Skip to main content

Impact of early tumor shrinkage on quality of life in patients treated with first-line cetuximab plus chemotherapy for unresectable metastatic colorectal cancer: results of Phase II QUACK trial

Abstract

Purpose

Although early tumor shrinkage (ETS) is a predictor of improved overall survival (OS), the association between ETS and health-related quality of life (HRQOL) remains unclear for patients with metastatic colorectal cancer (mCRC) treated with first-line cetuximab plus chemotherapy.

Methods

The data were collected from a prospective trial that assessed HRQOL using the EORTC QLQ-C30. The impact of ETS on HRQOL was estimated using a linear mixed-effects model for repeated measures.

Results

ETS was achieved in 82 (64.1%) of 128 mCRC patients treated with first-line cetuximab plus chemotherapy, and these patients had a significantly longer OS than those without ETS (HR, 0.38; 95% CI, 0.20–0.72; P = .002). Asymptomatic patients with ETS had a favorable OS, while symptomatic patients without ETS had a worse OS (2-year OS rates, 77.8% vs. 42.5%). Symptomatic patients with ETS had similar outcomes as asymptomatic patients without ETS (2-year OS rates, 64.1% vs. 67.0%). For symptomatic patients, ETS was associated with improved HRQOL scores between baseline and 8 weeks: the mean changes for patients with and without ETS were 5.86 and -4.94 for global health status (GHS)/QOL, 26.73 and 3.79 for physical functioning, and 13.58 and -3.10 for social functioning, respectively. The improved HRQOL was comparable to that of asymptomatic patients without ETS. For asymptomatic patients, ETS showed a decreased deterioration in HRQOL.

Conclusion

Our findings highlight the importance of ETS for HRQOL and prognostic estimates, and assessing ETS may provide clinically useful information for physicians and patients to make more informed decisions.

Peer Review reports

Introduction

Colorectal cancer (CRC) risk is predominantly driven by environmental factors [1]. CRC has the third highest incidence among cancers and is the second leading cause of cancer-related deaths worldwide; thus, it remains a major clinical challenge [2]. Almost one-fourth of CRC patients have metastatic disease at the time of diagnosis [3, 4]. Despite advances in the treatment of patients with metastatic CRC (mCRC), treatment at this stage is essentially palliative [4, 5]. Accordingly, in addition to the prevention of tumor progression and the prolongation of survival, both improving tumor-related symptoms and maintaining health-related quality of life (HRQOL) should be taken into consideration when planning treatment [5]. In fact, cancer patients tend to request more information on not only survival estimates but also HRQOL [6]. Therefore, an assessment of the impacts of both the treatment and disease on HRQOL is urgently needed.

HRQOL comprises mental, physical, and social well-being, all of which are affected by the tumor burden and/or adverse events [7, 8]. It is difficult for physicians to accurately determine how a patient feels or functions with respect to a health condition because of their dependence on the patient’s perception [9,10,11]. In fact, the U.S. The Food and Drug Administration stated that some treatment effects are known only to the patient, and such information can be lost when the patient’s perspective is filtered through a physician’s evaluation of the patient’s responses to clinical interview questions [12]. A patient-reported outcome (PRO) is defined as any report about a health condition and treatment obtained directly from the patient, without interpretation of the patient’s response by a physician or anyone else, via standardized questionnaires designed to measure a conceptual framework of HRQOL, including symptoms, satisfaction, or functioning [12, 13]. Thus, PROs are an umbrella term encompassing patient self-reported outcomes related to a patient’s health status and perceptions, and PROs can bridge the considerable gap in reported HRQOL between patients and physicians [9], representing an effective approach to improve the quality of care for patients.

Cetuximab, an antibody targeting epidermal growth factor receptor (EGFR), has been demonstrated to have promising efficacy when combined with chemotherapy as a first-line treatment for left-sided and RAS wild-type mCRC [14,15,16,17]. Of note, there is a growing body of evidence demonstrating that cetuximab plus chemotherapy more effectively promotes early tumor shrinkage (ETS) than chemotherapy alone or bevacizumab plus chemotherapy and that ETS is associated with long-term OS, possibly by achieving a maximal depth of response [16, 18]. As HRQOL is a major concern for mCRC patients because their tumor burden often results in them being symptomatic at the time of diagnosis [19, 20], ETS may rapidly improve HRQOL for symptomatic patients at baseline. However, the clinical impact of ETS on HRQOL has not yet been determined in mCRC patients treated with first-line cetuximab plus chemotherapy.

The QUACK study was prospectively performed to assess the HRQOL of mCRC patients treated with cetuximab plus chemotherapy using a PRO assessment tool [9, 19, 21]. The aim of the present study was to assess the association of ETS with HRQOL and prognostic outcomes according to the baseline symptom status by performing a post hoc analysis of the QUACK trial data. Our findings will provide additional relevant information that may help both patients and physicians make more informed clinical decisions.

Patients and Methods

Study design and treatment

The QUACK study was a prospective, multicenter, phase II study that assessed the associations of QOL with adverse events and treatment efficacy for mCRC patients treated with first-line cetuximab plus standard chemotherapy (FOLFOX or FOLFIRI). Detailed information on the study design has been provided previously [22]. In total, 149 patients with KRAS wild-type mCRC were enrolled from 49 institutions between July 2013 and April 2015, of which 140 patients received cetuximab plus chemotherapy at least once. The chemotherapy regimen (FOLFOX or FOLFIRI) was selected according to the treating physician’s discretion and institutional guidelines. The primary results have already been published elsewhere [21].

This study was registered with the University Hospital Medical Information Network (UMIN) Clinical Trial Registry (UMIN000010985) on July 19, 2013 and was conducted in accordance with the Declaration of Helsinki and the Ethics Guidelines for Clinical Research issued by the Ministry of Health, Labor, and Welfare in Japan. The study protocol was approved by the institutional review board, and written informed consent was obtained from all patients before registration.

