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.

  • Protocol
  • Published:

Stable isotope tracing to assess tumor metabolism in vivo

Abstract

Cancer cells undergo diverse metabolic adaptations to meet the energetic demands imposed by dysregulated growth and proliferation. Assessing metabolism in intact tumors allows the investigator to observe the combined metabolic effects of numerous cancer cell-intrinsic and -extrinsic factors that cannot be fully captured in culture models. We have developed methods to use stable isotope-labeled nutrients (e.g., [13C]glucose) to probe metabolic activity within intact tumors in vivo, in mice and humans. In these methods, the labeled nutrient is introduced to the circulation through an intravenous catheter prior to surgical resection of the tumor and adjacent nonmalignant tissue. Metabolism within these tissues during the infusion transfers the isotope label into metabolic intermediates from pathways supplied by the infused nutrient. Extracting metabolites from surgical specimens and analyzing their isotope labeling patterns provides information about metabolism in the tissue. We provide detailed information about this technique, from introduction of the labeled tracer through data analysis and interpretation, including streamlined approaches to quantify isotope labeling in informative metabolites extracted from tissue samples. We focus on infusions with [13C]glucose and the application of mass spectrometry to assess isotope labeling in intermediates from central metabolic pathways, including glycolysis, the tricarboxylic acid cycle and nonessential amino acid synthesis. We outline practical considerations to apply these methods to human subjects undergoing surgical resections of solid tumors. We also discuss the method’s versatility and consider the relative advantages and limitations of alternative approaches to introduce the tracer, harvest the tissue and analyze the data.

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: Patient infusion workflow.
Fig. 2: Mouse infusion workflow.
Fig. 3: Mouse infusion apparatus.
Fig. 4: Confirmation of [13C] peaks in HRMS.
Fig. 5: Data from [U-13C]glucose infusions in patients and mice.
Fig. 6: Examining metabolic features among tumor and tissue types.

Similar content being viewed by others

Data availability

Previously unpublished data are included in the Supplementary Information. The remaining supporting data can be found in refs. 4,5,21.

Code availability

The software can be found at https://github.com/wencgu/nac.

References

  1. Faubert, B. & DeBerardinis, R. J. Analyzing tumor metabolism in vivo. Annu. Rev. Cancer Biol. 1, 99–117 (2016).

    Article  Google Scholar 

  2. Buescher, J. M. et al. A roadmap for interpreting (13)C metabolite labeling patterns from cells. Curr. Opin. Biotechnol. 34, 189–201 (2015).

    Article  CAS  Google Scholar 

  3. Maher, E. A. et al. Metabolism of [U-13 C]glucose in human brain tumors in vivo. NMR Biomed. 25, 1234–1244 (2012).

    Article  CAS  Google Scholar 

  4. Hensley, C. T. et al. Metabolic heterogeneity in human lung tumors. Cell 164, 681–694 (2016).

    Article  CAS  Google Scholar 

  5. Faubert, B. et al. Lactate metabolism in human lung tumors. Cell 171, 358–371 e359 (2017).

    Article  CAS  Google Scholar 

  6. Courtney, K. D. et al. Isotope tracing of human clear cell renal cell carcinomas demonstrates suppressed glucose oxidation in vivo. Cell Metab. 28, 793–800 (2018).

    Article  CAS  Google Scholar 

  7. Johnston, K. et al. Isotope tracing reveals glycolysis and oxidative metabolism in childhood tumors of multiple histologies. Med 2, 395–410 (2021).

    Article  Google Scholar 

  8. Oizel, K. et al. Glutamine uptake and utilization of human mesenchymal glioblastoma in orthotopic mouse model. Cancer Metab. 8, 9 (2020).

    Article  Google Scholar 

  9. Momcilovic, M. et al. The GSK3 signaling axis regulates adaptive glutamine metabolism in lung squamous cell carcinoma. Cancer Cell 33, 905–921 e905 (2018).

    Article  CAS  Google Scholar 

  10. Davidson, S. M. et al. Environment impacts the metabolic dependencies of Ras-driven non-small cell lung cancer. Cell Metab. 23, 517–528 (2016).

    Article  CAS  Google Scholar 

  11. Shi, X. et al. The abundance of metabolites related to protein methylation correlates with the metastatic capacity of human melanoma xenografts. Sci. Adv. 3, eaao5268 (2017).

