• No results found

VU Research Portal

N/A
N/A
Protected

Academic year: 2021

Share "VU Research Portal"

Copied!
25
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Assessment of health-related quality of life in cancer clinical trials

Ediebah, D.E.

2020

document version

Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)

Ediebah, D. E. (2020). Assessment of health-related quality of life in cancer clinical trials: clinical relevance and

methodological barriers.

General rights

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain

• You may freely distribute the URL identifying the publication in the public portal ?

Take down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address:

vuresearchportal.ub@vu.nl

(2)

Methodological barriers in

assessing health-related quality

(3)
(4)

| 87

the analysis of health-related quality

of life outcomes in cancer patients

Annals of Oncology 24: 231–237, 2013 Divine E. Ediebah Corneel Coens John T. Maringwa Chantal Quinten Efstathios Zikos Jolie Ringash Madaleine T. king Carolyn Gotay Hans-H. Flechtner Joseph S. von Koch Joachim Weis Egbert F. Smit

Claus-Henning Kohne Andrew Bottomley

(5)

ABTRACT

Background: We examined if cancer patients’ health-related quality of life

(HRQOL) scores on the European Organisation for Research and Treatment of Cancer (EORTC) QLQ-C30 are affected by the specific time point, before or during treatment, at which the questionnaire is completed, and whether this could bias the overall treatment comparison analyses.

Patient and methods: A ‘completion-time window’ variable was created on three

closed EORTC randomised control trials in lung (non-small cell lung cancer, NSCLC) and colorectal cancer (CRC) to indicate when the QLQ-30 was com-pleted relative to chemotherapy cycle dates, defined as ‘before’, ‘on’ and ‘after’. HRQOL mean scores were calculated using a linear mixed model.

Results: Statistically significant differences (P < 0.05) were observed on 6 and

5 scales for ‘on’ and ‘after’ comparisons in the NSCLC and two-group CRC trial, respectively. As for the three-group CRC trial, several statistical differences were observed in the ‘before’ to ‘on’ and the ‘on’ to ‘after’ comparisons. For all three trials, including the ‘completion-time window’ variable in the model resulted in a better fit, but no substantial changes in the treatment effects were noted.

Conclusion: We showed that considering the exact timing of completion within

specified windows resulted in statistical and potentially clinically significant dif-ferences, but it did not alter the conclusions of treatment comparison in these studies.

(6)

INTRODUCTION

The assessment of health-related quality of life (HRQOL) in cancer patients has increased over recent years, and it has become an integral part of many ran-domised clinical trials. However, careful assessments of HRQOL and appropriate analysis techniques are required. Appropriate techniques have been described in the literature, including linear mixed models for analysing longitudinal data.1 A key aspect in the analysis of HRQOL data is the timing of the assessments. Osoba pointed out that when studying the effects of a treatment that is cyclic and toxic, the planning and timing of HRQOL assessment has particular rel-evance.2 For example, the most severe side-effects related to cancer chemo-therapy occur during and immediately after treatment.3 Data in clinical trials are often most conveniently collected when the patient attends a scheduled clinic visit, or is admitted to hospital for treatment. In these circumstances, HRQOL is generally assessed just before receiving a cycle of chemotherapy.3

In an appropriately designed clinical trial, the protocol will state exactly when the HRQOL assessments are scheduled. For example, as noted above, it could be stated that patients should complete the HRQOL questionnaire before receiving chemotherapy on each of the scheduled cycles. However, it is clear that deviations from this schedule are not unexpected. Patients may, for some reason, fail to complete the questionnaire at the scheduled time, but then return it by post a few days later. In order not to lose patients, analyses of HRQOL fre-quently use ‘completion-time windows’ around the expected completion-time.4 For example, a certain number of days before and after the scheduled treat-ment cycle date may be allowed, such that all questionnaires completed within that period are assumed to belong to that particular cycle. The time scheduled for patients to come for clinical visits may not capture the clinically important period, because the severe side-effects from the previous cycle may no longer be present. Although the importance of timing of HRQOL seems apparent, the issue has received relatively limited attention in the literature.5, 6

Our study aims to evaluate whether HRQOL scores on the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 (EORTC QLQ-C30) are affected by the specific time point, before or during treatment, at which the questionnaire was completed, and whether this could unduly bias the treatment comparisons.

