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:

Cancer-associated malnutrition

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

References

  1. WHO. Cancer Fact sheet [online], http://www.who.int/mediacentre/factsheets/fs297/en/ (2017).

  2. Hébuterne X, et al. Prevalence of malnutrition and current use of nutrition support in patients with cancer. JPEN J Parenter Enteral Nutr. 2014;38:196–204.

    Article  Google Scholar 

  3. WHO. Obesity and overweight fact sheet [online], http://www.who.int/mediacentre/factsheets/fs311/en/ (2016).

  4. Fearon K, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12:489–95.

    Article  Google Scholar 

  5. Jensen GL, et al. International Consensus Guideline Committee. Adult starvation and disease-related malnutrition: a proposal for etiology-based diagnosis in the clinical practice setting from the International Consensus Guideline Committee. JPEN J Parenter Enteral Nutr. 2010;34:156–9.

    Article  Google Scholar 

  6. Dechaphunkul T, et al. Malnutrition assessment in patients with cancers of the head and neck: a call to action and consensus. Crit Rev Oncol Hematol. 2013;88:459–76.

    Article  Google Scholar 

  7. Baracos VE, et al. Cancer-associated cachexia. Nat Rev Dis Prim. 2018;4:17105.

    Article  Google Scholar 

  8. de van der Schueren MAE, et al. Systematic review and meta-analysis of the evidence for oral nutritional intervention on nutritional and clinical outcomes during chemo(radio)therapy: current evidence and guidance for design of future trials. Ann Oncol. 2018;29:1141–53.

    Article  Google Scholar 

  9. Mourtzakis M, et al. A practical and precise approach to quantification of body composition in cancer patients using computed tomography images acquired during routine care. Appl Physiol Nutr Metab. 2008;33:997–1006.

    Article  Google Scholar 

  10. Kazemi-Bajestani SMR, et al. Computed tomography-defined muscle and fat wasting are associated with cancer clinical outcomes. Semin Cell Dev Biol. 2016;54:2–10.

    Article  Google Scholar 

  11. Arends J, et al. ESPEN guidelines on nutrition in cancer patients. Clin Nutr. 2017;36:11–48.

    Article  Google Scholar 

  12. Bozzetti F, et al. The prognosis of incurable cachectic cancer patients on home parenteral nutrition: a multi-centre observational study with prospective follow-up of 414 patients. Ann Oncol. 2014;25:487–93.

    Article  CAS  Google Scholar 

  13. Bozzetti F, et al. Development and validation of a nomogram to predict survival in incurable cachectic cancer patients on home parenteral nutrition. Ann Oncol. 2015;26:2335–40.

    Article  CAS  Google Scholar 

  14. Crowder SL, et al. Nutrition impact symptoms and associated outcomes in post-chemoradiotherapy head and neck cancer survivors: a systematic review. J Cancer Surviv. 2018. https://doi.org/10.1007/s11764-018-0687-7.

    Article  Google Scholar 

  15. Scott D, et al. Health care professionals’ experience, understanding and perception of need of advanced cancer patients with cachexia and their families: the benefits of a dedicated clinic. BMC Palliat Care. 2016;15:100.

    Article  Google Scholar 

  16. Dev R, et al. Hypermetabolism and symptom burden in advanced cancer patients evaluated in a cachexia clinic. J Cachex- Sarcopenia Muscle. 2015;6:95–98.

    Article  Google Scholar 

  17. Vigano A, et al. The Cachexia Clinic: from staging to managing nutritional and functional problems in advanced cancer patients. Crit Rev Oncog. 2012;17:293–304.

    Article  Google Scholar 

  18. Cederholm T, et al. To create a consensus on malnutrition diagnostic criteria. JPEN J Parenter Enteral Nutr. 2017;41:311–4.

    Article  Google Scholar 

  19. Martin L, et al. Diagnostic criteria for the classification of cancer-associated weight loss. J Clin Oncol. 2015;33:90–9.

    Article  Google Scholar 

  20. Schiessel DL, et al. Barriers to cancer nutrition therapy: excess catabolism of muscle and adipose tissues induced by tumour products and chemotherapy. Proc Nutr Soc. 2018;30:1–9.

    Google Scholar 

  21. Loomans HA, et al. Intertwining of activin a and TGFβ signaling: dual roles in cancer progression and cancer cell invasion. Cancers. 2014;7:70–91.

