Lipidomic-Based Advances in Diagnosis and Modulation of Immune Response to Cancer
Abstract
:1. Introduction
2. Lipid Metabolism Impacts Immune Activation against Tumor Progression
2.1. Lipid Interplay with Immune Regulation
2.2. Short-Chain Fatty Acids from Gut Microbiota as Effectors of the Immune System
3. Lipids as Biomarkers of Immune Response to Cancer
4. Active Modulation of Lipid Metabolism to Improve CAR T Cell Therapy
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Watson, J.D.; Crick, F.H.C. Molecular Structure of Nucleic Acids: A Structure for Deoxyribose Nucleic Acid. Nature 1953, 171, 737–738. [Google Scholar] [CrossRef] [PubMed]
- Check Hayden, E. Technology: The $1,000 genome. Nature 2014, 507, 294–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rattray, N.J.W.; Deziel, N.C.; Wallach, J.D.; Khan, S.A.; Vasiliou, V.; Ioannidis, J.P.A.; Johnson, C.H. Beyond genomics: Understanding exposotypes through metabolomics. Hum. Genom. 2018, 12, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Romero, R.; Espinoza, J.; Gotsch, F.; Kusanovic, J.; Friel, L.; Erez, O.; Mazaki-Tovi, S.; Than, N.; Hassan, S.; Tromp, G. The use of high-dimensional biology (genomics, transcriptomics, proteomics, and metabolomics) to understand the preterm parturition syndrome. BJOG Int. J. Obstet. Gynaecol. 2006, 113, 118–135. [Google Scholar] [CrossRef]
- Lam, S.M.; Tian, H.; Shui, G. Lipidomics, en route to accurate quantitation. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2017, 1862, 752–761. [Google Scholar] [CrossRef]
- Mancini, R.; Noto, A.; Pisanu, M.E.; De Vitis, C.; Maugeri-Saccà, M.; Ciliberto, G. Metabolic features of cancer stem cells: The emerging role of lipid metabolism. Oncogene 2018, 37, 2367–2378. [Google Scholar] [CrossRef]
- Mohamed, A.; Collins, J.; Jiang, H.; Molendijk, J.; Stoll, T.; Torta, F.; Wenk, M.R.; Bird, R.J.; Marlton, P.; Mollee, P.; et al. Concurrent lipidomics and proteomics on malignant plasma cells from multiple myeloma patients: Probing the lipid metabolome. PLoS ONE 2020, 15, e0227455. [Google Scholar] [CrossRef] [Green Version]
- Domblides, C.; Lartigue, L.; Faustin, B. Control of the Antitumor Immune Response by Cancer Metabolism. Cells 2019, 8, 104. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- 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.; et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood 2010, 115, 4742–4749. [Google Scholar] [CrossRef] [Green Version]
- Lauterbach, M.A.; Hanke, J.E.; Serefidou, M.; Mangan, M.S.J.; Kolbe, C.C.; Hess, T.; Rothe, M.; Kaiser, R.; Hoss, F.; Gehlen, J.; et al. Toll-like Receptor Signaling Rewires Macrophage Metabolism and Promotes Histone Acetylation via ATP-Citrate Lyase. Immunity 2019, 51, 997–1011. [Google Scholar] [CrossRef] [PubMed]
- Márquez, S.; Fernández, J.J.; Mancebo, C.; Herrero-Sánchez, C.; Alonso, S.; Sandoval, T.A.; Rodríguez Prados, M.; Cubillos-Ruiz, J.R.; Montero, O.; Fernández, N.; et al. Tricarboxylic Acid Cycle Activity and Remodeling of Glycerophosphocholine Lipids Support Cytokine Induction in Response to Fungal Patterns. Cell Rep. 2019, 27, 525–536. [Google Scholar] [CrossRef] [Green Version]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.M.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and Activity of Anti–PD-L1 Antibody in Patients with Advanced Cancer. New Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [Green Version]
- Donini, C.; D’Ambrosio, L.; Grignani, G.; Aglietta, M.; Sangiolo, D. Next generation immune-checkpoints for cancer therapy. J. Thorac. Dis. 2018, 10, S1581–S1601. [Google Scholar] [CrossRef]
