Strategies to Interfere with Tumor Metabolism through the Interplay of Innate and Adaptive Immunity
Abstract
:1. The Delicate Interplay between the Host Immunity and the Tumor
2. Cancer Cell Metabolism—Nutrient Plasticity at a Glance
2.1. Adaptations of Tumor Cell Metabolism under Hypoxia
2.2. Impact of Iron on OXPHOS and TCA
2.3. Tumor Cell Iron Metabolism
3. Anti-Tumor Therapies—Immunotherapeutic Approaches and Beyond
3.1. Targeting Iron Metabolism in Cancer—A Tug of War
3.2. Tumor-Associated Macrophages—Promising Targets or Major Players in Tumor Immunotherapy?
3.3. T Cells at the Center Stage of Effective Anti-Tumor Immunity
Funding
Conflicts of Interest
Abbreviations
References
- Hui, L.; Chen, Y. Tumor microenvironment: Sanctuary of the devil. Cancer Lett. 2015, 368, 7–13. [Google Scholar] [CrossRef]
- Xu, L.; Vaupel, P.; Bai, S.; Menze, B.; Shi, K. Computational Simulation of Tumor Hypoxia Based on In Vivo Microvasculature Assessed in a Dorsal Skin Window Chamber. Adv. Exp. Med. Biol. 2017, 977, 109–117. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Bae, J.-S. Tumor-Associated Macrophages and Neutrophils in Tumor Microenvironment. Mediat. Inflamm. 2016, 2016, 6058147. [Google Scholar] [CrossRef] [PubMed]
- Brüne, B.; Weigert, A.; Dehne, N. Macrophage Polarization In The Tumor Microenvironment. Redox Biol. 2015, 5, 419. [Google Scholar] [CrossRef] [Green Version]
- Dehne, N.; Mora, J.; Namgaladze, D.; Weigert, A.; Brüne, B. Cancer cell and macrophage cross-talk in the tumor microenvironment. Curr. Opin. Pharmacol. 2017, 35, 12–19. [Google Scholar] [CrossRef] [PubMed]
- Zhou, L.; Zhao, B.; Zhang, L.; Wang, S.; Dong, D.; Lv, H.; Shang, P. Alterations in Cellular Iron Metabolism Provide More Therapeutic Opportunities for Cancer. Int. J. Mol. Sci. 2018, 19, 1545. [Google Scholar] [CrossRef] [PubMed]
- Yagi, T.; Baba, Y.; Okadome, K.; Kiyozumi, Y.; Hiyoshi, Y.; Ishimoto, T.; Iwatsuki, M.; Miyamoto, Y.; Yoshida, N.; Watanabe, M.; et al. Tumour-associated macrophages are associated with poor prognosis and programmed death ligand 1 expression in oesophageal cancer. Eur. J. Cancer 2019, 111, 38–49. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.; He, H.; Liu, H.; Li, R.; Chen, Y.; Qi, Y.; Jiang, Q.; Chen, L.; Zhang, P.; Zhang, H.; et al. Tumour-associated macrophages-derived CXCL8 determines immune evasion through autonomous PD-L1 expression in gastric cancer. Gut 2019. [Google Scholar] [CrossRef] [PubMed]
- Yang, M.; Li, Z.; Ren, M.; Li, S.; Zhang, L.; Zhang, X.; Liu, F. Stromal Infiltration of Tumor-Associated Macrophages Conferring Poor Prognosis of Patients with Basal-Like Breast Carcinoma. J. Cancer 2018, 9, 2308–2316. [Google Scholar] [CrossRef] [PubMed]
- Zheng, X.; Turkowski, K.; Mora, J.; Brüne, B.; Seeger, W.; Weigert, A.; Savai, R. Redirecting tumor-associated macrophages to become tumoricidal effectors as a novel strategy for cancer therapy. Oncotarget 2017, 8, 48436–48452. [Google Scholar] [CrossRef] [Green Version]
- Weigert, A.; Weichand, B.; Brune, B. S1P regulation of macrophage functions in the context of cancer. Anticancer. Agents Med. Chem. 2011, 11, 818–829. [Google Scholar] [CrossRef] [PubMed]
- Olesch, C.; Sha, W.; Angioni, C.; Sha, L.K.; Açaf, E.; Patrignani, P.; Jakobsson, P.-J.; Radeke, H.H.; Grösch, S.; Geisslinger, G.; et al. MPGES-1-derived PGE2 suppresses CD80 expression on tumor-associated phagocytes to inhibit anti-tumor immune responses in breast cancer. Oncotarget 2015, 6, 10284–10296. [Google Scholar] [CrossRef] [Green Version]
- Weigert, A.; Mora, J.; Sekar, D.; Syed, S.; Brüne, B. Killing Is Not Enough: How Apoptosis Hijacks Tumor-Associated Macrophages to Promote Cancer Progression. Adv. Exp. Med. Biol. 2016, 930, 205–239. [Google Scholar] [CrossRef] [PubMed]
- Gregory, C.D.; Pound, J.D. Cell death in the neighbourhood: Direct microenvironmental effects of apoptosis in normal and neoplastic tissues. J. Pathol. 2011, 223, 177–194. [Google Scholar] [CrossRef] [PubMed]
- Bosurgi, L.; Cao, Y.G.; Cabeza-Cabrerizo, M.; Tucci, A.; Hughes, L.D.; Kong, Y.; Weinstein, J.S.; Licona-Limon, P.; Schmid, E.T.; Pelorosso, F.; et al. Macrophage function in tissue repair and remodeling requires IL-4 or IL-13 with apoptotic cells. Science 2017, 356, 1072–1076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cummings, R.J.; Barbet, G.; Bongers, G.; Hartmann, B.M.; Gettler, K.; Muniz, L.; Furtado, G.C.; Cho, J.; Lira, S.A.; Blander, J.M. Different tissue phagocytes sample apoptotic cells to direct distinct homeostasis programs. Nature 2016, 539, 565–569. [Google Scholar] [CrossRef] [PubMed]
