Linking Serine/Glycine Metabolism to Radiotherapy Resistance
Abstract
:Simple Summary
Abstract
1. Introduction
2. Activation of De Novo Serine/Glycine Biosynthesis in Cancer
2.1. De Novo Serine/Glycine Biosynthesis Pathway
2.2. De Novo Serine/Glycine Biosynthesis Activation in Cancer
2.3. Regulatory Mechanisms of De Novo Serine/Glycine Biosynthesis in Cancer
3. Activation of De Novo Serine/Glycine Biosynthesis and Treatment Resistance
3.1. Targeted Therapies
3.2. Chemotherapy
4. Radiotherapy Resistance and the Link with De Novo Serine/Glycine Biosynthesis
4.1. CSC Fate and Functions
4.2. Tumor Microenvironment
4.2.1. Hypoxia and ROS Homeostasis
4.2.2. Immunomodulatory and Angiogenic Functions
4.3. DNA Damage Response (DDR)
5. Future Perspectives: Therapeutic Targeting of De Novo Serine/Glycine Biosynthesis to Improve Radiotherapy Response
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.-W.; Dang, C.V. Cancer’s molecular sweet tooth and the Warburg effect. Cancer Res. 2006, 66, 8927–8930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wise, D.R.; Thompson, C.B. Glutamine addiction: A new therapeutic target in cancer. Trends Biochem. Sci. 2010, 35, 427–433. [Google Scholar] [CrossRef] [Green Version]
- Akins, N.S.; Nielson, T.C.; Le, H.V. Inhibition of glycolysis and glutaminolysis: An emerging drug discovery approach to combat cancer. Curr. Top. Med. Chem. 2018, 18, 494–504. [Google Scholar] [CrossRef] [PubMed]
- Amelio, I.; Cutruzzolá, F.; Antonov, A.; Agostini, M.; Melino, G. Serine and glycine metabolism in cancer. Trends Biochem. Sci. 2014, 39, 191–198. [Google Scholar] [CrossRef] [PubMed]
- Mattaini, K.R.; Sullivan, M.R.; Vander Heiden, M.G. The importance of serine metabolism in cancer. J. Cell Biol. 2016, 214, 249–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Locasale, J.W. Serine, glycine and one-carbon units: Cancer metabolism in full circle. Nat. Rev. Cancer 2013, 13, 572–583. [Google Scholar] [CrossRef] [Green Version]
- Yang, M.; Vousden, K.H. Serine and one-carbon metabolism in cancer. Nat. Rev. Cancer 2016, 16, 650–662. [Google Scholar] [CrossRef]
- Reina-Campos, M.; Diaz-Meco, M.T.; Moscat, J. The complexity of the serine glycine one-carbon pathway in cancer. J. Cell Biol. 2019, 219, e201907022. [Google Scholar] [CrossRef] [PubMed]
- Geeraerts, S.L.; Heylen, E.; De Keersmaecker, K.; Kampen, K.R. The ins and outs of serine and glycine metabolism in cancer. Nat. Metab. 2021. [Google Scholar] [CrossRef]
- Snell, K. Enzymes of serine metabolism in normal, developing and neoplastic rat tissues. Adv. Enzym. Regul. 1984, 22, 325–400. [Google Scholar] [CrossRef]
- Kit, S. The biosynthesis of free glycine and serine by tumors. Cancer Res. 1955, 15, 715–718. [Google Scholar]
- Locasale, J.W.; Grassian, A.R.; Melman, T.; Lyssiotis, C.A.; Mattaini, K.R.; Bass, A.J.; Heffron, G.; Metallo, C.M.; Muranen, T.; Sharfi, H. Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nat. Genet. 2011, 43, 869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Labuschagne, C.F.; Van Den Broek, N.J.; Mackay, G.M.; Vousden, K.H.; Maddocks, O.D. Serine, but not glycine, supports one-carbon metabolism and proliferation of cancer cells. Cell Rep. 2014, 7, 1248–1258. [Google Scholar] [CrossRef] [Green Version]
- Maddocks, O.D.; Athineos, D.; Cheung, E.C.; Lee, P.; Zhang, T.; van den Broek, N.J.; Mackay, G.M.; Labuschagne, C.F.; Gay, D.; Kruiswijk, F. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature 2017, 544, 372–376. [Google Scholar] [CrossRef] [PubMed]
- DeNicola, G.M.; Chen, P.-H.; Mullarky, E.; Sudderth, J.A.; Hu, Z.; Wu, D.; Tang, H.; Xie, Y.; Asara, J.M.; Huffman, K.E. NRF2 regulates serine biosynthesis in non–small cell lung cancer. Nat. Genet. 2015, 47, 1475. [Google Scholar] [CrossRef] [Green Version]
- Kim, D.; Fiske, B.P.; Birsoy, K.; Freinkman, E.; Kami, K.; Possemato, R.L.; Chudnovsky, Y.; Pacold, M.E.; Chen, W.W.; Cantor, J.R. SHMT2 drives glioma cell survival in ischaemia but imposes a dependence on glycine clearance. Nature 2015, 520, 363–367. [Google Scholar] [CrossRef] [Green Version]
- Ross, K.C.; Andrews, A.J.; Marion, C.D.; Yen, T.J.; Bhattacharjee, V. Identification of the serine biosynthesis pathway as a critical component of BRAF inhibitor resistance of melanoma, pancreatic, and non–small cell lung cancer cells. Mol. Cancer Ther. 2017, 16, 1596–1609. [Google Scholar] [CrossRef] [Green Version]
- Lindell, E.J.; Erngren, I.; Engskog, M.; Haglöf, J.; Arvidsson, T.; Hedeland, M.; Petterson, C.; Laurell, G.; Nestor, M. Exploring Radiation Response in Two Head and Neck Squamous Carcinoma Cell Lines Through Metabolic Profiling. Front. Oncol. 2019, 9, 825. [Google Scholar] [CrossRef] [Green Version]
- Baumann, M.; Krause, M.; Hill, R. Exploring the role of cancer stem cells in radioresistance. Nat. Rev. Cancer 2008, 8, 545–554. [Google Scholar] [CrossRef]
- Yaromina, A.; Krause, M.; Thames, H.; Rosner, A.; Krause, M.; Hessel, F.; Grenman, R.; Zips, D.; Baumann, M. Pre-treatment number of clonogenic cells and their radiosensitivity are major determinants of local tumour control after fractionated irradiation. Radiother. Oncol. 2007, 83, 304–310. [Google Scholar] [CrossRef] [PubMed]
- De Koning, T.J.; Snell, K.; Duran, M.; Berger, R.; Surtees, R. L-serine in disease and development. Biochem. J. 2003, 371, 653–661. [Google Scholar] [CrossRef]
