A prospective study examining cachexia predictors in patients with incurable cancer

Ola Magne Vagnildhaug, Cinzia Brunelli, Marianne J Hjermstad, Florian Strasser, Vickie Baracos, Andrew Wilcock, Maria Nabal, Stein Kaasa, Barry Laird, Tora S Solheim, Ola Magne Vagnildhaug, Cinzia Brunelli, Marianne J Hjermstad, Florian Strasser, Vickie Baracos, Andrew Wilcock, Maria Nabal, Stein Kaasa, Barry Laird, Tora S Solheim

Abstract

Background: Early intervention against cachexia necessitates a predictive model. The aims of this study were to identify predictors of cachexia development and to create and evaluate accuracy of a predictive model based on these predictors.

Methods: A secondary analysis of a prospective, observational, multicentre study was conducted. Patients, who attended a palliative care programme, had incurable cancer and did not have cachexia at baseline, were amenable to the analysis. Cachexia was defined as weight loss (WL) > 5% (6 months) or WL > 2% and body mass index< 20 kg/m2. Clinical and demographic markers were evaluated as possible predictors with Cox analysis. A classification and regression tree analysis was used to create a model based on optimal combinations and cut-offs of significant predictors for cachexia development, and accuracy was evaluated with a calibration plot, Harrell's c-statistic and receiver operating characteristic curve analysis.

Results: Six-hundred-twenty-eight patients were included in the analysis. Median age was 65 years (IQR 17), 359(57%) were female and median Karnofsky performance status was 70(IQR 10). Median follow-up was 109 days (IQR 108), and 159 (25%) patients developed cachexia. Initial WL, cancer type, appetite and chronic obstructive pulmonary disease were significant predictors (p ≤ 0.04). A five-level model was created with each level carrying an increasing risk of cachexia development. For Risk-level 1-patients (WL < 3%, breast or hematologic cancer and no or little appetite loss), median time to cachexia development was not reached, while Risk-level 5-patients (WL 3-5%) had a median time to cachexia development of 51 days. Accuracy of cachexia predictions at 3 months was 76%.

Conclusion: Important predictors of cachexia have been identified and used to construct a predictive model of cancer cachexia.

Trial registration: ClinicalTrials.gov Identifier: NCT01362816 .

Keywords: Cachexia; Cancer; Palliative care; Pre-cachexia; Weight loss.

Conflict of interest statement

BL has received honoraria from Helsinn.

FS has had punctual advisorships (boards, expert meetings) for Danone, Grünenthal, Helsinn, ISIS Global, Mundipharma, Novartis, Novelpharm, Obexia, Ono Pharmaceutical, Psioxus Therapeutics, PrIME Oncology, Sunstone Captial, Vifor. On behalf of his institution, he has received unrestricted industry grants for clinical research from Celgene, Fresenius and Helsinn. He has participated in a clinical cachexia trial lead by Novartis.

OMV, CB, MJH, VEB, AW, MN, SK and TSS declare that they have no competing interest.

Figures

Fig. 1
Fig. 1
Flow chart
Fig. 2
Fig. 2
Classification and regression tree (CART) analysis. The study population is divided repeatedly according to optimal cut-offs of the variables weight loss (rounded to the nearest integer), cancer type and appetite loss into subdivisions of significantly different hazard rates. Adjacent subdivisions from different branches with similar hazard rates are combined resulting in five risk-levels. Hazard ratios (HR) are reported relative to the branch with neutral risk cancer type and no or little appetite loss
Fig. 3
Fig. 3
Kaplan-Meier plot of time to cachexia development depending on risk-level. Median time to cachexia development was not reached in level 1, 249 days for level 2, 175 days for level 3, 145 days for level 4 and 51 days for level 5. Log-rank test and test for trend in failure time-analysis were both significant (p <  0.0001)
Fig. 4
Fig. 4
Calibration plot showing the risk of cachexia development after 3 months, as predicted by the risk-level model, plotted against the observed risk
Fig. 5
Fig. 5
Sensitivity and specificity of cachexia prediction at 3 months when using different cut-offs of risk-level to divide patients into a high or low risk group of cachexia development. Risk-level ≥ 2 yields a high sensitivity (95%), while risk-level ≥ 3 yields a high specificity (88%). No single cut-off yields both a high sensitivity and high specificity

