Power Comparisons and Clinical Meaning of Outcome Measures in Assessing Treatment Effect in Cancer Cachexia: Secondary Analysis From a Randomized Pilot Multimodal Intervention Trial

Trude R Balstad, Cinzia Brunelli, Caroline H Pettersen, Svanhild A Schønberg, Frank Skorpen, Marie Fallon, Stein Kaasa, Asta Bye, Barry J A Laird, Guro B Stene, Tora S Solheim, Trude R Balstad, Cinzia Brunelli, Caroline H Pettersen, Svanhild A Schønberg, Frank Skorpen, Marie Fallon, Stein Kaasa, Asta Bye, Barry J A Laird, Guro B Stene, Tora S Solheim

Abstract

Background: New clinical trials in cancer cachexia are essential, and outcome measures with high responsiveness to detect meaningful changes are crucial. This secondary analysis from a multimodal intervention trial estimates sensitivity to change and between treatment effect sizes (ESs) of outcome measures associated with body composition, physical function, metabolism, and trial intervention. Methods: The study was a multicenter, open-label, randomized pilot study investigating the feasibility of a 6-week multimodal intervention [exercise, non-steroidal anti-inflammatory drugs, and oral nutritional supplements containing polyunsaturated fatty acids (n-3 PUFAs)] vs. standard cancer care in non-operable non-small-cell lung cancer and advanced pancreatic cancer. Body composition measures from computerized tomography scans and circulating biomarkers were analyzed. Results: Forty-six patients were randomized, and the analysis included 22 and 18 patients in the treatment and control groups, respectively. The between-group ESs were high for body weight (ES = 1.2, p < 0.001), small for body composition and physical function [handgrip strength (HGS)] measures (ES < 0.25), moderate to high for n-3 PUFAs and 25-hydroxyvitamin D (25-OH vitamin D) (ES range 0.64-1.37, p < 0.05 for all), and moderate for serum C-reactive protein (ES = 0.53, p = 0.12). Analysis within the multimodal treatment group showed high sensitivity to change for adiponectin (ES = 0.86, p = 0.001) and n-3 PUFAs (ES > 0.8, p < 0.05 for all) and moderate for 25-OH vitamin D (ES = 0.49, p = 0.03). In the control group, a moderate sensitivity to change for body weight (ES = -0.84, p = 0.002) and muscle mass (ES = -0.67, p = 0.016) and a high sensitivity to change for plasma levels of 25-OH vitamin D (ES = -0.88, p = 0.002) were found. Conclusion: Demonstrating high sensitivity to change and between treatment ES and body composition measures, body weight still stands out as a clinical and relevant outcome measure in cancer cachexia. Body composition and physical function measures clearly are important to address but demand large sample sizes to detect treatment group differences. Trial registration: ClinicalTrials.gov identifier: NCT01419145.

Keywords: biomarkers; body composition; cachexia; effect size; multimodal management; outcome measures; sample size (n).

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Balstad, Brunelli, Pettersen, Schønberg, Skorpen, Fallon, Kaasa, Bye, Laird, Stene and Solheim.

Figures

Figure 1
Figure 1
Sample size per treatment arm by effect size values. Sample size by treatment arm by effect size (ES) values (black curve). Dashed vertical lines indicate reference value for small (0.8) ESs (17). Colored vertical lines indicate ESBG for each outcome measure: body weight (orange, n = 1), body composition (blue, n = 6, two overlap, one overlaps with metabolism outcome), physical function (black, n = 1), metabolic mediators (pink, n = 6, two overlap), and nutrient components (green, n = 4) (exact values are reported in Table 2). Sample size values for ES < 0.2 are higher than 1,000 and not shown in the figure.