- Maddocks, O.D.; Labuschagne, C.F.; Adams, P.D.; Vousden, K.H. Serine metabolism supports the methionine cycle and DNA/RNA methylation through de novo ATP synthesis in cancer cells. Mol. Cell 2016, 61, 210–221. [Google Scholar] [CrossRef] [Green Version]
- Gao, X.; Lee, K.; Reid, M.A.; Sanderson, S.M.; Qiu, C.; Li, S.; Liu, J.; Locasale, J.W. Serine availability influences mitochondrial dynamics and function through lipid metabolism. Cell Rep. 2018, 22, 3507–3520. [Google Scholar] [CrossRef] [Green Version]
- Kottakis, F.; Nicolay, B.N.; Roumane, A.; Karnik, R.; Gu, H.; Nagle, J.M.; Boukhali, M.; Hayward, M.C.; Li, Y.Y.; Chen, T. LKB1 loss links serine metabolism to DNA methylation and tumorigenesis. Nature 2016, 539, 390–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tedeschi, P.M.; Markert, E.K.; Gounder, M.; Lin, H.; Dvorzhinski, D.; Dolfi, S.; Chan, L.L.; Qiu, J.; DiPaola, R.; Hirshfield, K. Contribution of serine, folate and glycine metabolism to the ATP, NADPH and purine requirements of cancer cells. Cell Death Dis. 2013, 4, e877. [Google Scholar] [CrossRef] [Green Version]
- Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef] [PubMed]
- Du, M.Q.; Carmichael, P.L.; Phillips, D.H. Induction of activating mutations in the human c-Ha-ras-1 proto-oncogene by oxygen free radicals. Mol. Carcinog. 1994, 11, 170–175. [Google Scholar] [CrossRef] [PubMed]
- Maciag, A.; Sithanandam, G.; Anderson, L.M. Mutant K-ras V12 increases COX-2, peroxides and DNA damage in lung cells. Carcinogenesis 2004, 25, 2231–2237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chandel, N.S.; McClintock, D.S.; Feliciano, C.E.; Wood, T.M.; Melendez, J.A.; Rodriguez, A.M.; Schumacker, P.T. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1α during hypoxia: A mechanism of O2 sensing. J. Biol. Chem. 2000, 275, 25130–25138. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; McMillan-Ward, E.; Kong, J.; Israels, S.; Gibson, S. Oxidative stress induces autophagic cell death independent of apoptosis in transformed and cancer cells. Cell Death Differ. 2008, 15, 171–182. [Google Scholar] [CrossRef]
- Gotoh, Y.; Cooper, J.A. Reactive oxygen species-and dimerization-induced activation of apoptosis signal-regulating kinase 1 in tumor necrosis factor-α signal transduction. J. Biol. Chem. 1998, 273, 17477–17482. [Google Scholar] [CrossRef] [Green Version]
- Fan, J.; Ye, J.; Kamphorst, J.J.; Shlomi, T.; Thompson, C.B.; Rabinowitz, J.D. Quantitative flux analysis reveals folate-dependent NADPH production. Nature 2014, 510, 298–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ducker, G.S.; Chen, L.; Morscher, R.J.; Ghergurovich, J.M.; Esposito, M.; Teng, X.; Kang, Y.; Rabinowitz, J.D. Reversal of cytosolic one-carbon flux compensates for loss of the mitochondrial folate pathway. Cell Metab. 2016, 23, 1140–1153. [Google Scholar] [CrossRef] [Green Version]
- Meiser, J.; Tumanov, S.; Maddocks, O.; Labuschagne, C.F.; Athineos, D.; Van Den Broek, N.; Mackay, G.M.; Gottlieb, E.; Blyth, K.; Vousden, K. Serine one-carbon catabolism with formate overflow. Sci. Adv. 2016, 2, e1601273. [Google Scholar] [CrossRef] [Green Version]
- Moreira, J.D.V.; Hamraz, M.; Abolhassani, M.; Bigan, E.; Pérès, S.; Paulevé, L.; Nogueira, M.L.; Steyaert, J.-M.; Schwartz, L. The redox status of cancer cells supports mechanisms behind the Warburg effect. Metabolites 2016, 6, 33. [Google Scholar] [CrossRef]
- Ju, H.-Q.; Lin, J.-F.; Tian, T.; Xie, D.; Xu, R.-H. NADPH homeostasis in cancer: Functions, mechanisms and therapeutic implications. Signal Transduct. Target. Ther. 2020, 5, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Possemato, R.; Marks, K.M.; Shaul, Y.D.; Pacold, M.E.; Kim, D.; Birsoy, K.; Sethumadhavan, S.; Woo, H.-K.; Jang, H.G.; Jha, A.K. Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature 2011, 476, 346–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, L.; Song, L.; Wan, Q.; Wu, G.; Li, X.; Wang, Y.; Wang, J.; Liu, Z.; Zhong, X.; He, X. cMyc-mediated activation of serine biosynthesis pathway is critical for cancer progression under nutrient deprivation conditions. Cell Res. 2015, 25, 429–444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kerr, E.M.; Gaude, E.; Turrell, F.K.; Frezza, C.; Martins, C.P. Mutant Kras copy number defines metabolic reprogramming and therapeutic susceptibilities. Nature 2016, 531, 110–113. [Google Scholar] [CrossRef] [Green Version]
- Harding, H.P.; Novoa, I.; Zhang, Y.; Zeng, H.; Wek, R.; Schapira, M.; Ron, D. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol. Cell 2000, 6, 1099–1108. [Google Scholar] [CrossRef]
- Ye, J.; Mancuso, A.; Tong, X.; Ward, P.S.; Fan, J.; Rabinowitz, J.D.; Thompson, C.B. Pyruvate kinase M2 promotes de novo serine synthesis to sustain mTORC1 activity and cell proliferation. Proc. Natl. Acad. Sci. USA 2012, 109, 6904–6909. [Google Scholar] [CrossRef] [Green Version]
- Chaneton, B.; Hillmann, P.; Zheng, L.; Martin, A.C.; Maddocks, O.D.; Chokkathukalam, A.; Coyle, J.E.; Jankevics, A.; Holding, F.P.; Vousden, K.H. Serine is a natural ligand and allosteric activator of pyruvate kinase M2. Nature 2012, 491, 458–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, S.; Ge, A.; Xu, S.; You, Z.; Ning, S.; Zhao, Y.; Pang, D. PSAT1 is regulated by ATF4 and enhances cell proliferation via the GSK3β/β-catenin/cyclin D1 signaling pathway in ER-negative breast cancer. J. Exp. Clin. Cancer Res. 2017, 36, 179. [Google Scholar] [CrossRef] [Green Version]
