Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

A proposed framework to evaluate the quality and reliability of targeted metabolomics assays from the UK Consortium on Metabolic Phenotyping (MAP/UK)

Abstract

Targeted metabolite assays that measure tens or hundreds of pre-selected metabolites, typically using liquid chromatography–mass spectrometry, are increasingly being developed and applied to metabolic phenotyping studies. These are used both as standalone phenotyping methods and for the validation of putative metabolic biomarkers obtained from untargeted metabolomics studies. However, there are no widely accepted standards in the scientific community for ensuring reliability of the development and validation of targeted metabolite assays (referred to here as ‘targeted metabolomics’). Most current practices attempt to adopt, with modifications, the strict guidance provided by drug regulatory authorities for analytical methods designed largely for measuring drugs and other xenobiotic analytes. Here, the regulatory guidance provided by the European Medicines Agency, US Food and Drug Administration and International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use are summarized. In this Perspective, we have adapted these guidelines and propose a less onerous ‘tiered’ approach to evaluate the reliability of a wide range of metabolomics analyses, addressing the need for community-accepted, harmonized guidelines for tiers other than full validation. This ‘fit-for-purpose’ tiered approach comprises four levels—discovery, screening, qualification and validation—and is discussed in the context of a range of targeted and untargeted metabolomics assays. Issues arising with targeted multiplexed metabolomics assays, and how these might be addressed, are considered. Furthermore, guidance is provided to assist the community with selecting the appropriate degree of reliability for a series of well-defined applications of metabolomics.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Validation workflow steps and positioning of the suggested tiered framework.

Similar content being viewed by others

References

  1. Roberts, L. D., Souza, A. L., Gerszten, R. E. & Clish, C. B. Targeted metabolomics. Curr. Protoc. Mol. Biol. 98, 30.2.1–30.2.24 (2012).

    Article  Google Scholar 

  2. Goodacre, R., Vaidyanathan, S., Dunn, W. B., Harrigan, G. G. & Kell, D. B. Metabolomics by numbers: acquiring and understanding global metabolite data. Trends Biotechnol. 22, 245–252 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Wei, R., Guodong, L. & Seymour, A. B. High-throughput and multiplexed LC/MS/MRM method for targeted metabolomics. Anal. Chem. 82, 5527–5533 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Dunn, W. B. et al. Molecular phenotyping of a UK population: defining the human serum metabolome. Metabolomics 11, 9–26 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Guijas, C., Montenegro-Burke, J. R., Warth, B., Spilker, M. E. & Siuzdak, G. Metabolomics activity screening for identifying metabolites that modulate phenotype. Nat. Biotechnol. 36, 316–320 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rinschen, M. M., Ivanisevic, J., Giera, M. & Siuzdak, G. Identification of bioactive metabolites using activity metabolomics. Nat. Rev. Mol. Cell Biol. 20, 353–367 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Viant, M. R. et al. Use cases, best practice and reporting standards for metabolomics in regulatory toxicology. Nat. Commun. 10, 3041 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Gertsman, I. & Barshop, B. A. Promises and pitfalls of untargeted metabolomics. J. Inherit. Metab. Dis. 41, 355–366 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rattray, N. J. W. et al. Metabolic dysregulation in vitamin E and carnitine shuttle energy mechanisms associate with human frailty. Nat. Commun. 10, 5027 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ribbenstedt, A., Ziarrusta, H. & Benskin, J. P. Development, characterization and comparisons of targeted and non-targeted metabolomics methods. PLoS One 13, e0207082 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Want, E. J., Cravatt, B. F. & Siuzdak, G. The expanding role of mass spectrometry in metabolite profiling and characterization. Chem. BioChem. 6, 1941–1951 (2005).

    CAS  Google Scholar 

  12. Dunn, W. B. et al. Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and liquid chromatography coupled to mass spectrometry. Nat. Protoc. 6, 1060–1083 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Goodman, J. et al. Update to the European Bioanalysis Forum recommendation on biomarkers assays; bringing context of use into practice. Bioanalysis 12, 1427–1437 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Tabassum, R. et al. Genetic architecture of human plasma lipidome and its link to cardiovascular disease. Nat. Commun. 10, 4329 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Goutman, S. A. et al. Untargeted metabolomics yields insight into ALS disease mechanisms. J. Neurol. Neurosurg. Psychiatry 91, 1329–1338 (2020).

    Article  PubMed  Google Scholar 

  16. Palmer, J. A. et al. A targeted metabolomics-based assay using human induced pluripotent stem cell-derived cardiomyocytes identifies structural and functional cardiotoxicity potential. Toxicol. Sci. 174, 218–240 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Michel, F., Pirotte, B., Fillet, M. & Tullio, P. Metabolomics as a challenging approach for medicinal chemistry and personalized medicine. J. Med. Chem. 59, 8649–8666 (2016).

    Article  Google Scholar 

  18. Beger, R. D. et al. Metabolomics enables precision medicine: “A White Paper, Community Perspective. Metabolomics 12, 149 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Mandal, R., Chamot, D. & Wishart, D. S. The role of the human metabolome database in inborn errors of metabolism. J. Inherit. Metab. Dis. 41, 329–336 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Burla, B. et al. MS-based lipidomics of human blood plasma: a community-initiated position paper to develop accepted guidelines. J. Lipid Res. 59, 2001–2017 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Thompson, J. W. et al. International ring trial of a high resolution targeted metabolomics and lipidomics platform for serum and plasma analysis. Anal. Chem. 91, 14407–14416 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. König, R., Cave, A., Goldammer, M. & Meulendijks, D. Bioanalytical Omics Subgroup Report (Heads of Medicines Agencies (HMA) and European Medicines Agency (EMA)); https://www.ema.europa.eu/en/documents/report/bioanalytical-omics-subgroup-report_en.pdf

  23. Harrill, J. A. et al. Progress towards an OECD reporting framework for transcriptomics and metabolomics in regulatory toxicology. Regul. Toxicol. Pharmacol. 125, 105020 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Dunn, W. B., Broadhurst, D. I., Atherton, H. J., Goodacre, R. & Griffin, J. L. Systems level studies of mammalian metabolomes: the Roles of mass spectrometry and nuclear magnetic resonance spectroscopy. Chem. Soc. Rev. 40, 387–426 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Dunn, W. B. et al. Quality assurance and quality control processes: summary of a metabolomics community questionnaire. Metabolomics 13, 50 (2017).

    Article  Google Scholar 

  26. Broadhurst, D. et al. Guidelines and considerations for the use of system suitability and quality control samples in mass spectrometry assays applied in untargeted clinical metabolomic studies. Metabolomics 14, 72 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Hyötyläinen, T., Ahonen, L., Pöhö, P. & Orešič, M. Lipidomics in biomedical research-practical considerations. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 1862, 800–803 (2017).

    Article  PubMed  Google Scholar 

  28. Beger, R. D. et al. Towards quality assurance and quality control in untargeted metabolomics studies. Metabolomics 15, 4 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Theodorsson, E. Validation and verification of measurement methods in clinical chemistry. Bioanalysis 4, 305–320 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. Bioanalytical Method Validation, Guidance for Industry (US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), and Center for Veterinary Medicine (CVM), 2018); https://www.fda.gov/media/70858/download

  31. Guideline on Bioanalytical Method Validation (European Medicines Agency, 2011); https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-bioanalytical-method-validation_en.pdf

  32. ICH Guideline M10 on Bioanalytical Method Validation (European Medicines Agency, 2019); https://www.ema.europa.eu/en/documents/scientific-guideline/draft-ich-guideline-m10-bioanalytical-method-validation-step-2b_en.pdf

  33. Draft Guideline on Bioanalytical Method Validation in Pharmaceutical Development (Ministry of Health and Welfare, Japan, 2013); https://www.nihs.go.jp/drug/BMV/BMV_draft_130415_E.pdf

  34. Good Manufacturing Practice for Drugs (2010 Revision) MOH Decree No. 79. (China Food and Drug Administration (CFDA), 2010); https://www.gmpsop.com/RegulatoryReference/CFDA/China_SFDA_Good_Manufacturing_Practice_for_Drugs.pdf

  35. Finished Product (Medicine) Analytical Procedure Validations for Complementary Medicines, March 2006 (Austrailian Government, Therapeutic Goods Administration, 2006); https://www.tga.gov.au/sites/default/files/cm-analytical-procedure-finished.pdf

  36. ANVISA Guide for Validation of Analytical and Bioanalytical Methods Resolution RE no.899 (Brazilian Sanitary Surveillance Agency, 2012).

