Manipulation of Metabolic Pathways and Its Consequences for Anti-Tumor Immunity: A Clinical Perspective
Abstract
1. The Tumor Microenvironment and Immune Checkpoint Inhibitors
2. The Implications of Metabolic Syndrome for Targeting Metabolic Pathways within the Tumor Microenvironment
3. The Metabolic Pathways in the Tumor Microenvironment: Glucose and Lipid
4. The Metabolic Pathways in the Tumor Microenvironment: Amino Acids and Metabolic Intermediates
5. The Effects of Exosomes and Immune Checkpoint Inhibitors on Metabolic Pathways
6. Conclusions
Funding
Conflicts of Interest
Abbreviations
TME | tumor microenvironment |
Treg | regulatory T |
TGF | transforming growth factor |
IL | interleukin |
CTLA-4 | cytotoxic T-lymphocyte-associated antigen 4 |
PD-1 | programmed death 1 |
DC | dendritic cell |
TAM | tumor-associated macrophage |
ICI | immune checkpoint inhibitor |
IGF | insulin-like growth factor |
PI3K | phosphatidylinositol 3-kinase |
APC | antigen-presenting cell |
FAO | fatty acid oxidation |
FAS | fatty acid synthesis |
ACC1 | acetyl-CoA carboxylase 1 |
ACAT | acetyl-CoA acetyltransferase |
ASCT | alanine serine cysteine transporter |
DON | 6-diazo-5-oxo-L-norleucine |
IDO | indoleamine-2,3-dioxygenase |
LDH | lactate dehydrogenase |
OXPHOS | oxidative phosphorylation |
References
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
- Zhang, Y.; Ertl, H.C. Starved and asphyxiated: How can CD8+ T cells within a tumor microenvironment prevent tumor progression. Front. Immunol. 2016, 7, 32. [Google Scholar] [CrossRef] [PubMed]
- Gilkes, D.M.; Semenza, G.L.; Wirtz, D. Hypoxia and the extracellular matrix: Drivers of tumour metastasis. Nat. Rev. Cancer 2014, 14, 430–439. [Google Scholar] [CrossRef] [PubMed]
- Meseure, D.; Alsibai, K.D.; Nicolas, A. Pivotal role of pervasive neoplastic and stromal cells reprogramming in circulating tumor cells dissemination and metastatic colonization. Cancer Microenviron. 2014, 7, 95–115. [Google Scholar] [CrossRef] [PubMed]
- Alkasalias, T.; Moyano-Galceran, L.; Arsenian-Henriksson, M.; Lehti, K. Fibroblasts in the tumor microenvironment: Shield or spear? Int. J. Mol. Sci. 2018, 19, 1532. [Google Scholar] [CrossRef]
- Poltavets, V.; Kochetkova, M.; Pitson, S.M.; Samuel, M.S. The role of the extracellular matrix and its molecular and cellular regulators in cancer cell plasticity. Front. Oncol. 2018, 8, 431. [Google Scholar] [CrossRef]
- Almendros, I.; Gileles-Hillel, A.; Khalyfa, A.; Wang, Y.; Zhang, S.X.; Carreras, A.; Farré, R.; Gozal, D. Adipose tissue macrophage polarization by intermittent hypoxia in a mouse model of OSA: Effect of tumor microenvironment. Cancer Lett. 2015, 361, 233–239. [Google Scholar] [CrossRef]
- Wang, M.; Zhao, J.; Zhang, L.; Wei, F.; Lian, Y.; Wu, Y.; Gong, Z.; Zhang, S.; Zhou, J.; Cao, K. Role of tumor microenvironment in tumorigenesis. J. Cancer 2017, 8, 761. [Google Scholar] [CrossRef]
- Ghajar, C.M.; Peinado, H.; Mori, H.; Matei, I.R.; Evason, K.J.; Brazier, H.; Almeida, D.; Koller, A.; Hajjar, K.A.; Stainier, D.Y. The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 2013, 15, 807–817. [Google Scholar] [CrossRef]
- Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
- Fridman, W.H.; Pagès, F.; Sautes-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306. [Google Scholar] [CrossRef]
- Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef] [PubMed]
- Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
- Nishikawa, H.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Curr. Opin. Immunol. 2014, 27, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Sakaguchi, S.; Yamaguchi, T.; Nomura, T.; Ono, M. Regulatory T cells and immune tolerance. Cell 2008, 133, 775–787. [Google Scholar] [CrossRef] [PubMed]
- deLeeuw, R.J.; Kost, S.E.; Kakal, J.A.; Nelson, B.H. The prognostic value of FoxP3+ tumor-infiltrating lymphocytes in cancer: A critical review of the literature. Clin. Cancer Res. 2012, 18, 3022–3029. [Google Scholar] [CrossRef]
- Shevach, E.M. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity 2009, 30, 636–645. [Google Scholar] [CrossRef] [PubMed]
- Schmidt, A.; Oberle, N.; Krammer, P.H. Molecular mechanisms of treg-mediated T cell suppression. Front. Immunol. 2012, 3, 51. [Google Scholar] [CrossRef] [PubMed]
- Engelhardt, J.J.; Boldajipour, B.; Beemiller, P.; Pandurangi, P.; Sorensen, C.; Werb, Z.; Egeblad, M.; Krummel, M.F. Marginating dendritic cells of the tumor microenvironment cross-present tumor antigens and stably engage tumor-specific T cells. Cancer Cell 2012, 21, 402–417. [Google Scholar] [CrossRef] [PubMed]
- Cao, X.; Cai, S.F.; Fehniger, T.A.; Song, J.; Collins, L.I.; Piwnica-Worms, D.R.; Ley, T.J. Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity 2007, 27, 635–646. [Google Scholar] [CrossRef]
- Veldhoen, M.; Moncrieffe, H.; Hocking, R.J.; Atkins, C.J.; Stockinger, B. Modulation of dendritic cell function by naive and regulatory CD4+ T cells. J. Immunol. 2006, 176, 6202–6210. [Google Scholar] [CrossRef] [PubMed]
- Grossman, W.J.; Verbsky, J.W.; Barchet, W.; Colonna, M.; Atkinson, J.P.; Ley, T.J. Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity 2004, 21, 589–601. [Google Scholar] [CrossRef] [PubMed]
- Yan, Z.; Garg, S.K.; Banerjee, R. Regulatory T cells interfere with glutathione metabolism in dendritic cells and T cells. J. Biol. Chem. 2010, 285, 41525–41532. [Google Scholar] [CrossRef] [PubMed]
- Vignali, D.A.; Collison, L.W.; Workman, C.J. How regulatory T cells work. Nat. Rev. Immunol. 2008, 8, 523–532. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Franco, F.; Ho, P.-C. Metabolic regulation of Tregs in cancer: Opportunities for immunotherapy. Trends Cancer 2017, 3, 583–592. [Google Scholar] [CrossRef]
- DeNardo, D.G.; Barreto, J.B.; Andreu, P.; Vasquez, L.; Tawfik, D.; Kolhatkar, N.; Coussens, L.M. CD4+ T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 2009, 16, 91–102. [Google Scholar] [CrossRef]
- Shiao, S.L.; Ruffell, B.; DeNardo, D.G.; Faddegon, B.A.; Park, C.C.; Coussens, L.M. TH2-polarized CD4+ T cells and macrophages limit efficacy of radiotherapy. Cancer Immunol. Res. 2015, 3, 518–525. [Google Scholar] [CrossRef]
- De Monte, L.; Wörmann, S.; Brunetto, E.; Heltai, S.; Magliacane, G.; Reni, M.; Paganoni, A.M.; Recalde, H.; Mondino, A.; Falconi, M. Basophil recruitment into tumor-draining lymph nodes correlates with Th2 inflammation and reduced survival in pancreatic cancer patients. Cancer Res. 2016, 76, 1792–1803. [Google Scholar] [CrossRef]
- Kumar, V.; Cheng, P.; Condamine, T.; Mony, S.; Languino, L.R.; McCaffrey, J.C.; Hockstein, N.; Guarino, M.; Masters, G.; Penman, E. CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity 2016, 44, 303–315. [Google Scholar] [CrossRef]
- Franklin, R.A.; Liao, W.; Sarkar, A.; Kim, M.V.; Bivona, M.R.; Liu, K.; Pamer, E.G.; Li, M.O. The cellular and molecular origin of tumor-associated macrophages. Science 2014, 344, 921–925. [Google Scholar] [CrossRef]
- Sangaletti, S.; Di Carlo, E.; Gariboldi, S.; Miotti, S.; Cappetti, B.; Parenza, M.; Rumio, C.; Brekken, R.A.; Chiodoni, C.; Colombo, M.P. Macrophage-derived SPARC bridges tumor cell-extracellular matrix interactions toward metastasis. Cancer Res. 2008, 68, 9050–9059. [Google Scholar] [CrossRef] [PubMed]
- Nucera, S.; Biziato, D.; De Palma, M. The interplay between macrophages and angiogenesis in development, tissue injury and regeneration. Int. J. Dev. Biol. 2011, 55, 495–503. [Google Scholar] [CrossRef] [PubMed]
- Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [PubMed]
- Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399. [Google Scholar] [CrossRef] [PubMed]
- Johnstone, R.M.; Adam, M.; Hammond, J.; Orr, L.; Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar]
- Kim, H.K.; Song, K.; Park, Y.; Kang, Y.; Lee, Y.; Lee, K.; Ryu, K.; Bae, J.; Kim, S. Elevated levels of circulating platelet microparticles, VEGF, IL-6 and RANTES in patients with gastric cancer: Possible role of a metastasis predictor. Eur. J. Cancer 2003, 39, 184–191. [Google Scholar] [CrossRef]
- Kharaziha, P.; Ceder, S.; Li, Q.; Panaretakis, T. Tumor cell-derived exosomes: A message in a bottle. Biochim. Et Biophys. Acta (Bba)-Rev. Cancer 2012, 1826, 103–111. [Google Scholar] [CrossRef]
- Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423. [Google Scholar] [CrossRef]
- Zhang, H.-G.; Grizzle, W.E. Exosomes: A novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. Am. J. Pathol. 2014, 184, 28–41. [Google Scholar] [CrossRef]
- Gatalica, Z.; Snyder, C.; Maney, T.; Ghazalpour, A.; Holterman, D.A.; Xiao, N.; Overberg, P.; Rose, I.; Basu, G.D.; Vranic, S. Programmed cell death 1 (PD-1) and its ligand (PD-L1) in common cancers and their correlation with molecular cancer type. Cancer Epidemiol. Prev. Biomark. 2014, 23, 2965–2970. [Google Scholar] [CrossRef] [PubMed]