(7)

PATIENTS AND METHODS

For our analysis, we used three closed and published, multi-centre, EORTC ran-domised phase III trials consisting of one three-group non-small cell lung can-cer (NSCLC) trial (trial 1), a two-group colorectal cancan-cer (CRC) trial (trial 2) and a three-group CRC trial (trial 3). In all three trials, HRQOL was a secondary end point measured at baseline and during treatment. The NSCLC trial enrolled 480 palliative, locally advanced and/or metastatic cancer patients randomised into three different chemotherapy groups [paclitaxel (Taxol, Bristol-Myers Squibb, Belgium) 175 mg/m2—cisplatin 80 mg/m2 (group A), gemcitabine 1250 mg/m2 cisplatin 80 mg/ m2 (group B) and paclitaxel 175 mg/m2—gemcitabine 1250 mg/ m2 (group C)].7 The two colorectal trials enrolled 430 (trial 2) and 498 (trial 3) patients with metastatic CRC. In trial 2, patients were randomised into folinic acid 500 mg/m2 followed by fluorouracil 2.6 g/m2 (reference group A), or fluo-rouracil 2.3 g/m2, later reduced to fluorouracil 2.0 g/m2 (experimental group B). For trial 3, patients were randomised into: leucovorin 20 mg/m2 as intravenous (i.v.) bolus followed by fluorouracil 425 mg/m2 as i.v. bolus (reference group C); or one of two experimental groups, one of which received 2600 mg/m2 of i.v. flu-orouracil as a 24-h infusion with (fluflu-orouracil24H+ leucovorin) (group B) and the other received leucovorin 500 mg/m2 i.v. alone as a 2-h infusion (group A) before each fluorouracil administration.8,9

HRQOL was assessed with the EORTC QLQ-C30 (versions 2 and 3). The EORTC QLQ-C30 is a validated multi-domain instrument, containing both sin-gle- and multi-item scales. Of the 30 items, 24 aggregate into nine multi-item scales representing various HRQOL dimensions: five functioning scales (phys-ical, role, emotional, cognitive and social), three symptom scales (fatigue, pain and nausea) and the global health status/QoL scale. The remaining six sin-gle-item scales assess symptoms: dyspnoea, appetite loss, sleep disturbance, constipation and diarrhoea and the perceived financial impact of the disease treatment. The recall period is the past week. All scales and item scores are linearly transformed to a scale from 0 to 100 for ease of statistical interpreta-tion and psychometric validainterpreta-tion. High scores indicate better HRQOL for the five functional scales and the global health status/QoL scale but worse HRQOL for the symptoms scales and items. This instrument has been extensively tested

(8)

for reliability and validity.10–18 Based on the work by Osoba et al.19 the minimum clinically meaningful difference in an HRQOL variable was classified as a differ-ence of at least 10 points on the scale, categorising patients as improved, stable or worsened on the EORTC QLQ-C30 scales and items. Similarly, Ringash et al.20 reported that one rule of thumb for interpreting a difference in HRQOL scores is to consider a change of ~5–10. More information on the trials design can be found in supplementary Table S1, available at Annals of Oncology online.

Statistical Analysis

To investigate the effect of questionnaire completion before and during treat-ment, a ‘completion-time window’ variable was created to indicate when the EORTC QLQ-C30 was completed relative to cycle dates, defined as ‘before’ (up to 10 days before the cycle date), ‘on’ (on the cycle date) and ‘after’ (up to 10 days after the cycle date). HRQOL mean scores were estimated using linear mixed models for longitudinal data analysis21 with first-order autoregressive covariance structure. The first-order autoregressive covariance structure was used based on the Akaike information criterion (AIC)/Bayesian information criterion (BIC) values from comparing different covariance structure in the mixed model (smallest AIC/ BIC value).22,23 The variables treatment, cycle of treatment and treatment-by-cycle interaction were included as fixed effects with subject-specific random intercepts.

The global health status/QoL scale was used as the main HRQOL outcome to assess the added value for including the completion-time variable in the model. To do this, change in global health status/QoL score from baseline was compared at each assessment time point, with the level of significance set at P = 0.011, 24 between groups with different relative completion-time windows; all other tests were at P = 0.05. This scale can discriminate between groups of patients assumed to differ in their overall HRQOL, and is responsive to changes in health status.10,15,25–28 Models with and without the completion-time variables were fitted, and their AIC/BIC values were compared to identify the model with the best fit (smallest AIC/BIC value). A likelihood ratio test (LRT) was also car-ried out to complement the model fit using the AIC/BIC. Additional sensitivity analysis was carried out using completion-time windows reduced to 8 days29 and extended from 20 to 30 days to verify the stability of the model. All analyses were carried out with the Statistical Analysis Software.30

(9)

RESULTS

Clinical results of these studies have been reported previously.7–9 Briefly, for trial 1, there were no statistically significant differences in survival times between the reference group (group A) and the two experimental groups (groups B and C), and most HRQOL parameters were similar in all three groups. In trial 2, pro-gression-free survival was superior in the experimental group, but no HRQOL differences were seen between groups at any time except for financial problems at day 150 (11, P = 0.04) and day 200 (12, P = 0.03), and diarrhoea at day 150 (13, P = 0.04) all in favour of the reference group. For trial 3, neither survival nor HRQOL differed significantly among the three treatment groups.

