Metabolic Barriers to Glioblastoma Immunotherapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. Effects of Increased Tumor Cell Reliance on Glycolysis on the Immune Microenvironment of Glioblastoma
3. Anti-Tumoral Immunologic Effects of Targeting Glutamine Metabolism in Cancer
4. Inhibition of Tryptophan Degrading Enzymes as a Strategy to Promote an Anti-Tumoral Immune Response
5. Exogenous Induction of Lipid Peroxidation as a Strategy to Promote an Anti-Tumoral Immune Response
6. Implications of GBM Metabolic Alterations for Specific Immunotherapy Strategies
7. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Tan, A.C.; Ashley, D.M.; López, G.Y.; Malinzak, M.; Friedman, H.S.; Khasraw, M. Management of glioblastoma: State of the art and future directions. CA Cancer J. Clin. 2020, 70, 299–312. [Google Scholar] [CrossRef]
- Stupp, R.; Lukas, R.V.; Hegi, M.E. Improving survival in molecularly selected glioblastoma. Lancet 2019, 393, 615–617. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Yu, M.W.; Quail, D.F. Immunotherapy for Glioblastoma: Current Progress and Challenges. Front. Immunol. 2021, 12, 1637. [Google Scholar] [CrossRef]
- Bausart, M.; Préat, V.; Malfanti, A. Immunotherapy for glioblastoma: The promise of combination strategies. J. Exp. Clin. Cancer Res. 2022, 41, 35. [Google Scholar] [CrossRef]
- Leone, R.D.; Powell, J.D. Metabolism of immune cells in cancer. Nat. Rev. Cancer 2020, 20, 516–531. [Google Scholar] [CrossRef]
- DePeaux, K.; Delgoffe, G.M. Metabolic barriers to cancer immunotherapy. Nat. Rev. Immunol. 2021, 21, 785–797. [Google Scholar] [CrossRef]
- Zhou, W.; Wahl, D.R. Metabolic Abnormalities in Glioblastoma and Metabolic Strategies to Overcome Treatment Resistance. Cancers 2019, 11, 1231. [Google Scholar] [CrossRef] [Green Version]
- Garcia, J.H.; Jain, S.; Aghi, M.K. Metabolic Drivers of Invasion in Glioblastoma. Front. Cell Dev. Biol. 2021, 9, 683276. [Google Scholar] [CrossRef]
- Marin-Valencia, I.; Yang, C.; Mashimo, T.; Cho, S.; Baek, H.; Yang, X.-L.; Rajagopalan, K.N.; Maddie, M.; Vemireddy, V.; Zhao, Z.; et al. Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo. Cell Metab. 2012, 15, 827–837. [Google Scholar] [CrossRef] [Green Version]
- Sanzey, M.; Rahim, S.A.A.; Oudin, A.; Dirkse, A.; Kaoma, T.; Vallar, L.; Herold-Mende, C.; Bjerkvig, R.; Golebiewska, A.; Niclou, S.P. Comprehensive analysis of glycolytic enzymes as therapeutic targets in the treatment of glioblastoma. PLoS ONE 2015, 10, e0123544. [Google Scholar] [CrossRef]
- Wolf, A.; Agnihotri, S.; Munoz, D.; Guha, A. Developmental profile and regulation of the glycolytic enzyme hexokinase 2 in normal brain and glioblastoma multiforme. Neurobiol. Dis. 2011, 44, 84–91. [Google Scholar] [CrossRef]
- Muir, A.; Vander Heiden, M.G. The nutrient environment affects therapy. Science 2018, 360, 962–963. [Google Scholar] [CrossRef]
- Bi, J.; Bi, F.; Pan, X.; Yang, Q. Establishment of a novel glycolysis-related prognostic gene signature for ovarian cancer and its relationships with immune infiltration of the tumor microenvironment. J. Transl. Med. 2021, 19, 382. [Google Scholar] [CrossRef]
- Li, F.; Simon, M.C. Cancer Cells Don’t Live Alone: Metabolic Communication within Tumor Microenvironments. Dev. Cell 2020, 54, 183–195. [Google Scholar] [CrossRef]
