4,250
Views
18
CrossRef citations to date
0
Altmetric
Editorial

Treatment switching in oncology trials and the acceptability of adjustment methods

Abstract

Treatment switching has become an important issue in the development and approval of new drugs, particularly in oncology. Randomized controlled trials (RCTs) represent the gold standard for evaluating the effectiveness of interventions, but often patients randomized to the control group are permitted to switch onto the experimental treatment at some point during the trial. This is important, because standard statistical approaches used to analyze RCTs compare groups as randomized, based upon an intention-to-treat principle. When patients in both groups receive the new drug, such analyses do not provide an accurate estimate of the comparative effectiveness of the two treatments. This may lead to inappropriate decision-making – cost-effective drugs may not be approved. Limited healthcare finances may be used inefficiently. Health-related quality-of-life and lives may be lost.

In an oncology setting, treatment switching often occurs after disease progression, particularly when progression-free survival (PFS) is the primary endpoint. Consequently, PFS is not affected, but intention-to-treat estimates of the overall survival treatment effect – critical for health technology assessment (HTA) decision-making – will be confounded.

Statistical methods are available for adjusting for treatment switching, and have been the focus of recent methodological and applied studies Citation[1–10]. Given the confounding caused by switching, it may seem obvious that adjustment methods should be used to inform HTA. However, frequently adjustment analyses have been rejected by decision makers. This editorial discusses why this is the case and offers thoughts on how this can be addressed.

Why does treatment switching occur?

Treatment switching is driven by ethical concerns Citation[1,3]. The Declaration of Helsinki dictates that generation of knowledge cannot take precedence over the interests of individual medical research subjects Citation[11]. If interim RCT analyses suggest that clinical equipoise no longer exists, it may not be defensible to disallow treatment switching for patients randomized to the inferior treatment – particularly if no other non-palliative treatments are available. Switching is also often incorporated in study designs, usually after primary endpoints (such as PFS) have been observed, to boost trial recruitment Citation[12].

How important is treatment switching?

Over half of the technology appraisals of cancer medicines completed by the National Institute for Health and Care Excellence (NICE) and the Australian Pharmaceutical Benefits Advisory Committee have been affected by treatment switching Citation[1], [Mitchell A, Pers. Comm.].

Switching can have a large impact on cost–effectiveness results. For instance, in NICE TA269 (vemurafenib for melanoma), adjusting for switching in 34% of control group patients, reduced the incremental cost–effectiveness ratio from £75,500 per quality-adjusted life-year gained to £51,800 per quality-adjusted life-year gained Citation[13]. At an incremental cost–effectiveness ratio of £75,500 per quality-adjusted life-year gained vemurafenib would almost certainly not have been deemed cost-effective Citation[14]. However, the adjusted analysis was accepted and vemurafenib was recommended.

Adjustment methods

It is commonly accepted that simple adjustment methods (excluding switchers from the analysis, or censoring them at the time of switch) are prone to severe selection bias, because switching is usually related to prognosis Citation[1]. Although these have been commonly used in HTA historically Citation[1], current recommendations are that they should be avoided Citation[1,15]. Attention has turned to more complex methods, primarily the Rank Preserving Structural Failure Time Model (RPSFTM) Citation[16], inverse probability of censoring weights (IPCW) Citation[17] and two-stage adjustment Citation[2]. These may produce unbiased adjustments providing their assumptions hold. To consider the acceptability of these methods, it is important to reflect upon their key assumptions.

The RPSFTM assumes that if no patient in an RCT receives treatment, survival times will be equal, on average, in the randomized groups. In a well-designed RCT, this seems reasonable. The RPSFTM also assumes that the relative treatment effect is equal for all patients no matter when treatment is received (the ‘common treatment effect’ assumption, which is impossible to perfectly test). This is often regarded as a problematic assumption, given that switchers receive treatment later, often after disease progression. Attempts have been made to relax this assumption by developing multi-parameter RPSFTMs, but with little success Citation[18].

The IPCW extends the simple censoring approach. Switchers are censored, but remaining observations are weighted using baseline and time-dependent covariates to remove selection bias. This requires that the ‘no unmeasured confounders’ assumption holds – data must be available on all prognostic factors for mortality that independently predict the probability of switching – otherwise censoring-related selection bias will remain. The plausibility of this assumption depends upon the data collected during the trial. Further insight may be gathered from clinical experts, and by examining data collected in similar trials. However, we can never be sure that all confounders are measured. Nevertheless, as long as the most important confounders are measured, the IPCW may be prone to minimal bias. A frequent problem for IPCW applications to RCT data is that data are collected until disease progression but not beyond. To be unbiased, data collection is required up to the point of switch in switchers, and for the entire trial period for non-switchers.

