Metabolomics enables precision medicine: "A White Paper, Community Perspective"

Richard D Beger, Warwick Dunn, Michael A Schmidt, Steven S Gross, Jennifer A Kirwan, Marta Cascante, Lorraine Brennan, David S Wishart, Matej Oresic, Thomas Hankemeier, David I Broadhurst, Andrew N Lane, Karsten Suhre, Gabi Kastenmüller, Susan J Sumner, Ines Thiele, Oliver Fiehn, Rima Kaddurah-Daouk, for “Precision Medicine and Pharmacometabolomics Task Group”-Metabolomics Society Initiative, Richard D Beger, Warwick Dunn, Michael A Schmidt, Steven S Gross, Jennifer A Kirwan, Marta Cascante, Lorraine Brennan, David S Wishart, Matej Oresic, Thomas Hankemeier, David I Broadhurst, Andrew N Lane, Karsten Suhre, Gabi Kastenmüller, Susan J Sumner, Ines Thiele, Oliver Fiehn, Rima Kaddurah-Daouk, for “Precision Medicine and Pharmacometabolomics Task Group”-Metabolomics Society Initiative

Abstract

Introduction background to metabolomics: Metabolomics is the comprehensive study of the metabolome, the repertoire of biochemicals (or small molecules) present in cells, tissues, and body fluids. The study of metabolism at the global or "-omics" level is a rapidly growing field that has the potential to have a profound impact upon medical practice. At the center of metabolomics, is the concept that a person's metabolic state provides a close representation of that individual's overall health status. This metabolic state reflects what has been encoded by the genome, and modified by diet, environmental factors, and the gut microbiome. The metabolic profile provides a quantifiable readout of biochemical state from normal physiology to diverse pathophysiologies in a manner that is often not obvious from gene expression analyses. Today, clinicians capture only a very small part of the information contained in the metabolome, as they routinely measure only a narrow set of blood chemistry analytes to assess health and disease states. Examples include measuring glucose to monitor diabetes, measuring cholesterol and high density lipoprotein/low density lipoprotein ratio to assess cardiovascular health, BUN and creatinine for renal disorders, and measuring a panel of metabolites to diagnose potential inborn errors of metabolism in neonates.

Objectives of white paper—expected treatment outcomes and metabolomics enabling tool for precision medicine: We anticipate that the narrow range of chemical analyses in current use by the medical community today will be replaced in the future by analyses that reveal a far more comprehensive metabolic signature. This signature is expected to describe global biochemical aberrations that reflect patterns of variance in states of wellness, more accurately describe specific diseases and their progression, and greatly aid in differential diagnosis. Such future metabolic signatures will: (1) provide predictive, prognostic, diagnostic, and surrogate markers of diverse disease states; (2) inform on underlying molecular mechanisms of diseases; (3) allow for sub-classification of diseases, and stratification of patients based on metabolic pathways impacted; (4) reveal biomarkers for drug response phenotypes, providing an effective means to predict variation in a subject's response to treatment (pharmacometabolomics); (5) define a metabotype for each specific genotype, offering a functional read-out for genetic variants: (6) provide a means to monitor response and recurrence of diseases, such as cancers: (7) describe the molecular landscape in human performance applications and extreme environments. Importantly, sophisticated metabolomic analytical platforms and informatics tools have recently been developed that make it possible to measure thousands of metabolites in blood, other body fluids, and tissues. Such tools also enable more robust analysis of response to treatment. New insights have been gained about mechanisms of diseases, including neuropsychiatric disorders, cardiovascular disease, cancers, diabetes and a range of pathologies. A series of ground breaking studies supported by National Institute of Health (NIH) through the Pharmacometabolomics Research Network and its partnership with the Pharmacogenomics Research Network illustrate how a patient's metabotype at baseline, prior to treatment, during treatment, and post-treatment, can inform about treatment outcomes and variations in responsiveness to drugs (e.g., statins, antidepressants, antihypertensives and antiplatelet therapies). These studies along with several others also exemplify how metabolomics data can complement and inform genetic data in defining ethnic, sex, and gender basis for variation in responses to treatment, which illustrates how pharmacometabolomics and pharmacogenomics are complementary and powerful tools for precision medicine.