The mean baseline scores for the different treatment groups within each of the three trials were similar. To assess whether patients from these trials were representative of patients with advanced NSCLC and metastatic CRC, baseline HRQOL scores were compared with reference values for patients with advanced NSCLC and recurrent/metastatic CRC from the EORTC QLQ-C30 reference sample of 1262 (NSCLC) and 653 (CRC) patients.31 The HRQOL mean scores of patients in these trials were similar to the reference values at baseline with small differences. Data for the trial 1 are shown in Table 1.

Compliance for trial 1 at baseline and throughout the active treatment period was >60% but decreased dramatically at cycle 6 and during follow-up. Compliance for trial 2 was low: 52% at baseline, decreasing to 17% after year 1. Compliance for trial 3 was also low. At baseline, questionnaires were collected from 54%, 56% and 70% of patients in groups A, B, and C, respectively, and grad-ually decreased over time to 21, 21, and 28% at year 1. There was no statistical significant difference in compliance rates for the global health status/QoL scale at the different assessment points between the treatment groups in any of the three trials. To assess the effect of the completion-time windows, the analyses were limited to six cycles.

(10)

Table 1. Baseline HRQOL scores for trial 1 (Smit et al.7) and reference data Reference data NSCLC Mean (SD) Group A Mean (SD) Group B Mean (SD) Group C Mean (SD) Global health status / QoL 58.8 (22.5) 55.0 (22.2) 59.4 (20.6) 57.8 (25.1) Physical Functioning 78.4 (19.3) 71.9 (23.9) 71.6 (24.4) 71.2 (24.0) Role Functioning 60.7 (33.1) 56.4 (23.9) 58.1 (31.5) 58.5 (33.7) Emotional Functioning 68.1 (24.2) 64.9 (23.6) 62.1 (24.3) 63.4 (25.1) Cognitive Functioning 84.0 (21.1) 81.7 (22.3) 83.1 (19.8) 84.9 (20.2) Social Functioning 73.6 (28.9) 69.8 (32.6) 76.6 (26.0) 72.3 (29.0) Fatigue 40.4 (27.0) 40.8 (26.3) 40.3 (27.7) 38.4 (27.5) Nausea / Vomiting 9.7 (18.3) 9.8 (17.56) 10.9 (18.1) 7.1 (13.5) Pain 29.7 (30.3) 32.9 (32.5) 34.5 (32.5) 38.1 (33.2) Dyspnoea 38.5 (31.7) 33.8 (28.5) 33.8 (31.7) 34.1 (33.6) Insomnia 32.4 (32.7) 31.4 (32.5) 33.1 (33.5) 40.2 (34.8) Appetite loss 27.9 (33.5) 26.8 (35.8) 28.8 (32.5) 29.6 (32.2) Constipation 17.4 (27.9) 14.1 (26.2) 11.4 (23.2) 13.7 (24.8) Diarrhoea 6.8 (17.4) 6.1 (17.4) 6.0 (14.1) 6.8 (17.8) Financial Problems 12.8 (25.8) 11.7 (25.6) 11.6 (26.4) 14.0 (28.9) Reference data: Scott et al.31

A total of 863 questionnaires were completed before the cycle date, 573 on the cycle date and 120 after the cycle date for trial 1. In trial 2, 381 question-naires were completed before the cycle date, 332 on the cycle date and 69 after the cycle date. Similarly, in trial 3, 173 questionnaires were completed before the cycle date, 286 on the cycle date and 82 after the cycle date. Some patients tend to stay in the same category with respect to completion-time window over the six cycles. The detail distributions of completed questionnaires in the given time frames are shown in supplementary Tables S1-S4, available at Annals of Oncology online. No statistically significant differences were observed in scores between the ‘before’ and ‘on’ comparisons in trials 1 and 2. However, statisti-cally significant differences (P < 0.05) were observed on six subscales (social functioning [effect size = 2.0], cognitive functioning [effect size = 3.2], fatigue [effect size = −2.1], nausea/vomiting [effect size = −3.5], appetite loss [effect size = −1.9] and constipation [effect size = −2.2] for ‘on’ and ‘after’ comparisons

(11)

in trial 1 and five subscales (social functioning [effect size = 2.0], global health status/QoL [effect size = 2.2], nausea/ vomiting [effect size = −2.2], appetite loss [effect size = −2.5] and fatigue [effect size = −2.5]) in trial 2. We thus formed two groups of patients: one in which questionnaires were completed ‘before-or-on’, and the other in which they were completed ‘after’. As for trial 3, statistical dif-ferences were observed in the ‘before’ to ‘on’ and the ‘on’ to ‘after’ comparisons. Figures 1–3 show the raw mean HRQOL profiles for the global health sta-tus/QoL scores based on a pooled analysis of all treatment groups together for the NSCLC trial, two-group CRC trial and three-group CRC trial, respectively, divided into ‘before-or-on’ and ‘after’ over the six cycles. The ‘before-or-on’ mean scores seem to be higher at most time points in the NSCLC and two-group CRC trials whereas in the three-two-group CRC trial the mean profiles depict a fluctuating pattern.