    Article  Google Scholar 

  12. Grinde, M. T. et al. Glutamine to proline conversion is associated with response to glutaminase inhibition in breast cancer. Breast Cancer Res. 21, 61 (2019).

    Article  Google Scholar 

  13. Fan, T. W. et al. Altered regulation of metabolic pathways in human lung cancer discerned by (13)C stable isotope-resolved metabolomics (SIRM). Mol. Cancer 8, 41 (2009).

    Article  Google Scholar 

  14. Sellers, K. et al. Pyruvate carboxylase is critical for non-small-cell lung cancer proliferation. J Clin. Invest. 125, 687–698 (2015).

    Article  Google Scholar 

  15. Sun, R. C. et al. Noninvasive liquid diet delivery of stable isotopes into mouse models for deep metabolic network tracing. Nat. Commun. 8, 1646 (2017).

    Article  Google Scholar 

  16. Jang, C., Chen, L. & Rabinowitz, J. D. Metabolomics and isotope tracing. Cell 173, 822–837 (2018).

    Article  CAS  Google Scholar 

  17. Roussel, R., Carlier, P. G., Robert, J. J., Velho, G. & Bloch, G. 13C/31P NMR studies of glucose transport in human skeletal muscle. Proc. Natl Acad. Sci. USA 95, 1313–1318 (1998).

    Article  CAS  Google Scholar 

  18. Romijn, J. A., Coyle, E. F., Sidossis, L. S., Rosenblatt, J. & Wolfe, R. R. Substrate metabolism during different exercise intensities in endurance-trained women. J. Appl. Physiol. 88, 1707–1714 (2000).

    Article  CAS  Google Scholar 

  19. Coggan, A. R., Kohrt, W. M., Spina, R. J., Bier, D. M. & Holloszy, J. O. Endurance training decreases plasma glucose turnover and oxidation during moderate-intensity exercise in men. J. Appl. Physiol. 68, 990–996 (1990).

    Article  CAS  Google Scholar 

  20. Ma, E. H. et al. Metabolic profiling using stable isotope tracing reveals distinct patterns of glucose utilization by physiologically activated CD8(+) T cells. Immunity 51, 856–870 e855 (2019).

    Article  CAS  Google Scholar 

  21. Tasdogan, A. et al. Metabolic heterogeneity confers differences in melanoma metastatic potential. Nature 577, 115–120 (2020).

    Article  CAS  Google Scholar 

  22. Marin-Valencia, I. et al. Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo. Cell Metab. 15, 827–837 (2012).

    Article  CAS  Google Scholar 

  23. Hui, S. et al. Glucose feeds the TCA cycle via circulating lactate. Nature 551, 115–118 (2017).

    Article  Google Scholar 

  24. Pan, M. et al. Regional glutamine deficiency in tumours promotes dedifferentiation through inhibition of histone demethylation. Nat. Cell Biol. 18, 1090–1101 (2016).

    Article  CAS  Google Scholar 

  25. Zhang, J. et al. 13C isotope-assisted methods for quantifying glutamine metabolism in cancer cells. Methods Enzymol. 542, 369–389 (2014).

    Article  CAS  Google Scholar 

  26. Long, C. P. & Antoniewicz, M. R. High-resolution (13)C metabolic flux analysis. Nat. Protoc. 14, 2856–2877 (2019).

    Article  CAS  Google Scholar 

  27. Yuan, M. et al. Ex vivo and in vivo stable isotope labelling of central carbon metabolism and related pathways with analysis by LC-MS/MS. Nat. Protoc. 14, 313–330 (2019).

    Article  CAS  Google Scholar 

  28. Broekaert, D. & Fendt, S. M. Measuring in vivo tissue metabolism using (13)C glucose infusions in mice. Methods Mol. Biol. 1862, 67–82 (2019).

    Article  CAS  Google Scholar 

  29. Su, X., Lu, W. & Rabinowitz, J. D. Metabolite spectral accuracy on orbitraps. Anal. Chem. 89, 5940–5948 (2017).

    Article  CAS  Google Scholar 

  30. Antoniewicz, M. R., Kelleher, J. K. & Stephanopoulos, G. Elementary metabolite units (EMU): a novel framework for modeling isotopic distributions. Metab. Eng. 9, 68–86 (2007).