The two-stage adjustment method was designed according to the switching commonly observed in oncology RCTs – where switching is permitted after a disease-related time-point (e.g., disease progression) Citation[2]. Treatment effects are estimated separately for switchers and for patients originally randomized to the experimental group. The method is only applicable if switching occurs after a specific disease-related time-point, and requires the ‘no unmeasured confounders’ assumption at this time-point. Compared to the RPSFTM, the two-stage method has the advantage of not assuming a common treatment effect. Compared to the IPCW method, it has the advantage of only requiring prognostic data availability at the specified disease-related time-point, not beyond. However, to be unbiased, it requires that switching occurs immediately at the disease-related time-point. This is unlikely to be exactly true, but if switching occurs soon after this time-point, resulting bias may be small.

Evidence suggests that each of these adjustment methods can produce low bias across a range of scenarios Citation[1,2,6], producing better estimates of the true comparative treatment effect than an intention-to-treat analysis. However, the RPSFTM is sensitive to violations of the ‘common treatment effect’ assumption. The IPCW, and to a slightly lesser extent, two-stage methods, are particularly prone to convergence issues and error when the switching proportion is very high, and when sample sizes and event numbers are small. No single method is optimal for all scenarios.

Are adjustment analyses being accepted?

At first sight, it might appear that adjustment analyses are being used inconsistently by healthcare decision makers. Consider the recent NICE technology appraisals affected by treatment switching. In TA263 (bevacizumab for breast cancer), an RPSFTM analysis was not considered appropriate, whereas in TA269 (vemurafenib for melanoma), TA326 (imatinib for gastrointestinal stromal tumors) and TA296 (crizotinib for non-small-cell lung cancer) RPSFTM and IPCW adjustments were accepted, and informed decision-making Citation[13,19–21].

A closer investigation of these appraisals reveals that consideration of adjustment methods may indeed have been consistent. In TA263, the RPSFTM was the only complex adjustment analysis presented, and was considered inappropriate for the case being investigated, largely because it adjusted only for ‘direct’ treatment switching (from the control group to the experimental treatment), while, in fact, ‘indirect’ switches (to other post-study treatments) also occurred Citation[19]. In contrast, in TA269, TA326 and TA296 a comprehensive range of adjustment methods were discussed in detail. Acceptance of the adjustment analyses was associated with consideration of the clinical plausibility of their results, supported by analyses of external data Citation[13,20,21].

This indicates that NICE is willing to accept adjustment analyses, provided methods used are appropriately justified, a range of potentially appropriate methods are considered, and, particularly, if results are shown to be clinically plausible. A similar position is taken by the Australian Pharmaceutical Benefits Advisory Committee and the Scottish Medicines Consortium, where recent examples show a consideration of RPSFTM and IPCW analyses, their assumptions, external evidence and clinical plausibility Citation[22–25].

However, these agencies may be further advanced in their use of adjustment methods than others around the world. For instance, RPSFTM analyses have been submitted to the Pan-Canadian Oncology Drug Review, but have not been considered in subsequent final review documents Citation[26–29]. Further, in Germany, the Institute for Quality and Efficiency in Healthcare rejected an RPSFTM analysis, stating that the method is ‘based on strong assumptions, the fulfillment of which cannot be checked with the available data’ (pg.22, Citation[30]). All complex adjustment methods make untestable assumptions, and thus none may be acceptable to the Institute for Quality and Efficiency in Healthcare. However, in 2014, the Institute for Quality and Efficiency in Healthcare held an ‘In Dialogue’ session specifically discussing adjustment methods Citation[31], and therefore their current position may alter.

The way forward

There are examples of adjustment analyses being accepted and rejected by HTA agencies around the world. This is not necessarily a sign of inconsistency, because adjustment analyses may not always be acceptable. Agencies such as NICE, the Australian Pharmaceutical Benefits Advisory Committee and the Scottish Medicines Consortium seem likely to reject adjustment analyses when they have not been well explained or justified, or when they provide implausible results. It is a concern that some HTA agencies do not appear to have come to terms with the use of adjustment methods. However, this may change with better submissions to these agencies. The emphasis is on manufacturers to ensure that analyses submitted to decision makers are comprehensive and robust, justifying and explaining the methods used.

Uncertainties about the adjustment methods themselves remain, and the confidence that decision makers have in them may be enhanced by further research. It is not possible to perfectly test the ‘common treatment effect’ assumption, but methods could be developed to investigate its approximate plausibility, or to quantify the bias associated with violations of it. Also, IPCW and two-stage analyses are reliant on comprehensive data collection over time, and trial design could be altered to increase the likelihood that adjustment methods can be successfully – and acceptably – applied. Furthermore, although treatment switching may be required in many circumstances to treat study participants ethically, early switching should be avoided when clinical equipoise remains, in order to protect the scientific validity of the trial.