  37. Huynh-Ba, K. & Beumer Sassi, A. Anvisa: an introduction to a new regulatory agency with many challenges. AAPS Open 4, 9 (2018).

    Article  Google Scholar 

  38. Essential Considerations for Successful Qualification of Novel Methodologies (EMA, 2017); https://www.ema.europa.eu/en/documents/other/essential-considerations-successful-qualification-novel-methodologies_en.pdf

  39. Points to Consider Document: Scientific and Regulatory Considerations for the Analytical Validation of Assays Used in the Qualification of Biomarkers in Biological Matrices (Critical Path Institute, 2019); https://c-path.org/wp-content/uploads/2019/06/evidconsid-whitepaper-analyticalsection2019.pdf

  40. Viswanathan, C. T. et al. Quantitative bioanalytical methods validation and implementation: best practices for chromatographic and ligand binding assays. Pharm. Res. 24, 1962–1973 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Timmerman, P. et al. Best practices in a tiered approach to metabolite quantification: views and recommendations of the European Bioanalysis Forum. Bioanalysis 2, 1185–1194 (2010).

    Article  PubMed  Google Scholar 

  42. Lowes, S. et al. Tiered approaches to chromatographic bioanalytical method performance evaluation: recommendation for best practices and harmonization from the global bioanalysis consortium harmonization team. AAPS J. 17, 17–23 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Nakamura, T. 7th Japan Bioanalysis Forum symposium: regulated bioanalysis, to a new stage. Bioanalysis 8, 2097–2102 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Kadian, N. et al. Comparative assessment of bioanalytical method validation guidelines for pharmaceutical industry. J. Pharm. Biomed. Anal. 126, 83–97 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. Booth, B. et al. Workshop report: Crystal City V—quantitative bioanalytical method validation and implementation: the 2013 Revised FDA Guidance. AAPS J. 17, 277–288 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Global Bioanalysis Consortium on Harmonisation of Bioanalytical Guidance (GBC, 2020); https://sites.google.com/site/globalbioanalysisconsortium/

  47. Timmerman, P. et al. Tiered approach into practice: scientific validation for chromatography-based assays in early development—a recommendation from the European Bioanalysis Forum. Bioanalysis 7, 2387–2398 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Lavezzari, G. & Womack, A. W. Industry perspectives on biomarker qualification. Clin. Pharmacol. Ther. 99, 208–213 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Shah, V. P. et al. Bioanalytical method validation—a revisit with a decade of progress. Pharm. Res. 17, 1551–1557 (2004).

    Article  Google Scholar 

  50. Safavi A. Exploratory Biomarker Testing—to Qualify or Validate the Assay? (BioAgilityx, 2021); https://www.bioagilytix.com/blog/2019/08/28/webinar-sneak-peek-exploratory-biomarker-testing-to-qualify-or-validate-the-assay/

  51. Lewis, M. et al. An open platform for large scale LC-MS-based metabolomics. Preprint at ChemRxiv https://doi.org/10.26434/chemrxiv-2022-nq9k0 (2022).

  52. Uwe, C. et al. Mass spectrometry-based multiplexing for the analysis of biomarkers in drug development and clinical diagnostics—how much is too much? Microchem. J. 105, 32–38 (2012).

    Article  Google Scholar 

  53. Thway, T. M. et al. Assessment of incurred sample reanalysis for macromolecules to evaluate bioanalytical method robustness: effects from imprecision. AAPS J. 13, 291–298 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Van de Merbel, N. C. Quantitative determination of endogenous compounds in biological samples using chromatographic techniques. Trends Anal. Chem. 27, 924–933 (2008).

    Article  Google Scholar 

  55. Thakare, R. et al. Quantitative analysis of endogenous compounds. J. Pharm. Biomed. Anal. 128, 426–437 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Tsikas, D. Bioanalytical method validation of endogenous substances according to guidelines by the FDA and other organizations: basic need to specify concentration ranges. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 1093–1094, 80–81 (2018).