- Wang, A.; Wang, H.; Liu, Y.; Zhao, M.; Zhang, H.; Lu, Z.; Fang, Y.; Chen, X.; Liu, G. The prognostic value of PD-L1 expression for non-small cell lung cancer patients: A meta-analysis. Eur. J. Surg. Oncol. (Ejso) 2015, 41, 450–456. [Google Scholar] [CrossRef] [PubMed]
- Muenst, S.; Schaerli, A.; Gao, F.; Däster, S.; Trella, E.; Droeser, R.; Muraro, M.; Zajac, P.; Zanetti, R.; Gillanders, W. Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res. Treat. 2014, 146, 15–24. [Google Scholar] [CrossRef] [PubMed]
- Kuang, D.-M.; Zhao, Q.; Peng, C.; Xu, J.; Zhang, J.-P.; Wu, C.; Zheng, L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 2009, 206, 1327–1337. [Google Scholar] [CrossRef] [PubMed]
- Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell 2015, 27, 450–461. [Google Scholar] [CrossRef]
- Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef]
- Han, Y.; Yang, Y.; Chen, Z.; Jiang, Z.; Gu, Y.; Liu, Y.; Xu, S.; Lin, C.; Pan, Z.; Zhou, W. Human hepatocellular carcinoma-infiltrating CD4+ CD69+ Foxp3− regulatory T cell suppresses T cell response via membrane-bound TGF-β1. J. Mol. Med. 2014, 92, 539–550. [Google Scholar] [CrossRef]
- Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E. Nivolumab versus docetaxel in advanced nonsquamous non–small-cell lung cancer. New Engl. J. Med. 2015, 373, 1627–1639. [Google Scholar] [CrossRef]
- Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L. Pembrolizumab for the treatment of non–small-cell lung cancer. New Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef]
- Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C. Improved survival with ipilimumab in patients with metastatic melanoma. New Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. New Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef]
- Schadendorf, D.; Hodi, F.S.; Robert, C.; Weber, J.S.; Margolin, K.; Hamid, O.; Patt, D.; Chen, T.-T.; Berman, D.M.; Wolchok, J.D. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J. Clin. Oncol. 2015, 33, 1889. [Google Scholar] [CrossRef] [PubMed]
- Robert, C.; Thomas, L.; Bondarenko, I.; O’Day, S.; Weber, J.; Garbe, C.; Lebbe, C.; Baurain, J.-F.; Testori, A.; Grob, J.-J. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. New Engl. J. Med. 2011, 364, 2517–2526. [Google Scholar] [CrossRef] [PubMed]
- Jenkins, R.W.; Barbie, D.A.; Flaherty, K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 2018, 118, 9–16. [Google Scholar] [CrossRef]
- Darvin, P.; Toor, S.M.; Nair, V.S.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Aguilar, M.; Bhuket, T.; Torres, S.; Liu, B.; Wong, R.J. Prevalence of the metabolic syndrome in the United States, 2003-2012. Jama 2015, 313, 1973–1974. [Google Scholar] [CrossRef]
- Ranasinghe, P.; Mathangasinghe, Y.; Jayawardena, R.; Hills, A.; Misra, A. Prevalence and trends of metabolic syndrome among adults in the asia-pacific region: A systematic review. Bmc Public Health 2017, 17, 101. [Google Scholar] [CrossRef]
- Dunkley, A.; Charles, K.; Gray, L.; Camosso-Stefinovic, J.; Davies, M.; Khunti, K. Effectiveness of interventions for reducing diabetes and cardiovascular disease risk in people with metabolic syndrome: Systematic review and mixed treatment comparison meta-analysis. Diabetesobesity Metab. 2012, 14, 616–625. [Google Scholar] [CrossRef]
- Lorenzo, C.; Okoloise, M.; Williams, K.; Stern, M.P.; Haffner, S.M. The metabolic syndrome as predictor of type 2 diabetes: The San Antonio heart study. Diabetes Care 2003, 26, 3153–3159. [Google Scholar] [CrossRef] [PubMed]
- Grundy, S.M. Metabolic syndrome: Connecting and reconciling cardiovascular and diabetes worlds. J. Am. Coll. Cardiol. 2006, 47, 1093–1100. [Google Scholar] [CrossRef]
- Wilson, P.W.; D’Agostino, R.B.; Parise, H.; Sullivan, L.; Meigs, J.B. Metabolic syndrome as a precursor of cardiovascular disease and type 2 diabetes mellitus. Circulation 2005, 112, 3066–3072. [Google Scholar] [CrossRef] [PubMed]
- Arnold, M.; Pandeya, N.; Byrnes, G.; Renehan, A.G.; Stevens, G.A.; Ezzati, M.; Ferlay, J.; Miranda, J.J.; Romieu, I.; Dikshit, R. Global burden of cancer attributable to high body-mass index in 2012: A population-based study. Lancet Oncol. 2015, 16, 36–46. [Google Scholar] [CrossRef]
- Calle, E.E.; Kaaks, R. Overweight, obesity and cancer: Epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 2004, 4, 579–591. [Google Scholar] [CrossRef]
- Pearson-Stuttard, J.; Zhou, B.; Kontis, V.; Bentham, J.; Gunter, M.J.; Ezzati, M. Retracted: Worldwide Burden of Cancer Attributable to Diabetes and High Body-Mass Index: A Comparative Risk Assessment; Elsevier: Amsterdam, The Netherlands, 2018. [Google Scholar]
- Calle, E.E.; Rodriguez, C.; Walker-Thurmond, K.; Thun, M.J. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of US adults. New Engl. J. Med. 2003, 348, 1625–1638. [Google Scholar] [CrossRef] [PubMed]
- Barone, B.B.; Yeh, H.-C.; Snyder, C.F.; Peairs, K.S.; Stein, K.B.; Derr, R.L.; Wolff, A.C.; Brancati, F.L. Long-term all-cause mortality in cancer patients with preexisting diabetes mellitus: A systematic review and meta-analysis. Jama 2008, 300, 2754–2764. [Google Scholar] [CrossRef] [PubMed]
- Lipscombe, L.L.; Goodwin, P.J.; Zinman, B.; McLaughlin, J.R.; Hux, J.E. The impact of diabetes on survival following breast cancer. Breast Cancer Res. Treat. 2008, 109, 389–395. [Google Scholar] [CrossRef]
- Chen, Y.; Wu, F.; Saito, E.; Lin, Y.; Song, M.; Luu, H.N.; Gupta, P.C.; Sawada, N.; Tamakoshi, A.; Shu, X.-O. Association between type 2 diabetes and risk of cancer mortality: A pooled analysis of over 771,000 individuals in the Asia Cohort Consortium. Diabetologia 2017, 60, 1022–1032. [Google Scholar] [CrossRef]
- Vigneri, P.; Frasca, F.; Sciacca, L.; Pandini, G.; Vigneri, R. Diabetes and cancer. Endocr. -Relat. Cancer 2009, 16, 1103–1123. [Google Scholar] [CrossRef]
- Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
- Catrina, S.-B.; Okamoto, K.; Pereira, T.; Brismar, K.; Poellinger, L. Hyperglycemia regulates hypoxia-inducible factor-1α protein stability and function. Diabetes 2004, 53, 3226–3232. [Google Scholar] [CrossRef]
- Ma, J.; Li, H.; Giovannucci, E.; Mucci, L.; Qiu, W.; Nguyen, P.L.; Gaziano, J.M.; Pollak, M.; Stampfer, M.J. Prediagnostic body-mass index, plasma C-peptide concentration, and prostate cancer-specific mortality in men with prostate cancer: A long-term survival analysis. Lancet Oncol. 2008, 9, 1039–1047. [Google Scholar] [CrossRef]
- Wolpin, B.M.; Meyerhardt, J.A.; Chan, A.T.; Ng, K.; Chan, J.A.; Wu, K.; Pollak, M.N.; Giovannucci, E.L.; Fuchs, C.S. Insulin, the insulin-like growth factor axis, and mortality in patients with nonmetastatic colorectal cancer. J. Clin. Oncol. 2009, 27, 176. [Google Scholar] [CrossRef] [PubMed]
- Perseghin, G.; Calori, G.; Lattuada, G.; Ragogna, F.; Dugnani, E.; Garancini, M.P.; Crosignani, P.; Villa, M.; Bosi, E.; Ruotolo, G. Insulin resistance/hyperinsulinemia and cancer mortality: The Cremona study at the 15th year of follow-up. Acta Diabetol. 2012, 49, 421–428. [Google Scholar] [CrossRef] [PubMed]
- Tsujimoto, T.; Kajio, H.; Sugiyama, T. Association between hyperinsulinemia and increased risk of cancer death in nonobese and obese people: A population-based observational study. Int. J. Cancer 2017, 141, 102–111. [Google Scholar] [CrossRef] [PubMed]
- Coppack, S.W. Pro-inflammatory cytokines and adipose tissue. Proc. Nutr. Soc. 2001, 60, 349–356. [Google Scholar] [CrossRef]
- Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003, 112, 1796–1808. [Google Scholar] [CrossRef]
- Pollak, M. Insulin and insulin-like growth factor signalling in neoplasia. Nat. Rev. Cancer 2008, 8, 915–928. [Google Scholar] [CrossRef]
- Brahmkhatri, V.P.; Prasanna, C.; Atreya, H.S. Insulin-like growth factor system in cancer: Novel targeted therapies. Biomed Res. Int. 2015, 2015. [Google Scholar] [CrossRef]
- Le Roith, D. Insulin-like growth factors. New Engl. J. Med. 1997, 336, 633–640. [Google Scholar] [CrossRef]
- Rinderknecht, E.; Humbel, R.E. The amino acid sequence of human insulin-like growth factor I and its structural homology with proinsulin. J. Biol. Chem. 1978, 253, 2769–2776. [Google Scholar]
- LeRoith, D.; Werner, H.; Beitner-Johnson, D.; Roberts, C.T., Jr. Molecular and cellular aspects of the insulin-like growth factor I receptor. Endocr. Rev. 1995, 16, 143–163. [Google Scholar] [CrossRef]
- Kim, J.J.; Accili, D. Signalling through IGF-I and insulin receptors: Where is the specificity? Growth Horm. Igf Res. 2002, 12, 84–90. [Google Scholar] [CrossRef]