Conclusions key scientific concepts and recommendations for precision medicine: Our metabolomics community believes that inclusion of metabolomics data in precision medicine initiatives is timely and will provide an extremely valuable layer of data that compliments and informs other data obtained by these important initiatives. Our Metabolomics Society, through its "Precision Medicine and Pharmacometabolomics Task Group", with input from our metabolomics community at large, has developed this White Paper where we discuss the value and approaches for including metabolomics data in large precision medicine initiatives. This White Paper offers recommendations for the selection of state of-the-art metabolomics platforms and approaches that offer the widest biochemical coverage, considers critical sample collection and preservation, as well as standardization of measurements, among other important topics. We anticipate that our metabolomics community will have representation in large precision medicine initiatives to provide input with regard to sample acquisition/preservation, selection of optimal omics technologies, and key issues regarding data collection, interpretation, and dissemination. We strongly recommend the collection and biobanking of samples for precision medicine initiatives that will take into consideration needs for large-scale metabolic phenotyping studies.

Keywords: Metabolomics; Metabonomics; Personalized medicine; Pharmacometabolomics; Pharmacometabonomics; Precision medicine.

Conflict of interest statement

The authors declare that they have no conflict of interest Ethical approval This is a review and does not contain any studies with human or animal subjects. Disclaimer for Dr. Beger The views expressed in this paper are solely those of the author, and they do not represent official policy of the U.S. Food and Drug Administration. Funding National Institute of General Medical Sciences (US); Award Numbers: R24 GM078233 and RC2GM092729 “The Pharmacometabolomics Research Network”; National Institute on Aging; Award numbers: 1R01AG046171; RF1AG051550 Grant Recipient: Rima Kaddurah-Daouk. In addition, this work was supported by funding to Phenome Centre Birmingham by the Medical Research Council in the UK (MR/M009157/1).

Figures

Fig. 1
Fig. 1
Metabolomics, a global biochemical approach for disease sub classification and drug response phenotyping
Fig. 2
Fig. 2
Example of a genetically influenced metabotype (GIM). Fatty acid desaturase 1 (FADS1) catalyzes the desaturation of dihomolinolenoyl-CoA to arachidonoyl-CoA (C20:3 to C20:4). Minor allele homozygotes (7.6 % of the population) of the rs174548 variant convert C20:3 to C20:4 poly-unsaturated fatty acids (PUFAs) about 50 % slower than homozygous carriers of the major allele (52.6 % of the population). The FADS locus has been implicated in multiple GWAS with different cancers, Crohn’s disease and cardiovascular disease traits. This example shows how genetic variance in metabolic traits can be linked to complex disorders to provide a functional understanding of the underlying disease mechanism. Figure from Suhre et al.
Fig. 3
Fig. 3
The metabolic trait is an intermediate phenotype that links the genome, lifestyle and environmental factors to the clinical endpoint. The general concept (a) and an example using information from actual genome-wide association studies with metabolic traits (b). The association of a genetic variant is strongest with its closest intermediate phenotype [IP; for example, the association of fatty acid desaturase 1 (FADS1) with its product–substrate pair], while the association with the clinical end point may be hard to detect at a level of genome-wide significance in a GWAS (P = 0.021 for FADS1 with coronary heart disease). The ensemble of all genetic associations with metabolic traits defines our metabolic individuality and thereby our predisposition to disease. Deep metabolic phenotyping of large precision medicine initiatives allows to identify key factors for the development of complex disorders, which can then serve as biomarkers and targets for clinical intervention. Figure reproduced from Suhre and Gieger
Fig. 4
Fig. 4
Precision medicine approach using metabolomics as compared to treatment-failure evidence-based medicine approach in clinical practice. ‘Personalized profile’ based on metabolomics as well as other clinical and lifestyle data will be used to predict the patients’ responses to specific treatments and thus help select the best treatment regimens

References

    1. Abo R, Hebbring S, Ji Y, Zhu H, Zeng ZB, Batzler A, et al. Merging pharmacometabolomics with pharmacogenomics using ‘1000 Genomes’ single-nucleotide polymorphism imputation: Selective serotonin reuptake inhibitor response pharmacogenomics. Pharmacogenetics and Genomics. 2012;22(4):247–253. doi: 10.1097/FPC.0b013e32835001c9.
    1. Aurich, M.K., Fleming, R.M.T., Thiele, I. (2016). A systems approach reveals distinct metabolic strategies among the NCI-60 cancer cell lines (under review).
    1. Aurich MK, Thiele I. Computational modeling of human metabolism and its application to systems biomedicine. Methods in Molecular Biology. 2016;1386:253–281. doi: 10.1007/978-1-4939-3283-2_12.