- Dvorak, H.F. Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing. N. Engl. J. Med. 1986, 315, 1650–1659. [Google Scholar] [CrossRef]
- Han, C.Z.; Ravichandran, K.S. Metabolic connections during apoptotic cell engulfment. Cell 2011, 147, 1442–1445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vats, D.; Mukundan, L.; Odegaard, J.I.; Zhang, L.; Smith, K.L.; Morel, C.R.; Wagner, R.A.; Greaves, D.R.; Murray, P.J.; Chawla, A. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metabolism 2006, 4, 13–24. [Google Scholar] [CrossRef]
- Colegio, O.R.; Chu, N.-Q.; Szabo, A.L.; Chu, T.; Rhebergen, A.M.; Jairam, V.; Cyrus, N.; Brokowski, C.E.; Eisenbarth, S.C.; Phillips, G.M.; et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014, 513, 559–563. [Google Scholar] [CrossRef] [Green Version]
- Kratochvill, F.; Neale, G.; Haverkamp, J.M.; van de Velde, L.-A.; Smith, A.M.; Kawauchi, D.; McEvoy, J.; Roussel, M.F.; Dyer, M.A.; Qualls, J.E.; et al. TNF Counterbalances the Emergence of M2 Tumor Macrophages. Cell Rep. 2015, 12, 1902–1914. [Google Scholar] [CrossRef] [PubMed]
- Carmona-Fontaine, C.; Deforet, M.; Akkari, L.; Thompson, C.B.; Joyce, J.A.; Xavier, J.B. Metabolic origins of spatial organization in the tumor microenvironment. Proc. Natl. Acad. Sci. USA 2017, 114, 2934–2939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munn, D.H.; Shafizadeh, E.; Attwood, J.T.; Bondarev, I.; Pashine, A.; Mellor, A.L. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J. Exp. Med. 1999, 189, 1363–1372. [Google Scholar] [CrossRef]
- Hobson-Gutierrez, S.A.; Carmona-Fontaine, C. The metabolic axis of macrophage and immune cell polarization. Dis. Model. Mech. 2018, 11. [Google Scholar] [CrossRef] [PubMed]
- Warburg, O. The Metabolism of Tumors in the Body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef]
- Potter, M.; Newport, E.; Morten, K.J. The Warburg effect: 80 years on. Biochem. Soc. Trans. 2016, 44, 1499–1505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, H.; Wang, J.; Yan, W.; Cui, Y.; Chen, Z.; Gao, X.; Wen, X.; Chen, J. GLUT1 regulates cell glycolysis and proliferation in prostate cancer. Prostate 2018, 78, 86–94. [Google Scholar] [CrossRef]
- Corbet, C.; Feron, O. Cancer cell metabolism and mitochondria: Nutrient plasticity for TCA cycle fueling. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 7–15. [Google Scholar] [CrossRef]
- Anderson, N.M.; Mucka, P.; Kern, J.G.; Feng, H. The emerging role and targetability of the TCA cycle in cancer metabolism. Protein Cell 2018, 9, 216–237. [Google Scholar] [CrossRef]
- Bonuccelli, G.; Tsirigos, A.; Whitaker-Menezes, D.; Pavlides, S.; Pestell, R.G.; Chiavarina, B.; Frank, P.G.; Flomenberg, N.; Howell, A.; Martinez-Outschoorn, U.E.; et al. Ketones and lactate “fuel” tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle 2010, 9, 3506–3514. [Google Scholar] [CrossRef]
- Martinez-Outschoorn, U.E.; Lin, Z.; Whitaker-Menezes, D.; Howell, A.; Sotgia, F.; Lisanti, M.P. Ketone body utilization drives tumor growth and metastasis. Cell Cycle 2012, 11, 3964–3971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, Y.; Oeh, J.; Hitz, A.; Hedehus, M.; Eastham-Anderson, J.; Peale, F.V.; Hamilton, P.; O’Brien, T.; Sampath, D.; Carano, R.A.D. Monitoring and Targeting Anti-VEGF Induced Hypoxia within the Viable Tumor by 19F-MRI and Multispectral Analysis. Neoplasia 2017, 19, 950–959. [Google Scholar] [CrossRef]
- Appelhoff, R.J.; Tian, Y.-M.; Raval, R.R.; Turley, H.; Harris, A.L.; Pugh, C.W.; Ratcliffe, P.J.; Gleadle, J.M. Differential Function of the Prolyl Hydroxylases PHD1, PHD2, and PHD3 in the Regulation of Hypoxia-inducible Factor. J. Biol. Chem. 2004, 279, 38458–38465. [Google Scholar] [CrossRef] [Green Version]
- Lu, H.; Dalgard, C.L.; Mohyeldin, A.; McFate, T.; Tait, A.S.; Verma, A. Reversible Inactivation of HIF-1 Prolyl Hydroxylases Allows Cell Metabolism to Control Basal HIF-1. J. Biol. Chem. 2005, 280, 41928–41939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bharti, S.K.; Mironchik, Y.; Wildes, F.; Penet, M.-F.; Goggins, E.; Krishnamachary, B.; Bhujwalla, Z.M. Metabolic consequences of HIF silencing in a triple negative human breast cancer xenograft. Oncotarget 2018, 9, 15326–15339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, J.-W.; Tchernyshyov, I.; Semenza, G.L.; Dang, C.V. HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006, 3, 177–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Semenza, G.L.; Jiang, B.H.; Leung, S.W.; Passantino, R.; Concordet, J.P.; Maire, P.; Giallongo, A. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J. Biol. Chem. 1996, 271, 32529–32537. [Google Scholar] [CrossRef]