    Article  Google Scholar 

  57. Wakamatsu, A. et al. Proposed selection strategy of surrogate matrix to quantify endogenous substances by Japan Bioanalysis Forum DG2015-15. Bioanalysis 10, 1349–1360 (2018).

    Article  CAS  PubMed  Google Scholar 

  58. ICH Guideline Q2(R2) on Validation of Analytical Procedures EMA/CHMP/ICH/82072/2006 (European Medicines Agency, 2022); https://www.ema.europa.eu/en/ich-q2-r1-validation-analytical-procedures-text-methodology

  59. Analytical Method Validation (WHO, 2018); https://pharmaguidances.com/analytical-method-validation-as-per-who-technical-report-series-no-937/

  60. Araujo, P. Key aspects of analytical method validation and linearity evaluation. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 877, 2224–2234 (2009).

    Article  CAS  Google Scholar 

  61. Boque, R. & Heyden, Y. V. The limit of detection. LC GC Eur. 22, 82–85 (2009).

    CAS  Google Scholar 

  62. Desimoni, E. & Brunetti, B. Signal to noise ratio; limit of detection; standard error of the regression; chromatographic/voltammetric/spectroscopic signals. Pharm. Anal. Acta. 6, 355 (2015).

    Google Scholar 

  63. U.S. Pharmacopeia. Document 1225, validation of compendial methods. Pharmacoepial Forum 31, 549 (2005).

    Google Scholar 

  64. Analytical Procedures and Methods Validation for Drugs and Biologics: Guidance for Industry (Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER), 2015); https://www.fda.gov/media/87801/download

  65. Robouch, P., Stroka, J., Haedrich, J., Schaechtele, A., & Wenzl, T. Guidance Document on the Estimation of LOD and LOQ for Measurements in the Field of Contaminants in Feed and Food (European Commission, Centre Joint Research, 2016).

  66. 2002/657/EC: Commission Decision of 12 August 2002 Implementing Council Directive 96/23/EC Concerning the Performance of Analytical Methods and the Interpretation of Results (Text with EEA Relevance) (Notified under Document Number C (2002) 3044) (European Commission, 2002); https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2002:221:0008:0036:EN:PDF

  67. Watson, D. G. Pharmaceutical Analysis 5th edn (Elsevier, 2020).

  68. Bliesner, D. M. Validating Chromatographic Methods: A Practical Guide (Wiley Interscience, 2006).

  69. Lee, J. W. et al. Fit-for-purpose method development and validation for successful biomarker measurement. Pharm. Res. 23, 312–328 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Simón-Manso, Y. et al. Metabolite profiling of a NIST Standard Reference Material for human plasma (SRM 1950): GC-MS, LC-MS, NMR, and clinical laboratory analyses, libraries, and web-based resources. Anal. Chem. 85, 11725–11731 (2013).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Funding was provided by the Medical Research Council (grant reference: MR/S010483/1) to the MAP/UK project. Wellcome Trust provided support for LIPID MAPS (203014/Z/16/Z) to V.B.O’D.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

The content of the present manuscript was discussed by MAP/UK consortium members and then written up by S.S. and T.S. under the latter’s supervision and guidance. Scientific input and critical review were provided by M.R.L., I.D.W., M.R.V., R.G., W.B.D, J.L.G., V.B.O’D., K.E.C. and B.N.

Corresponding author

Correspondence to Toru Suzuki.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Jianguo Xia and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Thompson, J. W. Anal. Chem. 91, 14407–14416 (2019): https://doi.org/10.1021/acs.analchem.9b02908

Lewis, M. et al. Preprint at ChemRxiv https://doi.org/10.26434/chemrxiv-2022-nq9k0 (2022).

Israr, M. Z. Am. Heart. J. 234, 71–80 (2021): https://doi.org/10.1016/j.ahj.2021.01.006

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2

Supplementary Note

Core members of the MAP/UK consortium

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sarmad, S., Viant, M.R., Dunn, W.B. et al. A proposed framework to evaluate the quality and reliability of targeted metabolomics assays from the UK Consortium on Metabolic Phenotyping (MAP/UK). Nat Protoc 18, 1017–1027 (2023). https://doi.org/10.1038/s41596-022-00801-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-022-00801-8

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research