    1. Backshall A, Sharma R, Clarke SJ, Keun HC. Pharmacometabonomic profiling as a predictor of toxicity in patients with inoperable colorectal cancer treated with capecitabine. Clinical Cancer Research. 2011;17:3019–3028. doi: 10.1158/1078-0432.CCR-10-2474.
    1. Barton RH, Nicholson JK, Elliott P, Holmes E. High-throughput 1H NMR-based metabolic analysis of human serum and urine for large-scale epidemiological studies: Validation study. International Journal of Epidemiology. 2008;37(suppl 1):i31–i40. doi: 10.1093/ije/dym284.
    1. Beger RD, Bhattacharyya S, Yang X, Gill PS, Schnackenberg LK, Sun J, et al. Translational biomarkers of acetaminophen-induced acute liver injury. Archives of Toxicology. 2015;89(9):1497–1522. doi: 10.1007/s00204-015-1519-4.
    1. Bictash M, Ebbels TM, Chan Q, Loo RL, Yap IK, Brown IJ, et al. Opening up the “black box”: Metabolic phenotyping in epidemiology and metabolome-wide association studies. Journal of Clinical Epidemiology. 2010;63(9):970–979. doi: 10.1016/j.jclinepi.2009.10.001.
    1. Biocrates Life Sciences. (2016). Research products: The essence of metabolic phenotyping—accurate metabolic measurement. Biocrates Life Sciences. .
    1. Cacciatore S, Loda M. Innovation in metabolomics to improve personalized healthcare. Annals of the New York Academy of Sciences. 2015;1346(1):57–62. doi: 10.1111/nyas.12775.
    1. Cajka T, Fiehn O. Comprehensive analysis of lipids in biological systems by liquid chromatography-mass spectrometry. Trends in Analytical Chemistry. 2014;61:192–206. doi: 10.1016/j.trac.2014.04.017.
    1. Chen Q, Park H-Q, Goligorsky MS, Chander P, Fischer SM, Gross SS. Untargeted plasma metabolite profiling reveals the broad systemic consequences of xanthine oxidoreductase inactivation in mice. PLoS One. 2012;7(6):e37149. doi: 10.1371/journal.pone.0037149.
    1. Chumpitazi BP, Hollister EB, Cope JL, Luna RA, Versalovic J, Shulman RJ. 164 gut microbiome biomarkers are associated with clinical response to a low FODMAP diet in children with irritable bowel syndrome. Gastroenterology. 2015;148(4, Supplement 1):S-44. doi: 10.1016/S0016-5085(15)30153-0.
    1. . (2014). Testing service for 25-hydroxy vitamin D2 and D3. Pathology Department, City Hospital, Dudley Road, Birmingham. .
    1. Clayton TA, Baker D, Lindon JC, Everrett JR, Nicholson JK. Pharmacometabolomics identification of a significant host-microbiome metabolic interaction affecting human drug metabolism. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(34):14728–14733. doi: 10.1073/pnas.0904489106.
    1. Clayton TA, Lindon JC, Cloarec O, Antti H, Charuel C, Hanton G, et al. Pharmaco-metabonomic phenotyping and personalized drug treatment. Nature. 2006;440(7087):1073–1077. doi: 10.1038/nature04648.
    1. Collins FS, Varmus H. A new initiative on precision medicine. New England Journal of Medicine. 2015;372:793–795. doi: 10.1056/NEJMp1500523.
    1. Cooper-Dehoff RM, Hou W, Weng L, Baillie RA, Beitelshees AL, Gong Y, et al. Is diabetes mellitus-linked amino acid signature associated with β-blocker-induced impaired fasting glucose? Circulation Cardiovascular Genetics. 2014;7(2):199–205. doi: 10.1161/CIRCGENETICS.113.000421.
    1. Dhurandhar NV, Schoeller D, Brown AW, Heymsfield SB, Thomas D, Sørensen TI, Energy Balance Measurement Working Group et al. Energy balance measurement: when something is not better than nothing. International Journal of Obesity (London) 2015;39(7):1109–1113. doi: 10.1038/ijo.2014.199.
    1. Draisma HH, Pool R, Kobl M, Jansen R, Petersen AK, Vaarhorst AA, et al. Genome-wide association study identifies novel genetic variants contributing to variation in blood metabolite levels. Nature Communication. 2015;6:7208. doi: 10.1038/ncomms8208.