- Chitnis, M.M.; Yuen, J.S.; Protheroe, A.S.; Pollak, M.; Macaulay, V.M. The type 1 insulin-like growth factor receptor pathway. Clin. Cancer Res. 2008, 14, 6364–6370. [Google Scholar] [CrossRef]
- Hakuno, F.; Takahashi, S.-I. 40 years of IGF1: IGF1 receptor signaling pathways. J. Mol. Endocrinol. 2018, 61, T69–T86. [Google Scholar] [CrossRef]
- Robey, R.B.; Hay, N. Is Akt the “Warburg kinase”?—Akt-energy metabolism interactions and oncogenesis. Semin. Cancer Biol. 2009, 19, 25–31. [Google Scholar] [CrossRef]
- Cully, M.; You, H.; Levine, A.J.; Mak, T.W. Beyond PTEN mutations: The PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat. Rev. Cancer 2006, 6, 184–192. [Google Scholar] [CrossRef]
- Dai, N.; Ji, F.; Wright, J.; Minichiello, L.; Sadreyev, R.; Avruch, J. IGF2 mRNA binding protein-2 is a tumor promoter that drives cancer proliferation through its client mRNAs IGF2 and HMGA1. Elife 2017, 6, e27155. [Google Scholar] [CrossRef]
- Shukla, A.; Grisouard, J.; Ehemann, V.; Hermani, A.; Enzmann, H.; Mayer, D. Analysis of signaling pathways related to cell proliferation stimulated by insulin analogs in human mammary epithelial cell lines. Endocr. -Relat. Cancer 2009, 16, 429–441. [Google Scholar] [CrossRef]
- Shen, K.; Cui, D.; Sun, L.; Lu, Y.; Han, M.; Liu, J. Inhibition of IGF-IR increases chemosensitivity in human colorectal cancer cells through MRP-2 promoter suppression. J. Cell. Biochem. 2012, 113, 2086–2097. [Google Scholar] [CrossRef]
- Rochester, M.A.; Riedemann, J.; Hellawell, G.O.; Brewster, S.F.; Macaulay, V.M. Silencing of the IGF1R gene enhances sensitivity to DNA-damaging agents in both PTEN wild-type and mutant human prostate cancer. Cancer Gene Ther. 2005, 12, 90–100. [Google Scholar] [CrossRef]
- Chitnis, M.M.; Lodhia, K.A.; Aleksic, T.; Gao, S.; Protheroe, A.S.; Macaulay, V.M. IGF-1R inhibition enhances radiosensitivity and delays double-strand break repair by both non-homologous end-joining and homologous recombination. Oncogene 2014, 33, 5262–5273. [Google Scholar] [CrossRef]
- Ireland, L.; Santos, A.; Ahmed, M.S.; Rainer, C.; Nielsen, S.R.; Quaranta, V.; Weyer-Czernilofsky, U.; Engle, D.D.; Perez-Mancera, P.A.; Coupland, S.E. Chemoresistance in pancreatic cancer is driven by stroma-derived insulin-like growth factors. Cancer Res. 2016, 76, 6851–6863. [Google Scholar] [CrossRef]
- Nowak-Sliwinska, P.; van Beijnum, J.R.; Huijbers, E.J.; Gasull, P.C.; Mans, L.; Bex, A.; Griffioen, A.W. Oncofoetal insulin receptor isoform A marks the tumour endothelium; an underestimated pathway during tumour angiogenesis and angiostatic treatment. Br. J. Cancer 2019, 120, 218–228. [Google Scholar] [CrossRef]
- Ooi, G.T.; Tseng, L.; Tran, M.Q.; Rechler, M.M. Insulin rapidly decreases insulin-like growth factor-binding protein-1 gene transcription in streptozotocin-diabetic rats. Mol. Endocrinol. 1992, 6, 2219–2228. [Google Scholar]
- Powell, D.R.; Suwanichkul, A.; Cubbage, M.L.; DePaolis, L.A.; Snuggs, M.B.; Lee, P. Insulin inhibits transcription of the human gene for insulin-like growth factor-binding protein-1. J. Biol. Chem. 1991, 266, 18868–18876. [Google Scholar]
- Renehan, A.G.; Frystyk, J.; Flyvbjerg, A. Obesity and cancer risk: The role of the insulin–IGF axis. Trends Endocrinol. Metab. 2006, 17, 328–336. [Google Scholar] [CrossRef]
- Weinstein, D.; Simon, M.; Yehezkel, E.; Laron, Z.; Werner, H. Insulin analogues display IGF-I-like mitogenic and anti-apoptotic activities in cultured cancer cells. Diabetes/Metab. Res. Rev. 2009, 25, 41–49. [Google Scholar] [CrossRef]
- van Niekerk, G.; Christowitz, C.; Conradie, D.; Engelbrecht, A.-M. Insulin as an immunomodulatory hormone. Cytokine Growth Factor Rev. 2019. [Google Scholar] [CrossRef]
- Wensveen, F.M.; Šestan, M.; Turk Wensveen, T.; Polić, B. ‘Beauty and the beast’in infection: How immune–endocrine interactions regulate systemic metabolism in the context of infection. Eur. J. Immunol. 2019, 49, 982–995. [Google Scholar] [CrossRef]
- Bilbao, D.; Luciani, L.; Johannesson, B.; Piszczek, A.; Rosenthal, N. Insulin-like growth factor-1 stimulates regulatory T cells and suppresses autoimmune disease. Embo Mol. Med. 2014, 6, 1423–1435. [Google Scholar] [CrossRef]
- Johannesson, B.; Sattler, S.; Semenova, E.; Pastore, S.; Kennedy-Lydon, T.M.; Sampson, R.D.; Schneider, M.D.; Rosenthal, N.; Bilbao, D. Insulin-like growth factor-1 induces regulatory T cell-mediated suppression of allergic contact dermatitis in mice. Dis. Models Mech. 2014, 7, 977–985. [Google Scholar] [CrossRef]
- Miyagawa, I.; Nakayamada, S.; Nakano, K.; Yamagata, K.; Sakata, K.; Yamaoka, K.; Tanaka, Y. Induction of regulatory T cells and its regulation with insulin-like growth factor/insulin-like growth factor binding protein-4 by human mesenchymal stem cells. J. Immunol. 2017, 199, 1616–1625. [Google Scholar] [CrossRef]
- Kooijman, R.; Coppens, A. Insulin-like growth factor-I stimulates IL-10 production in human T cells. J. Leukoc. Biol. 2004, 76, 862–867. [Google Scholar] [CrossRef]
- Lu, M.C.; Yu, C.L.; Chen, H.C.; Yu, H.C.; Huang, H.B.; Lai, N.S. Increased miR-223 expression in T cells from patients with rheumatoid arthritis leads to decreased insulin-like growth factor-1-mediated interleukin-10 production. Clin. Exp. Immunol. 2014, 177, 641–651. [Google Scholar] [CrossRef]
- Barrett, J.P.; Minogue, A.M.; Falvey, A.; Lynch, M.A. Involvement of IGF-1 and Akt in M1/M2 activation state in bone marrow-derived macrophages. Exp. Cell Res. 2015, 335, 258–268. [Google Scholar] [CrossRef]
- Schabbauer, G.; Sharif, O.; Brunner, J.S.; Vogel, A. Macrophage rewiring by nutrient associated PI3K dependent pathways. Front. Immunol. 2019, 10, 2002. [Google Scholar]
- Ma, J.; Giovannucci, E.; Pollak, M.; Leavitt, A.; Tao, Y.; Gaziano, J.M.; Stampfer, M.J. A prospective study of plasma C-peptide and colorectal cancer risk in men. J. Natl. Cancer Inst. 2004, 96, 546–553. [Google Scholar] [CrossRef]
- Goodwin, P.J.; Ennis, M.; Pritchard, K.I.; Trudeau, M.E.; Koo, J.; Madarnas, Y.; Hartwick, W.; Hoffman, B.; Hood, N. Fasting insulin and outcome in early-stage breast cancer: Results of a prospective cohort study. J. Clin. Oncol. 2002, 20, 42–51. [Google Scholar] [CrossRef]
- Cummings, D.E.; Overduin, J.; Foster-Schubert, K.E. Gastric bypass for obesity: Mechanisms of weight loss and diabetes resolution. J. Clin. Endocrinol. Metab. 2004, 89, 2608–2615. [Google Scholar] [CrossRef]
- Lau, D.C.; Teoh, H. Benefits of modest weight loss on the management of type 2 diabetes mellitus. Can. J. Diabetes 2013, 37, 128–134. [Google Scholar] [CrossRef]
- Pendyala, S.; Neff, L.M.; Suarez-Farinas, M.; Holt, P.R. Diet-induced weight loss reduces colorectal inflammation: Implications for colorectal carcinogenesis. Am. J. Clin. Nutr. 2011, 93, 234–242. [Google Scholar] [CrossRef]
- Rapp, K.; Klenk, J.; Ulmer, H.; Concin, H.; Diem, G.; Oberaigner, W.; Schroeder, J. Weight change and cancer risk in a cohort of more than 65 000 adults in Austria. Ann. Oncol. 2008, 19, 641–648. [Google Scholar] [CrossRef] [PubMed]
- Sjöström, L.; Gummesson, A.; Sjöström, C.D.; Narbro, K.; Peltonen, M.; Wedel, H.; Bengtsson, C.; Bouchard, C.; Carlsson, B.; Dahlgren, S. Effects of bariatric surgery on cancer incidence in obese patients in Sweden (Swedish Obese Subjects Study): A prospective, controlled intervention trial. Lancet Oncol. 2009, 10, 653–662. [Google Scholar] [CrossRef]
- Bai, Y.; Sun, Q. Macrophage recruitment in obese adipose tissue. Obes. Rev. 2015, 16, 127–136. [Google Scholar] [CrossRef] [PubMed]
- Park, E.J.; Lee, J.H.; Yu, G.-Y.; He, G.; Ali, S.R.; Holzer, R.G.; Österreicher, C.H.; Takahashi, H.; Karin, M. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 2010, 140, 197–208. [Google Scholar] [CrossRef]
- Ceccarini, G.; Pelosini, C.; Ferrari, F.; Magno, S.; Vitti, J.; Salvetti, G.; Moretto, C.; Marioni, A.; Buccianti, P.; Piaggi, P. Serum IGF-binding protein 2 (IGFBP-2) concentrations change early after gastric bypass bariatric surgery revealing a possible marker of leptin sensitivity in obese subjects. Endocrine 2019, 65, 86–93. [Google Scholar] [CrossRef]
- Bailey, C.J. Metformin: Historical overview. Diabetologia 2017, 60, 1566–1576. [Google Scholar] [CrossRef]
- Kay, J.P.; Alemzadeh, R.; Langley, G.; D’angelo, L.; Smith, P.; Holshouser, S. Beneficial effects of metformin in normoglycemic morbidly obese adolescents. Metab. -Clin. Exp. 2001, 50, 1457–1461. [Google Scholar] [CrossRef]
- Fontbonne, A.; Charles, M.A.; Juhan-Vague, I.; Bard, J.M.; André, P.; Isnard, F.; Cohen, J.M.; Grandmottet, P.; Vague, P.; Safar, M.E. The effect of metformin on the metabolic abnormalities associated with upper-body fat distribution. BIGPRO Study Group. Diabetes Care 1996, 19, 920–926. [Google Scholar] [CrossRef]