Figure 1. Raw mean profiles for the global health status/QoL scale for trial 1 by ‘before-or-on’ and

(12)

Figure 2. Raw mean profiles for the global health status/QoL scale for trial 2 by ‘before-or-on’ and

‘after’.

Figure 3. Raw mean profiles for the global health status/QoL scale for trial 3 by ‘before’, ‘on’ and

(13)

We observed the following statistically significant differences in the esti-mated HRQOL mean values between ‘before-or-on’ and ‘after’ completions in trial 1: compared with patients assessed ‘before-or-on’ treatment, those assessed ‘after’ treatment had worse social functioning (−4, P = 0.02), cognitive functioning (−5, P = 0.0001), fatigue (4, P = 0.03), nausea/vomiting (6, P = 0.001), appetite loss (5, P = 0.04) and constipation (5, P = 0.02). Similarly, in trial 2, patients assessed ‘after’ treatment had worse social functioning (−6, P = 0.05), global health status/QoL (−6, P = 0.03), nausea/vomiting (5, P = 0.03), appetite loss (7, P = 0.03) and fatigue (8, P = 0.01) than those assessed ‘before-or-on’ treatment. These results were driven largely by the large difference at cycle 4, as shown by Figure 2. For trial 3, only within group C were significant (P < 0.05) HRQOL mean score differences reported. Compared with HRQOL mean scores ‘on’ the scheduled cycle data, the ‘before’ HRQOL mean scores were worse for physical functioning (3; P = 0.0003) and fatigue (6; P = 0.01) and the ‘after’ HRQOL mean scores were worse for nausea and vomiting (3; P = 0.03) and physical functioning (2; P = 0.01). These results confirm those in earlier studies of patients with lung cancer and CRC.17, 18 All of the mean HRQOL estimates were below the 10 points difference generally accepted as clinically meaningful although some of the scores were within the range of 5–10.20

The performance of models with and without the completion-time window for the global health status scale is shown in Table 2. From this table, we see that the estimated treatment effects on global health status/QoL in each of the trials remain largely unchanged by including or excluding the completion-time window variable. We found that there were no associations between comple-tion-time window and treatment groups. Owing to the lack of longitudinal trends based on completion-time windows, we carried out a cross-sectional analy-sis32, 33 to verify the effect of cross-sectional differences between the treatment groups with respect to global health status/QoL score at each individual cycle using a Wilcoxon test. We considered a completion-time window of 5, 8 and 10 days for the analysis. There were no significant cross-sectional differences between treatment groups on global health status/QoL in all the three trials at each individual cycle except for trial 2 where there was a significant difference between treatment groups at cycle 2 (P = 0.02).

(14)

Table 2. Parameter estimates with SEs for the three trials for global health

sta-tus/QoL scale

Parameters Model with

treatment, cycle, treatment-by-cycle interaction and completion-time Model with treatment, cycle and treatment-by-cycle interaction Parameter Estimate SE* Parameter Estimate SE* Trial 1 Treatment B 3.4 3.4 3.4 3.4 Treatment C –1.8 3.7 –1.9 3.7 Cycle 1 –3.7 2.5 –3.6 2.5 Cycle 2 3.4 2.5 3.4 2.6 Cycle 3 5.7 2.6 5.7 2.6 Cycle 4 4.8 2.6 4.7 2.6 Cycle 5 3.7 2.4 3.6 2.4

Interaction between Cycle 1 and treatment B 0.8 3.5 0.7 3.5 Interaction between Cycle 1 and treatment C 5.3 3.8 5.2 3.8 Interaction between Cycle 2 and treatment B –3.4 3.5 –3.5 3.6 Interaction between Cycle 2 and treatment C 4.1 3.9 4.0 3.9 Interaction between Cycle 3 and treatment B –4.5 3.6 –4.5 3.6 Interaction between Cycle 3 and treatment C –1.9 4.0 –2.0 4.0 Interaction between Cycle 4 and treatment B –3.4 3.6 –3.3 3.6 Interaction between Cycle 4 and treatment C –2.0 4.0 –1.8 4.0 Interaction between Cycle 5 and treatment B –4.8 3.4 –4.8 3.4 Interaction between Cycle 5 and treatment C –3.0 3.7 –3.0 3.7

Completion-time 2.8 1.5 –2 Log Likelihood 12895.9 12901.9 AIC 12901.9 12907.9 BIC 12914.2 12920.2 Trial 2 Treatment 2.0 7.0 2.0 7.0

(15)

Cycle 1 –9.6 5.4 –10.0 5.4

Cycle 2 –1.9 5.6 –2.4 5.6

Cycle 3 0.4 5.6 –0.3 5.7

Cycle 4 –2.5 5.8 –2.9 5.8

Cycle 5 –1.8 6.2 –2.8 6.2

Interaction between Cycle 1 and treatment –2.8 7.1 –2.7 7.2 Interaction between Cycle 2 and treatment –7.9 7.4 –7.9 7.4 Interaction between Cycle 3 and treatment –5.4 7.4 –4.9 7.5 Interaction between Cycle 4 and treatment –1.5 7.8 –0.9 7.8 Interaction between Cycle 5 and treatment –2.6 8.2 –2.0 8.2

Completion-time 5.5 2.6 –2 Log Likelihood 6734.8 6743.0 AIC 6740.8 6749.0 BIC 6752.4 6760.6 Trial 3 Treatment FUFACI –7.0 16.7 –7.3 16.7 Treatment FUFAB 1.7 10.9 2.1 10.9 Cycle 1 –4.9 10.8 –5.6 10.8 Cycle 2 –4.0 10.7 –4.4 10.7 Cycle 3 –3.3 10.7 –3.5 10.7 Cycle 4 –10.0 11.1 –10.2 11.1 Cycle 5 –5.1 11.9 –4.8 11.9

Interaction between Cycle 1 and FUFACI –0.5 17.5 –0.3 17.5 Interaction between Cycle 1 and FUFAB –2.7 11.7 –3.0 11.7 Interaction between Cycle 2 and FUFACI 3.8 17.2 4.7 17.2 Interaction between Cycle 2 and FUFAB –4.2 11.3 –4.1 11.3 Interaction between Cycle 3 and FUFACI 1.5 17.3 3.0 17.3 Interaction between Cycle 3 and FUFAB 2.5 11.4 2.4 11.4 Interaction between Cycle 4 and FUFACI 17.3 17.4 18.7 17.4 Interaction between Cycle 4 and FUFAB 7.8 11.7 8.0 11.7 Interaction between Cycle 5 and FUFACI 8.8 19.3 9.5 19.3

(16)

Interaction between Cycle 5 and FUFAB 2.5 12.4 1.7 12.4

Completion-time 3.0 2.0

–2 Log Likelihood 4593.9 4599.5

AIC 4599.9 4605.5

BIC 4610.7 4616.3

SE, standard error.

All the models including the completion-time window variable had lower AIC/BIC values, implying a better fit to the data. Similar results were observed from the likelihood ratio test statistics when comparing the reduced (without completion-time variable) and the full (including completion-time variable) models. Results from the sensitivity analyses were similar to those of the main analysis (data not shown).

(17)

DISCUSSION

Our main aim was to evaluate if scores on the EORTC QLQC30 are affected by the specific time point at which the questionnaire was completed, and whether this could bias the treatment comparisons. We found that HRQOL scores were sta-tistically significantly different between ‘before-or-on’ and ‘after’ chemotherapy administration for several domains in all the three chemotherapy studies. Worse outcomes were reported when collected after the start of the cycle for nausea and vomiting in all three studies; and in two of the three studies, fatigue, appetite loss and social functioning all worsened. In one of the studies, we also showed that HRQOL scores ‘on’ the scheduled cycle date differed from both ‘before’ and ‘after’ completions, with worse physical functioning and fatigue seen ‘before’ treatment. Although all of the ‘after’ treatment differences were in the ‘worse’ direction, they were less than the 10 points generally accepted as clinically meaningful. However, some scores may have represented small, potentially noticeable changes in the range of 5–1020 which could possibly be important to the individual patient and warrant clinical attention. Also, by averaging over sev-eral cycles, cross-sectional differences (such as at cycle 4 in Figure 2) might be reduced, but this cannot be formally tested in this study, due to the multiplicity of cross-sectional testing on limited amount of data.

Adding completion-time windows to longitudinal mixed models, as com-monly used in analysing HRQOL data from randomised controlled trials, pro-duced a better fit, with lower AIC/BIC values and slightly smaller standard errors. However, this improved fit did not substantially alter the treatment estimates in any of the three trials. The effect of cross-sectional differences between the treatment groups on global health status/QoL in all the three trials at each indi-vidual cycle was not significant except for trial 2 where there was a significant difference between treatment groups at cycle 2. Hence, adjusting for the com-pletion-time windows does not seem to be required for evaluating treatment effects in these settings. The results from LRT support the model fit findings from the AIC/BIC values.

This study had several limitations. HRQOL data were obtained only for patients who did not progress, implying that the sample reflects patients with good health status, who experienced fewer symptoms than patients with

(18)

pro-gressive disease. Such missing data, which are common in HRQOL studies, may have biased our findings. There is no standard method to deal with missing data; however, modern approaches do exist, such as maximum likelihood and multiple imputations. Another limitation is that our ability to extrapolate these findings to the general cancer population is limited. Also, we could not use the exact time point in the longitudinal model since it was not uncommon for some patients not to complete the questionnaires on the exact planned date. Instead, we used the cycle number with fixed time points but with the disadvantage that it might not really reflect the exact time the patient completed the question-naires due to delays by some patients. Baseline scores for all three trials were very similar to reference data, suggesting that the findings from our trials are generally representative of patients with advanced NSCLC and recurrent/meta-static CRC. However, on-treatment scores and the effect of time windows may differ in other treatment settings (e.g. different chemotherapy regimens, radio-therapy, surgery), ultimately even affecting treatment effect estimation.

In conclusion, allowing completion-time windows in cancer clinical trials can improve the validity of the result by allowing inclusion of an increased pro-portion of HRQOL questionnaires and improving goodness-of-fit. However, we have shown that the exact timing of completion within such windows can result in statistical significant differences. While the magnitude of the differences we observed fell below commonly accepted standards for clinical significance, even small differences may be perceptible and important to some patients. Accounting for this information did not alter treatment comparisons in the trials we examined, but might do so in different settings. Thus, we recommend that the possible effects of the timing of completion within time windows should be further examined in future cancer clinical trials before overall conclusions about the need to consider this factor in analysis can be drawn. In particular, exam-ination of the effects of smaller time windows may be informative, as well as assessing effects in trials where there were differences between the treatment groups.

(19)

REFERENCE

1. Fairclough DL. Design and Analysis of Quality of Life Studies in the Clinical Trials. London: Chapman and hall/CRC 2002.

2. Osoba D. Rationale for the timing of health-related quality-of-life (HQL) assessments in oncological palliative therapy. Cancer Treat Rev 1996; 22: 69–73.

3. Klee MC, King MT, Machin D et al. A clinical model for quality of life assess-mentin cancer patients receiving chemotherapy. Ann Oncol 2000; 11(1): 23–30.

4. Hopwood P, Stephens RJ, Machin D. Approaches to the analysis of quality of lifedata: experiences gained from a medical research council lung can-cer working party palliative chemotherapy trial. Qual Life Res. 1994; 3(5): 339–352.

5. Hürny C, Bernhard J, Coates A et al. The International Breast Cancer Study Group: timing of baseline quality of life assessment in an international adju-vant breast cancer trial: its effect on patient self-estimation. Ann Oncol 1994; 5: 65–74.

6. Pater J, Osoba D, Zee B et al. Effects of altering the time of administration andthe time frame of quality of life assessments in clinical trials: an example using the EORTC QLQ-C30 in a large anti-emetic trial. Qual Life Res 1998; 7: 273–278.

7. Smit EF, van Meerbeeck JPAM, Lianes P et al. Three-arm randomised study oftwo cisplatin-based regimens and paclitaxel plus gemcitabine in advanced nonsmall cell lung cancer: a phase III trial of the European Organization for Research and Treatment of Cancer Lung Cancer Group-EORTC 08975. J Clin Oncol 2003; 21: 3909–3917.

8. Köhne C-H, van Cutsem E, Wils J et al Phase III study of weekly high-dosein-fusional fluorouracil plus folinic acid with or without irinotecan in patients with metastatic colorectal cancer: European Organisation for Research and Treatment of Cancer Gastrointestinal Group Study 40986. J Clin Oncol 2005; 23(22): 4856–4865.

9. Köhne CH, Wils J, Lorenz M et al Randomised phase III study of high-dose-fluorouracil given as a weekly 24-hour infusion with or without leucovorin

(20)

versus bolus fluorouracil plus leucovorin in advanced colorectal cancer: European Organization of Research and Treatment of Cancer Gastrointestinal Group Study 40952. J Clin Oncol 2003; 21(20): 3721–3728.

10. Aaronson NK, Ahmedzai S, Bullinger M et al. The EORTC core quali-ty-of-lifequestionnaire: interim results of an international field study. In Osoba D (ed), Effect of Cancer on Quality of Life. Boston: CRC Press 1991; 185–203.

11. Aaronson NK, Ahmedzai S, Bergman B 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. J Natl Cancer Inst 1993; 85: 365–376.

12. Groenvold M, Klee MC, Sprangers MA et al. Validation of the EORTC QLQ-C30 quality of life questionnaire through combined qualitative and quantita-tive assessment of patient-observer agreement. J Clin Epidemiol 1997; 50: 441–450.

13. Hjermstad MJ, Fossa SD, Bjordal K et al. Test/retest study of the EuropeanOrganization for Research and Treatment of Cancer Core Quality-of-Life Questionnaire. J Clin Oncol 1995; 13: 1249–1254.

14. Bjordal K, Kaasa S. Psychometric validation of the EORTC core quality of lifequestionnaire, 30-item version and a diagnosis-specific module for head and neck cancer patients. Acta Oncol 1992; 31: 311–321.

15. Ringdal GI, Ringdal K. Testing the EORTC Quality of Life Questionnaire on cancerpatients with heterogeneous diagnoses. Quality Life Res 1993; 2: 129–140.

16. Fayers PM, Aaronson NK, Bjordal K et al. on behalf of the EORTC Quality of LifeGroup. EORTC QLQ-C30 Scoring Manual, 3rd edition. Brussels, EORTC 2001.

17. Osoba D, Zee B, Pater J et al Psychometric properties and responsiveness of theEORTC quality of Life Questionnaire (QLQ-C30) in patients with breast, ovarian and lung cancer. Qual Life Res 1994; 3(5): 353–364.

18. Uwer L, Rotonda C, Guillemin F et al. Responsiveness of EORTC QLQ-C30, QLQCR38 and FACT-C quality of life questionnaires in patients with colorec-tal cancer. Health Qual Life Outcomes 2011; 9: 70.

(21)

19. Osoba D, Rodrigues G, Myles J et al. Interpreting the significance of changes in health-related quality-of-life scores. J Clin Oncol 1998; 16: 139–144. 20. Ringash J, Redelmeier DA, O’Sullivan B et al Quality of life and utility in

irra-diated laryngeal cancer patients. Int J Radiat Oncol Biol Phys 2000; 47(4): 875–881.

21. Verbeke, G, Molenberghs G. Linear Mixed Models for Longitudinal Data. New York: Springer-Verlag 2000.

22. Akaike H. “Information theory and an extension of the maximum likelihood principle”. In Petrov BN, Csaki F (eds), Second International Symposium on Information Theory. Budapest: Akademiai Kiado 1973; 267–281.

23. Schwarz, G. “Estimating the dimension of a model”. Ann Stat 1978; 6: 461–464.

24. Bottomley A, Therasse P, Piccart M et al. Health-related quality of life in sur-vivorsof locally advanced breast cancer: an international randomised con-trolled phase III trial. Lancet Oncol 2005; 6: 287–294.

25. Fayers P, Aaronson N, Bjordal K et al. on behalf of EORTC Quality of Life StudyGroup. EORTC QLQ-C30 Scoring Manual. EORTC Study Group on Quality of Life. Brussels, Belgium: EORTC Data Centre 1995.

26. Bergman B, Sullivan M, Sörenson S. Quality of life during chemotherapy forsmall cell lung cancer. I. An evaluation with generic health measures. Acta Oncol 1991; 30: 947–957.

27. Sigurdadóttir V, Bolund C, Sullivan M. Quality of life evaluation by the EORTCquestionnaire technique in patients with generalized malignant mel-anoma on chemotherapy. Acta Oncol 1996; 35: 149–159.

28. Curran D, Fossa S, Aaronson N et al. Baseline quality of life of patients with-advanced prostate cancer. Eur J Cancer 1997; 33: 1809–1814.

29. Ballatori E, Roila F. Impact of nausea and vomiting on quality of life in can-cerpatients during chemotherapy. Health Qual Life Outcomes 2003; 1: 46. 30. SAS Institute Inc. 2010. SAS/GRAPH® 9.2 Reference, 2nd Edition. Cary, NC:

SAS Institute Inc.

31. Scott NW, Fayers P, Bottomley A et al. EORTC QLQ-C30 Reference ValuesManual. Brussels, Belgium: EORTC Quality of Life Group Publications 2008.

(22)

32. Fairclough DL. Summary measures and statistics for comparison of quality of lifein a clinical trial of cancer therapy. Stat Med 1997; 15: 1197–1209. 33. Curran D, Aaronson N, Standaert B et al. Summary measures and statistics

inthe analysis of quality of life data: an example from an EORTCNCIC-SAKK locally advanced breast cancer study. Eur J Cancer 2000; 36: 834–844.

(23)

SUPPLEMENTARY DATA

S1. Protocol summary of the three trials

Trial 1 Trial 2 Trial 3

Reference Smit et al (2003)7 Köhne et al (2005)8 Köhne et al (2003)9

Cancer type NSCLC Colorectal Colorectal Stage Locally advance

and/or metastatic metastatic colorectal metastatic colorectal Patients randomised Intervention Yes N=150 N=150 N=150 Yes N=215 N=215 Yes N=159 N=159 N=159 Regimen Intervention • Cisplatin • Gemcitabine hydrochloride • Paclitaxel • FOLFIRI • Fluorouracil • Irinotecan hydrochloride • Leucovorin calcium • Fluorouracil • Leucovorin calcium

Brief title Randomised study with new combination chemotherapies in advanced NSCLC

CPT-11 in combination with weekly 24 hour infusion 5-fu plus folinic acid relative to weekly 24 hour infusion 5-fu plus folinic acid alone in patients with advanced colorectal cancer

Phase III randomised study of weekly, 24-hour, high-dose 5-fu with vs without cf vs bolus 5-fu/ cf for advanced colorectal cancer

Brief summary RATIONALE: Drugs used in chemotherapy use different ways to stop tumor cells from dividing so they stop growing or die. Combining more than one drug may kill more tumor cells. It is not yet known which combination chemotherapy regimen is more effective in treating advanced non-small cell lung cancer.

RATIONALE: Drugs used in chemotherapy use different ways to stop tumor cells from dividing so they stop growing or die. It is not yet known whether fluorouracil and leucovorin plus irinotecan is more effective than fluorouracil and leucovorin alone for colorectal cancer.

(24)

PURPOSE:

Randomised phase III trial to compare the effectiveness of three different combination chemotherapy regimens in treating patients who have advanced non-small cell lung cancer.

PURPOSE: Randomised phase III trial to compare the effectiveness of fluorouracil and leucovorin with or without irinotecan in treating patients who have metastatic colorectal cancer.

PURPOSE:

I. Assess the survival, quality of life, and time to progression associated with weekly high-dose fluorouracil (5-FU) given by 24-hour infusion with vs. without leucovorin (CF) vs. bolus 5-FU with low-dose CF in patients with advanced colorectal cancer. II. Assess the response rates produced by these 3 treatments. III. Evaluate the toxicity and cost effectiveness of these 3 treatments. Cycles Planned Completed 66 99 99 Timing of HRQOL Baseline During treatment Follow-up Prior to randomisation Before each cycle

Prior to randomisation Before each cycle

Prior to randomisation Before each cycle

Compliance (HRQOL) Baseline During treatment Follow-up 80.2% 61.6% 25.0% 52% 34% -61% 24% -Endpoints Primary HRQOL as secondary Overall survival Yes Progression free survival Yes Overall survival Yes Missing data

(HRQOL) Missing HRQOL data were ignored during the analysis

Missing HRQOL data were ignored during the analysis

Missing HRQOL data were ignored during the analysis Protocol Ids from

National Cancer Institute (NCI) CDR0000066658 EORTC-08975, NCT00003589 CDR0000067560 EORTC-40986, NCT00004885 EORTC-40952 GER-AIO-01/95, EU-95049

(25)

S2. Number of completed HRQOL questionnaire in given time window (%, trial 1)

Cycle Number of forms completed Before cycle date Number of forms completed On cycle date Number of forms completed After cycle date 1 284 (70.5) 100 (24.8) 19 (4.7) 2 178 (54.6) 116 (35.6) 32 (9.8) 3 129 (50.8) 107 (42.1) 18 (7.1) 4 115 (49.6) 99 (42.7) 18 (7.8) 5 85 (45.5) 82 (43.9) 20 (10.7) 6 72 (46.8) 69 (44.8) 13 (8.4) Row total 863 (55.5) 573 (36.8) 120 (7.7)

S3: Number of completed HRQOL questionnaire in given time window (%, trial 2)

Cycle Number of forms completed Before cycle date Number of forms completed On cycle date Number of forms completed After cycle date 1 154 (46.8) 148 (45.0) 27 (8.2) 2 87 (52.4) 60 (36.1) 19 (11.5) 3 61 (44.5) 62 (45.3) 14 (10.2) 4 39 (50.7) 34 (44.2) 4 (5.2) 5 22 (52.4) 15 (35.7) 5 (11.9) 6 18 (58.1) 13 (41.9) -Row total 381 (48.7) 332 (42.5) 69 (8.8)

S4. Number of completed HRQOL questionnaire in given time window (%, trial 3)

Cycle Number of forms completed Before cycle date Number of forms completed On cycle date Number of forms completed After cycle date 1 57 (52.8) 40 (37.0) 11 (10.2) 2 50 (30.5) 88 (53.7) 26 (15.9) 3 28 (25.5) 58 (52.7) 24 (21.8) 4 16 (20.5) 50 (64.1) 12 (15.4) 5 13 (28.9) 27 (60.0) 5 (11.1) 6 9 (25.0) 23 (63.9) 4 (11.1) Row total 173 (32.0) 286 (52.9) 82 (15.2)

Referenties

GERELATEERDE DOCUMENTEN

The primary aims of this population- based, observational study were to (a) investigate the associa- tion between the novel European Organisation for Research and Treatment of

The following questionnaires were used: the ‘EORTC core quality of life questionnaire’ (QLQ-C30), ‘International Index of Erectile Function 15’ (IIEF-15; containing five areas:

(‘Perceived Stress Scale’/de OR ‘Insomnia Severity Index’/de OR ‘International Index of Erectile Function’/de OR ((cancer NEAR/3 worr* NEAR/3 scale*) OR (patient NEAR/3

The European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30) (version 3.0) was used to assess HRQoL in cancer patients [17, 18]..

ANOVAs were conducted to assess differences in EORTC QLQ-C30 scores by sex, by age groups and sex and by self-reported morbidities in (1) mean EORTC baseline sub- scale scores; (2)

CPILS: Cancer Problems in Living Scale; EORTC: European Organisation for Research and Treatment of Cancer; FACT-G: Functional Assessment of Cancer Therapy – General;

CVZ: Dutch Health Care Insurance Board; eHIQ: e-Health Impact Questionnaire; EORTC QLQ-C30/BR23/CR29/H&amp;N43/HL27/NHL-HG29/NHL-LG20: 30-item core European Organisation for

BEP: Bleomycin, etoposide, and cisplatin; EORTC: The European Organisation for Research and Treatment of Cancer; ES: Effect size; HRQoL: Health-related quality of life; IOCv2: