Ovarian cancer screening and mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): a randomised controlled trial

Ian J Jacobs, Usha Menon, Andy Ryan, Aleksandra Gentry-Maharaj, Matthew Burnell, Jatinderpal K Kalsi, Nazar N Amso, Sophia Apostolidou, Elizabeth Benjamin, Derek Cruickshank, Danielle N Crump, Susan K Davies, Anne Dawnay, Stephen Dobbs, Gwendolen Fletcher, Jeremy Ford, Keith Godfrey, Richard Gunu, Mariam Habib, Rachel Hallett, Jonathan Herod, Howard Jenkins, Chloe Karpinskyj, Simon Leeson, Sara J Lewis, William R Liston, Alberto Lopes, Tim Mould, John Murdoch, David Oram, Dustin J Rabideau, Karina Reynolds, Ian Scott, Mourad W Seif, Aarti Sharma, Naveena Singh, Julie Taylor, Fiona Warburton, Martin Widschwendter, Karin Williamson, Robert Woolas, Lesley Fallowfield, Alistair J McGuire, Stuart Campbell, Mahesh Parmar, Steven J Skates, Ian J Jacobs, Usha Menon, Andy Ryan, Aleksandra Gentry-Maharaj, Matthew Burnell, Jatinderpal K Kalsi, Nazar N Amso, Sophia Apostolidou, Elizabeth Benjamin, Derek Cruickshank, Danielle N Crump, Susan K Davies, Anne Dawnay, Stephen Dobbs, Gwendolen Fletcher, Jeremy Ford, Keith Godfrey, Richard Gunu, Mariam Habib, Rachel Hallett, Jonathan Herod, Howard Jenkins, Chloe Karpinskyj, Simon Leeson, Sara J Lewis, William R Liston, Alberto Lopes, Tim Mould, John Murdoch, David Oram, Dustin J Rabideau, Karina Reynolds, Ian Scott, Mourad W Seif, Aarti Sharma, Naveena Singh, Julie Taylor, Fiona Warburton, Martin Widschwendter, Karin Williamson, Robert Woolas, Lesley Fallowfield, Alistair J McGuire, Stuart Campbell, Mahesh Parmar, Steven J Skates

Abstract

Background: Ovarian cancer has a poor prognosis, with just 40% of patients surviving 5 years. We designed this trial to establish the effect of early detection by screening on ovarian cancer mortality.

Methods: In this randomised controlled trial, we recruited postmenopausal women aged 50-74 years from 13 centres in National Health Service Trusts in England, Wales, and Northern Ireland. Exclusion criteria were previous bilateral oophorectomy or ovarian malignancy, increased risk of familial ovarian cancer, and active non-ovarian malignancy. The trial management system confirmed eligibility and randomly allocated participants in blocks of 32 using computer-generated random numbers to annual multimodal screening (MMS) with serum CA125 interpreted with use of the risk of ovarian cancer algorithm, annual transvaginal ultrasound screening (USS), or no screening, in a 1:1:2 ratio. The primary outcome was death due to ovarian cancer by Dec 31, 2014, comparing MMS and USS separately with no screening, ascertained by an outcomes committee masked to randomisation group. All analyses were by modified intention to screen, excluding the small number of women we discovered after randomisation to have a bilateral oophorectomy, have ovarian cancer, or had exited the registry before recruitment. Investigators and participants were aware of screening type. This trial is registered with ClinicalTrials.gov, number NCT00058032.

Findings: Between June 1, 2001, and Oct 21, 2005, we randomly allocated 202,638 women: 50,640 (25·0%) to MMS, 50,639 (25·0%) to USS, and 101,359 (50·0%) to no screening. 202,546 (>99·9%) women were eligible for analysis: 50,624 (>99·9%) women in the MMS group, 50,623 (>99·9%) in the USS group, and 101,299 (>99·9%) in the no screening group. Screening ended on Dec 31, 2011, and included 345,570 MMS and 327,775 USS annual screening episodes. At a median follow-up of 11·1 years (IQR 10·0-12·0), we diagnosed ovarian cancer in 1282 (0·6%) women: 338 (0·7%) in the MMS group, 314 (0·6%) in the USS group, and 630 (0·6%) in the no screening group. Of these women, 148 (0·29%) women in the MMS group, 154 (0·30%) in the USS group, and 347 (0·34%) in the no screening group had died of ovarian cancer. The primary analysis using a Cox proportional hazards model gave a mortality reduction over years 0-14 of 15% (95% CI -3 to 30; p=0·10) with MMS and 11% (-7 to 27; p=0·21) with USS. The Royston-Parmar flexible parametric model showed that in the MMS group, this mortality effect was made up of 8% (-20 to 31) in years 0-7 and 23% (1-46) in years 7-14, and in the USS group, of 2% (-27 to 26) in years 0-7 and 21% (-2 to 42) in years 7-14. A prespecified analysis of death from ovarian cancer of MMS versus no screening with exclusion of prevalent cases showed significantly different death rates (p=0·021), with an overall average mortality reduction of 20% (-2 to 40) and a reduction of 8% (-27 to 43) in years 0-7 and 28% (-3 to 49) in years 7-14 in favour of MMS.