- Patsoukis, N.; Bardhan, K.; Chatterjee, P.; Sari, D.; Liu, B.; Bell, L.N.; Karoly, E.D.; Freeman, G.J.; Petkova, V.; Seth, P.; et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat. Commun. 2015, 6, 6692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ogando, J.; Sáez, M.E.; Santos, J.; Nuevo-Tapioles, C.; Gut, M.; Esteve-Codina, A.; Heath, S.; González-Pérez, A.; Cuezva, J.M.; Lacalle, R.A.; et al. PD-1 signaling affects cristae morphology and leads to mitochondrial dysfunction in human CD8+ T lymphocytes. J. Immunother. Cancer 2019, 7, 151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Varga, T.; Czimmerer, Z.; Nagy, L. PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2011, 1812, 1007–1022. [Google Scholar] [CrossRef]
- Cipolletta, D.; Feuerer, M.; Li, A.; Kamei, N.; Lee, J.; Shoelson, S.E.; Benoist, C.; Mathis, D. PPAR-γ is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 2012, 486, 549–553. [Google Scholar] [CrossRef]
- Zhao, Y.; Lee, C.K.; Lin, C.H.; Gassen, R.B.; Xu, X.; Huang, Z.; Xiao, C.; Bonorino, C.; Lu, L.F.; Bui, J.D.; et al. PD-L1:CD80 Cis-Heterodimer Triggers the Co-stimulatory Receptor CD28 While Repressing the Inhibitory PD-1 and CTLA-4 Pathways. Immunity 2019, 51, 1059–1073. [Google Scholar] [CrossRef]
- Chowdhury, P.S.; Chamoto, K.; Kumar, A.; Honjo, T. PPAR-Induced Fatty Acid Oxidation in T Cells Increases the Number of Tumor-Reactive CD8+ T Cells and Facilitates Anti-PD-1 Therapy. Cancer Immunol. Res. 2018, 6, 1375–1387. [Google Scholar] [CrossRef] [PubMed]
- Saibil, S.D.; St. Paul, M.; Laister, R.C.; Garcia-Batres, C.R.; Israni-Winger, K.; Elford, A.R.; Grimshaw, N.; Robert-Tissot, C.; Roy, D.G.; Jones, R.G.; et al. Activation of Peroxisome Proliferator-Activated Receptors α and δ Synergizes with Inflammatory Signals to Enhance Adoptive Cell Therapy. Cancer Res. 2019, 79, 445–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gutiérrez, S.; Svahn, S.L.; Johansson, M.E. Effects of Omega-3 Fatty Acids on Immune Cells. Int. J. Mol. Sci. 2019, 20, 5028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mukaro, V.R.; Costabile, M.; Murphy, K.J.; Hii, C.S.; Howe, P.R.; Ferrante, A. Leukocyte numbers and function in subjects eating n-3 enriched foods: Selective depression of natural killer cell levels. Arthritis Res. Ther. 2008, 10, R57. [Google Scholar] [CrossRef] [Green Version]
- Miles, E.A.; Banerjee, T.; Wells, S.J.; Calder, P.C. Limited effect of eicosapentaenoic acid on T-lymphocyte and natural killer cell numbers and functions in healthy young males. Nutrition 2006, 22, 512–519. [Google Scholar] [CrossRef]
- Ichimura, A.; Hirasawa, A.; Poulain-Godefroy, O.; Bonnefond, A.; Hara, T.; Yengo, L.; Kimura, I.; Leloire, A.; Liu, N.; Iida, K.; et al. Dysfunction of lipid sensor GPR120 leads to obesity in both mouse and human. Nature 2012, 483, 350–354. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Hopkins, M.M.; Zhang, Z.; Quisenberry, C.B.; Fix, L.C.; Galvan, B.M.; Meier, K.E. Omega-3 fatty acids and other FFA4 agonists inhibit growth factor signaling in human prostate cancer cells. J. Pharmacol. Exp. Ther. 2015, 352, 380–394. [Google Scholar] [CrossRef]
- Thirunavukkarasan, M.; Wang, C.; Rao, A.; Hind, T.; Teo, Y.R.; Siddiquee, A.A.M.; Goghari, M.A.I.; Kumar, A.P.; Herr, D.R. Short-chain fatty acid receptors inhibit invasive phenotypes in breast cancer cells. PLoS ONE 2017, 12, e0186334. [Google Scholar] [CrossRef]
- Berod, L.; Friedrich, C.; Nandan, A.; Freitag, J.; Hagemann, S.; Harmrolfs, K.; Sandouk, A.; Hesse, C.; Castro, C.N.; Bähre, H.; et al. De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat. Med. 2014, 20, 1327–1333. [Google Scholar] [CrossRef]