- Dupuy, F.; Tabariès, S.; Andrzejewski, S.; Dong, Z.; Blagih, J.; Annis, M.G.; Omeroglu, A.; Gao, D.; Leung, S.; Amir, E.; et al. PDK1-Dependent Metabolic Reprogramming Dictates Metastatic Potential in Breast Cancer. Cell Metabolism 2015, 22, 577–589. [Google Scholar] [CrossRef] [Green Version]
- Chen, Z.; Zuo, X.; Zhang, Y.; Han, G.; Zhang, L.; Wu, J.; Wang, X. MiR-3662 suppresses hepatocellular carcinoma growth through inhibition of HIF-1α-mediated Warburg effect. Cell Death Dis. 2018, 9, 549. [Google Scholar] [CrossRef] [Green Version]
- De Saedeleer, C.J.; Copetti, T.; Porporato, P.E.; Verrax, J.; Feron, O.; Sonveaux, P. Lactate activates HIF-1 in oxidative but not in Warburg-phenotype human tumor cells. PLoS ONE 2012, 7, e46571. [Google Scholar] [CrossRef]
- Fuhrmann, D.C.; Brüne, B. Mitochondrial composition and function under the control of hypoxia. Redox Biol. 2017, 12, 208–215. [Google Scholar] [CrossRef]
- Wei, R.; Mao, L.; Xu, P.; Zheng, X.; Hackman, R.M.; Mackenzie, G.G.; Wang, Y. Suppressing glucose metabolism with epigallocatechin-3-gallate (EGCG) reduces breast cancer cell growth in preclinical models. Food Funct. 2018, 9, 5682–5696. [Google Scholar] [CrossRef] [PubMed]
- De Padua, M.C.; Delodi, G.; Vučetić, M.; Durivault, J.; Vial, V.; Bayer, P.; Noleto, G.R.; Mazure, N.M.; Ždralević, M.; Pouysségur, J. Disrupting glucose-6-phosphate isomerase fully suppresses the “Warburg effect” and activates OXPHOS with minimal impact on tumor growth except in hypoxia. Oncotarget 2017, 8, 87623–87637. [Google Scholar] [CrossRef] [Green Version]
- Ždralević, M.; Brand, A.; Di Ianni, L.; Dettmer, K.; Reinders, J.; Singer, K.; Peter, K.; Schnell, A.; Bruss, C.; Decking, S.-M.; et al. Double genetic disruption of lactate dehydrogenases A and B is required to ablate the “Warburg effect” restricting tumor growth to oxidative metabolism. J. Biol. Chem. 2018, 293, 15947–15961. [Google Scholar] [CrossRef]
- Fuhrmann, D.C.; Wittig, I.; Heide, H.; Dehne, N.; Brüne, B. Chronic hypoxia alters mitochondrial composition in human macrophages. Biochim. Biophys. Acta Proteins Proteom. 2013, 1834, 2750–2760. [Google Scholar] [CrossRef]
- Hernansanz-Agustín, P.; Ramos, E.; Navarro, E.; Parada, E.; Sánchez-López, N.; Peláez-Aguado, L.; Cabrera-García, J.D.; Tello, D.; Buendia, I.; Marina, A.; et al. Mitochondrial complex I deactivation is related to superoxide production in acute hypoxia. Redox Biol. 2017, 12, 1040–1051. [Google Scholar] [CrossRef] [Green Version]
- Lucarelli, G.; Rutigliano, M.; Sallustio, F.; Ribatti, D.; Giglio, A.; Lepore Signorile, M.; Grossi, V.; Sanese, P.; Napoli, A.; Maiorano, E.; et al. Integrated multi-omics characterization reveals a distinctive metabolic signature and the role of NDUFA4L2 in promoting angiogenesis, chemoresistance, and mitochondrial dysfunction in clear cell renal cell carcinoma. Aging (Albany NY) 2018, 10, 3957–3985. [Google Scholar] [CrossRef]
- Tello, D.; Balsa, E.; Acosta-Iborra, B.; Fuertes-Yebra, E.; Elorza, A.; Ordóñez, Á.; Corral-Escariz, M.; Soro, I.; López-Bernardo, E.; Perales-Clemente, E.; et al. Induction of the Mitochondrial NDUFA4L2 Protein by HIF-1α Decreases Oxygen Consumption by Inhibiting Complex I Activity. Cell Metabol. 2011, 14, 768–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fuhrmann, D.C.; Wittig, I.; Dröse, S.; Schmid, T.; Dehne, N.; Brüne, B. Degradation of the mitochondrial complex I assembly factor TMEM126B under chronic hypoxia. Cell. Mol. Life Sci. 2018, 75, 3051–3067. [Google Scholar] [CrossRef]
- Semenza, G.L. Oxygen-dependent regulation of mitochondrial respiration by hypoxia-inducible factor 1. Biochem. J. 2007, 405, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Wise, D.R.; Ward, P.S.; Shay, J.E.S.; Cross, J.R.; Gruber, J.J.; Sachdeva, U.M.; Platt, J.M.; DeMatteo, R.G.; Simon, M.C.; Thompson, C.B. Hypoxia promotes isocitrate dehydrogenase-dependent carboxylation of -ketoglutarate to citrate to support cell growth and viability. Proc. Natl. Acad. Sci. USA 2011, 108, 19611–19616. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Bai, C.; Ruan, Y.; Liu, M.; Chu, Q.; Qiu, L.; Yang, C.; Li, B. Coordinative metabolism of glutamine carbon and nitrogen in proliferating cancer cells under hypoxia. Nat. Commun. 2019, 10, 201. [Google Scholar] [CrossRef] [PubMed]