- Shime, H.; Yabu, M.; Akazawa, T.; Kodama, K.; Matsumoto, M.; Seya, T.; Inoue, N. Tumor-secreted lactic acid promotes IL-23/IL-17 proinflammatory pathway. J. Immunol. Baltim. Md. 1950 2008, 180, 7175–7183. [Google Scholar] [CrossRef] [Green Version]
- Dietl, K.; Renner, K.; Dettmer, K.; Timischl, B.; Eberhart, K.; Dorn, C.; Hellerbrand, C.; Kastenberger, M.; Kunz-Schughart, L.A.; Oefner, P.J.; et al. Lactic acid and acidification inhibit TNF secretion and glycolysis of human monocytes. J. Immunol. Baltim. Md. 1950 2010, 184, 1200–1209. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.; Wang, G.; Xu, Z.-G.; Tu, H.; Hu, F.; Dai, J.; Chang, Y.; Chen, Y.; Lu, Y.; Zeng, H.; et al. Lactate Is a Natural Suppressor of RLR Signaling by Targeting MAVS. Cell 2019, 178, 176–189.e15. [Google Scholar] [CrossRef]
- Donnelly, R.P.; Loftus, R.M.; Keating, S.E.; Liou, K.T.; Biron, C.A.; Gardiner, C.M.; Finlay, D.K. mTORC1-dependent metabolic reprogramming is a prerequisite for NK cell effector function. J. Immunol. Baltim. Md. 1950 2014, 193, 4477–4484. [Google Scholar] [CrossRef] [Green Version]
- Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef] [Green Version]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef] [Green Version]
- 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.; et al. Phosphoenolpyruvate Is a Metabolic Checkpoint of Anti-tumor T Cell Responses. Cell 2015, 162, 1217–1228. [Google Scholar] [CrossRef] [Green Version]
- Cascone, T.; McKenzie, J.A.; Mbofung, R.M.; Punt, S.; Wang, Z.; Xu, C.; Williams, L.J.; Wang, Z.; Bristow, C.A.; Carugo, A.; et al. Increased Tumor Glycolysis Characterizes Immune Resistance to Adoptive T Cell Therapy. Cell Metab. 2018, 27, 977–987.e4. [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] [Green Version]
- Calcinotto, A.; Filipazzi, P.; Grioni, M.; Iero, M.; De Milito, A.; Ricupito, A.; Cova, A.; Canese, R.; Jachetti, E.; Rossetti, M.; et al. Modulation of Microenvironment Acidity Reverses Anergy in Human and Murine Tumor-Infiltrating T Lymphocytes. Cancer Res. 2012, 72, 2746–2756. [Google Scholar] [CrossRef] [Green Version]
- Pilon-Thomas, S.; Kodumudi, K.; El-Kenawi, A.; Russell, S.; Weber, A.; Luddy, K.; Damaghi, M.; Wojtkowiak, J.; Mulé, J.; Ibrahim-Hashim, A.; et al. Neutralization of tumor acidity improves anti-tumor responses to immunotherapies. Cancer Res. 2016, 76, 1381–1390. [Google Scholar] [CrossRef] [Green Version]
- 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. Baltim. Md. 1950 2013, 191, 1486–1495. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Angelin, A.; Gil-De-Gómez, L.; Dahiya, S.; Jiao, J.; Guo, L.; Levine, M.H.; Wang, Z.; Quinn, W.J., III; Kopinski, P.K.; Wang, L.; et al. Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. Cell Metab. 2017, 25, 1282–1293.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodríguez-Espinosa, O.; Rojas-Espinosa, O.; Moreno-Altamirano, M.M.B.; López-Villegas, E.O.; Sánchez-García, F.J. Metabolic requirements for neutrophil extracellular traps formation. Immunology 2015, 145, 213–224. [Google Scholar] [CrossRef] [Green Version]
- Schmielau, J.; Finn, O.J. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of t-cell function in advanced cancer patients. Cancer Res. 2001, 61, 4756–4760. [Google Scholar]
- Bambury, R.M.; Teo, M.Y.; Power, D.G.; Yusuf, A.; Murray, S.; Battley, J.E.; Drake, C.; O’Dea, P.; Bermingham, N.; Keohane, C.; et al. The association of pre-treatment neutrophil to lymphocyte ratio with overall survival in patients with glioblastoma multiforme. J. Neurooncol. 2013, 114, 149–154. [Google Scholar] [CrossRef]