References

    1. Vagnildhaug OM, Balstad TR, Almberg SS, Brunelli C, Knudsen AK, Kaasa S, et al. A cross-sectional study examining the prevalence of cachexia and areas of unmet need in patients with cancer. Support Care Cancer. 2018;26(6):1871–1880. doi: 10.1007/s00520-017-4022-z.
    1. Fearon K, Arends J, Baracos V. Understanding the mechanisms and treatment options in cancer cachexia. Nat Rev Clin Oncol. 2013;10(2):90–99. doi: 10.1038/nrclinonc.2012.209.
    1. Awad S, Tan BH, Cui H, Bhalla A, Fearon KC, Parsons SL, et al. Marked changes in body composition following neoadjuvant chemotherapy for oesophagogastric cancer. Clin Nutr. 2012;31(1):74–77. doi: 10.1016/j.clnu.2011.08.008.
    1. Paulsen O, Aass N, Kaasa S, Dale O. Do corticosteroids provide analgesic effects in cancer patients? A systematic literature review. J Pain Symptom Manag. 2013;46(1):96–105. doi: 10.1016/j.jpainsymman.2012.06.019.
    1. Solheim TS, Laird BJ. Evidence base for multimodal therapy in cachexia. Curr Opin Support Palliat Care. 2012;6(4):424–431. doi: 10.1097/SPC.0b013e328359b668.
    1. Crawford J, Dalton JT, Hancock MA, Johnston MA, Steiner M. Enobosarm, A selective androgen receptor modulator (SARM), increases lean body mass (LBM) in advanced non-small cell lung cancer patients in two pivotal, international phase 3 trials. J Cachex Sarcopenia Muscle. 2014;5(1):35–78. doi: 10.1007/s13539-014-0129-y.
    1. Temel JS, Abernethy AP, Currow DC, Friend J, Duus EM, Yan Y, 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(4):519–531. doi: 10.1016/S1470-2045(15)00558-6.
    1. Laird B, Fallon M. Treating cancer cachexia: an evolving landscape. Ann Oncol. 2017;28(9):2055–2056. doi: 10.1093/annonc/mdx345.
    1. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12(5):489–495. doi: 10.1016/S1470-2045(10)70218-7.
    1. Blum D, Stene GB, Solheim TS, Fayers P, Hjermstad MJ, Baracos VE, et al. Validation of the consensus-definition for Cancer Cachexia and evaluation of a classification model--a study based on data from an international multicentre project (EPCRC-CSA) Ann Oncol. 2014;25(8):1635–1642. doi: 10.1093/annonc/mdu086.
    1. van der Meij BS, Schoonbeek CP, Smit EF, Muscaritoli M, van Leeuwen PA, Langius JA. Pre-cachexia and cachexia at diagnosis of stage III non-small-cell lung carcinoma: an exploratory study comparing two consensus-based frameworks. Br J Nutr. 2013;109(12):2231–2239. doi: 10.1017/S0007114512004527.
    1. Vigano AAL, Morais JA, Ciutto L, Rosenthall L, di Tomasso J, Khan S, et al. Use of routinely available clinical, nutritional, and functional criteria to classify cachexia in advanced cancer patients. Clin Nutr. 2017;36(5):1378–1390. doi: 10.1016/j.clnu.2016.09.008.
    1. Hjermstad MJ, Aass N, Aielli F, Bennett M, Brunelli C, Caraceni A, et al. Characteristics of the case mix, organisation and delivery in cancer palliative care: a challenge for good-quality research. BMJ Support Palliat Care. 2016. 10.1136/bmjspcare-2015-000997.
    1. Dewys WD, Begg C, Lavin PT, Band PR, Bennett JM, Bertino JR, et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern cooperative oncology group. Am J Med. 1980;69(4):491–497. doi: 10.1016/S0149-2918(05)80001-3.
    1. Sun L, Quan XQ, Yu S. An epidemiological survey of Cachexia in advanced Cancer patients and analysis on its diagnostic and treatment status. Nutr Cancer. 2015;67(7):1056–1062. doi: 10.1080/01635581.2015.1073753.
    1. Vigano AL, di Tomasso J, Kilgour RD, Trutschnigg B, Lucar E, Morais JA, et al. The abridged patient-generated subjective global assessment is a useful tool for early detection and characterization of cancer cachexia. J Acad Nutr Diet. 2014;114(7):1088–1098. doi: 10.1016/j.jand.2013.09.027.
    1. Groenvold M, Petersen MA, Aaronson NK, Arraras JI, Blazeby JM, Bottomley A, et al. The development of the EORTC QLQ-C15-PAL: a shortened questionnaire for cancer patients in palliative care. Eur J Cancer. 2006;42(1):55–64. doi: 10.1016/j.ejca.2005.06.022.
    1. Mouronte-Roibas C, Leiro-Fernandez V, Fernandez-Villar A, Botana-Rial M, Ramos-Hernandez C, Ruano-Ravina A. COPD, emphysema and the onset of lung cancer. A systematic review. Cancer Lett. 2016;382(2):240–244. doi: 10.1016/j.canlet.2016.09.002.
    1. Van Putten W. CART: Stata module to perform classification and regression tree analysis. Stat Softw Components 2006; Available from: . Accessed 24 Aug 2018.
    1. Grossberg AJ, Scarlett JM, Marks DL. Hypothalamic mechanisms in cachexia. Physiol Behav. 2010;100(5):478–489. doi: 10.1016/j.physbeh.2010.03.011.
    1. Solheim TS, Blum D, Fayers PM, Hjermstad MJ, Stene GB, Strasser F, et al. Weight loss, appetite loss and food intake in cancer patients with cancer cachexia: three peas in a pod? - analysis from a multicenter cross sectional study. Acta Oncol. 2014;53(4):539–546. doi: 10.3109/0284186X.2013.823239.
    1. von Haehling S, Anker SD. Prevalence, incidence and clinical impact of cachexia: facts and numbers-update 2014. J Cachexia Sarcopenia Muscle. 2014;5(4):261–263. doi: 10.1007/s13539-014-0164-8.
    1. Fearon KC, Glass DJ, Guttridge DC. Cancer cachexia: mediators, signaling, and metabolic pathways. Cell Metab. 2012;16(2):153–166. doi: 10.1016/j.cmet.2012.06.011.

Source: PubMed

3
S'abonner