    Article  CAS  Google Scholar 

  31. Millard, P., Letisse, F., Sokol, S. & Portais, J. C. IsoCor: correcting MS data in isotope labeling experiments. Bioinformatics 28, 1294–1296 (2012).

    Article  CAS  Google Scholar 

  32. Young, J. D. INCA: a computational platform for isotopically non-stationary metabolic flux analysis. Bioinformatics 30, 1333–1335 (2014).

    Article  CAS  Google Scholar 

  33. Alger, J. R., Sherry, A. D. & Malloy, C. R. tcaSIM: a simulation program for optimal design of (13)C tracer experiments for analysis of metabolic flux by NMR and mass spectroscopy. Curr. Metabolomics 6, 176–187 (2018).

    Article  CAS  Google Scholar 

  34. Yoo, H., Antoniewicz, M. R., Stephanopoulos, G. & Kelleher, J. K. Quantifying reductive carboxylation flux of glutamine to lipid in a brown adipocyte cell line. J. Biol. Chem. 283, 20621–20627 (2008).

    Article  CAS  Google Scholar 

  35. Weitzel, M. et al. 13CFLUX2-high-performance software suite for (13)C-metabolic flux analysis. Bioinformatics 29, 143–145 (2013).

    Article  CAS  Google Scholar 

  36. Hui, S. et al. Quantitative fluxomics of circulating metabolites. Cell Metab. 32, 676–688 e674 (2020).

    Article  CAS  Google Scholar 

  37. Jang, C. et al. Metabolite exchange between mammalian organs quantified in pigs. Cell Metab. 30, 594–606 e593 (2019).

    Article  CAS  Google Scholar 

  38. Liu, S., Dai, Z., Cooper, D. E., Kirsch, D. G. & Locasale, J. W. Quantitative analysis of the physiological contributions of glucose to the TCA cycle. Cell Metab. 32, 619–628 e621 (2020).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

R.J.D. is a Howard Hughes Medical Institute (HHMI) Investigator, the Robert L. Moody, Sr. Faculty Scholar at UT Southwestern and Joel B. Steinberg, M.D. Distinguished Chair in Pediatrics. S.J.M. is an HHMI Investigator, the Mary McDermott Cook Chair in Pediatric Genetics, the Kathryn and Gene Bishop Distinguished Chair in Pediatric Research, the director of the Hamon Laboratory for Stem Cells and Cancer, and a Cancer Prevention and Research Institute of Texas Scholar. The research was supported by the Cancer Prevention and Research Institute of Texas (RP170114 and RP180778), the National Institutes of Health (R35 CA220449; U01 CA228608) and the Robert A. Welch Foundation (I-1733). A.T. was supported by the Leopoldina Fellowship (LPDS 2016-16) from the German National Academy of Sciences and the Fritz Thyssen Foundation. B.F. is supported by the National Institutes of Health (K99/R00 CA237724-01A1). Figures were generated using BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

B.F., A.T., S.J.M., T.P.M and R.J.D. wrote and edited the manuscript. T.P.M established HRMS methodology.

Corresponding authors

Correspondence to Brandon Faubert or Ralph J. DeBerardinis.

Ethics declarations

Competing interests

R.J.D. is an adviser for Agios Pharmaceuticals and Vida Ventures, and a founder and adviser for Atavistik Bio. S.J.M. is an adviser for Frequency Therapeutics and Protein Fluidics.

Additional information

Peer review information Nature Protocols thanks Monica Montopoli and the other, anonymous reviewer(s) for their contribution to the peer review of this work.

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

Related links

Key data used in this protocol

Faubert, B. et al. Cell 171, 358–371 (2017): https://doi.org/10.1016/j.cell.2017.09.019

Tasdogan, A. et al. Nature 577, 115–120 (2020): https://doi.org/10.1038/s41586-019-1847-2

Hensley, CT. et al. Cell 164, 681–694 (2016): https://doi.org/10.1016/j.cell.2015.12.034

Supplementary information

Reporting Summary

Supplementary Data 1

Supplementary data tables for patient and mouse 13C glucose infusions

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Faubert, B., Tasdogan, A., Morrison, S.J. et al. Stable isotope tracing to assess tumor metabolism in vivo. Nat Protoc 16, 5123–5145 (2021). https://doi.org/10.1038/s41596-021-00605-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-021-00605-2

This article is cited by

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: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer