Notes
Article history paragraph text
The research reported in this issue of the journal was funded by the HTA programme as project number 09/126/01. The contractual start date was in February 2010. The draft report began editorial review in April 2012 and was accepted for publication in September 2012. The authors have been wholly responsible for all data collection, analysis and interpretation, and for writing up their work. The HTA editors and publisher have tried to ensure the accuracy of the authors' report and would like to thank the reviewers for their constructive comments on the draft document. However, they do not accept liability for damages or losses arising from material published in this report.
Declared competing interests of authors
none
Corrections
-
This article was corrected in November 2013. See Brown T, Pilkington G, Bagust A, Boland A, Oyee J, Tudur Smith C, et al. Corrigendum. Health Technol Assess 2013;17(31):279–280. http://dx/doi.org/10.3310/hta17310-c201505
-
This article was corrected in May 2015. See Brown T, Pilkington G, Bagust A, Boland A, Oyee J, Tudur Smith C, et al. Corrigendum: Clinical effectiveness and cost-effectiveness of first-line chemotherapy for adult patients with locally advanced or metastatic non-small cell lung cancer: a systematic review and economic evaluation. Health Technol Assess 2015;17(31):281–282. http://dx/doi.org/10.3310/hta17310-c201505
Permissions
Copyright statement
© Queen's Printer and Controller of HMSO 2013. This work was produced by Brown et al. under the terms of a commissioning contract issued by the Secretary of State for Health. This issue may be freely reproduced for the purposes of private research and study and extracts (or indeed, the full report) may be included in professional journals provided that suitable acknowledgement is made and the reproduction is not associated with any form of advertising. Applications for commercial reproduction should be addressed to: NIHR Journals Library, National Institute for Health Research, Evaluation, Trials and Studies Coordinating Centre, Alpha House, University of Southampton Science Park, Southampton SO16 7NS, UK.
Scientific summary
Background
Lung cancer is the most common cancer in the world and the second most common cancer diagnosed in the UK after breast cancer. In 2008, 40,806 new cases of lung cancer were diagnosed in the UK: 32,546 in England and 2403 in Wales. Lung cancer is rarely diagnosed in people aged < 40 years and 86% of cases occur in people aged > 60 years. In both men and women, smoking is the primary cause of lung cancer and prognosis is poor. Early-stage lung cancer is often asymptomatic, with two-thirds of patients diagnosed at a late stage.
In 2005 in the UK, the National Institute for Health and Care Excellence (NICE) produced comprehensive guidelines on the management of patients with lung cancer; these guidelines recommended chemotherapy for patients with non-small cell lung cancer (NSCLC): docetaxel (Taxotere®, Sanofi-aventis; DOC), gemcitabine (Gemzar®, Eli Lilly and Company; GEM), paclitaxel (Abraxane®, Celgene Corporation; PAX) or vinorelbine (Navelbine®, Pierre Fabre Pharmaceuticals Inc.; VNB) in combination with either cisplatin (CIS) or carboplatin (CARB) as standard first-line treatments for patients with locally advanced or metastatic disease. Further guidance has been published which recommends pemetrexed (Alimta®, Eli Lilly and Company; PEM) in combination with CIS as first-line treatment for patients with non-squamous locally advanced or metastatic disease and gefitinib (Iressa®, AstraZeneca; GEF) as a suitable first-line treatment for patients with epidermal growth factor receptor (EGFR) mutation-positive (M+) locally advanced or metastatic disease. The NICE guidelines for the diagnosis and treatment of lung cancer were partially updated in 2011. However, the current guidance on chemotherapy for patients with NSCLC has not been updated and there is therefore a need for the synthesis of current NICE guidelines with NICE guidance resulting from recent single technology appraisals. The objective of this report is to provide such a synthesis.
Objectives
The objective of the study was to evaluate the clinical effectiveness and cost-effectiveness of first-line chemotherapy currently licensed in Europe and recommended by NICE, for adult patients with locally advanced or metastatic NSCLC. The results in this report relate solely to first-line systemic therapy for patients with locally advanced or metastatic NSCLC. No inference should be drawn from them regarding chemotherapy in any other context; this includes adjuvant therapy, combination therapy (with radiotherapy or surgery) or second-line and maintenance therapy. It is also important to recognise that, as in the delivery of all clinical care, there is a need to tailor treatments to the needs of individual patients and this will include the exploration of options and consideration of the risks and benefits of the various treatments available by the clinician in consultation with his or her patients.
Methods
Search strategy
Three electronic databases (MEDLINE, EMBASE and The Cochrane Library) were searched from January 1990 to August 2010 for randomised controlled trials (RCTs), systematic reviews and economic evaluations.
Patient populations
Chemotherapy-naive adult patients with locally advanced or metastatic NSCLC were included.
Interventions and comparators
Studies that compared any first-line chemotherapy currently licensed in Europe and recommended by NICE were considered.
Outcomes
Data on any of the following outcomes were included in the assessment of clinical effectiveness: overall survival (OS), OS at 1 and 2 years, progression-free survival (PFS), time to progression (TTP), tumour overall response rate, quality of life (QoL) and adverse events (AEs). For the assessment of cost-effectiveness, outcomes included incremental cost per life-year gained and incremental cost per quality-adjusted life-year (QALY) gained.
Application of inclusion/exclusion criteria
Two reviewers independently screened all titles and/or abstracts including economic evaluations. The full manuscript of any publication judged to be relevant by a reviewer was obtained and assessed for inclusion or exclusion. Two reviewers assessed the relevance of each publication; any discrepancies were resolved by consensus and, where necessary, a third reviewer was consulted.
Data extraction and quality assessment
Data were extracted into a Microsoft Access 2007 database (Microsoft Corporation, Redmond, WA, USA). All trials were assessed for methodological quality using criteria based on the Centre for Reviews and Dissemination guidance. The results of clinical and economic data extraction and quality assessment are summarised in the tables and narrative description.
Evidence synthesis
Where appropriate, relative treatment effects for OS, PFS, TTP and survival risk at years 1 and 2 were estimated using a standard meta-analysis for head-to-head comparisons between interventions based on intention-to-treat analyses. Mixed-treatment comparison methodology was used for the clinical effectiveness outcomes of OS, PFS, TTP and survival risk at 1 and 2 years.
Results
Of the 193 identified trials published since 2000, 23 trials compared chemotherapy drug regimens that are currently licensed in Europe and are recommended by NICE in a monotherapy or in combination with a platinum (PLAT) drug for the first-line treatment of patients with locally advanced or metastatic NSCLC.
Seven economic evaluations were identified from a possible 15 potential publications.
Quality assessment
Overall, the quality of the included RCTs was poorer than expected: there were few trials with fully reported methods and the definitions of the health outcomes used often differed between trials.
Clinical effectiveness review: efficacy data
All 23 clinical trials were published between 2001 and 2010 and included a total of 11,428 randomised patients. Of the 20 multicentre trials, six were international; the three single-centre trials were based in Taiwan. Seventeen trials were assessed as being sufficiently powered to evaluate OS. Median follow-up of patients ranged from 8 to 45 months. Doses of chemotherapy drugs varied, median number of chemotherapy cycles ranged from 2.6 to 6 and chemotherapy treaments were administered either by intravenous (i.v.) infusion or orally.
When the three GEF trials were compared with the other included trials, the proportion of males to females was much less; the percentage of males in the GEF trials ranged from 21% to 37%. These three trials were conducted in East Asian countries and had somewhat different patient populations compared with the other trials. Two of these trials included only patients with EGFR M+ tumour status, and one trial included patients with pulmonary adenocarcinoma who were never-smokers or were former light smokers.
Twenty-three trials were included within the network of trials for the clinical analysis. The direct evidence for the NSCLC population with squamous disease included 18 trials (> 7000 patients and > 6000 deaths). These same 18 trials plus subgroup data from an additional two studies were included in the analysis of the NSCLC population with non-squamous disease. Participants of three studies, conducted entirely within East Asian countries, constituted the EGFR M+ NSCLC population. In general, there was consistency between the results of the direct meta-analyses and the mixed-treatment comparison analyses, and also very good consistency across individual trials in the within-group comparisons.
Among NSCLC patients with squamous disease, there were no statistically significant differences between any of the four chemotherapy regimens (DOC + PLAT, GEM + PLAT, PAX + PLAT, VNB + PLAT) in terms of increasing OS. However, both the direct and indirect evidence suggests a potential non-statistically significant advantage in terms of OS for GEM + PLAT [direct meta-analysis 1: hazard ratio (HR) = 1.08; 95% confidence interval (CI) 0.98 to 1.20] and for DOC + PLAT (direct meta-analysis 1: HR = 0.89; 95% CI 0.78 to 1.00; mixed-treatment comparison 1, HR = 0.92; 95% CI 0.81 to 1.03) compared with VNB + PLAT. Analyses of 1- and 2-year survival support this conclusion.
For patients with non-squamous NSCLC there is borderline statistically significant evidence to suggest that PEM + PLAT increases OS compared with GEM + PLAT (direct meta-analysis 1, HR = 0.85; 95% CI 0.73 to 1.00). However, there is no statistically significant evidence to suggest that PEM + PLAT compared with GEM + PLAT increases PFS (mixed-treatment comparison 1, HR = 0.85; 95% CI 0.74 to 0.98).
Among patients with EGFR M+ status, OS was not statistically significantly different in those treated with GEF and those receiving PAX + PLAT or in those treated with GEF compared with those treated with DOC + PLAT. There was a statistically significant improvement in PFS among those patients treated with GEF compared with those treated with DOC + PLAT or PAX + PLAT. However, there was significant quantitative heterogeneity between the two trials comparing GEF with PAX + PLAT, which requires further exploration.
It remains unknown whether or not the clinical effectiveness of PEM + PLAT is superior to that of GEF monotherapy for patients with non-squamous disease. The relative clinical effectiveness of PEM + PLAT in patients who are EGFR M+ is unknown.
Clinical effectiveness review: adverse events
Across all the chemotherapy arms of the included trials, the most common AEs were neutropenia, anaemia and leucopenia. Rates of haematological AEs were similar for all the chemotherapy drugs with the exception of GEF, which appears to be associated with a significantly lower severe AE rate than some of the other drugs. The trials often varied in the way that AEs were defined, measured and reported.
Clinical effectiveness review: quality of life
Twelve trials reported QoL outcomes using a variety of instruments/tools. Seven trials reported no significant difference in QoL and four trials reported some significant differences between treatment groups. A lack of reporting of QoL data is a feature of the great majority of trials assessing outcomes of treatment for patients with NSCLC. This, despite its relevance to patients and clinicians, is a major shortcoming of lung cancer research. Measuring QoL outcomes in patients with advanced NSCLC is difficult mainly because of the severity of symptoms, the side effects of chemotherapy and early deaths associated with NSCLC. However, the British Thoracic Oncology Group Trial 2 has shown that it is feasible to collect QoL data in patients with performance status (PS) 0–2, stage IIIB/IV NSCLC disease within a clinical trial setting.
Cost-effectiveness review: summary
None of the seven included studies were directly relevant to decision-making in the NHS because they are not UK focused and/or they do not estimate incremental cost-effectiveness ratios (ICERs) in terms of cost per QALY gained.
Summary of Assessment Group's cost-effectiveness results
A total of 12 first-line chemotherapy regimens were incorporated into the economic model developed by the Assessment Group (AG): five primary licensed products [DOC, GEM, PAX, VNB (i.v. and oral)] used in combination with either CIS or CARB, PEM in combination with CIS, and GEF monotherapy. First-line chemotherapy regimens with the same primary agent but different PLAT therapy differ only in terms of treatment costs. A lifetime perspective is taken in the model and costs and benefits are discounted at 3.5% per annum. In the base-case analysis, British National Formulary (BNF) prices are used and in the sensitivity analysis, electronic market information tool (eMIT) prices are used; probabilistic sensitivity analysis results are also provided.
Economic results: patients with squamous disease
The four third-generation chemotherapy agents, when used in combination with PLAT for first-line treatment of advanced or metastatic NSCLC, are often considered to exhibit similar effectiveness, when compared in terms of standard statistical measures (e.g. p-values). However, the mixed-treatment comparison analysis undertaken by the AG which informs the current model does indicate important differences which, when combined with differences in the management of the condition and acquisition cost, provide a basis for differentiating between treatment options and arriving at some robust conclusions:
-
In both deterministic and probabilistic analyses for both the base-case and alternative pricing scenarios, VNB doublets yield the least patient benefit (as measured by expected discounted QALYs), and are not the least expensive option. As a result, VNB cannot be considered to provide either optimal effective or cost-effective chemotherapy treatment.
-
PAX doublets are consistently minimum cost options and therefore represent the initial ‘good value’ treatment, to be supplanted only if an alternative option yields greater benefit at an acceptable ‘willingness-to-pay’ (WTP) threshold.
-
The choice of preferred alternative main agent to PAX generally favours DOC over GEM as its greater effectiveness appears to outweigh the additional acquisition cost, although both lie on the efficiency frontier.
Economic results: patients with non-squamous disease
The addition of a PEM doublet to the four third-generation chemotherapy agents changes the relationship between the regimens, because of the clear outcome advantage of PEM therapy in terms of the improved expected survival of patients with non-squamous disease. However, the high price of branded PEM compared with the other drugs (in most cases available generically) means that PEM is preferred on cost-effectiveness grounds only if the WTP threshold is set > £37,000 per QALY (or £50,000 per QALY if eMIT prices are assumed). This means that PAX remains a viable treatment (and possibly GEM and DOC). However, VNB is clearly not cost-effective in either scenario.
Economic results: patients who are epidermal growth factor receptor mutation positive
The base-case analyses for GEF compared with the two chemotherapy doublets (PAX and DOC) for which evidence is available show poor cost-effectiveness for GEF. Results are improved somewhat by disaggregating the three GEF trials, but even then cost-effective ICERs (< £30,000 per QALY gained) are obtained only for the second alternative scenario [Western Japan Thoracic Oncology Group (WJTOG) trial only] based on the smallest RCT comparing GEF with the DOC + CIS doublet.
Discussion
Using BNF prices the AG has demonstrated that CIS doublets are preferred to CARB doublets. For patients with squamous disease, moving from low to moderate WTP thresholds, preferred drugs are: PAX → GEM → DOC. For patients with non-squamous disease, a similar pattern of ranking applies: PAX → GEM → DOC. However, PEM + CIS has improved OS compared with all other recommended treatments in patients with non-squamous disease, but PEM + CIS is relatively expensive and a high threshold is required before PEM + CIS can be considered cost-effective (up to £35,000 per QALY gained). For patients with EGFR M+, comparing GEF to PAX and DOC yields very high ICERs. For all populations, using eMIT prices means that CARB doublets are generally preferred to CIS doublets and drug administration costs become more important than drug acquisition costs. The AG is aware that the economic results rely on the limited clinical data available. Modelling of costs and benefits reveals that there are often only slight differences between treatments in terms of clinical effectiveness yet when these differences are modelled over the longer term (> 12 months) and the costs of the treatments are taken into consideration, then differences in cost-effectiveness begin to appear.
The treatment of patients with NSCLC is complex. In contrast to previous research, recent clinical effectiveness evidence from RCTs demonstrates that patient health outcomes depend not only on the treatment received but also on the characteristics of the patient population participating in the trial and of the cancer subtypes. Patients with NSCLC are not a homogeneous group; increasingly trials are distinguishing between three populations of patients (patients with squamous disease, patients with non-squamous disease and patients who are EGFR M+). The clinical effectiveness and cost-effectiveness evidence for each of the three patient populations needs to be reviewed separately.
As the prices of generic chemotherapy fall and new treatments become available, it is also prudent to consider cost-effectiveness using both BNF and eMIT prices. From the results of the economic evaluations described in this report it is clear that the size of the decision-makers' WTP threshold influences the range of treatments considered to be cost-effective.
Limitations
The limitations of the report can be summarised as follows: very few trials reported QoL data; AEs from the different trials were difficult to compare; CARB and CIS were treated as being similarly effective in the clinical analyses; and owing to the large volumes of data available for patients with lung cancer, the methods employed in the review do not always match the methods stated in the original protocol. Finally, the quality of the included trials was poorer than anticipated and this finding must be taken into consideration when interpreting the results of the clinical and economic analyses presented.
Conclusion
This comprehensive Health Technology Assessment review is unique to the field of NSCLC research in that it compares all of the regimens currently licensed in Europe and approved by NICE for the first-line systemic treatment of patients with advanced NSCLC. This review may assist clinicians to make decisions regarding the treatment of patients with advanced NSCLC as new evidence related to the important subgroups of patients becomes available in published form.
Research recommendations
The design of future lung cancer trials needs to reflect the influence of factors such as histology, genetics and the new prognostic biomarkers that are currently being identified. In addition, trials will need to be adequately powered so as to be able to test for statistically significant clinical effectiveness differences within patient populations. New initiatives are in place to record detailed information on the precise chemotherapy (and targeted chemotherapy) regimens being used, together with data on age, cell type, stage of disease and PS, allowing for very detailed observational audits of management and outcomes at a population level. It would be useful if these initiatives could be expanded to include the collection of health economics data.
Implications for practice
Closer examination of clinical effectiveness and cost-effectiveness data means that we have been able to provide a comprehensive framework of information for three subpopulations of patients with NSCLC that clinicians can refer to as they attempt to balance patient factors, available treatments, treatment costs and AEs in their daily decision-making.
Concluding remarks
The completion of this review has taken a significant length of time and during that period there has been explicit acknowledgement in the published literature of the important differences in the characteristics of patients who previously were identified as having NSCLC. It is anticipated that no further RCTs will be carried out involving patients with NSCLC as a homogeneous group, but that consideration of the important patient subgroups will take precedence and allow for the development of more specialised and targeted treatments which, in turn, will require RCTs of increasingly sophisticated design.
Funding
Funding for this study was provided by the Health Technology Assessment programme of the National Institute for Health Research.
Chapter 1 Background
Description of health problem
Incidence and prevalence
Lung cancer is the most common cancer in the world and the second most common cancer diagnosed in the UK after breast cancer. In 2008, 40,806 new cases were diagnosed in the UK: 32,546 in England and 2403 in Wales. 1 Lung cancer is rarely diagnosed in people aged < 40 years and 86% of cases occur in people aged > 60 years. 1 Table 1 provides an overview of lung cancer statistics in the UK. The European age-standardised incidence rate of lung cancer in 2008 was 45.6 per 100,000 population in England and 52.2 per 100,000 population in Wales. 1 The UK incidence rate in males is similar to incidence rates in most of Western Europe and lower than those in most of Eastern Europe. The UK incidence rate in females is one of the highest rates in the European Union (EU). 1 There is an increased incidence of lung cancer in individuals from the lowest socioeconomic strata. 2–4 In 2008, around 65,000 individuals were living with lung cancer in the UK,1 the majority of them male. 1
Lung cancer – UK | Males | Females | Total |
---|---|---|---|
Number of new cases (UK 2008) | 22,846 | 17,960 | 40,806 |
Rate per 100,000 populationa | 59.4 | 37.6 | 47.8 |
Number of deaths (UK 2008) | 19,868 | 15,393 | 35,261 |
Rate per 100,000 populationa | 51.0 | 32.0 | 40.3 |
One-year survival rate (for patients diagnosed 2004–6, England) | 27% | 30% | – |
Five-year survival rate (for patients diagnosed 2004–6, England) | 7% | 9% | – |
Causation
Smoking causes around 90% of lung cancer deaths in men and > 80% of lung cancer deaths in women in the UK. 5 Other causes include radon exposure, air pollution, heredity and occupational exposures such as asbestos and industrial chemicals. 6
Survival
There were 35,261 lung cancer-related deaths in the UK in 2008. 1 Prognosis is very poor; lung cancer is usually asymptomatic in the early stages and two-thirds of patients are diagnosed at a late stage when curative treatment is not possible. Twenty-seven per cent of male and 30% of female lung cancer patients in England and Wales survive for 1 year; 7% and 9%, respectively, survive 5 years. 1 According to the National Lung Cancer Data Audit (LUCADA) 2006–8, the median survival for individuals with lung cancer in England is 203 days (interquartile range 62–545 days). 7
There are many factors that affect lung cancer survival rates, including smoking status, general health, sex, race and cancer treatments. For example, survival rates at 1 and 3 years are significantly higher among Asian than white lung cancer patients, regardless of age. 1
Diagnosis
Lung cancer at an early stage is usually asymptomatic and, thus, diagnosis is often at a late stage. Unfortunately, two-thirds of patients are diagnosed when the cancer has already metastasised. Across England and Wales a significant proportion of each age group presents with late-stage metastatic disease. 8 According to recently updated National Institute for Health and Care Excellence (NICE) guidelines,7 urgent referral for a chest radiograph should be offered when a patient presents with haemoptysis or any of the following unexplained or persistent (i.e. lasting > 3 weeks) symptoms or signs:
-
cough
-
chest/shoulder pain
-
dyspnoea
-
weight loss
-
chest signs
-
hoarseness
-
finger clubbing
-
features suggestive of metastasis from a lung cancer (e.g. in brain, bone, liver or skin)
-
cervical/supraclavicular lymphadenopathy.
There are various techniques for diagnosing and staging non-small cell lung cancer (NSCLC) in the UK. The updated guidelines7 for the diagnosis and treatment of lung cancer recommend that if a chest radiograph or computerised tomography (CT) scan suggests lung cancer, patients should be offered an urgent referral usually to a chest physician, who should choose further investigations that give the most information about diagnosis and staging with the least risk to the patient.
Within this diagnostic process there are a number of key issues that need to be addressed including histology, epidermal growth factor receptor (EGFR) mutation status, disease staging, performance status (PS) and the presence of comorbid disease.
Disease staging
The stage of lung cancer at diagnosis reflects the degree of spread of cancer and is crucially important to determine which patients have potentially curative disease, and which do not, and this helps to define a patient's prognosis. TNM (tumour, node and metastasis) classification provides a system for staging the extent of cancer. Table 2 shows the seventh edition of the Union for International Cancer Control (UICC) TNM system9 for classification of NSCLC disease stage. T refers to the size of the primary tumour, N refers to the involvement of the lymph nodes and M refers to the presence of metastases or distant spread of the disease. It should be noted that all of the trial evidence in this review would have used the UICC sixth edition (or lower), as the seventh edition has only been implemented in the UK since January 2010. Table 2 compares the stage from the sixth edition, which has been modified, with the new stage in the seventh edition. Table 3 shows the surgical stage groupings in the seventh TNM classification.
Sixth edition | Seventh edition | |
---|---|---|
TNM stage | TNM stage | Descriptor |
T1 | T1a | Maximum dimension ≤ 2 cm |
T1b | Maximum dimension 2–3 cm | |
T2 | T2a | Maximum dimension 3–5 cm |
T2b | Maximum dimension 5 –7 cm | |
T3 | Maximum dimension > 7 cm | |
T4 | T3 | Additional nodule in same lobe |
M1 | T4 | Additional nodule in ipsilateral different lobe |
M1 | M1a | Additional nodules in contralateral lung |
M1 | M1a | Ipsilateral pleural effusion |
Stage | T | N | M |
---|---|---|---|
0 | Tis | N0 | M0 |
IA | T1a, b | N0 | M0 |
IB | T2a | N0 | M0 |
IIA | T1a, b | N1 | M0 |
T2a | N1 | M0 | |
T2b | N0 | M0 | |
IIB | T2b | N1 | M0 |
T3 | N0 | M0 | |
IIIA | T1, 2 | N2 | M0 |
T3 | N1, N2 | M0 | |
T4 | N0, N1 | M0 | |
IIIB | T4 | N2 | M0 |
Any T | N3 | M0 | |
IV | Any T | Any N | M1a, b |
Performance status
Performance status is used to quantify cancer patients' general well-being and may be used to determine whether or not a patient is fit enough to receive chemotherapy, whether or not a chemotherapy dose adjustment is necessary, and to quantify how much supportive care a patient may require. There are three main scales used to measure PS: the World Health Organization (WHO) PS scale,10 the Karnofsky Performance Status (KPS) scale10 and the Eastern Cooperative Oncology Group (ECOG) PS scale. 11 A summary of the WHO PS scale is shown in Table 4 as this is the most commonly used scale in clinical practice in the UK. 10 A score of 0 on the WHO scale indicates a patient is completely able to look after him/herself and a score of 4 indicates that a patient requires a lot of support.
Scale | WHO criteria10 |
---|---|
0 | Patient is fully active and more or less the same as before illness |
1 | Patient is unable to carry out heavy physical work, but can do anything else |
2 | Patient is up and about more than half the day, able to look after him/herself, but not well enough to work |
3 | Patient is in bed or sitting in a chair for more than half the day, needs some help in looking after him/herself |
4 | Patient is in bed or a chair all the time and needs a lot of looking after |
Histology
Non-small cell lung cancer accounts for approximately 84% of all lung cancers diagnosed and the remaining 16% are small cell lung cancers. The main subtypes of NSCLC are squamous cell carcinoma (33%) and non-squamous cell carcinoma (29%); the latter is made up of adenocarcinoma (25%) and large cell carcinoma (4%). Approximately 36% of patients are listed as having NSCLC ‘not-otherwise specified’ and 1% as having carcinoma in situ. 12
Squamous cell carcinoma commonly begins in the bronchi, centrally in the lungs. Adenocarcinoma starts in the periphery of the lungs and can tend to be present for a long time before it is detected. It is the type of lung cancer usually found in non-smokers and is the most common type seen in women. Large cell carcinomas often occur in the outer regions of the lungs, and tend to grow rapidly and spread more quickly than some other forms of NSCLC. 13
Histological confirmation (i.e. a diagnosis made by taking a sample of tissue or cells) is an important element of diagnosis because it helps to determine a patient's treatment pathway. However, it is noted that histological confirmation is not always straightforward and there are several key issues that must be noted. For example, tumour heterogeneity in the context of small histological or cytological samples size, interobserver variation, the absence of centralised pathology review, and the lack of any tested biological hypothesis which explains this observation that some drugs do better when the term squamous is applied to the biopsy and some when it is not.
Despite these cautions, more and more treatments are being recommended for different types of patients as recent evidence suggests that newer drugs are beneficial in certain histological subtypes of NSCLC. For example, pemetrexed (Alimta®, Eli Lilly and Company; PEM) is beneficial in patients with non-squamous carcinoma. 13 (Note: PEM is licensed and recommended for use in patients with adenocarcinoma and large cell carcinoma; however, for the purposes of this report we refer to the use of PEM in patients with non-squamous disease.) A significant proportion of patients are diagnosed based on clinical examination and radiological investigations alone, without histological evidence. According to LUCADA, in England and Wales, histological confirmation of the cancer diagnosis is made in 72% of cases, although there is wide (regional) variation from 25% to > 85%. 14 Given that more chemotherapy options are becoming available which alter the potential treatment pathway of a patient, histological testing is expected to be carried out more frequently and in a more standardised way in the future. Recent NICE guidance for the first-line treatment of NSCLC recommends histological testing and, therefore, histological testing rates are expected to increase. 7 The proportion of patients diagnosed as having disease ‘not otherwise specified’ will decrease in time [and the proportion of patients with non-squamous and EGFR mutation-positive (M+) disease will gradually rise], although regional variation will inevitably continue.
Epidermal growth factor receptor mutation status
Improvements in the understanding of the molecular and biological basis of lung cancer have led to the identification of a number of drugs that target proteins on cancer cells for the treatment of lung cancer. Among the most studied is EGFR tyrosine kinase inhibitors (TKIs), which target proteins on cancer cells and are an effective treatment for patients with tumours with activating mutations of the epidermal growth factor receptor-tyrosine kinase (EGFR-TK)15 (EGFR M+) and are often referred to as part of a group of treatments labelled ‘targeted chemotherapy’. Analysis of predictive tumour markers is necessary to identify patients with EGFR M+ who would then be candidates for such targeted treatment. 16
Clinical consensus is that EGFR M+ status will be present in around 10% of patients within the overall population. A study by Rossell et al. 17 found that 17% of Spanish patients with non-squamous NSCLC were EGFR M+. There are various clinical and lifestyle factors associated with the likelihood of the presence of EGFR mutation; for example, the rate of EGFR M+ status is higher among East Asian female non-smokers with adenocarcinoma than among white British male smokers with squamous cell carcinoma. EGFR mutation status can act as both a predictor of response to chemotherapy treatment (identification of subgroups of populations that would benefit from EGFR-targeted therapy) and a prognostic factor (indicator or the likely natural course of the disease).
Current service provision
Two linked but independent processes guide provision of care for patients with NSCLC in the UK. The European Medicines Agency (EMA) is a centralised agency of the EU that is responsible for the scientific evaluation of medicines developed by pharmaceutical companies for use in the EU. In the UK, EMA approval, through the granting of marketing authorisation, does not automatically guarantee patient access to those medicines. At the request of the Department of Health, NICE provides guidance to the NHS in England and Wales on the clinical effectiveness and cost-effectiveness of selected new and established technologies for NSCLC by undertaking appraisals of these technologies. The NHS is legally obliged to fund and resource medicines and treatments that are recommended based on the results of NICE technology appraisals.
The National Institute for Health and Care Excellence produces clinical guidance and guidelines recommending appropriate treatments and care for people with NSCLC, the recommendations are based on the best available clinical effectiveness and cost-effectiveness evidence. Comprehensive guidelines18 on the management of patients with NSCLC published by NICE in 2005 recommended docetaxel (Taxotere®, Sanofi-aventis; DOC), gemcitabine (Gemzar®, Eli Lilly and Company; GEM), paclitaxel (Abraxane®, Celgene Corporation; PAX) and vinorelbine (Navelbine®, Pierre Fabre Pharmaceuticals Inc.; VNB) for the first-line treatment of patients with locally advanced or metastatic NSCLC.
However, since the release of the guidelines a number of NICE single technology appraisals (STAs) have evaluated other treatment regimens. STAs evaluate a single technology for a single indication. These have included PEM for patients with adenocarcinoma and large cell carcinoma,13 GEF as a first-line treatment for EGFR M+ patients,16 PEM for patients with adenocarcinoma and large cell carcinoma in the maintenance setting,19 erlotinib (Tarceva®, Roche Products Limited and Roche Diagnostics Limited; ERL) in the second-line setting20 and ERL in the maintenance setting. 21 Planned STAs include cetuximab (Erbitux®, Merck Serono) in the first-line setting22 and ERL for the first-line treatment of patients with EGFR-TK M+ NSCLC.
There has been no systematic or comprehensive examination of the clinical effectiveness and cost-effectiveness of the current chemotherapy recommendations. New and updated guidelines7 include recommendations on communication, diagnosis and staging, selection of patients for treatment with curative intent, surgical techniques, smoking cessation, combination treatment for NSCLC, treatment of small cell lung cancer, managing endobronchial obstruction, managing brain metastases, and follow-up and patient perspectives. Given that the guidelines7 reflect the status of treatment preferences reflected in a number of recent NICE appraisals and the complexity of the clinical issues and changes in drug prices (as generics become available and Patient Access Schemes are applied), it can be confusing for health professionals to determine the most cost-effective chemotherapy for an individual patient.
Treatment options for non-small cell lung cancer
It would be useful to define chemotherapy options before discussing treatment options in detail. Chemotherapy is the treatment of cancer using chemical substances. Chemotherapy drugs work to destroy cancer cells by preventing them from multiplying. Treatment consists of either a chemotherapeutic agent or a molecularly targeted agent such as EGFR. Chemotherapies are generally non-specific in cellular action; they preferentially target rapidly proliferating cells and do not discriminate between malignant and non-malignant cells.
Figure 1 shows a treatment pathway for patients with NSCLC and shows estimates of the proportions and numbers of patients with NSCLC along the treatment pathway in England and Wales based on histology and staging data, NICE guidelines7 and NICE guidance. 26,27 Recommendations for patients with small cell lung cancer are not discussed in this report.
Thirty per cent of patients with NSCLC are diagnosed with stage I–IIIA disease (personal communication with Dr Michael Peake, Glenfield Hospital, using unpublished LUCADA data from 2009). These patients are suitable for potentially curative surgery or radical radiotherapy. Surgery for NSCLC consists of lobectomy, pneumonectomy and wedge resection. Approximately 50% of patients undergoing these procedures will relapse and will then be eligible for further treatment. 18 Patients with stage IIIA–IIIB disease who are not amenable to surgery can be treated with potentially curative chemoradiation.