- Bernhardt, S.; Tönsing, C.; Mitra, D.; Erdem, N.; Müller-Decker, K.; Korf, U.; Kreutz, C.; Timmer, J.; Wiemann, S. Functional proteomics of breast cancer metabolism identifies GLUL as responder during hypoxic adaptation. J. Proteome Res. 2019. [Google Scholar] [CrossRef] [PubMed]
- Fuhrmann, D.C.; Olesch, C.; Kurrle, N.; Schnütgen, F.; Zukunft, S.; Fleming, I.; Brüne, B. Chronic Hypoxia Enhances β-Oxidation-Dependent Electron Transport via Electron Transferring Flavoproteins. Cells 2019, 8, 172. [Google Scholar] [CrossRef] [PubMed]
- Chowdhury, R.; McDonough, M.A.; Mecinović, J.; Loenarz, C.; Flashman, E.; Hewitson, K.S.; Domene, C.; Schofield, C.J. Structural basis for binding of hypoxia-inducible factor to the oxygen-sensing prolyl hydroxylases. Structure 2009, 17, 981–989. [Google Scholar] [CrossRef] [PubMed]
- Baek, J.H.; Reiter, C.E.N.; Manalo, D.J.; Buehler, P.W.; Hider, R.C.; Alayash, A.I. Induction of hypoxia inducible factor (HIF-1α) in rat kidneys by iron chelation with the hydroxypyridinone, CP94. Biochim. Biophys. Acta 2011, 1809, 262–268. [Google Scholar] [CrossRef] [PubMed]
- Miles, A.L.; Burr, S.P.; Grice, G.L.; Nathan, J.A. The vacuolar-ATPase complex and assembly factors, TMEM199 and CCDC115, control HIF1α prolyl hydroxylation by regulating cellular iron levels. Elife 2017, 6. [Google Scholar] [CrossRef] [Green Version]
- Siegert, I.; Schödel, J.; Nairz, M.; Schatz, V.; Dettmer, K.; Dick, C.; Kalucka, J.; Franke, K.; Ehrenschwender, M.; Schley, G.; et al. Ferritin-Mediated Iron Sequestration Stabilizes Hypoxia-Inducible Factor-1α upon LPS Activation in the Presence of Ample Oxygen. Cell Rep. 2015, 13, 2048–2055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Volani, C.; Doerrier, C.; Demetz, E.; Haschka, D.; Paglia, G.; Lavdas, A.A.; Gnaiger, E.; Weiss, G. Dietary iron loading negatively affects liver mitochondrial function. Metallomics 2017, 9, 1634–1644. [Google Scholar] [CrossRef]
- Dziegala, M.; Kobak, K.A.; Kasztura, M.; Bania, J.; Josiak, K.; Banasiak, W.; Ponikowski, P.; Jankowska, E.A. Iron Depletion Affects Genes Encoding Mitochondrial Electron Transport Chain and Genes of Non-Oxidative Metabolism, Pyruvate Kinase and Lactate Dehydrogenase, in Primary Human Cardiac Myocytes Cultured upon Mechanical Stretch. Cells 2018, 7, 175. [Google Scholar] [CrossRef]
- Oexle, H.; Gnaiger, E.; Weiss, G. Iron-dependent changes in cellular energy metabolism: Influence on citric acid cycle and oxidative phosphorylation. Biochim. Biophys. Acta 1999, 1413, 99–107. [Google Scholar] [CrossRef]
- Pfeifhofer-Obermair, C.; Tymoszuk, P.; Petzer, V.; Weiss, G.; Nairz, M. Iron in the Tumor Microenvironment—Connecting the Dots. Front. Oncol. 2018, 8. [Google Scholar] [CrossRef]
- Bonkovsky, H.L. Iron and the liver. Am. J. Med. Sci. 1991, 301, 32–43. [Google Scholar] [CrossRef]
- Rolfs, A.; Kvietikova, I.; Gassmann, M.; Wenger, R.H. Oxygen-regulated transferrin expression is mediated by hypoxia-inducible factor-1. J. Biol. Chem. 1997, 272, 20055–20062. [Google Scholar] [CrossRef]
- Kalgaonkar, S.; Lönnerdal, B. Receptor-mediated uptake of ferritin-bound iron by human intestinal Caco-2 cells. J. Nutr. Biochem. 2009, 20, 304–311. [Google Scholar] [CrossRef]
- Torti, S.V.; Torti, F.M. Iron and cancer: More ore to be mined. Nat. Rev. Cancer 2013, 13, 342–355. [Google Scholar] [CrossRef]
- Torti, S.V.; Torti, F.M. Cellular iron metabolism in prognosis and therapy of breast cancer. Crit. Rev. Oncog. 2013, 18, 435–448. [Google Scholar] [CrossRef]
- Cortes, D.F.; Sha, W.; Hower, V.; Blekherman, G.; Laubenbacher, R.; Akman, S.; Torti, S.V.; Shulaev, V. Differential gene expression in normal and transformed human mammary epithelial cells in response to oxidative stress. Free Radic. Biol. Med. 2011, 50, 1565–1574. [Google Scholar] [CrossRef] [Green Version]
- Muckenthaler, M.U.; Galy, B.; Hentze, M.W. Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu. Rev. Nutr. 2008, 28, 197–213. [Google Scholar] [CrossRef]
- Ganz, T.; Nemeth, E. The hepcidin-ferroportin system as a therapeutic target in anemias and iron overload disorders. Hematol. Am. Soc. Hematol. Educ. Program 2011, 2011, 538–542. [Google Scholar] [CrossRef]
- Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.B.; Donovan, A.; Ward, D.M.; Ganz, T.; Kaplan, J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306, 2090–2093. [Google Scholar] [CrossRef]
- Sanchez, M.; Galy, B.; Dandekar, T.; Bengert, P.; Vainshtein, Y.; Stolte, J.; Muckenthaler, M.U.; Hentze, M.W. Iron regulation and the cell cycle: Identification of an iron-responsive element in the 3’-untranslated region of human cell division cycle 14A mRNA by a refined microarray-based screening strategy. J. Biol. Chem. 2006, 281, 22865–22874. [Google Scholar] [CrossRef]
- Gardenghi, S.; Ramos, P.; Marongiu, M.F.; Melchiori, L.; Breda, L.; Guy, E.; Muirhead, K.; Rao, N.; Roy, C.N.; Andrews, N.C.; et al. Hepcidin as a therapeutic tool to limit iron overload and improve anemia in β-thalassemic mice. J. Clin. Investig. 2010, 120, 4466–4477. [Google Scholar] [CrossRef] [Green Version]
- Ganz, T.; Nemeth, E. Hepcidin and iron homeostasis. Biochim. Biophys. Acta 2012, 1823, 1434–1443. [Google Scholar] [CrossRef] [Green Version]
- Nemeth, E.; Rivera, S.; Gabayan, V.; Keller, C.; Taudorf, S.; Pedersen, B.K.; Ganz, T. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J. Clin. Investig. 2004, 113, 1271–1276. [Google Scholar] [CrossRef] [Green Version]
- Nicolas, G.; Chauvet, C.; Viatte, L.; Danan, J.L.; Bigard, X.; Devaux, I.; Beaumont, C.; Kahn, A.; Vaulont, S. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J. Clin. Investig. 2002, 110, 1037–1044. [Google Scholar] [CrossRef] [Green Version]
- Lok, C.N.; Ponka, P. Identification of a hypoxia response element in the transferrin receptor gene. J. Biol. Chem. 1999, 274, 24147–24152. [Google Scholar] [CrossRef]
- Tacchini, L.; Bianchi, L.; Bernelli-Zazzera, A.; Cairo, G. Transferrin receptor induction by hypoxia. HIF-1-mediated transcriptional activation and cell-specific post-transcriptional regulation. J. Biol. Chem. 1999, 274, 24142–24146. [Google Scholar] [CrossRef]
- Mukhopadhyay, C.K.; Mazumder, B.; Fox, P.L. Role of hypoxia-inducible factor-1 in transcriptional activation of ceruloplasmin by iron deficiency. J. Biol. Chem. 2000, 275, 21048–21054. [Google Scholar] [CrossRef]
- Semenza, G.L.; Roth, P.H.; Fang, H.M.; Wang, G.L. Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J. Biol. Chem. 1994, 269, 23757–23763. [Google Scholar]
- Jun, J.C.; Rathore, A.; Younas, H.; Gilkes, D.; Polotsky, V.Y. Hypoxia-Inducible Factors and Cancer. Curr. Sleep Med. Rep. 2017, 3, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Nagaraju, G.P.; Bramhachari, P.V.; Raghu, G.; El-Rayes, B.F. Hypoxia inducible factor-1α: Its role in colorectal carcinogenesis and metastasis. Cancer Lett. 2015, 366, 11–18. [Google Scholar] [CrossRef]
- Tsuzuki, Y.; Fukumura, D.; Oosthuyse, B.; Koike, C.; Carmeliet, P.; Jain, R.K. Vascular endothelial growth factor (VEGF) modulation by targeting hypoxia-inducible factor-1alpha-- hypoxia response element-- VEGF cascade differentially regulates vascular response and growth rate in tumors. Cancer Res. 2000, 60, 6248–6252. [Google Scholar]
- Fang, J.; Yan, L.; Shing, Y.; Moses, M.A. HIF-1alpha-mediated up-regulation of vascular endothelial growth factor, independent of basic fibroblast growth factor, is important in the switch to the angiogenic phenotype during early tumorigenesis. Cancer Res. 2001, 61, 5731–5735. [Google Scholar]
- Xue, X.; Taylor, M.; Anderson, E.; Hao, C.; Qu, A.; Greenson, J.K.; Zimmermann, E.M.; Gonzalez, F.J.; Shah, Y.M. Hypoxia-inducible factor-2α activation promotes colorectal cancer progression by dysregulating iron homeostasis. Cancer Res. 2012, 72, 2285–2293. [Google Scholar] [CrossRef] [Green Version]
- Lee, P.J.; Jiang, B.H.; Chin, B.Y.; Iyer, N.V.; Alam, J.; Semenza, G.L.; Choi, A.M. Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia. J. Biol. Chem. 1997, 272, 5375–5381. [Google Scholar] [CrossRef]
- Fraser, S.T.; Midwinter, R.G.; Berger, B.S.; Stocker, R. Heme Oxygenase-1: A Critical Link between Iron Metabolism, Erythropoiesis, and Development. Adv. Hematol. 2011, 2011, 473709. [Google Scholar] [CrossRef]
- Peyssonnaux, C.; Nizet, V.; Johnson, R.S. Role of the hypoxia inducible factors HIF in iron metabolism. Cell Cycle 2008, 7, 28–32. [Google Scholar] [CrossRef] [Green Version]
- Le, N.T.V.; Des Richardson, R. The role of iron in cell cycle progression and the proliferation of neoplastic cells. Biochim. Biophys. Acta 2002, 1603, 31–46. [Google Scholar] [CrossRef]
- Edgren, G.; Reilly, M.; Hjalgrim, H.; Tran, T.N.; Rostgaard, K.; Adami, J.; Titlestad, K.; Shanwell, A.; Melbye, M.; Nyrén, O. Donation frequency, iron loss, and risk of cancer among blood donors. J. Natl. Cancer Inst. 2008, 100, 572–579. [Google Scholar] [CrossRef]
- Kotze, M.J.; van Velden, D.P.; van Rensburg, S.J.; Erasmus, R. Pathogenic Mechanisms Underlying Iron Deficiency and Iron Overload: New Insights for Clinical Application. EJIFCC 2009, 20, 108–123. [Google Scholar]
- Dixon, S.J.; Stockwell, B.R. The role of iron and reactive oxygen species in cell death. Nat. Chem. Biol. 2014, 10, 9–17. [Google Scholar] [CrossRef]
- Neufeld, E.J. Oral chelators deferasirox and deferiprone for transfusional iron overload in thalassemia major: New data, new questions. Blood 2006, 107, 3436–3441. [Google Scholar] [CrossRef]
- Blatt, J.; Boegel, F.; Hedlund, B.E.; Arena, V.C.; Shadduck, R.K. Failure to alter the course of acute myelogenous leukemia in the rat with subcutaneous deferoxamine. Leuk. Res. 1991, 15, 391–394. [Google Scholar] [CrossRef]
- Akam, E.A.; Chang, T.M.; Astashkin, A.V.; Tomat, E. Intracellular reduction/activation of a disulfide switch in thiosemicarbazone iron chelators. Metallomics 2014, 6, 1905–1912. [Google Scholar] [CrossRef] [Green Version]
- Akam, E.A.; Tomat, E. Targeting Iron in Colon Cancer via Glycoconjugation of Thiosemicarbazone Prochelators. Bioconjug. Chem. 2016, 27, 1807–1812. [Google Scholar] [CrossRef]
- Chang, T.M.; Tomat, E. Disulfide/thiol switches in thiosemicarbazone ligands for redox-directed iron chelation. Dalton Trans. 2013, 42, 7846–7849. [Google Scholar] [CrossRef]
- Vander Heiden, M.G. Targeting cancer metabolism: A therapeutic window opens. Nat. Rev. Drug Discov. 2011, 10, 671–684. [Google Scholar] [CrossRef]
- Lehenkari, P.P.; Kellinsalmi, M.; Näpänkangas, J.P.; Ylitalo, K.V.; Mönkkönen, J.; Rogers, M.J.; Azhayev, A.; Väänänen, H.K.; Hassinen, I.E. Further insight into mechanism of action of clodronate: Inhibition of mitochondrial ADP/ATP translocase by a nonhydrolyzable, adenine-containing metabolite. Mol. Pharmacol. 2002, 61, 1255–1262. [Google Scholar] [CrossRef]
- Kitamura, T.; Qian, B.-Z.; Soong, D.; Cassetta, L.; Noy, R.; Sugano, G.; Kato, Y.; Li, J.; Pollard, J.W. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J. Exp. Med. 2015, 212, 1043–1059. [Google Scholar] [CrossRef] [Green Version]
- Pienta, K.J.; Machiels, J.-P.; Schrijvers, D.; Alekseev, B.; Shkolnik, M.; Crabb, S.J.; Li, S.; Seetharam, S.; Puchalski, T.A.; Takimoto, C.; et al. Phase 2 study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 (CCL2), in metastatic castration-resistant prostate cancer. Invest. New Drugs 2013, 31, 760–768. [Google Scholar] [CrossRef] [PubMed]
- Brana, I.; Calles, A.; LoRusso, P.M.; Yee, L.K.; Puchalski, T.A.; Seetharam, S.; Zhong, B.; de Boer, C.J.; Tabernero, J.; Calvo, E. Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: An open-label, multicenter phase 1b study. Target. Oncol. 2015, 10, 111–123. [Google Scholar] [CrossRef] [PubMed]
- Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [PubMed]
- Mantovani, A.; Allavena, P. The interaction of anticancer therapies with tumor-associated macrophages. J. Exp. Med. 2015, 212, 435–445. [Google Scholar] [CrossRef] [Green Version]
- Komohara, Y.; Fujiwara, Y.; Ohnishi, K.; Takeya, M. Tumor-associated macrophages: Potential therapeutic targets for anti-cancer therapy. Adv. Drug Deliv. Rev. 2016, 99, 180–185. [Google Scholar] [CrossRef] [PubMed]
- Genard, G.; Lucas, S.; Michiels, C. Reprogramming of Tumor-Associated Macrophages with Anticancer Therapies: Radiotherapy versus Chemo- and Immunotherapies. Front. Immunol. 2017, 8, 828. [Google Scholar] [CrossRef] [PubMed]
- Edwards, J.P.; Emens, L.A. The multikinase inhibitor sorafenib reverses the suppression of IL-12 and enhancement of IL-10 by PGE2 in murine macrophages. Int. Immunopharmacol. 2010, 10, 1220–1228. [Google Scholar] [CrossRef]
- Hussain, S.F.; Kong, L.-Y.; Jordan, J.; Conrad, C.; Madden, T.; Fokt, I.; Priebe, W.; Heimberger, A.B. A novel small molecule inhibitor of signal transducers and activators of transcription 3 reverses immune tolerance in malignant glioma patients. Cancer Res. 2007, 67, 9630–9636. [Google Scholar] [CrossRef]
- Hsu, P.P.; Sabatini, D.M. Cancer cell metabolism: Warburg and beyond. Cell 2008, 134, 703–707. [Google Scholar] [CrossRef]
- Domblides, C.; Lartigue, L.; Faustin, B. Metabolic Stress in the Immune Function of T Cells, Macrophages and Dendritic Cells. Cells 2018, 7, 68. [Google Scholar] [CrossRef]
- Mazzone, M.; Menga, A.; Castegna, A. Metabolism and TAM functions-it takes two to tango. FEBS J. 2018, 285, 700–716. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.C.-C.; Smith, A.M.; Everts, B.; Colonna, M.; Pearce, E.L.; Schilling, J.D.; Pearce, E.J. Metabolic Reprogramming Mediated by the mTORC2-IRF4 Signaling Axis Is Essential for Macrophage Alternative Activation. Immunity 2016, 45, 817–830. [Google Scholar] [CrossRef] [PubMed]
- Pathria, P.; Louis, T.L.; Varner, J.A. Targeting Tumor-Associated Macrophages in Cancer. Trends Immunol. 2019, 40, 310–327. [Google Scholar] [CrossRef] [PubMed]