- Rice, C.M.; Davies, L.C.; Subleski, J.J.; Maio, N.; Gonzalez-Cotto, M.; Andrews, C.; Patel, N.L.; Palmieri, E.M.; Weiss, J.M.; Lee, J.-M.; et al. Tumour-elicited neutrophils engage mitochondrial metabolism to circumvent nutrient limitations and maintain immune suppression. Nat. Commun. 2018, 9, 5099. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Yu, G.; Chu, H.; Wang, X.; Xiong, L.; Cai, G.; Liu, R.; Gao, H.; Tao, B.; Li, W.; et al. Macrophage-Associated PGK1 Phosphorylation Promotes Aerobic Glycolysis and Tumorigenesis. Mol. Cell 2018, 71, 201–215.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gong, L.; Ji, L.; Xu, D.; Wang, J.; Zou, J. TGF-β links glycolysis and immunosuppression in glioblastoma. Histol. Histopathol. 2021, 36, 1111–1124. [Google Scholar] [PubMed]
- Miska, J.; Lee-Chang, C.; Rashidi, A.; Muroski, M.E.; Chang, A.L.; Lopez-Rosas, A.; Zhang, P.; Panek, W.K.; Cordero, A.; Han, Y.; et al. HIF-1α Is a Metabolic Switch between Glycolytic-Driven Migration and Oxidative Phosphorylation-Driven Immunosuppression of Tregs in Glioblastoma. Cell Rep. 2019, 27, 226–237.e4. [Google Scholar] [CrossRef] [Green Version]
- Liang, X.; Wang, Z.; Dai, Z.; Zhang, H.; Zhang, J.; Luo, P.; Liu, Z.; Liu, Z.; Yang, K.; Cheng, Q.; et al. Glioblastoma glycolytic signature predicts unfavorable prognosis, immunological heterogeneity, and ENO1 promotes microglia M2 polarization and cancer cell malignancy. Cancer Gene Ther. 2022, 22, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Liu, X.; Jiang, H.; Chen, B. A Promising Glycolysis- and Immune-Related Prognostic Signature for Glioblastoma. World Neurosurg. 2022, 161, e363–e375. [Google Scholar] [CrossRef]
- Son, J.; Lyssiotis, C.A.; Ying, H.; Wang, X.; Hua, S.; Ligorio, M.; Perera, R.M.; Ferrone, C.R.; Mullarky, E.; Shyh-Chang, N.; et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 2013, 496, 101–105. [Google Scholar] [CrossRef] [Green Version]
- Willems, L.; Jacque, N.; Jacquel, A.; Neveux, N.; Maciel, T.T.; Lambert, M.; Schmitt, A.; Poulain, L.; Green, A.S.; Uzunov, M.; et al. Inhibiting glutamine uptake represents an attractive new strategy for treating acute myeloid leukemia. Blood 2013, 122, 3521–3532. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.-B.; Erickson, J.W.; Fuji, R.; Ramachandran, S.; Gao, P.; Dinavahi, R.; Wilson, K.F.; Ambrosio, A.L.; Dias, S.M.; Dang, C.V.; et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell 2010, 18, 207–219. [Google Scholar] [CrossRef] [Green Version]
- Gu, Y.; Albuquerque, C.P.; Braas, D.; Zhang, W.; Villa, G.R.; Bi, J.; Ikegami, S.; Masui, K.; Gini, B.; Yang, H.; et al. mTORC2 Regulates Amino Acid Metabolism in Cancer by Phosphorylation of the Cystine-Glutamate Antiporter xCT. Mol. Cell 2017, 67, 128–138.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wise, D.R.; DeBerardinis, R.J.; Mancuso, A.; Sayed, N.; Zhang, X.-Y.; Pfeiffer, H.K.; Nissim, I.; Daikhin, E.; Yudkoff, M.; McMahon, S.B.; et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc. Natl. Acad. Sci. USA 2008, 105, 18782–18787. [Google Scholar] [CrossRef] [Green Version]
- DeBerardinis, R.J.; Mancuso, A.; Daikhin, E.; Nissim, I.; Yudkoff, M.; Wehrli, S.; Thompson, C.B. Beyond aerobic glycolysis: Transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc. Natl. Acad. Sci. USA 2007, 104, 19345–19350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takano, T.; Lin, J.H.-C.; Arcuino, G.; Gao, Q.; Yang, J.; Nedergaard, M. Glutamate release promotes growth of malignant gliomas. Nat. Med. 2001, 7, 1010–1015. [Google Scholar] [CrossRef]
- Jin, L.; Li, D.; Alesi, G.N.; Fan, J.; Kang, H.-B.; Lu, Z.; Boggon, T.J.; Jin, P.; Yi, H.; Wright, E.R.; et al. Glutamate Dehydrogenase 1 Signals through Antioxidant Glutathione Peroxidase 1 to Regulate Redox Homeostasis and Tumor Growth. Cancer Cell 2015, 27, 257–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, C.; Sudderth, J.; Dang, T.; Bachoo, R.G.; McDonald, J.G.; DeBerardinis, R.J. Glioblastoma cells require glutamate dehydrogenase to survive impairments of glucose metabolism or Akt signaling. Cancer Res. 2009, 69, 7986–7993. [Google Scholar] [CrossRef] [Green Version]
- Rosati, A.; Poliani, P.L.; Todeschini, A.; Cominelli, M.; Medicina, D.; Cenzato, M.; Simoncini, E.L.; Magrini, S.M.; Buglione, M.; Grisanti, S.; et al. Glutamine synthetase expression as a valuable marker of epilepsy and longer survival in newly diagnosed glioblastoma multiforme. Neuro-Oncol. 2013, 15, 618–625. [Google Scholar] [CrossRef]
- Tardito, S.; Oudin, A.; Ahmed, S.U.; Fack, F.; Keunen, O.; Zheng, L.; Miletic, H.; Sakariassen, P.Ø.; Weinstock, A.; Wagner, A.; et al. Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma. Nat. Cell Biol. 2015, 17, 1556–1568. [Google Scholar] [CrossRef] [Green Version]
- Wang, R.; Dillon, C.P.; Shi, L.Z.; Milasta, S.; Carter, R.; Finkelstein, D.; McCormick, L.L.; Fitzgerald, P.; Chi, H.; Munger, J.; et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 2011, 35, 871–882. [Google Scholar] [CrossRef] [Green Version]
- 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.; et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 2019, 366, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Liu, R.; Qu, X.; Yu, H.; Chu, H.; Zhang, Y.; Zhu, W.; Wu, X.; Gao, H.; Tao, B.; et al. α-Ketoglutarate-Activated NF-κB Signaling Promotes Compensatory Glucose Uptake and Brain Tumor Development. Mol. Cell 2019, 76, 148–162.e7. [Google Scholar] [PubMed]
- Liu, P.-S.; Wang, H.; Li, X.; Chao, T.; Teav, T.; Christen, S.; Di Conza, G.; Cheng, W.-C.; Chou, C.-H.; Vavakova, M.; et al. α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 2017, 18, 985–994. [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.; et al. Targeting glutamine metabolism enhances tumor-specific immunity by modulating suppressive myeloid cells. J. Clin. Investig. 2020, 130, 3865–3884. [Google Scholar] [CrossRef] [Green Version]
- Guastella, A.R.; Michelhaugh, S.K.; Klinger, N.V.; Fadel, H.A.; Kiousis, S.; Ali-Fehmi, R.; Kupsky, W.J.; Juhász, C.; Mittal, S. Investigation of the aryl hydrocarbon receptor and the intrinsic tumoral component of the kynurenine pathway of tryptophan metabolism in primary brain tumors. J. Neurooncol. 2018, 139, 239–249. [Google Scholar] [CrossRef]
- Kim, M.; Tomek, P. Tryptophan: A Rheostat of Cancer Immune Escape Mediated by Immunosuppressive Enzymes IDO1 and TDO. Front. Immunol. 2021, 12, 636081. [Google Scholar]
- The Role of IDO in Brain Tumor Immunotherapy|SpringerLink. Available online: https://link.springer.com/article/10.1007/s11060-014-1687-8 (accessed on 15 December 2022).
- 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] [PubMed]
- Wainwright, D.A.; Balyasnikova, I.V.; Chang, A.L.; Ahmed, A.U.; Moon, K.-S.; Auffinger, B.; Tobias, A.L.; Han, Y.; Lesniak, M.S. IDO expression in brain tumors increases the recruitment of regulatory T cells and negatively impacts survival. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012, 18, 6110–6121. [Google Scholar] [CrossRef] [Green Version]
- Locasale, J.W.; Melman, T.; Song, S.; Yang, X.; Swanson, K.D.; Cantley, L.C.; Wong, E.T.; Asara, J.M. Metabolomics of Human Cerebrospinal Fluid Identifies Signatures of Malignant Glioma*. Mol. Cell. Proteom. 2012, 11, M111.014688. [Google Scholar] [CrossRef] [Green Version]
- Mitsuka, K.; Kawataki, T.; Satoh, E.; Asahara, T.; Horikoshi, T.; Kinouchi, H. Expression of indoleamine 2,3-dioxygenase and correlation with pathological malignancy in gliomas. Neurosurgery 2013, 72, 1031–1038; discussion 1038–1039. [Google Scholar] [CrossRef]
- Campesato, L.F.; Budhu, S.; Tchaicha, J.; Weng, C.-H.; Gigoux, M.; Cohen, I.J.; Redmond, D.; Mangarin, L.; Pourpe, S.; Liu, C.; et al. Blockade of the AHR restricts a Treg-macrophage suppressive axis induced by L-Kynurenine. Nat. Commun. 2020, 11, 4011. [Google Scholar] [CrossRef] [PubMed]
- Fallarino, F.; Grohmann, U.; Vacca, C.; Bianchi, R.; Orabona, C.; Spreca, A.; Fioretti, M.C.; Puccetti, P. T cell apoptosis by tryptophan catabolism. Cell Death Differ. 2002, 9, 1069–1077. [Google Scholar] [CrossRef] [PubMed]
- Munn, D.H.; Sharma, M.D.; Baban, B.; Harding, H.P.; Zhang, Y.; Ron, D.; Mellor, A.L. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity 2005, 22, 633–642. [Google Scholar] [CrossRef] [Green Version]
- van Baren, N.; Van den Eynde, B.J. Tumoral Immune Resistance Mediated by Enzymes That Degrade Tryptophan. Cancer Immunol. Res. 2015, 3, 978–985. [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] [PubMed]
- Brandacher, G.; Perathoner, A.; Ladurner, R.; Schneeberger, S.; Obrist, P.; Winkler, C.; Werner, E.R.; Werner-Felmayer, G.; Weiss, H.G.; G√∂Bel, G.; et al. Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: Effect on tumor-infiltrating T cells. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006, 12, 1144–1151. [Google Scholar] [CrossRef] [Green Version]
- 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. Baltim. Md. 1950 2013, 190, 3783–3797. [Google Scholar] [CrossRef] [Green Version]
- Holmgaard, R.B.; Zamarin, D.; Li, Y.; Gasmi, B.; Munn, D.H.; Allison, J.P.; Merghoub, T.; Wolchok, J.D. Tumor-expressed IDO recruits and activates MDSCs in a Treg-dependent manner. Cell Rep. 2015, 13, 412–424. [Google Scholar] [CrossRef] [Green Version]
- Beatty, G.L.; O’Dwyer, P.J.; Clark, J.; Shi, J.G.; Newton, R.C.; Schaub, R.; Maleski, J.; Leopold, L.; Gajewski, T. Phase I study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of the oral inhibitor of indoleamine 2,3-dioxygenase (IDO1) INCB024360 in patients (pts) with advanced malignancies. J. Clin. Oncol. 2013, 31, 3025. [Google Scholar] [CrossRef]
- Opitz, C.A.; Litzenburger, U.M.; Sahm, F.; Ott, M.; Tritschler, I.; Trump, S.; Schumacher, T.; Jestaedt, L.; Schrenk, D.; Weller, M.; et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 2011, 478, 197–203. [Google Scholar] [CrossRef] [Green Version]
- Moyer, B.J.; Rojas, I.Y.; Murray, I.A.; Lee, S.; Hazlett, H.F.; Perdew, G.H.; Tomlinson, C.R. Indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors activate the aryl hydrocarbon receptor. Toxicol. Appl. Pharmacol. 2017, 323, 74–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wirth, L.J.; Burtness, B.; Mehra, R.; Bauman, J.R.; Lee, J.; Smith, N.M.S.; Lefranc-Torres, A.; Westra, W.H.; Bishop, J.A.; Faquin, W.C.; et al. IDO1 as a mechanism of adaptive immune resistance to anti-PD1 monotherapy in HNSCC. J. Clin. Oncol. 2017, 35, 6053. [Google Scholar] [CrossRef]
- Botticelli, A.; Cerbelli, B.; Lionetto, L.; Zizzari, I.; Salati, M.; Pisano, A.; Federica, M.; Simmaco, M.; Nuti, M.; Marchetti, P. Can IDO activity predict primary resistance to anti-PD-1 treatment in NSCLC? J. Transl. Med. 2018, 16, 219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Wainwright, D.A.; Chang, A.L.; Dey, M.; Balyasnikova, I.V.; Kim, C.K.; Tobias, A.; Cheng, Y.; Kim, J.W.; Qiao, J.; Zhang, L.; et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 5290–5301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Long, G.V.; Dummer, R.; Hamid, O.; Gajewski, T.; Caglevic, C.; Dalle, S.; Arance, A.; Carlino, M.; Grob, J.-J.; Kim, T.M.; et al. Epacadostat (E) plus pembrolizumab (P) versus pembrolizumab alone in patients (pts) with unresectable or metastatic melanoma: Results of the phase 3 ECHO-301/KEYNOTE-252 study. J. Clin. Oncol. 2018, 36, 108. [Google Scholar] [CrossRef]
- Fujiwara, Y.; Kato, S.; Nesline, M.K.; Conroy, J.M.; DePietro, P.; Pabla, S.; Kurzrock, R. Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy. Cancer Treat. Rev. 2022, 110, 102461. [Google Scholar] [CrossRef]
- Zhai, L.; Bell, A.; Ladomersky, E.; Lauing, K.L.; Bollu, L.; Nguyen, B.; Genet, M.; Kim, M.; Chen, P.; Mi, X.; et al. Tumor Cell IDO Enhances Immune Suppression and Decreases Survival Independent of Tryptophan Metabolism in Glioblastoma. Clin. Cancer Res. 2021, 27, 6514–6528. [Google Scholar] [CrossRef]
- Kesarwani, P.; Prabhu, A.; Kant, S.; Chinnaiyan, P. Metabolic remodeling contributes towards an immune-suppressive phenotype in glioblastoma. Cancer Immunol. Immunother. 2019, 68, 1107–1120. [Google Scholar] [CrossRef]
- Pascual, G.; Avgustinova, A.; Mejetta, S.; Martín, M.; Castellanos, A.; Attolini, C.S.-O.; Berenguer, A.; Prats, N.; Toll, A.; Hueto, J.A.; et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 2017, 541, 41–45. [Google Scholar] [CrossRef]
- Hoang-Minh, L.B.; Siebzehnrubl, F.; Yang, C.; Suzuki-Hatano, S.; Dajac, K.; Loche, T.; Andrews, N.; Massari, M.S.; Patel, J.; Amin, K.; et al. Infiltrative and drug-resistant slow-cycling cells support metabolic heterogeneity in glioblastoma. EMBO J. 2018, 37, e98772. [Google Scholar] [CrossRef] [PubMed]
- Shakya, S.; Gromovsky, A.D.; Hale, J.S.; Knudsen, A.M.; Prager, B.; Wallace, L.C.; Penalva, L.O.F.; Ivanova, P.; Brown, H.A.; Kristensen, B.W.; et al. Altered lipid metabolism marks glioblastoma stem and non-stem cells in separate tumor niches. Acta Neuropathol. Commun. 2021, 9, 101. [Google Scholar] [CrossRef] [PubMed]
- Cheng, X.; Geng, F.; Pan, M.; Wu, X.; Zhong, Y.; Wang, C.; Tian, Z.; Cheng, C.; Zhang, R.; Puduvalli, V.; et al. Targeting DGAT1 Ameliorates Glioblastoma by Increasing Fat Catabolism and Oxidative Stress. Cell Metab. 2020, 32, 229–242.e8. [Google Scholar] [CrossRef] [PubMed]
- Taïb, B.; Aboussalah, A.M.; Moniruzzaman, M.; Chen, S.; Haughey, N.J.; Kim, S.F.; Ahima, R.S. Lipid accumulation and oxidation in glioblastoma multiforme. Sci. Rep. 2019, 9, 19593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, P.; Xia, S.; Lal, B.; Shi, X.; Yang, K.S.; A Watkins, P.; Laterra, J. Lipid metabolism enzyme ACSVL3 supports glioblastoma stem cell maintenance and tumorigenicity. BMC Cancer 2014, 14, 401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, F.; Zhao, Z.; Chai, R.C.; Liu, Y.Q.; Li, G.Z.; Jiang, H.Y.; Jiang, T. Prognostic power of a lipid metabolism gene panel for diffuse gliomas. J. Cell. Mol. Med. 2019, 23, 7741–7748. [Google Scholar] [CrossRef] [Green Version]
- 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. Baltim. Md. 1950 2011, 186, 3299–3303. [Google Scholar] [CrossRef] [Green Version]
- Howie, D.; Ten Bokum, A.; Necula, A.S.; Cobbold, S.P.; Waldmann, H. The Role of Lipid Metabolism in T Lymphocyte Differentiation and Survival. Front. Immunol. 2017, 8, 1949. [Google Scholar] [CrossRef] [Green Version]
- Abuhusain, H.J.; Matin, A.; Qiao, Q.; Shen, H.; Kain, N.; Day, B.W.; Stringer, B.; Daniels, B.; Laaksonen, M.A.; Teo, C.; et al. A Metabolic Shift Favoring Sphingosine 1-Phosphate at the Expense of Ceramide Controls Glioblastoma Angiogenesis. J. Biol. Chem. 2013, 288, 37355–37364. [Google Scholar] [CrossRef] [Green Version]
- Kempkes, R.W.M.; Joosten, I.; Koenen, H.J.P.M.; He, X. Metabolic Pathways Involved in Regulatory T Cell Functionality. Front. Immunol. 2019, 10, 2839. [Google Scholar] [CrossRef]
- Wang, H.; Franco, F.; Tsui, Y.-C.; Xie, X.; Trefny, M.P.; Zappasodi, R.; Mohmood, S.R.; Fernández-García, J.; Tsai, C.-H.; Schulze, I.; et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 2020, 21, 298–308. [Google Scholar] [CrossRef] [PubMed]
- Hossain, F.; Al-Khami, A.A.; Wyczechowska, D.; Hernandez, C.; Zheng, L.; Reiss, K.; Del Valle, L.; 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] [PubMed] [Green Version]
- Turubanova, V.D.; Balalaeva, I.V.; Mishchenko, T.A.; Catanzaro, E.; Alzeibak, R.; Peskova, N.N.; Efimova, I.; Bachert, C.; Mitroshina, E.V.; Krysko, O.; et al. Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine. J. Immunother. Cancer 2019, 7, 350. [Google Scholar] [CrossRef]
- Dastmalchi, F.; Karachi, A.; Yang, C.; Azari, H.; Sayour, E.J.; Dechkovskaia, A.; Vlasak, A.L.; Saia, M.E.; Lovaton, R.E.; Mitchell, D.A.; et al. Sarcosine promotes trafficking of dendritic cells and improves efficacy of anti-tumor dendritic cell vaccines via CXC chemokine family signaling. J. Immunother. Cancer 2019, 7, 321. [Google Scholar] [CrossRef]
- Jiang, N.; Xie, B.; Xiao, W.; Fan, M.; Xu, S.; Duan, Y.; Hamsafar, Y.; Evans, A.C.; Huang, J.; Zhou, W.; et al. Fatty acid oxidation fuels glioblastoma radioresistance with CD47-mediated immune evasion. Nat. Commun. 2022, 13, 1511. [Google Scholar] [CrossRef]
- Miska, J.; Chandel, N.S. Targeting fatty acid metabolism in glioblastoma. J. Clin. Investig. 2023, 133, e163448. [Google Scholar] [CrossRef] [PubMed]
- Chiocca, E.A.; Yu, J.S.; Lukas, R.V.; Solomon, I.H.; Ligon, K.L.; Nakashima, H.; Triggs, D.A.; Reardon, D.A.; Wen, P.; Stopa, B.M.; et al. Regulatable interleukin-12 gene therapy in patients with recurrent high-grade glioma: Results of a phase 1 trial. Sci. Transl. Med. 2019, 11, eaaw5680. [Google Scholar] [CrossRef]
- Iorgulescu, J.B.; Gokhale, P.C.; Speranza, M.C.; Eschle, B.K.; Poitras, M.J.; Wilkens, M.K.; Soroko, K.M.; Chhoeu, C.; Knott, A.; Gao, Y.; et al. Concurrent Dexamethasone Limits the Clinical Benefit of Immune Checkpoint Blockade in Glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 276–287. [Google Scholar] [CrossRef]
- Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.S.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef] [Green Version]
- Nduom, E.K.; Weller, M.; Heimberger, A.B. Immunosuppressive mechanisms in glioblastoma. Neuro-Oncol. 2015, 17 (Suppl. S7), vii9–vii14. [Google Scholar] [CrossRef] [Green Version]
- Lim, M.; Xia, Y.; Bettegowda, C.; Weller, M. Current state of immunotherapy for glioblastoma. Nat. Rev. Clin. Oncol. 2018, 15, 422–442. [Google Scholar] [CrossRef] [PubMed]
- Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F.; et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [PubMed] [Green Version]