Seventy per cent of patients with NSCLC have stage IIIB or IV disease and a PS of 0 or 1 at the time of diagnosis. These patients are assessed for their suitability for first-line chemotherapy; less than half (48%) of patients who are assessed actually receive it. 8 Among those who receive chemotherapy, almost half will respond to treatment and have either a complete or a partial response. Of these patients, a relatively small proportion can go on to have maintenance treatment and only 28% are suitable for second-line chemotherapy. 23
The majority of patients with NSCLC are diagnosed late and have metastatic or locally advanced disease. Therefore, up to 50% of patients are treated with best supportive care (BSC) alone. During all stages of treatment, patients receive BSC or ‘active supportive care’ in addition to any anticancer treatment. In the recently published lung cancer guidelines,7 NICE defines ‘supportive care’ as ‘the multidisciplinary holistic care offered to all patients and their carers throughout the pathway to help them cope with cancer and treatment of it. Best supportive care packages include options for information giving, symptom control and psychological, social and spiritual support. Palliative care provides a similar holistic approach, but is specific to those patients with advanced progressive illness’ (p. 98). 7
First-line treatment options for patients with NSCLC are shown in Figure 2. Less than 70% of patients with NSCLC have stage IIIB or stage IV disease, which equates to 20,433 patients. The percentage of patients in each stage is from 2009 audit data (M Peake, personal communication). The proportion of patients receiving BSC, chemotherapy, radiotherapy and surgery, stratified by stage is derived from LUCADA data for 2009 (personal communication with Dr Paul Beckett, Queens Hospital, using unpublished LUCADA data from 2009). These proportions have been applied to the most up-to-date incidence rates for NSCLC in England and Wales. 24 It should be noted that disease stage was recorded in 81% of cases and that these cases represent 98% of expected incidence cases for 2009; therefore, as a result of these missing data the total percentage of patients receiving BSC, radiotherapy, chemotherapy and surgery ranges from 70% to 94% within each disease stage (and does not equal 100%). In addition, the percentage of people receiving radiotherapy includes both those receiving radical and those receiving palliative treatment.
Outcome measures
Survival is considered the most reliable cancer end point within a randomised controlled trial (RCT), and when trials can be conducted to adequately assess survival it is usually the preferred end point. Overall survival (OS) is measured as the time from randomisation to death from any cause; median survival is the point in time at which 50% of people with a condition will have died and 50% are still alive. Year-1 and -2 survival risks are defined as the probability of survival in intervals of time elapsed from randomisation to years 1 and 2, respectively.
The majority of trials also report progression-free survival (PFS) as an intermediate surrogate measure of survival. PFS measures the length of time between randomisation until tumour progression or death from any cause; unlike OS, PFS is not an unequivocal outcome measure and is often determined by how frequently patients are monitored.
Tumour progression is defined as at least a 20% growth in the size of the tumour or spread of the tumour since the beginning of treatment. 28 Time to progression (TTP) is defined as the time from randomisation until tumour progression (and does not include death). The majority of RCTs also measure overall response rate (ORR), which is the proportion of people who show a response (the tumour shrinks), which can be complete or partial. Stable disease is recorded when there is no response and the tumour does not change in size. Stable disease also means that no new tumours have developed and that the cancer has not spread to any new regions of the body. 28
Adverse event (AE) and quality-of-life (QoL) data are also measures of important clinical benefit and provide information on how well chemotherapy is tolerated. In patients with advanced NSCLC, palliative chemotherapy is given to improve QoL. The EuroQol 5D (European Quality of Life-5 Dimensions; EQ-5D) is a standardised generic instrument for measuring health-related quality of life (HRQoL). It provides a utility score for health and a self-rating of HRQoL. Other commonly used QoL tools within NSCLC trials are the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ)-C3029 and the lung cancer-specific module QLQ-LC13,30 the Lung Cancer Symptom Scale (LCSS)31 and the Functional Assessment of Cancer Therapy – Lung (FACT-L) questionnaire. 32 Both AEs resulting from the disease itself and those due to chemotherapy have a considerable impact on HRQoL. 33
Despite QoL being both a vitally important measure of a patient's general emotional, physical and mental well-being and a very relevant measure of the ‘success’ of chemotherapy treatment primarily because advanced stage NSCLC is not curable, a minority of trials address QoL issues. When QoL has been examined, patients receiving chemotherapy report better scores compared with patients receiving BSC alone. 34
Current UK guidelines and guidance
In terms of first-line treatment, NICE guidelines7 recommend that chemoradiation is the first choice of treatment for patients with stage IIIA disease and is also an option for patients with stage IIIB disease who have good PS (WHO 0–1 or KPS 80–100) and localised disease that can be safely encompassed in a radical radiotherapy treatment volume. These patients are a very different and much smaller group of patients than patients receiving palliative chemotherapy. Chemotherapy is the first choice of treatment for patients with stage IIIB and stage IV NSCLC assessed as being of WHO PS 0–1. 7 BSC (including palliative radiotherapy) is the first choice of treatment for patients with stage IV WHO PS 3–4, following medical optimisation.
The NICE guidelines7 recommend that first-line chemotherapy for advanced NSCLC should be a combination of a third-generation drug (DOC, GEM, PAX or VNB) and a platinum (PLAT) – either carboplatin (CARB) or cisplatin (CIS). According to the updated NICE guidelines,7 whether CARB or CIS is used depends on the balance of toxicity, efficacy and convenience. Patients who are unable to tolerate a PLAT combination may be offered single-agent chemotherapy with a third-generation drug. 7
Following STAs of PEM and gefitinib (Iressa®, AstraZeneca; GEF), NICE also recommends that PEM plus CIS be considered as a first-line therapy for patients with locally advanced or metastatic NSCLC who are histologically confirmed as having large cell or adenocarcinoma. 13 GEF as a single agent is recommended as an option for the first-line treatment of patients with locally advanced or metastatic NSCLC who test positive for the EGFR-TK mutation. 16 Table 5 summarises the licensed indications and recommendations set out by NICE which govern the use of chemotherapy as a first-line treatment for patients with locally advanced or metastatic NSCLC in England and Wales.
Treatment | NICE recommendations | Licensed indication (Summary of Product Characteristics) | Common AEs |
---|---|---|---|
Docetaxel (DOC) (Taxotere®, Sanofi-aventis; Docetaxel Teva Pharma®, Actavis UK Ltd) Taxane Blocks the growth of cancer cells by preventing cell division i.v. Steroids given to prevent allergic reaction |
DOC + PLAT22 (Single agent if intolerant) First line Stages III and IV Good PS (WHO 0–1/KPS 80–100) |
Taxotere in combination with CIS is indicated for the treatment of patients with unresectable, locally advanced or metastatic NSCLC, in patients who have not previously received chemotherapy for this condition | Allergic reaction presenting as flushing, skin reactions, itching, chest tightness, back pain, difficulty breathing, fever or chills, swelling, weight gain, stomach upsets, alopecia, cardiac irregularities and tiredness |
Docetaxel Teva Pharma in combination with CIS is indicated for the treatment of patients with unresectable, locally advanced or metastatic NSCLC, in patients who have not previously received chemotherapy for this condition | |||
Gefitinib (GEF) (Iressa®, AstraZeneca) Protein kinase inhibitor Blocks the protein EGFR which is involved in the growth and spread of cancer cells Oral |
NICE TA 19227 First line Patients with EGFR-TK M+ status Stages III and IV Manufacturer must provide at fixed price agreed under Patient Access Scheme |
Iressa is indicated for the treatment of adult patients with locally advanced or metastatic NSCLC with activating mutations of EGFR-TK | Diarrhoea, vomiting, nausea, skin reactions such as an acne-like rash, which is sometimes itchy with dry and/or cracked skin, loss of appetite, weakness, dry, red or sore mouth and increased level of alanine aminotransferase |
Gemcitabine (GEM) (Gemzar®, Eli Lilly and Company) Cytotoxic agent Kills dividing cells including cancer cells. i.v |
GEM + PLAT22 (Single agent if intolerant) First line Stages III and IV Good PS (WHO 0–1/KPS 80–100) |
Gemzar in combination with CIS is indicated as a first-line treatment of patients with locally advanced (inoperable stage IIIA or IIIB) or metastatic (stage IV) NSCLC | Myelosuppression, lethargy, flu-like symptoms, rashes, nausea and vomiting and hair loss |
Gemzar is indicated for the palliative treatment of adult patients with locally advanced or metastatic NSCLC | |||
Paclitaxel (PAX) (Abraxane®, Celgene Corporation) Taxane Blocks the growth of cancer cells by preventing cell division i.v. Steroids given to prevent allergic reaction |
PAX + PLAT22 (Single agent if intolerant) First line Stages III and IV Good PS (WHO 0–1/KPS 80–100) |
PAX in combination with CIS is indicated for the treatment of NSCLC in patients who are not candidates for potentially curative surgery and/or radiotherapy | Allergic reactions: blood disorders, fever, unusual bleeding or unexplained bruising, heart problems, high or low blood pressure, numbness, joint or muscle pain, liver disorders, nausea, diarrhoea, sore mouth and tongue, hair loss, skin reactions and swelling at injection site |
Pemetrexed (PEM) (Alimta®, Eli Lilly and Company) Antifolate Blocks three separate enzyme targets vital to the survival of cancer cells i.v. Given with folic acid tablets and vitamin B12 injections |
NICE TA 18126 PEM + CIS Patients with non-squamous histology: adenocarcinoma and large cell First line Stages III and IV |
Alimta in combination with CIS is indicated for the first-line treatment of patients with locally advanced or metastatic NSCLC other than predominantly squamous cell histology | Fatigue, nausea, loss of appetite, rash, diarrhoea and sore mouth |
Vinorelbine (VNB) (Navelbine®, Pierre Fabre Pharmaceuticals Inc.) Vinca alkaloid Stops cell division of affected cells and causes cell death i.v./oral |
VNB + PLAT22 (Single agent if intolerant) First line Stages III and IV Good PS (WHO 0–1/KPS 80–100) |
Navelbine as a single agent or in combination is indicated for the first-line treatment of stage III or IV NSCLC | Myelosuppression, nausea and vomiting, constipation, weakness, peripheral neuropathy, alopecia and injection site pain |
The patient chemotherapy treatment pathway
Following the publication of guidelines by NICE in 2005,18 PLAT-based doublet chemotherapy has become established as the standard first-line treatment for patients with advanced NSCLC and good PS in the UK. Data from a large observational pan-European trial35 show that four cycles of PLAT-based chemotherapy treatment is standard practice in England and Wales. Figure 3 presents a flow diagram of the patient treatment pathway for first-line treatment of NSCLC and an estimate of the proportions of patients along the pathway. The proportion of patients who have non-squamous disease and are treated with PEM or GEM is unknown.
Availability of therapeutic agents
Table 6 lists the costs of available branded and generic preparations as taken from the British National Formulary (BNF). 38 AstraZeneca provides a Patient Access Scheme decreasing the cost of GEF to the NHS. In addition, clinical centres frequently negotiate prices below those listed in the BNF. 38 Over the past few years, the patents for a number of these agents have expired and they are now available in generic formulations which are less expensive. The currently available data on costs are discussed more fully in Chapter 4.
Chemotherapy agent | Manufacturer | Strength | Presentation | Cost | Comments |
---|---|---|---|---|---|
DOC | Sanof-aventis | 20 mg/ml | Vials: 20 mg/1 ml, 80 mg/4 ml, 160 mg/8 ml | 20 mg = £162.75, 80 mg = £543.75, 160 mg = £1069.50 | Taxotere First licensed DOC product |
Actavis | 10 mg/ml | Vials: 20 mg/0.5 ml, 40 mg/1 ml, 80 mg/2 ml | Requires reconstitution, solvent included | ||
Actavis | 20 mg/ml | Vials: 20 mg/1 ml, 80 mg/4 ml, 140 mg/7 ml | |||
Hospira | 10 mg/ml | Vials: 20 mg/2 ml, 80 mg/8 ml, 160 mg/16 ml | |||
medac | 20 mg/ml | Vials: 20 mg/1 ml, 80 mg/4 ml, 140 mg/7 ml | Taxceus | ||
GEF | AstraZeneca | 250 mg | Tablets | 30 = £2167.71 | Iressa |
GEM | Eli Lilly and Company | 38 mg/ml after reconstitution | Vials: 200 mg, 1 g | 200 mg = £32.55, 1 g = £162.76 | Gemzar First licensed GEM product |
Actavis | Vials: 200 mg, 1 g, 2 g | 200 mg = £32, 1 g = £162, 1.5 g = £213.93, 2 g = £324.00 | |||
Hospira | Vials: 200 mg, 1 g, 2 g | ||||
medac | Vials: 200 mg, 1 g, 1.5 mg | ||||
Sun | Vials: 200 mg, 1 g | ||||
PAX | Bristol-Myers Squibb | 6 mg/ml | Taxol First licensed PAX product, discontinued in 2008 |
||
Celgene Corporation | 5 mg/ml | Vials: 100 mg | Not licensed for lung cancer | ||
Actavis Hospira |
6 mg/ml | Vials: 30 mg/5 ml, 100 mg/16.7 ml, 150 mg/25 ml, 300 mg/50 ml | 30 mg = £66.85, 100 mg = £200.35, 150 mg = £300.52, 300 mg = £601.03 | ||
medac | Vials: 30 mg/5 ml, 100 mg/16.7 ml, 300 mg/50 ml | ||||
PEM | Eli Lilly and Company | 25 mg/ml | Vials: 100 mg, 500 mg | 100 mg = £160.00, 500 mg = £800.00 | Alimta |
VNB | Pierre Fabre Pharmaceuticals Inc. | 10 mg/ml | Vials: 10 mg/1 ml, 40 mg/4 ml, 50 mg/5 ml | 10 mg = £29.75, 50 mg = £139.98 | Navelbine First licensed VNB product |
20 mg, 30 mg, 80 mg | Capsules | 20 mg = £43.98, 30 mg = £65.98, 80 mg = £175.92 | |||
Actavis medac |
10 mg/ml | Vials: 10 mg/1 ml, 50 mg/5 ml | 10 mg = £29.00, 50 mg = £139.00 |
Reasons for conducting this review
The most recent comprehensive review of chemotherapy treatments for patients with NSCLC was conducted in 2001 by Clegg et al. 39 and was integral to the development of the NICE guidelines for the diagnosis and treatment of lung cancer in 2005. 18 The Clegg et al. review39 focused on three first-line drugs and their use in all patients with NSCLC: PAX, GEM and VNB. At the time of the Clegg et al. review,39 DOC was not licensed for the first-line treatment of patients with lung cancer in Europe. However, in 2005 when NICE's lung cancer guidelines were first published,18 DOC had received a licence for use in this patient population and was therefore included in the guidelines alongside PAX, GEM and VNB and recommended as a standard first-line treatment.
Since 2005, the NICE appraisal process has evolved and additional recommendations from the STA process have made the clinical pathway more complicated. In addition, generic preparations for a number of the chemotherapy agents have become available resulting in a need to re-examine the cost-effectiveness of some of the drugs. Finally, research in this area appears to be at a crossroads because recent research related to histology and genetics has demonstrated important differences within the NSCLC population and the focus of clinical trials is changing. The Clegg et al. review39 served as a basis for decisions related to the clinical effectiveness of chemotherapy treatments compared with BSC. For ethical reasons, new chemotherapy drugs used in the first-line setting will not be compared with BSC, they will need to be compared with currently available therapies. In addition, there has been recent identification of specific subgroups of patients who may respond to treatment in different ways and it is expected that future research will identify more, and the clinical pathway will become even more complex.
The goal of the review is to provide a succinct overview of the now complex clinical evidence relating to clinical effectiveness and AEs and match this to the cost-effectiveness evidence for the first-line treatment of patients with NSCLC. This review aims to inform current and future guidelines, assist policy makers in deciding how the newer chemotherapy agents (e.g. PEM and GEF) fit into the current treatment pathway for patients in the NHS in England and Wales, and provide clinicians with a framework for decision-making related to the treatment options available for patients with NSCLC.
Chapter 2 Definition of the decision problem
Decision problem
The population of interest is adult patients who are chemotherapy-naive, with locally advanced or metastatic NSCLC, who are not suitable for treatment with curative intent.
Analysis was restricted to chemotherapy drugs currently licensed in Europe and approved by NICE for the first-line treatment of patients with locally advanced and metastatic NSCLC:
-
PLAT-based chemotherapy (CARB or CIS) in combination with DOC, GEM, PAX, VNB
-
PEM + CIS
-
single-agent therapy – GEF.
The primary outcome was OS. Secondary outcomes were:
-
PFS
-
time to disease progression
-
survival risk
-
ORRs
-
AEs
-
HRQoL
-
incremental cost per life-year gained (LYG)
-
incremental cost per quality-adjusted life-year (QALY) gained.
Overall aims and objectives of assessment
The objectives of the assessment are to evaluate the clinical effectiveness and cost-effectiveness of first-line chemotherapy for adult patients with locally advanced or metastatic NSCLC. Second-line, third-line and maintenance treatments are not included in the assessment.
Chapter 3 Assessment of clinical effectiveness
Methods for reviewing effectiveness
In order to ensure that adequate clinical input into the review was obtained an Advisory Panel, comprised of clinicians and experts in the field, was established. The role of this panel was to answer specific clinical questions and comment on the draft report.
Identification of trials
The systematic review was guided by the general principles recommended in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. 40 A comprehensive search strategy was developed; search terms included a combination of index terms (e.g. non-small-cell lung carcinoma) and free-text words (e.g. lung cancer or lung tumour or lung carcinoma). The search of MEDLINE and EMBASE was restricted to papers with abstracts published in the English language. MEDLINE was searched from January 1990 to March week 3 2009 and EMBASE was searched from January 1990 to week 13 2009. The Cochrane Library (including Cochrane Reviews, Database of Abstracts of Reviews of Effects, Cochrane Central Register of Controlled Trials and Health Technology Assessments) was searched up to Issue 3, July 2010. An updated search was performed of MEDLINE and EMBASE to identify trials published up until August 2010. All references were exported to the EndNote version X4 (Thomson Reuters, CA, USA). Searches have been limited to these databases based on the evidence related to searching presented by Royle and Waugh,41 which demonstrates that wider searching is not always effective in retrieving additional trials for inclusion in a specific group of diseases including cancer. Details of the search strategies are available in Appendix 1.
The protocol was revised to exclude trials that had been published prior to the year 2000, owing to the large number of references identified by the searches and to reflect recent advances in chemotherapy treatments (e.g. third-generation chemotherapy drugs). The protocol is available in Appendix 2.
A number of hand searches were carried out to ensure the completeness of the review including the database of the American Society for Clinical Oncology (ASCO) and the US Food and Drug Administration (FDA) and EMA websites. A key review of chemotherapy treatments for patients with NSCLC by Clegg et al. 39 was searched for relevant trials. Reference lists of included trials were also searched to identify any further relevant trials.
Inclusion and exclusion
The inclusion/exclusion assessment by each reviewer was recorded on a pretested, standardised form. The citations identified by the search strategy were assessed for inclusion; reviewers independently screened all the titles and abstracts identified by electronic searching of MEDLINE, EMBASE and The Cochrane Library (Issue 3, July 2010). The search of MEDLINE and EMBASE was updated to August 2010. Potentially relevant references were obtained as full-text copies and each reference was assessed independently by two reviewers using the inclusion and exclusion criteria outlined in Table 7.
Patient population | Chemotherapy-naive adult patients with locally advanced or metastatic NSCLC |
Intervention | Any first-line chemotherapy treatment currently licensed in Europe and approved by NICE including:
|
Comparators | Any first-line chemotherapy treatment currently licensed in Europe and approved by NICE for the first-line treatment of patients with locally advsanced and metastatic NSCLC |
Study Design | RCTs |
Systematic reviews | |
Outcomes | OS |
PFS | |
Time to disease progression | |
1- and 2-year survival rates | |
ORRs | |
AEs | |
HRQoL |
Data extraction strategy
Data extraction forms were developed and piloted on a sample of included trials. Data were extracted on trial design, population characteristics and outcomes by one reviewer and independently checked for accuracy by a second reviewer. Microsoft Access software (Microsoft Corporation, Redmond, WA, USA) was used to store extracted data from the included trials. Appendix 3 contains details of data extraction.
Critical appraisal strategy
All included trials were assessed for methodological quality using criteria based on the Centre for Reviews and Dissemination (CRD) guidance42 for undertaking reviews in health care and adapted to reflect the characteristics of patients with NSCLC. Data relating to quality assessment were extracted by one reviewer and independently checked for accuracy by a second reviewer. Appendix 4 contains the quality assessment criteria. Where necessary, disagreements between reviewers were discussed in consultation with a third reviewer to achieve consensus.
Evidence synthesis
Analysis was restricted to chemotherapy drugs currently licensed in Europe and approved by NICE. Recent clinical evidence from PEM-based trials has suggested significant interaction between PEM efficacy and tumour histology and indicates that histology is critical in choosing the appropriate therapy for patients with NSCLC. Based on these data, NICE recommends that PEM in combination with CIS may also be considered as a first-line therapy for patients with locally advanced or metastatic NSCLC with disease histologically confirmed as large cell or adenocarcinoma. 13 Similarly, evidence from GEF-based trials indicates that GEF efficacy depends on the presence of sensitising EGFR mutations in the tumour. GEF is recommended by NICE as an option for the first-line treatment of locally advanced or metastatic NSCLC that tests positive for the EGFR-TK mutation. 16 Recent trial evidence, therefore, supports the concept that treatment choices in the first-line management of advanced NSCLC should no longer be the same for all patients with NSCLC, but rather decisions must take into consideration tumour histology subtyping and also molecular profiling.
To reflect current UK treatment pathways, analyses were undertaken and reported for three subpopulations of NSCLC: patients with predominantly squamous disease, patients with predominantly non-squamous disease and patients who were EGFR M+. In the main, all analyses were conducted on the total population according to randomisation; however, subpopulation data were included in our analyses if used previously for international or national decision-making.
Patients with squamous disease can be treated with any of the third-generation drugs (DOC, GEM, PAX or VNB) in combination with PLAT. Very few published RCTs differentiate between subpopulations of patients. We assume that the results of all studies that do not differentiate between subpopulations are equally applicable to patients with squamous disease and non-squamous disease. Before adopting this approach, we identified four third-generation studies43–46 that reported multivariate statistical testing and included histology as a candidate explanatory variable. From our critique of these studies, we concluded that there was no significant influence of histology on outcomes for patients with squamous or non-squamous disease. In this review, all data applicable to the squamous population were derived from mixed population studies; however, none of the studies included in the review investigated the use of chemotherapy solely for patients with squamous disease.
Patients with non-squamous disease who are not EGFR M+ can be treated with either third-generation drugs in combination with PLAT or PEM + CIS. This means that the data available to support treatment decisions for patients with non-squamous disease may be derived from analyses of total (mixed) population studies as well as from RCTs where survival analyses by histology may have been undertaken. Use of subpopulation data means that survival analyses were not conducted on the total trial population according to randomisation. Subpopulation data regarding the use of PEM have been used as the basis for the award of European marketing authorisation and regulatory decision-making, we therefore considered use of these subpopulation data in our analyses to be reasonable and appropriate.
Patients who are EGFR M+ can be treated with either third-generation drugs in combination with PLAT or GEF. Again, subpopulation data regarding the use of GEF have been used as the basis for the award of European marketing authorisation and regulatory decision-making, we therefore considered use of subpopulation data in our analyses to be reasonable and appropriate.
Data on any of the following outcomes were included in the meta-analyses: OS; survival at 1 and 2 years, PFS and TTP. Definitions of PFS and TTP varied between trials and the PFS and TTP outcomes in all of the included trials were assessed for eligibility for inclusion in analyses. No evidence synthesis was attempted for QoL, AEs and ORR owing to limited data or variability in outcome assessment.
Both direct head-to-head meta-analysis and mixed-treatment comparison approaches were undertaken in order to integrate information on the relative efficacy of all included drugs as an insufficient number of trials were available that directly compared all treatment options. When sufficient data permitted, analyses were undertaken for the squamous population, the non-squamous population and the EGFR M+ population.
For the analyses of the population with squamous disease, trials for PEM and GEF were excluded. Therefore, the following analyses on OS, PFS and TTP were planned. The primary analyses in the population with squamous disease were direct meta-analysis 1 and mixed-treatment comparison 1. The remaining analyses were all sensitivity analyses to explore the impact of particular trials or characteristics on results from the primary analyses: the sensitivity analyses were undertaken to explore the impact of six cycles of chemotherapy, different combinations of chemotherapy and PLAT, and trials with < 24 months follow-up.
Primary analyses: population with squamous disease:
-
Direct meta-analysis 1: standard direct head-to-head meta-analysis using data from four licensed third-generation agents (PAX, VNB, DOC and GEM) from 18 trials. 47–60 This included four pair-wise meta-analyses for the following comparisons: GEM + PLAT compared with VNB + PLAT, GEM + PLAT compared with PAX + PLAT, VNB + PLAT compared with PAX + PLAT and VNB + PLAT compared with DOC + PLAT. Data for two comparisons (GEM + PLAT vs DOC + PLAT, PAX + PLAT vs DOC + PLAT) were available from single trials and, therefore, no direct meta-analysis was undertaken.
-
Mixed-treatment comparison 1: mixed-treatment comparison using data from four licensed third-generation agents (PAX, VNB, DOC and GEM) from 18 trials. 47–60 The analysis included direct and indirect evidence from all six pair-wise comparisons: GEM + PLAT compared with VNB + PLAT, GEM + PLAT compared with PAX + PLAT, VNB + PLAT compared with PAX + PLAT, VNB + PLAT compared with DOC + PLAT, GEM + PLAT compared with DOC + PLAT and PAX + PLAT compared with DOC + PLAT.
Sensitivity analyses: population with squamous disease:
-
Direct meta-analysis A and mixed-treatment comparison A: sensitivity analysis excluding Chen et al. 52 (< 24 months follow-up time) and Tan et al. 59 (used six cycles of chemotherapy).
-
Direct meta-analysis B and mixed-treatment comparison B: sensitivity analysis using data from PAX + CIS instead of PAX + CARB from the Schiller et al. 47 trial.
-
Direct meta-analysis C and mixed-treatment comparison C: sensitivity analysis using data from DOC + CARB instead of DOC + CIS from the Fossella et al. 44 trial.
-
Direct meta-analysis D and mixed-treatment comparison D: sensitivity analysis excluding Tan et al. 59 (used six cycles of chemotherapy).
-
Direct meta-analysis E and mixed-treatment comparison E: sensitivity analysis excluding Chen et al. 52 (< 24 months follow-up time). This affects VNB + PLAT compared with DOC + PLAT pair-wise comparison.
For the population with non-squamous disease, the following analyses on OS and PFS were planned.
Primary analyses: population with non-squamous disease:
-
Direct meta-analysis 1: standard direct head-to-head meta-analysis using data from four licensed third-generation agents (PAX, VNB, DOC and GEM) from 18 trials47–60 and two PEM studies. 61,62 This included five pair-wise meta-analyses for the following comparisons: GEM + PLAT compared with VNB + PLAT, GEM + PLAT compared with PAX + PLAT, VNB + PLAT compared with PAX + PLAT, VNB + PLAT compared with DOC + LAT and GEM + PLAT compared with PEM + PLAT. Data for two comparisons (GEM + PLAT vs DOC + PLAT and PAX + PLAT vs DOC + PLAT) were available from single trials and, therefore, no direct meta-analysis was undertaken.
-
Mixed-treatment comparison 1: mixed-treatment comparison using data from four licensed third-generation agents (PAX, VNB, DOC and GEM) from 18 trials47–60 and two PEM studies. 61,62 The analysis included direct and indirect evidence from all 10 pair-wise comparisons: GEM + PLAT compared with VNB + PLAT, GEM + PLAT compared with PAX + PLAT, GEM + PLAT compared with DOC + PLAT, GEM + PLAT compared with PEM + PLAT, VNB + PLAT compared with PAX + PLAT, VNB + PLAT compared with DOC + PLAT, VNB + PLAT compared with PEM + PLAT, PAX + LAT compared with DOC + PLAT, PAX + PLAT compared with PEM + PLAT and DOC + PLAT compared with PEM + PLAT.
The following sensitivity analyses were undertaken to explore the impact of six cycles of chemotherapy, different combinations of chemotherapy and PLAT, trials with < 24 months follow-up and the one study62 with PEM + CARB which is not licensed in the UK.
Sensitivity analyses: population with non-squamous disease:
-
Direct meta-analysis A and mixed-treatment comparison A: sensitivity analysis excluding Chen et al. 52 (< 24 months follow-up time) and Tan et al. 59 (used six cycles of chemotherapy).
-
Direct meta-analysis B and mixed-treatment comparison B: sensitivity analysis using data from PAX + CIS instead of PAX + CARB from the Schiller et al. 47 trial.
-
Direct meta-analysis C and mixed-treatment comparison C: sensitivity analysis using data from DOC + CARB instead of DOC + CIS from the Fossella et al. 44 trial.
-
Direct meta-analysis D and mixed-treatment comparison D: sensitivity analysis excluding Tan et al. 59 (used six cycles of chemotherapy).
-
Direct meta-analysis E and mixed-treatment comparison E: sensitivity analysis excluding Chen et al. 52 (< 24 months follow-up time).
-
Direct meta-analysis F and mixed-treatment comparison F: sensitivity analysis excluding Gronberg et al. 62 (contains PEM + CARB which is not licensed in the UK).
For the EGFR M+ population, the following analyses on OS and PFS were planned:
-
Direct meta-analysis 1: standard direct head-to-head meta-analysis including two trials. 15,63,64
-
Mixed-treatment comparison A: analysis including three GEF trials. 15,63–65
-
This analysis includes both direct and indirect evidence for all three trials.
Adverse events
This review focuses on AEs that were categorised in the published trials as being grades 3 and 4. It was anticipated that the AEs would be reported in a disparate fashion that would not be amenable to meta-analysis and, if this was the case, then AEs would be summarised in tabular format. Significant differences in AEs between chemotherapy treatment groups within trials are highlighted, as well as the top 10 AEs that occurred within chemotherapy treatment regimens. These top 10 AEs have been summarised by extracting all AE data from each trial, grouping similar AEs and calculating the weighted average of the proportion of each AE according to each chemotherapy treatment.
Direct evidence synthesis
All analyses for the NSCLC population with squamous disease were based on the intention-to-treat (ITT) population where possible and as noted above, included a mix of patients with squamous and non-squamous disease. For non-squamous and EGFR M+ populations, data from trials15,62,63 were based on the results of subgroup analyses. Where appropriate, standard meta-analysis were undertaken for each pair-wise treatment comparison using the ‘metan’ command within Stata Version 9.2 (StataCorp LP, College Station, TX, USA). For time-to-event outcomes (OS, PFS and TTP), the trial level estimate of log-hazard ratio (HR) and its variance were extracted directly from trial publications if available. Additional data were requested whenever needed from the authors of trials that directly compared first-line chemotherapy treatments currently licensed in Europe and approved by NICE. These additional data were requested in order to include as many relevant trials as possible in the meta-analysis. Details of additional data requested and provided are presented in Appendix 5. In the absence of direct estimates from published papers or requested from the authors, previously reported methods that use published data such as Kaplan–Meier survival curves or log-rank statistics were used to estimate the required trial-level log-HR and its variance. 66,67 A random-effects (frequentist) inverse variance-weighted approach was used to pool estimates of log-HR across trials.
In economic modelling, both short- and long-term survival data are always preferred when projecting survival benefits for a technology over a lifetime period using the best available evidence. This evidence is normally derived from a meta-analysis as the most appropriate summary statistic because this takes into account both the number of events and the time to these events, and also the data from those patients who have been censored. However, trial reports do not always report time to event data. Therefore, in order to address this, 1-year and 2-year survival data were extracted from trial reports, along with the number randomised to each treatment group to estimate the risk ratio within each trial and used a random effects Mantel–Haenszel approach to calculate the pooled risk ratio with a 95% confidence interval (CI). Although 1-year and 2-year survival analysis was specifically designed to inform economic modelling, this summary measure has some limitations, especially when follow-up and censoring patterns vary from trial to trial. One approach would be to adjust for variable follow-up and censoring across trials. However, in this analysis, not all trials reported the censoring rates and for simplicity, we assumed that these factors were comparable across trials.
Statistical heterogeneity was assessed by considering the chi-squared test for heterogeneity with a 10% level of significance, and the I2-statistic with a value of 50% representing moderate heterogeneity. 68,69
Mixed-treatment comparison – direct and indirect comparisons
As trials conducting head-to-head comparisons of all treatments under evaluation were not available or insufficient for some comparisons, the possibility of conducting an indirect comparison was investigated. This approach fulfils the objective of providing simultaneous comparison of all the relevant treatment alternatives, and can provide information about the associated decision uncertainty or sufficient information for economic evaluation. Hence, for the purposes of decision-making, a Bayesian mixed-treatment comparison framework was adopted to synthesise information on all technologies simultaneously using Markov Chain Monte Carlo (MCMC) methods to estimate the posterior distributions for our outcomes of interest. The MCMC simulation begins with an approximate distribution and, if the model is a good fit to the data, the distribution converges to the true distribution. The mixed-treatment comparison analysis allows for the synthesis of data from direct and indirect comparisons and allows for the ranking of different treatments in order of efficacy and estimation of the relative treatment effect of competing interventions. This approach assumes exchangeability of treatment effect across all included trials, such that the observed treatment effect for any comparison could have been expected to arise if it had been measured in all other included trials. This was assessed informally through examination of the trial populations and comparability of outcomes in the common treatment group facilitating the comparison. Inconsistency in the treatment effects between pair-wise comparisons were investigated by comparing the direct and indirect evidence together with the 95% CIs.
