Handling informative dropout in longitudinal analysis of health-related quality of life: application of three approaches to data from the esophageal cancer clinical trial PRODIGE 5/ACCORD 17

B Cuer, C Mollevi, A Anota, E Charton, B Juzyna, T Conroy, C Touraine, B Cuer, C Mollevi, A Anota, E Charton, B Juzyna, T Conroy, C Touraine

Abstract

Background: Health-related quality of life (HRQoL) has become a major endpoint to assess the clinical benefit of new therapeutic strategies in oncology clinical trials. Typically, HRQoL outcomes are analyzed using linear mixed models (LMMs). However, longitudinal analysis of HRQoL in the presence of missing data remains complex and unstandardized. Our objective was to compare the modeling alternatives that account for informative dropout.

Methods: We investigated three alternative methods-the selection model (SM), pattern-mixture model (PMM), and shared-parameters model (SPM)-in relation to the LMM. We first compared them on the basis of methodological arguments highlighting their advantages and drawbacks. Then, we applied them to data from a randomized clinical trial that included 267 patients with advanced esophageal cancer for the analysis of four HRQoL dimensions evaluated using the European Organisation for Research and Treatment of Cancer (EORTC) QLQ-C30 questionnaire.

Results: We highlighted differences in terms of outputs, interpretation, and underlying modeling assumptions; this methodological comparison could guide the choice of method according to the context. In the application, none of the four models detected a significant difference between the two treatment arms. The estimated effect of time on HRQoL varied according to the method: for all analyzed dimensions, the PMM estimated an effect that contrasted with those estimated by the SM and SPM; the LMM estimated effects were confirmed by the SM (on two of four HRQoL dimensions) and SPM (on three of four HRQoL dimensions).

Conclusions: The PMM, SM, or SPM should be used to confirm or invalidate the results of LMM analysis when informative dropout is suspected. Of these three alternative methods, the SPM appears to be the most interesting from both theoretical and practical viewpoints.

Trial registration: This study is registered with ClinicalTrials.gov , number NCT00861094 .

Keywords: Cancer clinical trial; Health-related quality of life; Informative dropout; Joint modeling; Pattern-mixture model; Selection model; Shared-parameters model.

Conflict of interest statement

The authors have declared no conflicts of interest.

Figures

Fig. 1
Fig. 1
Patients who dropped out after visits V0 to V6, or did not drop out (V7). Legend: Ratio calculated by treatment arm in evaluable patients for each of the four dimensions of EORTC QLQ-C30 (QL, global health status; PF, physical functioning; PA, pain; and FA, fatigue) during radiochemotherapy (RT), chemotherapy (CT), and follow-up visits (V)
Fig. 2
Fig. 2
Estimated parameters and 95% confidence intervals. Legend: Time effect β1 (slope in the control arm) and interaction effect β2 (slope difference between the experimental and control arm) for the four dimensions of the EORTC QLQ-C30 (QL, PF, PA, and FA) according to the LMM, PMM, SM, SPM
Fig. 3
Fig. 3
Predicted HRQoL score trajectories of the pattern-mixture model. Legend: Predictions over time by treatment arm regimen for the four dimensions of EORTC QLQ-C30 (QL, PF, PA, and FA). The linear trajectories are shown in pattern 1 (last measurement at visits V0 = 0, V1 = 1.25, or V2 = 3 months), pattern 2 (last measurement at visits V3 = 4 or V4 = 6 months), pattern 3 (last measurement at visits V5 = 12 months), pattern 4 (last measurement at visits V6 = 24 or V7 = 36 months, i.e., no dropout) and the overall patterns (marginal HRQoL scores). The solid line refers to the control fluorouracil-cisplatin regimen and the dashed line refers the experimental FOLFOX regimen
Fig. 4
Fig. 4
Predicted HRQoL score trajectories. Legend: Predictions for the four dimensions of the EORTC QLQ-C30 (QL, PF, PA, and FA) according to the LMM, PMM, SM, and SPM. The solid line refers to the control fluorouracil-cisplatin regimen and the dashed line refers the experimental FOLFOX regimen