- Xia, Y.; Ye, B.; Ding, J.; Yu, Y.; Alptekin, A.; Thangaraju, M.; Prasad, P.D.; Ding, Z.-C.; Park, E.J.; Choi, J.-H. Metabolic reprogramming by MYCN confers dependence on the serine-glycine-one-carbon biosynthetic pathway. Cancer Res. 2019, 79, 3837–3850. [Google Scholar] [CrossRef] [Green Version]
- Li, K.; Wu, J.-l.; Qin, B.; Fan, Z.; Tang, Q.; Lu, W.; Zhang, H.; Xing, F.; Meng, M.; Zou, S. ILF3 is a substrate of SPOP for regulating serine biosynthesis in colorectal cancer. Cell Res. 2020, 30, 163–178. [Google Scholar] [CrossRef]
- Kinnaird, A.; Zhao, S.; Wellen, K.E.; Michelakis, E.D. Metabolic control of epigenetics in cancer. Nat. Rev. Cancer 2016, 16, 694–707. [Google Scholar] [CrossRef]
- Wong, C.; Qian, Y.; Yu, J. Interplay between epigenetics and metabolism in oncogenesis: Mechanisms and therapeutic approaches. Oncogene 2017, 36, 3359–3374. [Google Scholar] [CrossRef]
- Ding, J.; Li, T.; Wang, X.; Zhao, E.; Choi, J.-H.; Yang, L.; Zha, Y.; Dong, Z.; Huang, S.; Asara, J.M. The histone H3 methyltransferase G9A epigenetically activates the serine-glycine synthesis pathway to sustain cancer cell survival and proliferation. Cell Metab. 2013, 18, 896–907. [Google Scholar] [CrossRef] [Green Version]
- Zhao, E.; Ding, J.; Xia, Y.; Liu, M.; Ye, B.; Choi, J.-H.; Yan, C.; Dong, Z.; Huang, S.; Zha, Y. KDM4C and ATF4 cooperate in transcriptional control of amino acid metabolism. Cell Rep. 2016, 14, 506–519. [Google Scholar] [CrossRef] [Green Version]
- Riscal, R.; Schrepfer, E.; Arena, G.; Cissé, M.Y.; Bellvert, F.; Heuillet, M.; Rambow, F.; Bonneil, E.; Sabourdy, F.; Vincent, C. Chromatin-bound MDM2 regulates serine metabolism and redox homeostasis independently of p53. Mol. Cell 2016, 62, 890–902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cissé, M.Y.; Pyrdziak, S.; Firmin, N.; Gayte, L.; Heuillet, M.; Bellvert, F.; Fuentes, M.; Delpech, H.; Riscal, R.; Arena, G. Targeting MDM2-dependent serine metabolism as a therapeutic strategy for liposarcoma. Sci. Transl. Med. 2020. [Google Scholar] [CrossRef]
- Efimova, E.V.; Takahashi, S.; Shamsi, N.A.; Wu, D.; Labay, E.; Ulanovskaya, O.A.; Weichselbaum, R.R.; Kozmin, S.A.; Kron, S.J. Linking cancer metabolism to DNA repair and accelerated senescence. Mol. Cancer Res. 2016, 14, 173–184. [Google Scholar] [CrossRef] [Green Version]
- Najjar, Y.G.; Menk, A.V.; Sander, C.; Rao, U.; Karunamurthy, A.; Bhatia, R.; Zhai, S.; Kirkwood, J.M.; Delgoffe, G.M. Tumor cell oxidative metabolism as a barrier to PD-1 blockade immunotherapy in melanoma. JCI Insight 2019. [Google Scholar] [CrossRef] [PubMed]
- Baenke, F.; Chaneton, B.; Smith, M.; Van Den Broek, N.; Hogan, K.; Tang, H.; Viros, A.; Martin, M.; Galbraith, L.; Girotti, M.R. Resistance to BRAF inhibitors induces glutamine dependency in melanoma cells. Mol. Oncol. 2016, 10, 73–84. [Google Scholar] [CrossRef] [PubMed]
- Ascierto, P.A.; Schadendorf, D.; Berking, C.; Agarwala, S.S.; van Herpen, C.M.; Queirolo, P.; Blank, C.U.; Hauschild, A.; Beck, J.T.; St-Pierre, A. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: A non-randomised, open-label phase 2 study. Lancet Oncol. 2013, 14, 249–256. [Google Scholar] [CrossRef]
- Zimmer, L.; Barlesi, F.; Martinez-Garcia, M.; Dieras, V.; Schellens, J.H.; Spano, J.-P.; Middleton, M.R.; Calvo, E.; Paz-Ares, L.; Larkin, J. Phase I expansion and pharmacodynamic study of the oral MEK inhibitor RO4987655 (CH4987655) in selected patients with advanced cancer with RAS–RAF mutations. Clin. Cancer Res. 2014, 20, 4251–4261. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, M.Q.; Teh, J.L.; Purwin, T.J.; Chervoneva, I.; Davies, M.A.; Nathanson, K.L.; Cheng, P.F.; Levesque, M.P.; Dummer, R.; Aplin, A.E. Targeting PHGDH upregulation reduces glutathione levels and re-sensitizes resistant NRAS mutant melanoma to MEK inhibition. J. Investig. Dermatol. 2020, 140, 2242–2252. [Google Scholar] [CrossRef]
- Jin, N.; Bi, A.; Lan, X.; Xu, J.; Wang, X.; Liu, Y.; Wang, T.; Tang, S.; Zeng, H.; Chen, Z. Identification of metabolic vulnerabilities of receptor tyrosine kinases-driven cancer. Nat. Commun. 2019, 10, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Dong, J.-K.; Lei, H.-M.; Liang, Q.; Tang, Y.-B.; Zhou, Y.; Wang, Y.; Zhang, S.; Li, W.-B.; Tong, Y.; Zhuang, G. Overcoming erlotinib resistance in EGFR mutation-positive lung adenocarcinomas through repression of phosphoglycerate dehydrogenase. Theranostics 2018, 8, 1808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, L.; Lee, D.; Law, C.-T.; Zhang, M.S.; Shen, J.; Chin, D.W.-C.; Zhang, A.; Tsang, F.H.-C.; Wong, C.L.-S.; Ng, I.O.-L. Genome-wide CRISPR/Cas9 library screening identified PHGDH as a critical driver for Sorafenib resistance in HCC. Nat. Commun. 2019, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Nwosu, Z.C.; Piorońska, W.; Battello, N.; Zimmer, A.D.; Dewidar, B.; Han, M.; Pereira, S.; Blagojevic, B.; Castven, D.; Charlestin, V. Severe metabolic alterations in liver cancer lead to ERK pathway activation and drug resistance. EBioMedicine 2020, 54, 102699. [Google Scholar] [CrossRef] [PubMed]
- Zaal, E.A.; Wu, W.; Jansen, G.; Zweegman, S.; Cloos, J.; Berkers, C.R. Bortezomib resistance in multiple myeloma is associated with increased serine synthesis. Cancer Metab. 2017, 5, 7. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Xia, J.; Zhang, J.; Zhu, Y.; Wu, Y.; Guo, J.; Chen, S.; Lei, Q.; Meng, B.; Kuang, C. Phosphoglycerate dehydrogenase promotes proliferation and bortezomib resistance through increasing reduced glutathione synthesis in multiple myeloma. Br. J. Haematol. 2020, 190, 52–66. [Google Scholar] [CrossRef]
- Lipchick, B.C.; Fink, E.E.; Nikiforov, M.A. Oxidative stress and proteasome inhibitors in multiple myeloma. Pharmacol. Res. 2016, 105, 210–215. [Google Scholar] [CrossRef] [Green Version]
- Niewerth, D.; Jansen, G.; Assaraf, Y.G.; Zweegman, S.; Kaspers, G.J.; Cloos, J. Molecular basis of resistance to proteasome inhibitors in hematological malignancies. Drug Resist. Updates 2015, 18, 18–35. [Google Scholar] [CrossRef] [PubMed]
- Yoshino, H.; Nohata, N.; Miyamoto, K.; Yonemori, M.; Sakaguchi, T.; Sugita, S.; Itesako, T.; Kofuji, S.; Nakagawa, M.; Dahiya, R. PHGDH as a key enzyme for serine biosynthesis in HIF2α-targeting therapy for renal cell carcinoma. Cancer Res. 2017, 77, 6321–6329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samanta, D.; Park, Y.; Andrabi, S.A.; Shelton, L.M.; Gilkes, D.M.; Semenza, G.L. PHGDH expression is required for mitochondrial redox homeostasis, breast cancer stem cell maintenance, and lung metastasis. Cancer Res. 2016, 76, 4430–4442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.; Bai, W. Repression of phosphoglycerate dehydrogenase sensitizes triple-negative breast cancer to doxorubicin. Cancer Chemother. Pharmacol. 2016, 78, 655–659. [Google Scholar] [CrossRef]
- Chen, C.-Y.; Chang, Y.-L.; Shih, J.-Y.; Lin, J.-W.; Chen, K.-Y.; Yang, C.-H.; Yu, C.-J.; Yang, P.-C. Thymidylate synthase and dihydrofolate reductase expression in non-small cell lung carcinoma: The association with treatment efficacy of pemetrexed. Lung Cancer 2011, 74, 132–138. [Google Scholar] [CrossRef] [PubMed]
- Daidone, F.; Florio, R.; Rinaldo, S.; Contestabile, R.; di Salvo, M.L.; Cutruzzolà, F.; Bossa, F.; Paiardini, A. In silico and in vitro validation of serine hydroxymethyltransferase as a chemotherapeutic target of the antifolate drug pemetrexed. Eur. J. Med. Chem. 2011, 46, 1616–1621. [Google Scholar] [CrossRef] [PubMed]
- Parker, W.B.; Cheng, Y.C. Metabolism and mechanism of action of 5-fluorouracil. Pharmacol. Ther. 1990, 48, 381–395. [Google Scholar] [CrossRef]
- Carbone, A.; Rodeck, U.; Mauri, F.A.; Sozzi, M.; Gaspari, F.; Smirne, C.; Prati, A.; Addeo, A.; Novarino, A.; Robecchi, A. Human pancreatic carcinoma cells secrete bioactive interleukin-18 after treatment with 5-fluorouracil: Implications for anti-tumor immune response. Cancer Biol. Ther. 2005. [Google Scholar] [CrossRef] [Green Version]
- Montrose, D.C.; Saha, S.; Foronda, M.; McNally, E.M.; Chen, J.; Zhou, X.K.; Ha, T.; Krumsiek, J.; Buyukozkan, M.; Verma, A. Exogenous and endogenous sources of serine contribute to colon cancer metabolism, growth, and resistance to 5-fluorouracil. Cancer Res. 2021. [Google Scholar] [CrossRef] [PubMed]
- Lewanski, C.R.; Gullick, W.J. Radiotherapy and cellular signalling. Lancet Oncol. 2001, 2, 366–370. [Google Scholar] [CrossRef]
- Delaney, G.; Jacob, S.; Featherstone, C.; Barton, M. The role of radiotherapy in cancer treatment: Estimating optimal utilization from a review of evidence-based clinical guidelines. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2005, 104, 1129–1137. [Google Scholar] [CrossRef]
- Gewirtz, D.A. Growth arrest and cell death in the breast tumor cell in response to ionizing radiation and chemotherapeutic agents which induce DNA damage. Breast Cancer Res. Treat. 2000, 62, 223–235. [Google Scholar] [CrossRef]
- Hawkins, A.J.; Golding, S.E.; Khalil, A.; Valerie, K. DNA double-strand break–induced pro-survival signaling. Radiother. Oncol. 2011, 101, 13–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raleigh, D.R.; Haas-Kogan, D.A. Molecular targets and mechanisms of radiosensitization using DNA damage response pathways. Future Oncol. 2013, 9, 219–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menon, S.S.; Uppal, M.; Randhawa, S.; Cheema, M.S.; Aghdam, N.; Usala, R.L.; Ghosh, S.P.; Cheema, A.K.; Dritschilo, A. Radiation metabolomics: Current status and future directions. Front. Oncol. 2016, 6, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhatt, A.N.; Chauhan, A.; Khanna, S.; Rai, Y.; Singh, S.; Soni, R.; Kalra, N.; Dwarakanath, B.S. Transient elevation of glycolysis confers radio-resistance by facilitating DNA repair in cells. Bmc Cancer 2015, 15, 335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Quennet, V.; Yaromina, A.; Zips, D.; Rosner, A.; Walenta, S.; Baumann, M.; Mueller-Klieser, W. Tumor lactate content predicts for response to fractionated irradiation of human squamous cell carcinomas in nude mice. Radiother. Oncol. 2006, 81, 130–135. [Google Scholar] [CrossRef]
- Shimura, T.; Noma, N.; Sano, Y.; Ochiai, Y.; Oikawa, T.; Fukumoto, M.; Kunugita, N. AKT-mediated enhanced aerobic glycolysis causes acquired radioresistance by human tumor cells. Radiother. Oncol. 2014, 112, 302–307. [Google Scholar] [CrossRef]
- Sattler, U.G.; Meyer, S.S.; Quennet, V.; Hoerner, C.; Knoerzer, H.; Fabian, C.; Yaromina, A.; Zips, D.; Walenta, S.; Baumann, M. Glycolytic metabolism and tumour response to fractionated irradiation. Radiother. Oncol. 2010, 94, 102–109. [Google Scholar] [CrossRef]
- Liu, R.; Fan, M.; Candas, D.; Qin, L.; Zhang, X.; Eldridge, A.; Zou, J.X.; Zhang, T.; Juma, S.; Jin, C. CDK1-mediated SIRT3 activation enhances mitochondrial function and tumor radioresistance. Mol. Cancer Ther. 2015, 14, 2090–2102. [Google Scholar] [CrossRef] [Green Version]
- Tang, L.; Wei, F.; Wu, Y.; He, Y.; Shi, L.; Xiong, F.; Gong, Z.; Guo, C.; Li, X.; Deng, H. Role of metabolism in cancer cell radioresistance and radiosensitization methods. J. Exp. Clin. Cancer Res. 2018, 37, 87. [Google Scholar] [CrossRef]
- Fu, S.; Li, Z.; Xiao, L.; Hu, W.; Zhang, L.; Xie, B.; Zhou, Q.; He, J.; Qiu, Y.; Wen, M. Glutamine synthetase promotes radiation resistance via facilitating nucleotide metabolism and subsequent DNA damage repair. Cell Rep. 2019, 28, 1136–1143.e1134. [Google Scholar] [CrossRef] [Green Version]
- Wu, L.; Hu, Z.; Huang, Y.; Yu, Y.; Liang, W.; Zheng, Q.; Huang, X.; Huang, Y.; Lu, X.; Zhao, Y. Radiation changes the metabolic profiling of melanoma cell line B16. PLoS ONE 2016. [Google Scholar] [CrossRef] [PubMed]
- Bhute, V.J.; Ma, Y.; Bao, X.; Palecek, S.P. The poly (ADP-ribose) polymerase inhibitor veliparib and radiation cause significant cell line dependent metabolic changes in breast cancer cells. Sci. Rep. 2016, 6, 36061. [Google Scholar] [CrossRef] [Green Version]
- Arenas, M.; Rodríguez, E.; García-Heredia, A.; Fernández-Arroyo, S.; Sabater, S.; Robaina, R.; Gascón, M.; Rodríguez-Pla, M.; Cabré, N.; Luciano-Mateo, F. Metabolite normalization with local radiotherapy following breast tumor resection. PLoS ONE 2018, 13, e0207474. [Google Scholar] [CrossRef] [PubMed]
- Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Wakeman, T.P.; Lathia, J.D.; Hjelmeland, A.B.; Wang, X.F.; White, R.R.; Rich, J.N.; Sullenger, B.A. Notch promotes radioresistance of glioma stem cells. Stem Cells 2010, 28, 17–28. [Google Scholar] [CrossRef] [Green Version]
- Phillips, T.M.; McBride, W.H.; Pajonk, F. The response of CD24−/low/CD44+ breast cancer–initiating cells to radiation. J. Natl. Cancer Inst. 2006, 98, 1777–1785. [Google Scholar] [CrossRef] [Green Version]
- Desai, A.; Webb, B.; Gerson, S.L. CD133+ cells contribute to radioresistance via altered regulation of DNA repair genes in human lung cancer cells. Radiother. Oncol. 2014, 110, 538–545. [Google Scholar] [CrossRef] [Green Version]
- Diehn, M.; Cho, R.W.; Lobo, N.A.; Kalisky, T.; Dorie, M.J.; Kulp, A.N.; Qian, D.; Lam, J.S.; Ailles, L.E.; Wong, M. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009, 458, 780–783. [Google Scholar] [CrossRef] [PubMed]
- Sharif, T.; Martell, E.; Dai, C.; Ghassemi-Rad, M.S.; Lee, K.; Singh, S.K.; Weaver, I.C.; Gujar, S. Phosphoglycerate dehydrogenase inhibition induces p-mTOR-independent autophagy and promotes multilineage differentiation in embryonal carcinoma stem-like cells. Cell Death Dis. 2018, 9, 990. [Google Scholar] [CrossRef]
- Hwang, I.-Y.; Kwak, S.; Lee, S.; Kim, H.; Lee, S.E.; Kim, J.-H.; Kim, Y.A.; Jeon, Y.K.; Chung, D.H.; Jin, X. Psat1-dependent fluctuations in α-ketoglutarate affect the timing of ESC differentiation. Cell Metab. 2016, 24, 494–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ge, R.; Wang, Z.; Montironi, R.; Jiang, Z.; Cheng, M.; Santoni, M.; Huang, K.; Massari, F.; Lu, X.; Cimadamore, A. Epigenetic modulations and lineage plasticity in advanced prostate cancer. Ann. Oncol. 2020, 31, 470–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palapattu, G.S.; Wu, C.; Silvers, C.R.; Martin, H.B.; Williams, K.; Salamone, L.; Bushnell, T.; Huang, L.S.; Yang, Q.; Huang, J. Selective expression of CD44, a putative prostate cancer stem cell marker, in neuroendocrine tumor cells of human prostate cancer. Prostate 2009, 69, 787–798. [Google Scholar] [CrossRef]
- Wang, H.T.; Yao, Y.H.; Li, B.G.; Tang, Y.; Chang, J.W.; Zhang, J. Neuroendocrine Prostate Cancer (NEPC) progressing from conventional prostatic adenocarcinoma: Factors associated with time to development of NEPC and survival from NEPC diagnosis—a systematic review and pooled analysis. J. Clin. Oncol. 2014, 32, 3383–3390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reina-Campos, M.; Linares, J.F.; Duran, A.; Cordes, T.; L’Hermitte, A.; Badur, M.G.; Bhangoo, M.S.; Thorson, P.K.; Richards, A.; Rooslid, T. Increased serine and one-carbon pathway metabolism by PKCλ/ι deficiency promotes neuroendocrine prostate cancer. Cancer Cell 2019, 35, 385–400.e389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baksh, S.C.; Todorova, P.K.; Gur-Cohen, S.; Hurwitz, B.; Ge, Y.; Novak, J.S.; Tierney, M.T.; dela Cruz-Racelis, J.; Fuchs, E.; Finley, L.W. Extracellular serine controls epidermal stem cell fate and tumour initiation. Nat. Cell Biol. 2020, 22, 779–790. [Google Scholar] [CrossRef] [PubMed]
- Tran, T.Q.; Hanse, E.A.; Habowski, A.N.; Li, H.; Gabra, M.B.I.; Yang, Y.; Lowman, X.H.; Ooi, A.M.; Liao, S.Y.; Edwards, R.A. α-Ketoglutarate attenuates Wnt signaling and drives differentiation in colorectal cancer. Nat. Cancer 2020, 1, 345–358. [Google Scholar] [CrossRef]
- Prager, B.C.; Xie, Q.; Bao, S.; Rich, J.N. Cancer stem cells: The architects of the tumor ecosystem. Cell Stem Cell 2019, 24, 41–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carcereri de Prati, A.; Butturini, E.; Rigo, A.; Oppici, E.; Rossin, M.; Boriero, D.; Mariotto, S. Metastatic breast cancer cells enter into dormant state and express cancer stem cells phenotype under chronic hypoxia. J. Cell. Biochem. 2017, 118, 3237–3248. [Google Scholar] [CrossRef]
- Wu, C.-T.; Lin, W.-Y.; Chang, Y.-H.; Chen, W.-C.; Chen, M.-F. Impact of CD44 expression on radiation response for bladder cancer. J. Cancer 2017, 8, 1137. [Google Scholar] [CrossRef] [Green Version]
- Dong, C.; Yuan, T.; Wu, Y.; Wang, Y.; Fan, T.W.; Miriyala, S.; Lin, Y.; Yao, J.; Shi, J.; Kang, T. Loss of FBP1 by Snail-mediated repression provides metabolic advantages in basal-like breast cancer. Cancer Cell 2013, 23, 316–331. [Google Scholar] [CrossRef] [Green Version]
- Conley, S.J.; Gheordunescu, E.; Kakarala, P.; Newman, B.; Korkaya, H.; Heath, A.N.; Clouthier, S.G.; Wicha, M.S. Antiangiogenic agents increase breast cancer stem cells via the generation of tumor hypoxia. Proc. Natl. Acad. Sci. USA 2012, 109, 2784–2789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bao, S.; Wu, Q.; Sathornsumetee, S.; Hao, Y.; Li, Z.; Hjelmeland, A.B.; Shi, Q.; McLendon, R.E.; Bigner, D.D.; Rich, J.N. Stem cell–like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006, 66, 7843–7848. [Google Scholar] [CrossRef] [Green Version]
- Du, R.; Lu, K.V.; Petritsch, C.; Liu, P.; Ganss, R.; Passegué, E.; Song, H.; VandenBerg, S.; Johnson, R.S.; Werb, Z. HIF1α induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 2008, 13, 206–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hasmim, M.; Noman, M.Z.; Messai, Y.; Bordereaux, D.; Gros, G.; Baud, V.; Chouaib, S. Cutting edge: Hypoxia-induced Nanog favors the intratumoral infiltration of regulatory T cells and macrophages via direct regulation of TGF-β1. J. Immunol. 2013, 191, 5802–5806. [Google Scholar] [CrossRef] [Green Version]
- Ye, J.; Fan, J.; Venneti, S.; Wan, Y.-W.; Pawel, B.R.; Zhang, J.; Finley, L.W.; Lu, C.; Lindsten, T.; Cross, J.R. Serine catabolism regulates mitochondrial redox control during hypoxia. Cancer Discov. 2014, 4, 1406–1417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Engel, A.L.; Lorenz, N.I.; Klann, K.; Münch, C.; Depner, C.; Steinbach, J.P.; Ronellenfitsch, M.W.; Luger, A.-L. Serine-dependent redox homeostasis regulates glioblastoma cell survival. Br. J. Cancer 2020, 122, 1391–1398. [Google Scholar] [CrossRef] [PubMed]
- Xiang, L.; Xie, G.; Liu, C.; Zhou, J.; Chen, J.; Yu, S.; Li, J.; Pang, X.; Shi, H.; Liang, H. Knock-down of glutaminase 2 expression decreases glutathione, NADH, and sensitizes cervical cancer to ionizing radiation. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Res. 2013, 1833, 2996–3005. [Google Scholar] [CrossRef] [Green Version]
- Amelio, I.; Markert, E.; Rufini, A.; Antonov, A.; Sayan, B.; Tucci, P.; Agostini, M.; Mineo, T.; Levine, A.; Melino, G. p73 regulates serine biosynthesis in cancer. Oncogene 2014, 33, 5039–5046. [Google Scholar] [CrossRef] [Green Version]
- Singleton, D.C.; Harris, A.L. ATF4, hypoxia and treatment resistance in cancer. In The Unfolded Protein Response in Cancer; Springer: Cham, Switzerland, 2019; pp. 75–108. [Google Scholar]
- Rouschop, K.M.; Dubois, L.J.; Keulers, T.G.; van den Beucken, T.; Lambin, P.; Bussink, J.; van der Kogel, A.J.; Koritzinsky, M.; Wouters, B.G. PERK/eIF2α signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS. Proc. Natl. Acad. Sci. USA 2013, 110, 4622–4627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verfaillie, T.; Rubio, N.; Garg, A.; Bultynck, G.; Rizzuto, R.; Decuypere, J.; Piette, J.; Linehan, C.; Gupta, S.; Samali, A. PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ. 2012, 19, 1880–1891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dadey, D.Y.; Kapoor, V.; Khudanyan, A.; Thotala, D.; Hallahan, D.E. PERK regulates glioblastoma sensitivity to ER stress although promoting radiation resistance. Mol. Cancer Res. 2018, 16, 1447–1453. [Google Scholar] [CrossRef] [Green Version]
- Barker, H.E.; Paget, J.T.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef]
- Formenti, S.C.; Demaria, S. Systemic effects of local radiotherapy. Lancet Oncol. 2009, 10, 718–726. [Google Scholar] [CrossRef] [Green Version]
- Boustani, J.; Grapin, M.; Laurent, P.-A.; Apetoh, L.; Mirjolet, C. The 6th R of radiobiology: Reactivation of anti-tumor immune response. Cancers 2019, 11, 860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P. Toll-like receptor 4–dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef]
- Kroemer, G.; Galluzzi, L.; Kepp, O.; Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 2013, 31, 51–72. [Google Scholar] [CrossRef] [PubMed]
- Dewan, M.Z.; Galloway, A.E.; Kawashima, N.; Dewyngaert, J.K.; Babb, J.S.; Formenti, S.C.; Demaria, S. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti–CTLA-4 antibody. Clin. Cancer Res. 2009, 15, 5379–5388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, J.; Escamilla, J.; Mok, S.; David, J.; Priceman, S.; West, B.; Bollag, G.; McBride, W.; Wu, L. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res. 2013, 73, 2782–2794. [Google Scholar] [CrossRef] [Green Version]
- Colton, M.; Cheadle, E.J.; Honeychurch, J.; Illidge, T.M. Reprogramming the tumour microenvironment by radiotherapy: Implications for radiotherapy and immunotherapy combinations. Radiat. Oncol. 2020, 15, 254. [Google Scholar] [CrossRef]
- Gough, M.J.; Young, K.; Crittenden, M. The impact of the myeloid response to radiation therapy. Clin. Dev. Immunol. 2013, 2013, 281958. [Google Scholar] [CrossRef] [Green Version]
- Meyer, C.; Cagnon, L.; Costa-Nunes, C.M.; Baumgaertner, P.; Montandon, N.; Leyvraz, L.; Michielin, O.; Romano, E.; Speiser, D.E. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol. Immunother. 2014, 63, 247–257. [Google Scholar] [CrossRef] [Green Version]
- Dovedi, S.J.; Cheadle, E.J.; Popple, A.L.; Poon, E.; Morrow, M.; Stewart, R.; Yusko, E.C.; Sanders, C.M.; Vignali, M.; Emerson, R.O. Fractionated radiation therapy stimulates antitumor immunity mediated by both resident and infiltrating polyclonal T-cell populations when combined with PD-1 blockade. Clin. Cancer Res. 2017, 23, 5514–5526. [Google Scholar] [CrossRef] [Green Version]
- Shi, X.; Shiao, S.L. The role of macrophage phenotype in regulating the response to radiation therapy. Transl. Res. 2018, 191, 64–80. [Google Scholar] [CrossRef] [PubMed]
- Okubo, M.; Kioi, M.; Nakashima, H.; Sugiura, K.; Mitsudo, K.; Aoki, I.; Taniguchi, H.; Tohnai, I. M2-polarized macrophages contribute to neovasculogenesis, leading to relapse of oral cancer following radiation. Sci. Rep. 2016, 6, 27548. [Google Scholar] [CrossRef] [PubMed]
- De Palma, M.; Lewis, C.E. Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 2013, 23, 277–286. [Google Scholar] [CrossRef] [Green Version]
- Hughes, R.; Qian, B.-Z.; Rowan, C.; Muthana, M.; Keklikoglou, I.; Olson, O.C.; Tazzyman, S.; Danson, S.; Addison, C.; Clemons, M. Perivascular M2 macrophages stimulate tumor relapse after chemotherapy. Cancer Res. 2015, 75, 3479–3491. [Google Scholar] [CrossRef] [Green Version]
- Chaudary, N.; Pintilie, M.; Jelveh, S.; Lindsay, P.; Hill, R.P.; Milosevic, M. Plerixafor improves primary tumor response and reduces metastases in cervical cancer treated with radio-chemotherapy. Clin. Cancer Res. 2017, 23, 1242–1249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilson, J.L.; Nägele, T.; Linke, M.; Demel, F.; Fritsch, S.D.; Mayr, H.K.; Cai, Z.; Katholnig, K.; Sun, X.; Fragner, L. Inverse data-driven modeling and multiomics analysis reveals phgdh as a metabolic checkpoint of macrophage polarization and proliferation. Cell Rep. 2020, 30, 1542–1552.e1547. [Google Scholar] [CrossRef]
- Rodriguez, A.E.; Ducker, G.S.; Billingham, L.K.; Martinez, C.A.; Mainolfi, N.; Suri, V.; Friedman, A.; Manfredi, M.G.; Weinberg, S.E.; Rabinowitz, J.D. Serine metabolism supports macrophage IL-1β production. Cell Metab. 2019, 29, 1003–1011.e1004. [Google Scholar] [CrossRef] [Green Version]
- Yu, W.; Wang, Z.; Zhang, K.; Chi, Z.; Xu, T.; Jiang, D.; Chen, S.; Li, W.; Yang, X.; Zhang, X. One-carbon metabolism supports S-adenosylmethionine and histone methylation to drive inflammatory macrophages. Mol. Cell 2019, 75, 1147–1160.e1145. [Google Scholar] [CrossRef]
- Kaplanov, I.; Carmi, Y.; Kornetsky, R.; Shemesh, A.; Shurin, G.V.; Shurin, M.R.; Dinarello, C.A.; Voronov, E.; Apte, R.N. Blocking IL-1β reverses the immunosuppression in mouse breast cancer and synergizes with anti–PD-1 for tumor abrogation. Proc. Natl. Acad. Sci. USA 2019, 116, 1361–1369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, W.; Ding, I.; Chen, K.; Olschowka, J.; Xu, J.; Hu, D.; Morrow, G.R.; Okunieff, P. Interleukin 1β (IL1B) signaling is a critical component of radiation-induced skin fibrosis. Radiat. Res. 2006, 165, 181–191. [Google Scholar] [CrossRef] [PubMed]
- O’Brien-Ladner, A.; Nelson, M.E.; Kimler, B.F.; Wesselius, L.J. Release of interleukin-1 by human alveolar macrophages after in vitro irradiation. Radiat. Res. 1993, 136, 37–41. [Google Scholar] [CrossRef] [PubMed]
- Fuks, Z.; Kolesnick, R. Engaging the vascular component of the tumor response. Cancer Cell 2005, 8, 89–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soto, I.C.; Fontanesi, F.; Myers, R.S.; Hamel, P.; Barrientos, A. A heme-sensing mechanism in the translational regulation of mitochondrial cytochrome c oxidase biogenesis. Cell Metab. 2012, 16, 801–813. [Google Scholar] [CrossRef] [Green Version]
- Vandekeere, S.; Dubois, C.; Kalucka, J.; Sullivan, M.R.; García-Caballero, M.; Goveia, J.; Chen, R.; Diehl, F.F.; Bar-Lev, L.; Souffreau, J. Serine synthesis via PHGDH is essential for heme production in endothelial cells. Cell Metab. 2018, 28, 573–587.e513. [Google Scholar] [CrossRef] [Green Version]
- Gray, L.H.; Conger, A.D.; Ebert, M.; Hornsey, S.; Scott, O. The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br. J. Radiol. 1953, 26, 638–648. [Google Scholar] [CrossRef]
- Markkanen, E.; Fischer, R.; Ledentcova, M.; Kessler, B.M.; Dianov, G.L. Cells deficient in base-excision repair reveal cancer hallmarks originating from adjustments to genetic instability. Nucleic Acids Res. 2015, 43, 3667–3679. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Zheng, A.; Hydbring, P.; Ambroise, G.; Ouchida, A.T.; Goiny, M.; Vakifahmetoglu-Norberg, H.; Norberg, E. PHGDH defines a metabolic subtype in lung adenocarcinomas with poor prognosis. Cell Rep. 2017, 19, 2289–2303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lukas, J.; Lukas, C.; Bartek, J. Mammalian cell cycle checkpoints: Signalling pathways and their organization in space and time. DNA Repair 2004, 3, 997–1007. [Google Scholar] [CrossRef]
- Gudkov, A.V.; Komarova, E.A. The role of p53 in determining sensitivity to radiotherapy. Nat. Rev. Cancer 2003, 3, 117–129. [Google Scholar] [CrossRef]
- Maddocks, O.D.; Berkers, C.R.; Mason, S.M.; Zheng, L.; Blyth, K.; Gottlieb, E.; Vousden, K.H. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature 2013, 493, 542–546. [Google Scholar] [CrossRef]
- Jain, M.; Nilsson, R.; Sharma, S.; Madhusudhan, N.; Kitami, T.; Souza, A.L.; Kafri, R.; Kirschner, M.W.; Clish, C.B.; Mootha, V.K. Metabolite profiling identifies a key role for glycine in rapid cancer cell proliferation. Science 2012, 336, 1040–1044. [Google Scholar] [CrossRef] [Green Version]
- Tajan, M.; Hennequart, M.; Cheung, E.C.; Zani, F.; Hock, A.K.; Legrave, N.; Maddocks, O.D.; Ridgway, R.A.; Athineos, D.; Suárez-Bonnet, A.; et al. Serine synthesis pathway inhibition cooperates with dietary serine and glycine limitation for cancer therapy. Nat. Commun. 2021, 12, 366. [Google Scholar] [CrossRef] [PubMed]
- Méndez-Lucas, A.; Lin, W.; Driscoll, P.C.; Legrave, N.; Novellasdemunt, L.; Xie, C.; Charles, M.; Wilson, Z.; Jones, N.P.; Rayport, S. Identifying strategies to target the metabolic flexibility of tumours. Nat. Metab. 2020, 2, 335–350. [Google Scholar] [CrossRef] [PubMed]
- Sullivan, M.R.; Mattaini, K.R.; Dennstedt, E.A.; Nguyen, A.A.; Sivanand, S.; Reilly, M.F.; Meeth, K.; Muir, A.; Darnell, A.M.; Bosenberg, M.W. Increased serine synthesis provides an advantage for tumors arising in tissues where serine levels are limiting. Cell Metab. 2019, 29, 1410–1421.e1414. [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. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [Green Version]
- Farnsworth, R.H.; Lackmann, M.; Achen, M.G.; Stacker, S.A. Vascular remodeling in cancer. Oncogene 2014, 33, 3496–3505. [Google Scholar] [CrossRef] [Green Version]
- Gravel, S.-P.; Hulea, L.; Toban, N.; Birman, E.; Blouin, M.-J.; Zakikhani, M.; Zhao, Y.; Topisirovic, I.; St-Pierre, J.; Pollak, M. Serine deprivation enhances antineoplastic activity of biguanides. Cancer Res. 2014, 74, 7521–7533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bridges, H.R.; Jones, A.J.; Pollak, M.N.; Hirst, J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem. J. 2014, 462, 475–487. [Google Scholar] [CrossRef] [Green Version]
- Geeraerts, S.L.; Kampen, K.R.; Rinaldi, G.; Gupta, P.; Planque, M.; Louros, N.; Heylen, E.; De Cremer, K.; De Brucker, K.; Vereecke, S. Repurposing the Antidepressant Sertraline as SHMT Inhibitor to Suppress Serine/Glycine Synthesis–Addicted Breast Tumor Growth. Mol. Cancer Ther. 2020, 20, 50–63. [Google Scholar] [CrossRef]
- Bao, X.R.; Ong, S.-E.; Goldberger, O.; Peng, J.; Sharma, R.; Thompson, D.A.; Vafai, S.B.; Cox, A.G.; Marutani, E.; Ichinose, F. Mitochondrial dysfunction remodels one-carbon metabolism in human cells. elife 2016, 5, e10575. [Google Scholar] [CrossRef]
- McCann, E.; O’Sullivan, J.; Marcone, S. Targeting cancer-cell mitochondria and metabolism to improve radiotherapy response. Transl. Oncol. 2020, 14, 100905. [Google Scholar] [CrossRef] [PubMed]
- Zannella, V.E.; Dal Pra, A.; Muaddi, H.; McKee, T.D.; Stapleton, S.; Sykes, J.; Glicksman, R.; Chaib, S.; Zamiara, P.; Milosevic, M. Reprogramming metabolism with metformin improves tumor oxygenation and radiotherapy response. Clin. Cancer Res. 2013, 19, 6741–6750. [Google Scholar] [CrossRef] [Green Version]
- Lynam-Lennon, N.; Maher, S.G.; Maguire, A.; Phelan, J.; Muldoon, C.; Reynolds, J.V.; O’Sullivan, J. Altered mitochondrial function and energy metabolism is associated with a radioresistant phenotype in oesophageal adenocarcinoma. PLoS ONE 2014, 9, e100738. [Google Scholar] [CrossRef]
- Feng, T.; Li, L.; Ling, S.; Fan, N.; Fang, M.; Zhang, H.; Fang, X.; Lan, W.; Hou, Z.; Meng, Q. Metformin enhances radiation response of ECa109 cells through activation of ATM and AMPK. Biomed. Pharmacother. 2015, 69, 260–266. [Google Scholar] [CrossRef] [PubMed]
- Storozhuk, Y.; Hopmans, S.; Sanli, T.; Barron, C.; Tsiani, E.; Cutz, J.; Pond, G.; Wright, J.; Singh, G.; Tsakiridis, T. Metformin inhibits growth and enhances radiation response of non-small cell lung cancer (NSCLC) through ATM and AMPK. Br. J. Cancer 2013, 108, 2021–2032. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Storr, S.J.; Johnson, K.; Green, A.R.; Rakha, E.A.; Ellis, I.O.; Morgan, D.A.; Martin, S.G. Involvement of metformin and AMPK in the radioresponse and prognosis of luminal versus basal-like breast cancer treated with radiotherapy. Oncotarget 2014, 5, 12936. [Google Scholar] [CrossRef] [Green Version]
- Skinner, H.D.; McCurdy, M.R.; Echeverria, A.E.; Lin, S.H.; Welsh, J.W.; O’Reilly, M.S.; Hofstetter, W.L.; Ajani, J.A.; Komaki, R.; Cox, J.D. Metformin use and improved response to therapy in esophageal adenocarcinoma. Acta Oncol. 2013, 52, 1002–1009. [Google Scholar] [CrossRef] [Green Version]
- Ron-Harel, N.; Santos, D.; Ghergurovich, J.M.; Sage, P.T.; Reddy, A.; Lovitch, S.B.; Dephoure, N.; Satterstrom, F.K.; Sheffer, M.; Spinelli, J.B. Mitochondrial biogenesis and proteome remodeling promote one-carbon metabolism for T cell activation. Cell Metab. 2016, 24, 104–117. [Google Scholar] [CrossRef] [Green Version]
- Ma, E.H.; Bantug, G.; Griss, T.; Condotta, S.; Johnson, R.M.; Samborska, B.; Mainolfi, N.; Suri, V.; Guak, H.; Balmer, M.L. Serine is an essential metabolite for effector T cell expansion. Cell Metab. 2017, 25, 345–357. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sánchez-Castillo, A.; Vooijs, M.; Kampen, K.R. Linking Serine/Glycine Metabolism to Radiotherapy Resistance. Cancers 2021, 13, 1191. https://doi.org/10.3390/cancers13061191
Sánchez-Castillo A, Vooijs M, Kampen KR. Linking Serine/Glycine Metabolism to Radiotherapy Resistance. Cancers. 2021; 13(6):1191. https://doi.org/10.3390/cancers13061191
Chicago/Turabian StyleSánchez-Castillo, Anaís, Marc Vooijs, and Kim R. Kampen. 2021. "Linking Serine/Glycine Metabolism to Radiotherapy Resistance" Cancers 13, no. 6: 1191. https://doi.org/10.3390/cancers13061191
APA StyleSánchez-Castillo, A., Vooijs, M., & Kampen, K. R. (2021). Linking Serine/Glycine Metabolism to Radiotherapy Resistance. Cancers, 13(6), 1191. https://doi.org/10.3390/cancers13061191