As with all meta-analyses, mixed-treatment comparison may be conducted using either fixed- or random-effects models. Random-effects models allow for the possibility that the true treatment effect may differ between trials. In our analyses, random-effects models were used throughout. Model fit was assessed based on residual deviance and deviance information criteria. Adjustment for multiarm trials was performed since estimates of relative treatment effects from trials with more than two treatment arms will be correlated owing to their joint dependence with the reference treatment arm.
In each MCMC simulation, we ranked the absolute log-hazard then used it to calculate the probability that each treatment was best across all simulations. 70,71 If a treatment is significantly better than all other treatments in the mixed-treatment comparison, the probability of it being the most effective treatment will be at least 95%. A probability < 95% indicates that there is at least one other treatment which is not significantly different to the best treatment (at the 5% level). A non-informative (flat prior) normal distribution was used for the log-HR and log-relative risk (RR) of each relative comparison; thus, the observed results are completely influenced by the data and not the choice of prior.
WinBUGS version 1.4 statistical software (MRC Biostatistics Unit, Cambridge, UK) was used for the mixed-treatment comparison analysis by adapting code (presented in Appendix 6) from the Multi-Parameter Evidence Synthesis Research Group. 72 Two chains were used to ensure that model convergence was met after 90,000 iterations with a burn-in of 10,000. Formal convergence of the models was assessed using trace plots and the Gelman–Rubin approach73 and through inspection of the history plots. OS, PFS and TTP results were expressed as HRs with 95% CI.
Results of review of clinical effectiveness
Quantity of research available
As shown in Figure 4, the electronic searches identified 5378 citations (Table 8 describes in detail the results of the database searching). Initial screening identified 330 potentially relevant references; these were obtained as full-text copies, and the 240 references that were published post 2000 were assessed for eligibility for inclusion. Of the 223 trials, 30 trials were excluded because they were not chemotherapy versus chemotherapy comparisons. Information regarding the 17 RCTs that were excluded from the 240 references found from electronic searching are listed, with reasons for exclusion, in Appendix 7.
Database | Dates | Number | Deduplicated | First screen | Second screen | Chemotherapy unlicensed in Europe or not recommended by NICE | Excluded | Chemotherapy vs chemotherapy |
---|---|---|---|---|---|---|---|---|
MEDLINE | 1990 to March week 3 2009 | 2594 | 3848 | 329 | 265 | 136 references, 126 trials and 10 linked references | 10 | 18 references, 17 trials and one linked reference |
EMBASE | 1990 to week 13 2009 | 3034 | ||||||
MEDLINE | 2009 to August week 3 2010 | 316 | 455 | 35 | 34 | 23 | 0 | 6 |
EMBASE | 2009 to week 34 2010 | 370 | ||||||
CCTR | 2000 to Issue 3 of 4 July 2010 | 1034 | 1034 | 174 | 31 | 21 | 7 | 0 |
CDSR | 2000 to Issue 3 of 4 July 2010 | 4 | 4 | 0 | 0 | 0 | 0 | 0 |
DARE | 2000 to Issue 3 of 4 July 2010 | 22 | 22 | 0 | 0 | 0 | 0 | 0 |
HTA | 2000 to Issue 3 of 4 July 2010 | 15 | 15 | 0 | 0 | 0 | 0 | 0 |
Total no. of references | 7389 | 5378 | 538 | 330 | 180 | 17 | 24 | |
Total no. of RCTs (see Figure 4) | 170 | 17 | 23 |
The database of abstracts from the ASCO annual NSCLC meetings up to and including 2010 was searched to identify any relevant trials from details of conference abstracts. Three abstracts were identified as potentially relevant for inclusion; however, full-text articles could not be found of any of the three abstracts. 74–76
Overall, 193 trials compared chemotherapy with chemotherapy, of which 23 trials (reported in 24 publications15,47–65) compared chemotherapy drugs currently licensed in Europe and recommended by NICE for the first-line treatment of patients with locally advanced or metastatic NSCLC.
Assessment of clinical effectiveness
Quality
Full details of the quality assessment criteria are presented in Appendix 4. Results of the methodological quality assessment of trials are presented in Table 9. Overall, methodological quality of included trials was poor. Only six15,43,45,55,57,64,65 of the 23 included trials reported sufficient information for them to be assessed as adequately randomised and with adequate concealment of random allocation.
Trial | Randomisation | Baseline comparability | Eligibility criteria specified | Co-interventions identified | Blinding | Withdrawals | ITT | Other outcomes | |||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Truly random | Allocation concealment | Number stated | Presented | Achieveda | Assessors | Administration | Participants | Procedure assessed | > 80% in final analysis | Reasons stated | |||||
Kelly 200148 | NS | NS | ✓ | ✓/✗ | NS | ✓ | NS | NS | NS | NS | NS | ✓ | ✓ | ✓ | ✓ |
Scagliotti 200243 | ✓ | ✓ | ✓ | ✓ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Schiller 200247 | NS | NS | ✓ | ✓/✗ | NS | ✓ | ✓/✗ | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Fossella 200344 | NS | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✗ | ✗ | ✗ | NA | ✓ | ✓ | ✓ | ✗ |
Gebbia 200349 | NS | ✓ | ✓ | ✓ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✓ | ✗ |
Gridelli 200345 | ✓ | ✓ | ✓ | ✓ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Smit 200346 | ✓ | NS | ✓ | ✓ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Chen 200451 | NS | NS | ✓ | ✓ | ✓ | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✓ | ✗ |
Douillard 200553 | NS | NS | ✓ | ✓/✗ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✓ | ✓ |
bMartoni 200554 | NS | NS | ✓ | ✓ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Thomas 200658 | NS | NS | ✓ | ✓ | ✓/✗ | ✓ | NS | ✗ | ✗ | ✗ | NA | ✓ | ✓ | ✗ | ✗ |
Chen 200752 | NS | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✓ | ✗ |
Helbekkmo 200755 | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Langer 200756 | ✓ | NS | ✓ | ✓ | NS | ✓ | NS | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Ohe 200757 | ✓ | ✓ | ✓ | ✓ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✓ |
Chang 200850 | NS | NS | ✓ | ✓ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Scagliotti 200861 | ✓ | NS | ✓ | ✓ | NS | ✓ | ✓ | NS | NS | NS | NS | ✓ | ✓ | ✓ | ✓ |
Gronberg 200962 | NS | ✓ | ✓ | ✓ | NS | ✓ | ✓ | ✗ | ✗ | ✗ | NA | ✓ | ✓ | ✗ | ✗ |
IPASS: Mok 200915 and Fukuoka 201164 | ✓ | ✓ | ✓ | ✓ | NS | ✓ | ✓ | ✗ | ✗ | ✗ | NA | ✓ | ✓ | ✓ | ✗ |
Tan 200959 | ✓ | NS | ✓ | ✓ | NS | ✓ | ✓ | ✓ | ✗ | ✗ | NA | ✓ | ✓ | ✓ | ✗ |
Maemondo 201063 | NS | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | ✓ | NS | NS | NS | ✓ | ✓ | ✗ | ✗ |
Mitsudom 201065 | ✓ | ✓ | ✓ | ✓ | NS | ✓ | ✓ | ✗ | ✗ | ✗ | NA | ✓ | ✓ | ✗ | ✗ |
Treat 201060 | NS | NS | ✓ | ✓ | NS | ✓ | NS | NS | NS | NS | NS | ✓ | ✓ | ✓ | ✓ |
All trials clearly reported the number of participants randomised. All trials reported eligibility criteria and, with the exception of four trials,48,56,58,60 all trials reported details about co-interventions, for example palliative radiotherapy and/or second-line chemotherapy (in one trial54 a minority of patients had surgery following chemotherapy). In the trial by Douillard et al. , 53 second-line therapy was built into the trial design; however, nearly all trials (appropriately) allowed second-line treatments which could potentially confound results. Five trials15,44,58,62,64,65 were reported as open, i.e. assessors, administrators and participants were not blinded; two studies59,63 blinded the assessors. In 16 studies43,45–57,60,61 the authors did not state whether or not blinding of participants, investigators or outcome assessors was carried out. The outcomes of > 80% of patients were assessed in all studies and all studies reported reasons for dropout; 10 trials15,44,48,49,51–53,59–61,64 used an ITT approach to assess OS. Five of the trials48,53,57,60,61 appeared to report fewer outcomes than initially stated.
Bias can occur as a result of the early closure of trials;77 it is noted that three of the included trials were stopped early. 47,49,65 In the trial by Gebbia et al. ,49 further accrual into the two ‘sequential chemotherapy’ arms [in this case meaning GEM + ifosfamide (Mitoxana®, Baxter Healthcare Ltd) followed by VNB + CIS or the opposite sequence, which are two group comparisons not included within this review] was stopped because the VNB + CIS arm appeared to be more effective. Owing to initial slow accrual, the protocol of the trial by Mitsudomi et al. 65 was amended to include patients with stage IIIB/IV disease and to allow outsourcing of EGFR genetic testing in order to further facilitate patient accrual. Accrual was halted when investigators considered the trial to be sufficiently powered, making further accrual of patients unnecessary, and final analyses were done on the available data. In the trial by Schiller et al. ,47 after the first 68 patients, accrual of the PS 2 cohort was halted owing to a high incidence of AEs, including five deaths (subsequent analysis showed that only two of the five deaths were clearly treatment related). All trials provided reasons for withdrawal; see Table 23 for actual numbers of patients treated.
Trial characteristics
Trial characteristics are presented in Appendix 8. The 23 trials were published between 2001 and 2010. Of the 20 multicentre trials, six have international centres. 15,44–46,59,61,64 The three single-centre trials were all located in Taiwan. 50–52 All included trials were published in English.
There are five Phase II trials,51–53,56,58 16 Phase III trials15,43–46,48,49,54,55,57,59–65 and two trials47,50 with phase undefined. Ten trials15,43,44,53,57–62,64 were funded solely by pharmaceutical companies, five trials46,47,56,63,65 were funded by research grants, two trials45,48 were funded by both pharmaceutical companies and research grants and funding was not stated in six trials. 49–52,54,55
Seventeen trials15,43–48,51–54,57,60–62,64,65 were sufficiently powered to evaluate OS, four trials49,56,59,63 were inadequately powered and the power of two trials50,58 was unclear. (If a trial reported an estimated sample size and then randomised at least this number of patients, then the trial was assessed as sufficiently powered.) Median follow-up ranged from 11 to 40 months.
Details of trial interventions are presented in Appendix 9. Four trials43,44,46,60 compared three treatment arms and three trials47,49,57 compared four treatment arms (not all arms met the inclusion criteria for this analysis). Table 10 shows the chemotherapy comparisons which were available from the 23 included trials. Trials using either CARB or CIS are both described as including PLAT.
Pair-wise comparison | Trials | Number of comparisons |
---|---|---|
GEM + PLAT vs VNB + PLAT | Chang 2008;50 Gebbia 2003;49 Gridelli 2003;45 Helbekkmo 2007;55 Martoni 2005;54 Ohe 2007;57 Scagliotti 2002;43 and Thomas 200658 | 8 |
GEM + PLAT vs PAX + PLAT | Langer 2007;56 Ohe 2007;57 Scagliotti 2002;43 Schiller 2002;47 Smit 2003;46 and Treat 201060 | 6 |
GEM + PLAT vs DOC + PLAT | Schiller 200247 | 1 |
GEM + PLAT vs PEM + PLAT | Gronberg 2009;62 and Scagliotti 200861 | 2 |
VNB + PLAT vs PAX + PLAT | Chen 2004;51 Kelly 2001;48 Ohe 2007;57 and Scagliotti 200243 | 4 |
VNB + PLAT vs DOC + PLAT | Chen 2007;52 Douillard 2005;53 Fossella 2003;44 and Tan 200959 | 4 |
PAX + PLAT vs DOC + PLAT | Schiller 200247 | 1 |
PAX + PLAT vs GEF | Maemondo 2010;63 and IPASS15,64 | 2 |
DOC + PLAT vs GEF | Mitsudomi 201065 | 1 |
Doses of chemotherapy drugs used varied, the median number of chemotherapy cycles ranged from 2.6 to 6, and route of administration was intravenous (i.v.) or oral. The majority of trials reported second-line chemotherapy and/or palliative radiotherapy, with the exception of one trial47 that reported that second-line treatment data were not collected, and four trials49,56,58,65 in which it was unclear whether or not patients received any second-line treatment. Two trials48,54 reported that patients who went on to have radiotherapy were excluded from the analysis.
Patient characteristics
Patient characteristics are presented in Appendix 10. Trial patients are generally younger and have better PS and fewer comorbidities than patients in a clinical setting. Full details of individual trial inclusion criteria can be found in Appendix 11. The majority of patients were male with adenocarcinoma stage IIIB or IV and a PS of 1. Only five trials46,48,55,58,60 reported details of the staging system used to classify patients and given the variety in the dates and settings of the trials, the staging systems used is most likely to have varied across trials.
The number of patients randomised into trial arms ranged from 39 to 863. Median age ranged from 56 to 67 years within the clinical trials, which is younger than routinely found in clinical practice. The most common age group at diagnosis of ‘malignant neoplasm of bronchus or lung’ for men and women in England in 2008 was 75–79 years. 78 The percentage of males within each trial arm ranged from 56% to 84% for trials with PLAT-based doublets incorporating third-generation chemotherapy drugs. In the three GEF trials,15,63–65 the proportion of males to females is much less; the percentage of males ranged from 21% to 37%, this is because sex is a factor in the likelihood of the presence of EGFR mutation (mutation found more often in females).
The patient populations in the trials by Maemondo et al. 63 and Mitsudomi et al. 65 were quite different from those in the other trials. These two trials63,65 were based on molecular selection, and only patients with EGFR M+ tumours were eligible for inclusion. The Iressa Pan ASian Study (IPASS)15,64 restricted the patient population to those with adenocarcinoma who were never-smokers or former light smokers in order to increase the likelihood of the presence of the EGFR mutation. All three trials15,63–65 were multicentre, but conducted within East Asian countries. Patients assigned to the experimental arms received oral GEF at the standard dose (250 mg daily). Patients assigned to the chemotherapy arm received different PLAT-based doublets (PAX + CARB in two trials15,63,64 and DOC + CIS in one trial65).
The majority of patients within the trials had stage IIIB or IV disease; at least twice as many patients had stage IV disease as stage IIIB disease.
Outcomes
Non-small cell lung cancer population with squamous disease
Eighteen trials are included for outcomes in this patient population; these 18 trials43–60 reported outcomes in trials with mixed-patient populations (i.e. a mix of squamous and non-squamous disease). The results of these 18 trials are data available for patients with squamous disease and patients with non-squamous disease.
Median OS was reported in all 18 trials; two papers43,44 also directly reported HR for OS. All 18 trials reported median PFS or TTP (as defined by each individual trial); one trial reported HRs for PFS45 and one trial reported HRs for TTP. 43 Sixteen trials43,44,46–57,59,60 reported survival rates at 1 year and nine trials44,47,48,53–57,60 reported survival rates at 2 years. Sixteen trials43,44,46–54,56–60 reported tumour ORR. Full details of the outcomes assessed in each trial are presented in Table 11.
Trial | Treatment | OS | PFS | Survival 1 year | Survival 2 years | Tumour ORR | |||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Median, months | 95% CI | HR (CI) | Median, months | 95% CI | HR (95% CI) | % | 95% CI | % | 95% CI | % | CI | ||
Kelly 200148 | VNB + CIS | 8.1 | 6.7 to 9.6 | NR | 4 | NR | NR | 36 | NR | 16 | NR | 28 | NR |
PAX + CARB | 8.6 | 7.2 to 10.7 | NR | 4 | NR | NR | 38 | NR | 15 | NR | 24 | NR | |
Scagliotti 200243 | GEM + CIS | 9.8 | 8.6 to 11.2 | 0.87 (95% CI 0.69 to 1.09) | 5.3 (TTP) | 4.4 to 6.3 | 0.95 (0.77 to 1.77) | 37 | NR | NR | NR | 30 | 95% CI 24 to 37 |
PAX + CARB | 9.9 | 9.0 to 12.5 | 0.84 (95% CI 0.67 to 1.05) | 5.5 (TTP) | 4.6 to 6.4 | 0.91 (0.74 to 1.22) | 43 | NR | NR | NR | 32 | 95% CI 25 to 38 | |
VNB + CIS | 9.5 | 8.3 to 11.0 | NR | 4.6 (TTP) | 3.9 to 5.6 | 37 | NR | NR | NR | 30 | 95% CI 24 to 36 | ||
Schiller 200247 | PAX + CIS | 7.8 | 7.0 to 8.9 | NR | 3.4 (TTP) | 2.8 to 3.9 | NR | 31 | 26 to 36 | 10 | 5 to 12 | 21 | NR |
GEM + CIS | 8.1 | 7.2 to 9.4 | NR | 4.2 (TTP), p = 0.008 vs PAX + CIS | 3.7 to 4.8 | NR | 36 | 31 to 42 | 13 | 7 to 15 | 22 | NR | |
DOC + CIS | 7.4 | 6.6 to 8.8 | NR | 3.7 (TTP) | 2.9 to 4.2 | NR | 31 | 26 to 36 | 11 | 7 to 14 | 17 | NR | |
PAX + CARB | 8.1 | 7.0 to 9.5 | NR | 3.1 (TTP) | 2.8 to 3.9 | NR | 34 | 29 to 40 | 11 | 7 to 14 | 17 | NR | |
aFossella 200344 | DOC + CIS | 11.3, p = 0.044 vs VNB + CIS | 10.1 to 12.4 | 1.183 (97.2% CI 0.989 to 1.416) | 5.07 (TTP) | 4.84 to 5.76 | NR | 46 | 42 to 51 | 21 | 16 to 25 | 31.6, p = 0.028 vs VNB + CIS | 95% CI 27.1 to 36.4 |
DOC + CARB | 9.4 | 8.7 to 10.6 | 1.048 (97.2% CI 0.877 to 1.253) | 4.61 (TTP) | 4.38 to 5.30 | NR | 38 | 33 to 43 | 18 | 13 to 22 | 23.9 | 95% CI 19.8 to 28.3 | |
VNB + CIS | 10.1 | 9.2 to 11.3 | 5.30 (TTP) | 4.84 to 6.22 | NR | 41 | 36 to 46 | 14 | 10 to 18 | 24.5 | 95% CI 20.4 to 29.0 | ||
Gebbia 200349 | GEM + CIS | 8.2 | NR | NR | 4.0 (TTP) | NR | NR | 24 | NR | NR | NR | 34 | 95% CI 26 to 42 |
VNB + CIS | 9.0 | NR | NR | 4.1 (TTP) | NR | NR | 20 | NR | NR | NR | 44, p = 0.032 vs GEM + CIS | 95% CI 36 to 53 | |
bGridelli 200345 | GEM + CIS | NR | NR | NR | NR | NR | 0.91 (0.70 to 1.18) | NR | NR | NR | NR | NR | NR |
VNB + CIS | NR | NR | NR | NR | NR | NR | NR | NR | NR | NR | NR | NR | |
Smit 200346 | PAX + CIS | 8.1 | 6.2 to 9.9 | NR | 4.2 | 3.2 to 4.5 | NR | 35.9 | 28.4 to 43.4 | NR | NR | 31.8 | 95% CI 24.4 to 39.2 |
GEM + CIS | 8.9 | 7.8 to 10.5 | NR | 5.1 | 4.5 to 5.7 | NR | 33.1 | 25.8 to 40.4 | NR | NR | 36.8 | 95% CI 29.2 to 44.5 | |
Chen 200451 | PAX + CIS | 11.7 | 7.8 to 15.6 | 1.13 (95% CI 0.76 to 1.68)c | 6 | NR | NR | 46.3 | NR | NR | NR | 38.6 | 95% CI 29.3 to 47.9 |
VNB + CIS | 15.4 | 13.9 to 16.8 | 8.4, p = 0.0344 vs PAX + CIS | NR | NR | 60.9 | NR | NR | NR | 38.6 | 95% CI 29.3 to 47.9 | ||
bDouillard 200553 | DOC + CIS | 8.3 | NR | 0.89 (95% CI 0.66 to 1.20)c | 5 (TTP) | NR | NR | 37 | NR | 17 | NR | 33.9 | 95% CI 25.4 to 43.3 |
VNB + CIS | 9 | NR | NR | 5 (TTP) | NR | NR | 36 | NR | 10 | NR | 26.3 | 95% CI 18.6 to 35.2 | |
Marton 200554 | VNB + CIS | 11 | 9.0 to 13.0 | NR | 5 (TTP) | 4.0 to 6.0 | NR | 39.7 | NR | 13.7 | NR | 32.1 | 95% CI 24.5 to 40.5 |
GEM + CIS | 11 | 9.0 to 13.0 | NR | 5 (TTP) | 4.0 to 6.0 | NR | 44.4 | NR | 16.6 | NR | 26.7 | 95% CI 19.5 to 35.1 | |
Thomas 200658 | GEM + CARB | 10.99 | 6.28 to 13.75 | 4.61 (TTP) | 2.37 to 7.17 | NR | NR | NR | NR | NR | 19.6 | 95% CI 9.8 to 33.1 | |
VNB + CIS | 10.0 | 6.45 to 12.93 | 1.05 (95% CI 0.67 to 1.66)c | 4.87 (TTP) | 3.88 to 6.91 | NR | NR | NR | NR | NR | 29.2 | 95% CI 17.0 to 44.1 | |
Chen 200752 | VNB + CIS | 13.8 | 9.7 to 17.8 | 6.3 | 4.4 to 8.2 | NR | 51.7 | NR | NR | NR | 45.8 | 95% CI 31.7 to 59.9 | |
DOC + CIS | 13 | 5.4 to 20.6 | 0.99 (95% CI 0.58 to 1.69)c | 4.7 | 3.9 to 5.4 | NR | 55.5 | NR | NR | NR | 43.5 | 95% CI 29.2 to 57.8 | |
Helbekkmo 200755 | VNB + CARB | 7.3 | NR | NR | NR | NR | NR | 28 | NR | 7 | NR | NR | NR |
GEM + CARB | 6.4 | NR | NR | NR | NR | NR | 30 | NR | 7 | NR | NR | NR | |
Langer 200756 | GEM + CARB | 6.9 | NR | NR | 3.0 | 1.7 to 4.8 | NR | 25.5 | 13.1 to 38.0 | 13.0 | 3.3 to 22.0 | 23 | 90% CI 13.1 to 34.4 |
PAX + CARB | 6.2 | NR | NR | 3.5 | 2.6 to 6.0 | NR | 19.6 | 8.7 to 30.5 | 7.8 | 0.5 to 15.2 | 14 | 90% CI 6.4 to 23.4 | |
Ohe 200757 | GEM + CIS | 14.0 | NR | NR | 4.0 | NR | NR | 59.6 | NR | 31.5 | NR | 30.1 | NR |
PAX + CARB | 12.3 | NR | NR | 4.5 | NR | NR | 51.0 | NR | 25.5 | NR | 32.4 | NR | |
VNB + CIS | 11.4 | NR | NR | 4.1 | NR | NR | 48.3 | NR | 21.4 | NR | 33.1 | NR | |
Chang 200850 | VNB + CIS | 9.0 | NR | NR | 5.3 | 4.7 to 8.5 | NR | 33.3 | NR | NR | NR | 31 | 95% CI 16 to 46 |
GEM + CIS | 12.9 | NR | NR | 6.6 | 5.2 to 7.6 | NR | 55.9 | NR | NR | NR | 38 | 95% CI 21 to 55 | |
Tan 200959 | VNB + CIS | 9.9 | 8.41 to 11.6 | NR | 4.9 | 4.44 to 5.95 | NR | 39.4 | NR | NR | NR | 27.4 | 95% CI 21.2 to 34.2 |
DOC + CIS | 9.8 | 8.8 to 11.5 | NR | 5.1 | 4.34 to 6.14 | NR | 40.9 | NR | NR | NR | 27.2 | 95% CI 21.0 to 34.2 | |
Treat 201060 | GEM + CARB | 7.9 | 7.1 to 9.2 | NR | 4.3 (TTP) | 4.1 to 5.1 | NR | 33.9 | 29.1 to 38.7 | 11.5 | 8.1 to 14.9 | 25.3 | 95% CI 21.0 to 30.0 |
PAX + CARB | 8.7 | 7.7 to 9.9 | NR | 4.7 (TTP) | 4.2 to 5.5 | NR | 35.6 | 30.7 to 40.4 | 13.3 | 9.7 to 16.9 | 29.8 | 95% CI 25.3 to 34.7 |
Across the trials, median OS ranged from 6.2 to 15.4 months and median PFS/TTP ranged from 3.0 to 8.4 months. Definitions of PFS and TTP varied between trials and are reviewed in more detail in Results of evidence synthesis. Survival rates at 1 year ranged from 19.6% to 60.9% and at 2 years ranged from 7% to 31.5%. Tumour ORR ranged from 14% to 45.8%.
In terms of OS, one trial44 demonstrated statistically significant differences between chemotherapy drug regimens; patients in the DOC + CIS arm had a significantly longer median OS than those in the VNB + CIS arm. However, the HR was not considered to be statistically significant.
In terms of PFS, two trials47,51,61 demonstrated statistically significant differences between chemotherapy drug regimens. In one trial,47 patients treated with GEM + CIS had a significantly longer median PFS than those on PAX + CIS (4.2 months compared with 3.4 months, respectively). In another trial,51 patients treated with VNB + CIS had a significantly longer median PFS than patients treated with PAX + CIS (8.4 months compared with 6.0 months, respectively).
Two trials44,49 showed statistically significant differences for tumour ORR; one trial44 showed that DOC + CIS was associated with a beneficial partial response of the tumour compared with VNB + CIS and another trial49 was associated with a beneficial partial response of the tumour to VNB + CIS compared with GEM + CIS.
Patients with non-squamous disease
The results of the 18 trials included in the NSCLC population with squamous disease are equally applicable for inclusion the NSCLC population with non-squamous disease. However, two additional trials61,62 reported outcomes specifically in subgroups of patients with non-squamous disease. Details of the outcomes assessed in each trial are presented in Table 12.
Trial | Treatment | OS | PFS | Survival 1 year | Survival 2 years | Tumour ORR | |||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Median (months) | 95% CI | HR (95% CI) | Median (months) | 95% CI | HR (95% CI) | % | 95% CI | % | 95% CI | % | 95% CI | ||
Scagliotti 200861 | PEM + CIS | 11.8 | 10.4 to13.2 | 0.81 (0.70 to 0.94); p = 0.005 | 5.3 | 4.8 to 5.7 | 0.90 (0.79 to1.02) | NR | NR | NR | NR | NR | NR |
GEM + CIS | 10.4 | 9.6 to11.2 | 4.7 | 4.4 to 5.4 | NR | NR | NR | NR | NR | NR | |||
Gronberg 200962 | PEM + CARB | 7.8 | 5.4 to 10.1 | 0.96 (0.75 to 1.23)a | NR | NR | NR | NR | NR | NR | NR | NR | NR |
GEM + CARB | 7.5 | 6.0 to 9.4 | NR | NR | NR | NR | NR | NR | NR | NR | NR |
In a trial by Scagliotti et al. 61 comparing PEM + CIS compared with GEM + CIS, OS was statistically significantly superior for patients with non-squamous disease who received PEM + CIS compared with GEM + CIS.
Another PEM trial62 did not find any significant difference in survival when analysing patients with non-squamous disease separately (n = 248: PEM + CARB, 7.8 months; GEM + CARB, 7.5 months; p = 0.77).
Epidermal growth factor receptor mutation-positive population
Three trials were included: two trials63,65 specifically included only patients with EGFR M+ status and in the third, the IPASS,15,64 patients were selected in order to produce a relatively high proportion of patients with EGFR M+ status and reported outcomes by the subgroup of patients with EGFR M+ status. Details of the outcomes assessed in each trial are presented in Table 13.
Trial | Treatment | Median OS | Median PFS | Survival 1 year | Survival 2 years | Tumour ORR | |||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Months | 95% CI | HR (CI) | Months | 95% CI | HR (CI) | % | CI | % | CI | % | CI | ||
IPASS: Mok 200915 and Fukuoka 201164 | GEF | 21.6 | NR | 1.00 (0.76 to 1.33); p = 0.990 | 9.5 | NR | 0.48 (0.36 to 0.64); p< 0.001 | NR | NR | NR | NR | 71.2; p< 0.001 | NR |
PAX + CARB | 21.9 | NR | 6.3 | NR | NR | NR | NR | NR | 47.3 | NR | |||
Maemondo 201063 | GEF | 30.5 | NR | 0.80 (0.52 to 1.23)a | 10.8 | NR | 0.30 (0.22 to 0.41); p< 0.001 | NR | NR | 61.4 | NR | 73.7; p< 0.001 | NR |
PAX + CARB | 23.6 | NR | NR | 5.4 | NR | NR | NR | NR | 46.7 | NR | 30.7 | NR | |
Mitsudomi 201065 | GEF | 30.9 | 24.1b | 1.638 (0.75 to 3.58); p = 0.211 | 9.2 | 8.0 to 13.9 | 0.489 (0.336 to 0.710); p< 0.0001 | NR | NR | NR | NR | 62.1; p< 0.0001 | NR |
DOC + CIS | Not reached | 15.0b | 6.3 | 5.8 to 7.8 | NR | NR | NR | NR | 32.2 | NR |
During the production of this review, final results from the IPASS were published by Fukuoka et al. ;64 the IPASS had already been included in this review as Mok et al. 15 The PFS and ORR outcomes are derived from Mok et al. ;15 however, the OS results were immature and so mature OS outcomes are derived from Fukuoka et al. 64
In terms of median OS, there was no significant difference between GEF and PAX + CARB in two trials15,63,64 and there was no significant difference between GEF and DOC + CIS. 65 The three GEF trials15,63–65 demonstrated a statistically significant benefit of GEF in terms of HR for PFS compared with PAX + CARB, and DOC + CIS.
In the subgroup of 261 patients in the IPASS15,64 who were EGFR M+, median PFS was significantly longer among those who received GEF than among those who received PAX + CARB (HR = 0.48; 95% CI 0.36 to 0.64; p< 0.001). In the subgroup of 176 patients who were EGFR mutation negative (M–), PFS was significantly longer among those who received PAX + CARB (HR = 2.85; 95% CI 2.05 to 3.98; p< 0.001).
In the Maemondo et al. trial,63 median PFS was significantly longer in the GEF group (10.8 months vs 5.4 months) than in the PAX + CARB group (HR = 0.30; 95% CI 0.22 to 0.41; p< 0.001); ORR was also higher in the GEF group (73.7% vs 30.7%; p< 0.001). However, median OS was not significantly different between the treatment arms.
In the Mitsudomi et al. trial,65 PFS was significantly longer in the GEF group than in the DOC + CIS group, with a median PFS time of 9.2 (95% CI 8.0 to 13.9) months compared with 6.3 (95% CI 5.8 to 7.8) months and a HR of 0.489 (95% CI 0.34 to 0.71; log-rank p< 0.0001).
The three GEF trials15,63–65 demonstrated a statistically significant benefit of GEF in terms of tumour ORR compared with PAX + CARB and DOC + CIS.
Results of evidence synthesis
Twenty-three trials15,43–65 were eligible for inclusion in the direct meta-analysis and mixed-treatment comparison analyses, with 11,428 randomised patients.
Eighteen trials43–60 were included in the analyses of the NSCLC population with squamous disease and are included in meta-analysis 1 and mixed-treatment comparison 1. The same 18 trials plus subgroup data from the two PEM trials61,62 were included in the analyses of the NSCLC population with non-squamous disease and are included in meta-analysis 1 and mixed-treatment comparison 1. Two highly selective trials63,65 as well as the IPASS15,64 with an EGFR M+-only subgroup were included in the analyses of the NSCLC population with EGFR M+ status.