Treatment efficacy

Radiologic assessments were performed using computed tomography at baseline and every 8 weeks during the treatment period. The investigator at each institution assessed the tumor response based on the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. The overall response rate (ORR) was calculated as the proportion of patients with a complete response or a partial response according to the RECIST criteria. ETS was defined as a relative reduction of ≥ 20% in the sum of the longest diameters of target lesions between the values at baseline and 8 weeks [18]. The ETS cutoff value of ≥ 20% was determined in a time-dependent receiver operating characteristic curve analysis [18], and the 8-week time point was expected to minimize the influence of early study termination due to the first radiologic assessment.

Progression-free survival (PFS) was defined as the time interval between registration and the date of tumor progression or death. Time to treatment failure (TTF) was defined as the time interval between registration and the date of treatment discontinuation for any reason, including treatment toxicity, tumor progression, patient withdrawal, or death. Overall survival (OS) was defined as the time from registration to the date of death from any cause.

HRQOL and symptom assessments

Because the QUACK study was specifically designed to assess the associations of HRQOL with adverse events and treatment efficacy, HRQOL assessments were typically performed at baseline and after 2, 4, 8, 16, and 24 weeks. The survey sheets were collected at registration and after 4, 8, 16, and 24 weeks, and these surveys also included the patients’ assessments of safety, compliance with treatment, and treatment efficacy.

HRQOL was evaluated using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire C30 (EORTC QLQ-C30) version 3.0, which is a valid and reliable PRO instrument for assessing HRQOL in cancer settings [23, 24]. The EORTC QLQ-C30 questionnaire is composed of both single- and multi-item scales, including a global health status (GHS)/QOL scale, five functional scales (role, physical, cognitive, emotional, and social), and eight symptom scales (appetite loss, pain, diarrhea, constipation, dyspnea, fatigue, insomnia, and nausea/vomiting) [23]. The observed raw data were standardized through a linear transformation, and the scores ranged from 0 to 100, with a higher score indicating better levels of GHS/QOL and functioning [25]. The symptom scales have four response categories for each question (“very much,” “quite a bit,” “a little,” and “not at all”). Patients were defined as symptomatic when they reported “very much” or “quite a bit” to at least one of the symptom questions at baseline and as asymptomatic when they reported only “a little” or “not at all” to all eight symptom scales [21, 26].

Statistical analysis

All analyses performed in this study included eligible patients who underwent any intervention after registration and who answered the HRQOL questionnaire at both baseline and at least once postbaseline. To assess the impact of ETS on HRQOL, the association of ETS with changes in the EORTC QLQ-C30 scores from baseline throughout the observation period of 24 weeks was analyzed using a linear mixed-effects model for repeated measures, with the intercept and slope for the study week treated as random effects to estimate the least squares means of the change from baseline. A statistical analysis was also performed to evaluate the association between ETS and HRQOL according to the baseline symptom status.

The distribution of the prognostic outcomes was estimated using the Kaplan–Meier method, and the log-rank test was used to compare the distribution between the populations. The Cox proportional hazard model was used to analyze the association between ETS and the time-to-event endpoints, for which the adjusted hazard ratios (HRs) and the 95% confidence intervals (CIs) were calculated.

The continuous data variables were expressed as the mean ± the standard error of the mean (SEM), and they were compared using a two-tailed Student’s t test. Fisher’s exact test was used for categorical variables. The JMP 14 software package (SAS Institute, Cary, NC, USA) was used to conduct all statistical analyses.

Results

Clinicopathological characteristics associated with ets following cetuximab plus chemotherapy

This study analyzed a dataset collected from 128 of 140 mCRC patients treated with first-line cetuximab plus chemotherapy in the prospective QUACK study (Table 1). The median age was 66 years (range, 27–89 years), 87 (68.0%) were male, and 107 (83.6%) had an Eastern Cooperative Oncology Group Performance Status (ECOG PS) of 0. At baseline, 51 patients (39.8%) were symptomatic, reporting “very much” or “quite a bit” for at least one of the eight symptom items (i.e., appetite loss, pain, diarrhea, constipation, dyspnea, fatigue, insomnia, and nausea/vomiting) on the EORTC QLQ-C30 questionnaire. Although ETS may depend on the location and size of metastases [27], ETS was achieved in 82 (64.1%) of 128 patients treated with first-line cetuximab plus chemotherapy (Fig. 1A), consistent with previous reports, including an ETS rate of 61.5–80% [16, 18, 28,29,30,31]. Baseline clinicopathological characteristics were well balanced between patients with and without ETS, except for differentiation (Table 1). ETS was associated with a higher ORR (76.8% vs. 21.7%; P < .001). The rate of surgical resection of metastases was significantly higher among patients with ETS than among those without ETS (28.6% vs. 11.4%; P = .031).

Table 1 Correlation of clinicopathologic characteristics with ETS in 128 metastatic colorectal cancer patients
Fig. 1
figure 1

Association between ETS and prognostic outcomes in 128 mCRC patients treated with cetuximab plus chemotherapy. A) Waterfall plots of individual patient tumor shrinkage at 8 weeks from the initiation of treatment. B) Kaplan–Meier curves of PFS according to ETS status. C) Kaplan–Meier curves of OS according to ETS status

Association of ETS with prognosis according to the symptomatic status at baseline

At the data cutoff point on April 20, 2016, 103 and 39 events were observed in relation to PFS and OS, respectively. The median follow-up time was 18.1 months (95% CI, 17.2–19.7), and the median PFS was 10.8 months (95% CI, 9.6–12.1). The median OS was not reached at the data cutoff point, and the 2-year estimated OS rate was 66.7%. Patients with ETS had a significantly longer PFS than those without ETS: the median PFS values were 12.2 months for patients with ETS and 6.9 months for those without ETS (log-rank test, P = .001, HR, 0.52; 95% CI, 0.35–0.78; Fig. 1B). Achieving ETS was also associated with a better OS (2-year OS rates, 73.5% vs. 54.1%; log-rank test, P = .002; HR, 0.38; 95% CI, 0.20–0.72; Fig. 1C) and TTF (median TTF, 7.7 vs. 4.1 months; log-rank test, P < .001; HR, 0.48; 95% CI, 0.33–0.70; Fig. S1). In addition, higher ETS values were significantly associated with longer times for PFS, OS, and TTF when ETS was analyzed as a continuous variable (i.e., the percentage of tumor shrinkage at 8 weeks) (Fig. S2).

