cancer: One Coin With Two Sides? Front. Oncol. 2020, 10, 576399. [Google Scholar] [CrossRef]
- Nelson, J.K.; Thin, M.Z.; Evan, T.; Howell, S.; Wu, M.; Almeida, B.; Legrave, N.; Koenis, D.S.; Koifman, G.; Sugimoto, Y.; et al. USP25 promotes pathological HIF-1-driven metabolic reprogramming and is a potential therapeutic target in pancreatic cancer. Nat. Commun 2022, 13, 2070. [Google Scholar] [CrossRef]
- Scafoglio, C.; Hirayama, B.A.; Kepe, V.; Liu, J.; Ghezzi, C.; Satyamurthy, N.; Moatamed, N.A.; Huang, J.; Koepsell, H.; Barrio, J.R.; et al. Functional expression of sodium-glucose transporters in cancer. Proc. Natl. Acad. Sci. USA 2015, 112, E4111–E4119. [Google Scholar] [CrossRef] [Green Version]
- Paredes, F.; Williams, H.C.; San Martin, A. Metabolic adaptation in hypoxia and cancer. Cancer Lett. 2021, 502, 133–142. [Google Scholar] [CrossRef]
- Chen, S.; Ning, B.; Song, J.; Yang, Z.; Zhou, L.; Chen, Z.; Mao, L.; Liu, H.; Wang, Q.; He, S.; et al. Enhanced pentose phosphate pathway activity promotes pancreatic ductal adenocarcinoma progression via activating YAP/MMP1 axis under chronic acidosis. Int. J. Biol. Sci. 2022, 18, 2304–2316. [Google Scholar] [CrossRef]
- Kong, S.C.; Nohr-Nielsen, A.; Zeeberg, K.; Reshkin, S.J.; Hoffmann, E.K.; Novak, I.; Pedersen, S.F. Monocarboxylate Transporters MCT1 and MCT4 Regulate Migration and Invasion of Pancreatic Ductal Adenocarcinoma Cells. Pancreas 2016, 45, 1036–1047. [Google Scholar] [CrossRef]
- Baek, G.; Tse, Y.F.; Hu, Z.; Cox, D.; Buboltz, N.; McCue, P.; Yeo, C.J.; White, M.A.; DeBerardinis, R.J.; Knudsen, E.S.; et al. MCT4 defines a glycolytic subtype of pancreatic cancer with poor prognosis and unique metabolic dependencies. Cell Rep. 2014, 9, 2233–2249. [Google Scholar] [CrossRef] [Green Version]
- Quinn, W.J., III; Jiao, J.; TeSlaa, T.; Stadanlick, J.; Wang, Z.; Wang, L.; Akimova, T.; Angelin, A.; Schafer, P.M.; Cully, M.D.; et al. Lactate Limits T Cell Proliferation via the NAD(H) Redox State. Cell Rep. 2020, 33, 108500. [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]
- Shukla, S.K.; Purohit, V.; Mehla, K.; Gunda, V.; Chaika, N.V.; Vernucci, E.; King, R.J.; Abrego, J.; Goode, G.D.; Dasgupta, A.; et al. MUC1 and HIF-1alpha Signaling Crosstalk Induces Anabolic Glucose Metabolism to Impart Gemcitabine Resistance to Pancreatic Cancer. Cancer Cell 2017, 32, 392. [Google Scholar] [CrossRef]
- Riethdorf, S.; Reimers, N.; Assmann, V.; Kornfeld, J.W.; Terracciano, L.; Sauter, G.; Pantel, K. High incidence of EMMPRIN expression in human tumors. Int. J. Cancer 2006, 119, 1800–1810. [Google Scholar] [CrossRef]
- Kirk, P.; Wilson, M.C.; Heddle, C.; Brown, M.H.; Barclay, A.N.; Halestrap, A.P. CD147 is tightly associated with lactate transporters MCT1 and MCT4 and facilitates their cell surface expression. EMBO J. 2000, 19, 3896–3904. [Google Scholar] [CrossRef]
- Schneiderhan, W.; Scheler, M.; Holzmann, K.H.; Marx, M.; Gschwend, J.E.; Bucholz, M.; Gress, T.M.; Seufferlein, T.; Adler, G.; Oswald, F. CD147 silencing inhibits lactate transport and reduces malignant potential of pancreatic cancer cells in in vivo and in vitro models. Gut 2009, 58, 1391–1398. [Google Scholar] [CrossRef]
- Patra, K.C.; Hay, N. The pentose phosphate pathway and cancer. Trends Biochem. Sci. 2014, 39, 347–354. [Google Scholar] [CrossRef] [Green Version]
- Bechard, M.E.; Word, A.E.; Tran, A.V.; Liu, X.; Locasale, J.W.; McDonald, O.G. Pentose conversions support the tumorigenesis of pancreatic cancer distant metastases. Oncogene 2018, 37, 5248–5256. [Google Scholar] [CrossRef]
- Kruger, N.J.; von Schaewen, A. The oxidative pentose phosphate pathway: Structure and organisation. Curr. Opin. Plant. Biol. 2003, 6, 236–246. [Google Scholar] [CrossRef]
- Stincone, A.; Prigione, A.; Cramer, T.; Wamelink, M.M.; Campbell, K.; Cheung, E.; Olin-Sandoval, V.; Gruning, N.M.; Kruger, A.; Tauqeer Alam, M.; et al. The return of metabolism: Biochemistry and physiology of the pentose phosphate pathway. Biol. Rev. Camb. Philos. Soc. 2015, 90, 927–963. [Google Scholar] [CrossRef] [Green Version]