- Ecker, C.; Guo, L.; Voicu, S.; Gil-de-Gómez, L.; Medvec, A.; Cortina, L.; Pajda, J.; Andolina, M.; Torres-Castillo, M.; Donato, J.L.; et al. Differential Reliance on Lipid Metabolism as a Salvage Pathway Underlies Functional Differences of T Cell Subsets in Poor Nutrient Environments. Cell Rep. 2018, 23, 741–755. [Google Scholar] [CrossRef] [Green Version]
- Essig, K.; Hu, D.; Guimaraes, J.C.; Alterauge, D.; Edelmann, S.; Raj, T.; Kranich, J.; Behrens, G.; Heiseke, A.; Floess, S.; et al. Roquin Suppresses the PI3K-mTOR Signaling Pathway to Inhibit T Helper Cell Differentiation and Conversion of Treg to Tfr Cells. Immunity 2017, 47, 1067–1082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Angelin, A.; Gil-de-Gómez, L.; Dahiya, S.; Jiao, J.; Guo, L.; Levine, M.H.; Wang, Z.; Quinn, W.J.; Kopinski, P.K.; Wang, L.; et al. Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. Cell Metab. 2017, 25, 1282–1293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mizuno, R.; Kawada, K.; Sakai, Y. Prostaglandin E2/EP Signaling in the Tumor Microenvironment of Colorectal Cancer. Int. J. Mol. Sci. 2019, 20, 6254. [Google Scholar] [CrossRef] [Green Version]
- Gil-de-Gómez, L.; Astudillo, A.M.; Lebrero, P.; Balboa, M.A.; Balsinde, J. Essential Role for Ethanolamine Plasmalogen Hydrolysis in Bacterial Lipopolysaccharide Priming of Macrophages for Enhanced Arachidonic Acid Release. Front. Immunol. 2017, 8, 1251. [Google Scholar] [CrossRef]
- Li, J.; Feng, G.; Liu, J.; Rong, R.; Luo, F.; Guo, L.; Zhu, T.; Wang, G.; Chu, Y. Renal cell carcinoma may evade the immune system by converting CD4+Foxp3- T cells into CD4+CD25+Foxp3+ regulatory T cells: Role of tumor COX-2-derived PGE2. Mol. Med. Rep. 2010, 3, 959–963. [Google Scholar] [CrossRef] [Green Version]
- Kalinski, P. Regulation of Immune Responses by Prostaglandin E2. J. Immunol. 2012, 188, 21–28. [Google Scholar] [CrossRef] [Green Version]
- Trinath, J.; Hegde, P.; Sharma, M.; Maddur, M.S.; Rabin, M.; Vallat, J.M.; Magy, L.; Balaji, K.N.; Kaveri, S.V.; Bayry, J. Intravenous immunoglobulin expands regulatory T cells via induction of cyclooxygenase-2–dependent prostaglandin E2 in human dendritic cells. Blood 2013, 122, 1419–1427. [Google Scholar] [CrossRef]
- Loew, A.; Köhnke, T.; Rehbeil, E.; Pietzner, A.; Weylandt, K.H. A Role for Lipid Mediators in Acute Myeloid Leukemia. Int. J. Mol. Sci. 2019, 20, 2425. [Google Scholar] [CrossRef] [Green Version]
- Prima, V.; Kaliberova, L.N.; Kaliberov, S.; Curiel, D.T.; Kusmartsev, S. COX2/mPGES1/PGE 2 pathway regulates PD-L1 expression in tumor-associated macrophages and myeloid-derived suppressor cells. Proc. Natl. Acad. Sci. USA 2017, 114, 1117–1122. [Google Scholar] [CrossRef] [Green Version]
- Miao, J.; Lu, X.; Hu, Y.; Piao, C.; Wu, X.; Liu, X.; Huang, C.; Wang, Y.; Li, D.; Liu, J. Prostaglandin E2 and PD-1 mediated inhibition of antitumor CTL responses in the human tumor microenvironment. Oncotarget 2017, 8, 89802–89810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mock, A.; Zschäbitz, S.; Kirsten, R.; Scheffler, M.; Wolf, B.; Herold-Mende, C.; Kramer, R.; Busch, E.; Jenzer, M.; Jäger, D.; et al. Serum very long-chain fatty acid-containing lipids predict response to immune checkpoint inhibitors in urological cancers. Cancer Immunol. Immunother. 2019, 68, 2005–2014. [Google Scholar] [CrossRef] [PubMed]
- Wu, A.; Wu, Q.; Deng, Y.; Liu, Y.; Lu, J.; Liu, L.; Li, X.; Liao, C.; Zhao, B.; Song, H. Loss of VGLL4 Suppresses Tumor PD-L1 Expression and Immune Evasion. EMBO J. 2019, 38. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.C.C.; Everts, B.; Ivanova, Y.; O’Sullivan, D.; Nascimento, M.; Smith, A.M.; Beatty, W.; Love-Gregory, L.; Lam, W.Y.; O’Neill, C.M.; et al. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat. Immunol. 2014, 15, 846–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oishi, Y.; Spann, N.J.; Link, V.M.; Muse, E.D.; Strid, T.; Edillor, C.; Kolar, M.J.; Matsuzaka, T.; Hayakawa, S.; Tao, J.; et al. SREBP1 Contributes to Resolution of Pro-inflammatory TLR4 Signaling by Reprogramming Fatty Acid Metabolism. Cell Metab. 2017, 25, 412–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Griffiths, B.; Lewis, C.A.; Bensaad, K.; Ros, S.; Zhang, Q.; Ferber, E.C.; Konisti, S.; Peck, B.; Miess, H.; East, P.; et al. Sterol regulatory element binding protein-dependent regulation of lipid synthesis supports cell survival and tumor growth. Cancer Metab. 2013, 1, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, C.; Chikina, M.; Deshpande, R.; Menk, A.V.; Wang, T.; Tabib, T.; Brunazzi, E.A.; Vignali, K.M.; Sun, M.; Stolz, D.B.; et al. Treg Cells Promote the SREBP1-Dependent Metabolic Fitness of Tumor-Promoting Macrophages via Repression of CD8+ T Cell-Derived Interferon-γ. Immunity 2019, 51, 381–397. [Google Scholar] [CrossRef] [PubMed]
- Wu, S.; Näär, A.M. SREBP1-dependent de novo fatty acid synthesis gene expression is elevated in malignant melanoma and represents a cellular survival trait. Sci. Rep. 2019, 9, 10369. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Hua, L.; Fan, H.; He, Y.; Xu, W.; Zhang, L.; Yang, J.; Deng, F.; Zeng, F. Interplay of PKD3 with SREBP1 Promotes Cell Growth via Upregulating Lipogenesis in Prostate Cancer Cells. J. Cancer 2019, 10, 6395–6404. [Google Scholar] [CrossRef] [Green Version]
- Wang, B.; Tontonoz, P. Liver X receptors in lipid signalling and membrane homeostasis. Nat. Rev. Endocrinol. 2018, 14, 452–463. [Google Scholar] [CrossRef]
- Shimano, H.; Sato, R. SREBP-regulated lipid metabolism: Convergent physiology–divergent pathophysiology. Nat. Rev. Endocrinol. 2017, 13, 710–730. [Google Scholar] [CrossRef] [PubMed]
- Costales, P.; Castellano, J.; Revuelta-López, E.; Cal, R.; Aledo, R.; Llampayas, O.; Nasarre, L.; Juarez, C.; Badimon, L.; Llorente-Cortés, V. Lipopolysaccharide downregulates CD91/low-density lipoprotein receptor-related protein 1 expression through SREBP-1 overexpression in human macrophages. Atherosclerosis 2013, 227, 79–88. [Google Scholar] [CrossRef] [PubMed]
- Im, S.S.; Yousef, L.; Blaschitz, C.; Liu, J.Z.; Edwards, R.A.; Young, S.G.; Raffatellu, M.; Osborne, T.F. Linking Lipid Metabolism to the Innate Immune Response in Macrophages through Sterol Regulatory Element Binding Protein-1a. Cell Metab. 2011, 13, 540–549. [Google Scholar] [CrossRef] [Green Version]
- Joseph, S.B.; Castrillo, A.; Laffitte, B.A.; Mangelsdorf, D.J.; Tontonoz, P. Reciprocal regulation of inflammation and lipid metabolism by liver X receptors. Nat. Med. 2003, 9, 213–219. [Google Scholar] [CrossRef] [PubMed]
- Chausse, B.; Kakimoto, P.A.; Caldeira-Da-Silva, C.C.; Chaves-Filho, A.B.; Yoshinaga, M.Y.; Da Silva, R.P.; Miyamoto, S.; Kowaltowski, A.J. Distinct metabolic patterns during microglial remodeling by oleate and palmitate. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oh, Y.T.; Lee, J.Y.; Lee, J.; Kim, H.; Yoon, K.S.; Choe, W.; Kang, I. Oleic acid reduces lipopolysaccharide-induced expression of iNOS and COX-2 in BV2 murine microglial cells: Possible involvement of reactive oxygen species, p38 MAPK, and IKK/NF-κB signaling pathways. Neurosci. Lett. 2009, 464, 93–97. [Google Scholar] [CrossRef] [PubMed]
- Krycer, J.R.; Brown, A.J. Cross-talk between the androgen receptor and the liver X receptor: Implications for cholesterol homeostasis. J. Biol. Chem. 2011, 286, 20637–20647. [Google Scholar] [CrossRef] [Green Version]
- Balgoma, D.; Zelleroth, S.; Grönbladh, A.; Hallberg, M.; Pettersson, C.; Hedeland, M. Anabolic androgenic steroids exert a selective remodeling of the plasma lipidome that mirrors the decrease of the de novo lipogenesis in the liver. Metabolomics 2020, 16, 12. [Google Scholar] [CrossRef] [Green Version]
- Zhu, P.; Baek, S.H.; Bourk, E.M.; Ohgi, K.A.; Garcia-Bassets, I.; Sanjo, H.; Akira, S.; Kotol, P.F.; Glass, C.K.; Rosenfeld, M.G.; et al. Macrophage/Cancer Cell Interactions Mediate Hormone Resistance by a Nuclear Receptor Derepression Pathway. Cell 2006, 124, 615–629. [Google Scholar] [CrossRef] [Green Version]
- Perucha, E.; Melchiotti, R.; Bibby, J.A.; Wu, W.; Frederiksen, K.S.; Roberts, C.A.; Hall, Z.; LeFriec, G.; Robertson, K.A.; Lavender, P.; et al. The cholesterol biosynthesis pathway regulates IL-10 expression in human Th1 cells. Nat. Commun. 2019, 10, 498. [Google Scholar] [CrossRef] [Green Version]
- Rodrigues, N.V.; Correia, D.V.; Mensurado, S.; Nóbrega-Pereira, S.; DeBarros, A.; Kyle-Cezar, F.; Tutt, A.; Hayday, A.C.; Norell, H.; Silva-Santos, B.; et al. Low-Density Lipoprotein Uptake Inhibits the Activation and Antitumor Functions of Human Vγ9Vδ2 T Cells. Cancer Immunol. Res. 2018, 6, 448–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chaudhary, J.; Bower, J.; Corbin, I.R. Lipoprotein Drug Delivery Vehicles for Cancer: Rationale and Reason. Int. J. Mol. Sci. 2019, 20, 6327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, H.; Lan, J.; Li, C.; Shi, H.; Brosseau, J.P.; Wang, H.; Lu, H.; Fang, C.; Zhang, Y.; Liang, L.; et al. Inhibiting PD-L1 palmitoylation enhances T-cell immune responses against tumours. Nat. Biomed. Eng. 2019, 3, 306–317. [Google Scholar] [CrossRef] [PubMed]
- Sharma, C.; Wang, H.X.; Li, Q.; Knoblich, K.; Reisenbichler, E.S.; Richardson, A.L.; Hemler, M.E. Protein Acyltransferase DHHC3 Regulates Breast Tumor Growth, Oxidative Stress, and Senescence. Cancer Res. 2017, 77, 6880–6890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef] [Green Version]
- Kim, C.H. Immune regulation by microbiome metabolites. Immunology 2018, 154, 220–229. [Google Scholar] [CrossRef]
- Botticelli, A.; Vernocchi, P.; Marini, F.; Quagliariello, A.; Cerbelli, B.; Reddel, S.; Del Chierico, F.; Di Pietro, F.; Giusti, R.; Tomassini, A.; et al. Gut metabolomics profiling of non-small cell lung cancer (NSCLC) patients under immunotherapy treatment. J. Transl. Med. 2020, 18, 49. [Google Scholar] [CrossRef]
- Belcheva, A.; Irrazabal, T.; Robertson, S.J.; Streutker, C.; Maughan, H.; Rubino, S.; Moriyama, E.H.; Copeland, J.K.; Surendra, A.; Kumar, S.; et al. Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells. Cell 2014, 158, 288–299. [Google Scholar] [CrossRef] [Green Version]
- Ruemmele, F.M. Butyrate induced Caco-2 cell apoptosis is mediated via the mitochondrial pathway. Gut 2003, 52, 94–100. [Google Scholar] [CrossRef] [Green Version]
- Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, Receptor for Niacin and the Commensal Metabolite Butyrate, Suppresses Colonic Inflammation and Carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef] [Green Version]
- Park, J.; Kim, M.; Kang, S.G.; Jannasch, A.H.; Cooper, B.; Patterson, J.; Kim, C.H. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR–S6K pathway. Mucosal Immunol. 2015, 8, 80–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bilotta, A.J.; Cong, Y. Gut microbiota metabolite regulation of host defenses at mucosal surfaces: Implication in precision medicine. Precis. Clin. Med. 2019, 2, 110–119. [Google Scholar] [CrossRef]
- Bachem, A.; Makhlouf, C.; Binger, K.J.; De Souza, D.P.; Tull, D.; Hochheiser, K.; Whitney, P.G.; Fernandez-Ruiz, D.; Dähling, S.; Kastenmüller, W.; et al. Microbiota-Derived Short-Chain Fatty Acids Promote the Memory Potential of Antigen-Activated CD8+ T Cells. Immunity 2019, 51, 285–297. [Google Scholar] [CrossRef] [PubMed]
- O’Sullivan, D.; Van Der Windt, G.J.W.; Huang, S.C.C.; Curtis, J.D.; Chang, C.H.; Buck, M.D.; Qiu, J.; Smith, A.M.; Lam, W.Y.; DiPlato, L.M.; et al. Memory CD8+ T Cells Use Cell-Intrinsic Lipolysis to Support the Metabolic Programming Necessary for Development. Immunity 2014, 41, 75–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Der Windt, G.J.W.; 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] [PubMed] [Green Version]
- Nastasi, C.; Candela, M.; Bonefeld, C.M.; Geisler, C.; Hansen, M.; Krejsgaard, T.; Biagi, E.; Andersen, M.H.; Brigidi, P.; Ødum, N.; et al. The effect of short-chain fatty acids on human monocyte-derived dendritic cells. Sci. Rep. 2015, 5, 16148. [Google Scholar] [CrossRef]
- Nastasi, C.; Fredholm, S.; Willerslev-Olsen, A.; Hansen, M.; Bonefeld, C.M.; Geisler, C.; Andersen, M.H.; Ødum, N.; Woetmann, A. Butyrate and propionate inhibit antigen-specific CD8+ T cell activation by suppressing IL-12 production by antigen-presenting cells. Sci. Rep. 2017, 7, 14516. [Google Scholar] [CrossRef]
- Coutzac, C.; Jouniaux, J.-M.; Paci, A.; Schmidt, J.; Mallardo, D.; Seck, A.; Asvatourian, V.; Cassard, L.; Saulnier, P.; Lacroix, L.; et al. Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat. Commun. 2020, 11, 2168. [Google Scholar] [CrossRef]
- Zhang, W.Q.; Zhao, S.K.; Luo, J.W.; Dong, X.P.; Hao, Y.T.; Li, H.; Shan, L.; Zhou, Y.; Shi, H.B.; Zhang, Z.Y.; et al. Alterations of fecal bacterial communities in patients with lung cancer. Am. J. Transl. Res. 2018, 10, 3171–3185. [Google Scholar]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Man Lei, Y.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [Green Version]
- Chalmin, F.; Bruchard, M.; Vegran, F.; Ghiringhelli, F. Regulation of T cell antitumor immune response by tumor induced metabolic stress. Cell Stress 2019, 3, 9–18. [Google Scholar] [CrossRef] [PubMed] [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. 2011, 186, 3299–3303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santaren, I.D.; Bazinet, R.P.; Liu, Z.; Johnston, L.W.; Sievenpiper, J.L.; Giacca, A.; Retnakaran, R.; Harris, S.B.; Zinman, B.; Hanley, A.J. The Distribution of Fatty Acid Biomarkers of Dairy Intake across Serum Lipid Fractions: The Prospective Metabolism and Islet Cell Evaluation (PROMISE) Cohort. Lipids 2019, 54, 617–627. [Google Scholar] [CrossRef] [PubMed]
- Olazarán, J.; Gil-de-Gómez, L.; Rodríguez-Martín, A.; Valentí-Soler, M.; Frades-Payo, B.; Marín-Muñoz, J.; Antúnez, C.; Frank-García, A.; Acedo-Jiménez, C.; Morlán-Gracia, L.; et al. A Blood-Based, 7-Metabolite Signature for the Early Diagnosis of Alzheimer’s Disease. J. Alzheimer’s Dis. 2015, 45, 1157–1173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stuani, L.; Riols, F.; Millard, P.; Sabatier, M.; Batut, A.; Saland, E.; Viars, F.; Tonini, L.; Zaghdoudi, S.; Linares, L.; et al. Stable Isotope Labeling Highlights Enhanced Fatty Acid and Lipid Metabolism in Human Acute Myeloid Leukemia. Int. J. Mol. Sci. 2018, 19, 3325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pabst, T.; Kortz, L.; Fiedler, G.M.; Ceglarek, U.; Idle, J.R.; Beyoğlu, D. The plasma lipidome in acute myeloid leukemia at diagnosis in relation to clinical disease features. BBA Clin. 2017, 7, 105–114. [Google Scholar] [CrossRef]
- Stefanko, A.; Thiede, C.; Ehninger, G.; Simons, K.; Grzybek, M. Lipidomic approach for stratification of acute myeloid leukemia patients. PLoS ONE 2017, 12, e0168781. [Google Scholar] [CrossRef] [Green Version]
- Lavoie, J.M.; Black, P.C.; Eigl, B.J. Predictive Biomarkers for Checkpoint Blockade in Urothelial Cancer: A Systematic Review. J. Urol. 2019, 202, 49–56. [Google Scholar] [CrossRef] [Green Version]
- Wang, B.; Tontonoz, P. Phospholipid Remodeling in Physiology and Disease. Annu. Rev. Physiol. 2019, 81, 165–188. [Google Scholar] [CrossRef]
- Yang, L.; Bai, Y.; Han, X.; Shi, Y.; Liu, H. Plasma Lipidomic Analysis to Identify Novel Biomarkers for Hepatocellular Carcinoma. J. Anal. Test. 2017, 1, 223–232. [Google Scholar] [CrossRef]
- Lee, J.W.; Mok, H.J.; Lee, D.Y.; Park, S.C.; Kim, G.S.; Lee, S.E.; Lee, Y.S.; Kim, K.P.; Kim, H.D. UPLC-QqQ/MS-Based Lipidomics Approach To Characterize Lipid Alterations in Inflammatory Macrophages. J. Proteome Res. 2017, 16, 1460–1469. [Google Scholar] [CrossRef]
- Balgoma, D.; Pettersson, C.; Hedeland, M. Common Fatty Markers in Diseases with Dysregulated Lipogenesis. Trends Endocrinol. Metab. 2019, 30, 283–285. [Google Scholar] [CrossRef] [PubMed]
- Balgoma, D.; Guitton, Y.; Evans, J.J.; Le Bizec, B.; Dervilly-Pinel, G.; Meynier, A. Modeling the fragmentation patterns of triacylglycerides in mass spectrometry allows the quantification of the regioisomers with a minimal number of standards. Anal. Chim. Acta 2019, 1057, 60–69. [Google Scholar] [CrossRef] [PubMed]
- O’Keefe, S.J.D.; Ou, J.; Aufreiter, S.; O’Connor, D.; Sharma, S.; Sepulveda, J.; Fukuwatari, T.; Shibata, K.; Mawhinney, T. Products of the colonic microbiota mediate the effects of diet on colon cancer risk. J. Nutr. 2009, 139, 2044–2048. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.M.; Yu, Y.N.; Wang, J.L.; Lin, Y.W.; Kong, X.; Yang, C.Q.; Yang, L.; Liu, Z.J.; Yuan, Y.Z.; Liu, F.; et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am. J. Clin. Nutr. 2013, 97, 1044–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uribe-Herranz, M.; Rafail, S.; Beghi, S.; Gil-de-Gómez, L.; Verginadis, I.; Bittinger, K.; Pustylnikov, S.; Pierini, S.; Perales-Linares, R.; Blair, I.A.; et al. Gut microbiota modulate dendritic cell antigen presentation and radiotherapy-induced antitumor immune response. J. Clin. Investig. 2020, 130, 466–479. [Google Scholar] [CrossRef]
- Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [Green Version]
- Milone, M.C.; Fish, J.D.; Carpenito, C.; Carroll, R.G.; Binder, G.K.; Teachey, D.; Samanta, M.; Lakhal, M.; Gloss, B.; Danet-Desnoyers, G.; et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol. Ther. 2009, 17, 1453–1464. [Google Scholar] [CrossRef]
- George, P.; Dasyam, N.; Giunti, G.; Mester, B.; Bauer, E.; Andrews, B.; Perera, T.; Ostapowicz, T.; Frampton, C.; Li, P.; et al. Third-generation anti-CD19 chimeric antigen receptor T-cells incorporating a TLR2 domain for relapsed or refractory B-cell lymphoma: A phase I clinical trial protocol (ENABLE). BMJ Open 2020, 10, e034629. [Google Scholar] [CrossRef] [Green Version]
- June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science 2018, 359, 1361–1365. [Google Scholar] [CrossRef] [Green Version]
- Wang, D.; Starr, R.; Chang, W.C.; Aguilar, B.; Alizadeh, D.; Wright, S.L.; Yang, X.; Brito, A.; Sarkissian, A.; Ostberg, J.R.; et al. Chlorotoxin-directed CAR T cells for specific and effective targeting of glioblastoma. Sci. Transl. Med. 2020, 12. [Google Scholar] [CrossRef] [PubMed]
- Schulz, G.; Cheresh, D.A.; Varki, N.M.; Yu, A.; Staffileno, L.K.; Reisfeld, R.A. Detection of ganglioside GD2 in tumor tissues and sera of neuroblastoma patients. Cancer Res. 1984, 44, 5914–5920. [Google Scholar] [PubMed]