References

    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) 12:489–95. 10.1016/S1470-2045(10)70218-7
    1. Fearon K, Argiles JM, Baracos VE, Bernabei R, Coats A, Crawford J, et al. . Request for regulatory guidance for cancer cachexia intervention trials. J Cachexia Sarcopenia Muscle. (2015) 6:272–4. 10.1002/jcsm.12083
    1. Laird BJA, Balstad TR, Solheim TS. Endpoints in clinical trials in cancer cachexia: where to start? Curr Opin Support Palliat Care. (2018) 12:445–52. 10.1097/SPC.0000000000000387
    1. Bing C. Lipid mobilization in cachexia: mechanisms and mediators. Curr Opin Support Palliat Care. (2011) 5:356–60. 10.1097/SPC.0b013e32834bde0e
    1. Tsoli M, Swarbrick MM, Robertson GR. Lipolytic and thermogenic depletion of adipose tissue in cancer cachexia. Semin Cell Dev Biol. (2016) 54:68–81. 10.1016/j.semcdb.2015.10.039
    1. Loumaye A, Thissen JP. Biomarkers of cancer cachexia. Clin Biochem. (2017) 50:1281–8. 10.1016/j.clinbiochem.2017.07.011
    1. Solheim TS, Laird BJA, Balstad TR, Stene GB, Bye A, Johns N, et al. . A randomized phase II feasibility trial of a multimodal intervention for the management of cachexia in lung and pancreatic cancer. J Cachexia Sarcopenia Muscle. (2017) 8:778–88. 10.1002/jcsm.12201
    1. Solheim TS, Laird BJA, Balstad TR, Bye A, Stene G, Baracos V, et al. . Cancer cachexia: rationale for the MENAC (multimodal-exercise, nutrition and anti-inflammatory medication for cachexia) trial. BMJ Support Palliat Care. (2018) 8:258–65. 10.1136/bmjspcare-2017-001440
    1. Mourtzakis M, Prado CM, Lieffers JR, Reiman T, McCargar LJ, Baracos VE. 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. 10.1139/H08-075
    1. Shen W, Punyanitya M, Wang Z, Gallagher D, St-Onge MP, Albu J, et al. . Total body skeletal muscle and adipose tissue volumes: estimation from a single abdominal cross-sectional image. J Appl Physiol. (2004) 97:2333–8. 10.1152/japplphysiol.00744.2004
    1. Popuri K, Cobzas D, Esfandiari N, Baracos V, Jagersand M. Body composition assessment in axial CT images using FEM-based automatic segmentation of skeletal muscle. IEEE Trans Med Imaging. (2016) 35:512–20. 10.1109/TMI.2015.2479252
    1. Miller KD, Jones E, Yanovski JA, Shankar R, Feuerstein I, Falloon J. Visceral abdominal-fat accumulation associated with use of indinavir. Lancet. (1998) 351:871–5. 10.1016/S0140-6736(97)11518-5
    1. Mitsiopoulos N, Baumgartner RN, Heymsfield SB, Lyons W, Gallagher D, Ross R. Cadaver validation of skeletal muscle measurement by magnetic resonance imaging and computerized tomography. J Appl Physiol. (1998) 85:115–22. 10.1152/jappl.1998.85.1.115
    1. Stene GB, Balstad TR, Leer ASM, Bye A, Kaasa S, Fallon M, et al. . Deterioration in muscle mass and physical function differs according to weight loss history in cancer cachexia. Cancers (Basel). (2019) 11:1925. 10.3390/cancers11121925
    1. Agustsson T, Ryden M, Hoffstedt J, van Harmelen V, Dicker A, Laurencikiene J, et al. . Mechanism of increased lipolysis in cancer cachexia. Cancer Res. (2007) 67:5531–7. 10.1158/0008-5472.CAN-06-4585
    1. Lakens D. Calculating and reporting effect sizes to facilitate cumulative science: a practical primer for t-tests and ANOVAs. Front Psychol. (2013) 4:863. 10.3389/fpsyg.2013.00863
    1. Cohen J. Statistical Power Analysis for the Behavioral Sciences, 2nd edn. New York, NY: Taylor & Francis Inc; (1988).
    1. Prado CM, Purcell SA, Alish C, Pereira SL, Deutz NE, Heyland DK, et al. . Implications of low muscle mass across the continuum of care: a narrative review. Ann Med. (2018) 50:675–93. 10.1080/07853890.2018.1511918