- Santana-Codina, N.; Roeth, A.A.; Zhang, Y.; Yang, A.; Mashadova, O.; Asara, J.M.; Wang, X.; Bronson, R.T.; Lyssiotis, C.A.; Ying, H.; et al. Oncogenic KRAS supports pancreatic cancer through regulation of nucleotide synthesis. Nat. Commun. 2018, 9, 4945. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Ishak Gabra, M.B.; Hanse, E.A.; Lowman, X.H.; Tran, T.Q.; Li, H.; Milman, N.; Liu, J.; Reid, M.A.; Locasale, J.W.; et al. MiR-135 suppresses glycolysis and promotes pancreatic cancer cell adaptation to metabolic stress by targeting phosphofructokinase-1. Nat. Commun. 2019, 10, 809. [Google Scholar] [CrossRef] [Green Version]
- Wu, D.H.; Liang, H.; Lu, S.N.; Wang, H.; Su, Z.L.; Zhang, L.; Ma, J.Q.; Guo, M.; Tai, S.; Yu, S. miR-124 Suppresses Pancreatic Ductal Adenocarcinoma Growth by Regulating Monocarboxylate Transporter 1-Mediated Cancer Lactate Metabolism. Cell Physiol. Biochem. 2018, 50, 924–935. [Google Scholar] [CrossRef]
- Abue, M.; Yokoyama, M.; Shibuya, R.; Tamai, K.; Yamaguchi, K.; Sato, I.; Tanaka, N.; Hamada, S.; Shimosegawa, T.; Sugamura, K.; et al. Circulating miR-483-3p and miR-21 is highly expressed in plasma of pancreatic cancer. Int. J. Oncol. 2015, 46, 539–547. [Google Scholar] [CrossRef] [Green Version]
- Daoud, A.Z.; Mulholland, E.J.; Cole, G.; McCarthy, H.O. MicroRNAs in Pancreatic Cancer: Biomarkers, prognostic, and therapeutic modulators. BMC Cancer 2019, 19, 1130. [Google Scholar] [CrossRef] [Green Version]
- Xu, R.; Yang, J.; Ren, B.; Wang, H.; Yang, G.; Chen, Y.; You, L.; Zhao, Y. Reprogramming of Amino Acid Metabolism in Pancreatic Cancer: Recent Advances and Therapeutic Strategies. Front. Oncol. 2020, 10, 572722. [Google Scholar] [CrossRef]
- Cruzat, V.; Macedo Rogero, M.; Noel Keane, K.; Curi, R.; Newsholme, P. Glutamine: Metabolism and Immune Function, Supplementation and Clinical Translation. Nutrients 2018, 10, 1564. [Google Scholar] [CrossRef] [Green Version]
- Altman, B.J.; Stine, Z.E.; Dang, C.V. From Krebs to clinic: Glutamine metabolism to cancer therapy. Nat. Rev. Cancer 2016, 16, 619–634. [Google Scholar] [CrossRef] [Green Version]
- Cluntun, A.A.; Lukey, M.J.; Cerione, R.A.; Locasale, J.W. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer 2017, 3, 169–180. [Google Scholar] [CrossRef] [PubMed] [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.; et al. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res. 2015, 75, 544–553. [Google Scholar] [CrossRef] [Green Version]
- Bott, A.J.; Shen, J.; Tonelli, C.; Zhan, L.; Sivaram, N.; Jiang, Y.P.; Yu, X.; Bhatt, V.; Chiles, E.; Zhong, H.; et al. Glutamine Anabolism Plays a Critical Role in Pancreatic Cancer by Coupling Carbon and Nitrogen Metabolism. Cell Rep. 2019, 29, 1287–1298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reyes-Castellanos, G.; Abdel Hadi, N.; Carrier, A. Autophagy Contributes to Metabolic Reprogramming and Therapeutic Resistance in Pancreatic Tumors. Cells 2022, 11, 426. [Google Scholar] [CrossRef] [PubMed]
- Li, C.J.; Liao, W.T.; Wu, M.Y.; Chu, P.Y. New Insights into the Role of Autophagy in Tumor Immune Microenvironment. Int. J. Mol. Sci. 2017, 18, 1566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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.; et al. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature 2013, 497, 633–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wise, D.R.; DeBerardinis, R.J.; Mancuso, A.; Sayed, N.; Zhang, X.Y.; Pfeiffer, H.K.; Nissim, I.; Daikhin, E.; Yudkoff, M.; McMahon, S.B.; et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc. Natl. Acad. Sci. USA 2008, 105, 18782–18787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bar-Sagi, D.; Feramisco, J.R. Induction of membrane ruffling and fluid-phase pinocytosis in quiescent fibroblasts by ras proteins. Science 1986, 233, 1061–1068. [Google Scholar] [CrossRef]
- Porat-Shliom, N.; Kloog, Y.; Donaldson, J.G. A unique platform for H-Ras signaling involving clathrin-independent endocytosis. Mol. Biol. Cell 2008, 19, 765–775. [Google Scholar] [CrossRef] [Green Version]
- Seo, J.W.; Choi, J.; Lee, S.Y.; Sung, S.; Yoo, H.J.; Kang, M.J.; Cheong, H.; Son, J. Autophagy is required for PDAC glutamine metabolism. Sci. Rep. 2016, 6, 37594. [Google Scholar] [CrossRef]
- Carey, B.W.; Finley, L.W.; Cross, J.R.; Allis, C.D.; Thompson, C.B. Intracellular alpha-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature 2015, 518, 413–416. [Google Scholar] [CrossRef] [Green Version]
- Tran, T.Q.; Ishak Gabra, M.B.; Lowman, X.H.; Yang, Y.; Reid, M.A.; Pan, M.; O’Connor, T.R.; Kong, M. Glutamine deficiency induces DNA alkylation damage and sensitizes cancer cells to alkylating agents through inhibition of ALKBH enzymes. PLoS Biol. 2017, 15, e2002810. [Google Scholar] [CrossRef]
- Parker, S.J.; Amendola, C.R.; Hollinshead, K.E.R.; Yu, Q.; Yamamoto, K.; Encarnacion-Rosado, J.; Rose, R.E.; LaRue, M.M.; Sohn, A.S.W.; Biancur, D.E.; et al. Selective Alanine Transporter Utilization Creates a Targetable Metabolic Niche in Pancreatic Cancer. Cancer Discov. 2020, 10, 1018–1037. [Google Scholar] [CrossRef]
- Sperb, N.; Tsesmelis, M.; Wirth, T. Crosstalk between Tumor and Stromal Cells in Pancreatic Ductal Adenocarcinoma. Int. J. Mol. Sci. 2020, 21, 5486. [Google Scholar] [CrossRef]
- Riedl, J.M.; Posch, F.; Prager, G.; Eisterer, W.; Oehler, L.; Sliwa, T.; Wilthoner, K.; Petzer, A.; Pichler, P.; Hubmann, E.; et al. The AST/ALT (De Ritis) ratio predicts clinical outcome in patients with pancreatic cancer treated with first-line nab-paclitaxel and gemcitabine: Post hoc analysis of an Austrian multicenter, noninterventional study. Ther. Adv. Med. Oncol. 2020, 12, 1758835919900872. [Google Scholar] [CrossRef] [Green Version]
- Olivares, O.; Mayers, J.R.; Gouirand, V.; Torrence, M.E.; Gicquel, T.; Borge, L.; Lac, S.; Roques, J.; Lavaut, M.N.; Berthezene, P.; et al. Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions. Nat. Commun. 2017, 8, 16031. [Google Scholar] [CrossRef]
- Tajan, M.; Hock, A.K.; Blagih, J.; Robertson, N.A.; Labuschagne, C.F.; Kruiswijk, F.; Humpton, T.J.; Adams, P.D.; Vousden, K.H. A Role for p53 in the Adaptation to Glutamine Starvation through the Expression of SLC1A3. Cell Metab. 2018, 28, 721–736. [Google Scholar] [CrossRef] [Green Version]
- Chen, R.; Lai, L.A.; Sullivan, Y.; Wong, M.; Wang, L.; Riddell, J.; Jung, L.; Pillarisetty, V.G.; Brentnall, T.A.; Pan, S. Disrupting glutamine metabolic pathways to sensitize gemcitabine-resistant pancreatic cancer. Sci. Rep. 2017, 7, 7950. [Google Scholar] [CrossRef] [Green Version]
- Chini, C.C.; Guerrico, A.M.; Nin, V.; Camacho-Pereira, J.; Escande, C.; Barbosa, M.T.; Chini, E.N. Targeting of NAD metabolism in pancreatic cancer cells: Potential novel therapy for pancreatic tumors. Clin. Cancer Res. 2014, 20, 120–130. [Google Scholar] [CrossRef] [Green Version]
- Maddocks, O.D.K.; Athineos, D.; Cheung, E.C.; Lee, P.; Zhang, T.; van den Broek, N.J.F.; Mackay, G.M.; Labuschagne, C.F.; Gay, D.; Kruiswijk, F.; et al. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature 2017, 544, 372–376. [Google Scholar] [CrossRef]
- Badgley, M.A.; Kremer, D.M.; Maurer, H.C.; DelGiorno, K.E.; Lee, H.J.; Purohit, V.; Sagalovskiy, I.R.; Ma, A.; Kapilian, J.; Firl, C.E.M.; et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 2020, 368, 85–89. [Google Scholar] [CrossRef] [PubMed]
- Bianchi, M.E. DAMPs, PAMPs and alarmins: All we need to know about danger. J. Leukoc. Biol. 2007, 81, 1–5. [Google Scholar] [CrossRef] [PubMed]
- Genkinger, J.M.; Kitahara, C.M.; Bernstein, L.; Berrington de Gonzalez, A.; Brotzman, M.; Elena, J.W.; Giles, G.G.; Hartge, P.; Singh, P.N.; Stolzenberg-Solomon, R.Z.; et al. Central adiposity, obesity during early adulthood, and pancreatic cancer mortality in a pooled analysis of cohort studies. Ann. Oncol. 2015, 26, 2257–2266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oni, T.E.; Biffi, G.; Baker, L.A.; Hao, Y.; Tonelli, C.; Somerville, T.D.D.; Deschenes, A.; Belleau, P.; Hwang, C.I.; Sanchez-Rivera, F.J.; et al. SOAT1 promotes mevalonate pathway dependency in pancreatic cancer. J. Exp. Med. 2020, 217, e20192389. [Google Scholar] [CrossRef]
- Gabitova-Cornell, L.; Surumbayeva, A.; Peri, S.; Franco-Barraza, J.; Restifo, D.; Weitz, N.; Ogier, C.; Goldman, A.R.; Hartman, T.R.; Francescone, R.; et al. Cholesterol Pathway Inhibition Induces TGF-beta Signaling to Promote Basal Differentiation in Pancreatic Cancer. Cancer Cell 2020, 38, 567–583.e511. [Google Scholar] [CrossRef]
- Silvente-Poirot, S.; Poirot, M. Cancer. Cholesterol and cancer, in the balance. Science 2014, 343, 1445–1446. [Google Scholar] [CrossRef]
- Clerc, P.; Bensaadi, N.; Pradel, P.; Estival, A.; Clemente, F.; Vaysse, N. Lipid-dependent proliferation of pancreatic cancer cell lines. Cancer Res. 1991, 51, 3633–3638. [Google Scholar]
- Swierczynski, J.; Hebanowska, A.; Sledzinski, T. Role of abnormal lipid metabolism in development, progression, diagnosis and therapy of pancreatic cancer. World J. Gastroenterol. 2014, 20, 2279–2303. [Google Scholar] [CrossRef]
- Walter, K.; Hong, S.M.; Nyhan, S.; Canto, M.; Fedarko, N.; Klein, A.; Griffith, M.; Omura, N.; Medghalchi, S.; Kuhajda, F.; et al. Serum fatty acid synthase as a marker of pancreatic neoplasia. Cancer Epidemiol. Biomark. Prev. 2009, 18, 2380–2385. [Google Scholar] [CrossRef] [Green Version]
- Sun, Y.; He, W.; Luo, M.; Zhou, Y.; Chang, G.; Ren, W.; Wu, K.; Li, X.; Shen, J.; Zhao, X.; et al. SREBP1 regulates tumorigenesis and prognosis of pancreatic cancer through targeting lipid metabolism. Tumour Biol. 2015, 36, 4133–4141. [Google Scholar] [CrossRef]
- Tadros, S.; Shukla, S.K.; King, R.J.; Gunda, V.; Vernucci, E.; Abrego, J.; Chaika, N.V.; Yu, F.; Lazenby, A.J.; Berim, L.; et al. De Novo Lipid Synthesis Facilitates Gemcitabine Resistance through Endoplasmic Reticulum Stress in Pancreatic Cancer. Cancer Res. 2017, 77, 5503–5517. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Liu, H.; Li, Z.; Zhao, Z.; Yip-Schneider, M.; Fan, Q.; Schmidt, C.M.; Chiorean, E.G.; Xie, J.; Cheng, L.; et al. Role of fatty acid synthase in gemcitabine and radiation resistance of pancreatic cancers. Int. J. Biochem. Mol. Biol. 2011, 2, 89–98. [Google Scholar]
- Rozeveld, C.N.; Johnson, K.M.; Zhang, L.; Razidlo, G.L. KRAS Controls Pancreatic Cancer Cell Lipid Metabolism and Invasive Potential through the Lipase HSL. Cancer Res. 2020, 80, 4932–4945. [Google Scholar] [CrossRef]
- Dalen, K.T.; Ulven, S.M.; Arntsen, B.M.; Solaas, K.; Nebb, H.I. PPARalpha activators and fasting induce the expression of adipose differentiation-related protein in liver. J. Lipid Res. 2006, 47, 931–943. [Google Scholar] [CrossRef] [Green Version]
- Chang, B.H.; Li, L.; Paul, A.; Taniguchi, S.; Nannegari, V.; Heird, W.C.; Chan, L. Protection against fatty liver but normal adipogenesis in mice lacking adipose differentiation-related protein. Mol. Cell Biol. 2006, 26, 1063–1076. [Google Scholar] [CrossRef] [Green Version]
- Hashimoto, Y.; Ishida, M.; Ryota, H.; Yamamoto, T.; Kosaka, H.; Hirooka, S.; Yamaki, S.; Kotsuka, M.; Matsui, Y.; Yanagimoto, H.; et al. Adipophilin expression is an indicator of poor prognosis in patients with pancreatic ductal adenocarcinoma: An immunohistochemical analysis. Pancreatology 2019, 19, 443–448. [Google Scholar] [CrossRef]
- Chen, E.; Tsai, T.H.; Li, L.; Saha, P.; Chan, L.; Chang, B.H. PLIN2 is a Key Regulator of the Unfolded Protein Response and Endoplasmic Reticulum Stress Resolution in Pancreatic beta Cells. Sci. Rep. 2017, 7, 40855. [Google Scholar] [CrossRef]
- Schmidt, S.M.; Schag, K.; Muller, M.R.; Weinschenk, T.; Appel, S.; Schoor, O.; Weck, M.M.; Grunebach, F.; Kanz, L.; Stevanovic, S.; et al. Induction of adipophilin-specific cytotoxic T lymphocytes using a novel HLA-A2-binding peptide that mediates tumor cell lysis. Cancer Res. 2004, 64, 1164–1170. [Google Scholar] [CrossRef] [Green Version]
- Seifert, A.M.; Reiche, C.; Heiduk, M.; Tannert, A.; Meinecke, A.C.; Baier, S.; von Renesse, J.; Kahlert, C.; Distler, M.; Welsch, T.; et al. Detection of pancreatic ductal adenocarcinoma with galectin-9 serum levels. Oncogene 2020, 39, 3102–3113. [Google Scholar] [CrossRef] [Green Version]
- He, Y.; Dong, Y.; Zhang, X.; Ding, Z.; Song, Y.; Huang, X.; Chen, S.; Wang, Z.; Ni, Y.; Ding, L. Lipid Droplet-Related PLIN2 in CD68(+) Tumor-Associated Macrophage of Oral Squamous Cell Carcinoma: Implications for Cancer Prognosis and Immunotherapy. Front. Oncol. 2022, 12, 824235. [Google Scholar] [CrossRef]
- Philip, B.; Roland, C.L.; Daniluk, J.; Liu, Y.; Chatterjee, D.; Gomez, S.B.; Ji, B.; Huang, H.; Wang, H.; Fleming, J.B.; et al. 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] [Green Version]
- Chung, K.M.; Singh, J.; Lawres, L.; Dorans, K.J.; Garcia, C.; Burkhardt, D.B.; Robbins, R.; Bhutkar, A.; Cardone, R.; Zhao, X.; et al. Endocrine-Exocrine Signaling Drives Obesity-Associated Pancreatic Ductal Adenocarcinoma. Cell 2020, 181, 832–847.E18. [Google Scholar] [CrossRef]
- Greenberg, A.S.; Coleman, R.A.; Kraemer, F.B.; McManaman, J.L.; Obin, M.S.; Puri, V.; Yan, Q.W.; Miyoshi, H.; Mashek, D.G. The role of lipid droplets in metabolic disease in rodents and humans. J. Clin. Investig. 2011, 121, 2102–2110. [Google Scholar] [CrossRef] [Green Version]
- Olzmann, J.A.; Carvalho, P. Dynamics and functions of lipid droplets. Nat. Rev. Mol. Cell Biol. 2019, 20, 137–155. [Google Scholar] [CrossRef]
- Guillaumond, F.; Bidaut, G.; Ouaissi, M.; Servais, S.; Gouirand, V.; Olivares, O.; Lac, S.; Borge, L.; Roques, J.; Gayet, O.; et al. 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]
- 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]
- Padanad, M.S.; Konstantinidou, G.; Venkateswaran, N.; Melegari, M.; Rindhe, S.; Mitsche, M.; Yang, C.; Batten, K.; Huffman, K.E.; Liu, J.; et al. Fatty Acid Oxidation Mediated by Acyl-CoA Synthetase Long Chain 3 Is Required for Mutant KRAS Lung Tumorigenesis. Cell Rep. 2016, 16, 1614–1628. [Google Scholar] [CrossRef] [Green Version]
- Kamp, F.; Hamilton, J.A. How fatty acids of different chain length enter and leave cells by free diffusion. Prostaglandins Leukot Essent Fat. Acids 2006, 75, 149–159. [Google Scholar] [CrossRef]
- Rossi Sebastiano, M.; Konstantinidou, G. Targeting Long Chain Acyl-CoA Synthetases for Cancer Therapy. Int. J. Mol. Sci. 2019, 20, 3624. [Google Scholar] [CrossRef] [Green Version]
- Saliakoura, M.; Sebastiano, M.R.; Nikdima, I.; Pozzato, C.; Konstantinidou, G. Restriction of extracellular lipids renders pancreatic cancer dependent on autophagy. J. Exp. Clin. Cancer Res. 2022, 41, 16. [Google Scholar] [CrossRef]
- Brandi, J.; Dando, I.; Pozza, E.D.; Biondani, G.; Jenkins, R.; Elliott, V.; Park, K.; Fanelli, G.; Zolla, L.; Costello, E.; et al. Proteomic analysis of pancreatic cancer stem cells: Functional role of fatty acid synthesis and mevalonate pathways. J. Proteomics 2017, 150, 310–322. [Google Scholar] [CrossRef] [PubMed]
- Di Carlo, C.; Sousa, B.C.; Manfredi, M.; Brandi, J.; Dalla Pozza, E.; Marengo, E.; Palmieri, M.; Dando, I.; Wakelam, M.J.O.; Lopez-Clavijo, A.F.; et al. Integrated lipidomics and proteomics reveal cardiolipin alterations, upregulation of HADHA and long chain fatty acids in pancreatic cancer stem cells. Sci. Rep. 2021, 11, 13297. [Google Scholar] [CrossRef] [PubMed]
- El-Hafidi, M.; Correa, F.; Zazueta, C. Mitochondrial dysfunction in metabolic and cardiovascular diseases associated with cardiolipin remodeling. Biochim. Biophys. Acta. Mol. Basis Dis. 2020, 1866, 165744. [Google Scholar] [CrossRef] [PubMed]
- Pfeiffer, K.; Gohil, V.; Stuart, R.A.; Hunte, C.; Brandt, U.; Greenberg, M.L.; Schagger, H. Cardiolipin stabilizes respiratory chain supercomplexes. J. Biol. Chem. 2003, 278, 52873–52880. [Google Scholar] [CrossRef] [Green Version]
- Wolrab, D.; Jirasko, R.; Cifkova, E.; Horing, M.; Mei, D.; Chocholouskova, M.; Peterka, O.; Idkowiak, J.; Hrnciarova, T.; Kuchar, L.; et al. Lipidomic profiling of human serum enables detection of pancreatic cancer. Nat. Commun. 2022, 13, 124. [Google Scholar] [CrossRef]
- Mayerle, J.; Kalthoff, H.; Reszka, R.; Kamlage, B.; Peter, E.; Schniewind, B.; Gonzalez Maldonado, S.; Pilarsky, C.; Heidecke, C.D.; Schatz, P.; et al. Metabolic biomarker signature to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Gut 2018, 67, 128–137. [Google Scholar] [CrossRef] [Green Version]
- Fahrmann, J.F.; Bantis, L.E.; Capello, M.; Scelo, G.; Dennison, J.B.; Patel, N.; Murage, E.; Vykoukal, J.; Kundnani, D.L.; Foretova, L.; et al. A Plasma-Derived Protein-Metabolite Multiplexed Panel for Early-Stage Pancreatic Cancer. J. Natl. Cancer Inst. 2019, 111, 372–379. [Google Scholar] [CrossRef]
- Wang, G.; Yao, H.; Gong, Y.; Lu, Z.; Pang, R.; Li, Y.; Yuan, Y.; Song, H.; Liu, J.; Jin, Y.; et al. Metabolic detection and systems analyses of pancreatic ductal adenocarcinoma through machine learning, lipidomics, and multi-omics. Sci. Adv. 2021, 7, eabh2724. [Google Scholar] [CrossRef]
- Macias, R.I.R.; Munoz-Bellvis, L.; Sanchez-Martin, A.; Arretxe, E.; Martinez-Arranz, I.; Lapitz, A.; Gutierrez, M.L.; La Casta, A.; Alonso, C.; Gonzalez, L.M.; et al. A Novel Serum Metabolomic Profile for the Differential Diagnosis of Distal Cholangiocarcinoma and Pancreatic Ductal Adenocarcinoma. Cancers 2020, 12, 1433. [Google Scholar] [CrossRef]
- Doll, S.; Proneth, B.; Tyurina, Y.Y.; Panzilius, E.; Kobayashi, S.; Ingold, I.; Irmler, M.; Beckers, J.; Aichler, M.; Walch, A.; et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 2017, 13, 91–98. [Google Scholar] [CrossRef]
- Hu, N.; Bai, L.; Dai, E.; Han, L.; Kang, R.; Li, H.; Tang, D. Pirin is a nuclear redox-sensitive modulator of autophagy-dependent ferroptosis. Biochem. Biophys. Res. Commun. 2021, 536, 100–106. [Google Scholar] [CrossRef]
- Okawa, T.; Nagai, M.; Hase, K. Dietary Intervention Impacts Immune Cell Functions and Dynamics by Inducing Metabolic Rewiring. Front. Immunol. 2020, 11, 623989. [Google Scholar] [CrossRef]
- Loftus, R.M.; Finlay, D.K. Immunometabolism: Cellular Metabolism Turns Immune Regulator. J. Biol. Chem. 2016, 291, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Chang, C.H.; Qiu, J.; O’Sullivan, D.; Buck, M.D.; Noguchi, T.; Curtis, J.D.; Chen, Q.; Gindin, M.; Gubin, M.M.; van der Windt, G.J.; et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [Green Version]
- Hirayama, A.; Kami, K.; Sugimoto, M.; Sugawara, M.; Toki, N.; Onozuka, H.; Kinoshita, T.; Saito, N.; Ochiai, A.; Tomita, M.; et al. Quantitative metabolome profiling of colon and stomach cancer microenvironment by capillary electrophoresis time-of-flight mass spectrometry. Cancer Res. 2009, 69, 4918–4925. [Google Scholar] [CrossRef] [Green Version]
- Ochi, A.; Nguyen, A.H.; Bedrosian, A.S.; Mushlin, H.M.; Zarbakhsh, S.; Barilla, R.; Zambirinis, C.P.; Fallon, N.C.; Rehman, A.; Pylayeva-Gupta, Y.; et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. J. Exp. Med. 2012, 209, 1671–1687. [Google Scholar] [CrossRef]
- Choi, S.Y.; 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]
- Fukunaga, A.; Miyamoto, M.; Cho, Y.; Murakami, S.; Kawarada, Y.; Oshikiri, T.; Kato, K.; Kurokawa, T.; Suzuoki, M.; Nakakubo, Y.; et al. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas 2004, 28, e26–e31. [Google Scholar] [CrossRef]
- Ino, Y.; Yamazaki-Itoh, R.; Shimada, K.; Iwasaki, M.; Kosuge, T.; Kanai, Y.; Hiraoka, N. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br. J. Cancer 2013, 108, 914–923. [Google Scholar] [CrossRef]
- Kim, J.S.; Park, Y.S.; Kim, J.Y.; Kim, Y.G.; Kim, Y.J.; Lee, H.K.; Kim, H.S.; Hong, J.T.; Kim, Y.; Han, S.B. Inhibition of human pancreatic tumor growth by cytokine-induced killer cells in nude mouse xenograft model. Immune. Netw. 2012, 12, 247–252. [Google Scholar] [CrossRef] [Green Version]
- Yamamoto, K.; Venida, A.; Yano, J.; Biancur, D.E.; Kakiuchi, M.; Gupta, S.; Sohn, A.S.W.; Mukhopadhyay, S.; Lin, E.Y.; Parker, S.J.; et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 2020, 581, 100–105. [Google Scholar] [CrossRef]
- Liu, L.; Zhao, G.; Wu, W.; Rong, Y.; Jin, D.; Wang, D.; Lou, W.; Qin, X. Low intratumoral regulatory T cells and high peritumoral CD8(+) T cells relate to long-term survival in patients with pancreatic ductal adenocarcinoma after pancreatectomy. Cancer Immunol. Immunother. 2016, 65, 73–82. [Google Scholar] [CrossRef]
- Kurts, C. Th17 cells: A third subset of CD4+ T effector cells involved in organ-specific autoimmunity. Nephrol. Dial. Transplant. 2008, 23, 816–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wormann, S.M.; Diakopoulos, K.N.; Lesina, M.; Algul, H. The immune network in pancreatic cancer development and progression. Oncogene 2014, 33, 2956–2967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gnerlich, J.L.; Mitchem, J.B.; Weir, J.S.; Sankpal, N.V.; Kashiwagi, H.; Belt, B.A.; Porembka, M.R.; Herndon, J.M.; Eberlein, T.J.; Goedegebuure, P.; et al. Induction of Th17 cells in the tumor microenvironment improves survival in a murine model of pancreatic cancer. J. Immunol. 2010, 185, 4063–4071. [Google Scholar] [CrossRef] [Green Version]
- Helderman, J.H.; Reynolds, T.C.; Strom, T.B. The insulin receptor as a universal marker of activated lymphocytes. Eur. J. Immunol. 1978, 8, 589–595. [Google Scholar] [CrossRef] [PubMed]
- Fischer, H.J.; Sie, C.; Schumann, E.; Witte, A.K.; Dressel, R.; van den Brandt, J.; Reichardt, H.M. The Insulin Receptor Plays a Critical Role in T Cell Function and Adaptive Immunity. J. Immunol. 2017, 198, 1910–1920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singer, K.; Cheng, W.C.; Kreutz, M.; Ho, P.C.; Siska, P.J. Immunometabolism in cancer at a glance. Dis. Model. Mech. 2018, 11, dmm034272. [Google Scholar] [CrossRef] [Green Version]
- Sinclair, L.V.; Rolf, J.; Emslie, E.; Shi, Y.B.; Taylor, P.M.; Cantrell, D.A. Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat. Immunol. 2013, 14, 500–508. [Google Scholar] [CrossRef] [Green Version]
- Pilotte, L.; Larrieu, P.; Stroobant, V.; Colau, D.; Dolusic, E.; Frederick, R.; De Plaen, E.; Uyttenhove, C.; Wouters, J.; Masereel, B.; et al. Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase. Proc. Natl. Acad. Sci. USA 2012, 109, 2497–2502. [Google Scholar] [CrossRef] [Green Version]
- Yamamoto, T.; Yanagimoto, H.; Satoi, S.; Toyokawa, H.; Hirooka, S.; Yamaki, S.; Yui, R.; Yamao, J.; Kim, S.; Kwon, A.H. Circulating CD4+CD25+ regulatory T cells in patients with pancreatic cancer. Pancreas 2012, 41, 409–415. [Google Scholar] [CrossRef]
- Cai, S.W.; Yang, S.Z.; Gao, J.; Pan, K.; Chen, J.Y.; Wang, Y.L.; Wei, L.X.; Dong, J.H. Prognostic significance of mast cell count following curative resection for pancreatic ductal adenocarcinoma. Surgery 2011, 149, 576–584. [Google Scholar] [CrossRef]
- Deicher, A.; Andersson, R.; Tingstedt, B.; Lindell, G.; Bauden, M.; Ansari, D. Targeting dendritic cells in pancreatic ductal adenocarcinoma. Cancer Cell Int. 2018, 18, 85. [Google Scholar] [CrossRef]
- Dietl, K.; Renner, K.; Dettmer, K.; Timischl, B.; Eberhart, K.; Dorn, C.; Hellerbrand, C.; Kastenberger, M.; Kunz-Schughart, L.A.; Oefner, P.J.; et al. Lactic acid and acidification inhibit TNF secretion and glycolysis of human monocytes. J. Immunol. 2010, 184, 1200–1209. [Google Scholar] [CrossRef]
- Yang, S.; Liu, Q.; Liao, Q. Tumor-Associated Macrophages in Pancreatic Ductal Adenocarcinoma: Origin, Polarization, Function, and Reprogramming. Front. Cell Dev. Biol. 2020, 8, 607209. [Google Scholar] [CrossRef]
- Mills, E.L.; O’Neill, L.A. Reprogramming mitochondrial metabolism in macrophages as an anti-inflammatory signal. Eur. J. Immunol. 2016, 46, 13–21. [Google Scholar] [CrossRef]
- Rodriguez-Prados, J.C.; Traves, P.G.; Cuenca, J.; Rico, D.; Aragones, J.; Martin-Sanz, P.; Cascante, M.; Bosca, L. Substrate fate in activated macrophages: A comparison between innate, classic, and alternative activation. J. Immunol. 2010, 185, 605–614. [Google Scholar] [CrossRef] [Green Version]
- Haschemi, A.; Kosma, P.; Gille, L.; Evans, C.R.; Burant, C.F.; Starkl, P.; Knapp, B.; Haas, R.; Schmid, J.A.; Jandl, C.; et al. The sedoheptulose kinase CARKL directs macrophage polarization through control of glucose metabolism. Cell Metab. 2012, 15, 813–826. [Google Scholar] [CrossRef] [Green Version]
- O’Neill, L.A.; Kishton, R.J.; Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 2016, 16, 553–565. [Google Scholar] [CrossRef] [Green Version]
- Amedei, A.; Niccolai, E.; Prisco, D. Pancreatic cancer: Role of the immune system in cancer progression and vaccine-based immunotherapy. Hum. Vaccin. Immunother. 2014, 10, 3354–3368. [Google Scholar] [CrossRef] [Green Version]
- De Monte, L.; Reni, M.; Tassi, E.; Clavenna, D.; Papa, I.; Recalde, H.; Braga, M.; Di Carlo, V.; Doglioni, C.; Protti, M.P. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J. Exp. Med. 2011, 208, 469–478. [Google Scholar] [CrossRef] [PubMed]
- O’Sullivan, D.; van der Windt, G.J.; Huang, S.C.; Curtis, J.D.; Chang, C.H.; Buck, M.D.; Qiu, J.; Smith, A.M.; Lam, W.Y.; DiPlato, L.M.; et al. Memory CD8(+) T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 2014, 41, 75–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Esposito, I.; Menicagli, M.; Funel, N.; Bergmann, F.; Boggi, U.; Mosca, F.; Bevilacqua, G.; Campani, D. Inflammatory cells contribute to the generation of an angiogenic phenotype in pancreatic ductal adenocarcinoma. J. Clin. Pathol. 2004, 57, 630–636. [Google Scholar] [CrossRef] [PubMed] [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]
- Arnold, C.; Demuth, P.; Seiwert, N.; Wittmann, S.; Boengler, K.; Rasenberger, B.; Christmann, M.; Huber, M.; Brunner, T.; Linnebacher, M.; et al. The Mitochondrial Disruptor Devimistat (CPI-613) Synergizes with Genotoxic Anticancer Drugs in Colorectal Cancer Therapy in a Bim-Dependent Manner. Mol. Cancer Ther. 2022, 21, 100–112. [Google Scholar] [CrossRef]