    1. Dunn WB, Broadhurst DI, Atherton HJ, Goodacre R, Griffin JL. Systems level studies of mammalian metabolomes: the roles of mass spectrometry and nuclear magnetic resonance spectroscopy. Chemical Society Reviews. 2011;40:387. doi: 10.1039/B906712B.
    1. Dunn WB, Broadhurst D, Ellis DI, Brown M, Halsall A, O’Hagan S, et al. A GC-TOF-MS study of the stability of serum and urine metabolomes during the UK Biobank sample collection and preparation protocols. International Journal of Epidemiology. 2008;37:23–30. doi: 10.1093/ije/dym281.
    1. Dunn WB, Erban A, Ralf JM, Weber DJ, Creek MB, Breitling R, et al. Mass appeal: metabolite identification in mass spectrometry-focused untargeted metabolomics. Metabolomics. 2013;9(suppl):44–66. doi: 10.1007/s11306-012-0434-4.
    1. Dunn WB, Lin W, Broadhurst D, Begley P, Brown M, Zelena E, et al. Molecular phenotyping of a UK population: defining the human serum metabolome. Metabolomics. 2015;11:9–26. doi: 10.1007/s11306-014-0707-1.
    1. Dunn WB, Wilson ID, Nicholls AW, Broadhurst D. The importance of experimental design and QC samples in large-scale and MS-driven untargeted metabolomic studies of humans. Bioanalysis. 2012;4:2249–2264. doi: 10.4155/bio.12.204.
    1. Ellero-Simatos S, Beitelshees AL, Lewis JP, Yerges-Armstrong LM, Georgiades A, Dane A, Network Pharmacometabolomics Research, et al. Oxylipid profile of low-dose aspirin exposure: a pharmacometabolomics study. Journal of the American Heart Association. 2015;4(10):e002203. doi: 10.1161/JAHA.115.002203.
    1. Ellero-Simatos S, Lewis JP, Georgiades A, Yerges-Armstrong LM, Beitelshees AL, Horenstein RB, et al. Pharmacometabolomics reveals that serotonin is implicated in aspirin response variability. CPT: Pharmacometrics & Systems Pharmacology. 2014;3:e125.
    1. Erazo, M.A., Garcia, A., Ruperez, F.J., Barbas, C. (2013). Metabolomics of diet-related diseases. In: A. Cifuentes (Ed.), Foodomics: advanced mass spectrometry in modern food science and nutrition (pp. 429–452). Hoboken: Wiley. doi: 10.1002/9781118537282.ch16.
    1. Everett JR. Pharmacometabolomics in humans: a new tool for personalized medicine. Pharmacogenomics. 2015;16(7):737–754. doi: 10.2217/pgs.15.20.
    1. Everett JR, Loo RL, Pullen FS. Pharmacometabonomics and personalized medicine. Annals of Clinical Biochemistry. 2013;50:523–545. doi: 10.1177/0004563213497929.
    1. Fan TW-M, Lorkiewicz P, Sellers K, Moseley HNB, Higashi RM, Lane AN. Stable isotope-resolved metabolomics and applications to drug development. Pharmacology & Therapeutics. 2012;133:366–391. doi: 10.1016/j.pharmthera.2011.12.007.
    1. Fiehn O. Metabolomics by gas chromatography-mass spectrometry: combined targeted and untargeted profiling. Current Protocols in Molecular Biology. 2016;114:30.4.1–30.4.32. doi: 10.1002/0471142727.mb3004s114.
    1. Garcia Boy R, Henseler J, Mattern R, Skopp G. Determination of morphine and 6-acetylmorphine in blood with use of dried blood spots. Therapeutic Drug Monitoring. 2008;30:733–739. doi: 10.1097/FTD.0b013e31818d9fdb.
    1. Genser D. Food and drug interaction: consequence for the nutrition/health status. Annals of Nutrition & Metabolism. 2008;52:29–32. doi: 10.1159/000115345.
    1. Gibbons H, O’Gorman A, Brennan L. Metabolomics as a tool in nutritional research. Current Opinion in Lipidology. 2015;26(1):30–34. doi: 10.1097/MOL.0000000000000140.
    1. Godzien J, Alonso-Herranz V, Barbas C, Armitage EG. Controlling the quality of metabolomics data: new strategies to get the best out of the QC sample. Metabolomics. 2015;11(3):518–528. doi: 10.1007/s11306-014-0712-4.