    1. Faber J, Fonseca LM. How sample size influences research outcomes. Dental Press J Orthod. (2014) 19:27–9. 10.1590/2176-9451.19.4.027-029.ebo
    1. MacDonald AJ, Greig CA, Baracos V. The advantages and limitations of cross-sectional body composition analysis. Curr Opin Support Palliat Care. (2011) 5:342–9. 10.1097/SPC.0b013e32834c49eb
    1. Baracos V, Kazemi-Bajestani SM. Clinical outcomes related to muscle mass in humans with cancer and catabolic illnesses. Int J Biochem Cell Biol. (2013) 45:2302–8. 10.1016/j.biocel.2013.06.016
    1. Shen W, Chen J, Gantz M, Velasquez G, Punyanitya M, Heymsfield SB. A single MRI slice does not accurately predict visceral and subcutaneous adipose tissue changes during weight loss. Obesity (Silver Spring, Md). (2012) 20:2458–63. 10.1038/oby.2012.168
    1. Dobs AS, Boccia RV, Croot CC, Gabrail NY, Dalton JT, Hancock ML, et al. Effects of enobosarm on muscle wasting and physical function in patients with cancer: a double-blind, randomised controlled phase 2 trial. Lancet Oncol. (2013) 14:335–45. 10.1016/S1470-2045(13)70055-X
    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) 17:519–31. 10.1016/S1470-2045(15)00558-6
    1. Garcia JM, Boccia RV, Graham CD, Yan Y, Duus EM, Allen S, et al. . Anamorelin for patients with cancer cachexia: an integrated analysis of two phase 2, randomised, placebo-controlled, double-blind trials. Lancet Oncol. (2015) 16:108–16. 10.1016/S1470-2045(14)71154-4
    1. Ramage MI, Skipworth RJE. The relationship between muscle mass and function in cancer cachexia: smoke and mirrors? Curr Opin Support Palliat Care. (2018) 12:439–44. 10.1097/SPC.0000000000000381
    1. Tsoli M, Robertson G. Cancer cachexia: malignant inflammation, tumorkines, and metabolic mayhem. Trends Endocrinol Metab. (2013) 24:174–83. 10.1016/j.tem.2012.10.006
    1. Daas SI, Rizeq BR, Nasrallah GK. Adipose tissue dysfunction in cancer cachexia. J Cell Physiol. (2018) 234:13–22. 10.1002/jcp.26811
    1. Maeda N, Funahashi T, Matsuzawa Y, Shimomura I. Adiponectin, a unique adipocyte-derived factor beyond hormones. Atherosclerosis. (2020) 292:1–9. 10.1016/j.atherosclerosis.2019.10.021
    1. Kerem M, Ferahkose Z, Yilmaz UT, Pasaoglu H, Ofluoglu E, Bedirli A, et al. . Adipokines and ghrelin in gastric cancer cachexia. World J Gastroenterol. (2008) 14:3633–41. 10.3748/wjg.14.3633
    1. Diakowska D, Markocka-Maczka K, Szelachowski P, Grabowski K. Serum levels of resistin, adiponectin, and apelin in gastroesophageal cancer patients. Dis Markers. (2014) 2014:619649. 10.1155/2014/619649
    1. Smiechowska J, Utech A, Taffet G, Hayes T, Marcelli M, Garcia JM. Adipokines in patients with cancer anorexia and cachexia. J Investig Med. (2010) 58:554–9. 10.2310/JIM.0b013e3181cf91ca
    1. von Frankenberg AD, Silva FM, de Almeida JC, Piccoli V, do Nascimento FV, Sost MM, et al. Effect of dietary lipids on circulating adiponectin: a systematic review with meta-analysis of randomised controlled trials. Br J Nutr. (2014) 112:1235–50. 10.1017/S0007114514002013
    1. Wu JH, Cahill LE, Mozaffarian D. Effect of fish oil on circulating adiponectin: a systematic review and meta-analysis of randomized controlled trials. J Clin Endocrinol Metab. (2013) 98:2451–9. 10.1210/jc.2012-3899
    1. Abbott JH. The distinction between randomized clinical trials (RCTs) and preliminary feasibility and pilot studies: what they are and are not. J Orthop Sports Phys Ther. (2014) 44:555–8. 10.2519/jospt.2014.0110
    1. Bland JM, Altman DG. Comparisons against baseline within randomised groups are often used and can be highly misleading. Trials. (2011) 12:264. 10.1186/1745-6215-12-264

Source: PubMed

3
Tilaa