- Rebelo, R.; Polonia, B.; Santos, L.L.; Vasconcelos, M.H.; Xavier, C.P.R. Drug Repurposing Opportunities in Pancreatic Ductal Adenocarcinoma. Pharmaceuticals 2021, 14, 280. [Google Scholar] [CrossRef]
- Islam, M.M.; Goertzen, A.; Singh, P.K.; Saha, R. Exploring the metabolic landscape of pancreatic ductal adenocarcinoma cells using genome-scale metabolic modeling. iScience 2022, 25, 104483. [Google Scholar] [CrossRef]
Identifier ID | Study Title | Conditions | Interventions |
---|---|---|---|
NCT05296421 | Investigating Targetable Metabolic Pathways Sustaining Pancreatic Cancer | Primary | Procedure: Biopsy, Therapeutic Conventional Surgery Other: Uniformly-labeled [13C] glucose |
NCT04565327 | Hyperpolarized 13C Pyruvate MRI for Treatment Response Assessment in Patients With Locally Advanced or Metastatic Pancreatic Cancer | Primary | Drug: Hyperpolarized Carbon C 13 Pyruvate, Procedure: Magnetic Resonance Imaging (MRI) |
NCT04862260 | Cholesterol Disruption in Combination With FOLFIRINOX in Patients With Metastatic Pancreatic Adenocarcinoma | Primary and Metastatic | Drug: Cholesterol metabolism disruption |
NCT02978547 | The Effects of Neoadjuvant Metformin on Tumor Cell Proliferation and Tumor Progression in Pancreatic Ductal Adenocarcinoma | Primary | Drug: Metformin Hydrochloride 500 Mg Tablet |
NCT05254171 | Study of Nab-Paclitaxel and Gemcitabine With or Without SBP-101 in Pancreatic Cancer | Primary | Drug: SBP-101, Nab-paclitaxel, Gemcitabine and Placebo |
NCT03450018 | A Study of SLC-0111 and Gemcitabine for Metastatic Pancreatic Ductal Cancer in Subjects Positive for CAIX | Metastatic | Drug: SLC-0111, Gemcitabine Injection |
NCT05132244 | Monitoring and Managing Glucose Levels in People With Pancreatic Cancer | Primary | Procedure: Endocrinologist-directed target blood glucose level 4–10 mmol/L using data from a continuous glucose monitor (CGM). Other: Standard Care |
NCT04915417 | Neoadjuvant Stereotactic Ablative Radiotherapy for Pancreatic Ductal Adenocarcinoma | Primary | Radiation: Stereotactic Ablative Body Radiotherapy (SABR) |
NCT04662879 | Early Detection Initiative for Pancreatic Cancer | Primary | Other: Enriching New-onset Diabetes for Pancreatic Cancer (ENDPAC) score. Other: Abdominal imaging |
NCT03525392 | Study to Evaluate the Safety and Activity (Including Distribution) of 177Lu-3BP-227 in Subjects With Solid Tumors Expressing Neurotensin Receptor Type 1. | Primary | Drug: 177Lu-3BP-227 (also called 177Lu-IPN01087) |
NCT03410030 | Trial of Ascorbic Acid (AA) + Nanoparticle Paclitaxel Protein Bound + Cisplatin + Gemcitabine (AA NABPLAGEM) | Primary | Drug: Ascorbic Acid, Paclitaxel protein-bound, Cisplatin, Gemcitabine |
NCT04245644 | Efficacy of Chemopreventive Agents on Disease-free and Overall Survival in Patients With Pancreatic Ductal Adenocarcinoma: The CAOS Study (CAOS) | Primary | Behavioral: use of targeted drugs such as aspirin, B-Blockers, Metformin, ACE-inhibitors, Statins |
NCT03374852 | CPI-613 in Combination With Modified FOLFIRINOX in Patients With Locally Advanced Pancreatic Cancer | Primary | Drug: CPI-613 Drug: mFOLFIRNOX |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Ali, A.; Chianese, U.; Papulino, C.; Toraldo, A.; Abakar, M.E.A.; Passaro, E.; Cennamo, R.; Del Gaudio, N.; Altucci, L.; Benedetti, R. Metabolic Pathways as a Novel Landscape in Pancreatic Ductal Adenocarcinoma. Cancers 2022, 14, 3799. https://doi.org/10.3390/cancers14153799
Ali A, Chianese U, Papulino C, Toraldo A, Abakar MEA, Passaro E, Cennamo R, Del Gaudio N, Altucci L, Benedetti R. Metabolic Pathways as a Novel Landscape in Pancreatic Ductal Adenocarcinoma. Cancers. 2022; 14(15):3799. https://doi.org/10.3390/cancers14153799
Chicago/Turabian StyleAli, Ahmad, Ugo Chianese, Chiara Papulino, Antonella Toraldo, Mawada Elmagboul Abdalla Abakar, Eugenia Passaro, Rosario Cennamo, Nunzio Del Gaudio, Lucia Altucci, and Rosaria Benedetti. 2022. "Metabolic Pathways as a Novel Landscape in Pancreatic Ductal Adenocarcinoma" Cancers 14, no. 15: 3799. https://doi.org/10.3390/cancers14153799
APA StyleAli, A., Chianese, U., Papulino, C., Toraldo, A., Abakar, M. E. A., Passaro, E., Cennamo, R., Del Gaudio, N., Altucci, L., & Benedetti, R. (2022). Metabolic Pathways as a Novel Landscape in Pancreatic Ductal Adenocarcinoma. Cancers, 14(15), 3799. https://doi.org/10.3390/cancers14153799