We previously found that the presence of baseline patient-reported symptoms, as an independent predictor, was associated with a significantly worse OS in mCRC patients treated with cetuximab plus chemotherapy [19]. Because ETS was associated with a better OS, the association of ETS with prognostic outcomes based on the baseline symptom status was evaluated. ETS had a significant impact on PFS irrespective of the baseline symptom status: the median PFS was 13.0 months for symptomatic patients with ETS and 11.7 months for asymptomatic patients with ETS, while it was 5.5 months for symptomatic patients without ETS and 7.4 months for asymptomatic patients without ETS. Compared with symptomatic patients without ETS as a reference, the HRs were 0.36 (95% CI: 0.19–0.67) for symptomatic patients with ETS, 0.43 (95% CI: 0.26–0.72) for asymptomatic patients with ETS, and 0.61 (95% CI: 0.32–1.16) for asymptomatic patients without ETS (Fig. 2A). Next, survival estimates of OS were determined according to the baseline symptom status (Fig. 2B). In terms of the 2-year OS rates, asymptomatic patients with ETS had the most favorable outcomes (77.8%), while symptomatic patients without ETS had the worst outcomes (42.5%). Symptomatic patients who achieved ETS had similar prognostic characteristics as asymptomatic patients without ETS (64.1% vs. 67.0%). Compared with symptomatic patients who did not achieve ETS as a reference, the HRs were 0.18 (95% CI: 0.08–0.42) for asymptomatic patients with ETS, 0.40 (95% CI: 0.17–0.95) for symptomatic patients with ETS, and 0.39 (95% CI: 0.16–0.99) for asymptomatic patients without ETS.

Fig. 2
figure 2

Kaplan–Meier curves of A) PFS and B) OS according to ETS status based on symptoms reported by patients at baseline using the symptom scales of the EORTC QLQ-C30 questionnaire. Compared with symptomatic patients without ETS as a reference, the HR and 95% CI were calculated for each population

Association between ETS and HRQOL according to the symptomatic status at baseline

Although GHS/QOL and five functional (physical, social, emotional, role, and cognitive) scales are included among the HRQOL scales in the EORTC QLQ-C30 questionnaire, the association between ETS and each scale remains unclear. The impact of ETS on the changes in HRQOL scores from baseline was estimated using a linear mixed-effects model for repeated measures (Fig. 3). Less deterioration in all HRQOL scales was observed among patients with ETS than among those without ETS throughout the 24-week study period: the respective estimated mean score changes from baseline to 8 weeks were -2.78 vs. -8.05 for GHS/QOL, 5.04 vs. 0.51 for emotional functioning, -1.13 vs. -6.34 for cognitive functioning, -0.35 vs. -3.55 for physical functioning, 1.81 vs. -3.97 for social functioning, and -8.23 vs. -7.40 for role functioning.

Fig. 3
figure 3

Association of ETS with HRQOL from baseline throughout the study period of 24 weeks using a linear mixed-effects model for repeated measures. The least squares means of the score at each time point were determined for the GHS/QOL and five functional (physical, role, emotional, cognitive, social) scales

To evaluate the association between ETS and HRQOL based on the baseline symptom status, changes in HRQOL scores according to ETS were assessed in subgroups of patients with and without symptoms at baseline (Fig. 4 and S3). For symptomatic patients, ETS was associated with improved HRQOL scores from baseline to 8 weeks after initiation of treatment: the respective estimated mean score changes for patients with and without ETS were 5.86 and -4.94 for GHS/QOL, 11.73 and 3.15 for emotional functioning, 6.79 and -2.24 for cognitive functioning, 26.73 and 3.79 for physical functioning, 13.58 and -3.10 for social functioning, and 6.94 and 2.19 for role functioning. For asymptomatic patients, ETS was associated with the lowest degree of deterioration in all HRQOL scales throughout the study period. Of note, symptomatic patients with ETS had similar characteristics as asymptomatic patients without ETS in relation to HRQOL throughout the study period.

Fig. 4
figure 4

Association of ETS with HRQOL according to the baseline symptom status. A) The least squares means of the score at each time point throughout the study period for GHS/QOL, social functioning, and emotional functioning. B) The least squares means of the score change from baseline to 8 weeks after initiation of treatment for GHS/QOL, social functioning, and emotional functioning

Discussion

HRQOL provides a reasonably comprehensive picture of a patient’s experience during their disease and treatment [32]. PROs are becoming increasingly crucial for capturing the subjective aspects of a patient’s HRQOL because of the substantial disagreement between physician and patient assessments of a patient’s health status and perceptions [9, 33]. HRQOL may be even more relevant for mCRC patients because the aim of treatment is generally palliative rather than curative [5] and because they have many tumor-related symptoms, such as constipation, pain, fatigue, and appetite loss [19, 20]. Consequently, consideration should be given to not only the prognosis but also the rapid improvement of HRQOL when planning treatments for symptomatic patients. However, the lack of this invaluable information is among the main problems faced during the treatment of mCRC. Here, we analyzed the clinical impacts of ETS on HRQOL using a PRO assessment tool and on prognostic relevance in baseline symptomatic mCRC patients treated with first-line cetuximab plus chemotherapy.