References

    1. Osoba D. Health-related quality of life and cancer clinical trials. Ther Adv Med Oncol. 2011;3:57–71. doi: 10.1177/1758834010395342.
    1. Rubin DB. Inference and missing data. Biometrika. 1976;63:581–592. doi: 10.1093/biomet/63.3.581.
    1. Little RJA, Rubin DB. Statistical analysis with missing data. New York: Wiley; 1986.
    1. Fielding S, Ogbuagu A, Sivasubramaniam S, MacLennan G, Ramsay CR. Reporting and dealing with missing quality of life data in RCTs: has the picture changed in the last decade? Qual Life Res. 2016;25:2977–2983. doi: 10.1007/s11136-016-1411-6.
    1. Fairclough DL. Design and analysis of quality of life studies in clinical trials. 2010.
    1. Cnaan A, Laird NM, Slasor P. Using the general linear mixed model to analyse unbalanced repeated measures and longitudinal data. Stat Med. 1997;16:2349–2380. doi: 10.1002/(SICI)1097-0258(19971030)16:20<2349::AID-SIM667>;2-E.
    1. Fielding S, Fayers PM, Loge JH, Jordhøy MS, Kaasa S. Methods for handling missing data in palliative care research. Palliat Med. 2006;20:791–798. doi: 10.1177/0269216306072555.
    1. Hussain JA, White IR, Langan D, Johnson MJ, Currow DC, Torgerson DJ, et al. Missing data in randomized controlled trials testing palliative interventions pose a significant risk of bias and loss of power: a systematic review and meta-analyses. J Clin Epidemiol. 2016;74:57–65. doi: 10.1016/j.jclinepi.2015.12.003.
    1. Fairclough DL, Peterson HF, Chang V. Why are missing quality of life data a problem in clinical trials of cancer therapy? Stat Med. 1998;17:667–677. doi: 10.1002/(SICI)1097-0258(19980315/15)17:5/7<667::AID-SIM813>;2-6.
    1. DeSouza CM, Legedza ATR, Sankoh AJ. An overview of practical approaches for handling missing data in clinical trials. J Biopharm Stat. 2009;19:1055–1073. doi: 10.1080/10543400903242795.
    1. Ibrahim JG, Molenberghs G. Missing data methods in longitudinal studies: a review. Test Madr Spain. 2009;18:1–43.
    1. Hogan JW, Laird NM. Model-based approaches to analysing incomplete longitudinal and failure time data. Stat Med. 1997;16:259–272. doi: 10.1002/(SICI)1097-0258(19970215)16:3<259::AID-SIM484>;2-S.
    1. Little RJA. Modeling the drop-out mechanism in repeated-measures studies. J Am Stat Assoc. 1995;90:1112–1121. doi: 10.1080/01621459.1995.10476615.
    1. Tsiatis AA, Davidian M. Joint modeling of longitudinal and time-to-event data: an overview. Stat Sin. 2004:809–34.
    1. Vonesh EF, Greene T, Schluchter MD. Shared parameter models for the joint analysis of longitudinal data and event times. Stat Med. 2006;25:143–163. doi: 10.1002/sim.2249.
    1. Dupuy J-F. Joint modeling of survival and nonignorable missing longitudinal quality-of-life data. In: Mesbah M, Cole BF, Lee M-LT, editors. Statistical methods for quality of life studies: design, measurements and analysis. Boston: Springer US; 2002. pp. 309–322.
    1. Michiels B, Molenberghs G, Bijnens L, Vangeneugden T, Thijs H. Selection models and pattern-mixture models to analyse longitudinal quality of life data subject to drop-out. Stat Med. 2002;21:1023–1041. doi: 10.1002/sim.1064.
    1. Bell ML, Fairclough DL. Practical and statistical issues in missing data for longitudinal patient-reported outcomes. Stat Methods Med Res. 2013;23:440–459. doi: 10.1177/0962280213476378.
    1. Du H, Hahn EA, Cella D. The impact of missing data on estimation of health-related quality of life outcomes. Anal Randomized Longitud Clin Trial. 2011;11:134–144.
    1. Conroy T, Galais M-P, Raoul J-L, Bouché O, Gourgou-Bourgade S, Douillard J-Y, et al. Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol. 2014;15:305–314. doi: 10.1016/S1470-2045(14)70028-2.
    1. Bascoul-Mollevi C, Gourgou S, Galais M-P, Raoul J-L, Bouché O, Douillard J-Y, et al. Health-related quality of life results from the PRODIGE 5/ACCORD 17 randomised trial of FOLFOX versus fluorouracil–cisplatin regimen in oesophageal cancer. Eur J Cancer. 2017;84:239–249. doi: 10.1016/j.ejca.2017.07.038.
    1. Aaronson NK, Ahmedzai S, Bergman B, Bullinger M, Cull A, Duez NJ, et al. The European Organization for Research and Treatment of Cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. JNCI J Natl Cancer Inst. 1993;85:365–376. doi: 10.1093/jnci/85.5.365.
    1. Fayers P, Aaronson NK, Bjordal K, Groenvold M, Curran D, Bottomley A. EORTC QLQ-C30 scoring manual. 3 2001.
    1. Diggle P, Kenward MG. Informative drop-out in longitudinal data analysis. J R Stat Soc: Ser C: Appl Stat. 1994;43:49–93.
    1. Pauler DK, McCoy S, Moinpour C. Pattern mixture models for longitudinal quality of life studies in advanced stage disease. Stat Med. 2003;22:795–809. doi: 10.1002/sim.1397.
    1. Pinheiro JC, Bates DM. Mixed-effects models in S and S-PLUS. N. Y: Springer; 2000.
    1. Rizopoulos D. JM: an r package for the joint modelling of longitudinal and time-to-event data. J Stat Softw Artic. 2010;35:1–33.
    1. Thijs H, Molenberghs G, Michiels B, Verbeke G, Curran D. Strategies to fit pattern-mixture models. Biostatistics. 2002;3:245–265. doi: 10.1093/biostatistics/3.2.245.
    1. Little RJA. Pattern-mixture models for multivariate incomplete data. J Am Stat Assoc. 1993;88:125–134.
    1. Verbeke G, Molenberghs G, Thijs H, Lesaffre E, Kenward MG. Sensitivity analysis for nonrandom dropout: a local influence approach. Biometrics. 2001;57:7–14. doi: 10.1111/j.0006-341X.2001.00007.x.
    1. Rizopoulos D, Verbeke G, Molenberghs G. Shared parameter models under random effects misspecification. Biometrika. 2008;95:63–74. doi: 10.1093/biomet/asm087.
    1. Rizopoulos D. The r package JMbayes for fitting joint models for longitudinal and time-to-event data using MCMC. J Stat Softw Artic. 2016;72:1–46.
    1. Crowther MJ, Abrams KR, Lambert PC. Joint modeling of longitudinal and survival data. Stata J. 2013;13:165–184. doi: 10.1177/1536867X1301300112.
    1. Garcia-Hernandez A, Rizopoulos D. %JM: A SAS macro to fit jointly generalized mixed models for longitudinal data and time-to-event responses. J Stat Softw Artic. 2018;84:1–29.
    1. Molenberghs G, Kenward M. Missing data in clinical studies: Wiley; 2007.
    1. Calvert M, Blazeby J, Altman DG, Revicki DA, Moher D, Brundage MD, et al. Reporting of patient-reported outcomes in randomized trials: the CONSORT PRO extension. JAMA. 2013;309:814–822. doi: 10.1001/jama.2013.879.
    1. Little RJ, D’agostino R, Cohen ML, Dickersin K, Emerson SS, Farrar JT, et al. The prevention and treatment of missing data in clinical trials. N Engl J Med. 2012;367:1355–1360. doi: 10.1056/NEJMsr1203730.
    1. Bottomley A, Pe M, Sloan J, Basch E, Bonnetain F, Calvert M, et al. Moving forward toward standardizing analysis of quality of life data in randomized cancer clinical trials. Clin Trials. 2018;15:624–630. doi: 10.1177/1740774518795637.
    1. Bell ML, Fiero M, Horton NJ, Hsu C-H. Handling missing data in RCTs; a review of the top medical journals. BMC Med Res Methodol. 2014;14:118. doi: 10.1186/1471-2288-14-118.

Source: PubMed

3
Subscribe