    Article  Google Scholar 

  22. Agustsson T, et al. Mechanism of increased lipolysis in cancer cachexia. Cancer Res. 2007;67:5531–7.

    Article  CAS  Google Scholar 

  23. Xue Y, et al. Zinc-α-2-glycoprotein: a candidate biomarker for colon cancer diagnosis in chinese population. Int J Mol Sci. 2015;16:691–703.

    Article  Google Scholar 

  24. Cabassi A, et al. Zinc-α2-glycoprotein as a marker of fat catabolism in humans. Curr Opin Clin Nutr Metab Care. 2013;16:267–71.

    Article  CAS  Google Scholar 

  25. Adegoke OAJ, et al. mTORC1 and the regulation of skeletal muscle anabolism and mass. Appl Physiol Nutr Metab. 2012;37:395–406.

    Article  CAS  Google Scholar 

  26. Egerman MA, et al. Signaling pathways controlling skeletal muscle mass. Crit Rev Biochem Mol Biol. 2014;49:59–68.

    Article  CAS  Google Scholar 

  27. Martin L, et al. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol. 2013;31:1539–47.

    Article  Google Scholar 

  28. Blauwhoff-Buskermolen S, et al. Loss of muscle mass during chemotherapy is predictive for poor survival of patients with metastatic colorectal cancer. J Clin Oncol. 2016;34:1339–44.

    Article  CAS  Google Scholar 

  29. Ní Bhuachalla ÉB, et al. Computed tomography diagnosed cachexia and sarcopenia in 725 oncology patients: is nutritional screening capturing hidden malnutrition?. J Cachex- Sarcopenia Muscle. 2018;9:295–305.3.

    Article  Google Scholar 

  30. Purcell SA, et al. Key determinants of energy expenditure in cancer and implications for clinical practice. Eur J Clin Nutr. 2016;70:1230–8.

    Article  CAS  Google Scholar 

  31. Mitchell CJ, et al. The effects of dietary protein intake on appendicular lean mass and muscle function in elderly men: a 10-wk randomized controlled trial. Am J Clin Nutr. 2017;106:1375–83.

    Article  CAS  Google Scholar 

  32. Weyermann P, et al. Orally available selective melanocortin-4 receptor antagonists stimulate food intake and reduce cancer-induced cachexia in mice. PLoS ONE. 2009;4:e4774.

    Article  Google Scholar 

  33. Breit SN, et al. Targeting obesity and cachexia: identification of the GFRAL receptor-MIC-1/GDF15 pathway. Trends Mol Med. 2017;3:1065–7.

    Article  Google Scholar 

  34. Temel JS, et al. Anamorelin in patients with non-small-cell lung cancer and cachexia (ROMANA 1 and ROMANA 2): results from two randomised, double-blind, phase 3 trials. Lancet Oncol. 2016;17:519–31.

    Article  CAS  Google Scholar 

  35. Prado CMM, et al. Skeletal muscle anabolism is a side effect of therapy with the MEK inhibitor: selumetinib in patients with cholangiocarcinoma. Br J Cancer. 2012;106:1583–6.

    Article  CAS  Google Scholar 

  36. Pappalardo G, et al. Eicosapentaenoic acid in cancer improves body composition and modulates metabolism. Nutrition. 2015;31:549–55.

    Article  CAS  Google Scholar 

  37. Di Girolamo FG, et al. Omega-3 fatty acids and protein metabolism: enhancement of anabolic interventions for sarcopenia. Curr Opin Clin Nutr Metab Care. 2014;17:145–50.

    Article  Google Scholar 

  38. Xue H, et al. Nutrition modulation of gastrointestinal toxicity related to cancer chemotherapy: from preclinical findings to clinical strategy. JPEN J Parenter Enteral Nutr. 2011;35:74–90.

    Article  CAS  Google Scholar 

  39. Fridman WH, et al. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 2017;14:717–34.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The author receives funding from the Canadian Institutes of Health Research and the Alberta Cancer Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Vickie E. Baracos.

Ethics declarations

Conflict of interest

The author declares that they have no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Baracos, V.E. Cancer-associated malnutrition. Eur J Clin Nutr 72, 1255–1259 (2018). https://doi.org/10.1038/s41430-018-0245-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41430-018-0245-4

This article is cited by

Search

Quick links