ETS was associated with maintaining HRQOL in an exploratory analysis of the phase III ABSOLUTE trial that assessed the treatment efficacy of nanoparticle albumin-bound paclitaxel vs. solvent-based paclitaxel in second-line chemotherapy for advanced gastric cancer [34]. We previously reported that response to treatment was associated with a clinically meaningful improvement in HRQOL for symptomatic mCRC patients [21]. Therefore, we hypothesized that ETS may result in faster symptom relief because of the rapid reduction of the tumor burden for mCRC patients with baseline symptoms. However, there have been few in-depth assessments of the effects of ETS on the HRQOL of mCRC patients according to their baseline symptom status [35]. This exploratory analysis used the EORTC questionnaire and found that ETS was associated with less deterioration of GHS/QOL and functioning. In a retrospective analysis of data from three trials of first-line chemotherapy plus the anti-EGFR antibody panitumumab in RAS-wild-type mCRC, the onset of new tumor-related symptoms, defined as new opiate use, weight loss, anemia, asthenia, and decline in ECOG PS, was delayed by achieving ETS, regardless of the treatment regimen received [36]. These findings suggest that HRQOL may remain relatively constant in mCRC patients with ETS. Of note, for symptomatic patients at baseline, ETS was associated with an improved HRQOL, and their status was comparable to that of asymptomatic patients without ETS at 8 weeks. Thus, achieving ETS may be of particular benefit to mCRC patients with baseline symptoms in terms of HRQOL. Considering the high proportion of ETS in patients receiving FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab [37], it is also important to evaluate whether ETS could be a predictor of PROs even in mCRC patients treated with bevacizumab plus chemotherapy. Future studies are needed to clarify the benefit of ETS for HRQOL.

In clinical trials, tumor response based on the RECIST criteria is defined as at least a 30% reduction in the tumor, but this definition does not consider the timing of the response. Importantly, the objective response rate is not a reliable surrogate for PFS or OS [38]. On the other hand, ETS is defined as a minimum tumor reduction of 20% 6–8 weeks after the initiation of treatment, and it has been found to be associated with favorable prognostic outcomes in post hoc analyses in several trials, independent of the agents used and cancer types [16, 18, 28, 29, 31, 36, 37, 39,40,41,42,43,44,45,46]. We confirmed that ETS was significantly associated with prolonged PFS, OS, and TTF. Although the presence of baseline patient-reported symptoms has been reported as an independent prognostic factor in mCRC patients [19, 47], ETS has been associated with an improved OS even for symptomatic patients, who had similar outcomes as asymptomatic patients without ETS. Consistent with previous reports [39, 48], ETS was associated with successful conversion surgery. Collectively, ETS may be a hallmark feature of sensitivity to treatment and will offer several clinical advantages, including serving as an early predictor of treatment efficacy, guiding treatment strategies regarding surgical resection for potentially resectable disease, and providing a means for rapidly improving HRQOL.

The phase III CRYSTAL study reported a significant survival advantage of first-line cetuximab plus chemotherapy over chemotherapy alone in the treatment of KRAS wild-type mCRC [49]. Subsequently, in two phase III trials, FIRE-3/AIO KRK0306 and CALGB/SWOG 80405, evaluating the first-line treatment efficacy of cetuximab versus an antibody targeting vascular endothelial growth factor (bevacizumab) in combination with chemotherapy, the median OS was significantly better in the cetuximab plus chemotherapy group than in the bevacizumab plus chemotherapy group for patients with left-sided and RAS-wild type mCRC [16, 17]. Cetuximab maintenance treatment also tended to have a superior survival benefit when compared with bevacizumab maintenance in RAS-wild type mCRC [50]. Of note, the rate of ETS was markedly higher in the cetuximab plus chemotherapy group than in the chemotherapy alone group in the CRYSTAL trial (61.5% vs. 49.1%) [18] or in the bevacizumab plus chemotherapy group in the FIRE-3/AIO KRK0306 trial (68.2% vs. 49.1%) [16], consistent with the findings of this study (64.1% for cetuximab plus chemotherapy). In addition, our previous findings indicated that there was no deterioration in HRQOL following the addition of cetuximab to chemotherapy in the CRYSTAL study [51] and that the toxicity profiles and effects on PFS and ORR were similar for patients receiving cetuximab plus chemotherapy irrespective of their baseline symptom status in the QUACK study [19]. Taken together, cetuximab plus chemotherapy can notably accelerate ETS, suggesting that it may be the most preferred first-line regimen for left-sided and RAS wild-type mCRC patients, especially for those with baseline symptoms, from the point of view of HRQOL, prognosis, and safety.

To our knowledge, this is the first study to report the clinical impacts of ETS on HRQOL according to the baseline symptom status in mCRC patients treated with first-line cetuximab plus chemotherapy. However, the presented results are limited by the exploratory retrospective nature of the analysis and the relatively small sample size. In addition, the interpretation of the findings is limited by the design of comparisons between subgroups of patients treated with cetuximab plus chemotherapy because of the single-arm study. Because the differences in the efficacy of cetuximab according to the location of the primary tumor had not yet been demonstrated when this study began [14], there were no data on tumor sidedness. On the other hand, the main strengths of this study are the use of data from questionnaires with high completion rates, the prospective design for studying HRQOL, and the use of a well-established global PRO assessment tool [21].

In conclusion, ETS may be useful not only as an early-on-treatment predictor of treatment efficacy but also to rapidly improve HRQOL for symptomatic patients, which will facilitate patient-centered care in clinical practice.

Availability of data and materials

All data analyzed during this study are included in this published article and its supplementary information files.

References

  1. Ocvirk S, O'Keefe SJD. Dietary fat, bile acid metabolism and colorectal cancer. Semin Cancer Biol. 2021;73:347–55.

    CAS  PubMed  Google Scholar 

  2. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.

    Article  Google Scholar 

  3. Van Cutsem E, Cervantes A, Nordlinger B, Arnold D. Metastatic colorectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2014;25(Suppl 3):iii1–9.

    PubMed  Google Scholar 

  4. Andrei P, Battuello P, Grasso G, Rovera E, Tesio N, Bardelli A. Integrated approaches for precision oncology in colorectal cancer: The more you know, the better. Semin Cancer Biol. 2021. https://0-doi-org.brum.beds.ac.uk/10.1016/j.semcancer.2021.04.007.

  5. Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27(8):1386–422.

    PubMed  Google Scholar 

  6. Hagerty RG, Butow PN, Ellis PA, Lobb EA, Pendlebury S, Leighl N, et al. Cancer patient preferences for communication of prognosis in the metastatic setting. J Clin Oncol. 2004;22(9):1721–30.

    PubMed  Google Scholar 

  7. van Leeuwen M, Husson O, Alberti P, Arraras JI, Chinot OL, Costantini A, et al. Understanding the quality of life (QOL) issues in survivors of cancer: towards the development of an EORTC QOL cancer survivorship questionnaire. Health Qual Life Outcomes. 2018;16(1):114.

    PubMed  PubMed Central  Google Scholar 

  8. Gray NM, Hall SJ, Browne S, Macleod U, Mitchell E, Lee AJ, et al. Modifiable and fixed factors predicting quality of life in people with colorectal cancer. Br J Cancer. 2011;104(11):1697–703.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Ooki A, Morita S, Tsuji A, Iwamoto S, Hara H, Tanioka H, et al. Disagreement between patient- and physician-reported outcomes on symptomatic adverse events as poor prognosis in patients treated with first-line cetuximab plus chemotherapy for unresectable metastatic colorectal cancer: Results of Phase II QUACK trial. Cancer Med. 2020;9:9419.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Basch E. The missing voice of patients in drug-safety reporting. New Engl J Med. 2010;362(10):865–9.

    CAS  PubMed  Google Scholar 

  11. LeBlanc TW, Abernethy AP. Patient-reported outcomes in cancer care - hearing the patient voice at greater volume. Nat Rev Clin Oncol. 2017;14(12):763–72.

    PubMed  Google Scholar 

  12. U.S. Department of Health and Human Services FDA Center for Drug Evaluation and Research, et al. Guidance for industry: patient-reported outcome measures: use in medical product development to support labeling claims: draft guidance. Health Qual Life Outcomes. 2006;4:79.

    PubMed Central  Google Scholar 

  13. Doward LC, Gnanasakthy A, Baker MG. Patient reported outcomes: looking beyond the label claim. Health Qual Life Outcomes. 2010;8:89.

    PubMed  PubMed Central  Google Scholar 

  14. Yoshino T, Arnold D, Taniguchi H, Pentheroudakis G, Yamazaki K, Xu RH, et al. Pan-Asian adapted ESMO consensus guidelines for the management of patients with metastatic colorectal cancer: a JSMO-ESMO initiative endorsed by CSCO, KACO, MOS, SSO and TOS. Ann Oncol. 2018;29(1):44–70.

    CAS  PubMed  Google Scholar 

  15. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology, Colon Cancer. http://www.nccn.org/professionals/physician_gls/f_guidelinesasp 2021, version 3.

  16. Stintzing S, Modest DP, Rossius L, Lerch MM, von Weikersthal LF, Decker T, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab for metastatic colorectal cancer (FIRE-3): a post-hoc analysis of tumour dynamics in the final RAS wild-type subgroup of this randomised open-label phase 3 trial. Lancet Oncol. 2016;17(10):1426–34.

    CAS  PubMed  Google Scholar 

  17. Arnold D, Lueza B, Douillard JY, Peeters M, Lenz HJ, Venook A, et al. Prognostic and predictive value of primary tumour side in patients with RAS wild-type metastatic colorectal cancer treated with chemotherapy and EGFR directed antibodies in six randomized trials. Ann Oncol. 2017;28(8):1713–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Piessevaux H, Buyse M, Schlichting M, Van Cutsem E, Bokemeyer C, Heeger S, et al. Use of early tumor shrinkage to predict long-term outcome in metastatic colorectal cancer treated with cetuximab. J Clin Oncol. 2013;31(30):3764–75.

    CAS  PubMed  Google Scholar 

  19. Ooki A, Morita S, Iwamoto S, Hara H, Tanioka H, Satake H, et al. Patient-reported symptom burden as a prognostic factor in treatment with first-line cetuximab plus chemotherapy for unresectable metastatic colorectal cancer: Results of Phase II QUACK trial. Cancer Med. 2020. https://0-doi-org.brum.beds.ac.uk/10.1002/cam1004.2826.

  20. Marventano S, Forjaz M, Grosso G, Mistretta A, Giorgianni G, Platania A, et al. Health related quality of life in colorectal cancer patients: state of the art. BMC Surg. 2013;13(Suppl 2):S15.

    PubMed  PubMed Central  Google Scholar 

  21. Iwamoto S, Ooki A, Morita S, Hara H, Tanioka H, Satake H, et al. A prospective Phase II study to examine the relationship between quality of life and adverse events of first-line chemotherapy plus cetuximab in patients with KRAS wild-type unresectable metastatic colorectal cancer: QUACK trial. Cancer Med. 2018;7(9):4217–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Ooki A, Ando M, Sakamoto J, Sato A, Fujii H, Yamaguchi K. A prospective observational study to examine the relationship between quality of life and adverse events of first-line chemotherapy plus cetuximab in patients with KRAS wild-type unresectable metastatic colorectal cancer: QUACK Trial. Jpn J Clin Oncol. 2014;44(4):383–7.

    PubMed  Google Scholar 

  23. 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. J Natl Cancer Inst. 1993;85(5):365–76.

    CAS  PubMed  Google Scholar 

  24. Byrne C, Griffin A, Blazeby J, Conroy T, Efficace F. Health-related quality of life as a valid outcome in the treatment of advanced colorectal cancer. Eur J Surg Oncol. 2007;33 Suppl 2(2):S95–104.

    Google Scholar 

  25. Fayers PM, Aaronson NK, Bjordal K, Groenvold M, Curran D, Bottomley A, on behalf of the EORTC Quality of Life Group. The EORTC QLQ-C30 Scoring Manual (3rd Edition). Brussels: European Organisation for Research and Treatment of Cancer; 2001. https://www.eortc.org/app/uploads/sites/2/2018/02/SCmanual.pdf.

  26. Lang I, Kohne CH, Folprecht G, Rougier P, Curran D, Hitre E, et al. Quality of life analysis in patients with KRAS wild-type metastatic colorectal cancer treated first-line with cetuximab plus irinotecan, fluorouracil and leucovorin. Eur J Cancer (Oxford, England:1990). 2013;49(2):439–48.

    CAS  Google Scholar 

  27. Froelich MF, Petersen EL, Heinemann V, Nörenberg D, Hesse N, Gesenhues AB, et al. Impact of Size and Location of Metastases on Early Tumor Shrinkage and Depth of Response in Patients With Metastatic Colorectal Cancer: Subgroup Findings of the Randomized, Open-Label Phase 3 Trial FIRE-3/AIO KRK-0306. Clin Colorectal Cancer. 2020;19(4):291–300.e295.

    PubMed  Google Scholar 

  28. Cheng AL, Cornelio G, Shen L, Price T, Yang TS, Chung IJ, et al. Efficacy, Tolerability, and Biomarker Analyses of Once-Every-2-Weeks Cetuximab Plus First-Line FOLFOX or FOLFIRI in Patients With KRAS or All RAS Wild-Type Metastatic Colorectal Cancer: The Phase 2 APEC Study. Clin Colorectal Cancer. 2017;16(2):e73–88.

    PubMed  Google Scholar 

  29. Tsuji A, Sunakawa Y, Ichikawa W, Nakamura M, Kochi M, Denda T, et al. Early Tumor Shrinkage and Depth of Response as Predictors of Favorable Treatment Outcomes in Patients with Metastatic Colorectal Cancer Treated with FOLFOX Plus Cetuximab (JACCRO CC-05). Target Oncol. 2016;11(6):799–806.

    PubMed  Google Scholar 

  30. Ye LC, Wei Y, Zhu DX, Chen T, Xu J. Impact of early tumor shrinkage on clinical outcome in wild-type-KRAS colorectal liver metastases treated with cetuximab. J Gastroenterol Hepatol. 2015;30(4):674–9.

    CAS  PubMed  Google Scholar 

  31. Modest DP, Laubender RP, Stintzing S, Giessen C, Schulz C, Haas M, et al. Early tumor shrinkage in patients with metastatic colorectal cancer receiving first-line treatment with cetuximab combined with either CAPIRI or CAPOX: an analysis of the German AIO KRK 0104 trial. Acta Oncologica (Stockholm, Sweden). 2013;52(5):956–62.

    CAS  Google Scholar 

  32. Kluetz PG, Chingos DT, Basch EM, Mitchell SA. Patient-Reported Outcomes in Cancer Clinical Trials: Measuring Symptomatic Adverse Events With the National Cancer Institute's Patient-Reported Outcomes Version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE). Am Soc Clin Oncol Educ Book. 2016;35:67–73.

    PubMed  Google Scholar 

  33. Basch E, Dueck AC, Rogak LJ, Minasian LM, Kelly WK, O'Mara AM, et al. Feasibility Assessment of Patient Reporting of Symptomatic Adverse Events in Multicenter Cancer Clinical Trials. JAMA Oncol. 2017;3(8):1043–50.

    PubMed  PubMed Central  Google Scholar 

  34. Fujitani K, Shitara K, Takashima A, Koeda K, Hara H, Nakayama N, et al. Effect of early tumor response on the health-related quality of life among patients on second-line chemotherapy for advanced gastric cancer in the ABSOLUTE trial. Gastric Cancer. 2021;24(2):467–76.

  35. Siena S, Tabernero J, Bodoky G, Cunningham D, Rivera F, Ruff P, et al. Quality of life during first-line FOLFOX4+/-panitumumab in RAS wild-type metastatic colorectal carcinoma: results from a randomised controlled trial. ESMO Open. 2016;1(2):e000041.

    PubMed  PubMed Central  Google Scholar 

  36. Taieb J, Geissler M, Rivera F, Karthaus M, Wilson R, Loupakis F, et al. Relationship Between Tumor Response and Tumor-Related Symptoms in RAS Wild-Type Metastatic Colorectal Cancer: Retrospective Analyses From 3 Panitumumab Trials. Clin Colorectal Cancer. 2019;18(4):245–256.e245.

    PubMed  Google Scholar 

  37. Cremolini C, Loupakis F, Antoniotti C, Lonardi S, Masi G, Salvatore L, et al. Early tumor shrinkage and depth of response predict long-term outcome in metastatic colorectal cancer patients treated with first-line chemotherapy plus bevacizumab: results from phase III TRIBE trial by the Gruppo Oncologico del Nord Ovest. Ann Oncol. 2015;26(6):1188–94.

    CAS  PubMed  Google Scholar 

  38. Cooper K, Tappenden P, Cantrell A, Ennis K. A systematic review of meta-analyses assessing the validity of tumour response endpoints as surrogates for progression-free or overall survival in cancer. Br J Cancer. 2020;123(11):1686–96.

    PubMed  PubMed Central  Google Scholar 

  39. Heinemann V, Stintzing S, Modest DP, Giessen-Jung C, Michl M, Mansmann UR. Early tumour shrinkage (ETS) and depth of response (DpR) in the treatment of patients with metastatic colorectal cancer (mCRC). Eur J Cancer (Oxford, England: 1990). 2015;51(14):1927–36.

    Google Scholar 

  40. Petrelli F, Pietrantonio F, Cremolini C, Di Bartolomeo M, Coinu A, Lonati V, et al. Early tumour shrinkage as a prognostic factor and surrogate end-point in colorectal cancer: a systematic review and pooled-analysis. Eur J Cancer (Oxford, England : 1990). 2015;51(7):800–7.