- Zakikhani, M.; Dowling, R.; Fantus, I.G.; Sonenberg, N.; Pollak, M. Metformin is an AMP kinase–dependent growth inhibitor for breast cancer cells. Cancer Res. 2006, 66, 10269–10273. [Google Scholar] [CrossRef]
- Alimova, I.N.; Liu, B.; Fan, Z.; Edgerton, S.M.; Dillon, T.; Lind, S.E.; Thor, A.D. Metformin inhibits breast cancer cell growth, colony formation and induces cell cycle arrest in vitro. Cell Cycle 2009, 8, 909–915. [Google Scholar] [CrossRef] [PubMed]
- Liu, B.; Fan, Z.; Edgerton, S.M.; Deng, X.-S.; Alimova, I.N.; Lind, S.E.; Thor, A.D. Metformin induces unique biological and molecular responses in triple negative breast cancer cells. Cell Cycle 2009, 8, 2031–2040. [Google Scholar] [CrossRef] [PubMed]
- Dowling, R.J.; Zakikhani, M.; Fantus, I.G.; Pollak, M.; Sonenberg, N. Metformin inhibits mammalian target of rapamycin–dependent translation initiation in breast cancer cells. Cancer Res. 2007, 67, 10804–10812. [Google Scholar] [CrossRef] [PubMed]
- Algire, C.; Zakikhani, M.; Blouin, M.-J.; Shuai, J.H.; Pollak, M. Metformin attenuates the stimulatory effect of a high-energy diet on in vivo LLC1 carcinoma growth. Endocr. -Relat. Cancer 2008, 15, 833–839. [Google Scholar] [CrossRef]
- Algire, C.; Amrein, L.; Bazile, M.; David, S.; Zakikhani, M.; Pollak, M. Diet and tumor LKB1 expression interact to determine sensitivity to anti-neoplastic effects of metformin in vivo. Oncogene 2011, 30, 1174–1182. [Google Scholar] [CrossRef]
- Algire, C.; Amrein, L.; Zakikhani, M.; Panasci, L.; Pollak, M. Metformin blocks the stimulative effect of a high-energy diet on colon carcinoma growth in vivo and is associated with reduced expression of fatty acid synthase. Endocr. -Relat. Cancer 2010, 17, 351. [Google Scholar] [CrossRef]
- Gallagher, E.J.; LeRoith, D. Minireview: IGF, insulin, and cancer. Endocrinology 2011, 152, 2546–2551. [Google Scholar] [CrossRef]
- Tsugane, S.; Inoue, M. Insulin resistance and cancer: Epidemiological evidence. Cancer Sci. 2010, 101, 1073–1079. [Google Scholar] [CrossRef]
- Evans, J.M.; Donnelly, L.A.; Emslie-Smith, A.M.; Alessi, D.R.; Morris, A.D. Metformin and reduced risk of cancer in diabetic patients. Bmj 2005, 330, 1304–1305. [Google Scholar] [CrossRef]
- Bowker, S.L.; Majumdar, S.R.; Veugelers, P.; Johnson, J.A. Increased cancer-related mortality for patients with type 2 diabetes who use sulfonylureas or insulin. Diabetes Care 2006, 29, 254–258. [Google Scholar] [CrossRef]
- Currie, C.; Poole, C.; Gale, E. The influence of glucose-lowering therapies on cancer risk in type 2 diabetes. Diabetologia 2009, 52, 1766–1777. [Google Scholar] [CrossRef] [PubMed]
- Monami, M.; Lamanna, C.; Balzi, D.; Marchionni, N.; Mannucci, E. Sulphonylureas and cancer: A case–control study. Acta Diabetol. 2009, 46, 279. [Google Scholar] [CrossRef] [PubMed]
- Wright, J.L.; Stanford, J.L. Metformin use and prostate cancer in Caucasian men: Results from a population-based case–control study. Cancer Causes Control 2009, 20, 1617. [Google Scholar] [CrossRef]
- Landman, G.W.; Kleefstra, N.; van Hateren, K.J.; Groenier, K.H.; Gans, R.O.; Bilo, H.J. Metformin associated with lower cancer mortality in type 2 diabetes: ZODIAC-16. Diabetes Care 2010, 33, 322–326. [Google Scholar] [CrossRef] [PubMed]
- Jonasson, J.; Ljung, R.; Talbäck, M.; Haglund, B.; Gudbjörnsdòttir, S.; Steineck, G. Insulin glargine use and short-term incidence of malignancies—a population-based follow-up study in Sweden. Diabetologia 2009, 52, 1745–1754. [Google Scholar] [CrossRef] [PubMed]
- Colhoun, H. Use of insulin glargine and cancer incidence in Scotland: A study from the Scottish Diabetes Research Network Epidemiology Group. Diabetologia 2009, 52, 1755–1765. [Google Scholar] [CrossRef]
- Hemkens, L.G.; Grouven, U.; Bender, R.; Günster, C.; Gutschmidt, S.; Selke, G.W.; Sawicki, P.T. Risk of malignancies in patients with diabetes treated with human insulin or insulin analogues: A cohort study. Diabetologia 2009, 52, 1732–1744. [Google Scholar] [CrossRef]
- DeCensi, A.; Puntoni, M.; Goodwin, P.; Cazzaniga, M.; Gennari, A.; Bonanni, B.; Gandini, S. Metformin and cancer risk in diabetic patients: A systematic review and meta-analysis. Cancer Prev. Res. 2010, 3, 1451–1461. [Google Scholar] [CrossRef]
- Jabbour, M. Primary care physicians and insulin initiation: Multiple barriers, lack of knowledge or both? Int. J. Clin. Pract. 2008, 62, 845–847. [Google Scholar] [CrossRef]
- Kurtzhals, P.; Schäffer, L.; Sørensen, A.; Kristensen, C.; Jonassen, I.; Schmid, C.; Trüb, T. Correlations of receptor binding and metabolic and mitogenic potencies of insulin analogs designed for clinical use. Diabetes 2000, 49, 999–1005. [Google Scholar] [CrossRef]
- Liefvendahl, E.; Arnqvist, H. Mitogenic effect of the insulin analogue glargine in malignant cells in comparison with insulin and IGF-I. Horm. Metab. Res. 2008, 40, 369–374. [Google Scholar] [CrossRef] [PubMed]
- Cheong, J.-H.; Park, E.S.; Liang, J.; Dennison, J.B.; Tsavachidou, D.; Nguyen-Charles, C.; Cheng, K.W.; Hall, H.; Zhang, D.; Lu, Y. Dual inhibition of tumor energy pathway by 2-deoxyglucose and metformin is effective against a broad spectrum of preclinical cancer models. Mol. Cancer Ther. 2011, 10, 2350–2362. [Google Scholar] [CrossRef]
- Sahra, I.B.; Laurent, K.; Giuliano, S.; Larbret, F.; Ponzio, G.; Gounon, P.; Le Marchand-Brustel, Y.; Giorgetti-Peraldi, S.; Cormont, M.; Bertolotto, C. Targeting cancer cell metabolism: The combination of metformin and 2-deoxyglucose induces p53-dependent apoptosis in prostate cancer cells. Cancer Res. 2010, 70, 2465–2475. [Google Scholar] [CrossRef] [PubMed]
- Buzzai, M.; Jones, R.G.; Amaravadi, R.K.; Lum, J.J.; DeBerardinis, R.J.; Zhao, F.; Viollet, B.; Thompson, C.B. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res. 2007, 67, 6745–6752. [Google Scholar] [CrossRef]
- Yap, T.A.; Bjerke, L.; Clarke, P.A.; Workman, P. Drugging PI3K in cancer: Refining targets and therapeutic strategies. Curr. Opin. Pharmacol. 2015, 23, 98–107. [Google Scholar] [CrossRef] [PubMed]
- Ma, C.X.; Sanchez, C.; Gao, F.; Crowder, R.; Naughton, M.; Pluard, T.; Creekmore, A.; Guo, Z.; Hoog, J.; Lockhart, A.C. A phase I study of the AKT inhibitor MK-2206 in combination with hormonal therapy in postmenopausal women with estrogen receptor–positive metastatic breast cancer. Clin. Cancer Res. 2016, 22, 2650–2658. [Google Scholar] [CrossRef] [PubMed]
- Sprinzl, M.F.; Puschnik, A.; Schlitter, A.M.; Schad, A.; Ackermann, K.; Esposito, I.; Lang, H.; Galle, P.R.; Weinmann, A.; Heikenwälder, M. Sorafenib inhibits macrophage-induced growth of hepatoma cells by interference with insulin-like growth factor-1 secretion. J. Hepatol. 2015, 62, 863–870. [Google Scholar] [CrossRef]
- Bowers, L.W.; Rossi, E.L.; O’Flanagan, C.H.; deGraffenried, L.A.; Hursting, S.D. The role of the insulin/IGF system in cancer: Lessons learned from clinical trials and the energy balance-cancer link. Front. Endocrinol. 2015, 6, 77. [Google Scholar] [CrossRef]
- Simpson, A.; Petnga, W.; Macaulay, V.M.; Weyer-Czernilofsky, U.; Bogenrieder, T. Insulin-like growth factor (IGF) pathway targeting in cancer: Role of the IGF axis and opportunities for future combination studies. Target. Oncol. 2017, 12, 571–597. [Google Scholar] [CrossRef]
- Pollak, M. The insulin and insulin-like growth factor receptor family in neoplasia: An update. Nat. Rev. Cancer 2012, 12, 159–169. [Google Scholar] [CrossRef]
- Busaidy, N.L.; Farooki, A.; Dowlati, A.; Perentesis, J.P.; Dancey, J.E.; Doyle, L.A.; Brell, J.M.; Siu, L.L. Management of metabolic effects associated with anticancer agents targeting the PI3K-Akt-mTOR pathway. J. Clin. Oncol. 2012, 30, 2919. [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]
- Quail, D.F.; Bowman, R.L.; Akkari, L.; Quick, M.L.; Schuhmacher, A.J.; Huse, J.T.; Holland, E.C.; Sutton, J.C.; Joyce, J.A. The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas. Science 2016, 352, aad3018. [Google Scholar] [CrossRef]
- Andrews, D.W.; Resnicoff, M.; Flanders, A.E.; Kenyon, L.; Curtis, M.; Merli, G.; Baserga, R.; Iliakis, G.; Aiken, R.D. Results of a pilot study involving the use of an antisense oligodeoxynucleotide directed against the insulin-like growth factor type I receptor in malignant astrocytomas. J. Clin. Oncol. 2001, 19, 2189–2200. [Google Scholar] [CrossRef] [PubMed]
- Resnicoff, M.; Sell, C.; Rubini, M.; Coppola, D.; Ambrose, D.; Baserga, R.; Rubin, R. Rat glioblastoma cells expressing an antisense RNA to the insulin-like growth factor-1 (IGF-1) receptor are nontumorigenic and induce regression of wild-type tumors. Cancer Res. 1994, 54, 2218–2222. [Google Scholar] [PubMed]