    1. Goodacre R. Metabolomics of a superorganism. Journal of Nutrition. 2007;137:259S–266S.
    1. Gooding J, Jensen MV, Dai X, Wenner BR, Lu D, Arumugam R, et al. Adenylosuccinate is an insulin secretagogue derived from glucose-induced purine metabolism. Cell Reports. 2015;13:157–167. doi: 10.1016/j.celrep.2015.08.072.
    1. Gupta M, Neavin D, Liu D, Biernacka J, Hall-Flavin D, Bobo WV, et al. TSPAN5, ERICH3 and selective serotonin reuptake inhibitors in major depressive disorder: pharmacometabolomics-informed pharmacogenomics. Molecular Psychiatry. 2016
    1. Heinken A, Thiele I. Systematic prediction of health-relevant human-microbial co-metabolism through a computational framework. Gut Microbes. 2015;6(2):120–130. doi: 10.1080/19490976.2015.1023494.
    1. Huang Q, Aa J, Jia H, Xin X, Tao C, Liu L, et al. A pharmacometabonomic approach to predicting metabolic phenotypes and pharmacokinetic parameters of atorvastatin in healthy volunteers. Journal of Proteome Research. 2015;14(9):3970–3981. doi: 10.1021/acs.jproteome.5b00440.
    1. Hviid A, Svanstrom H, Frisch M. Antibiotic use and inflammatory bowel diseases in childhood. Gut. 2011;60:49–54. doi: 10.1136/gut.2010.219683.
    1. Institute of Medicine. (2012). Evolution of translational omics: lessons learned and the path forward. In: C. M. Micheel, S. Nass, G. S. Omenn (Eds.), Washington DC: The National Academies Press.
    1. Jansson J, Willing B, Lucio M, Fekete A, Dicksved J, Halfvarson J, et al. Metabolomics reveals metabolic biomarkers of Crohn’s disease. PLoS ONE. 2009;4(7):e6386. doi: 10.1371/journal.pone.0006386.
    1. Ji Y, Hebbring S, Zhu H, Jenkins GD, Biernacka J, Snyder K, et al. Glycine and a glycine dehydrogenase (GLDC) SNP as citalopram/escitalopram response biomarkers in depression: pharmacometabolomics-informed pharmacogenomics. Clinical Pharmacology and Therapeutics. 2011;89(1):97–104. doi: 10.1038/clpt.2010.250.
    1. Johnson CH, Patterson AD, Idle JR, Gonzalez FJ. Xenobiotic metabolomics: major impact on the metabolome. Annual Review of Pharmacology and Toxicology. 2012;52:37–56. doi: 10.1146/annurev-pharmtox-010611-134748.
    1. Jones PM, Bennett MJ. The changing face of newborn screening: diagnosis of inborn errors of metabolism by tandem mass spectrometry. Clinica Chimica Acta International Journal of Clinical Chemistry. 2002;324(1–2):121–128. doi: 10.1016/S0009-8981(02)00238-3.
    1. Kaddurah-Daouk R, Baillie RA, Zhu H, Zeng ZB, Wiest MM, Nguyen UT, et al. Lipidomic analysis of variation in response to simvastatin in the Cholesterol and Pharmacogenetics Study. Metabolomics. 2010;6(2):191–201. doi: 10.1007/s11306-010-0207-x.
    1. Kaddurah-Daouk R, Baillie RA, Zhu H, Zeng ZB, Wiest MM, Nguyen UT, et al. Enteric microbiome metabolites correlate with response to simvastatin treatment. PLoS ONE. 2011;6(10):e25482. doi: 10.1371/journal.pone.0025482.
    1. Kaddurah-Daouk R, Bogdanov MB, Wikoff WR, Zhu H, Boyle SH, Churchill E, et al. Pharmacometabolomic mapping of early biochemical changes induced by sertraline and placebo. Translational Psychiatry. 2013;3:e223. doi: 10.1038/tp.2012.142.
    1. Kaddurah-Daouk R, Boyle SH, Matson W, Sharma S, Matson S, Zhu H, et al. Pretreatment metabotype as a predictor of response to sertraline or placebo in depressed outpatients: A proof of concept. Translational Psychiatry. 2011;1:26. doi: 10.1038/tp.2011.22.
    1. Kaddurah-Daouk R, Kristal BS, Weinshilboum RM. Metabolomics: A global biochemical approach to drug response and disease. Annual Review of Pharmacology and Toxicology. 2008;48:653–683. doi: 10.1146/annurev.pharmtox.48.113006.094715.
    1. Kaddurah-Daouk, R., McEvoy, J., Baillie, R. A., Lee, D., Yao, J. K., Doraiswamy, P. M., et al. (2007). Metabolomic mapping of atypical antipsychotic effects in schizophrenia. Molecular Psychiatry, 12, 934–945. doi:10.1038/sj.mp.4002000.
    1. Kaddurah-Daouk R, Weinshilboum RM, Pharmacometabolomics Research Network Pharmacometabolomics: implications for clinical pharmacology and systems pharmacology. Clinical Pharmacology and Therapeutics. 2014;95(2):154–167. doi: 10.1038/clpt.2013.217.
    1. Kaddurah-Daouk R, Weinshilboum R, Pharmacometabolomics Research Network Metabolomic signatures for drug response phenotypes-pharmacometabolomics enables precision medicine. Clinical Pharmacology and Therapeutics. 2015;98(1):71–75. doi: 10.1002/cpt.134.
    1. Kastenmüller G, Raffler J, Gieger C, Suhre K. Genetics of human metabolism: An update. Human Molecular Genetics. 2015;24(R1):R93–R101. doi: 10.1093/hmg/ddv263.
    1. Kim S, Kim J, Yun EJ, Kim KH. Food metabolomics: from farm to human. Current Opinion in Biotechnology. 2016;37:16–23. doi: 10.1016/j.copbio.2015.09.004.
    1. Koulman A, Prentice P, Wong MC, Matthews L, Bond NJ, Eiden M, et al. The development and validation of a fast and robust dried blood spot based lipid profiling method to study infant metabolism. Metabolomics. 2014;10:1018–1025. doi: 10.1007/s11306-014-0628-z.
    1. Krauss RM, Zhu H, Kaddurah-Daouk R. Pharmacometabolomics of statin response. Clinical Pharmacology and Therapeutics. 2013;94(5):562–565. doi: 10.1038/clpt.2013.164.
    1. Lewis JP, Yerges-Armstrong LM, Ellero-Simatos S, Georgiades A, Kaddurah-Daouk R, Hankemeier T. Integration of pharmacometabolomic and pharmacogenomic approaches reveals novel insights into antiplatelet therapy. Clinical Pharmacology and Therapeutics. 2013;94(5):570–573. doi: 10.1038/clpt.2013.153.
    1. Lindon JC, Nicholson JK. The emergent role of metabolic phenotyping in dynamic patient stratification. Expert Opinion on Drug Metabolism and Toxicology. 2014;10(7):915–919. doi: 10.1517/17425255.2014.922954.
    1. Mente A, Chalcraft K, Ak H, Davis AD, Lonn E, Miller R, et al. The relationship between trimethylamine-N-oxide and prevalent cardiovascular disease in a multiethnic population living in Canada. The Canadian Journal of Cardiology. 2015;31(9):1189–1194. doi: 10.1016/j.cjca.2015.06.016.
    1. Metabolomics Society. (2014). Metabolite identification task group. Metabolomics Society. .
    1. Miolo G, Muraro E, Caruso D, Crivellari D, Ash A, Scalone S, et al. Phamacometabolomics study identifies circulating spermidine and tryptophan as potential biomarkers associated with the complete pathological response to trastuzumab-paclitaxel neoadjuvant therapy in HER-2 positive breast cancer. Oncotarget. 2016
    1. Moco S, Martin FPJ, Rezzi S. Metabolomics view on gut microbiome modulation by polyphenol-rich foods. Journal of Proteome Research. 2012;11(10):4781–4790. doi: 10.1021/pr300581s.
    1. Nicholson JK, Everett JR, Lindon JC. Longitudinal pharmacometabonomics for predicting patient responses to therapy: drug metabolism, toxicity and efficacy. Expert Opinion on Drug Metabolism and Toxicology. 2012;8(2):135–139. doi: 10.1517/17425255.2012.646987.
    1. Nicholson JK, Holmes E, Lindon JC, Wilson ID. The challenges of modeling mammalian biocomplexity. Nature Biotechnology. 2004;22:1268–1274. doi: 10.1038/nbt1015.
    1. Nicholson JK, Holmes E, Wilson ID. Gut microorganisms, mammalian metabolism, and personalized health care. Nature Reviews Microbiology. 2005;3:431–438. doi: 10.1038/nrmicro1152.
    1. Odriozola L, Corrales FJ. Discovery of nutritional biomarker: future directions based on omics technologies. International Journal of Food Sciences and Nutrition. 2015;66:S31–S40. doi: 10.3109/09637486.2015.1038224.
    1. Patel, C. J., Manrai, A. K. (2015). Development of exposome correlation globes to map out environment-wide associations. Pacific Symposium on Biocomputing.
    1. Patti GJ, Yanes O, Siuzdak G. Innovation: metabolomics: the apogee of the omics trilogy. Nature Reviews Molecular Cell Biology. 2012;13:263–269. doi: 10.1038/nrm3314.
    1. Peterson J, Dwyer J, Adlercreutz H, Scalbert A, Jacques P, McCullough ML. Dietary lignans: physiology and potential for cardiovascular disease risk reduction. Nutrition Reviews. 2010;68(10):571–603. doi: 10.1111/j.1753-4887.2010.00319.x.
    1. Purnbaugh PJ, Gordon JI. An invitation to the marriage of metagenomics and metabolomics. Cell. 2008;134(5):708–713. doi: 10.1016/j.cell.2008.08.025.
    1. Rotroff DM, Oki NO, Liang X, Yee SW, Stocker SL, Corum DG, et al. Pharmacometabolomic assessment of metformin in non-diabetic, African Americans. Frontiers in Pharmacology. 2016;7:135. doi: 10.3389/fphar.2016.00135.
    1. Rotroff DM, Shahin MH, Gurley SB, Zhu H, Motsinger-Reif A, Meisner M, et al. Pharmacometabolomic assessments of atenolol and hydrochlorothiazide treatment reveal novel drug response phenotypes. CPT: Pharmacometrics & Systems Pharmacology. 2015;4(11):669–679.
    1. Sanseau P, Agarwal P, Barnes MR, Pastinen T, Richards JB, Cardon LR, et al. Use of genome-wide association studies for drug repositioning. Nature Biotechnology. 2012;30:317–320. doi: 10.1038/nbt.2151.
    1. Scalbert A, Brennan L, Fiehn O, Hankemeier T, Kristal BS, van Ommen B, et al. Mass-spectrometry-based metabolomics: limitations and recommendations for future progress with particular focus on nutrition research. Metabolomics. 2009;5:435–458. doi: 10.1007/s11306-009-0168-0.
    1. Schmedes M, Aadland EK, Sundekilde UK, Jacques H, Lavigne C, Graff IE, et al. Lean-seafood intake decreases urinary markers of mitochondrial lipid and energy metabolism in healthy subjects: metabolomics results from a randomized crossover intervention study. Molecular Nutrition & Food Research. 2016;60(7):1661–1672. doi: 10.1002/mnfr.201500785.
    1. Schmidt MA, Goodwin TJ. Personalized medicine in human space flight: using omics based analyses to develop individualized countermeasures that enhance astronaut safety and performance. Metabolomics. 2013;9(6):1134–1156. doi: 10.1007/s11306-013-0556-3.
    1. Schulze A, Lindner M, Kohlmüller D, Olgemöller K, Mayatepek E, Hoffmann GF. Expanded newborn screening for inborn errors of metabolism by electrospray ionization-tandem mass spectrometry: results, outcome, and implications. Pediatrics. 2003;111(6 pt 1):1399–1406. doi: 10.1542/peds.111.6.1399.
    1. Shahin, M.H., Gong, Y., McDonough, C.W., Rotroff, D.M., Beitelshees, A.L., Garrett, T.J., et al. (2016). A genetic response score for hydrochlorothiazide use: insights from genomics and metabolomics integration. Hypertension. pii: HYPERTENSIONAHA.116.07328. [Epub ahead of print].
    1. Shin SY, Fauman EB, Petersen AK, Krumsiek J, Santos R, Huang J, et al. An atlas of genetic influences on human blood metabolites. Nature Genetics. 2014;46:543–550. doi: 10.1038/ng.2982.
    1. Stebbing J, Sharma A, North B, Athersuch TJ, Zebrowski A, Pchejetski D, et al. A metabolic phenotyping approach to understanding relationships between metabolic syndrome and breast tumour responses to chemotherapy. Annals of Oncology. 2012;23:860–866. doi: 10.1093/annonc/mdr347.
    1. Su LJ, Fiehn O, Maruvada P, Moore SC, O’Keefe SJ, Wishart DS, et al. The use of metabolomics in population-based research. Advances in Nutrition. 2014;5(6):785–788. doi: 10.3945/an.114.006494.
    1. Suhre K, Gieger C. Genetic variation in metabolic phenotypes: study designs and applications. Nature Reviews Genetics. 2012;13:759–769. doi: 10.1038/nrg3314.
    1. Suhre K, Raffler J, Kastenmüller G. Biochemical insights from population studies with genetics and metabolomics. Archives of Biochemistry and Biophysics. 2016;589:168–176. doi: 10.1016/j.abb.2015.09.023.
    1. Suhre K, Shin SY, Petersen AK, Mohney RP, Meredith D, Wägele B, et al. Human metabolic individuality in biomedical and pharmaceutical research. Nature. 2011;477:54–60. doi: 10.1038/nature10354.
    1. Suhre K, Wallaschofski H, Raffler J, Friedrich N, Haring R, Michael K, et al. A genome-wide association study of metabolic traits in human urine. Nature Genetics. 2011;43:565–569. doi: 10.1038/ng.837.
    1. Tannahill GM. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496:238–242. doi: 10.1038/nature11986.
    1. Thiele I, Swainston N, Fleming RMT, Hoppe A, Sahoo S, Aurich MK, et al. A community-driven global reconstruction of human metabolism. Nature Biotechnology. 2013;31(5):419–425. doi: 10.1038/nbt.2488.
    1. Trupp M, Zhu H, Wikoff WR, Baillie RA, Zeng ZB, Karp PD, et al. Metabolomics reveals amino acids contribute to variation in response to simvastatin treatment. PLoS ONE. 2012;7(7):e38386. doi: 10.1371/journal.pone.0038386.
    1. Vernon HJ. Inborn errors of metabolism: advances in diagnosis and therapy. Journal of the American Medical Association Pediatrics. 2015;169(8):778–782.
    1. Walter-Sack I, Klotz U. Influence of diet and nutritional status of drug metabolism. Clin-Pharmacokinet. 1996;31:47. doi: 10.2165/00003088-199631010-00004.
    1. Wikoff WR, Frye RF, Zhu H, Gong Y, Boyle S, Churchill E, et al. Pharmacometabolomics reveals racial differences in response to atenolol treatment. PLoS ONE. 2013;8(3):e57639. doi: 10.1371/journal.pone.0057639.
    1. Wilson ID. Drugs, bugs, and personalized medicine: Pharmacometabonomics enters the ring. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:14187–14188. doi: 10.1073/pnas.0907721106.
    1. Winnike JH, Li Z, Wright FA, Macdonald JM, O’Connell TM, Watkins PB. Use of pharmaco-metabonomics for early prediction of acetaminophen-induced hepatotoxicity in humans. Clinical Pharmacology and Therapeutics. 2010;88:45–51. doi: 10.1038/clpt.2009.240.
    1. Xia J, Broadhurst DI, Wilson M, Wishart DS. Translational biomarker discovery in clinical metabolomics: an introductory tutorial. Metabolomics. 2013;9(2):280–299. doi: 10.1007/s11306-012-0482-9.
    1. Xie G, Jia W. Metabonomics in translational research for personalized medicine and nutrition. In: Kochhar S, Martin F-P, editors. Metabonomics and Gut Microbiota in Nutrition and Disease. New York: Springer; 2015. pp. 63–82.
    1. Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161:264–276. doi: 10.1016/j.cell.2015.02.047.
    1. Yerges-Armstrong LM, Ellero-Simatos S, Georgiades A, Zhu H, Lewis JP, Horenstein RB, et al. Purine pathway implicated in mechanism of resistance to aspirin therapy: pharmacometabolomics-informed pharmacogenomics. Clinical Pharmacology and Therapeutics. 2013;94(4):525–532. doi: 10.1038/clpt.2013.119.
    1. Yizhak K, Gaude E, Le Dévédec S, Waldman YY, Stein GY, van de Water B, et al. Phenotype-based cell-specific metabolic modeling reveals metabolic liabilities of cancer. eLife. 2014;3:e03641. doi: 10.7554/eLife.03641.
    1. Yoon HR. Screening newborns for metabolic disorders based on targeted metabolomics using tandem mass spectrometry. Annals of Pediatric Endocrinology & Metabolism. 2015;20(3):119–124. doi: 10.6065/apem.2015.20.3.119.
    1. Zamboni N, Saghatelian A, Patti GJ. Defining the metabolome: size, flux, and regulation. Molecular Cell. 2015;58(4):699–706. doi: 10.1016/j.molcel.2015.04.021.
    1. Zheng H, Clausen MR, Dalsgaard TK, Bertram HC. Metabolomics to explore impact of dairy intake. Nutrients. 2015;7(6):4875–4896. doi: 10.3390/nu7064875.

Source: PubMed

3
購読する