Recent work has focused upon adjusting for ‘direct’ treatment switching. However, ‘indirect’ switching is important. If we adjust for ‘direct’ switching, we should also make adjustments if experimental group patients switch onto other investigational agents. Otherwise, decision makers are likely to disregard adjustment analyses, believing them to be biased in favor of the experimental treatment. In addition, research has focused on making adjustments to survival-time estimates. However other inputs to the economic model may also be affected by switching (e.g. costs, health-related quality-of-life). Research into adjustment methods that may be applied to these outcomes would be valuable – otherwise economic analyses may remain confounded.

Better description and justification of adjustment analyses, together with further research into the methods themselves, will allow adjustment methods to be used more confidently in healthcare decision making. Statistical adjustment methods are not perfect, but in the presence of confounded RCT data, they can be used to provide useful information to enable better decision making for the good of patients and society.

Financial & competing interests disclosure

N Latimer has led consultancy projects conducted within the University of Sheffield funded by GSK, Eisai and the Pharmaceutical Oncology Initiative. In the past 12 months, N Latimer has undertaken private consultancy for Astellas, Sanofi Aventis, Boehringer Ingelheim and Janssen. The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

References

  • Latimer NR, Abrams KR, Lambert PC, et al. Adjusting survival time estimates to account for treatment switching in randomized controlled trials – an economic evaluation context: Methods, limitations and recommendations. Med Decis Making 2014;34(3):387-402
  • Latimer N, Abrams K, Lambert PC, et al. Adjusting for treatment switching in randomized controlled trials – A simulation study and a simplified two-stage method. Stat Methods Med Res 2014. [Epub ahead of print]
  • Jonsson L, Sandin R, Ekman M, et al. Analyzing overall survival in randomized controlled trials with crossover and implications for economic evaluation. Value Health 2014;17(6):707-13
  • Ishak KJ, Proskorovsky I, Korytowsky B, et al. Methods for adjusting for bias due to crossover in oncology trials. Pharmacoeconomics 2014;32(6):533-46
  • Watkins C, Huang X, Latimer N, et al. Adjusting overall survival for treatment switches: commonly used methods and practical application. Pharm Stat 2013;12(6):348-57
  • Morden JP, Lambert PC, Latimer NR, et al. Assessing methods for dealing with treatment switching in randomized controlled trials: a simulation study. BMC Med Res Methodol 2011;11:4
  • Demetri GD, Garrett CR, Schoffski P, et al. Complete longitudinal analyses of the randomized, placebo-controlled, phase III trial of sunitinib in patients with gastrointestinal stromal tumor following imatinib failure. Clin Cancer Res 2012;18(11):3170-9
  • Korhonen P, Zuber E, Branson M, et al. Correcting overall survival for the impact of crossover via a rank-preserving structural failure time (RPSFT) model in the RECORD-1 trial of everolimus in metastatic renal-cell carcinoma. J Biopharm Stat 2012;22(6):1258-71
  • Colleoni M, Giobbie-Hurder A, Regan MM, et al. Analyses adjusting for selective crossover show improved overall survival with adjuvant letrozole compared with tamoxifen in the BIG 1-98 study. J Clin Oncol 2011;29:1117-24
  • Morgan G, Palumbo A, Dhanasiri S, et al. Overall survival of relapsed and refractory multiple myeloma patients after adjusting for crossover in the MM-003 trial for pomalidomide plus low-dose dexamethasone. Brit J Haematol 2015;168(6):820-3
  • World Medical Association. Declaration of helsinki – ethical principles for medical research involving human subjects. JAMA 2013;310(20):2191-4
  • Motzer RJ, Escudier B, Oudard S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomized, placebo-controlled phase III trial. Lancet 2008;372(9637):449-56
  • NICE. Final appraisal determination: Vemurafenib for treating locally advanced or metastatic BRAF V600 mutation-positive malignant melanoma. 2012. Available from: www.nice.org.uk/guidance/ta269/documents/melanoma-braf-v600-mutation-positive-unresectable-metastatic-vemurafenib-final-appraisal-determination-document2 [Last accessed on 23 February 2015]
  • National Institute for Health and Clinical Excellence. Guide to the methods of technology appraisal. NICE, London; 2013. Available from: www.nice.org.uk/article/pmg9/resources/non-guidance-guide-to-the-methods-of-technology-appraisal-2013-pdf [Last accessed on 23 February 2015]