- Trojan, J.; Johnson, T.R.; Rudin, S.D.; Ilan, J.; Tykocinski, M.L. Treatment and prevention of rat glioblastoma by immunogenic C6 cells expressing antisense insulin-like growth factor I RNA. Science 1993, 259, 94–97. [Google Scholar] [CrossRef] [PubMed]
- Somasundaram, R.; Zhang, G.; Wagner, S.N.; Fukunaga-Kalabis, M.; Herlyn, M. The role of tumor microenvironment in therapy resistance and melanoma progression. AACR 2015. [Google Scholar]
- Afzal, M.Z.; Mercado, R.R.; Shirai, K. Efficacy of metformin in combination with immune checkpoint inhibitors (anti-PD-1/anti-CTLA-4) in metastatic malignant melanoma. J. Immunother. Cancer 2018, 6, 64. [Google Scholar] [CrossRef]
- Pietras, R.; Xu, H.; Hu, X.; Matheny, C.; Sandler, A.; Patel, M. P1. 04-33 retrospective descriptive analysis of metformin with atezolizumab in advanced non-small cell lung cancer in the OAK trial. J. Thorac. Oncol. 2018, 13, S538–S539. [Google Scholar] [CrossRef]
- Bahrambeigi, S.; Shafiei-Irannejad, V. Immune-mediated anti-tumor effects of metformin; targeting metabolic reprogramming of T cells as a new possible mechanism for anti-cancer effects of metformin. Biochem. Pharmacol. 2020, 174, 113787. [Google Scholar] [CrossRef]
- Araki, K.; Turner, A.P.; Shaffer, V.O.; Gangappa, S.; Keller, S.A.; Bachmann, M.F.; Larsen, C.P.; Ahmed, R. mTOR regulates memory CD8 T-cell differentiation. Nature 2009, 460, 108–112. [Google Scholar] [CrossRef] [PubMed]
- Prlic, M.; Bevan, M.J. A metabolic switch to memory. Nature 2009, 460, 41–42. [Google Scholar] [CrossRef] [PubMed]
- Curry, J.M.; Johnson, J.; Mollaee, M.; Tassone, P.; Amin, D.; Knops, A.; Whitaker-Menezes, D.; Mahoney, M.G.; South, A.; Rodeck, U. Metformin Clinical Trial in HPV+ and HPV–Head and Neck Squamous Cell Carcinoma: Impact on Cancer Cell Apoptosis and Immune Infiltrate. Front. Oncol. 2018, 8, 436. [Google Scholar] [CrossRef] [PubMed]
- Ding, L.; Liang, G.; Yao, Z.; Zhang, J.; Liu, R.; Chen, H.; Zhou, Y.; Wu, H.; Yang, B.; He, Q. Metformin prevents cancer metastasis by inhibiting M2-like polarization of tumor associated macrophages. Oncotarget 2015, 6, 36441. [Google Scholar] [CrossRef]
- Uehara, T.; Eikawa, S.; Nishida, M.; Kunisada, Y.; Yoshida, A.; Fujiwara, T.; Kunisada, T.; Ozaki, T.; Udono, H. Metformin induces CD11b+-cell-mediated growth inhibition of an osteosarcoma: Implications for metabolic reprogramming of myeloid cells and anti-tumor effects. Int. Immunol. 2019, 31, 187–198. [Google Scholar] [CrossRef]
- Verdura, S.; Cuyàs, E.; Martin-Castillo, B.; Menendez, J.A. Metformin as an archetype immuno-metabolic adjuvant for cancer immunotherapy. OncoImmunology 2019, 8, e1633235. [Google Scholar] [CrossRef]
- Cha, J.-H.; Yang, W.-H.; Xia, W.; Wei, Y.; Chan, L.-C.; Lim, S.-O.; Li, C.-W.; Kim, T.; Chang, S.-S.; Lee, H.-H. Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1. Mol. Cell 2018, 71, 606–620.e607. [Google Scholar] [CrossRef]
- Garner, H.; de Visser, K.E. Immune crosstalk in cancer progression and metastatic spread: A complex conversation. Nat. Rev. Immunol. 2020, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Palucka, A.K.; Coussens, L.M. The basis of oncoimmunology. Cell 2016, 164, 1233–1247. [Google Scholar] [CrossRef] [PubMed]
- Elgert, K.D.; Alleva, D.G.; Mullins, D.W. Tumor-induced immune dysfunction: The macrophage connection. J. Leukoc. Biol. 1998, 64, 275–290. [Google Scholar] [CrossRef] [PubMed]
- Fu, C.; Jiang, A. Dendritic cells and CD8 T cell immunity in tumor microenvironment. Front. Immunol. 2018, 9, 3059. [Google Scholar] [CrossRef] [PubMed]
- Lewis, C.E.; Pollard, J.W. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006, 66, 605–612. [Google Scholar] [CrossRef] [PubMed]
- Almand, B.; Resser, J.R.; Lindman, B.; Nadaf, S.; Clark, J.I.; Kwon, E.D.; Carbone, D.P.; Gabrilovich, D.I. Clinical significance of defective dendritic cell differentiation in cancer. Clin. Cancer Res. 2000, 6, 1755–1766. [Google Scholar] [PubMed]
- Tang, M.; Diao, J.; Cattral, M.S. Molecular mechanisms involved in dendritic cell dysfunction in cancer. Cell. Mol. Life Sci. 2017, 74, 761–776. [Google Scholar] [CrossRef]
- Xu, M.M.; Pu, Y.; Han, D.; Shi, Y.; Cao, X.; Liang, H.; Chen, X.; Li, X.-D.; Deng, L.; Chen, Z.J. Dendritic cells but not macrophages sense tumor mitochondrial DNA for cross-priming through signal regulatory protein α signaling. Immunity 2017, 47, 363–373.e365. [Google Scholar] [CrossRef]
- 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]
- Michalek, R.D.; Gerriets, V.A.; Jacobs, S.R.; Macintyre, A.N.; MacIver, N.J.; Mason, E.F.; Sullivan, S.A.; Nichols, A.G.; Rathmell, J.C. Cutting edge: Distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J. Immunol. 2011, 186, 3299–3303. [Google Scholar] [CrossRef]
- Shi, L.Z.; Wang, R.; Huang, G.; Vogel, P.; Neale, G.; Green, D.R.; Chi, H. HIF1α–dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J. Exp. Med. 2011, 208, 1367–1376. [Google Scholar] [CrossRef]
- Gubser, P.M.; Bantug, G.R.; Razik, L.; Fischer, M.; Dimeloe, S.; Hoenger, G.; Durovic, B.; Jauch, A.; Hess, C. Rapid effector function of memory CD8+ T cells requires an immediate-early glycolytic switch. Nat. Immunol. 2013, 14, 1064. [Google Scholar] [CrossRef]
- Dang, E.V.; Barbi, J.; Yang, H.-Y.; Jinasena, D.; Yu, H.; Zheng, Y.; Bordman, Z.; Fu, J.; Kim, Y.; Yen, H.-R. Control of TH17/Treg balance by hypoxia-inducible factor 1. Cell 2011, 146, 772–784. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez-Prados, J.-C.; Través, P.G.; Cuenca, J.; Rico, D.; Aragonés, J.; Martín-Sanz, P.; Cascante, M.; Boscá, L. Substrate fate in activated macrophages: A comparison between innate, classic, and alternative activation. J. Immunol. 2010, 185, 605–614. [Google Scholar] [CrossRef] [PubMed]
- Krawczyk, C.M.; Holowka, T.; Sun, J.; Blagih, J.; Amiel, E.; DeBerardinis, R.J.; Cross, J.R.; Jung, E.; Thompson, C.B.; Jones, R.G. Toll-like receptor–induced changes in glycolytic metabolism regulate dendritic cell activation. Bloodthe J. Am. Soc. Hematol. 2010, 115, 4742–4749. [Google Scholar] [CrossRef] [PubMed]
- 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. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [PubMed]
- Ho, P.-C.; Bihuniak, J.D.; Macintyre, A.N.; Staron, M.; Liu, X.; Amezquita, R.; Tsui, Y.-C.; Cui, G.; Micevic, G.; Perales, J.C. Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell 2015, 162, 1217–1228. [Google Scholar] [CrossRef] [PubMed]
- Gottfried, E.; Kunz-Schughart, L.A.; Ebner, S.; Mueller-Klieser, W.; Hoves, S.; Andreesen, R.; Mackensen, A.; Kreutz, M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood 2006, 107, 2013–2021. [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. Lactic acid and acidification inhibit TNF secretion and glycolysis of human monocytes. J. Immunol. 2010, 184, 1200–1209. [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. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014, 513, 559–563. [Google Scholar] [CrossRef]
- Mukhopadhyay, R.; Jia, J.; Arif, A.; Ray, P.S.; Fox, P.L. The GAIT system: A gatekeeper of inflammatory gene expression. Trends Biochem. Sci. 2009, 34, 324–331. [Google Scholar] [CrossRef]
- Chang, C.-H.; Curtis, J.D.; Maggi Jr, L.B.; Faubert, B.; Villarino, A.V.; O’Sullivan, D.; Huang, S.C.-C.; Van Der Windt, G.J.; Blagih, J.; Qiu, J. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 2013, 153, 1239–1251. [Google Scholar] [CrossRef]
- Sukumar, M.; Liu, J.; Ji, Y.; Subramanian, M.; Crompton, J.G.; Yu, Z.; Roychoudhuri, R.; Palmer, D.C.; Muranski, P.; Karoly, E.D. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Investig. 2013, 123, 4479–4488. [Google Scholar] [CrossRef] [PubMed]
- Pollizzi, K.N.; Patel, C.H.; Sun, I.-H.; Oh, M.-H.; Waickman, A.T.; Wen, J.; Delgoffe, G.M.; Powell, J.D. mTORC1 and mTORC2 selectively regulate CD8+ T cell differentiation. J. Clin. Investig. 2015, 125, 2090–2108. [Google Scholar] [CrossRef] [PubMed]
- Jha, A.K.; Huang, S.C.-C.; Sergushichev, A.; Lampropoulou, V.; Ivanova, Y.; Loginicheva, E.; Chmielewski, K.; Stewart, K.M.; Ashall, J.; Everts, B. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 2015, 42, 419–430. [Google Scholar] [CrossRef] [PubMed]
- Gerriets, V.A.; Kishton, R.J.; Nichols, A.G.; Macintyre, A.N.; Inoue, M.; Ilkayeva, O.; Winter, P.S.; Liu, X.; Priyadharshini, B.; Slawinska, M.E. Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J. Clin. Investig. 2015, 125, 194–207. [Google Scholar] [CrossRef] [PubMed]
- Beier, U.H.; Angelin, A.; Akimova, T.; Wang, L.; Liu, Y.; Xiao, H.; Koike, M.A.; Hancock, S.A.; Bhatti, T.R.; Han, R. Essential role of mitochondrial energy metabolism in Foxp3+ T-regulatory cell function and allograft survival. Faseb J. 2015, 29, 2315–2326. [Google Scholar] [CrossRef]