- Mertens, C.; Mora, J.; Ören, B.; Grein, S.; Winslow, S.; Scholich, K.; Weigert, A.; Malmström, P.; Forsare, C.; Fernö, M.; et al. Macrophage-derived lipocalin-2 transports iron in the tumor microenvironment. Oncoimmunology 2018, 7, e1408751. [Google Scholar] [CrossRef]
- Jung, M.; Ören, B.; Mora, J.; Mertens, C.; Dziumbla, S.; Popp, R.; Weigert, A.; Grossmann, N.; Fleming, I.; Brüne, B. Lipocalin 2 from macrophages stimulated by tumor cell-derived sphingosine 1-phosphate promotes lymphangiogenesis and tumor metastasis. Sci. Signal. 2016, 9, ra64. [Google Scholar] [CrossRef]
- Costa da Silva, M.; Breckwoldt, M.O.; Vinchi, F.; Correia, M.P.; Stojanovic, A.; Thielmann, C.M.; Meister, M.; Muley, T.; Warth, A.; Platten, M.; et al. Iron Induces Anti-tumor Activity in Tumor-Associated Macrophages. Front. Immunol. 2017, 8, 1479. [Google Scholar] [CrossRef] [PubMed]
- Jung, M.; Mertens, C.; Brüne, B. Macrophage iron homeostasis and polarization in the context of cancer. Immunobiology 2015, 220, 295–304. [Google Scholar] [CrossRef]
- Jung, M.; Mertens, C.; Tomat, E.; Brüne, B. Iron as a Central Player and Promising Target in Cancer Progression. Int. J. Mol. Sci. 2019, 20, 273. [Google Scholar] [CrossRef] [PubMed]
- Munn, D.H.; Bronte, V. Immune suppressive mechanisms in the tumor microenvironment. Curr. Opin. Immunol. 2016, 39, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shevchenko, I.; Bazhin, A.V. Metabolic Checkpoints: Novel Avenues for Immunotherapy of Cancer. Front. Immunol. 2018, 9, 1816. [Google Scholar] [CrossRef]
- Finlay, D.K.; Rosenzweig, E.; Sinclair, L.V.; Feijoo-Carnero, C.; Hukelmann, J.L.; Rolf, J.; Panteleyev, A.A.; Okkenhaug, K.; Cantrell, D.A. PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells. J. Exp. Med. 2012, 209, 2441–2453. [Google Scholar] [CrossRef] [Green Version]
- Clever, D.; Roychoudhuri, R.; Constantinides, M.G.; Askenase, M.H.; Sukumar, M.; Klebanoff, C.A.; Eil, R.L.; Hickman, H.D.; Yu, Z.; Pan, J.H.; et al. Oxygen Sensing by T Cells Establishes an Immunologically Tolerant Metastatic Niche. Cell 2016, 166, 1117–1131. [Google Scholar] [CrossRef]
- Scharping, N.E.; Menk, A.V.; Moreci, R.S.; Whetstone, R.D.; Dadey, R.E.; Watkins, S.C.; Ferris, R.L.; Delgoffe, G.M. The Tumor Microenvironment Represses T Cell Mitochondrial Biogenesis to Drive Intratumoral T Cell Metabolic Insufficiency and Dysfunction. Immunity 2016, 45, 701–703. [Google Scholar] [CrossRef] [Green Version]
- Facciabene, A.; Peng, X.; Hagemann, I.S.; Balint, K.; Barchetti, A.; Wang, L.-P.; Gimotty, P.A.; Gilks, C.B.; Lal, P.; Zhang, L.; et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature 2011, 475, 226–230. [Google Scholar] [CrossRef] [PubMed]
- Ren, L.; Yu, Y.; Wang, L.; Zhu, Z.; Lu, R.; Yao, Z. Hypoxia-induced CCL28 promotes recruitment of regulatory T cells and tumor growth in liver cancer. Oncotarget 2016, 7, 75763–75773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yan, M.; Jene, N.; Byrne, D.; Millar, E.K.A.; O’Toole, S.A.; McNeil, C.M.; Bates, G.J.; Harris, A.L.; Banham, A.H.; Sutherland, R.L.; et al. Recruitment of regulatory T cells is correlated with hypoxia-induced CXCR4 expression, and is associated with poor prognosis in basal-like breast cancers. Breast Cancer Res. 2011, 13, R47. [Google Scholar] [CrossRef] [PubMed]
- Angelin, A.; Gil-de-Gómez, L.; Dahiya, S.; Jiao, J.; Guo, L.; Levine, M.H.; Wang, Z.; Quinn, W.J.; Kopinski, P.K.; Wang, L.; et al. Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. Cell Metabolism 2017, 25, 1282–1293. [Google Scholar] [CrossRef]
- Chang, C.-H.; Curtis, J.D.; Maggi, L.B.; Faubert, B.; Villarino, A.V.; O’Sullivan, D.; Huang, S.C.-C.; van der Windt, G.J.W.; Blagih, J.; Qiu, J.; et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 2013, 153, 1239–1251. [Google Scholar] [CrossRef]
- 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.W.; et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Ertl, H.C.J. Starved and Asphyxiated: How Can CD8(+) T Cells within a Tumor Microenvironment Prevent Tumor Progression. Front. Immunol. 2016, 7, 32. [Google Scholar] [CrossRef]
- Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016, 165, 35–44. [Google Scholar] [CrossRef] [PubMed]
- Scharping, N.E.; Menk, A.V.; Whetstone, R.D.; Zeng, X.; Delgoffe, G.M. Efficacy of PD-1 Blockade Is Potentiated by Metformin-Induced Reduction of Tumor Hypoxia. Cancer Immunol. Res. 2017, 5, 9–16. [Google Scholar] [CrossRef] [PubMed]
- Najjar, Y.G.; Menk, A.V.; Sander, C.; Rao, U.; Karunamurthy, A.; Bhatia, R.; Zhai, S.; Kirkwood, J.M.; Delgoffe, G.M. Tumor cell oxidative metabolism as a barrier to PD-1 blockade immunotherapy in melanoma. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed]
- Schiffmann, L.M.; Brunold, M.; Liwschitz, M.; Goede, V.; Loges, S.; Wroblewski, M.; Quaas, A.; Alakus, H.; Stippel, D.; Bruns, C.J.; et al. A combination of low-dose bevacizumab and imatinib enhances vascular normalisation without inducing extracellular matrix deposition. Br. J. Cancer 2017, 116, 600–608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jamieson, S.M.; Tsai, P.; Kondratyev, M.K.; Budhani, P.; Liu, A.; Senzer, N.N.; Chiorean, E.G.; Jalal, S.I.; Nemunaitis, J.J.; Kee, D.; et al. Evofosfamide for the treatment of human papillomavirus-negative head and neck squamous cell carcinoma. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed]
- Hatfield, S.M.; Kjaergaard, J.; Lukashev, D.; Schreiber, T.H.; Belikoff, B.; Abbott, R.; Sethumadhavan, S.; Philbrook, P.; Ko, K.; Cannici, R.; et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci. Transl. Med. 2015, 7, 277ra30. [Google Scholar] [CrossRef]
- Zhao, Y.; Wu, T.; Shao, S.; Shi, B.; Zhao, Y. Phenotype, development, and biological function of myeloid-derived suppressor cells. Oncoimmunology 2016, 5, e1004983. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
- Parker, K.H.; Beury, D.W.; Ostrand-Rosenberg, S. Myeloid-Derived Suppressor Cells: Critical Cells Driving Immune Suppression in the Tumor Microenvironment. Adv. Cancer Res. 2015, 128, 95–139. [Google Scholar] [CrossRef] [Green Version]
- Wesolowski, R.; Markowitz, J.; Carson, W.E. Myeloid derived suppressor cells—A new therapeutic target in the treatment of cancer. J. Immunother. Cancer 2013, 1, 10. [Google Scholar] [CrossRef]
- Marvel, D.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected. J. Clin. Investig. 2015, 125, 3356–3364. [Google Scholar] [CrossRef] [PubMed]
- Ugel, S.; de Sanctis, F.; Mandruzzato, S.; Bronte, V. Tumor-induced myeloid deviation: When myeloid-derived suppressor cells meet tumor-associated macrophages. J. Clin. Investig. 2015, 125, 3365–3376. [Google Scholar] [CrossRef] [PubMed]
- Fleming, V.; Hu, X.; Weber, R.; Nagibin, V.; Groth, C.; Altevogt, P.; Utikal, J.; Umansky, V. Targeting Myeloid-Derived Suppressor Cells to Bypass Tumor-Induced Immunosuppression. Front. Immunol. 2018, 9, 398. [Google Scholar] [CrossRef] [PubMed]
- DeNardo, D.G.; Brennan, D.J.; Rexhepaj, E.; Ruffell, B.; Shiao, S.L.; Madden, S.F.; Gallagher, W.M.; Wadhwani, N.; Keil, S.D.; Junaid, S.A.; et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011, 1, 54–67. [Google Scholar] [CrossRef] [PubMed]
- Hossain, F.; Al-Khami, A.A.; Wyczechowska, D.; Hernandez, C.; Zheng, L.; Reiss, K.; Valle, L.D.; Trillo-Tinoco, J.; Maj, T.; Zou, W.; et al. Inhibition of Fatty Acid Oxidation Modulates Immunosuppressive Functions of Myeloid-Derived Suppressor Cells and Enhances Cancer Therapies. Cancer Immunol. Res. 2015, 3, 1236–1247. [Google Scholar] [CrossRef] [Green Version]
- Kusmartsev, S.; Nagaraj, S.; Gabrilovich, D.I. Tumor-associated CD8+ T cell tolerance induced by bone marrow-derived immature myeloid cells. J. Immunol. 2005, 175, 4583–4592. [Google Scholar] [CrossRef]
- Yu, J.; Du, W.; Yan, F.; Wang, Y.; Li, H.; Cao, S.; Yu, W.; Shen, C.; Liu, J.; Ren, X. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J. Immunol. 2013, 190, 3783–3797. [Google Scholar] [CrossRef] [PubMed]
- Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [Green Version]
© 2019 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mora, J.; Mertens, C.; Meier, J.K.; Fuhrmann, D.C.; Brüne, B.; Jung, M. Strategies to Interfere with Tumor Metabolism through the Interplay of Innate and Adaptive Immunity. Cells 2019, 8, 445. https://doi.org/10.3390/cells8050445
Mora J, Mertens C, Meier JK, Fuhrmann DC, Brüne B, Jung M. Strategies to Interfere with Tumor Metabolism through the Interplay of Innate and Adaptive Immunity. Cells. 2019; 8(5):445. https://doi.org/10.3390/cells8050445
Chicago/Turabian StyleMora, Javier, Christina Mertens, Julia K. Meier, Dominik C. Fuhrmann, Bernhard Brüne, and Michaela Jung. 2019. "Strategies to Interfere with Tumor Metabolism through the Interplay of Innate and Adaptive Immunity" Cells 8, no. 5: 445. https://doi.org/10.3390/cells8050445
APA StyleMora, J., Mertens, C., Meier, J. K., Fuhrmann, D. C., Brüne, B., & Jung, M. (2019). Strategies to Interfere with Tumor Metabolism through the Interplay of Innate and Adaptive Immunity. Cells, 8(5), 445. https://doi.org/10.3390/cells8050445