  • Latimer N, Abrams K. NICE DSU Technical Support Document 16: Adjusting survival time estimates in the presence of treatment switching, Report by the Decision Support Unit. July 2014. Available from: www.nicedsu.org.uk/TSD16_Treatment_Switching.pdf [Last accessed on 23 February 2015]
  • Robins JM, Tsiatis AA. Correcting for noncompliance in randomized trials using rank preserving structural failure time models. Commun Stat Theory Methods 1991;20(8):2609-31
  • Robins JM, Finkelstein DM. Correcting for noncompliance and dependent censoring in an AIDS clinical trial with inverse probability of censoring weighted (IPCW) log-rank tests. Biometrics 2000;56(3):779-88
  • White IR, Babiker AG, Walker S, Darbyshire JH. Randomization-based methods for correcting for treatment changes: Examples from the Concorde trial. Stat Med 1999;18(19):2617-34
  • NICE. Final appraisal determination: Bevacizumab in combination with capecitabine for the first-line treatment of metastatic breast cancer. 2012. Available from: www.nice.org.uk/guidance/ta263/documents/breast-cancer-metastatic-bevacizumab-1st-line-with-capecitabine-final-appraisal-determination-guidance2 [Last accessed on 23 February 2015]
  • NICE. Final appraisal determination: Imatinib for the adjuvant treatment of gastrointestinal stromal tumors (review of NICE technology appraisal guidance 196). 2014. Available from: www.nice.org.uk/guidance/ta326/documents/gastrointestinal-stromal-tumours-imatinib-adjuvant-rev-ta196-id696-final-appraisal-determination-document2 [Last accessed on 23 February 2015]
  • NICE. Final appraisal determination: Crizotinib for previously treated non-small-cell lung cancer associated with an anaplastic lymphoma kinase fusion gene. Available from: www.nice.org.uk/guidance/ta296/documents/lung-cancer-nonsmallcell-anaplastic-lymphoma-kinase-fusion-gene-previously-treated-crizotinib-final-appraisal-determination3 [Last accessed on 23 February 2015]
  • The Pharmaceutical Benefits Scheme, Australian Government, Department of Health, Crizotinib, 200mg and 250mg, capsule, Xalkori®. November 2013. Available from: www.pbs.gov.au/info/industry/listing/elements/pbac-meetings/psd/2013-11/crizotinib [Last accessed on 23 February 2015]
  • PBAC. Public summary document – July 2014 PBAC Meeting: 7.8 Ruxolitinib, 5mg, 15mg, 20mg tablets, Jakavi®, Novartis Pharmaceuticals Australia Pty Ltd. Available from: www.pbs.gov.au/industry/listing/elements/pbac-meetings/psd/2014-07/ruxolitinib-psd-07-2014.pdf [Last accessed on 23 February 2015]
  • SMC. Resubmission: vemurafenib 240mg film-coated tablet (Zelboraf®), SMC No. (792/12). 2013. Available from: www.scottishmedicines.org.uk/files/advice/vemurafenib__Zelboraf__RESUBMISSION_FINAL_Nov_2013_for_Website.pdf [Last accessed on 23 February 2015]
  • SMC. Resubmission: crizotinib, 200mg and 250mg, hard capsule (Xalkori®), SMC No. (865/13). 2013. Available from: www.scottishmedicines.org.uk/files/advice/crizotinib_Xalkori_Resubmission_FINAL_September_2013_website.pdf [Last accessed on 23 February 2015]
  • pCODR. Expert review committee (pERC) final recommendation – Everolimus. 2012. Available from: www.pcodr.ca/idc/groups/pcodr/documents/pcodrdocument/pcodr-afinitor-fn-rec.pdf [Last accessed on 23 Febeuary 2015]
  • pCODR. Final economic guidance report: Everolimus (Afinitor) for pancreatic neuroendocrine tumors. 2012. Available from: www.pcodr.ca/idc/groups/pcodr/documents/pcodrdocument/pcodr-afinitor-pnets-fn-egr.pdf [Last accessed on 23 February 2015]
  • pCODR. Expert review committee (pERC) final recommendation – Pazopanib. 2012. Available from: www.pcodr.ca/idc/groups/pcodr/documents/pcodrdocument/pcodr-votrientmrcc-fn-rec.pdf [Last accessed on 23 February 2015]
  • pCODR. Final clinical guidance report: Pazopanib hydrochloride (Votrient) for metastatic renal cell carcinoma. 2012. Available from: www.pcodr.ca/idc/groups/pcodr/documents/pcodrdocument/pcodr-votrientmrcc-fn-cgr.pdf [Last accessed on 23 February 2015]
  • IQWiG. IQWiG Reports – Commission No. A13-35. Dabrafenib – benefit assessment according to § 35a social code book v. extract. 2013. Available from: www.iqwig.de/download/A13-35_Dabrafenib_Extract-of-dossier-assessment.pdf [Last accessed on 23 February 2015]
  • IQWiG. IQWiG in dialogue 2014. ‘Benefit assessment in studies with allowed treatment switching’. Abstracts of presentations. 2014. Available from: www.iqwig.de/download/Abstracts_Programme_en.pdf [Last accessed on 23 February 2015]

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.