- Liau, L.M.; Ashkan, K.; Tran, D.; Campian, J.; Trusheim, J.; Cobbs, C.; Heth, J.; Salacz, M.; Taylor, S.; D’Andre, S.D.; et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J. Transl. Med. 2018, 16, 142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lukas, R.V.; Rodon, J.; Becker, K.; Wong, E.T.; Shih, K.; Touat, M.; Fassò, M.; Osborne, S.; Molinero, L.; O’Hear, C.; et al. Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma. J. Neurooncol. 2018, 140, 317–328. [Google Scholar]
- Renner, K.; Bruss, C.; Schnell, A.; Koehl, G.; Becker, H.M.; Fante, M.; Menevse, A.-N.; Kauer, N.; Blazquez, R.; Hacker, L.; et al. Restricting Glycolysis Preserves T Cell Effector Functions and Augments Checkpoint Therapy. Cell Rep. 2019, 29, 135–150.e9. [Google Scholar]
- Todo, T.; Ito, H.; Ino, Y.; Ohtsu, H.; Ota, Y.; Shibahara, J.; Tanaka, M. Intratumoral oncolytic herpes virus G47∆ for residual or recurrent glioblastoma: A phase 2 trial. Nat. Med. 2022, 28, 1630–1639. [Google Scholar]
- Woller, N.; Gürlevik, E.; Fleischmann-Mundt, B.; Schumacher, A.; Knocke, S.; Kloos, A.M.; Saborowski, M.; Geffers, R.; Manns, M.P.; Wirth, T.C.; et al. Viral Infection of Tumors Overcomes Resistance to PD-1-immunotherapy by Broadening Neoantigenome-directed T-cell Responses. Mol. Ther. J. Am. Soc. Gene Ther. 2015, 23, 1630–1640. [Google Scholar] [CrossRef] [Green Version]
- O’Rourke, D.M.; Nasrallah, M.; Desai, A.; Melenhorst, J.; Mansfield, K.; Morrissette, J.; Martinez-Lage, M.; Brem, S.; Maloney, E.; Shen, A.; et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 2017, 9, eaaa0984. [Google Scholar] [CrossRef] [Green Version]
- Nabe, S.; Yamada, T.; Suzuki, J.; Toriyama, K.; Yasuoka, T.; Kuwahara, M.; Shiraishi, A.; Takenaka, K.; Yasukawa, M.; Yamashita, M. Reinforce the antitumor activity of CD8+ T cells via glutamine restriction. Cancer Sci. 2018, 109, 3737–3750. [Google Scholar] [CrossRef] [Green Version]
- Sukumar, M.; Liu, J.; Ji, Y.; Subramanian, M.; Crompton, J.G.; Yu, Z.; Roychoudhuri, R.; Palmer, D.C.; Muranski, P.; Karoly, E.D.; et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Investig. 2013, 123, 4479–4488. [Google Scholar]
- Hajji, N.; Garcia-Revilla, J.; Soto, M.S.; Perryman, R.; Symington, J.; Quarles, C.C.; Healey, D.R.; Guo, Y.; Orta-Vázquez, M.L.; Mateos-Cordero, S.; et al. Arginine deprivation alters microglial polarity and synergizes with radiation to eradicate non-arginine-auxotrophic glioblastoma tumors. J. Clin. Investig. 2022, 132, e142137. [Google Scholar] [CrossRef]
- Bunse, L.; Pusch, S.; Bunse, T.; Sahm, F.; Sanghvi, K.; Friedrich, M.; Alansary, D.; Sonner, J.K.; Green, E.; Deumelandt, K.; et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat. Med. 2018, 24, 1192–1203. [Google Scholar] [CrossRef]
- Safaee, M.M.; Wang, E.J.; Jain, S.; Chen, J.-S.; Gill, S.; Zheng, A.C.; Garcia, J.H.; Beniwal, A.S.; Tran, Y.; Nguyen, A.T.; et al. CD97 is associated with mitogenic pathway activation, metabolic reprogramming, and immune microenvironment changes in glioblastoma. Sci. Rep. 2022, 12, 1464. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Choudhary, N.; Osorio, R.C.; Oh, J.Y.; Aghi, M.K. Metabolic Barriers to Glioblastoma Immunotherapy. Cancers 2023, 15, 1519. https://doi.org/10.3390/cancers15051519
Choudhary N, Osorio RC, Oh JY, Aghi MK. Metabolic Barriers to Glioblastoma Immunotherapy. Cancers. 2023; 15(5):1519. https://doi.org/10.3390/cancers15051519
Chicago/Turabian StyleChoudhary, Nikita, Robert C. Osorio, Jun Y. Oh, and Manish K. Aghi. 2023. "Metabolic Barriers to Glioblastoma Immunotherapy" Cancers 15, no. 5: 1519. https://doi.org/10.3390/cancers15051519
APA StyleChoudhary, N., Osorio, R. C., Oh, J. Y., & Aghi, M. K. (2023). Metabolic Barriers to Glioblastoma Immunotherapy. Cancers, 15(5), 1519. https://doi.org/10.3390/cancers15051519