Interpretation: Although the mortality reduction was not significant in the primary analysis, we noted a significant mortality reduction with MMS when prevalent cases were excluded. We noted encouraging evidence of a mortality reduction in years 7-14, but further follow-up is needed before firm conclusions can be reached on the efficacy and cost-effectiveness of ovarian cancer screening.

Funding: Medical Research Council, Cancer Research UK, Department of Health, The Eve Appeal.

Copyright © 2016 Jacobs Menon et al. Open Access article published under the terms of CC BY. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile MMS=multimodal screening. USS=ultrasound screening. *Events occurred before recruitment, but discovered after randomisation.
Figure 2
Figure 2
(A) Cumulative ovarian cancer and (B) ovarian and peritoneal cancer deaths The Royston-Parmar model is shown in the appendix (p 12, 13). HR=hazard ratio. MMS=multimodal screening. USS=ultrasound screening.
Figure 3
Figure 3
Rates of ovarian cancer The figure including confidence limits is in the appendix (p 14). MMS=multimodal screening. USS=ultrasound screening.
Figure 4
Figure 4
(A) Cumulative ovarian cancer and (B) ovarian and peritoneal deaths in MMS and no screening groups after exclusion of prevalent cases HRs and mortality reductions for 0–7 years and 7–14 years calculated from the Royston-Parmar model. Cumulative mortality curves from the Royston-Parmar model are overlaid onto Kaplan-Meier curves. HR=hazard ratio. MMS=multimodal screening.

References

    1. Cancer Research UK. Ovarian cancer survival statistics. One-, five- and ten-year survival for ovarian cancer. (accessed Nov 3, 2015)
    1. Jacobs I, Stabile I, Bridges J, et al. Multimodal approach to screening for ovarian cancer. Lancet. 1988;1:268–271.
    1. Jacobs IJ, Skates S, Davies AP, et al. Risk of diagnosis of ovarian cancer after raised serum CA 125 concentration: a prospective cohort study. BMJ. 1996;313:1355–1358.
    1. Menon U, Talaat A, Jeyarajah AR, et al. Ultrasound assessment of ovarian cancer risk in postmenopausal women with CA125 elevation. Br J Cancer. 1999;80:1644–1647.
    1. Jacobs IJ, Skates SJ, MacDonald N, et al. Screening for ovarian cancer: a pilot randomised controlled trial. Lancet. 1999;353:1207–1210.
    1. Skates SJ, Pauler DK, Jacobs IJ. Screening based on the risk of cancer calculation from Bayesian hierarchical changepoint and mixture models of longitudinal markers. J Am Stat Assoc. 2001;96:429–439.
    1. Skates SJ, Menon U, MacDonald N, et al. Calculation of the risk of ovarian cancer from serial CA-125 values for preclinical detection in postmenopausal women. J Clin Oncol. 2003;21(suppl 10):206s–210s.
    1. Menon U, Talaat A, Rosenthal AN, et al. Performance of ultrasound as a second line test to serum CA125 in ovarian cancer screening. BJOG. 2000;107:165–169.
    1. Menon U, Skates SJ, Lewis S, et al. Prospective study using the risk of ovarian cancer algorithm to screen for ovarian cancer. J Clin Oncol. 2005;23:7919–7926.
    1. Cancer Research UK Ovarian cancer statistics. Ovarian cancer mortality. (accessed Oct 6, 2015)
    1. Cancer Research UK Ovarian cancer survival statistics. Ovarian cancer survival by stage at diagnosis. (accessed Oct 6, 2015)
    1. Menon U, Gentry-Maharaj A, Ryan A, et al. Recruitment to multicentre trials—lessons from UKCTOCS: descriptive study. BMJ. 2008;337:a2079.
    1. Menon U, Gentry-Maharaj A, Hallett R, et al. Sensitivity and specificity of multimodal and ultrasound screening for ovarian cancer, and stage distribution of detected cancers: results of the prevalence screen of the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) Lancet Oncol. 2009;10:327–340.
    1. Menon U, Ryan A, Kalsi J, et al. Risk algorithm using serial biomarker measurements doubles the number of screen-detected cancers compared with a single-threshold rule in the United Kingdom Collaborative Trial of Ovarian Cancer Screening. J Clin Oncol. 2015;33:2062–2071.
    1. Barrett J, Jenkins V, Farewell V, et al. Psychological morbidity associated with ovarian cancer screening: results from more than 23,000 women in the randomised trial of ovarian cancer screening (UKCTOCS) BJOG. 2014;121:1071–1079.
    1. Sharma A, Burnell M, Gentry-Maharaj A, et al. Quality assurance and its impact on ovarian visualisation rates in the multicentre United Kingdom Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) Ultrasound Obstet Gynecol. 2015 doi: 10.1002/uog.14929. published online June 19.
    1. Prat J, for the FIGO Committee on Gynecologic Oncology Staging classification for cancer of the ovary, fallopian tube, and peritoneum. Int J Gynaecol Obstet. 2014;124:1–5.
    1. Tavassoli FA, Devilee P, editors. World Health Organization Classification of Tumours. Pathology and genetics. Tumors of the breast and female genital organs. World Health Organization; Lyon: 2003.
    1. Daya D, Cheung AN, Khunamornpong S, et al. In: WHO classification of tumors of female reproductive organs. 4th edn. Kurman RJ, Carcangiu ML, Herrington CS, Young RH, editors. International Agency for Research on Cancer; Lyon: 2014. Tumors of the peritoneum: epithelial tumors of Müllerian type; pp. 92–93.
    1. Self SG. An adaptive weighted log-rank test with application to cancer prevention and screening trials. Biometrics. 1991;47:975–986.
    1. Self SG, Etzioni R. A likelihood ratio test for cancer screening trials. Biometrics. 1995;51:44–50.
    1. Shen Y, Cai J. Maximum of the weighted Kaplan-Meier tests with application to cancer prevention and screening trials. Biometrics. 2001;57:837–843.
    1. Warwick J, Duffy SW. A review of cancer screening evaluation techniques, with some particular examples in breast cancer screening. J R Stat Soc Ser A. 2005;168:657–677.
    1. Buys SS, Partridge E, Black A, et al. Effect of screening on ovarian cancer mortality: the Prostate, Lung, Colorectal and Ovarian (PLCO) cancer screening randomized controlled trial. JAMA. 2011;305:2295–2303.
    1. Andriole GL, Crawford ED, Grubb RL, 3rd, et al. Mortality results from a randomized prostate-cancer screening trial. N Engl J Med. 2009;360:1310–1319.
    1. Oken MM, Hocking WG, Kvale PA, et al. Screening by chest radiograph and lung cancer mortality: the Prostate, Lung, Colorectal, and Ovarian (PLCO) randomized trial. JAMA. 2011;306:1865–1873.
    1. Schoen RE, Pinsky PF, Weissfeld JL, et al. Colorectal-cancer incidence and mortality with screening flexible sigmoidoscopy. N Engl J Med. 2012;366:2345–2357.
    1. National Lung Screening Trial Research Team. Aberle DR, Adams AM, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365:395–409.
    1. Fleming TR, Harrington DP. Counting processes and survival analysis. John Wiley & Sons; Hoboken: 2011.
    1. Izmirlian G. Estimation of the relative risk following group sequential procedure based upon the weighted log-rank statistic. Stat Interface. 2014;7:27–42.
    1. Royston P, Parmar MK. Flexible parametric proportional-hazards and proportional-odds models for censored survival data, with application to prognostic modelling and estimation of treatment effects. Stat Med. 2002;21:2175–2197.
    1. Liu ZA, Hanley JA, Strumpf EC. Projecting the yearly mortality reductions due to a cancer screening programme. J Med Screen. 2013;20:157–164.
    1. Fine JP, Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc. 1999;94:496–509.
    1. Carpenter B, Gelman A, Hoffman M, et al. Stan: a probabilistic programming language. J Stat Softw (in press).
    1. Lagakos SW, Schoenfeld DA. Properties of proportional-hazards score tests under misspecified regression models. Biometrics. 1984;40:1037–1048.
    1. Schroder FH, Hugosson J, Roobol MJ, et al. Screening and prostate-cancer mortality in a randomized European study. N Engl J Med. 2009;360:1320–1328.
    1. Holme O, Loberg M, Kalager M, et al. Effect of flexible sigmoidoscopy screening on colorectal cancer incidence and mortality: a randomized clinical trial. JAMA. 2014;312:606–615.
    1. Loberg M, Lousdal ML, Bretthauer M, Kalager M. Benefits and harms of mammography screening. Breast Cancer Res. 2015;17:63.
    1. Gentry-Maharaj A, Sharma A, Burnell M, et al. Acceptance of transvaginal sonography by postmenopausal women participating in the United Kingdom Collaborative Trial of Ovarian Cancer Screening. Ultrasound Obstet Gynecol. 2013;41:73–79.
    1. Lu KH, Skates S, Hernandez MA, et al. A 2-stage ovarian cancer screening strategy using the risk of ovarian cancer algorithm (ROCA) identifies early-stage incident cancers and demonstrates high positive predictive value. Cancer. 2013;119:3454–3461.
    1. Iyer R, Gentry-Maharaj A, Nordin A, et al. Predictors of complications in gynaecological oncological surgery: a prospective multicentre study (UKGOSOC-UK gynaecological oncology surgical outcomes and complications) Br J Cancer. 2015;112:475–484.

Source: PubMed

3
Se inscrever