- Procaccini, C.; Carbone, F.; Di Silvestre, D.; Brambilla, F.; De Rosa, V.; Galgani, M.; Faicchia, D.; Marone, G.; Tramontano, D.; Corona, M. The proteomic landscape of human ex vivo regulatory and conventional T cells reveals specific metabolic requirements. Immunity 2016, 44, 406–421. [Google Scholar] [CrossRef]
- O’Sullivan, D.; van der Windt, G.J.; Huang, S.C.-C.; Curtis, J.D.; Chang, C.-H.; Buck, M.D.; Qiu, J.; Smith, A.M.; Lam, W.Y.; DiPlato, L.M. Memory CD8+ T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 2014, 41, 75–88. [Google Scholar] [CrossRef]
- O’Neill, L.A.; Kishton, R.J.; Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 2016, 16, 553. [Google Scholar] [CrossRef]
- Molodtsov, A.; Turk, M.J. Tissue resident CD8 memory T cell responses in cancer and autoimmunity. Front. Immunol. 2018, 9, 2810. [Google Scholar] [CrossRef]
- Menares, E.; Gálvez-Cancino, F.; Cáceres-Morgado, P.; Ghorani, E.; López, E.; Díaz, X.; Saavedra-Almarza, J.; Figueroa, D.A.; Roa, E.; Quezada, S.A. Tissue-resident memory CD8+ T cells amplify anti-tumor immunity by triggering antigen spreading through dendritic cells. Nat. Commun. 2019, 10, 1–12. [Google Scholar] [CrossRef]
- van der Windt, G.J.; O’Sullivan, D.; Everts, B.; Huang, S.C.-C.; Buck, M.D.; Curtis, J.D.; Chang, C.-H.; Smith, A.M.; Ai, T.; Faubert, B. CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability. Proc. Natl. Acad. Sci. USA 2013, 110, 14336–14341. [Google Scholar] [CrossRef] [PubMed]
- van der Windt, G.J.; Everts, B.; Chang, C.-H.; Curtis, J.D.; Freitas, T.C.; Amiel, E.; Pearce, E.J.; Pearce, E.L. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 2012, 36, 68–78. [Google Scholar] [CrossRef]
- Currie, E.; Schulze, A.; Zechner, R.; Walther, T.C.; Farese Jr, R.V. Cellular fatty acid metabolism and cancer. Cell Metab. 2013, 18, 153–161. [Google Scholar] [CrossRef]
- Svensson, R.U.; Parker, S.J.; Eichner, L.J.; Kolar, M.J.; Wallace, M.; Brun, S.N.; Lombardo, P.S.; Van Nostrand, J.L.; Hutchins, A.; Vera, L. Inhibition of acetyl-CoA carboxylase suppresses fatty acid synthesis and tumor growth of non-small-cell lung cancer in preclinical models. Nat. Med. 2016, 22, 1108. [Google Scholar] [CrossRef] [PubMed]
- Flaveny, C.A.; Griffett, K.; El-Gendy, B.E.-D.M.; Kazantzis, M.; Sengupta, M.; Amelio, A.L.; Chatterjee, A.; Walker, J.; Solt, L.A.; Kamenecka, T.M. Broad anti-tumor activity of a small molecule that selectively targets the Warburg effect and lipogenesis. Cancer Cell 2015, 28, 42–56. [Google Scholar] [CrossRef] [PubMed]
- Everts, B.; Amiel, E.; Huang, S.C.-C.; Smith, A.M.; Chang, C.-H.; Lam, W.Y.; Redmann, V.; Freitas, T.C.; Blagih, J.; Van Der Windt, G.J. TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKɛ supports the anabolic demands of dendritic cell activation. Nat. Immunol. 2014, 15, 323. [Google Scholar] [CrossRef]
- Chen, H.W.; Heiniger, H.-J.; Kandutsch, A.A. Relationship between sterol synthesis and DNA synthesis in phytohemagglutinin-stimulated mouse lymphocytes. Proc. Natl. Acad. Sci. USA 1975, 72, 1950–1954. [Google Scholar] [CrossRef]
- Dufort, F.J.; Gumina, M.R.; Ta, N.L.; Tao, Y.; Heyse, S.A.; Scott, D.A.; Richardson, A.D.; Seyfried, T.N.; Chiles, T.C. Glucose-dependent de novo lipogenesis in B lymphocytes a requirement for ATP-citrate lyase in lipopolysaccharide-induced differentiation. J. Biol. Chem. 2014, 289, 7011–7024. [Google Scholar] [CrossRef]
- Lee, J.; Walsh, M.C.; Hoehn, K.L.; James, D.E.; Wherry, E.J.; Choi, Y. Regulator of fatty acid metabolism, acetyl coenzyme a carboxylase 1, controls T cell immunity. J. Immunol. 2014, 192, 3190–3199. [Google Scholar] [CrossRef]
- Berod, L.; Friedrich, C.; Nandan, A.; Freitag, J.; Hagemann, S.; Harmrolfs, K.; Sandouk, A.; Hesse, C.; Castro, C.N.; Bähre, H. De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat. Med. 2014, 20, 1327. [Google Scholar] [CrossRef]
- Gullino, P.M.; Clark, S.H.; Grantham, F.H. The interstitial fluid of solid tumors. Cancer Res. 1964, 24, 780–797. [Google Scholar] [PubMed]
- Buck, M.D.; O’Sullivan, D.; Geltink, R.I.K.; Curtis, J.D.; Chang, C.-H.; Sanin, D.E.; Qiu, J.; Kretz, O.; Braas, D.; van der Windt, G.J. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell 2016, 166, 63–76. [Google Scholar] [CrossRef] [PubMed]
- Menk, A.V.; Scharping, N.E.; Rivadeneira, D.B.; Calderon, M.J.; Watson, M.J.; Dunstane, D.; Watkins, S.C.; Delgoffe, G.M. 4-1BB costimulation induces T cell mitochondrial function and biogenesis enabling cancer immunotherapeutic responses. J. Exp. Med. 2018, 215, 1091–1100. [Google Scholar] [CrossRef] [PubMed]
- Teijeira, A.; Labiano, S.; Garasa, S.; Etxeberria, I.; Santamaría, E.; Rouzaut, A.; Enamorado, M.; Azpilikueta, A.; Inoges, S.; Bolaños, E. Mitochondrial morphological and functional reprogramming following CD137 (4-1BB) costimulation. Cancer Immunol. Res. 2018. [Google Scholar] [CrossRef]
- Cascone, T.; McKenzie, J.A.; Mbofung, R.M.; Punt, S.; Wang, Z.; Xu, C.; Williams, L.J.; Wang, Z.; Bristow, C.A.; Carugo, A. Increased tumor glycolysis characterizes immune resistance to adoptive T cell therapy. Cell Metab. 2018, 27, 977–987.e974. [Google Scholar] [CrossRef]
- 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]
- Yang, W.; Bai, Y.; Xiong, Y.; Zhang, J.; Chen, S.; Zheng, X.; Meng, X.; Li, L.; Wang, J.; Xu, C. Potentiating the antitumour response of CD8+ T cells by modulating cholesterol metabolism. Nature 2016, 531, 651–655. [Google Scholar] [CrossRef]
- Dean, E.J.; Falchook, G.S.; Patel, M.R.; Brenner, A.J.; Infante, J.R.; Arkenau, H.-T.; Borazanci, E.H.; Lopez, J.S.; Pant, S.; Schmid, P. Preliminary activity in the first in human study of the first-in-class fatty acid synthase (FASN) inhibitor, TVB-2640. Am. Soc. Clin. Oncol. 2016. [Google Scholar] [CrossRef]
- Hosios, A.M.; Hecht, V.C.; Danai, L.V.; Johnson, M.O.; Rathmell, J.C.; Steinhauser, M.L.; Manalis, S.R.; Vander Heiden, M.G. Amino acids rather than glucose account for the majority of cell mass in proliferating mammalian cells. Dev. Cell 2016, 36, 540–549. [Google Scholar] [CrossRef]
- Altman, B.J.; Stine, Z.E.; Dang, C.V. From Krebs to clinic: Glutamine metabolism to cancer therapy. Nat. Rev. Cancer 2016, 16, 619. [Google Scholar] [CrossRef]
- Wang, R.; Dillon, C.P.; Shi, L.Z.; Milasta, S.; Carter, R.; Finkelstein, D.; McCormick, L.L.; Fitzgerald, P.; Chi, H.; Munger, J. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 2011, 35, 871–882. [Google Scholar] [CrossRef] [PubMed]
- Carr, E.L.; Kelman, A.; Wu, G.S.; Gopaul, R.; Senkevitch, E.; Aghvanyan, A.; Turay, A.M.; Frauwirth, K.A. Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J. Immunol. 2010, 185, 1037–1044. [Google Scholar] [CrossRef] [PubMed]
- Crawford, J.; Cohen, H.J. The essential role of L-glutamine in lymphocyte differentiation in vitro. J. Cell. Physiol. 1985, 124, 275–282. [Google Scholar] [CrossRef] [PubMed]
- Nakaya, M.; Xiao, Y.; Zhou, X.; Chang, J.-H.; Chang, M.; Cheng, X.; Blonska, M.; Lin, X.; Sun, S.-C. Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. Immunity 2014, 40, 692–705. [Google Scholar] [CrossRef]
- Klysz, D.; Tai, X.; Robert, P.A.; Craveiro, M.; Cretenet, G.; Oburoglu, L.; Mongellaz, C.; Floess, S.; Fritz, V.; Matias, M.I. Glutamine-dependent α-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. Sci. Signal. 2015, 8, ra97. [Google Scholar] [CrossRef]
- Lemberg, K.M.; Vornov, J.J.; Rais, R.; Slusher, B.S. We’re not “DON” yet: Optimal dosing and prodrug delivery of 6-Diazo-5-oxo-L-norleucine. Mol. Cancer Ther. 2018, 17, 1824–1832. [Google Scholar] [CrossRef]
- Pinkus, L.M. [45] Glutamine binding sites. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 1977; Volume 46, pp. 414–427. [Google Scholar]
- Magill, G.; Myers, W.; Reilly, H.; Putnam, R.; Magill, J.; Sykes, M.; Escher, G.; Karnofsky, D.; Burchenal, J. Pharmacological and initial therapeutic observations on 6-Diazo-5-Oxo-L-Norleucine (Don) in human neoplastic disease. Cancer 1957, 10, 1138–1150. [Google Scholar] [CrossRef]
- Leone, R.D.; Zhao, L.; Englert, J.M.; Sun, I.-M.; Oh, M.-H.; Sun, I.-H.; Arwood, M.L.; Bettencourt, I.A.; Patel, C.H.; Wen, J. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 2019, 366, 1013–1021. [Google Scholar] [CrossRef]
- Palmieri, E.M.; Menga, A.; Martín-Pérez, R.; Quinto, A.; Riera-Domingo, C.; De Tullio, G.; Hooper, D.C.; Lamers, W.H.; Ghesquière, B.; McVicar, D.W. Pharmacologic or genetic targeting of glutamine synthetase skews macrophages toward an M1-like phenotype and inhibits tumor metastasis. Cell Rep. 2017, 20, 1654–1666. [Google Scholar] [CrossRef]
- Oh, M.-H.; Sun, I.-H.; Zhao, L.; Leone, R.D.; Sun, I.-M.; Xu, W.; Collins, S.L.; Tam, A.J.; Blosser, R.L.; Patel, C.H. Targeting glutamine metabolism enhances tumor specific immunity by modulating suppressive myeloid cells. J. Clin. Investig. 2020. [Google Scholar] [CrossRef]
- Guerra, V.; Dinardo, C.D.; Konopleva, M.; Burger, J.A.; Borthakur, G.; Jabbour, E.; Pemmaraju, N.; Sheppard, K.; Garcia-Manero, G.; Kantarjian, H.M. Interim results from a phase Ib/II clinical study of the glutaminase inhibitor telaglenastat (CB-839) in combination with azacitidine in patients with advanced myelodysplastic syndrome (MDS). Am. Soc. Clin. Oncol. 2019. [Google Scholar] [CrossRef]
- Kizilbash, S.H.; McBrayer, S.; Port, J.; Reid, J.M.; Lanza, I.; Allred, J.B.; Chakravarti, A.; Kunos, C.; Adjei, A.A. A phase Ib trial of CB-839 (telaglenastat) in combination with radiation therapy and temozolomide in patients with IDH-mutated diffuse astrocytoma and anaplastic astrocytoma (NCT03528642). Am. Soc. Clin. Oncol. 2019. [Google Scholar] [CrossRef]
- Wang, E.S.; Frankfurt, O.; Orford, K.W.; Bennett, M.; Flinn, I.W.; Maris, M.; Konopleva, M. Phase 1 study of CB-839, a first-in-class, orally administered small molecule inhibitor of glutaminase in patients with relapsed/refractory leukemia. Blood 2015, 126, 2566. [Google Scholar] [CrossRef]
- Lee, G.K.; Park, H.J.; Macleod, M.; Chandler, P.; Munn, D.H.; Mellor, A.L. Tryptophan deprivation sensitizes activated T cells to apoptosis prior to cell division. Immunology 2002, 107, 452–460. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Liu, H.; Huang, L.; Bradley, J.; Liu, K.; Bardhan, K.; Ron, D.; Mellor, A.L.; Munn, D.H.; McGaha, T.L. GCN2-dependent metabolic stress is essential for endotoxemic cytokine induction and pathology. Mol. Cell. Biol. 2014, 34, 428–438. [Google Scholar] [CrossRef] [PubMed]
- Bessede, A.; Gargaro, M.; Pallotta, M.T.; Matino, D.; Servillo, G.; Brunacci, C.; Bicciato, S.; Mazza, E.M.; Macchiarulo, A.; Vacca, C. Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 2014, 511, 184–190. [Google Scholar] [CrossRef]
- Uyttenhove, C.; Pilotte, L.; Théate, I.; Stroobant, V.; Colau, D.; Parmentier, N.; Boon, T.; Van den Eynde, B.J. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2, 3-dioxygenase. Nat. Med. 2003, 9, 1269–1274. [Google Scholar] [CrossRef]
- Okamoto, A.; Nikaido, T.; Ochiai, K.; Takakura, S.; Saito, M.; Aoki, Y.; Ishii, N.; Yanaihara, N.; Yamada, K.; Takikawa, O. Indoleamine 2, 3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells. Clin. Cancer Res. 2005, 11, 6030–6039. [Google Scholar] [CrossRef]
- Minhas, P.S.; Liu, L.; Moon, P.K.; Joshi, A.U.; Dove, C.; Mhatre, S.; Contrepois, K.; Wang, Q.; Lee, B.A.; Coronado, M. Macrophage de novo NAD+ synthesis specifies immune function in aging and inflammation. Nat. Immunol. 2019, 20, 50–63. [Google Scholar] [CrossRef]
- Munn, D.H.; Sharma, M.D.; Hou, D.; Baban, B.; Lee, J.R.; Antonia, S.J.; Messina, J.L.; Chandler, P.; Koni, P.A.; Mellor, A.L. Expression of indoleamine 2, 3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J. Clin. Investig. 2004, 114, 280–290. [Google Scholar] [CrossRef] [PubMed]
- Weinlich, G.; Murr, C.; Richardsen, L.; Winkler, C.; Fuchs, D. Decreased serum tryptophan concentration predicts poor prognosis in malignant melanoma patients. Dermatology 2007, 214, 8–14. [Google Scholar] [CrossRef] [PubMed]
- Holmgaard, R.B.; Zamarin, D.; Munn, D.H.; Wolchok, J.D.; Allison, J.P. Indoleamine 2, 3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4. J. Exp. Med. 2013, 210, 1389–1402. [Google Scholar] [CrossRef] [PubMed]
- Cronin, S.J.; Seehus, C.; Weidinger, A.; Talbot, S.; Reissig, S.; Seifert, M.; Pierson, Y.; McNeill, E.; Longhi, M.S.; Turnes, B.L. The metabolite BH4 controls T cell proliferation in autoimmunity and cancer. Nature 2018, 563, 564–568. [Google Scholar] [CrossRef] [PubMed]
- Gu, T.; Rowswell-Turner, R.B.; Kilinc, M.O.; Egilmez, N.K. Central role of IFNγ–indoleamine 2, 3-dioxygenase axis in regulation of interleukin-12–mediated antitumor immunity. Cancer Res. 2010, 70, 129–138. [Google Scholar] [CrossRef] [PubMed]
- Friberg, M.; Jennings, R.; Alsarraj, M.; Dessureault, S.; Cantor, A.; Extermann, M.; Mellor, A.L.; Munn, D.H.; Antonia, S.J. Indoleamine 2, 3-dioxygenase contributes to tumor cell evasion of T cell-mediated rejection. Int. J. Cancer 2002, 101, 151–155. [Google Scholar] [CrossRef] [PubMed]
- Labadie, B.W.; Bao, R.; Luke, J.J. Reimagining IDO pathway inhibition in cancer immunotherapy via downstream focus on the tryptophan–kynurenine–aryl hydrocarbon axis. Clin. Cancer Res. 2019, 25, 1462–1471. [Google Scholar] [CrossRef] [PubMed]
- Geiger, R.; Rieckmann, J.C.; Wolf, T.; Basso, C.; Feng, Y.; Fuhrer, T.; Kogadeeva, M.; Picotti, P.; Meissner, F.; Mann, M. L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 2016, 167, 829–842.e813. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, P.C.; Zea, A.H.; Culotta, K.S.; Zabaleta, J.; Ochoa, J.B.; Ochoa, A.C. Regulation of t cell receptor cd3ζ chain expression byl-arginine. J. Biol. Chem. 2002, 277, 21123–21129. [Google Scholar] [CrossRef]
- Rodriguez, P.C.; Quiceno, D.G.; Ochoa, A.C. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood 2007, 109, 1568–1573. [Google Scholar] [CrossRef]
- El-Gayar, S.; Thüring-Nahler, H.; Pfeilschifter, J.; Röllinghoff, M.; Bogdan, C. Translational control of inducible nitric oxide synthase by IL-13 and arginine availability in inflammatory macrophages. J. Immunol. 2003, 171, 4561–4568. [Google Scholar] [CrossRef]
- Pesce, J.T.; Ramalingam, T.R.; Mentink-Kane, M.M.; Wilson, M.S.; El Kasmi, K.C.; Smith, A.M.; Thompson, R.W.; Cheever, A.W.; Murray, P.J.; Wynn, T.A. Arginase-1–expressing macrophages suppress Th2 cytokine–driven inflammation and fibrosis. Plos Pathog. 2009, 5. [Google Scholar] [CrossRef]
- Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [PubMed]
- Cobbold, S.P.; Adams, E.; Farquhar, C.A.; Nolan, K.F.; Howie, D.; Lui, K.O.; Fairchild, P.J.; Mellor, A.L.; Ron, D.; Waldmann, H. Infectious tolerance via the consumption of essential amino acids and mTOR signaling. Proc. Natl. Acad. Sci. USA 2009, 106, 12055–12060. [Google Scholar] [CrossRef] [PubMed]
- Doedens, A.L.; Stockmann, C.; Rubinstein, M.P.; Liao, D.; Zhang, N.; DeNardo, D.G.; Coussens, L.M.; Karin, M.; Goldrath, A.W.; Johnson, R.S. Macrophage expression of hypoxia-inducible factor-1α suppresses T-cell function and promotes tumor progression. Cancer Res. 2010, 70, 7465–7475. [Google Scholar] [CrossRef] [PubMed]
- Sharda, D.R.; Yu, S.; Ray, M.; Squadrito, M.L.; De Palma, M.; Wynn, T.A.; Morris, S.M.; Hankey, P.A. Regulation of macrophage arginase expression and tumor growth by the Ron receptor tyrosine kinase. J. Immunol. 2011, 187, 2181–2192. [Google Scholar] [CrossRef]
- Rodriguez, P.C.; Quiceno, D.G.; Zabaleta, J.; Ortiz, B.; Zea, A.H.; Piazuelo, M.B.; Delgado, A.; Correa, P.; Brayer, J.; Sotomayor, E.M. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004, 64, 5839–5849. [Google Scholar] [CrossRef] [PubMed]
- Steggerda, S.M.; Bennett, M.K.; Chen, J.; Emberley, E.; Huang, T.; Janes, J.R.; Li, W.; MacKinnon, A.L.; Makkouk, A.; Marguier, G. Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J. Immunother. Cancer 2017, 5, 101. [Google Scholar] [CrossRef]
- Papadopoulos, K.P.; Tsai, F.Y.-C.; Bauer, T.M.; Muigai, L.; Liang, Y.; Bennett, M.K.; Orford, K.W.; Fu, S. CX-1158-101: A first-in-human phase 1 study of CB-1158, a small molecule inhibitor of arginase, as monotherapy and in combination with an anti-PD-1 checkpoint inhibitor in patients (pts) with solid tumors. Am. Soc. Clin. Oncol. 2017. [Google Scholar] [CrossRef]
- Naing, A.; Bauer, T.; Papadopoulos, K.; Rahma, O.; Tsai, F.; Garralda, E.; Naidoo, J.; Pai, S.; Gibson, M.; Rybkin, I. Phase I study of the arginase inhibitor INCB001158 (1158) alone and in combination with pembrolizumab (PEM) in patients (Pts) with advanced/metastatic (adv/met) solid tumours. Ann. Oncol. 2019, 30, v160. [Google Scholar] [CrossRef]
- Hui, S.; Ghergurovich, J.M.; Morscher, R.J.; Jang, C.; Teng, X.; Lu, W.; Esparza, L.A.; Reya, T.; Zhan, L.; Guo, J.Y. Glucose feeds the TCA cycle via circulating lactate. Nature 2017, 551, 115–118. [Google Scholar] [CrossRef] [PubMed]
- Faubert, B.; Li, K.Y.; Cai, L.; Hensley, C.T.; Kim, J.; Zacharias, L.G.; Yang, C.; Do, Q.N.; Doucette, S.; Burguete, D. Lactate metabolism in human lung tumors. Cell 2017, 171, 358–371.e359. [Google Scholar] [CrossRef] [PubMed]
- Ippolito, L.; Morandi, A.; Giannoni, E.; Chiarugi, P. Lactate: A metabolic driver in the tumour landscape. Trends Biochem. Sci. 2019, 44, 153–166. [Google Scholar] [CrossRef] [PubMed]
- Husain, Z.; Huang, Y.; Seth, P.; Sukhatme, V.P. Tumor-derived lactate modifies antitumor immune response: Effect on myeloid-derived suppressor cells and NK cells. J. Immunol. 2013, 191, 1486–1495. [Google Scholar] [CrossRef]