- Dobrenkov, K.; Ostrovnaya, I.; Gu, J.; Cheung, I.Y.; Cheung, N.K.V. Oncotargets GD2 and GD3 are highly expressed in sarcomas of children, adolescents, and young adults. Pediatric Blood Cancer 2016, 63, 1780–1785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Richman, S.A.; Nunez-Cruz, S.; Moghimi, B.; Li, L.Z.; Gershenson, Z.T.; Mourelatos, Z.; Barrett, D.M.; Grupp, S.A.; Milone, M.C. High-Affinity GD2-Specific CAR T Cells Induce Fatal Encephalitis in a Preclinical Neuroblastoma Model. Cancer Immunol. Res. 2018, 6, 36–46. [Google Scholar] [CrossRef] [Green Version]
- Tasian, S.K.; Loh, M.L.; Hunger, S.P. Philadelphia chromosome–like acute lymphoblastic leukemia. Blood 2017, 130, 2064–2072. [Google Scholar] [CrossRef]
- Finney, O.C.; Brakke, H.; Rawlings-Rhea, S.; Hicks, R.; Doolittle, D.; Lopez, M.; Futrell, B.; Orentas, R.J.; Li, D.; Gardner, R.; et al. CD19 CAR T cell product and disease attributes predict leukemia remission durability. J. Clin. Investig. 2019, 129, 2123–2132. [Google Scholar] [CrossRef]
- Konjar, Š.; Veldhoen, M. Dynamic Metabolic State of Tissue Resident CD8 T Cells. Front. Immunol. 2019, 10, 1683. [Google Scholar] [CrossRef] [Green Version]
- Kondo, T.; Ando, M.; Nagai, N.; Tomisato, W.; Srirat, T.; Liu, B.; Mise-Omata, S.; Ikeda, M.; Chikuma, S.; Nishimasu, H.; et al. The NOTCH–FOXM1 Axis Plays a Key Role in Mitochondrial Biogenesis in the Induction of Human Stem Cell Memory–like CAR-T Cells. Cancer Res. 2020, 80, 471–483. [Google Scholar] [CrossRef]
- Martinez, M.; Moon, E.K. CAR T Cells for Solid Tumors: New Strategies for Finding, Infiltrating, and Surviving in the Tumor Microenvironment. Front. Immunol. 2019, 10, 128. [Google Scholar] [CrossRef] [Green Version]
- Fucà, G.; Reppel, L.; Landoni, E.; Savoldo, B.; Dotti, G. Enhancing Chimeric Antigen Receptor T-Cell Efficacy in Solid Tumors. Clin. Cancer Res. 2020, 26, 2444–2451. [Google Scholar] [CrossRef]
- Vartak, S.; Robbins, M.E.C.; Spector, A.A. Polyunsaturated fatty acids increase the sensitivity of 36B10 rat astrocytoma cells to radiation-induced cell kill. Lipids 1997, 32, 283–292. [Google Scholar] [CrossRef] [PubMed]
- Das, U.N. Gamma-linolenic acid therapy of human glioma-a review of in vitro, in vivo, and clinical studies. Med. Sci. Monit. 2007, 13, RA119–RA131. [Google Scholar] [PubMed]
- Belavgeni, A.; Bornstein, S.R.; Von Mässenhausen, A.; Tonnus, W.; Stumpf, J.; Meyer, C.; Othmar, E.; Latk, M.; Kanczkowski, W.; Kroiss, M.; et al. Exquisite sensitivity of adrenocortical carcinomas to induction of ferroptosis. Proc. Natl. Acad. Sci. USA 2019, 116, 22269–22274. [Google Scholar] [CrossRef] [PubMed]
- Das, U.N. Can Bioactive Lipids Augment Anti-cancer Action of Immunotherapy and Prevent Cytokine Storm? Arch. Med. Res. 2019, 50, 342–349. [Google Scholar] [CrossRef]
- Van Bruggen, J.A.C.; Martens, A.W.J.; Fraietta, J.A.; Hofland, T.; Tonino, S.H.; Eldering, E.; Levin, M.D.; Siska, P.J.; Endstra, S.; Rathmell, J.C.; et al. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8+ T cells and impede CAR T-cell efficacy. Blood 2019, 134, 44–58. [Google Scholar] [CrossRef]
© 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
Gil-de-Gómez, L.; Balgoma, D.; Montero, O. Lipidomic-Based Advances in Diagnosis and Modulation of Immune Response to Cancer. Metabolites 2020, 10, 332. https://doi.org/10.3390/metabo10080332
Gil-de-Gómez L, Balgoma D, Montero O. Lipidomic-Based Advances in Diagnosis and Modulation of Immune Response to Cancer. Metabolites. 2020; 10(8):332. https://doi.org/10.3390/metabo10080332
Chicago/Turabian StyleGil-de-Gómez, Luis, David Balgoma, and Olimpio Montero. 2020. "Lipidomic-Based Advances in Diagnosis and Modulation of Immune Response to Cancer" Metabolites 10, no. 8: 332. https://doi.org/10.3390/metabo10080332
APA StyleGil-de-Gómez, L., Balgoma, D., & Montero, O. (2020). Lipidomic-Based Advances in Diagnosis and Modulation of Immune Response to Cancer. Metabolites, 10(8), 332. https://doi.org/10.3390/metabo10080332