    Google Scholar 

  41. Grünwald V, Lin X, Kalanovic D, Simantov R. Early Tumour Shrinkage: A Tool for the Detection of Early Clinical Activity in Metastatic Renal Cell Carcinoma. Eur Urol. 2016;70(6):1006–15.

    PubMed  Google Scholar 

  42. Kogawa T, Doi A, Shimokawa M, Fouad TM, Osuga T, Tamura F, et al. Early skin toxicity predicts better outcomes, and early tumor shrinkage predicts better response after cetuximab treatment in advanced colorectal cancer. Target Oncol. 2015;10(1):125–33.

    CAS  PubMed  Google Scholar 

  43. Colloca GA, Venturino A, Guarneri D. Early tumor shrinkage after first-line medical treatment of metastatic colorectal cancer: a meta-analysis. Int J Clin Oncol. 2019;24(3):231–40.

    PubMed  Google Scholar 

  44. Osumi H, Shinozaki E, Mashima T, Wakatsuki T, Suenaga M, Ichimura T, et al. Phase II trial of biweekly cetuximab and irinotecan as third-line therapy for pretreated KRAS exon 2 wild-type colorectal cancer. Cancer Sci. 2018;109(8):2567–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Hu J, Zhang Z, Zheng R, Cheng L, Yang M, Li L, et al. On-treatment markers as predictors to guide anti-EGFR MoAb treatment in metastatic colorectal cancer: a systematic review with meta-analysis. Cancer Chemother Pharmacol. 2017;79(2):275–85.

    CAS  PubMed  Google Scholar 

  46. Piessevaux H, Buyse M, De Roock W, Prenen H, Schlichting M, Van Cutsem E, et al. Radiological tumor size decrease at week 6 is a potent predictor of outcome in chemorefractory metastatic colorectal cancer treated with cetuximab (BOND trial). Ann Oncol. 2009;20(8):1375–82.

    CAS  PubMed  Google Scholar 

  47. Rutherford C, Campbell R, White K, King M. Patient-reported outcomes as predictors of survival in patients with bowel cancer: a systematic review. Qual Life Res. 2019;28(11):2871–87.

    PubMed  Google Scholar 

  48. Modest DP, Heinemann V, Folprecht G, Denecke T, Pratschke J, Lang H, et al. Factors That Influence Conversion to Resectability and Survival After Resection of Metastases in RAS WT Metastatic Colorectal Cancer (mCRC): Analysis of FIRE-3- AIOKRK0306. Ann Surg Oncol. 2020;27(7):2389–401.

    PubMed  PubMed Central  Google Scholar 

  49. Van Cutsem E, Kohne CH, Lang I, Folprecht G, Nowacki MP, Cascinu S, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol. 2011;29(15):2011–9.

    PubMed  Google Scholar 

  50. Huang C, Gu X, Zeng X, Chen B, Yu W, Chen M. Cetuximab versus bevacizumab following prior FOLFOXIRI and bevacizumab in postmenopausal women with advanced KRAS and BRAF wild-type colorectal cancer: a retrospective study. BMC Cancer. 2021;21(1):30.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Yamaguchi K, Ando M, Ooki A, Beier F, Guenther S, von Hohnhorst P, et al. Quality of Life Analysis in Patients With RAS Wild-Type Metastatic Colorectal Cancer Treated With First-Line Cetuximab Plus Chemotherapy. Clin Colorectal Cancer. 2016.

Download references

Acknowledgments

The authors thank the patients who participated in the QUACK study and their families; the QUACK investigators, nurses, and other medical staff who contributed to the study; the clinical research coordination (Chikako Ikeda and Mai Hatta); and the Efficacy and Safety Monitoring Committee Dr Hirofumi Fujii (Jichi Medical University Hospital) and Dr Atsushi Sato (Hirosaki University Graduate School of Medicine). This study was also supported by the Epidemiological & Clinical Research Information Network (ECRIN).

Detailed conflict of interest disclosures for each author

KY received speaker honoraria from Chugai, Bristol-Myers Squibb, Merck Serono, Takeda, Daiichi Sankyo, and Eli Lilly. JS received consultant fees from Chugai Pharmaceutical Co. Ltd. AO received speaker honoraria from Merck Serono, Chugai, Ono Pharmaceutical and Daiichi Sankyo. SM received speaker honoraria from Chugai, Bristol-Myers Squibb, Eli Lilly, MSD. Masahito Kotaka received honoraria from Chugai Pharmaceutical Co. Ltd., Takeda Pharmaceutical Co. Ltd., Yakult Honsha., Taiho Pharmaceutical, and Merck Serono. HH performed paid consulting or advisory roles for Boehringer Ingelheim, Daiichi Sankyo, Dainippon Sumitomo, Lilly, MSD and Ono and honoraria from Bayer, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Kyowa Hakko Kirin, Lilly, Merck Biopharma, MSD, Ono, Sanofi, Taiho, Takeda and Yakult Pharmaceutical and research funding from Astellas, AstraZeneca, BeiGene, Boehringer Ingelheim, Chugai, Daiichi Sankyo, Dainippon Sumitomo, Eisai, Elevar Therapeutics, GSK, Incyte, Merck Biopharma, MSD, Ono Pharmaceutical, Pfizer, and Taiho Pharmaceutical. MN received honoraria from Bayer Yakuhin, Chugai Pharmaceutical, Daiichi Sankyo, Eli Lilly Japan, Merck Bio Pharma, Ono Pharamaceutical, Otsuka Pharamaceutical, Taiho Pharmaceutical, Takeda Pharmaceutical and Yakult Honsha. SI received honoraria from Chugai Pharmaceutical Co. Ltd., and Merck Serono. HS has received research funding from Ono Pharmaceutical Co. Ltd., Daiichi Sankyo, Taiho Pharmaceutical Co., Ltd., Takeda Pharmaceutical Co., Ltd. and Sanofi; and honoraria from Bayer Co., Ltd., Bristol-Myers Squibb Co., Ltd., Chugai Pharmaceutical Co., Ltd., Eli Lilly Japan Co., Ltd., Merck Bio Pharma Co., Ltd., MSD Co., Ltd., and Yakult Honsha Co., Ltd. YH received honoraria from Taiho Pharmaceutical., Chugai Pharmaceutical Co. Ltd., Merck Serono, Takeda, Pharmaceutical Co. Ltd, Sanofi, Bayer and Eli Lilly Japan. AT received honoraria from Daiichi Sankyo, Chugai Pharmaceutical Co. Ltd., Taiho Pharmaceutical., Merck Serono, Takeda Pharmaceutical Co. Ltd., and Bristol-Myers Squibb Japan; and served on speaker bureaus for Chugai Pharmaceutical Co. Ltd., Taiho Pharmaceutical., Merck Serono, and Takeda Pharmaceutical Co. Ltd. MA received consultant fees from Asahi Kasei Co. Ltd. YK received honoraria from Bayer, Bristol-Myers Squibb, Chugai Pharma, Daiichi Sankyo, Eli Lilly Japan, Merck Bio Pharma, MSD, Ono Pharmaceutical, Sanofi, Taiho Pharmaceutical, Takeda and Yakult Honsha.

Clinical trial

This study is not a parallel group randomized trial. This is exploratory research using data from a single-arm Phase II trial prospectively evaluating the association of survival outcome, HRQOL, and adverse events in patients treated with cetuximab plus chemotherapy (QUACK trial). It was registered with the University Hospital Medical Information Network (UMIN): Clinical trial registration numbers: UMIN000010985 (format-19/06/2013).

Funding

This study was sponsored by a nonprofit organization, the Epidemiological and Clinical Research Information Network (NPO-ECRIN), as a funded and contracted research study with Bristol-Myers Squibb Co., Ltd. and Merck Serono Co., Ltd., an affiliate of Merck KGaA, Darmstadt, Germany.

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: Akira Ooki, Kensei Yamaguchi. Financial support: Junichi Sakamoto. Administrative support: Junichi Sakamoto. Data management: Junichi Sakamoto. Statistical Analysis: Akira Ooki. Interpretation: Akira Ooki, Satoshi Morita. Manuscript writing: Akira Ooki. Provision of study materials or patients: All authors (A.O., S.M., A.T., S.I., H.H., H.T., H.S., Masato Kataoka, Masahito Kotaka, Y.K., M.N., T.S., M.I., Y.M., T.S., Y.H., T.Y., M.A., J.S., K.Y.). Final approval of the manuscript: All authors (A.O., S.M., A.T., S.I., H.H., H.T., H.S., Masato Kataoka, Masahito Kotaka, Y.K., M.N., T.S., M.I., Y.M., T.S., Y.H., T.Y., M.A., J.S., K.Y.). Accountable for all aspects of the work: All authors (A.O., S.M., A.T., S.I., H.H., H.T., H.S., Masato Kataoka, Masahito Kotaka, Y.K., M.N., T.S., M.I., Y.M., T.S., Y.H., T.Y., M.A., J.S., K.Y.). The authors read and approved the final manuscript.

Corresponding author

Correspondence to Akira Ooki.

Ethics declarations

Ethics approval and consent to participate

This study was conducted according to the criteria set by the Declaration of Helsinki and the Ethics Guidelines for Clinical Research by the Ministry of Health, Labor, and Welfare in Japan. Written informed consent was obtained from all patients before the start of study treatment. The study design was approved by the local ethics committees/institutional review boards of Kobe City Medical Center General Hospital, Saitama Cancer Center, Kansai Medical University Hospital, Okayama Rosai Hospital, National Hospital Organization Nagoya Medical Center, Kansai Rosai Hospital, Sano Hospital, Aizawa Hospital, Osaka Saiseikai Senri Hospital, Yamagata Prefectural Central Hospital, Nishinomiya Municipal Central Hospital, Shikoku Central Hospital, Teikyo University Hospital, Yokohama Municipal Citizen's Hospital, Tokai Central Hospital, Gifu University Hospital, Japanese Red Cross Medical Center, Ina Central Hospital, Keiyukai Sapporo Hospital, Kiyoukai Okamoto Hospital, Kyoto Miniren Chuo Hospital, Fukuiken Saiseikai Hospital, National Hospital Organization Kobe Medical Center, Matsuyama Red Cross Hospital, Higashiosaka City General Hospital, Machida Municipal Hospital, Kansai Medical University Takii Hospital, Matsuda Hospital, Gifu Prefectural Tajimi Hospital, Japan Community Health Care Organization Kanazawa Hospital, Japan Post Kyoto Teishin Hospital, Ogaki Municipal Hospital, Saiseikai Shigaken Hospital, Kanazawa Medical University, Matsushita Memorial Hospital, Kyoto Prefectural University of Medicine, Kinki Central Hospital of the Mutual Aid Association of Public School Teachers, Kawasaki Hospital, Kaizuka City Hospital, Tokyo Metropolitan Tama Medical Center, Kikugawa General Hospital, Hirakata kohsai Hospital, Kouseiren Takaoka Hospital, Tachikawa Medical Center, Japanese Red Cross Otsu Hospital, Kagawa University Hospital, National Hospital Organization Fukuyama Medical Center, Takarazuka City Hospital, and Kansai Medical University Kori Hospital.

Consent for publication

Not applicable.

Competing interest

Merck KGaA reviewed the manuscript for medical accuracy only before journal submission.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ooki, A., Morita, S., Tsuji, A. et al. Impact of early tumor shrinkage on quality of life in patients treated with first-line cetuximab plus chemotherapy for unresectable metastatic colorectal cancer: results of Phase II QUACK trial. BMC Cancer 22, 711 (2022). https://0-doi-org.brum.beds.ac.uk/10.1186/s12885-022-09811-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12885-022-09811-x

Keywords