- Angelin, A.; Gil-de-Gómez, L.; Dahiya, S.; Jiao, J.; Guo, L.; Levine, M.H.; Wang, Z.; Quinn III, W.J.; Kopinski, P.K.; Wang, L. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab. 2017, 25, 1282–1293.e1287. [Google Scholar] [CrossRef] [PubMed]
- Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef]
- 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]
- Huber, V.; Camisaschi, C.; Berzi, A.; Ferro, S.; Lugini, L.; Triulzi, T.; Tuccitto, A.; Tagliabue, E.; Castelli, C.; Rivoltini, L. Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin. Cancer Biol. 2017, 43, 74–89. [Google Scholar] [CrossRef]
- Lacroix, R.; Rozeman, E.A.; Kreutz, M.; Renner, K.; Blank, C.U. Targeting tumor-associated acidity in cancer immunotherapy. Cancer Immunol. Immunother. 2018, 67, 1331–1348. [Google Scholar] [CrossRef]
- Peng, M.; Yin, N.; Chhangawala, S.; Xu, K.; Leslie, C.S.; Li, M.O. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science 2016, 354, 481–484. [Google Scholar] [CrossRef]
- Farabegoli, F.; Vettraino, M.; Manerba, M.; Fiume, L.; Roberti, M.; Di Stefano, G. Galloflavin, a new lactate dehydrogenase inhibitor, induces the death of human breast cancer cells with different glycolytic attitude by affecting distinct signaling pathways. Eur. J. Pharm. Sci. 2012, 47, 729–738. [Google Scholar] [CrossRef]
- Patel, C.H.; Leone, R.D.; Horton, M.R.; Powell, J.D. Targeting metabolism to regulate immune responses in autoimmunity and cancer. Nat. Rev. Drug Discov. 2019, 18, 669–688. [Google Scholar] [CrossRef]
- Miller, W.L.; Thomas, R.A.; Berne, R.M.; Rubio, R. Adenosine production in the ischemic kidney. Circ. Res. 1978, 43, 390–397. [Google Scholar] [CrossRef] [PubMed]
- Sorrentino, C.; Miele, L.; Porta, A.; Pinto, A.; Morello, S. Myeloid-derived suppressor cells contribute to A2B adenosine receptor-induced VEGF production and angiogenesis in a mouse melanoma model. Oncotarget 2015, 6, 27478. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Huang, L.; Ye, H.; Song, S.P.; Bajwa, A.; Lee, S.J.; Moser, E.K.; Jaworska, K.; Kinsey, G.R.; Day, Y.J. Dendritic cells tolerized with adenosine A 2A R agonist attenuate acute kidney injury. J. Clin. Investig. 2012, 122, 3931–3942. [Google Scholar] [CrossRef] [PubMed]
- Vijayan, D.; Young, A.; Teng, M.W.; Smyth, M.J. Targeting immunosuppressive adenosine in cancer. Nat. Rev. Cancer 2017, 17, 709. [Google Scholar] [CrossRef]
- Cai, X.Y.; Wang, X.F.; Li, J.; Dong, J.N.; Liu, J.Q.; Li, N.P.; Yun, B.; Xia, R.L.; Qin, J.; Sun, Y.H. High expression of CD39 in gastric cancer reduces patient outcome following radical resection. Oncol. Lett. 2016, 12, 4080–4086. [Google Scholar] [CrossRef]
- Turcotte, M.; Spring, K.; Pommey, S.; Chouinard, G.; Cousineau, I.; George, J.; Chen, G.M.; Gendoo, D.M.; Haibe-Kains, B.; Karn, T. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 2015, 75, 4494–4503. [Google Scholar] [CrossRef]
- Inoue, Y.; Yoshimura, K.; Kurabe, N.; Kahyo, T.; Kawase, A.; Tanahashi, M.; Ogawa, H.; Inui, N.; Funai, K.; Shinmura, K. Prognostic impact of CD73 and A2A adenosine receptor expression in non-small-cell lung cancer. Oncotarget 2017, 8, 8738. [Google Scholar] [CrossRef]
- Maj, T.; Wang, W.; Crespo, J.; Zhang, H.; Wang, W.; Wei, S.; Zhao, L.; Vatan, L.; Shao, I.; Szeliga, W. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat. Immunol. 2017, 18, 1332. [Google Scholar] [CrossRef]
- Sun, X.; Wu, Y.; Gao, W.; Enjyoji, K.; Csizmadia, E.; Müller, C.E.; Murakami, T.; Robson, S.C. CD39/ENTPD1 expression by CD4+ Foxp3+ regulatory T cells promotes hepatic metastatic tumor growth in mice. Gastroenterology 2010, 139, 1030–1040. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.; Apasov, S.; Koshiba, M.; Sitkovsky, M. Role of A2a extracellular adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell activation and expansion. Bloodthe J. Am. Soc. Hematol. 1997, 90, 1600–1610. [Google Scholar]
- Ohta, A.; Kini, R.; Ohta, A.; Subramanian, M.; Madasu, M.; Sitkovsky, M. The development and immunosuppressive functions of CD4+ CD25+ FoxP3+ regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front. Immunol. 2012, 3, 190. [Google Scholar] [CrossRef] [PubMed]
- Allard, B.; Pommey, S.; Smyth, M.J.; Stagg, J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin. Cancer Res. 2013, 19, 5626–5635. [Google Scholar] [CrossRef] [PubMed]
- Iannone, R.; Miele, L.; Maiolino, P.; Pinto, A.; Morello, S. Adenosine limits the therapeutic effectiveness of anti-CTLA4 mAb in a mouse melanoma model. Am. J. Cancer Res. 2014, 4, 172. [Google Scholar]
- Beavis, P.A.; Milenkovski, N.; Henderson, M.A.; John, L.B.; Allard, B.; Loi, S.; Kershaw, M.H.; Stagg, J.; Darcy, P.K. Adenosine receptor 2A blockade increases the efficacy of anti–PD-1 through enhanced antitumor T-cell responses. Cancer Immunol. Res. 2015, 3, 506–517. [Google Scholar] [CrossRef]
- Fong, L.; Forde, P.M.; Powderly, J.D.; Goldman, J.W.; Nemunaitis, J.J.; Luke, J.J.; Hellmann, M.D.; Kummar, S.; Doebele, R.C.; Mahadevan, D. Safety and clinical activity of adenosine A2a receptor (A2aR) antagonist, CPI-444, in anti-PD1/PDL1 treatment-refractory renal cell (RCC) and non-small cell lung cancer (NSCLC) patients. Am. Soc. Clin. Oncol. 2017. [Google Scholar] [CrossRef]
- Hammami, A.; Allard, D.; Allard, B.; Stagg, J. Targeting the adenosine pathway for cancer immunotherapy. Semin. Immunol. 2019, 42, 101304. [Google Scholar] [CrossRef]
- Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 2009, 284, 34211–34222. [Google Scholar] [CrossRef]
- Qu, J.-L.; Qu, X.-J.; Zhao, M.-F.; Teng, Y.-E.; Zhang, Y.; Hou, K.-Z.; Jiang, Y.-H.; Yang, X.-H.; Liu, Y.-P. Gastric cancer exosomes promote tumour cell proliferation through PI3K/Akt and MAPK/ERK activation. Dig. Liver Dis. 2009, 41, 875–880. [Google Scholar] [CrossRef]
- Zhao, H.; Yang, L.; Baddour, J.; Achreja, A.; Bernard, V.; Moss, T.; Marini, J.C.; Tudawe, T.; Seviour, E.G.; San Lucas, F.A. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. elife 2016, 5, e10250. [Google Scholar] [CrossRef] [PubMed]
- Achreja, A.; Zhao, H.; Yang, L.; Yun, T.H.; Marini, J.; Nagrath, D. Exo-MFA–a 13C metabolic flux analysis framework to dissect tumor microenvironment-secreted exosome contributions towards cancer cell metabolism. Metab. Eng. 2017, 43, 156–172. [Google Scholar] [CrossRef] [PubMed]
- Lazar, I.; Clement, E.; Dauvillier, S.; Milhas, D.; Ducoux-Petit, M.; Legonidec, S.; Moro, C.; Soldan, V.; Dalle, S.; Balor, S. Adipocyte exosomes promote melanoma aggressiveness through fatty acid oxidation: A novel mechanism linking obesity and cancer. Cancer Res. 2016, 76, 4051–4057. [Google Scholar] [CrossRef] [PubMed]
- Patsoukis, N.; Bardhan, K.; Chatterjee, P.; Sari, D.; Liu, B.; Bell, L.N.; Karoly, E.D.; Freeman, G.J.; Petkova, V.; Seth, P. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat. Commun. 2015, 6, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Patsoukis, N.; Brown, J.; Petkova, V.; Liu, F.; Li, L.; Boussiotis, V.A. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci. Signal. 2012, 5, ra46. [Google Scholar] [CrossRef]
- Parry, R.V.; Chemnitz, J.M.; Frauwirth, K.A.; Lanfranco, A.R.; Braunstein, I.; Kobayashi, S.V.; Linsley, P.S.; Thompson, C.B.; Riley, J.L. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell. Biol. 2005, 25, 9543–9553. [Google Scholar] [CrossRef]
- Li, X.; Wenes, M.; Romero, P.; Huang, S.C.-C.; Fendt, S.-M.; Ho, P.-C. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 425–441. [Google Scholar] [CrossRef]
Glycolysis | OXPHOS | FAO | FAS | |
---|---|---|---|---|
Naïve T cell | ○ | ○ | ||
Effector T cell | ○ | ○ | ○ | |
Memory T cell | ○ | ○ | ○ | |
Regulatory T cell | ○ | ○ |
© 2020 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
Yang, H.-Y.; Wu, C.-Y.; Powell, J.D.; Lu, K.-L. Manipulation of Metabolic Pathways and Its Consequences for Anti-Tumor Immunity: A Clinical Perspective. Int. J. Mol. Sci. 2020, 21, 4030. https://doi.org/10.3390/ijms21114030
Yang H-Y, Wu C-Y, Powell JD, Lu K-L. Manipulation of Metabolic Pathways and Its Consequences for Anti-Tumor Immunity: A Clinical Perspective. International Journal of Molecular Sciences. 2020; 21(11):4030. https://doi.org/10.3390/ijms21114030
Chicago/Turabian StyleYang, Huang-Yu, Chao-Yi Wu, Jonathan D. Powell, and Kun-Lin Lu. 2020. "Manipulation of Metabolic Pathways and Its Consequences for Anti-Tumor Immunity: A Clinical Perspective" International Journal of Molecular Sciences 21, no. 11: 4030. https://doi.org/10.3390/ijms21114030
APA StyleYang, H.-Y., Wu, C.-Y., Powell, J. D., & Lu, K.-L. (2020). Manipulation of Metabolic Pathways and Its Consequences for Anti-Tumor Immunity: A Clinical Perspective. International Journal of Molecular Sciences, 21(11), 4030. https://doi.org/10.3390/ijms21114030