Population 1: non-small cell lung cancer patients with squamous disease
Eighteen trials43–60 were eligible for inclusion in the direct meta-analysis and mixed-treatment comparison analyses in the population with squamous disease with 7382 randomised patients. Comparisons between PLAT-based doublets incorporating third-generation chemotherapy drugs were available from 18 trials. 43–60 The characteristics of trials and patients included in these trials are described narratively in Assessment of clinical effectiveness. A summary of chemotherapy regimens showing treatment arms included in the evidence synthesis is presented in Table 14, and shows that the majority of trials (n = 16) had at least one treatment arm containing a CIS regimen; whereas, nine trials had at least one treatment arm containing a CARB regimen.
Trials | GEM + CIS | VNB+CIS | PAX + CIS | DOC + CIS | GEM + CARB | VNB + CARB | PAX + CARB | DOC + CARB |
---|---|---|---|---|---|---|---|---|
Kelly 200148 | ✗ | ✗ | ||||||
Schiller 200247 | ✗ | ✗ | ✗ | ✗ | ||||
Scagliotti 200243 | ✗ | ✗ | ✗ | |||||
Fossella 200344 | ✗ | ✗ | ✗ | |||||
Gebbia 200349 | ✗ | ✗ | ||||||
Gridelli 200345 | ✗ | ✗ | ||||||
Smit 200346 | ✗ | ✗ | ||||||
Chen 200451 | ✗ | ✗ | ||||||
Douillard 200553 | ✗ | ✗ | ||||||
Martoni 200554 | ✗ | ✗ | ||||||
Chen 200752 | ✗ | ✗ | ||||||
Helbekkmo 200755 | ✗ | ✗ | ||||||
Langer 200756 | ✗ | ✗ | ||||||
Ohe 200757 | ✗ | ✗ | ✗ | |||||
Thomas 200658 | ✗ | ✗ | ||||||
Chang 200850 | ✗ | ✗ | ||||||
Tan 200959 | ✗ | ✗ | ||||||
Treat 201060 | ✗ | ✗ | ||||||
Total trials | 9 | 13 | 3 | 5 | 3 | 1 | 6 | 1 |
Seven trials43,44,47,48,56–58 directly compared CIS and CARB when used in combination with one of the third-generation chemotherapy drugs. The non-PLAT-based chemotherapy arms from four trials45,46,49,60 were excluded from all analyses because these treatments are not currently recommended by NICE in the first-line management of patients with advanced NSCLC in the UK.
Overall survival
Overall survival was defined consistently across trials as the time from randomisation to death from any cause. The data points included in the direct meta-analysis and mixed-treatment comparison analyses for OS are presented in Table 15 and six pair-wise comparisons are summarised in Figure 5. Analyses for OS were based on 18 trials43–60 involving 7382 randomly assigned patients and 6081 deaths. The data sources for OS HRs used in the direct meta-analysis and mixed-treatment comparison analyses are also displayed in Table 15. This indicates that not all trials reported HRs for OS. Thus, pre-specified methods (see Evidence synthesis) were used to extract the HR and its variance for each trial that reported any information on OS outcome. The HRs for OS from three trials43,44,57 were extracted directly from the trial papers, data from four trials51–53,58 were obtained by contacting the investigators. In addition, HRs for four trials45–47,50 were extracted from a systematic review79 of the literature as the HRs were not reported in the primary trial publication (three out of four investigators of the primary trials were also co-authors of this systematic review). The remaining seven trials48,49,54–56,59,60 did not report HRs for OS and could not be obtained from trial authors. A network of 18 connected RCTs with six different treatment comparisons is presented in Table 15 and Figure 5. The circles in Figure 5 represent different treatments, and the lines represent direct head-to-head trials informing each comparison. Unconnected circles indicate a lack of direct randomised comparison.
Trials | Data source for HR and variance | GEM + PLAT | VNB + PLAT | PAX + PLAT | DOC + PLAT |
---|---|---|---|---|---|
Schiller 200247 | aLe Chevalier 200579 | ✗ | ✗ | ✗ | |
Kelly 200148 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Scagliotti 200243 | Published trial | ✗ | ✗ | ✗ | |
Fossella 200344 | Published trial | ✗ | ✗ | ||
Gebbia 200349 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Gridelli 200345 | aLe Chevalier 200579 | ✗ | ✗ | ||
Smit 200346 | aLe Chevalier 200579 | ✗ | ✗ | ||
Chen 200451 | Author through e-mail | ✗ | ✗ | ||
Douillard 200553 | Author through e-mail | ✗ | ✗ | ||
Martoni 200554 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Chen 200752 | Author through e-mail | ✗ | ✗ | ||
Helbekkmo 200755 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Langer 200756 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Ohe 200757 | Published trial | ✗ | ✗ | ✗ | |
Thomas 200658 | Author through e-mail | ✗ | ✗ | ||
Chang 200850 | aLe Chevalier 200579 | ✗ | ✗ | ||
Tan 200959 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Treat 201060 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Total trials | 12 | 14 | 8 | 5 |
Result summaries for all pair-wise comparisons between interventions from the direct meta-analysis and the mixed-treatment comparison primary analyses are presented in Table 16. Individual trial results and overall pooled results from both analyses, where available, are also displayed as forest plots for each pair-wise comparison. Results for the mixed-treatment comparison sensitivity analyses of OS are presented in Appendix 12.
Reference treatment vs comparator | Number of data points (trials with head-to-head comparison) | Number of patients in reference treatment/comparator | Number of events (deaths) in reference treatment/comparator | Direct meta-analysis 1 (n = 18), HR (95% CI) | Mixed-treatment comparison 1 (n = 18),HR (95% CI) |
---|---|---|---|---|---|
GEM + PLAT vs VNB + PLAT43,45,49,50,54,55,57,58 | 8 | 1075/1077 | 842/860 | 1.08 (0.98 to 1.20) | 1.09 (0.99 to 1.19) |
GEM + PLAT vs PAX + PLAT43,46,47,56,57,60 | 6 | 1245/1344 | 1053/1186 | 1.03 (0.94 to 1.13) | 1.05 (0.96 to 1.15) |
GEM + PLAT vs DOC + PLAT47 | 1 | 301/304 | 262/271 | 1.06 (0.89 to 1.28) | 1.00 (0.88 to 1.13) |
VNB + PLAT vs PAX + PLAT43,48,51,57 | 4 | 625/630 | 496/481 | 0.98 (0.83 to 1.16) | 0.96 (0.86 to 1.08) |
VNB + PLAT vs DOC + PLAT44,52,53,59 | 4 | 766/1175 | 607/920 | 0.89 (0.78 to 1.00) | 0.92 (0.81 to 1.03) |
PAX + PLAT vs DOC + PLAT47 | 1 | 602/304 | 538/271 | 0.98 (0.76 to 1.27) | 0.95 (0.82 to 1.10) |
Gemcitabine plus platinum compared with vinorelbine plus platinum
Eight head-to-head RCTs43,45,49,50,54,55,57,58 were eligible for inclusion in the direct meta-analysis and mixed-treatment comparison for GEM + PLAT compared with VNB + PLAT, with 2152 randomised patients and 1702 deaths. Seven trials43,45,49,50,54,57,58 used a CIS-based regimen in at least one treatment arm. CARB-based regimens were used in two trials, in one trial55 comparing GEM + CARB with VNB + CARB and in another trial58 comparing GEM + CARB with VNB + CIS.
The HR and 95% CI for each trial are displayed in Figure 6 together with pooled results from meta-analysis 1 and mixed-treatment comparison analyses. Visual examination of Figure 6, the chi-squared test for heterogeneity (p = 0.972), and the I2-statistic (0%) all suggest very good consistency. The pooled OS estimate from meta-analysis 1 (HR = 1.08; 95% CI 0.98 to 1.20) shows a trend in favour of GEM + PLAT, and this was similar to, and consistent with, results of mixed-treatment comparison analyses (see Table 16 and Figure 6). However, as the CI includes a HR of unity we cannot exclude the possibility of no evidence of difference between GEM + PLAT and VNB + PLAT. The median OS of GEM + PLAT ranged from 6.4 to 14 months compared with 7.3 to 11.4 months for VNB + PLAT, with smaller trials showing larger median OS than bigger trials.
Gemcitabine plus platinum compared with paclitaxel plus platinum
Six head-to-head RCTs43,46,47,56,57,60 were eligible under this comparison, with up to 2589 patients and 2239 deaths. In this comparison, GEM was most frequently combined with CIS;43,46,47,56,57 only one trial60 used the GEM + CARB combination. Five trials43,47,56,57,60 evaluated the efficacy of PAX + CARB and two trials46,47 evaluated PAX + CIS. One multiarm trial47 evaluated efficacy for both PAX + CIS and PAX + CARB; however, in our analyses we excluded the PAX + CIS arm from this trial because of limited data points and included the PAX + CARB arm for both direct meta-analysis and mixed-treatment comparison analyses. A sensitivity analysis using PAX + CIS data produced similar results to the direct meta-analysis and the mixed-treatment comparison analyses (see Appendix 12). The HR and 95% CI for each trial are displayed in Figure 7 together with pooled results from meta-analysis 1 and mixed-treatment comparison analyses. Visual examination of Figure 7, the chi-squared test for heterogeneity (p = 0.948), and the I2-statistic (0%) all suggest very good consistency. The pooled OS estimate from meta-analysis 1 (HR = 1.03; 95% CI 0.94 to 1.13) was not statistically significant, and this was similar to and consistent with results from meta-analysis 2 and mixed-treatment comparison analyses (see Table 16 and Figure 7). The results provide insufficient evidence to support a difference in OS between GEM + PLAT and PAX + PLAT treatment. The median OS of GEM + PLAT-treated patients ranged from 6.2 to 14 months compared with 6.9 to 12.3 months in PAX + PLAT trials.
Gemcitabine plus platinum compared with docetaxel plus platinum
One single trial47 provided direct evidence for GEM + PLAT vs DOC + PLAT with 605 patients and 533 deaths contributing to this comparison. The PLAT-based regimen was the same in both treatment arms, implying that results from this comparison can be treated as the efficacy between GEM + CIS compared with DOC + CIS. The HR and 95% CI from this trial are presented in Table 16 together with the pooled HR estimates from the mixed-treatment comparison analyses.
The direct estimate for OS from this trial was not statistically significant (HR = 1.06; 95% CI 0.89 to 1.28). There is insufficient evidence of a difference between GEM + PLAT and DOC + PLAT in terms of survival improvement. The median survival of patients in the GEM + PLAT arm was 8.1 months compared with 7.4 months in the DOC + PLAT arm.
Vinorelbine plus platinum compared with paclitaxel plus platinum
Four head-to-head RCTs43,48,51,57 were eligible for inclusion in this comparison, with 1228 patients and 977 deaths. CIS was used in combination with VNB in all trials; thus, the pooled results from this comparison can be treated as the efficacy of VNB + CIS. Three trials43,48,57 evaluated the efficacy of PAX + CARB; only one trial51 evaluated PAX + CIS.
The HR and 95% CI for individual trials are displayed in Figure 8 together with the pooled results from meta-analysis 1 and mixed-treatment comparison analyses. Visual examination of Figure 8, the chi-squared test for heterogeneity (p = 0.802), and the I2-statistic (0%) all suggest very good consistency. The pooled OS estimate from meta-analysis 1 (HR = 0.98; 95% CI 0.83 to 1.16) was not statistically significant, and was similar and consistent with results from mixed-treatment comparison analyses (see Figure 8). Overall, these results provide insufficient evidence to support a difference in OS between VNB + PLAT and PAX + PLAT. Median OS associated with PAX + PLAT ranged from 8.6 to 12.3 months compared with 8.1 to 15.4 months in the VNB + PLAT trials (see Figure 8).
Vinorelbine plus platinum compared with docetaxel plus platinum
Four head-to-head RCTs44,52,53,59 were eligible for inclusion in this comparison, with 1525 patients and 1941 deaths. The PLAT regimen used in the four trials was CIS combined with VNB or DOC; thus, the pooled results from this comparison can be treated as the efficacy of VNB + CIS compared with DOC + CIS. One of these trials44 also evaluated the efficacy of DOC + CARB in addition to two CIS-based arms; this arm was excluded from the analysis because of insufficient trial data on the DOC + CARB treatment combination. However, we tested the use of the DOC + CARB arm in a sensitivity analysis and the results of the analysis did not show any significant difference.
The HR and 95% CI for individual trials are displayed in the in Figure 9 together with the pooled results from meta-analysis 1 and mixed-treatment comparison analyses. Visual examination of Figure 9, the chi-squared test for heterogeneity (p = 0.906), and the I2-statistic (0%) all suggest very good consistency. The pooled OS estimate from meta-analysis 1 (HR = 0.89; 95% CI 0.78 to 1.00) suggest an advantage to DOC + PLAT with similar and consistent results from mixed-treatment comparison analyses (see Table 16 and Figure 8). However, the CI includes values of HR that may not be clinically important. Median OS for DOC + PLAT ranged from 8.3 to 13 months compared with 9 to 13.8 months in the VNB + PLAT trials (see Figure 9).
Paclitaxel plus platinum compared with docetaxel plus platinum
One head-to-head RCT47 was identified that compared PAX + PLAT and DOC + PLAT with 603 randomised patients and 537 deaths. There were two PAX treatment arms, one consisting of PAX + CIS and the other consisting of PAX + CARB. We used PAX + CARB throughout our analyses as this was the most frequently used combination across trials. The HR and 95% CI from this trial are presented in Table 16 together with the pooled HR estimates from the mixed-treatment comparison analyses.
The direct OS HR and 95% CI from this trial was not statistically significant (HR = 1.02; 95% CI 0.79 to 1.32). These findings indicate insufficient evidence to suggest any difference between PAX + PLAT and DOC + PLAT in terms of survival improvement. The median OS estimates in the treatment arms were similar, with 8.1 months in the PAX + PLAT arm and 7.4 months in the DOC + PLAT arm.
Progression-free survival
The PFS and TTP outcomes in all of the 18 trials considered to be eligible for inclusion in the analysis of the population with squamous disease were reviewed. Eleven trials43,44,46,48–50,55,57–60 were excluded from the PFS analysis since non-standard definitions of PFS appeared to be utilised by the investigators. Seven trials45,47,51–54,56 were included in the PFS analysis detailed in Table 17; however, most of these trials used slightly different definitions of PFS. For instance, in six trials45,47,51–54 PFS was referred to as TTP despite inclusion of death (as in the standard definition of PFS). This was particularly evident in trials designed before PFS was officially recognised as an appropriate surrogate for OS by the US FDA in 2007. 80
Trial | Trial definitions of PFS |
---|---|
Schiller 200247 | TTP was calculated from the date of enrolment to the date of progression or death |
Gridelli 200345 | TTP was defined as the interval from date of random assignment to treatment and date of progression or death |
Chen 200451 | TTP was calculated from the date of initiation of treatment to the date of disease progression or death |
Douillard 200553 | TTP was defined as the time from random assignment to the first evidence of progressive disease or death |
Martoni 200554 | TTP was defined as the time from random assignment to the first evidence of progressive disease or death, if progression was not documented |
Chen 200752 | TTP was calculated from the date of initiation of treatment to the date of disease progression or death |
Langer 200756 | PFS was defined as time from random assignment to tumour progression or death without documented disease progression |
A network of seven connected RCTs45,47,51–54,56 with six different treatment comparisons is presented in Table 18 and Figure 10. The circles in Figure 10 represent different treatments, and the lines represent direct head-to-head trials informing each comparison. Unconnected circles indicate a lack of direct randomised comparison.
Trials | Data source for HR and variance | GEM + PLAT | VNB + PLAT | PAX + PLAT | DOC + PLAT |
---|---|---|---|---|---|
Schiller 200247 | Le Chevalier 200579 | ✗ | ✗ | ✗ | |
Gridelli 200345 | Le Chevalier 200579 | ✗ | ✗ | ||
Chen 200451 | Author through e-mail | ✗ | ✗ | ||
Douillard 200553 | Author through e-mail | ✗ | ✗ | ||
Martoni 200554 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Chen 200752 | Author through e-mail | ✗ | ✗ | ||
Langer 200756 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Total trials | 4 | 5 | 3 | 3 |
The data points included in the direct meta-analyses and mixed-treatment comparison analyses for PFS are presented in Table 18 and six pair-wise comparisons are summarised in Figure 10. Analysis of PFS was based on seven trials involving 3523 patients. 45,47,51–54,56 The HRs for PFS for three trials51–53 were obtained by contacting the investigators. Data from two trials45,47 were extracted from a systematic review79 as HRs were not reported in the primary trial publication (as noted earlier, three out of four investigators in the primary trials were also co-authors for this systematic review). The remaining two trials54,56 did not report HRs for PFS and so the HRs and associated variance were extracted using information on PFS outcome by applying pre-specified methods66,67 as described in Evidence synthesis.
Table 19 shows pair-wise comparison results related to GEM + PLAT, VNB + PLAT, PAX + PLAT and DOC + PLAT from the direct meta-analyses and the mixed-treatment comparison analyses. Where appropriate, direct estimates for each included trial and overall pooled HRs from both sets of analyses are also displayed as forest plots within each pair-wise comparison section. Results for the mixed-treatment comparison sensitivity analyses of PFS are presented in Appendix 13.
Reference treatment vs comparator | Number of data points (trials with head-to-head comparison) | Number of patients in reference treatment/comparator | Number of PFS events in reference treatment/comparator | Direct meta-analysis 1 (n = 9), HR (95% CI) | Mixed-treatment comparison 1, HR (n = 9) (95% CI) |
---|---|---|---|---|---|
GEM + PLAT vs VNB + PLAT45,54 | 2 | 269/269 | 312a | 1.09 (0.87 to 1.38) | 1.06 (0.81 to 1.39) |
GEM + PLAT vs PAX + PLAT47,56 | 2 | 350/656 | 142/304b | 1.17 (1.00 to 1.36) | 1.23 (0.94 to 1.62) |
GEM + PLAT vs DOC + PLAT47 | 1 | 301/304 | 105/114 | 1.15 (0.96 to 1.37) | 1.08 (0.79 to 1.45) |
VNB + PLAT vs PAX + PLAT51 | 1 | 70/70 | 7/14b | 1.52 (1.06 to 2.17) | 1.16 (0.87 to 1.61) |
VNB + PLAT vs DOC + PLAT52,53 | 2 | 168/165 | 92/86 | 0.92 (0.74 to 1.16) | 1.02 (0.78 to 1.36) |
PAX + PLAT vs DOC + PLAT47 | 1 | 602/304 | 130/263b | 0.97 (0.75 to 1.24) | 0.88 (0.62 to 1.21) |
Gemcitabine plus platinum compared with vinorelbine plus platinum
Two head-to-head RCTs45,54 were eligible for this comparison, with 544 randomised patients. Both trials used CIS as the PLAT-based regimen; thus, this analysis can be considered as a comparison of GEM + CIS with VNB + CIS.
The HR and 95% CI for each trial are displayed in Figure 11 together with the pooled results from meta-analysis 1 and mixed-treatment comparison analyses. Visual examination of Figure 11, the chi-squared test for heterogeneity (p = 0.943), and the I2-statistic (0%) all suggest very good consistency. The pooled PFS estimate from meta-analysis 1 (HR = 1.09; 95% CI 0.87 to 1.38) was not statistically significant, and was similar and consistent with results from mixed-treatment comparison analyses (see Table 19 and Figure 11). These findings suggest lack of evidence to support any difference in PFS between GEM + CIS and VNB + CIS. Median PFS estimates were 5 months in both arms in one trial54 and appear to be similar across the two trials.
Gemcitabine plus platinum compared with paclitaxel plus platinum
Two head-to-head RCTs47,56 including 1001 patients contributed to this analysis. Both trials used CIS as the PLAT agent; thus, this analysis can be considered as a comparison of GEM + CIS with PAX + CIS. The HR and 95% CI for each trial are displayed in Figure 12 together with the pooled result from meta-analysis 1 and mixed-treatment comparison analyses. Visual examination of Figure 12, the non-significant chi-squared test for heterogeneity (p = 0.670), and the I2-statistic (0%) all suggest very good consistency. The pooled direct HR (HR = 1.17; 95% CI 1.00 to 1.36; meta-analysis 1) suggests improvement in PFS for GEM + CIS over PAX + CIS, with a borderline statistically significant difference. The direct evidence is consistent with the results of the mixed-treatment comparison analyses. In addition, median PFS estimates were similar in both trials ranging from 3.0 to 4.2 months.
Gemcitabine plus platinum compared with docetaxel plus platinum
One single trial40 provided direct evidence for GEM + PLAT compared with DOC + PLAT with 605 randomised patients and 219 PFS events contributing to this comparison. The PLAT-based regimen was the same in both arms (CIS); thus, this analysis can be considered as a comparison of GEM + CIS with DOC + CIS. The HR and 95% CI from this trial are presented in Table 19 together with the pooled HR estimates from the mixed-treatment comparison analyses.
The direct PFS estimate from this trial was not statistically significant (HR = 1.15; 95% CI 0.96 to 1.37). This appears to be similar and consistent with the results from the mixed-treatment comparison analyses. The trial results and the findings from the mixed-treatment comparison analyses show insufficient evidence to conclude whether or not there are differences in PFS between GEM + CIS and DOC + CIS. The median survival of patients in the GEM + CIS arm was 4.2 months compared with 3.7 months in the DOC + CIS arm.
Vinorelbine plus platinum compared with paclitaxel plus platinum
One single trial51 provided direct evidence for VNB + PLAT compared with PAX + PLAT, with 140 randomised patients contributing to this comparison. The PLAT-based regimen was the same in both arms (CIS); thus, this analysis can be considered as a comparison of VNB + CIS vs PAX + CIS. The HR and 95% CI from this trial are presented in Table 19 together with the pooled HR estimates from the mixed-treatment comparison analyses. The direct estimate of PFS HR from this trial was statistically significant (HR = 1.52; 95% CI 1.06 to 2.17), suggesting an advantage for VNB + CIS. However, although results from the mixed-treatment comparison analysis were consistent in direction of effect, the HR is pulled towards the null value by the dominating indirect evidence and the mixed-treatment comparison analysis is consequently not statistically significant. As the direct evidence comes from one small trial, these findings indicate a degree of uncertainty about the difference between VNB + CIS and PAX + CIS. The median PFS estimate was 8.4 months in the VNB + CIS arm and 6 months in the PAX + CIS arm.
Vinorelbine plus platinum compared with docetaxel plus platinum
Two trials52,53 involving 333 patients explored the role of VNB + PLAT compared with DOC + PLAT. Both trials used CIS; thus, this analysis can be considered as a comparison of VNB + CIS with DOC + CIS. The HR and 95% CI from each trial are displayed in Figure 13 together with the pooled HR estimates from meta-analysis 1 and the mixed-treatment comparison analyses. Visual examination of Figure 13, the non-significant chi-squared test for heterogeneity (p = 445) and the I2-statistic (0%) all suggest very good consistency. PFS appeared favourable to DOC + CIS over VNB + CIS, although this was not statistically significant (HR = 0.92; 95% CI 0.74 to 1.16). Similar findings were observed in the mixed-treatment comparison analyses (see Table 19 and Figure 13). These results indicate insufficient evidence to conclude whether or not there are differences in PFS between VNB + CIS and DOC + CIS. Median PFS estimates in both arms were similar and ranged from 5 to 6.3 months in the VNB arms and 4.7 to 5 months in the DOC arms.
Paclitaxel plus platinum compared with docetaxel plus platinum
One single trial47 provided direct evidence for PAX + PLAT compared with DOC + PLAT, with 906 randomised patients contributing to this comparison. The PLAT-based regimen was the same in both arms (CIS); thus, this analysis can be considered as a comparison of PAX + CIS with DOC + CIS. The HR and 95% CI from this trial are presented in Table 19 together with the pooled HR estimates from mixed-treatment comparison analyses. There is insufficient evidence to conclude whether or not there are differences in PFS between PAX + CIS and DOC + CIS (HR = 0.97; 95% CI 0.75 to 1.24). We observed similar results in the mixed-treatment comparison analyses (see Table 19).
Time to disease progression
Time to disease progression was reported in 7 out of 21 trials. 43,44,46,49,50,58,60 Time to disease progression in RCTs is usually defined as the time from randomisation until objective tumour progression, and does not include death from other causes. We allowed these seven trials to be analysed as a group as their definitions of TTP were similar (Table 20).
Trial | Trial definition of TTP |
---|---|
Scagliotti 200243 | TTP defined as interval between trial enrolment and progressive disease |
Gebbia 200349 | TTP was calculated as the time elapsed from the date of patient's registration until the date of progressive disease or last documented control |
Smit 200346 | Duration of survival and PFS were calculated from the date of randomisation to progression |
Fossella 200344 | TTP was defined as the time from random assignment to first documentation of progressive disease |
Thomas 200658 | TTP was measured from the date of first treatment administration until the time of progressive disease or relapse |
Chang 200850 | TTP was calculated for all patients from the date of randomisation until the date progressive disease was first reported |
Treat 201060 | TTP were assessed using the calculated from the date of randomisation to the date of documented progression |
The data points included in the direct meta-analysis and mixed-treatment comparison analyses for TTP are presented in Table 21. Analyses of TTP were based on outcomes for 3572 randomised patients and approximately 1258 progressive events. Data on log-HR and its variance from two trials43,44 were extracted directly from the published trials, data from two trials were obtained by contacting investigators50,58 and data from one trial46 were extracted from a previously published meta-analysis. 79 Two out of seven trials did not report HRs for TTP49,60 and these were estimated via the pre-specified approaches66,67 described in Evidence synthesis.
Trials | Data source for HR and variance | GEM + PLAT | VNB + PLAT | PAX + PLAT | DOC + PLAT |
---|---|---|---|---|---|
Scagliotti 200243 | Extracted from the published report | ✗ | ✗ | ✗ | |
Gebbia 200349 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Smit 200346 | Le Chevalier 200579 | ✗ | ✗ | ||
Fossella 200344 | Extracted from the published report | ✗ | ✗ | ||
Thomas 200658 | Author through e-mail | ✗ | ✗ | ||
Chang 200850 | Author through e-mail | ✗ | ✗ | ||
Treat 201060 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | ||
Number of trials for TTP analysis | 6 | 5 | 3 | 1 |
The network of comparisons for TTP showing seven connected RCTs43,44,46,49,50,58,60 with six different treatment comparisons are presented in Table 22 and Figure 14. The nodes in Figure 14 represent different treatments and the lines represent direct head-to-head trials informing each comparison. Unconnected nodes indicate lack of direct evidence. The data overview indicate that PLAT-based doublets incorporating GEM, VNB, PAX and DOC generally had at least one data point and had been compared against each other.
Pair-wise comparison (reference treatment/comparator) | Number of patients in reference treatment/comparator | Number of events (TTP) reference treatment/comparator | Number of data points |
---|---|---|---|
GEM + PLAT vs VNB + PLAT43,49,50,58 | 433/436 | 91/82 | 4 |
GEM + PLAT vs PAX + PLAT43,46,60 | 744/742 | 417/423 | 3 |
GEM + PLAT vs DOC + PLAT | XX | XX | 0 |
VNB + PLAT vs PAX + PLAT43 | 203/204 | 34/37 | 1 |
VNB + PLAT vs DOC + PLAT44 | 404/406 | 86/88 | 1 |
PAX + PLAT vs DOC + PLAT | XX | XX | 0 |
Total | 3572 | 1258 |
Table 23 shows pair-wise comparison results between GEM + PLAT, VNB + PLAT, PAX + PLAT and DOC + PLAT from the direct meta-analyses and the mixed-treatment comparison analyses. Direct estimates for each included trial and overall pooled HR from both analyses are also displayed as forest plots within each pair-wise comparison section. Results for the mixed-treatment comparison sensitivity analyses of TTP are presented in Appendix 14. Appendix 15 shows results for the mixed-treatment comparison sensitivity analyses of PFS and TTP combined.
Reference treatment vs comparator | Number of data points | Number of patients in reference treatment/comparator | Number of events (TTP) reference treatment/comparator | Direct meta-analysis 1 (n = 7), HR (95% CI) | Mixed-treatment comparison 1 (n = 7), HR (95% CI) |
---|---|---|---|---|---|
GEM + PLAT vs VNB + PLAT43,49,50,58 | 4 | 433/436 | 91a/82a | 1.03 (0.90 to 1.18) | 1.02 (0.83 to 1.25) |
GEM + PLAT vs PAX + PLAT43,46,60 | 3 | 744/742 | 417a/423a | 1.01 (0.90 to 1.13) | 1.21 (0.73 to 1.99 |
GEM + PLAT vs DOC + PLAT | 0 | XX | XX | XX | 0.98 (0.62 to 1.52) |
VNB + PLAT vs PAX + PLAT43 | 1 | 203/204 | 34a/37a | 0.90 (0.64 to 1.28)b | 0.99 (0.77 to 1.28) |
VNB + PLAT vs DOC + PLAT44 | 1 | 404/406 | 86a/88a | 0.96 (0.70 to 1.31)b | 0.96 (0.65 to 1.43) |
PAX + PLAT vs DOC + PLAT | 0 | XX | XX | XX | 0.98 (0.6 to 1.55) |
Gemcitabine plus platinum compared with vinorelbine plus platinum
Four head-to-head RCTs43,49,50,58 were eligible under this comparison with 869 randomised patients. The PLAT-based regimen for VNB combination in all trials was CIS. For the GEM combination, one trial58 had CARB as the PLAT-based regimen. The HR and 95% CI for each trial are displayed in Figure 15 together with the pooled results from meta-analysis 1 and mixed-treatment comparison analyses. Visual examination of Figure 15, the chi-squared test for heterogeneity (p = 0.992) and the I2-statistic (0%) all suggest very good consistency. The pooled TTP estimates from meta-analysis 1 (HR = 1.03; 95% CI 0.90 to 1.18) were not statistically significant, and this was similar and consistent with results from mixed-treatment comparison analyses (see Table 23 and Figure 15). The estimated point estimate for the TTP HR was slightly different but also consistent with results from meta-analysis 1 and mixed-treatment comparison analyses. These results suggest that there is no evidence to support any difference in TTP between GEM + PLAT and VNB + PLAT. Median TTP estimates ranged from 4.1 to 5.3 months and 4 to 6.6 months in the VNB + PLAT and GEM + PLAT arms, respectively (see Figure 15).
Gemcitabine plus platinum compared with paclitaxel plus platinum
Three head-to-head RCTs PLAT43,46,60 including 1486 patients contributed to this analysis. The treatment arm PLAT combinations used in the trials were not very common among the included trials. For instance, GEM + CIS was used in two trials,43,46 GEM + CARB in one trial,60 PAX + CIS in one trial46 and PAX + CARB in two trials. 43,60 The HR and 95% CI for each trial are displayed in Figure 16 together with the pooled meta-analysis 1 result and HRs from the mixed-treatment comparison analyses. Visual examination of Figure 16, the non-significant chi-squared test for heterogeneity (p = 0.691) and the I2-statistic (0%) all suggest very good consistency. The pooled TTP estimate from the meta-analysis 1 (HR = 1.01; 95% CI 0.90 to 1.13) was not statistically significant, and this was slightly different but consistent with result from mixed-treatment comparison 1 analysis (see Table 19). These results suggest there is no evidence to support any difference in TTP between GEM + PLAT and PAX + PLAT. Median TTP estimates ranged from 4.3 to 5.3 months and 4.2 to 5.5 months in the GEM + PLAT and PAX + PLAT arms, respectively (see Table 19 and Figure 16).
Gemcitabine plus platinum compared with docetaxel plus platinum
There was no trial that directly compared GEM + PLAT and DOC + PLAT and reported TTP outcome. Therefore, TTP comparison between these two drugs was based entirely on results from the mixed-treatment comparison analyses. Results from the mixed-treatment comparison analyses showed no significant difference in TTP between GEM + PLAT and DOC + PLAT (HR = 0.98; 95% CI 0.62 to 1.57; mixed-treatment comparison A). These findings, therefore, indicate insufficient evidence to conclude whether or not there are differences in TTP between GEM + PLAT and DOC + PLAT.
Vinorelbine plus platinum compared with paclitaxel plus platinum
There was one trial43 that directly compared VNB + PLAT and PAX + PLAT, with 407 randomised patients contributing to this comparison. The PLAT-based regimens were different in the two treatment arms (VNB + CIS vs PAX + CARB). The HR and 95% CI from this trial are presented in Table 23 together with the pooled HR estimates from the mixed-treatment comparison analyses. The direct HR estimate for TTP from this trial was 0.90 (95% CI 0.64 to 1.28) and was not statistically significant. The direct evidence appears to be slightly different from the mixed-treatment comparison results where the HRs were estimated at 0.99 (see Table 23). These findings indicate insufficient evidence to conclude whether or not there are differences in TTP between VNB + PLAT and PAX + PLAT. The median TTP estimates were similar between the treatment arms, with 4.6 months for patients in the VNB + PLAT arm compared with 5.5 months in the PAX + PLAT arm.
Vinorelbine plus platinum compared with docetaxel plus platinum
One trial44 involving 810 patients and 174 TTP events explored the role of VNB + PLAT compared with DOC + PLAT and reported TTP. This was a multiarm trial with three treatment arms in which DOC was used in combination with either CIS or CARB. VNB was used in combination with CIS. The combination of DOC + CARB was used in the presented main analyses; however, inclusion of the DOC + CIS arm showed similar results (not presented). The direct HR and 95% CI estimates for the TTP from this trial were not statistically significant (HR = 0.96; 95% CI 0.70 to 1.31). The direct evidence was similar and consistent with the mixed-treatment comparison results as shown in Table 23. These results indicate insufficient evidence to conclude whether or not there are differences in TTP between VNB + PLAT and DOC + PLAT. Median TTP in both arms was similar (approximately 5 months).
Paclitaxel plus platinum compared with docetaxel plus platinum
There was no trial that directly compared PAX + PLAT and DOC + PLAT. Therefore, TTP comparison between these two drugs was based entirely on results from the mixed-treatment comparison analyses. The results from the mixed-treatment comparison analyses showed no significant difference in TTP between PAX + PLAT and DOC + PLAT (HR = 0.98; 95% CI 0.60 to 1.55; mixed-treatment comparison A); this result was consistent across the mixed-treatment comparisons. These findings indicate insufficient evidence to conclude whether or not there are differences in TTP between PAX + PLAT and DOC + PLAT.
Survival risk at year 1 and year 2 post randomization
Year 1 and year 2 survival risk were defined as the probability of survival in intervals of time elapsed from randomisation to year 1 and year 2, respectively. Analyses were based on 17 trials43,44,46–60 involving 7136 randomly assigned patients. There was insufficient information on survival risk at 1 or 2 years for one trial. 45 The proportion of patients still alive or survival rates at year 1 or 2 for all trials were extracted directly from the published reports or indirectly from the survival curves in the published reports.
Results for meta-analysis and mixed-treatment comparison analyses for 1-year and 2-year survival are shown in Appendices 16 and 17, respectively. Analyses for 1-year survival show no evidence to suggest any difference in survival between the third-generation chemotherapy treatments. None of the results from the 2-year analyses were statistically significant with wide CIs that include clinically important values.
Population 2: non-small cell lung cancer patients with non-squamous disease
Current treatment pathways in the UK show that patients with non-squamous locally advanced or metastatic NSCLC can receive any of the four PLAT-based third-generation chemotherapies (GEM, VNB, PAX or DOC) as a first-line treatment. At the time of the initial guideline recommendations,18 the clinical effectiveness of these chemotherapy regimens for non-squamous histology was unknown as clinical effectiveness data were not assessed according to histology. In addition, patients with non-squamous disease can receive PEM + CIS. Despite recommendations for use of these treatments, no comprehensive trial has directly compared all of these treatments in this patient population. Moreover, there are currently only two trials that have direct evidence comparing PEM + CIS61 or PEM + CARB62 with any of the four PLAT-based combinations (i.e. GEM + CIS61 and GEM + CARB62). In this section, comparisons are attempted between the five drugs currently available to patients with non-squamous disease. It is assumed that the treatment effect for all PLAT-based third-generation chemotherapies is not dependent on histology as in the current NICE guideline. 7 The same data points for PLAT-based third-generation chemotherapies that were used for NSCLC patients with squamous disease (n = 18) with the addition of subgroup data from two PEM + PLAT trials were employed. 61,62 Analysis of TTP alone was not performed since none of the PEM studies presented data on TTP.
Table 24 presents a network of trials showing direct evidence on at least one of the outcomes of interest for all chemotherapy trials in the population with non-squamous disease.
Trials | GEM + CIS | VNB + CIS | PAX + CIS | DOC + CIS | PEM + CIS | GEM + CARB | VNB + CARB | PAX + CARB | DOC + CARB | PEM + CARB |
---|---|---|---|---|---|---|---|---|---|---|
Kelly 200148 | ✗ | ✗ | ||||||||
Schiller 200247 | ✗ | ✗ | ✗ | ✗ | ||||||
Scagliotti 200243 | ✗ | ✗ | ✗ | |||||||
Fossella 200344 | ✗ | ✗ | ✗ | |||||||
Gebbia 200349 | ✗ | ✗ | ||||||||
Gridelli 200345 | ✗ | ✗ | ||||||||
Smit 200346 | ✗ | ✗ | ||||||||
Chen 200451 | ✗ | ✗ | ||||||||
Douillard 200553: | ✗ | ✗ | ||||||||
Martoni 200554 | ✗ | ✗ | ||||||||
Chen 200752 | ✗ | ✗ | ||||||||
Helbekkmo 200755 | ✗ | ✗ | ||||||||
Langer 200756 | ✗ | ✗ | ||||||||
Ohe 200757 | ✗ | ✗ | ✗ | |||||||
Thomas 200658 | ✗ | ✗ | ||||||||
Chang 200850 | ✗ | ✗ | ||||||||
Scagliotti 200861 | ✗ | ✗ | ||||||||
Gronberg 200962 | ✗ | ✗ | ||||||||
Tan 200959 | ✗ | ✗ | ||||||||
Treat 201060 | ✗ | ✗ | ||||||||
Total trials | 10 | 13 | 3 | 5 | 1 | 4 | 1 | 6 | 1 | 1 |
Overall survival
The data points included in the direct meta-analysis and mixed-treatment comparison analyses for OS are described in the previous section on squamous population analyses (see Overall survival) in Table 15 and the 15 pair-wise comparisons are summarised in the network diagram (see Figure 5). Analyses for OS were based on all 20 trials, involving 9553 randomly assigned patients and 7608 deaths. OS data used in the meta-analyses and mixed-treatment comparisons for the analyses in the non-squamous disease population were derived from the same 18 trials as the data used in the analyses in the squamous disease population, except for PEM, in which case the data used reflect its licensed population (patients with adenocarcinoma and large cell). Table 25 presents a network of trials showing direct OS evidence for all chemotherapy trials in the population with non-squamous disease. Figure 17 presents a network of trials with OS data used in the mixed-treatment comparison and meta-analysis in the population with non-squamous disease. Result summaries for all pair-wise comparisons between interventions from the direct meta-analysis and the mixed-treatment comparison analyses are presented in Table 26. Individual trial results and overall pooled results from both analyses are displayed as forest plots for each pair-wise comparison where possible. Results from the mixed-treatment comparison sensitivity analyses for OS are shown in Appendix 18.
Trials | Data source for HR and variance | GEM + PLAT | VNB + PLAT | PAX + PLAT | DOC + PLAT | PEM + PLAT |
---|---|---|---|---|---|---|
Schiller 200247 | Le Chevalier 200579 | ✗ | ✗ | ✗ | ||
Kelly 200148 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Scagliotti 200243 | Published trial | ✗ | ✗ | ✗ | ||
Fossella 200344 | Published trial | ✗ | ✗ | |||
Gebbia 200349 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Gridelli 200345 | Le Chevalier 200579 | ✗ | ✗ | |||
Smit 200346 | Le Chevalier 200579 | ✗ | ✗ | |||
Chen 200451 | Author through e-mail | ✗ | ✗ | |||
Douillard 200553 | Author through e-mail | ✗ | ✗ | |||
Martoni 200554 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Chen 200752 | Author through e-mail | ✗ | ✗ | |||
Helbekkmo 200755 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Langer 200756 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Ohe 200757 | Published trial | ✗ | ✗ | ✗ | ||
Thomas 200658 | Author through e-mail | ✗ | ✗ | |||
Chang 200850 | Le Chevalier 200579 | ✗ | ✗ | |||
Scagliotti 200861 | Published trial | ✗ | ✗ | |||
Gronberg 200962 | Author through e-mail | ✗ | ✗ | |||
Tan 200959 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Treat 201060 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Total trials | 14 | 14 | 8 | 5 | 2 |
Results of direct meta-analysis and mixed-treatment comparison for OS in trials in the population with non-squamous disease are shown in Table 26. GEM + PLAT compared with VNB + PLAT had the greatest number of head-to-head trials (eight trials43,45,49,50,54,55,57,58). There was no direct head-to-head trial in population with non-squamous disease for three comparisons; thus, relative treatment effects for these three comparisons are derived entirely from the indirect estimates of the mixed-treatment comparison analysis.
Reference treatment vs comparator | Number of data points (trials with head-to-head comparison) | Number of patients in reference treatment/comparator | Number of events (deaths) in reference treatment/comparator | Direct meta-analysis 1 (n = 20), HR (95% CI) | Mixed-treatment comparison 1 (n = 20), HR (95% CI) |
---|---|---|---|---|---|
GEM + PLAT vs VNB + PLAT43,45,49,50,54,55,57,58 | 8 | 1075/1077 | 842/860 | 1.08 (0.98 to 1.20) | 1.08 (0.99 to 1.18) |
GEM + PLAT vs PAX + PLAT43,46,47,56,57,60 | 6 | 1245/1344 | 1053/1186 | 1.03 (0.94 to 1.13) | 1.06 (0.97 to 1.16) |
GEM + PLAT vs DOC + PLAT47 | 1 | 301/304 | 262/271 | 1.06 (0.89 to 1.28) | 0.99 (0.87 to 1.13) |
GEM + PLAT vs PEM + PLAT61,62 | 2 | 1084/1087 | 755/772 | 0.85 (0.73 to 1.00) | 0.85 (0.74 to 0.98) |
VNB + PLAT vs PAX + PLAT43,48,51,57 | 4 | 625/630 | 496/481 | 0.98 (0.83 to 1.16) | 0.92 (0.68 to 1.24) |
VNB + PLAT vs DOC + PLAT44,52,53,59 | 4 | 766/1175 | 607/920 | 0.89 (0.78 to 1.00) | 0.98 (0.87 to 1.09) |
VNB + PLAT vs PEM + PLAT | 0 | XX | XX | XX | 0.92 (0.82, 1.03) |
PAX + PLAT vs DOC + PLAT47 | 1 | 602/304 | 538/271 | 0.98 (0.76 to 1.27) | 0.79 (0.66 to 0.93) |
PAX + PLAT vs PEM + PLAT | 0 | XX | XX | XX | 0.85 (0.63 to 1.16) |
DOC + PLAT vs PEM + PLAT | 0 | XX | XX | XX | 0.94 (0.81 to 1.09) |
Effect of other PLAT-based third-generation chemotherapies in the population with non-squamous disease (overall survival)
Results from the meta-analysis 1 and mixed-treatment comparison 1 analyses for these pair-wise comparisons (GEM + PLAT vs VNB + PLAT; GEM + PLAT vs PAX + PLAT; GEM + PLAT vs DOC + PLAT; VNB + PLAT vs PAX + PLAT; VNB + PLAT vs DOC + PLAT; and PAX + PLAT vs DOC + PLAT) in the population with non-squamous disease are shown in Table 26. With one exception, these results were identical to the meta-analysis 1 results from the analyses of NSCLC population with squamous disease and similar to the mixed-treatment comparison 1 results from the analyses of NSCLC population with squamous disease (see Overall survival). The exception was PAX + PLAT compared with DOC + PLAT. The mixed-treatment comparison 1 analysis shows a statistically significant difference between PAX + PLAT and DOC + PLAT (HR = 0.79; 95% CI 0.66 to 0.93); although the results of the direct meta-analysis 1 were not significant. There is insufficient evidence to conclude whether or not there is any difference between PAX + PLAT and DOC + PLAT in terms of OS in the population with non-squamous disease.
Gemcitabine plus platinum compared with pemetrexed plus platinum
Two head-to-head RCTs61,62 with 2171 patients were eligible for comparison between GEM and PEM in combination with PLAT in the analysis of the population with non-squamous disease. The two PEM trials61,62 used different PLAT-based regimens, with one trial61 comparing PEM + CIS with GEM + CIS, and the other trial62 comparing PEM + CARB with GEM + CARB.
The HR and 95% CI for each trial are displayed in Figure 18 together with the pooled results from direct meta-analysis 1 and mixed-treatment comparison analyses. Visual examination of Figure 18, the chi-squared test for heterogeneity (p = 0.253) and the I2-statistic (23.4%) all suggest good consistency; the test for heterogeneity is non-significant and the I2-statistic is well below our pre-defined 50% cut-off point for moderate heterogeneity. The pooled OS estimate from meta-analysis 1 (HR = 0.85; 95% CI 0.73 to 1.00) was borderline statistically significant. The mixed-treatment comparison 1 result was statistically significant (HR = 0.85; 95% CI 0.74 to 0.98). These results suggest that there is evidence to support a difference in an OS benefit PEM + PLAT compared with GEM + PLAT. Median OS estimates were quite different across the two trials: in the Scagliotti et al. trial61 median OS was 11.8 compared with 10.4 months and in the Gronberg et al. trial62 median OS was 7.8 compared with 7.5 months for GEM + PLAT compared with PEM + PLAT, respectively. This difference could be because the proportion of patients who received postprogression treatment in the Scagliotti et al. trial61 was much higher than in the Gronberg et al. trial62 (54% and 32%, respectively). Alternatively, it could also be owing to other differences between the trials that we have not been able to explore.
Vinorelbine plus platinum compared with pemetrexed plus platinum
There was no trial that directly compared VNB + PLAT with PEM + PLAT in the population with non-squamous disease. Therefore, the OS comparison between these two treatment combinations was based entirely on results from the mixed-treatment comparison 1 analysis which suggest a non-significant improvement in OS on PEM + PLAT (HR = 0.92; 95% CI 0.82 to 1.03).
Paclitaxel plus platinum compared with pemetrexed plus platinum
There was no trial that directly compared PAX + PLAT and PEM + PLAT in the population with non-squamous disease. Therefore, the OS comparison between these two treatment combinations was based entirely on results from the mixed-treatment comparison 1 analysis which shows a non-statistically significant difference between PAX + PLAT and PEM + PLAT (HR = 0.85; 95% CI 0.63 to 1.16). There is insufficient evidence to conclude whether or not there is any difference between PEM + PLAT and PAX + PLAT in terms of OS in the population with non-squamous disease.
Paclitaxel plus platinum compared with pemetrexed plus platinum
There was no trial that directly compared PAX + PLAT and PEM + PLAT in the population with non-squamous disease. Therefore, the OS comparison between these two treatment combinations was based entirely on results from the mixed-treatment comparison 1 analysis which shows a non-statistically significant difference between PAX + PLAT and PEM + PLAT (HR = 0.85; 95% CI 0.63 to 1.16). There is insufficient evidence to conclude whether or not there is any difference between PEM + PLAT and PAX + PLAT in terms of OS in the population with non-squamous disease.
Progression-free survival
The same data points used in the PFS analysis for the NSCLC population with squamous disease were used in this analysis except that the non-squamous specific PFS estimates for PEM + PLAT compared with GEM + PLAT from the two PEM trials were used. 61,62 Table 27 shows pair-wise comparison results between GEM + PLAT, VNB + PLAT, PAX + PLAT, DOC + PLAT and PEM + PLAT from the direct meta-analyses and the mixed-treatment comparison analyses. Eight trials45,47,51–54,56,61 were included in the PFS analysis detailed in Table 28. Results from the mixed-treatment comparison sensitivity analyses for OS are shown in Appendix 19. Appendices 20 and 21 shows results for direct meta-analysis and mixed-treatment comparison for combined PFS/TTP analyses.
Reference treatment vs comparator | Number of data points (trials with head-to-head comparison) | Number of patients in reference treatment/comparator | Number of PFS events in reference treatment/comparator | Direct meta-analysis 1 (n = 8), HR (95% CI) | Mixed-treatment comparison 1 (n = 8), HR (95% CI) |
---|---|---|---|---|---|
GEM + PLAT vs VNB + PLAT45,54 | 2 | 269/269 | 312a | 1.09 (0.87 to 1.38) | 1.06 (0.78 to 1.66) |
GEM + PLAT vs PAX + PLAT47,56 | 2 | 350/651 | 142/304b | 1.17 (1.00 to 1.36) | 1.23 (0.77 to 1.65) |
GEM + PLAT vs DOC + PLAT47 | 1 | 301/304 | 105/114 | 1.15 (0.96 to 1.37) | 1.08 (0.7 to 1.61) |
GEM + PLAT vs PEM + PLAT61 | 1 | 1084/1087 | NR | 0.90 (0.79 to 1.02) | 0.90 (0.53 to 1.52) |
VNB + PLAT vs PAX + PLAT51 | 1 | 70/70 | 7/14b | 1.52 (1.06 to 2.17) | 1.16 (0.6 to 1.65) |
VNB + PLAT vs DOC + PLAT52,53 | 2 | 168/165 | 92/86 | 0.92 (0.74 to 1.16) | 1.02 (0.61 to 1.44) |
VNB + PLAT vs PEM + PLAT | XX | XX | XX | XX | 0.85 (0.42 to 1.51) |
PAX + PLAT vs DOC + PLAT47 | 1 | 602/304 | 130/263b | 0.97 (0.75 to 1.24) | 0.88 (0.59 to 1.52) |
PAX + PLAT vs PEM + PLAT | XX | XX | XX | XX | 0.73 (0.42 to 1.53) |
DOC + PLAT vs PEM + PLAT | XX | XX | XX | XX | 0.83 (0.43 to 1.65) |
Trial | Trial definitions of PFS |
---|---|
Schiller 200247 | TTP was calculated from the date of enrolment to the date of progression or death |
Gridelli 200345 | TTP was defined as the interval from date of random assignment to treatment and date of progression or death |
Chen 200451 | TTP was calculated from the date of initiation of treatment to the date of disease progression or death |
Douillard 200553 | TTP was defined as the time from random assignment to the first evidence of progressive disease or death |
Martoni 200554 | TTP was defined as the time from random assignment to the first evidence of progressive disease or death, if progression was not documented |
Chen 200752 | TTP was calculated from the date of initiation of treatment to the date of disease progression or death |
Langer 200756 | PFS was defined as time from random assignment to tumour progression or death without documented disease progression |
Scagliotti 200861 | PFS: disease status was assessed according to Response Evaluation Criteria in Solid Tumours (RECIST) |
The data points included in the direct meta-analyses and mixed-treatment comparison analyses for PFS are presented in Table 18 and 10 pair-wise comparisons are summarised in Figure 19. Analysis of PFS was based on eight trials involving 4396 patients. The HRs for PFS from one trial61 were extracted from the trial papers and data from three trials51–53 were obtained by contacting the investigators. Data from two trials45,47 were extracted from a systematic review,79 as HRs were not reported in the primary trial publication (as noted earlier, three out of four investigators in the primary trials were also co-authors for this systematic review). The remaining two trials did not report HRs for PFS54,56 and so the HRs and associated variance were extracted using information on PFS outcome by applying pre-specified methods66,67 as described in Evidence synthesis.
A network of eight connected RCTs45,47,51–54,56,61 with 10 different treatment comparisons are presented in Table 29 and Figure 19. The circles in Figure 19 represent different treatments, and the lines represent direct head-to-head trials informing each comparison. Unconnected circles indicate a lack of direct randomised comparison. There was no direct evidence for the following comparisons: VNB + PLAT compared with PEM + PLAT; PAX + PLAT compared with PEM + PLAT; and DOC + PLAT compared with PEM + PLAT. Relative treatment effects for these comparisons are estimated entirely from the indirect evidence available from the mixed-treatment comparison analysis.
Trials | Data source for HR and variance | GEM + PLAT | VNB + PLAT | PAX + PLAT | DOC + PLAT | PEM + PLAT |
---|---|---|---|---|---|---|
Schiller 200247 | Le Chevalier 200579 | ✗ | ✗ | ✗ | ||
Gridelli 200345 | Le Chevalier 200579 | ✗ | ✗ | |||
Chen 200451 | Author through e-mail | ✗ | ✗ | |||
Douillard 200553 | Author through e-mail | ✗ | ✗ | |||
Martoni 200554 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Chen 200752 | Author through e-mail | ✗ | ✗ | |||
Langer 200756 | Estimated HR from reported survival estimates using common approaches66,67 | ✗ | ✗ | |||
Scagliotti 200861 | Published trial | ✗ | ✗ | |||
Total trials | 5 | 5 | 3 | 3 | 1 |
Effect of other PLAT-based third-generation chemotherapies in the population with non-squamous disease (progression-free survival)
Results from the meta-analysis 1 and mixed-treatment comparison 1 analyses for these pair-wise comparisons (GEM + PLAT vs VNB + PLAT; GEM + PLAT vs PAX + PLAT; GEM + PLAT vs DOC + PLAT; VNB + PLAT vs PAX + PLAT; VNB + PLAT vs DOC + PLAT; and PAX + PLAT vs DOC + PLAT) in the non-squamous population are shown in Table 27.
Gemcitabine plus platinum compared with pemetrexed plus platinum
A single trial61 provided direct evidence for GEM + PLAT compared with PEM + PLAT, with 1725 patients contributing to this comparison. The PLAT-based regimen was the same in both arms; thus, this analysis can be considered as a comparison of GEM and PEM when in combination with CIS. The HR and 95% CI from this trial are presented in Table 27 together with the pooled HR estimates from mixed-treatment comparison analyses. The direct PFS estimated from this trial suggests a potential benefit for PEM + PLAT, although the difference was not statistically significant (HR = 0.90; 95% CI 0.79 to 1.02). This result appears to be similar and consistent with the results from the mixed-treatment comparison 1 analysis, albeit with a much wider CI (HR = 0.90; 95% CI 0.53 to 1.52). These findings, therefore, indicate that there is insufficient evidence to conclude whether or not there are differences in PFS between GEM + PLAT and PEM + PLAT in the population with non-squamous disease.
Vinorelbine plus platinum compared with pemetrexed plus platinum
There was no trial that directly compared VNB + PLAT with PEM + PLAT. Therefore, the PFS comparison between these two drugs was based entirely on the results from the mixed-treatment comparison analyses. The results from the mixed-treatment comparison analyses were not statistically significant (HR = 0.85; 95% CI 0.42 to 1.51; mixed-treatment comparison 1). These findings indicate that there is insufficient evidence to conclude whether or not there are differences in PFS between VNB + PLAT and PEM + PLAT in population with non-squamous disease.
Paclitaxel plus platinum compared with pemetrexed plus platinum
There was no trial that directly compared PAX + PLAT with PEM + PLAT. Therefore, the PFS comparison between these two drugs was based entirely on results from the mixed-treatment comparison 1 analysis. The results from this analysis showed no significant difference in PFS between PAX + PLAT and PEM + PLAT (HR = 0.73; 95% CI 0.42 to 1.53; mixed-treatment comparison 1). These findings indicate that there is insufficient evidence to conclude whether or not there are differences in PFS between PAX + PLAT and PEM + PLAT in the population with non-squamous disease.
Docetaxel plus platinum compared with pemetrexed plus platinum
There was no head-to-head trial that compared DOC + PLAT with PEM + PLAT. Therefore, the PFS comparison between these two drugs was based entirely on results from the mixed-treatment comparison 1 analyses. The results from this analysis showed no significant difference in PFS between DOC + PLAT and PEM + PLAT (HR = 0.83; 95% CI 0.43 to 1.65; mixed-treatment comparison 1). These findings indicate that there is insufficient evidence to conclude whether or not there are differences in PFS between DOC + PLAT and PEM + PLAT in the population with non-squamous disease.
Survival risk at year 1 and year 2 post randomization
Year 1 and year 2 survival risks were defined as the probability of survival in intervals of time elapsed from randomisation to years 1 and 2, respectively. The same data points used in the year 1 and year 2 analyses for the NSCLC population with squamous disease were used in this analysis except that the non-squamous specific estimates for PEM + PLAT compared with GEM + PLAT from the two PEM trials were used. 61,62 Results for meta-analysis and mixed-treatment comparison analyses for 1-year and 2-year survival are shown in Appendices 22 and 23, respectively. None of the results from the 2-year analyses were statistically significant with wide CIs that include clinically important values.
Epidermal growth factor receptor mutation-positive population
Three RCTs15,63–65 that compared GEF to PLAT-based chemotherapy as a first-line treatment of patients with advanced NSCLC were eligible for inclusion in the analysis of the EGFR M+ population. This is the first time that data from the Mitsudomi et al. 65 and Maemondo et al. 63 trials have been used in any meta-analysis or mixed-treatment comparison analyses within this report. All three trials15,63–65 were conducted in patients from East Asian populations who were identified as having adenocarcinoma, and being never or light smokers. Patients were randomised to GEF arms or to chemotherapy. Those in chemotherapy arms received different PLAT-based combinations: PAX + CARB in two trials15,63,64 and DOC + CIS in one trial. 65 Patient characteristics in the three trials15,63–65 appear to be similar; however, the selection of patients differed across the three trials. In the IPASS15,64 patient enrolment was not restricted to patients who were EGFR M+, whereas in the two trials63,65 only EGFR M+ patients were randomised. Therefore, the EGFR M+ data used in this report from the IPASS15,64 are restricted to the subgroup of patients classified as EGFR M+. OS and PFS were reported in all three trials; however, TTP and survival rates were not reported by EGFR M+ status.
Overall survival
Overall survival was defined consistently across the three trials15,63–65 as time from randomisation to death from any cause. The data points included in the direct meta-analyses and mixed-treatment comparison analyses for OS are presented in Table 30 and three pair-wise comparisons are summarised in Figure 20. Analysis for OS was based on the results of all three trials, involving 663 randomly assigned patients. HRs for OS for two trials15,64,65 were extracted from the trial papers and OS data for one trial63 were obtained by contacting the trial investigator. Three treatments (GEF, PAX + PLAT and DOC + PLAT) qualified for inclusion in the analysis of the EGFR M+ population. A network of connected RCTs with different treatment comparisons is presented in Table 30 and Figure 20. The nodes in Figure 20 represent different treatments, and the lines represent direct head-to-head trials informing each comparison. Unconnected nodes indicate lack of direct randomised comparison. There were two direct head-to-head comparisons PAX + PLAT compared with GEF15,63,64 and DOC + PLAT compared with GEF. 65 There was no direct evidence for the PAX + PLAT compared with DOC + PLAT comparison.
Trials | Data source for HR and variance | PAX + PLAT | DOC + PLAT | GEF |
---|---|---|---|---|
Mok 200915 and Fukuoka 201164 | Published trial | ✗ | ✗ | |
Maemondo 201063 | Author through e-mail | ✗ | ✗ | |
Mitsudomi 201065 | Published trial | ✗ | ✗ | |
Total trials | 2 | 1 | 3 | |
Total number of deaths | 95 a | NR | 104 a | |
Total number of patients | 242 | 86 | 246 |
Result summaries for all pair-wise comparisons between interventions from the direct meta-analyses and the mixed-treatment comparison analyses including individual trial results are presented in Table 31.
Reference treatment vs comparator | Total deaths/patients in both arms | Direct meta-analysis (n = 3) HR (95% CI) | Mixed-treatment comparison (n = 3), HR (95% CI) |
---|---|---|---|
PAX + PLAT vs GEF15,63,64 | 199a/448 | 0.94 (0.74 to 1.18) | 0.94 (0.67 to 1.3) |
DOC + PLAT vs GEF65 | NR/172 | 1.64 (0.75 to 3.58)b | 1.64 (0.54 to 4.96) |
PAX + PLAT vs DOC + PLAT | XX | XX | 0.57 (0.18 to 1.81) |
Paclitaxel plus platinum compared with gefitinib
Two head-to-head RCTs15,63,64 including 484 patients were available that compared GEF and PAX + PLAT and contributed to the OS analysis in the EGFR M+ population. Both trials used CARB as the PLAT agent; thus, this analysis can be considered to compare PAX + CARB with GEF. The HR and 95% CI for each trial are displayed in Table 31 and Figure 21 together with the pooled meta-analysis result and HRs from the mixed-treatment comparison analyses. Visual examination of Figure 21, the non-significant chi-squared test for heterogeneity (p = 0.394) and the I2-statistic (0%) all suggest very good consistency. The pooled direct HR (HR = 0.94; 95% CI 0.74 to 1.18; meta-analysis for PAX + PLAT vs GEF) shows no significant difference in OS between GEF and PAX + PLAT. The direct meta-analysis evidence is consistent with the results of the mixed-treatment comparison analyses.
Docetaxel plus platinum compared with gefitinib
One head-to-head RCT65 including 172 patients was available that compared GEF with DOC + PLAT and contributed to the OS analysis in the EGFR M+ population. This trial used CARB as the PLAT agent. The HR and 95% CI for this trial are displayed in Table 31 together with the pooled meta-analysis and HR results from the mixed-treatment comparison analyses. The direct HR (HR = 1.64; 95% CI 0.75 to 3.58) suggests a lack of evidence of any difference in OS between GEF and DOC + PLAT. The direct evidence is consistent with the results of the mixed-treatment comparison analyses in terms of HR and 95% CI (i.e. not statistically significant). The wide CI is a reflection of few deaths from immature OS data (only 10 deaths at data cut-off).
Paclitaxel plus platinum compared with docetaxel plus platinum
There was no trial that directly compared PAX + PLAT with DOC + PLAT in the EGFR M+ population. Therefore, OS comparison between these two drugs was estimated from the mixed-treatment comparison analysis that included three trials. 15,63–65 The results from the mixed-treatment comparison analysis showed no significant difference in OS between PAX + PLAT and DOC + PLAT (HR = 0.57; 95% CI 0.18 to 1.81). These findings indicate that there is insufficient evidence to conclude whether or not there are differences in OS between PAX +PLAT and DOC + PLAT in EGFR M+ patients; indeed, the wide CIs associated with the HR may point to clinically important differences in both directions.
Progression-free survival
The definition of PFS was consistent across the three trials15,63–65 and all trials adopted Response Evaluation Criteria in Solid Tumours (RECIST)81 to assess tumour progression. The HRs for PFS were available from all trials. The data points included in the direct meta-analyses and mixed-treatment comparison analyses for PFS are presented in Table 32 and three pair-wise comparisons are summarised in Figure 22. Analysis for PFS was based on outcome data for 660 randomly assigned patients. Three treatments (GEF, PAX + PLAT and DOC + PLAT) qualified for inclusion in the analysis in the EGFR M+ population. A network of connected RCTs with different treatment comparisons is presented in Table 32 and Figure 22. The nodes represent different treatments, and the lines represent direct head-to-head trials informing each comparison. Unconnected nodes indicate lack of direct randomised comparison. There were two direct head-to-head comparisons (PAX + PLAT vs GEF and DOC + PLAT vs GEF). There was no direct evidence for PAX + PLAT compare with DOC + PLAT.
Trials | Data source for HR and variance | PAX + PLAT | DOC + PLAT | GEF |
---|---|---|---|---|
Mok 200915 and Fukuoka 201164 | Published trial | ✗ | ✗ | |
Maemondo 201063 | Published trial | ✗ | ✗ | |
Mitsudomi 201065 | Published trial | ✗ | ✗ | |
Total trials | 2 | 1 | 3 | |
Total number of PFS events | Not reported in all trials63,65 | Not reported in all trials63,65 | Not reported in all trials63,65 | |
Total number of patients | 242 | 86 | 332 |
Result summaries for all pair-wise comparisons between interventions from the direct meta-analyses and the mixed-treatment comparison analyses including individual trials results are presented in Table 33 and Figure 23.
Reference treatment vs comparator | Total PFS events/patients in both arms | Direct meta-analysis (n = 3), HR (95% CI) | Mixed-treatment comparison (n = 3), HR (95% CI) |
---|---|---|---|
PAX + PLAT vs GEF15,63,64 | NR/488 | 0.38 (0.24 to 0.60) | 0.39 (0.29 to 0.52) |
DOC + PLAT vs GEF65 | NR/172 | 0.49 (0.33 to 0.73) a | 0.49 (0.28 to 0.86) |
PAX + PLAT vs DOC + PLAT | XX | XX | 0.79 (0.42 to 1.48) |
Paclitaxel plus platinum compared with gefitinib
Two head-to-head RCTs15,63,64 including 491 patients were available that compared GEF and PAX + PLAT and contributed to the PFS analysis in the EGFR M+ population. Both trials used CARB as the PLAT. The HR and 95% CI for each trial are displayed in Table 33 together with the pooled meta-analysis results and HRs from mixed-treatment comparison analyses. Visual examination of Figure 23, a statistically significant chi-squared test for heterogeneity (p = 0.03) and the I2-statistic (78.8%) all suggested inconsistency in the direct evidence from the two trials15,63,64 comparing PAX + PLAT and GEF. However, both trials showed that GEF is significantly better than PAX + PLAT in terms of improving PFS in patients with the EGFR M+ population. The pooled direct meta-analysis HR (HR = 0.38; 95% CI 0.24 to 0.60) suggests evidence of a significant difference in PFS between GEF and PAX + PLAT. The direct evidence is consistent with the results of the mixed-treatment comparison analysis in terms of HR and 95% CI (i.e. statistically significant). Further investigation is required to explore the heterogeneity between trials. Given the significant evidence of heterogeneity, which is above a pre-specified 50% (moderate heterogeneity68,69) in the direct meta-analysis between PAX + PLAT and GEF, the pooled PFS HR should be interpreted with a degree of caution.
Docetaxel plus platinum compared with gefitinib
One head-to-head RCT65 including 172 patients was available that compared GEF with DOC + PLAT and contributed to the PFS analysis in the EGFR M+ population. This trial used CARB as the PLAT. The HR and 95% CI for this trial are displayed in Table 33 together with the pooled result and HRs from mixed-treatment comparison analyses. The direct PFS HR (HR = 0.49; 95% CI 0.33 to 0.73) suggests evidence of a significant difference in PFS between GEF and DOC + PLAT. The direct evidence is consistent with mixed-treatment comparison analysis in terms of HR and 95% CI (i.e. statistically significant in favour of GEF). In addition, median PFS were 6.3 and 9.2 months in the DOC + PLAT and GEF arms, respectively.
Paclitaxel plus platinum compared with docetaxel plus platinum
There was no trial that directly compared PAX + PLAT with DOC + PLAT in the EGFR M+ population. Therefore, the PFS comparison between these two chemotherapy treatments was estimated from the mixed-treatment comparison analysis that included three trials. 15,63,65 The findings indicate that there is insufficient evidence to conclude whether or not there are differences in PFS between PAX + PLAT and DOC + PLAT; the wide CIs associated with the HR may point to clinically important differences in both directions (HR = 0.79; 95% CI 0.42 to 1.48).
Adverse events
This review presents data on AEs that were categorised in the published trials as being grade 3 and 4. Appendix 24–26 provides details of the proportion of patients who experience grade 3–4 AEs within each individual trial and toxic deaths reported within each trial.
The trials reported a diverse range of AEs and the definitions of AEs (including grading) varied between trials, making it difficult to summarise AE data. Tables 34–37 show statistically significant AEs reported within the trials by pair-wise group comparisons.
Grade 3–4 AEs | GEM + PLAT | VNB + PLAT |
---|---|---|
Haematological toxicity | ||
Anaemia | Helbekkmo 200755 | |
Leucopenia | Helbekkmo 200755 | |
Neutropenia | Chang 200850 | |
Martoni 200554 | ||
Scagliotti 200243 | ||
Thomas 200658 | ||
Platelets | Gebbia 200349 | |
Thrombocytopenia | Chang 200850 | |
Helbekkmo 200755 | ||
Martoni 200554 | ||
Scagliotti 200243 | ||
Thomas 200658 | ||
Non-haematological toxicity | ||
Asthenia | Gebbia 200349 | |
Phlebitis | Gebbia 200349 | |
Vomiting | Chang 200850 | |
Scagliotti 200243 |
Grade 3–4 | GEM + PLAT | PAX + PLAT |
---|---|---|
Haematological toxicity | ||
Anaemia | Schiller 200247 | |
Smit 200346 | ||
Treat 201060 | ||
Red blood cell transfusion | Scagliotti 200243 | |
Smit 200346 | ||
Platelet transfusion | Scagliotti 200243 | |
Treat 201060 | ||
Febrile neutropenia | Schiller 200247 | |
Haemorrhage | Smit 200346 Treat 201060 | |
Neutropenia | Treat 201060 | Langer 200756 |
Platelet count | Schiller 200247 | |
Thrombocytopenia | Langer 200756 | |
Scagliotti 200243 | ||
Smit 200346 | ||
Treat 201060 | ||
Non-haematological toxicity | ||
Alopecia | ||
Arthralgia | Treat 201060 | |
Myelosuppression | Smit 200346 | |
Nausea/vomiting | Langer 200756 | |
Renal toxic effects | Schiller 200247 | |
Sensory neuropathy | Langer 200756 | |
Treat 201060 | ||
Fatigue | Langer 200756 |
Grade 3–4 | VNB + PLAT | PAX + PLAT |
---|---|---|
Haematological toxicity | ||
Anaemia | Scagliotti 200243 | |
Blood transfusions | Scagliotti 200243 | |
Leucopenia | Chen 200451 | |
Kelly 200148 | ||
Neutropenia | Chen 200451 | |
Kelly 200148 | ||
Scagliotti 200243 | ||
Thrombocytopenia | Scagliotti 200243 | |
Non-haematological toxicity | ||
Constipation | ||
Myalgia | Scagliotti 200243 | Chen 200451 |
Myelosuppression | Chen 200451 | |
Nausea/vomiting | Kelly 200148 | |
Scagliotti 200243 | ||
Peripheral neuropathy | Chen 200451 | |
Kelly 200148 |
Grade 3–4 | VNB + PLAT | DOC + PLAT |
---|---|---|
Haematological toxicity | ||
Anaemia | Douillard 200553 | |
Fossella 200344 | ||
Tan 200959 | ||
Febrile neutropenia | Douillard 200553 | |
Tan 200959 | ||
Neutropenia | Douillard 200553 | Tan 200959 |
Non-haematological toxicity | ||
Alopecia | Chen 200752 | |
Douillard 200553 | ||
Diarrhoea | Chen 200752 | |
Fossella 200344 | ||
Infection | Douillard 200553 | |
Nail disorder | Douillard 200553 | |
Nausea/vomiting | Fossella 200344 | Douillard 200553 |
Table 34 shows that five trials43,50,54,55,58 report significantly higher levels of thrombocytopenia in GEM + PLAT arms compared with VNB + PLAT arms. However, four trials43,50,54,58 report significantly greater levels of neutropenia in VNB + PLAT arms compared with GEM + PLAT arms.
Table 35 indicates that haematological toxicity is more common in patients receiving GEM + PLAT (anaemia, blood transfusions, haemorrhage and thrombocytopenia) compared with patients receiving PAX + PLAT.
Table 36 indicates that haematological toxicity is more common in patients treated with VNB + PLAT compared with patients treated with PAX + PLAT.
Table 37 shows that anaemia and febrile neutropenia were significantly more common in patients treated with VNB + PLAT compared with patients treated with DOC + PLAT. Diarrhoea and alopecia are more common in patients treated with DOC + PLAT compared with patients treated with VNB + PLAT.
Other data relating to AEs, including details of treatment administration and relative dose intensity (RDI), are presented in Appendix 27. Trials reported details of median time to complete treatment, percentage of patients who completed treatment as per protocol, details of chemotherapy dose reductions and delays, and median number of chemotherapy cycles.
The number of patients discontinued who treatment because of toxicity was significantly higher in the VNB + CIS treatment arm than in the PAX + CARB or DOC + CIS arms. In the trial by Kelly et al. ,48 discontinuation was significantly higher, and completion of treatment and RDI significantly lower, in the VNB + CIS arm than in the PAX + CARB arm. In the trial by Fossella et al. ,44 patients in the DOC + CIS and the DOC + CARB arms had a higher median number of chemotherapy cycles, higher RDI and completion rates and fewer treatment delays than those in the VNB + CIS arm. Patients in the DOC + CIS arm of the trial by Douillard et al. 53 had a higher median RDI, fewer cycle delays and fewer chemotherapy dose reductions compared with the VNB + CIS arm.
There was higher RDI for GEM compared with VNB in the trial by Thomas et al. 58 However, in a trial by Helbekkmo et al. ,55 a significantly greater percentage of patients in the GEM arm had > 24 days between chemotherapy courses and delayed or cancelled chemotherapy at day 8 due to haematological toxicity compared with the VNB arm.
In the trial by Scagliotti et al. ,61 dose adjustments were less frequent and RDI was higher in the PEM arm than in the GEM arm. In the trial by Gronberg et al. ,62 the mean number of cycles was higher and significantly more patients in the PEM arm than in the GEM arm completed four cycles, and without delays.
In the trial by Schiller et al. ,47 treatment with GEM + CIS was more likely to cause grade 3, 4 or 5 renal toxicity and 27% of patients who received GEM + CIS were withdrawn from the trial owing to complications of therapy, compared with 15% of patients in the PAX + CIS arm (p< 0.001).
Gefitinib is associated with significantly lower severe toxic AEs compared with PAX + CARB15,63,64 and DOC + CIS65 with the exception of liver dysfunction. 65 In one trial, GEF15,64 was associated with a lower rate of AEs leading to discontinuation of the drug (6.9% vs 13.6%) and a lower rate of dose modification due to toxic effects (16.1% vs 35.2% for CARB and 37.5% for PAX). AEs leading to death occurred in 3.8% of the patients treated with GEF and in 2.7% of the patients treated with PAX + CARB. Interstitial lung disease was significantly more common in patients treated with GEF than in those treated with PAX + CARB or DOC + CIS, including one fatality in each trial. 15,63–65
Table 38 shows the top 10 AEs that occur in the greatest proportion of patients across all arms that use each chemotherapy. The AEs are all grades 3 and 4 (with the exception of one trial58 in Table 38 in which the grades for febrile neutropenia were not specified for either arm); however, reporting of AEs varied (for example grade 3 only, grade 4 only, grade 3 or grade 4 and grade 3 plus grade 4). Certain AEs were grouped together: anaemia haemoglobin was categorised into anaemia; neutrophils to neutropenia; sensory neuropathy, motor neuropathy and neurotoxic effects were all grouped into neuropathy.
DOC + PLAT | GEF | GEM + PLAT | PAX + PLAT | PEM + PLAT | VNB + PLAT |
---|---|---|---|---|---|
Neutropenia, 71.4% | Aminotransferase, elevation, 33.8% | Granulocytopenia, 48.8% | Neutropenia, 62.5% | Granulocytopenia, 37.9% | Neutropenia, 68.3% |
Leucopenia, 43.5% | Appetite loss, 5.3% | Asthenia, 40.3% | Leucopenia, 31.9% | Blood transfusions, 26.9% | Leucopenia, 47.2% |
Weakness, 16.0% | Rash/acne, 3.3% | Neutropenia, 36.4% | Weakness, 14.5% | Infection, 16.4% | Oedema, 24.0% |
Pneumonitis, 11.5% | Toxic deaths, 3.1% | Thrombocytopenia, 34.6% | Cancer pain, 13.2% | Neutropenia, 15.1% | Anaemia, 19.3% |
Anaemia, 11.2% | Diarrhoea, 3.1% | Anorexia, 27.0% | Nausea, 10.3% | Alopecia, 11.9% | Phlebitis, 15.7% |
Asthenia, 10.2% | Neutropenia, 2.8% | Leucopenia, 20.1% | Anaemia, 10.0% | Leucopenia, 8.2% | Nausea/vomiting, 11.5% |
Nausea, 9.9% | Pneumonitis, 2.6% | Transfusion, 18.5% | Lethargy, 9.4% | Thrombocytopenia, 8.1% | Vomiting, 10.3% |
Vomiting, 9.8% | Fatigue, 2.5% | Alopecia, 17.2% | Thrombocytopenia, 8.3% | Anaemia, 7.0% | Nausea, 9.9% |
Cancer pain, 8.4% | Infection, 1.8% | Weakness, 17.0% | Neuropathy, 7.9% | Fatigue, 6.7% | Asthenia, 9.4% |
Infection, 7.5% | Anaemia, 1.6% | Anaemia, 16.5% | Vomiting, 7.4% | Nausea, 6.2% | Pain, 8.3% |
Table 38 compares the profile of AEs within each chemotherapy regimen and should not be used to compare toxicities across the different drug regimens. Table 38 shows each drug regimen differs in toxicity profile in terms of percentage of AE.
Table 38 shows that the most common AEs are neutropenia, anaemia and leucopenia. Neutropenia is the top AE for VNB, PAX and DOC and granulocytopenia is the top AE for GEM and PEM. Neutropenia, leucopenia, granulocytopenia all describe a fall in the white blood count and so the common AEs are similar across all the chemotherapy drugs with the exception of GEF, which appears to have a different toxicity profile; the top AE for GEF is aminotransferase elevation. The highest proportion experiencing neutropenia (71%) was among those taking DOC.
Quality of life
Twelve trials15,43–46,48,51,52,55,57,59,62,64 reported QoL outcomes and are listed in Appendix 28. It is surprising, given the importance of QoL, that 1147,49,50,53,54,56,58,60,61,63,65 of the 23 trials do not report QoL data, including three trials60,63,65 that were published in 2010. This could indicate outcome reporting bias, with trial authors failing to present results because they are not statistically significant. QoL was the primary outcome in two trials45,62 and, in the trial by Gridelli et al. ,45 QoL data were assessed according to GEM + VNB compared with PLAT-based chemotherapy (the GEM + VNB combination is not included in this review). Meta-analysis was not performed for QoL data owing to limited data and variability in outcome assessment measures.
A number of instruments/tools that measure QoL were employed in the included trials. The EORTC QLQ-C3029 and the lung cancer-specific module QLQ-LC1330 were used in five trials, the LCSS31 by three trials, and the FACT-L32 questionnaire by three trials. 15,48,57,64
Seven trials48,45,51,52,55,59,62 reported no significant difference in QoL between treatment groups. Four trials15,43,44,46,64 reported some significant differences between treatment groups for QoL; however, in one of these trials,43 results after two cycles of chemotherapy favoured the PAX + CARB arm over the VNB + CIS arm, and results after four cycles favoured the VNB + CIS arm.
In one trial,15,64 significantly more patients in the GEF group than in the PAX + CARB group had a clinically relevant improvement in QoL, as assessed by scores on the FACT-L questionnaire (odds ratio = 1.34; 95% CI 1.06 to 1.69; p = 0.01) and by scores on the Trial Outcome Index (TOI) (which is the sum of the physical well-being, functional well-being and lung cancer subscale scores of FACT-L; odds ratio = 1.78; 95% CI 1.40 to 2.26; p< 0.001).
In another trial46 comparing GEM + CIS with PAX + CIS, no significant difference in global QoL was observed; however, a statistically and clinically significant overall improvement was observed for peripheral neuropathy and alopecia in the GEM + CIS arm compared with the PAX + CIS arm.
Patients treated with DOC + PLAT reported consistently improved global QoL compared with patients treated with VNB + CIS, who generally experienced deterioration in QoL in the trial by Fossella et al. 44
In summary, PAX + PLAT may be associated with worse QoL for alopecia and peripheral neuropathy compared with VNB + PLAT and GEM + PLAT. GEM + PLAT may be associated with better QoL for peripheral neuropathy compared with PAX + PLAT and VNB + PLAT; however, there is a paucity of QoL data available to draw any firm conclusion.
Discussion
Summary of key results
Twenty-three trials that compared any first-line chemotherapy treatment currently licensed in Europe and recommended by NICE were included within the analyses; publication dates ranged from 2001 to 2010. Of the 20 multicentre trials, six had international centres. 15,44–46,59,61,64 All included trials were published in English. There are five Phase II trials,51–53,56,58 16 Phase III trials15,43–46,48,49,54,55,57,59–65 and two trials47,50 with phase undefined. Ten trials15,43,44,53,57–62,64 were funded solely by pharmaceutical companies.
Evidence for the NSCLC population with squamous disease included 18 trials43–62 (> 7000 patients and > 6000 deaths); these same 18 trials plus an additional two trials of PEM + PLAT with subgroup data provided evidence for the population with non-squamous disease. Three trials15,63–65 conducted entirely within East Asian countries provided evidence for the NSCLC population with EGFR M+ status.
The PLAT-based doublets of DOC, GEM, PAX and VNB had relatively more data points for all outcomes than the newer PEM + PLAT regimen and GEF monotherapy. In general, there was consistency between the results of the direct meta-analyses and the mixed-treatment comparison analyses, and very good consistency across individual trials in the within-group comparisons.
Overall, the quality of the included RCTs was poorer than expected – there were few trials with fully reported methods and the definitions of the health outcomes used often differed between trials. In addition, it is generally agreed that RCTs typically include patients who are generally fitter and younger than patients receiving treatment in routine clinical practice and that outcomes from RCTs are not always of the same magnitude as those gained from routine care. Caution is therefore required when interpreting and comparing the results of these trials, in particular the results generated through meta-analysis and mixed-treatment comparison.
Non-small cell lung cancer population with squamous disease
The evidence related to outcomes for patients with squamous disease demonstrates that there are no statistically significant differences in OS between any of the four third-generation chemotherapy treatments (DOC + PLAT, GEM + PLAT, PAX + PLAT or VNB + PLAT). However, both the direct and indirect evidence suggest a potential advantage in terms of OS for GEM + PLAT (direct meta-analysis 1, HR = 1.08; 95% CI 0.98 to 1.20) and for DOC + PLAT (direct meta-analysis 1, HR = 0.89; 95% CI 0.78 to 1.00; mixed-treatment comparison 1, HR = 0.92; 95% CI 0.81 to 1.03) compared with VNB + PLAT, although this advantage is not statistically significant. Analyses of 1- and 2-year survival support this conclusion.
Only seven trials45,47,51–54,56 were included in the PFS analysis and the majority of these trials used slightly different definitions of PFS. There was no evidence of any significant difference in PFS for GEM + PLAT compared with VNB + PLAT. There was insufficient evidence to conclude whether or not there were any statistically significant differences in PFS between the other third-generation chemotherapy comparators.
A further seven trials43,44,46,49,50,58,60 reported results for the outcome TTP and there was no evidence of any statistically significant difference in TTP for GEM + PLAT compared with VNB + PLAT and GEM + PLAT compared with PAX + PLAT or between the other third-generation chemotherapy comparators.
Non-small cell lung cancer population with non-squamous disease
For patients with non-squamous disease there is evidence to suggest that PEM + PLAT increases OS compared with GEM + PLAT (direct meta-analysis 1, HR = 0.85; 95% CI 0.73 to 1.00; mixed-treatment comparison 1, HR = 0.85; 95% CI 0.74 to 0.98). There is no evidence to conclude that there is any statistically significant difference between any of the other chemotherapy treatments in terms of increasing OS for patients with non-squamous disease. Both the direct and indirect evidence suggest a potential advantage for GEM + PLAT compared with VNB + PLAT in terms of OS; however, this advantage is not statistically significant. Both the direct and indirect evidence suggest a potential advantage for DOC + PLAT compared with VNB + PLAT in terms of OS; however, this advantage is borderline statistically significant (direct meta-analysis 1, HR = 0.89; 95% CI 0.78 to 1.00; mixed-treatment comparison 1, HR = 0.92; 95% CI 0.81 to 1.03). The mixed-treatment comparison 1 analysis shows a statistically significant difference between PAX + PLAT and DOC + PLAT (HR = 0.79; 95% CI 0.66 to 0.93); however, the direct meta-analysis 1 was not significant.
Epidermal growth factor receptor mutation-positive population
For patients with EGFR M+ status, there is no statistically significant difference in OS between GEF compared with PAX + PLAT and between GEF compared with DOC + PLAT. There is evidence of a statistically significant improvement in PFS with GEF compared with DOC + PLAT. Although there is also evidence of a statistically significant improvement in PFS with GEF compared with PAX + PLAT the significant heterogeneity between trials means the PFS results should be viewed with caution.
Generalisability of results
A limitation to this review is the generalisability of the patients in the included trials to the population with NSCLC in the UK. In the earlier trials of third-generation chemotherapy drugs, patients with NSCLC were treated as a generic group when in fact it is now accepted that they are a mixed population comprising patients with squamous and non-squamous disease. Earlier trials that assessed the clinical effectiveness of the third-generation chemotherapy drugs did not differentiate on factors such as histology or genetic markers. The mix of patient population is now expected to be taken into consideration at the time of trial design as demonstrated in the PEM and GEF trials. Making comparisons across the six available first-line chemotherapy treatments is therefore limited by the comparability of the treatment populations in the published trials.
In addition, it is questionable whether or not the results from four trials based entirely in East Asian populations50–52,57 are generalisable to UK clinical practice. The evidence relating to the EGFR M+ populations is based entirely on patients within East Asian populations. There are no relevant UK-based trial data for patients with EGFR M+ status. Evidence suggests that East Asian populations with NSCLC have a more favourable prognosis compared with non-East Asian populations. 82 Although EGFR mutation rates are likely to be quite different in different countries, actual response to chemotherapy may not differ in patients with the same mutation status.
Strengths and limitations
This is the first comprehensive systematic review and economic evaluation of all first-line chemotherapy options that are currently licensed for use in the UK and recommended by NICE for patients with advanced NSCLC. This includes PLAT-based doublets with DOC, GEM, PAX, PEM and VNB and also GEF monotherapy. This review highlights that research in this area is evolving rapidly with advances seen in relation to histology and genetic subgroups within the NSCLC population.
There was no direct evidence identified for six different comparisons of chemotherapy drugs which was a limitation; however, a particular strength of this review is that it is the first review to use indirect There was no direct evidence identified for six different comparisons of chemotherapy drugs which was a limitation; however, a particular strength of this review is that it is the first review to use indirect
This report was limited in its analyses of AEs mainly because trials varied in the way AEs were defined, measured and reported. For example, grade 3 and 4 AEs were reported separately or in aggregate. For this reason, where trials reported within-trial significant differences between chemotherapy treatment groups, these differences were highlighted in the report, although this approach may be hampered by the potential for selective reporting bias by the authors. AE data are often sparse with wide CIs, which means that individual trials lack the power to detect significant differences.
This report highlights the top 10 AEs that occurred within each chemotherapy regimen and are produced by weighted average grade 3–4 AEs which are calculated by the number of events related to the toxicity in all arms from all included trials of each chemotherapy regimen, divided by the number of patients who experience these events in all arms. However, this approach loses all the benefits of randomisation and a comparative control group because it splits arm-level data. This approach does not provide information about the comparative harms of chemotherapy (which would assist in balancing the potential benefit and risk of each chemotherapy regimen) and it is merely intended to highlight the different toxicity profiles of the six chemotherapy regimens. AE data were not reported by the three populations used to assess survival data or for cost-effectiveness analysis (CEA); patients with squamous and non-squamous disease.
Further research is required regarding the clinical significance of any of the reported AEs, also the significance to patients in terms of QoL and any differences in terms of costs. AE reporting needs to be standardised and reported consistently across trials if future comparisons are going to be possible.
Overall survival is an important outcome in deciding which chemotherapy drug a patient should receive, but this needs to be considered alongside the toxicity of chemotherapy therapy and the symptomatic benefits of therapy (QoL). A lack of reporting of QoL data is a feature of the great majority of trials assessing outcomes of treatment for patients with NSCLC. This, despite its relevance to patients and clinicians, is a major shortcoming of lung cancer research. Measuring QoL outcomes in patients with advanced NSCLC is difficult mainly because of the severity of symptoms, the side effects of chemotherapy treatment and early deaths associated with NSCLC. However, a British Thoracic Oncology Group Phase III trial83 [British Thoracic Oncology Group Trial 2 (BTOG2)] comparing GEM (1250 mg/m2 day 1 and day 8) with either CIS 80 mg/m2, CIS 50 mg/m2 or CARB area under curve (AUC) 6 is the largest study to date to collect QoL data on patients with NSCLC. QoL was measured at each chemotherapy cycle and follow-up visit using standard, validated questionnaires. More than 8000 questionnaires were returned from 1363 patients with compliance around 90% during the treatment period. This trial shows that it is feasible to collect QoL data in patients with PS 0–2, stage IIIB/IV NSCLC disease within a clinical trial setting.
Carboplatin and CIS were grouped together and treated as similar for the clinical effectiveness analyses, based on NICE guidelines7 which recommend that either CARB or CIS may be administered depending on the balance of toxicity, efficacy and convenience. CIS and CARB do differ in their toxicity profiles and differ in the mode of administration particularly in the time required for delivery. The hydration needed for CIS, which requires more hospital time than CARB, deters some clinicians from using it. There is variation between oncologists (and hence variation in usage by centre) as to which PLAT is preferred. The results of recent meta-analyses84,85 suggest that CIS delivers greater efficacy than CARB, and subsequently use of CIS has increased, but overall clinical practice in the UK is still split between the two PLATs. CIS and CARB have, in general, been considered interchangeable in terms of efficacy because neither is consistently superior in terms of OS. However, the efficacy of CARB and CIS may vary according to the specific type of chemotherapy drug it is combined with and the histology and disease stage of the patient. The BTOG286 aimed to establish the optimal CIS dose and whether or not CARB can be effectively substituted for the CIS at this dose; and will help to clarify the evidence regarding the relative efficacy of CIS and CARB in terms of survival, QoL and costs associated with each drug and its delivery. Publication of results is expected in 2013.
The results in this report are based entirely on the analysis of published data from Phase II and Phase III clinical trials. It is well known that patients in such trials are not necessarily representative of patients seen in UK clinical practice. The National Lung Cancer Audit has been collecting activity, performance and outcome data since 2005 and provides data on treatment rates, including chemotherapy, for every managing hospital trust in the UK, by cell type, stage, age and PS of the patients. What it has not been able to do is collect data on the specific drug regimens or the number of chemotherapy cycles being administered. New initiatives to collect data related to UK patients and the treatment they receive are now in place through the emergence of the National Cancer Intelligence Network87 and the National Cancer Data Repository that underpins it. The National Systemic Anti-Cancer Therapy (SACT) data set became operational on the 1 April 2012 and will enable much more detailed analyses of treatment and outcomes in this patient population. Thus, we will soon have access to detailed information on the precise chemotherapy (and targeted chemotherapy) regimens being used, together with data on age, cell type, stage of disease and PS, allowing for very detailed observational audits of management and outcomes at a population level. It will also be feasible to include health economic data into such future analyses. We would strongly endorse the development of initiatives of this kind in the effort to provide data that can more accurately define the true cost–benefit ratio of treatment interventions in this patient population.
A limitation of this review is that there is a very large volume of related literature in this field and so pragmatic decisions had to be taken about the inclusion criteria and the focus of the data analyses; therefore, the methods employed in this review differ slightly from the methods described in the original review protocol. We restricted the analysis to papers published from 2000 onwards and decided to include only chemotherapy drugs that are currently licensed and recommended by NICE for use in patients with NSCLC; we believe this to be the best management of the data in order to make the result of the review useful to clinicians.
Another potential limitation of this report is that the elderly population with NSCLC may be under-represented in the included trials. The majority of trials have an upper age limit, whereas in clinical practice there are substantial proportions of treated patients > 75 years of age. The majority of trials also focus on fitter populations with less comorbidity (which may include a larger proportion of elderly patients) than the average UK patient with NSCLC. In addition, we excluded single-agent regimens of DOC, GEM, PAX and VNB. Although the included chemotherapy drugs are not licensed for single-agent use, NICE7 states that DOC, GEM, PAX and VNB can be used for single-agent use if patients are intolerant of a PLAT-based doublet regimen, and this may include a larger proportion of elderly patients. Trials of single-agent regimens have focused on the elderly population, for example the Elderly Lung Cancer Vinorelbine Italian Study (ELVIS)88 demonstrated a significant survival advantage for elderly patients taking single-agent VNB compared with BSC. The Multicenter Italian Lung Cancer in the Elderly Study (MILES) trial89 showed that VNB + GEM did not improve survival compared with single-agent use of VNB and single-agent use of GEM in elderly patients with NSCLC. The elderly are less likely to have chemotherapy treatment in clinical practice in the UK, which is not explained by poorer PS or increased comorbidity. 90 Authors of a LUCADA indicate that further work is warranted to determine how far this can be explained by patient preference, appropriate physician judgement and physician prejudice. 91
Chapter 4 Assessment of cost-effectiveness
Systematic review of existing cost-effectiveness evidence
A systematic review of the economic literature was conducted to identify the existing evidence assessing the cost-effectiveness of first-line chemotherapy for patients with advanced and/or metastatic NSCLC. The criteria shown in Table 39 were used to identify the relevant studies for inclusion in the review. The search included a combination of terms (e.g. carcinoma, non-small-cell lung, economics, costs and cost analysis, effectiveness) and was limited to English-language articles. The electronic databases, including MEDLINE, EMBASE and The Cochrane Library (Issue 3, July 2010), were searched for the period from January 1980 to August 2010. All references were exported to the EndNote® version X4. Full details of the search strategies are available in Appendix 29. Two reviewers independently screened all titles and abstracts of papers identified in the search. Discrepancies were resolved by discussion with involvement of a third reviewer where necessary.
Inclusion criteria | |
---|---|
Evaluation design | Full economic evaluations that consider both costs and consequences (CEA, CUA and cost–benefit analysis) |
Patient population | Chemotherapy-naive adult patients with locally advanced or metastatic NSCLC |
Interventions | Any first-line chemotherapy treatment currently licensed:
|
Comparators | It is envisaged that the interventions will be compared with active therapy as described above |
Outcomes | Incremental cost per LYG |
Incremental cost per quality-adjusted LYG | |
Exclusion criteria | |
Other considerations | Only studies published post 2000 in full and with English-language abstracts will be included |
Trial design | CMAs are excluded from the review as there have not been any clinical equivalence trials conducted in this area and so any CMA would involve the questionable assumption of clinical equivalence |
Time frame of searching
The electronic searches for the cost-effectiveness review were originally developed for the same time frame as the clinical-effectiveness review (1980–2010); however, it was later decided to include only those trials published after the year 2000 as active chemotherapy treatments for patients with lung cancer have been evolving rapidly since this date. The clinical effectiveness and cost-effectiveness review of lung cancer treatments by Clegg et al. 39 was published in May 2001 and included economic evaluations up to and including 2000. None of the individual studies identified by Clegg et al. 39 are therefore included in this systematic review.
Identification of economic evaluations
A total of 1510 publications were identified as a result of the electronic searches. During stage 1, these studies were screened and duplicated papers were removed. In stage 2, titles and abstracts were screened and 15 papers39,92–105 were selected for potential inclusion in the review. Inclusion and exclusion criteria were applied to these 15 full papers and seven reports39,93–95,97,99,101 were included in the review. The flow diagram in Figure 24 shows the number of reports available at each stage of the inclusion process.
The lung cancer costing model discussed in the two publications by Clegg et al. 39,93 are focused primarily on chemotherapy compared with BSC. However, as they do include two chemotherapy versus chemotherapy comparisons as part of their detailed economic analysis, all data have been extracted and included in this review for information purposes only.
Relevant data were extracted from six evaluations from seven included publications39,93–95,97,99,101 into evidence tables (see Tables 42–45). All data were checked for accuracy by a second reviewer. The eight full-text reports92,96,98,100,102–105 that were excluded during the latter stages of the inclusion process are listed in Table 40 alongside reasons for exclusion. Of these eight trials, five96,102–105 were excluded as they were cost-minimisation analyses (CMAs) only. CMAs were explicitly excluded from the literature review as there are no published results from clinical equivalence trials between chemotherapy regimens for patients with NSCLC in the first-line setting to support such an analysis.
Report | Reason for exclusion |
---|---|
Lievens 200598 | One comparator was radiotherapy |
Rubio-Terrés 2002104 | CMA |
Pimentel 2006103 | CMA |
Neubauer 2010100 | Comparators were a compound of first- and second-line treatments |
Chen 200292 | CEA is only a costing exercise |
Manidiakis 2010105 | CMA |
Novello 2005102 | CMA |
Le Lay 200796 | CMA |
The quality of the reports was assessed using the 35-item list described by Drummond and Jefferson,106 the results of the quality assessment exercise are shown in Table 41. All of the reports are of good/reasonable methodological quality. They typically include the key components of a credible economic evaluation. The key methodological weaknesses include the following: a lack of detail on costs (e.g. no separation of quantity of resources consumed from unit costs); non-explicit statement of length of time horizon or discount rate used; and, in some cases, the authors did not provide disaggregated outcomes or carry out incremental analyses. The main weaknesses of the reports included in the review stem not from their quality but from their limited relevance to UK decision-making. This is a result of the comparisons considered and choice of incremental cost-effectiveness ratio (ICER) rarely being cost per QALY gained.
Checklist item | Clegg 2001/239,93 | Dooms 200694 | Klein 200995 | Lees 200297 | Maniadakis 200799 | Neymark 2005101 |
---|---|---|---|---|---|---|
The research question is stated | Y | Y | Y | Y | Y | Y |
The economic importance of the research question is stated | Y | Y | Y | Y | Y | Y |
The viewpoint(s) of the analysis are clearly stated and justified | Y | Y | Y | NC | Y | Y |
The rationale for choosing the alternative programmes or interventions compared is stated | Y | Y | Y | Y | Y | Y |
The alternatives being compared are clearly described | Y | Y | Y | Y | Y | Y |
The form of economic evaluation used is stated | Y | Y | Y | NC | Y | Y |
The choice of form of economic evaluation is justified in relation to the questions addressed | Y | Y | Y | NC | Y | Y |
The source(s) of effectiveness estimates used are stated | Y | Y | Y | Y | Y | Y |
Details of the design and results of effectiveness trial are given (if based on a single trial) | Y | Y | Y | Y | Y | Y |
Details of the method of synthesis or meta-analysis of estimates are given (if based on an overview of a number of effectiveness trials) | Y | NA | Y | Y | NA | NA |
The primary outcome measure(s) for the economic evaluation are clearly stated | Y | Y | Y | Y | Y | Y |
Methods to value health states and other benefits are stated | Y | Y | Y | NA | NA | NA |
Details of the subjects from whom valuations were obtained are given | Y | NC | Y | Y | NA | NA |
Productivity changes (if included) are reported separately | NA | NA | NA | NA | NA | NA |
The relevance of productivity changes to the study question is discussed if included | NA | Y | NA | NA | NA | NA |
Quantities of resources are reported separately from their unit costs | Y | Y | N | N | Y | Y |
Methods for the estimation of quantities and unit costs are described | Y | NC | NC | N | Y | Y |
Currency and price data are recorded | Y | Y | Y | Y | Y | Y |
Details of currency price adjustments for inflation or currency conversion are given | Y | Y | Y | Y | N | NC |
Details of any model used are given | Y | NA | NC | N | NA | NA |
The choice of model used and the key parameters on which it is based are justified | Y | NA | NC | N | NA | NA |
Time horizon of costs and benefits is stated | NA | Y | Y | NC | N | Y |
The discount rate(s) is stated | NA | NA | NA | NA | NC | NC |
The choice of rate(s) is justified | NA | NA | NA | NA | NC | NC |
An explanation is given if costs or benefits are not discounted | Y | Y | N | Y | NC | NO |
Details of statistical tests and CIs are given for stochastic data | Y | N | N | Y | Y | Y |
The approach to sensitivity analysis is given | Y | Y | Y | Y | Y | Y |
The choice of variables for sensitivity analysis is justified | Y | Y | Y | NC | NA | Y |
The ranges over which the variables are varied are stated | Y | Y | Y | NC | NA | Y |
Relevant alternatives are compared | Y | Y | Y | Y | Y | Y |
Incremental analysis is reported | Y | Y | Y | NC | Y | NO |
Major outcomes are presented in a disaggregated as well as aggregated form | Y | Y | Y | NC | NC | Y |
The answer to the study question is given | Y | Y | Y | Y | Y | Y |
Conclusions follow from the data reported | Y | Y | Y | Y | Y | Y |
Conclusions are accompanied by the appropriate caveats | Y | Y | Y | Y | N | Y |
Study characteristics and model overview
Three97,99,101 of the seven included studies are CEAs. Two papers39,93 are based on the use of three different economic models: a pair-wise comparison between the regimens or BSC (model 1), a CMA (model 2) and a CEA with BSC as the comparator (model 3); only model 1 included a chemotherapy compared with chemotherapy comparison. The study by Klein et al. 95 presents results from both a CMA and a cost–utility analysis (CUA) and the study by Dooms et al. 94 is a CUA.
Klein et al. 95 uses a Markov framework with an initial simple decision tree covering a 6-month period followed by three 6-month cycles. The other studies use simple decision trees with time horizons of ≤ 1 year in four trials,39,93,94,97 3 years101 and 40 months. 99 Most of the economic evaluations have short time frames; this is because they are based on clinical trials with mean OS estimates of approximately 10 months. All of the reports, but one,94 have been conducted using a third-party payer perspective taking account of direct costs only. Dooms et al. 94 adopts a societal perspective using direct costs and costs related to travel expenses. Three39,93,97 are UK based, one is set in Belgium,94 one in Greece,99 one in the Netherlands101 and one in the USA. 95
Four reports39,93,94,99 were funded from public grants or from university funds, two95,97 were funded by a pharmaceutical company and one101 was funded jointly by a pharmaceutical company and several hospitals.
The comparators used in each of the studies and detailed information about design and trial characteristics are presented in Table 42.
Study | Source | Type of study | Interventions | Study population | Country | Duration of study | Industry/author affiliation |
---|---|---|---|---|---|---|---|
Clegg 2001/239,93 | Full text | Model 1: Pair-wise comparisons from published trials Model 2: CMA Model 3: CEA (vs BSC) |
PAX, DOC, GEM, VNB, BSC | Patients with NSCLC | UK | < 1 year | This study was supported by the NHS R&D HTA programme |
Dooms 200694 | Full text | CUA | GEM, CIS + VIN | Patients with symptomatic advanced NSCLC | Belgium | < 1 year | The funding source is not stated. Authors affiliation: Leuven University |
Klein 200995 | Full text | CEA/CUA | CIS + PEM, CIS + GEM, CARB + PAX, CARB + PAX + BEV | Patients with symptomatic advanced NSCLC | USA | 2 years | Authors were contracted by Eli Lilly and Company and to conduct the research or work in Eli Lilly and Company |
Lees 200297 | Full text | CEA | (1) GEM + BSC vs BSC (2) GEM + CIS vs ETOP + CIS vs MIC + CIS vs MVP (3a) GEM + CIS vs taxane/PLAT regimens (3b) GEM + CIS vs VNB |
Patients with NSCLC | UK | 1 year | This study was carried out with a research grant from Eli Lilly and Company |
Maniadakis 200799 | Full text | CEA | DOC + GEM DOC |
Patients with advanced/metastatic NSCLC | Greece | 40 months | Public health-care members. No conflict of interest or funding from industry declared |
Neymark 2005101 | Full text | CEA | CIS + PAX CIS + GEM GEM + PAX |
Patients with advanced/metastatic NSCLC | Netherlands | 3 years | Authors are staff from several Medical centers around the Netherlands. The study was partially supported by Bristol-Myers Squibb and Eli Lilly and Company (provision of drugs used in the RCT) |
Model inputs and data sources
Costs were typically divided into the following categories: costs of drug administration, side effects costs, acquisition costs of drugs, costs of BSC, costs of tests/investigations; and the costs of travel expenses were considered in the only study94 using a societal perspective. The sources included public costs databases, hospital costing data and Medicare reimbursement rates. In general, costs were extracted from publicly available documents, which adds transparency to the costing approaches described in the studies. The economic models, cost item and the sources used are summarised in Tables 43 and 44.
Study | Type of model | Perspective | Model assumptions | |
---|---|---|---|---|
Outcomes | Costs and resource use | |||
Clegg 2001/239,93 | Three simple decision trees | Third-payer perspective | No RCTs directly compared two or more regimens in terms of QoL; thus, side-by-side comparisons cannot be made Whether or not one cycle of a regimen is equivalent to one cycle of another remains unclear and it is addressed in the SA PAX doses varied markedly between trials and so several regimens were considered Data on effectiveness were pooled using mixed-treatment comparison |
Costs of antiemetic and diuretics were negligible and were excluded from the analysis Cost of administration of drug in case of AEs were £500 (expert advice) Costs from published and unpublished data |
Dooms 200694 | Simple decision tree | Societal perspective | No differences in survival were observed between the two regimens but differences in QoL and clinical benefit were observed. Based on this finding, authors decided to carry out CUA using QALYs QALYs have been estimated using a VAS score from the LCSS QoL instrument. This VAS score was considered to be a reasonable alternative to the VAS thermometer score in the EQ-5D instrument |
Direct non-medical costs = travel expenses Indirect costs are not taken into account because they are assumed to be equal in both groups Second-line chemotherapy and radiotherapy did not show any significant difference between groups and were not taken into account Costs of diagnostic procedures and the expenditure at the end of the treatment period were unavailable and no real differences were assumed between the two arms |
Klein 200995 | Semi-Markov model with three 6-month cycles | US payer | Health states of partial and complete response were not reached until the beginning of the third and fourth cycle, respectively Side effects probabilities assumed to be independent of the health state and response, and progression probabilities assumed to be independent of side effects Dose reductions/delays between chemotherapy cycles were not modelled State transition probabilities beyond the first year were assumed to be the same for all initial treatments |
As long as the overall proportions of AEs are correct and AE costs are independent of treatment or disease costs, not altering AE probabilities by health state should have no effect on cost results Costs and utilities for the health states without chemotherapy were assumed to be equivalent regardless of the first-line treatment used |
Lees 200297 | Simple decision tree | UK NHS | All outcomes are taken from head-to-head clinical trials | Only direct NHS health costs were included Wastage was incorporated into the analyses |
Maniadakis 200799 | Simple decision tree | Greek NHS | Data on several clinical outcomes were collected; only mean survival has been used in the CEA | Only (Greek) NHS direct costs are included Cost of chemotherapy in each cycle is calculated by multiplying the exact dose given by cost per mg. No drug wastage |
Neymark 2005101 | Simple decision tree | Third-party payer | Antiemetic agent doses selected by examining the literature | Costs of drugs are based on the amount reimbursed by the health insurance firm |
Study | Cost items and cost data sources | Currency and currency year | Discount rate |
---|---|---|---|
Clegg 2001/239,93 | Administration of drugs and side effect costs (Scottish Health Purchasing Information Centre; Scottish Health Service Cost's ‘blue book’; Ninewells Hospital) Chemotherapy counselling (Scottish Health Purchasing Information Centre; Scottish Health Service Cost's ‘blue book’; Southampton General Hospital) Costs of drugs (BNF) BSC costs (case notes of patients published elsewhere and checked with the data from South-East Lung Trial) |
£/1999–2000 | NA |
Dooms 200694 | Costs of chemotherapy drugs, concomitant medications, outpatient admissions and inpatient hospitalisation during chemotherapy (Leuven University Hospital Pharmacy) Direct non-medical costs related to the travel expenses Resource-cost data were calculated from the actual data collected during the prospective clinical trial |
€/2000 | NA |
Klein 200995 | NSCLC-related direct health-care costs including chemotherapy, costs of pre-medication, administration of chemotherapy, laboratory monitoring, treating common AEs, subsequent therapies, direct care for disease-related morbidity, and end-of-life care (Medicare reimbursement rates; Pharmetrics paid claims database107) | US$/NS | NA |
Lees 200297 | Costs associated with chemotherapy acquisition and use of concomitant medications (BNF 40108) Hospitalisation (2000 UK National Schedule of Reference Costs109) Infusion and visits to health professionals (UK-based source of unit costs in health care; University of Kent at Canterbury110) Radiotherapy (previous economic evaluation Bagust 1999111) |
£/2002 | NA |
Maniadakis 200799 | Treatment costs: initial chemotherapy, concomitant medications, radiotherapy, diagnostic and laboratory testing, any hospitalisation and resources necessary for the treatment of AEs, follow-up visits and second-line chemotherapy (Greek national sources and database from the University General Hospital Heraklion) | €/2005 | NS |
Neymark 2005101 | Hospital overnight stay (daily allowance); day clinic stay (daily allowance); outpatient specialist consultations; transfusions (red blood cells); transfusions (platelets); pre-medication before PAX; antiemetics/CIS regimens (per cycle); antiemetics/non-CIS regimens (per cycle); trial treatments (radiotherapy, second line); chemotherapy (second line); surgery (second line) All cost data collected from: College Tarieven Gezondheidszorg (CTG);112 Tarieven voor Medisch Specialisten; Tarieflijst Instellingen for Hospitals; Farmacotherapeutisch Kompas113 |
€/2002 | NS |
The most commonly used efficacy outcome was survival time with median survival time (MST) used in four39,93,97,101 and OS used in three. 94,97,99 Response rates or ORRs were also used. All efficacy data used in the included studies are shown in Table 45. Sources of efficacy data are varied; a single clinical trial was used in five94,95,97,99,101 of the seven reports and the remaining two39,93 took data from a collection of trials using a mixed-treatment comparison to summarise the data.
Study | Efficacy data | Efficacy data sources | Health outcomes | Health outcome data sources | Discount rate |
---|---|---|---|---|---|
Clegg 2001/239,93 | Median number of cycles MST |
Collection of 33 trials from the systematic review conducted by the authors | LYS | Collection of 33 trials found in a systematic review conducted by the authors | NA |
Dooms 200694 | ORR OS Clinical benefit |
Data were collected from a prospective Phase III RCT (Vansteenkiste 2001114) | QALYs LYS |
Patient responses to one item in the LCSS QoL instrument (from patients in RCT114) were converted to a single utility value | NA |
Klein 200995 | TRR Progression rate |
Data were obtained from a head-to-head trial (Scagliotti 200861) and from a mixed-treatment comparison presented by Vansteenkiste et al. in 2008115 in Dresden (Proceedings of the Internal Thoracic Oncologic Congress) | LYG QALYs |
Utility values were calculated using an algorithm by Nafees 2008116 | NA |
Lees 200297 | (1) PFS, OTR and TTR (2) OS, PFS, MST and OTR (3a) OS, RR and TTP (3b) MST and RR |
(1) Anderson 2000117 (2) Cardenal 1999,118 Crino 1999119 and results presented in the World Lung Conference Tokyo 2000 (3a) Schiller 200047 (3b) Comella 2000120 |
(1) PFS, TTR and OTR (2) OS, PFS, MST and OTR (3a) OS, RR and TTP (3b) MST and RR |
(1) Anderson 2000117 (2) Cardenal 1999,118 Crino 1999119 and results presented in the World Lung Conference Tokyo 2000 (3a) Schiller 200047 (3b) Comella 2000120 |
NA |
Maniadakis 200799 | TTP Number of deaths Response rates and duration OS |
Multicenter Phase III RCT conducted by the HORG | LYS | Multicenter Phase III RCT conducted by the HORG | NS |
Neymark 2005101 | MST | Data were collected prospectively as an integrated part of the clinical trial presented in the paper | None is stated apart from the use of bootstrapping techniques to get CI and ICER | NS | NS |
Five studies used data from trials39,93–95,99 and used life-year saved (LYS) or LYG as the primary health outcome, whereas two used QALYs. 4,116 Different approaches for calculating QALYs were used in each of the latter of these studies. Klein et al. 95 adopted a CUA approach and used utility values from the published study by Naffes et al. 116 to calculate the QALYs gained in each regimen. Dooms et al. 94 used one item from the LCSS QoL instrument and transformed this into a corresponding utility value; these utilities were then combined with the survival data from a RCT in order to obtain QALYs. Several studies expressed their incremental ratios in terms of cost per progression-free life-year, cost per tumour response or costs to improve mean survival.
Results and sensitivity analysis
Tables 46 and 47 show the cost-effectiveness results, sensitivity analyses and conclusions of the reports. The results of the economic evaluation by Clegg et al. 39,93 reveal that any chemotherapy regimen is cost-effective (vs BSC) at a threshold of £30,000 per LYS except PAX. In the chemotherapy versus chemotherapy comparisons, GEM + CIS dominates GEM and VNB + CIS is cost-effective when compared with VNB. The conclusion of the authors is that depending on the assumptions used, the new drugs range from being cost-effective, as conventionally accepted, to being cost saving. The results of the sensitivity analyses only slightly change the results from the base-case analysis.
Study | Total costs | Total outcomes | ICERs | Conclusion |
---|---|---|---|---|
Clegg 2001/239,93 | Average cost per patient model 1: BSC vs GEM: £3342 vs £4132 BSC vs VNB: £3342 vs £2812 GEM + CIS vs VNB + CIS: £3943 vs £4420 BSC vs PAX: £3342 vs £8293 BSC vs DOC: £3342 vs £5040 VNB vs VNB + CIS: £3675 vs £4448 BSC vs DOC (75) vs DOC (100): £3342 vs £4365 vs £5040 DOC (75) vs DOC (100): £4365 vs £5040 Average cost per patient models 2 and 3: BSC £3342; GEM £4132; GEM + CIS £6321; VNB £3675; VNB + CIS £4736; PAX £8293; PAX (135) + CIS £6304; PAX (175) + CIS £7550; PAX (250) + CIS £8147; DOC £5040; DOC (2L) £4365 |
MST (months) and LYS model 1: BSC vs GEM (MST; LYS): 5.9; 0.49 vs 5.7; 0.48 BSC vs VNB (MST; LYS): 4.8; 0.40 vs 6.5; 0.54 GEM + CIS vs VNB + CIS (MST; LYS): 8.1; 0.68 vs 6.7; 0.56 BSC vs PAX (MST; LYS): 4.8; 0.40 vs 6.8; 0.57 BSC vs DOC (MST; LYS): 5.7; 0.48 vs 6.0; 0.50 VNB vs VNB + CIS (MST; LYS): 7.2; 0.60 vs 9.2; 0.77 BSC vs DOC (75) vs DOC (100) (MST; LYS): 4.6; 0.38 vs 7.5; 0.63 vs 5.9; 0.49 DOC (75) vs DOC (100) (MST; LYS): 5.7; 0.48 vs 5.5; 0.46 Model 2: Assumes equal efficacy Median survival model 3: BSC 5.24; GEM 6.9; GEM + CIS 8.80; VNB 7.06; VNB + CIS8.45; PAX 6.51; PAX (135) + CIS 9.40; PAX (175) + CIS 8.81; PAX (250) + CIS 10.00; DOC 6.00; DOC (2L) 5.94 LYS model 3: BSC 0.44; GEM 0.58; GEM + CID 0.73; VNB 0.59; VNB + CID 0.70; PAX 0.54; PAX (135) + CIS 0.78; PAX (175) + CIS 0.73; PAX (250) + CIS 0.83; DOC 0.50; DOC (2L) 0.49 |
Incremental cost per LYS model 1: BSC vs GEM: GEM dominated BSC vs VNB: BSC dominated GEM + CIS vs VNB + CIS: VNB ± CIS dominated BSC vs PAX: £29,704 (PAX) BSC vs DOC: £67,926 (DOC) VNB vs VNB + CIS: £4638 (VNB ± CIS) BSC vs DOC (75) vs DOC (100): £4234 (DOC 75); DOC (100) dominated DOC (75) vs DOC (100): DOC (100) dominated Incremental cost per LYS model 3: (vs BSC): GEM £5690; GEM + CIS £10,041; VNB £4091; VNB + CIS £5206; PAX £46,610; PAX (135) + CIS £8537; PAX (175) + CIS £14,124; PAX (250) + CIS £12,104; DOC £26,707; DOC (2L) £17,546 |
The new drugs for NSCLC extend life by only a few months compared with BSC, but appear to do so without net loss in QoL and at a cost per LYG that is much lower than for many other NHS activities. Depending on assumptions used these new drugs range from being cost-effective, as conventionally accepted, to being cost saving |
Dooms 200694 | CIS + VIN: €4502 per patient GEM: €6024 per patient |
LYS: CIS + VIN: 0.53; GEM: 0.68 QALYs: CIS + VIN: 0.18; GEM: 0.29 |
ICER: €13,836/QALY |
Although the least expensive strategy is CIS + VIN, the greater clinical benefit of GEM balances its higher cost and generates an acceptable incremental cost–utility ratio |
Klein 200995 | CIS + GEM: $61,535 CARB + PAX: $50,283 CIS + PEM: $66,606 CARB + PAX + BEV: $90,004 |
LYG: CIS + GEM: 0.9102 CARB + PAX: 0.8882 CIS + PEM: 0.9587 CARB + PAX + BEV: 1.0379 QALYs: CIS + GEM: 0.4661 CARB + PAX: 0.4469 CIS + PEM: 0.4943 CARB + PAX + BEV: 0.5260 |
Incremental (ONLY NS) CARB + PAX + BEV to CIS + PEM: $337,179/LYG; $1,006,065/QALY Incremental CIS + PEM to CIS+GEM: $104,577/LYG; $179,597/QALY Incremental CIS + PEM to CARB + PAX: $231,291/LYG; $343,870/QALY |
Compared with commonly used and reimbursed regimens for first-line chemotherapy in advanced NSCLC, PEM + CIS may be considered cost-effective, particularly in patients with NS cell histology. This analysis emphasises the importance of histology in identifying the appropriate patients for PEM + CIS first-line chemotherapy |
Lees 200297 | (1) GEM + BSC: £5502; BSC: £3861 (2) GC: £4142–5084; EP: £3762; MIV: £4481; MVP: £4005 (3a) GEM + CIS: £5537; PAX + CIS: £9043; PAX + CARB: £8444; DOC + CIS: £5779 (3b) GEM + CIS: £4477; VNB + CIS: £5048 |
(1) TTR (days) GEM + BSC: 288; BSC: 173. PFS (years) GEM + BSC: 0.789; BSC: 0.474. OTR (%): GEM + BSC: 18.5; BSC: 0.0 (2) EP: PFS (years): 0.358. OTR (%): 21.9. MIC: OTR (%): 27.6. MVP: OS (%): 17. OTR (%): 36.7. GEM + CIS: PFS (years): 0.575. OTR (%): 39.6–54. OS (%): 36 (3a) RR (%): GEM + CIS: 21; PAX + CIS: 21.3; DOC + CIS: 17.3; PAX + CARB: 15.3. OS (years): GEM + CIS: 0.375; PAX + CIS: 2.92; DOC + CIS: 0.275; PAX + CARB: 0.3 (3b) MST (weeks): GEM + CIS: 42; VNB + CIS: 35. RR (%): GEM + CIS: 30; VNB + CIS: 25 |
(1) GEM + BSC vs BSC: Cost per progression-free life-year: £5228. Cost per tumour response: £8873 (2) GEM + CIS vs EP: Cost per progression-free life-year: £1751. Cost per tumour response: £2032. GEM + CIS vs MIC: Cost per tumour response: £5169. GEM + CIS vs MVP: Cost per % gain in 1-year survival: £5681. Cost per tumour response: £6240 (3a) Incremental costs only: GEM + CIS vs PAX + CIS: –£3506; GEM + CIS vs PAX + CARB: –£2907; GEM + CIS vs DOC + CIS: –£242 (3b) GEM + CIS vs VNB + CIS: –£571 |
GEM alone or in combination with CIS was assessed to be a cost-effective or cost-saving therapy when compared with BSC, standard and novel chemotherapy. Chemotherapy regimens containing GEM therefore represent good value for money and an efficient use of health-care resources in the treatment of advanced NSCLC |
Maniadakis 200799 | DOC: €5739 (95% CI €5037 to €6519)) DOC + GEM: €7255 (95% CI €6565 to €7970 |
DOC: 7.25 months (95% CI 6.29 to 8.23) DOC + GEM: 9.19 months (95% CI 8.98 to 10.51) |
Incremental cost per LYS of DOC + GEM vs DOC: €9538 | The data support that DOC + GEM represents a cost-effective treatment option in relation to DOC monotherapy for patients with NSCLC in the Greek setting |
Neymark 2005101 | CIS + PAX: €16,662 (95% CI €15,251 to €18,072) CIS + GEM: €13,944 (95% CI €12,829 to €15,060) GEM + PAX: €17,377 (95% CI €16,088 to €18,667) |
Mean MST in years: CIS + PAX: 0.94 (95% CI 0.82 to 1.07) CIS + GEM: 0.98 (95% CI 0.86 to 1.11) GEM + PAX: 0.80 (95% CI 0.69 to 0.92) |
CIS + GEM vs CIS + PAX: 72% of the replicates indicate CIS + GEM improves outcomes and reduces costs GEM + PAX vs CIS + PAX: 82% of the replicates indicate GEM + PAX reduces MST while increases the costs |
The two CIS-based regimens are equivalent in terms of survival, but CIS + GEM may reduce costs by approximately €2000 per patient compared with PAX + CIS. GEM + PAX is a dominated option with higher costs and a reduction in MST compared with PAX + CIS |
Trial | Sensitivity analysis |
---|---|
Clegg 2001/239,93 | One-way sensitivity analysis was carried out across a range of variables including number of cycles (advice from clinical colleagues was that in routine care a more realistic scenario would be to assume 60% of patients would have only 1–2 cycles, while 40% would continue towards the recommended number of cycles: three for GEM, VNB and DOC regimens and four for PAX); number of administrations per cycle of VNB; best and worst cycles from trials; effect of discounts on BNF prices; and cost of newer antiemetic regimens. Mean survival estimates calculated from single trials by Berthelot 2000121 and non-patient-based utility estimates were also examined. The cost of BSC, particularly the number of inpatient days (21 vs 19 days), was varied to reflect slight differences between sources. VNB, VNB + CIS and GEM retain their cost-effectiveness under a range of assumptions and may even be dominant under certain circumstances |
Dooms 200694 | Extensive univariate sensitivity analysis has been performed using different cost ranges (from −50% to +50%) and cost items. Reducing the QALY gain increases the size of the ICER. The ICER is > €50,000 only when costs were increased by 50% and a lower QALY value (0.04) is used |
Changing the cost of drug administration has no real impact on the ICER, whereas varying the cost of the drug has the most significant impact | |
Klein 200995 | Several univariate sensitivity analyses were conducted on: number of PEM vials; non-squamous vs all NSCLC; responders receiving fifth and sixth chemotherapy cycle; unequal AE costs, equal mild side effects and discounting |
The tornado diagram described in the text shows that most reasonable changes in costs changed the ICER for PEM + CIS vs GEM + CIS by < 10% | |
Lees 200297 | (1) Univariate sensitivity analyses were employed varying: costs of GEM acquisition and administration, outcomes measures using confidence limits and unit costs of chemotherapy administration. No significant changes in the ICERs were noted |
(2) Several univariate sensitivity analyses were performed using all non-chemotherapy costs (upper and lower bounds) resulting in no significant changes to the size of the ICER | |
(3a/b) Costs/doses of all drugs in the group of novel therapies were varied; none of which changed the results significantly | |
Maniadakis 200799 | A PSA was carried out and shows that the probability of DOC + GEM being cost-effective in relation to DOC monotherapy is 91% at a threshold of €20,000, 97% at €35,000 and 98% at €50,000 |
Neymark 2005101 | A univariate sensitivity analysis was conducted by varying hospital costs. The incremental costs between strategies did not vary |
Dooms et al. 94 estimate an ICER of €13,836 per QALY in favour of GEM when compared with VNB + CIS; in the sensitivity analysis, the results are robust to credible changes in both costs and utilities.
Klein et al. 95 show that, as there are only slight differences in the total QALYs gained from each of the different regimens, the estimated ICERs exceed $100,000 per QALY gained when PEM + CIS is compared with (1) GEM + CIS and (2) PAX + CARB. For the non-squamous population only, ICERs exceed $150,000 per QALY gained. Reasonable changes introduced by undertaking sensitivity analyses do not change the base-case results by > 10%.
Lees et al. 97 who do not use LYS or QALYs as a measure of health outcome, conclude that GEM alone or in combination with CIS is a cost-saving therapy when compared with BSC. The authors state that GEM + CIS is cost saving when compared with novel chemotherapies (PAX + CIS, PAX + CARB, DOC + CIS and VNB + CIS). No significant changes to the results were identified via sensitivity analysis.
Maniadakis et al. 99 found DOC + GEM to be a cost-effective regimen when compared with DOC alone at a threshold of €9538 per LYS with a 91% and 98% probability of being cost-effective when the threshold is set at €20,000 and €50,000, respectively.
Neymark et al. 101 did not find any differences in survival between patients receiving CIS + GEM and CIS + PAX, but concluded that the former may reduce costs by approximately €2000 per patient and stated that CIS + PAX is a dominant option when compared with GEM + PAX. The sensitivity analysis carried out on the base-case scenario did not lead to a change in the cost-effectiveness results.
Critique of published literature
This section provides a summary and a more detailed critique of the economic and clinical evidence used in the economic evaluation papers included in the review. The aim of the commentary set out in this section is to supplement the quality assessment exercise undertaken as part of the systematic review.
Clegg et al. 200139 and Clegg et al. 200293
Methods of deriving the effectiveness data
In model 1, only two of the comparisons reviewed by Clegg et al. 39,93 compared chemotherapy with chemotherapy: GEM + CIS compared with GEM and VNB + CIS compared with VNB. In model 1, the pair-wise comparisons were based on the results of single trials only. In model 2, the authors make the assumption that the regimens have equal efficacy. In model 3, all relevant and available clinical effectiveness data are pooled using a mixed-treatment comparison approach as, at the time of writing, there was a lack of head-to-head evidence in this area.
Measurement and valuation of resource data
The main effectiveness measures used LYS and median number of chemotherapy cycles. Number of deaths or death rates at certain time points were not reported.
Measurement and valuation of health benefits (utilities)
Utilities have not been used, LYS are used as the main measure of health outcome.
Method of synthesising the costs and effects
Three different economic models were described in detail. Only the model incorporating pair-wise comparisons is able to comment on chemotherapy compared with chemotherapy and reports an ICER (cost per LYS); however, a limitation of this model is that each cost-effectiveness estimate was based on data from a single trial.
Analysis of uncertainty
Only univariate sensitivity analysis has been used to test the uncertainty related to use of data from different publications. Probabilistic sensitivity analysis (PSA) was not carried out.
Generalisability of the results
The authors have attempted to make the results of their economic models as generalisable to a UK population as possible. However, the authors conclude that comparisons among the chemotherapy drugs using the results of the CEA in model 3 should be viewed with caution because of the way the data were combined.
Dooms et al. 200694
Methods of deriving the effectiveness data
Data from a Phase III RCT comparing GEM with VNB + CIS were used. Reliance on a single trial as a source of clinical effectiveness data may be seen as a limitation of the economic evaluation. The main clinical effectiveness measure used was OS. The author states that as small differences in OS were identified between regimens and bigger differences in QoL and clinical benefit were also identified, a CUA was performed using QALYs.
Measurement and valuation of resource data
Resource-cost data were calculated from a RCT. The RCT used for this economic evaluation is not fully reported in the economic paper, but is fully referenced. Some cost items were considered to be equivalent across the two interventions and were not included in the economic evaluation.
Measurement and valuation of health benefits (utilities)
Quality-adjusted life-years and LYS were used in the economic evaluation. The method used to convert a global visual analogue score into a utility score is not fully described. The authors acknowledge that they could be criticised for this approach.
Method of synthesising the costs and effects
A CUA has been used to synthesise both cost and health outcomes in the form of an ICER.
Analysis of uncertainty
The authors have explored the effect of varying costs and utilities on the size of the incremental cost–utility ratio. PSA was not carried out.
Generalisability of the results
The setting for the economic evaluation was Belgium which means that the results are unlikely to be generalisable to a UK setting without an additional description of Belgian clinical practice and estimation of costs. As GEM monotherapy and VNB + CIS combination therapy are not routinely used as standard chemotherapy regimens in the UK, it is unlikely that the results will help health professionals make decisions that are relevant to a UK population.
Klein et al. 200995
Methods of deriving the effectiveness data
Efficacy data were obtained from a head-to-head trial comparing PEM + CIS and GEM + CIS regimens; for comparisons between PEM + CIS and PAX + CARB and with PAX + CARB + bevacizumab (BEV) regimens, data were derived from the results of a mixed-treatment comparison exercise. The calculation of the transition probabilities in the semi-Markov model is not fully explained.
Measurement and valuation of resource data
Costs are taken from the Medicare reimbursement rates.
Measurement and valuation of health benefits (utilities)
Differential survival and response rates for PAX + CARB and PAX + CARB + BEV were taken from a mixed-treatment comparison model; very little data on this model were provided and the reference cited was from a conference abstract. QALYs were calculated using the utility values estimated by Nafees et al. 116 which take account of toxicities and response rates. However, there is insufficient information in the paper to explain how these utility values were derived which means it is not possible to assess the robustness of these calculations. As the values are central to the author's conclusions, the inability to assess the calculations limits the usefulness of their findings.
Method of synthesising the costs and effects
The author has used a semi-Markov model with an initial simple decision tree covering a 6-month period followed by three 6-month cycles. The base-case ICER was estimated for patients with non-squamous NSCLC only, a second analysis was presented which estimates ICERs for all patients.
Analysis of uncertainty
Several univariate sensitivity analyses have been performed by the authors and were presented as a tornado diagram showing that changes in costs do not lead to variations in the cost-effectiveness results by > 10%. A PSA has not been employed.
Generalisability of the results
The setting of this economic evaluation is the US Medicare system which differs to the NHS not only in the finance and provision of chemotherapy regimens but also in the costs of administration. Two of the treatment options considered in the economic evaluation are of interest to UK decision-makers (PEM + CIS and GEM + CIS) and it is particularly useful that the authors provide ICERs for the population with non-squamous disease as well as the overall population (but these are based on QALYs, where we are unsure how the authors have incorporated utility).
Lees et al. 200297
Methods of deriving the effectiveness data
The authors used several head-to-head trials to inform the clinical base of the economic model. However, the authors did not state reasons for selecting these particular trials. Outcome measures used in the selected trials included OS, PFS and response rates. Comparison of GEM + CIS with novel chemotherapy was based on clinical data derived from two large trials (one of which was an interim analysis).
Measurement and valuation of resource data
Quantities of health-care resource use have been derived from the RCTs described. Costs were derived from NHS reference costs.
Measurement and valuation of health benefits (utilities)
No utilities have been used in this economic evaluation.
Method of synthesising the costs and effects
The author has used ICERs in terms of cost per progression-free life-year or incremental costs only; the ICER, therefore, does not reflect QoL lost related to the toxicity of treatment.
Analysis of uncertainty
Several univariate sensitivity analyses were undertaken using upper and lower bounds of the cost parameters. No sensitivity or scenario analysis was undertaken on efficacy parameters.
Generalisability of the results
The economic evaluations use UK costs and as the RCTs are multicentre trials, they appear to make the economic results generalisable to the UK setting. The comparison of GEM + CIS with novel chemotherapy is the most interesting to UK decision-makers. However, close scrutiny of the assumptions used in the base-case scenarios is merited; for example, PAX is given as a 24-hour i.v. therapy which is rarely the case in the UK and cost of median compared with mean number of treatment cycles influences the size of the ICER.
Maniadakis et al. 200799
Methods of deriving the effectiveness data
This economic evaluation is conducted alongside a multicentre Phase III RCT in Greece. TTP, OS, RRs and number of deaths were collected from the trial but the author stated that only median OS was to be used in the economic evaluation.
Measurement and valuation of resource data
Resource-use data were collected from the key RCT. The economic evaluation assumes no drug wastage. A detailed description of unit cost data is presented in the paper; data were taken from Greek national sources and the database of the University General Hospital of Heraklion. For example, the cost of chemotherapy was calculated by multiplying the exact dose given with cost per mg.
Measurement and valuation of health benefits (utilities)
No utilities have been used in the economic evaluation, only LYS have been estimated.
Method of synthesising the costs and effects
Incremental cost per LYS was used as the cost-effectiveness ratio of interest. No toxicity or QoL results were incorporated into the economic evaluation.
Analysis of uncertainty
A PSA was performed to quantify data uncertainty and demonstrated that the DOC + GEM combination was very likely to be cost-effective compared with DOC monotherapy.
Generalisability of the results
The cost and benefit data used in the study was specific to the Greek NHS. As GEM and DOC are now off patent and DOC + GEM is not used as a standard chemotherapy regimen in the UK, it is unlikely that the results will help health professionals make decisions that are relevant to a UK population.
Neymark et al. 2005101
Methods of deriving the effectiveness data
Efficacy data were derived from a prospective RCT; the economic evaluation was conducted alongside the RCT. Only data on survival were used in the economic evaluation.
Measurement and valuation of resource data
Prospective collection of data on the use of medical resources was integrated in the case report forms of the trial. Where data were not sufficiently precise to allow measurement, assumptions were made using set protocols and published literature. Unit prices of resources used in the trial are detailed in the paper; resource utilisation, mean quantities and proportions of patients are also described.
Measurement and valuation of health benefits (utilities)
No utilities have been used. The objective of the economic evaluation was to estimate an average cost per patient related to survival. There is no discussion of toxicity or QoL in the paper.
Method of synthesising the costs and effects
Differences between the mean cost per patient in each regimen were calculated using bootstrapping techniques with 5000 iterations. No ICERs were presented.
Analysis of uncertainty
The limited sensitivity analysis conducted by the authors was focused on the impact of varying hospital costs on total costs.
Generalisability of the results
Study results are interpretable to decision-makers in a hospital setting in the Netherlands. However, owing to the lack of health outcome measurements and failure to report ICERs, these results are of limited validity to decision-makers in the UK NHS.
Are the results of the reports included in the systematic review relevant to UK decision-makers?
As shown in Table 48, the results of the seven papers39,93–95,97,99,101 considered in the systematic review of first-line chemotherapy for patients with NSCLC are unlikely to aid decision-makers in the UK. First, the comparisons that have been the focus of the papers are not all standard NHS treatments and, second, only two of the reports present their findings in terms of cost per QALY gained.
Studies described in the systematic review | UK setting? | Relevant comparisons? | ICER (cost per QALY) estimated? | Relevance to NHS decision-making? |
---|---|---|---|---|
Clegg 2001/239,93 | Yes | None | No | Limited |
Dooms 200694 | No | None | Yes | Poor |
Klein 200995 | No | PEM + CIS vs:
|
Yes | Limited |
Lees 200297 | Yes | GEM + CIS vs:
|
No | Limited |
Maniadakis 200799 | No | None | No | Poor |
Neymark 2005101 | No | PAC + CIS vs GEM + CIS | No | Limited |
Other sources of economic evidence
In order to inform the debate and make use of relevant clinical effectiveness and cost-effectiveness data we have summarised the findings from two recent Evidence Review Group (ERG) reports122,123 on first-line treatments for patients with NSCLC. The ERG reports inform the NICE STA process and are written prior to the first Appraisal Committee (AC) meeting. Neither of these reports were identified by the literature searches, as they are not published in a peer-reviewed journal. The ERG reports122,123 are focused on two subgroups of patients with NSCLC: (1) patients with non-squamous disease and (2) patients who are EGFR+.
Patients with non-squamous disease
The manufacturer's submission (MS) for this STA122 included a de novo economic evaluation comparing PEM + CIS with GEM + CIS in patients with non-squamous disease using clinical effectiveness data from the trial by Scagliotti et al. 61
Patients who are epidermal growth factor receptor mutation positive
The MS for the second STA123 included a de novo economic evaluation comparing GEF with PAX + CARB in patients who are EGFR+ using clinical effectiveness data from the IPASS. 15
Adherence to the National Institute for Health and Care Excellence reference case and critical appraisal of economic evaluations
Tables 49 and 50 provide the ERG summary/critique of the de novo economic evaluations performed by the manufacturers and show whether or not the approach adopted by the manufacturer adheres to the reference case outlined by NICE.
Item | ERG critique (PEM)122 | ERG critique (GEF)124 |
---|---|---|
Was a well-defined question posed in answerable form? | The manufacturer did not fully address the decision problem (VNB and PAX not included) | The manufacturer only partially answered the decision problem set by NICE as (1) DOC and (2) PEM were not included |
Comprehensive description of competing alternatives? | The manufacturer described the chosen comparators adequately | The manufacturer described the chosen comparators adequately |
Was the effectiveness of the programme or services established? | Evidence from the JMDB trial demonstrated the clinical non-inferiority of PEM + CIS compared with GEM + CIS. The trial was not powered to detect subgroup analyses, which the manufacturer relies on heavily in the model. Also, for the comparisons with DOC + CIS and GEM + CARB, the manufacturer conducted indirect analysis; however, the methodology employed to achieve this was flawed | It is unclear to what extent treatment effectiveness is established for a UK population primarily because patients in the IPASS are younger, predominantly female, oriental, have adenocarcinoma histology and include patients whose PS = 2; these patients do not represent patients eligible for treatment with GEF in England and Wales. The ERG has also expressed its concern regarding the methods used in the meta-analysis and in the mixed-treatment comparison which supply the main sources of clinical effectiveness evidence; in particular, the ERG questions the validity of assuming differential efficacy rates for the four doublet chemotherapy regimens considered in the economic evaluation |
All important/relevant costs/consequences identified? | Key costs and consequences were identified | The key costs and outcomes were identified. ERG proposed not to include g-CSF costs as this is not used in clinical practice in NHS |
Were costs/consequences measured accurately in appropriate physical units? | For example, the BSA value used to calculate chemotherapy costs does not represent NSCLC patients in the UK | The BSA value used to calculate chemotherapy costs does not represent patients with NSCLC in the UK; cost per cycle of chemotherapy and second-line chemotherapy were estimated incorrectly |
Costs/consequences valued credibly? | Modelled OS and PFS were inaccurate and overestimated for some trial values | OS was not adequately modelled; poor correspondence between parametric survival models and source data |
Were costs/consequences adjusted for differential timing? | The method of discounting was appropriate | Costs and outcomes were discounted after 1 year; method of discounting did not conform to UK convention of discounting annually after year 1 |
Was an incremental analysis of costs and consequences of alternatives performed? | ICERs (cost per QALY gained and cost per LYG) were presented for the base-case population and subgroups | Pair-wise incremental results presented for the base-case target population and subgroups (adeno vs non-adeno; females vs males; never smokers vs ever smokers) |
Was allowance made for uncertainty in the estimates of costs and consequences? | Univariate SA and PSA were undertaken by the manufacturer | PSA and univariate SA and scenario analysis were also undertaken by the manufacturer but only limited results of the one-way SA undertaken were presented in the MS |
Did the presentation and discussion of study results include all issues? | Not all comparators have been included | The results are presented and discussed in detail. Resources and infrastructure required to implement a universal EGFR mutation test for eligible patients is not fully discussed in the MS |
Attribute | Does the de novo economic evaluation match the reference case? (PEM)122 | Does the de novo economic evaluation match the reference case? (GEF)123 |
---|---|---|
Comparator(s) (therapies routinely used in the NHS) | Therapies routinely used in the NHS include GEM, VNB, DOC and PAX with a PLAT. VNB and PAX not included | Partially. Economic evaluation does not include DOC or PEM as comparators; both these comparators are routinely used in the NHS |
Perspective costs (NHS/PSS) | The economic evaluation is carried out from the perspective of the NHS. No social costs are described in the MS | The economic evaluation is carried out from the perspective of the NHS. No social costs are described in the MS |
Perspective benefits | Health effects to the individual are captured via QALYs | Health effects to the individual are captured via QALYs |
Economic evaluation (CEA) | CEA | CEA |
Time horizon (capture differences in costs/outcomes) | The time horizon chosen was a lifetime horizon (6 years). This appears appropriate | The time horizon chosen was a lifetime horizon, which for this patient group was believed to be 5 years. This appears to be appropriate |
Synthesis of evidence on outcomes (systematic review) | All outcome data are derived from RCTs. Indirect methodology was utilised, although this was not applied correctly | All survival data are derived (and where appropriate extrapolated) from a mix of clinical data sources: the IPASS RCT, meta-analysis (IPASS and NEJGSG) and mixed-treatment comparison; the meta-analysis and mixed-treatment comparison were based on systematic reviews of the literature |
Outcome measure (QALYs) | QALYs were used, which is appropriate | QALYs were used which is appropriate |
Health states for QALY (standardised and validated instrument) | QoL data were not available from any of the trials, therefore a published QoL study116 was utilised. This is not ideal, but the utility values appear to be reasonable | In the IPASS QoL was not measured in terms of utility. After a systematic review conducted by the manufacturer did not identify any relevant utility values for use in the economic evaluation, the utility values from Nafees 2008116 was used |
Benefit valuation | The QoL study116 utilised SG interview techniques, which is acceptable | The main QoL Nafees et al. 116 study utilised standard gamble interview techniques, which is acceptable |
Source of preference data for valuation of changes in HRQoL (TTO or SG) | The QoL study116 was based on responses from 100 members of the general public. It is not clear how representative this sample is | Main QoL study by Nafees et al. 116 was based on responses from 105 members of the general public. Unclear how representative this sample is of the UK adult population. Furthermore, the QoL study was not specifically designed to capture the QoL of patients requiring first-line treatment |
Discount rate (3.5%) | Benefits and costs, where appropriate, have been discounted using the 3.5% rate | Benefits and costs have been discounted using a rate of 3.5% |
Equity (QALYs have equal weight) | All QALYs estimated by the economic model have the same weight | All QALYs estimated by the economic model have the same weight |
Sensitivity analysis (PSA) | A PSA was conducted by the manufacturer | A PSA was conducted by the manufacturer |
In the case of PEM, according to the ERG,122 the manufacturer's economic evaluation did not fully adhere to the NICE reference case, in particular with regards to the inclusion of all relevant comparators. The ERG also found that the manufacturer's economic evaluation had quality issues identified by the Drummond and Jefferson checklist,106 again, owing to the omission of key comparators, but also because of problems with valuing outcomes.
In the case of GEF, according to the ERG,123 the manufacturer had attempted to adhere to the NICE reference case. However, as DOC and PEM are not included as comparators in the economic evaluation performed by the manufacturer, not all therapies routinely used in the NHS were considered. Furthermore, the ERG believed that the source of utility values used in the economic model might not be appropriate to the decision problem. The ERG reported that the manufacturer's submitted model failed on a number of issues including the exclusion of valid comparators and the incorrect identification and measurement of key costs and benefits. The ERG also highlighted that the manufacturer employed differential efficacy rates for the four chemotherapy regimens considered in the economic evaluation whereas the results of the manufacturer's own mixed-treatment comparison demonstrate equivalent efficacy rates for the same four chemotherapy regimens. Ultimately, the ERG questioned to what extent the clinical effectiveness of GEF is established for use in clinical practice in England and Wales.
The ERG reports are one of multiple sources of evidence for use in the first AC meeting. The recommendations set out in the appraisal consultation document and in the final appraisal document are not solely based on the ERG report. After the first AC meeting, second and/or third AC meetings may also take place to discuss any unresolved issues about the clinical effectiveness and cost-effectiveness evidence presented. The AC considered PEM + CIS at two AC meetings and GEF at three AC meetings. The final ICER estimates and conclusions of the AC for PEM + CIS and GEF are summarised in Table 51 and are described in the Final Appraisal Determination13,16 issued by NICE. For the non-squamous population, PEM + CIS appears to be cost-effective compared with GEM + CIS. For the EGFR+ population, GEF appears to be cost-effective compared with PAX + CARB when the manufacturer provides GEF at a reduced price.
Interventions | ERG (ICER estimate) | Manufacturer (ICER estimate) | NICE conclusions |
---|---|---|---|
PEM + CIS vs GEM + CIS13 | < £30,000 for patients with non-squamous disease; < £25,000 for patients with adenocarcinoma/large cell carcinoma | < £30,000 for patients with non-squamous disease; < £25,000 for patients with adenocarcinoma/large cell carcinoma | PEM + CIS is recommended as an option for the first-line treatment of patients with adenocarcinoma or large cell carcinoma |
GEF vs PAX + CARB16 | £23,000 to £64,000 | £19,000 to £23,000 | GEF is recommended as an option for the first-line treatment of people with locally advanced or metastatic NSCLC if:
|
Discussion and conclusions of economic evidence available
It is clear from the preceding sections that, although there exists published cost-effectiveness evidence comparing different first-line chemotherapy regimens for patients with NSCLC, very few studies are directly helpful to decision-makers in the NHS because the studies are not UK focused and/or they do not estimate ICERs in terms of cost per QALY gained.
The newer drugs that are now available to treat patients with NSCLC are not suitable for use in the overall NSCLC population and it is likely that the targeting of drugs to specific groups of patients will continue to play a role in the future development of drugs in this field. In contrast to the older drugs, newer drugs are subject to appraisal by NICE and the cost-effectiveness evidence submitted by manufacturers in support of these new drugs is more relevant to the needs of NHS decision-makers than ever before. However, there is a paucity of economic evaluations considering the use of the newer drugs for patients with NSCLC.
In summary, the conclusions of our systematic review echo the conclusions of the review by Carlson et al. 125 that was published in 2008. Carlson et al. 125 conclude that: ‘The results…reflect the large number of treatment strategies available in the treatment of NSCLC … given the absence of trials on newer therapeutics and the lack of CUAs, additional trials appear to be warranted, especially those that incorporate QoL considerations in the comparison of treatment strategies…’. 125
Independent economic assessment: methods
Assessment perspective
Costs and outcomes are assessed from the perspective of the UK NHS and Personal Social Services. Wider indirect costs and benefits (e.g. loss of productivity, value of informal care and impact on utility of patient's family) are not considered.
Relevant patient populations
Three distinct populations are modelled as follows:
-
chemotherapy-naive adult patients with locally advanced or metastatic NSCLC, which is not of predominantly non-squamous histology (referred to as ‘squamous disease population’)
-
chemotherapy-naive adult patients with locally advanced or metastatic NSCLC of predominantly non-squamous histology whose tumour(s) have not been shown to be EGFR M+ for activating mutations (referred to as ‘non-squamous disease population’)
-
chemotherapy-naive adult patients with locally advanced or metastatic NSCLC of predominantly non-squamous histology whose tumour(s) have been shown to be EGFR positive for activating mutations (referred to as ‘EGFR M+ population’).
Population (1) may only be treated with third-generation doublet chemotherapy. Population (2) may receive PEM + PLAT chemotherapy or a third-generation doublet chemotherapy. Population (3) has potentially the widest range of treatment options including those targeted for EGFR-activating mutations such as GEF, but no evidence is available for the efficacy of PEM + PLAT chemotherapy in this population subgroup.
Treatment options to be evaluated
A total of 12 first-line chemotherapy regimens are incorporated into the model (five primary licensed products used in combination with either CIS or CARB, PEM in combination with CIS, and GEF monotherapy). Details of these regimens are shown in Table 52 (together with two agents available for second-line chemotherapy), and correspond to the information contained in the Summary of Product Characteristics for each product. Information on the likely setting for treatment administration was provided by clinical advisors. CARB-based i.v. combination therapy is always delivered in a day-case unit. DOC is also administered in a day-case unit irrespective of the choice of PLAT compound. For other CIS-based combination regimens, there is variation in practice concerning the proportions of patients treated as inpatients or day cases.
Regimen | Chemotherapy | Population | Cycles | Doses per cycle | Dose given (mg/m2) | Comedications | Setting |
---|---|---|---|---|---|---|---|
First line | |||||||
1 A/B | DOC i.v. | Any NSCLC | 4 × 21 days | 1 | 75 | Dexamethasone | Day case |
2 A/B | GEM i.v. | Any NSCLC | 4 × 21 days | 2 | 1250 | No | A: Inpatient or day case B: Day case |
3 A/B | PAX i.v. | Any NSCLC | 4 × 21 days | 1 | 175 | Dexamethasone, chlorphenamine, ranitidine | A: Inpatient or day case B: Day case |
4 A/B | VNB i.v. | Any NSCLC | 4 × 21 days | 1 | 25 first cycle, 30 thereafter | No | A: Inpatient or day case B: Day case |
5 A/B | VNB oral | Any NSCLC | 4 × 21 days | 1 | 60 first cycle, 80 thereafter | No | A: Inpatient or day case B: Day case |
6 A | PEM i.v. | Non-squamous | 4 × 21 days | 1 | 500 | Dexamethasone, hydroxocobalamin, folic acid | Inpatient or day case |
7 | GEF oral | EGFR M+ | To progression | Daily | 250-mg tablet | No | Outpatient |
Second line | |||||||
8 | DOC i.v. monotherapy | Not previously treated with DOC | 4 × 21 days | 1 | 75 | Dexamethasone | Day case |
9 | ERL oral | Any NSCLC | To progression | Daily | 150-mg tablet | No | Outpatient |
Carboplatin has no licensed indication for use in combination therapy for advanced NSCLC, but is widely used as a less toxic alternative to CIS.
In the base-case analysis it is assumed that equal numbers of patients suitable for second-line chemotherapy receive DOC monotherapy and 50% receive ERL. However, patients receiving DOC as first-line chemotherapy will not be re-exposed to it, and therefore may only receive ERL in second-line treatment.
Although PEM is licensed as monotherapy for second-line chemotherapy, it has not been considered alongside DOC and ERL as an alternative i.v. treatment since it is substantially more expensive than DOC and is not recommended by NICE for use in the NHS. This limitation is likely to have a minimal effect on the cost-effectiveness of first-line regimens.
Model design
The decision model (Figure 25) is conceptually straightforward, involving three health states prior to death, and up to two lines of chemotherapy. Chemotherapy is treated as an extended event, normally restricted to a maximum of 12 weeks in duration (four cycles each of 3 weeks). The only exception is for orally administered treatments given continuously until the disease progresses (i.e. GEF and ERL) where treatment is assumed to be coterminous with the duration of the PFS state.
Disease progression after either first- or second-line therapy is also treated as an event, resulting in one of three possible transitions: to further active therapy (only after first-line chemotherapy), to supportive care only or to death.
The model is implemented as a Microsoft Excel workbook (Microsoft Corporation, Redmond, WA, USA), using macro programming to perform PSA to assess the relative probabilities of cost-effectiveness between the available first-line treatments.
Ideally, the model should be driven by evidence from clinical trials relating to each of the model's health states: the duration of PFS until first confirmed disease progression, the duration of PFS following second-line treatment, and the duration of postprogression survival (PPS) receiving only BSC. Unfortunately, the only outcomes routinely reported for clinical trials are PFS (first-line chemotherapy) and OS. Thus, the model can only be populated indirectly, by inferring the likely experience of patients in the intermediate states. This leads to potentially serious difficulties and inconsistencies in model implementation. In particular, the normal practice of treating PFS and OS as independent variables is naive, as PFS is a major component of OS. Not recognising this easily leads to situations where deriving an estimate for PPS by subtracting estimated PFS from estimated OS leads to erroneous negative values at some point during the simulation period. The modeller has to exercise great care at every stage of model development, calibration and use to guard against producing nonsensical results.
Synthesis of survival evidence: squamous and non-squamous disease
Effectiveness evidence from clinical trials identified as relevant to each population were synthesised in two stages: data from individual trial arms are pooled to produce a risk profile representative of each available treatment option, then a mixed-treatment comparison at a common time point was employed to estimate HRs to allow these risk profiles to be mutually calibrated while preserving randomisation within each trial.
Agent-specific outcome profiles
Kaplan–Meier estimates for OS and PFS/TTP for each regimen were compared across all trials and were pooled in order to obtain a standard cumulative hazard profile, which reflects the temporal changes in hazard typical of each chemotherapy agent. This involved extracting monthly survival estimates for 0–24 months from trial reports and then pooling these trends, weighting each data point by the number of patients in each included trial arm. The resulting survival estimates were then converted to cumulative hazards. The resulting hazard profile was then standardised to match the pooled value of a reference chemotherapy agent (PAX) at 12 months. These profiles do not distinguish between CIS and CARB doublets, which are assumed to be equivalent in terms of clinical effectiveness.
Table 53 details the PFS and OS profiles for months 0–24. In each case a piecewise profile model was fitted by least-squares regression using linear or quadratic segment functions, as described mathematically in Table 54. Constrained regression analysis (using SPSS 18; SPSS Inc., Chicago, IL, USA) was employed to generate parameter estimates for each model.
Month | Treatment | |||||||||
---|---|---|---|---|---|---|---|---|---|---|
PAX | DOC | GEM | VNB | PEM | ||||||
PFS | OS | PFS | OS | PFS | OS | PFS | OS | PFS | OS | |
0 | 0.000 | 0.000 | 0.000 | 0.000 | 0.000 | 0.000 | 0.000 | 0.000 | 0.000 | 0.000 |
1 | 0.103 | 0.045 | 0.096 | 0.055 | 0.076 | 0.038 | 0.087 | 0.042 | 0.057 | 0.035 |
2 | 0.373 | 0.098 | 0.379 | 0.108 | 0.255 | 0.094 | 0.306 | 0.080 | 0.209 | 0.079 |
3 | 0.530 | 0.172 | 0.506 | 0.172 | 0.392 | 0.157 | 0.525 | 0.144 | 0.314 | 0.139 |
4 | 0.670 | 0.247 | 0.641 | 0.235 | 0.542 | 0.218 | 0.720 | 0.212 | 0.452 | 0.207 |
5 | 0.936 | 0.328 | 0.901 | 0.316 | 0.771 | 0.311 | 0.847 | 0.287 | 0.687 | 0.267 |
6 | 1.117 | 0.419 | 1.092 | 0.388 | 1.002 | 0.401 | 1.142 | 0.370 | 0.923 | 0.394 |
7 | 1.377 | 0.523 | 1.257 | 0.491 | 1.277 | 0.504 | 1.347 | 0.455 | 1.287 | 0.507 |
8 | 1.615 | 0.615 | 1.542 | 0.594 | 1.527 | 0.585 | 1.449 | 0.573 | 1.589 | 0.594 |
9 | 1.792 | 0.703 | 1.811 | 0.693 | 1.791 | 0.676 | 1.800 | 0.659 | 1.810 | 0.687 |
10 | 2.003 | 0.804 | 1.913 | 0.799 | 1.992 | 0.767 | 2.019 | 0.762 | 1.991 | 0.801 |
11 | 2.214 | 0.891 | 2.214 | 0.883 | 2.196 | 0.879 | 2.169 | 0.866 | 2.195 | 0.888 |
12 | 2.379 | 0.983 | 2.379 | 0.983 | 2.379 | 0.983 | 2.379 | 0.983 | 2.379 | 0.983 |
13 | 2.527 | 1.096 | 2.556 | 1.114 | 2.548 | 1.083 | 2.450 | 1.064 | 2.516 | 1.071 |
14 | 2.638 | 1.193 | 2.671 | 1.195 | 2.705 | 1.203 | 2.678 | 1.182 | 2.876 | 1.161 |
15 | 2.723 | 1.278 | 2.817 | 1.236 | 2.802 | 1.288 | 2.768 | 1.296 | 2.876 | 1.295 |
16 | 2.856 | 1.349 | 3.135 | 1.320 | 2.970 | 1.370 | 2.944 | 1.398 | 2.968 | 1.413 |
17 | 2.979 | 1.410 | 3.445 | 1.374 | 3.090 | 1.443 | 2.993 | 1.490 | 3.071 | 1.460 |
18 | 3.110 | 1.493 | 3.605 | 1.457 | 3.180 | 1.515 | 3.561 | 1.601 | 3.071 | 1.538 |
19 | 3.266 | 1.566 | 3.605 | 1.492 | 3.289 | 1.606 | 3.637 | 1.681 | 3.247 | 1.656 |
20 | 3.389 | 1.661 | 3.994 | 1.567 | 3.372 | 1.685 | 3.729 | 1.785 | 3.247 | 1.717 |
21 | 3.389 | 1.754 | 4.218 | 1.627 | 3.413 | 1.756 | 3.976 | 1.876 | 3.247 | 1.810 |
22 | 3. 495 | 1.858 | 4.307 | 1.681 | 3.587 | 1.887 | 3.976 | 1.941 | 3.389 | 1.841 |
23 | 3.701 | 1.931 | 4.307 | 1.800 | 3.701 | 1.988 | 4.570 | 2.043 | 3.389 | 1.944 |
24 | 3.752 | 1.959 | 4.307 | 1.845 | 3.818 | 2.077 | 4.570 | 2.122 | 3.389 | 1.996 |
Sources | 47, 43, 46, 56, 57, 60 | 43, 46–48, 51, 56, 57, 60 | 47, 59 | 44, 47, 53, 65 | 43, 46, 47, 54, 56, 57, 60, 61 | 43, 46, 47, 50, 54–58, 60, 61 | 43, 54 58 | 43, 44, 48, 50, 51, 53, 54, 57, 58 | 61 | 61 |
Regimen | PFS profile cumulative hazard model | OS profile cumulative hazard model |
---|---|---|
PAX + PLAT | H(t) = 0.19821 × t [t< 12.40 months] H(t) = 2.457 + 0.11459 × (t−12.40) [t≥ 12.40 months] |
H(t) = 0.04465 × t [t< 1.53 months] H(t) = 0.087 + 0.08739 × (t−1.53) [t≥ 1.53 months] |
DOC + PLAT | H(t) = 0.19821 × t [t in months] |
H(t) = 0.06012 × t [t< 5.18 months] H(t) = 0.311 + 0.09852 × (t−5.18) [5.18 ≤ t<14.00 months] H(t) = 1.180 + 0.06557 × (t−14.00) [t≥ 14.00 months] |
GEM + PLAT | H(t) = 0.12583 × t [t < 3.19 months] H(t) = 0.401 + 0.22441 × (t− 3.19) [3.19 ≤ t < 13.14 months] H(t) = 2.635 + 0.10776 × (t−13.14) [t ≥ 13.14 months] |
H(t) = 0.04544 × t [t < 2.55 months] H(t) = 0.116 + 0.09181 × (t−2.55) [t ≥ 2.55 months] |
VNB + PLAT | H(t) = 0.19821 × t [t < 12.00 months] H(t) = 2.379 + 0.16717 × (t − 12.00) [t ≥ 12.00 months] |
H(t) = 0.04946 × t [t< 4.07 months] H(t) = 0.201 + 0.09862 × (t−4.07) [t ≥ 4.07 months] |
PEM + CIS | H(t) = 0.04051 × t + 0.01982 × t2 [t < 7.94 months] H(t) = 1.573 + 0.19870 × (t−7.94) [7.94 ≤ t< 14.62 months] H(t) = 2.899 + 0.06943 × (t−14.62) [t ≥ 14.62 months] |
H(t) = 0.02503 × t + 0.00650 × t2 [t <7.00 months] H(t) = 0.494 + 0.09793 × (t − 7.00) [7.00≤ t <17.68 months] H(t) = 1.540 + 0.07417 × (t−17.68) [t ≥ 17.68 months] |
The cumulative hazard profile models are illustrated in Figures 26–29 (see Figures 26 and 28 for PFS and Figures 27 and 29 for OS). These suggest that the third-generation agents generate outcomes in quite similar ways, though exhibiting more divergence in the second year of survival when some treatments show an apparent moderation of long-term risks.
When these profiles are examined in the form of traditional survival curves, differences in the short term are more easily seen, especially for the PFS models.
Hazard ratios
Derivation and application of hazard ratios
Hazard ratios for OS and PFS/TTP were obtained from a network meta-analysis of relevant trials based on determining the HR of each first-line regimen relative to a PAX doublet regimen. Table 55 shows the values obtained for use as model parameters; full details of the mixed-treatment comparison are shown in Chapter 3, Population 1: non-small cell lung cancer patients with squamous disease and Chapter 3, Population 2: non-small cell lung cancer patients with non-squamous disease. HRs only differed significantly from PAX for OS in the case of PEM.
Regimen | HR | LCL | UCL | SE(ln[HR]) | p-value |
---|---|---|---|---|---|
Squamous disease | PFS | ||||
PAX | 1.000 | – | – | – | – |
DOC | 0.966 | 0.785 | 1.168 | 0.101 | 0.365 |
VNB | 0.923 | 0.823 | 1.031 | 0.058 | 0.082 |
GEM | 0.971 | 0.829 | 1.134 | 0.080 | 0.355 |
Squamous disease | OS | ||||
PAX | 1.000 | – | – | – | – |
DOC | 0.942 | 0.805 | 1.106 | 0.081 | 0.232 |
VNB | 0.953 | 0.870 | 1.045 | 0.047 | 0.154 |
GEM | 1.040 | 0.928 | 1.168 | 0.059 | 0.745 |
Non-squamous disease | PFS | ||||
PAX | 1.000 | – | – | – | – |
DOC | 0.961 | 0.787 | 1.171 | 0.102 | 0.363 |
VNB | 0.922 | 0.823 | 1.033 | 0.058 | 0.080 |
GEM | 0.971 | 0.832 | 1.134 | 0.079 | 0.356 |
PEM | 0.831 | 0.653 | 1.049 | 0.121 | 0.063 |
Non-squamous disease | OS | ||||
PAX | 1.000 | – | – | – | – |
DOC | 0.942 | 0.803 | 1.106 | 0.082 | 0.230 |
VNB | 0.954 | 0.871 | 1.045 | 0.046 | 0.155 |
GEM | 1.040 | 0.931 | 1.168 | 0.058 | 0.748 |
PEM | 0.770 | 0.636 | 0.941 | 0.100 | 0.005 |
These HRs were then applied to adjust the standardised cumulative hazard profile of each regimen to obtain a final characterisation of treatment effectiveness of each regimen for use in the decision model.
Uncertainty in hazard ratios
Ideally, the model would have been constructed using PFS and PPS as the primary outcome measures, with OS used as a confirmation of model reliability. Unfortunately, PPS is not reported in clinical trials and the model was constructed to reflect the PFS and OS data available. This presents a difficulty for projective modelling, and also in the representation of parameter uncertainty when carrying out PSA. Patient numbers in PPS are usually estimated as the numerical difference between numbers in OS and PFS at each time point. This can easily generate a sequence of negative results which are clearly meaningless and lead to erroneous results.
To overcome this problem, uncertainty in the HRs was addressed by use of linked variations in PFS and OS HRs, based on the estimated proportion of OS contributed by PFS leading to correlated random variables for PFS and OS. The proportions used for each regimen are shown in Table 56.
Regimen | PFS:OS ratio |
---|---|
DOC | 0.41 |
PAX | 0.43 |
VNB | 0.43 |
GEM | 0.48 |
PEM | 0.57 |
Synthesis of survival evidence: epidermal growth factor receptor mutation-positive population
Consistency of third-generation trial data
In order to include clinical trial evidence into a network for mixed-treatment comparison analysis it is important to establish compatibility of the populations studied and consistent treatment effects across trials. The many important trials of third-generation chemotherapy agents were carried out prior to widespread use of histology testing and before any genetic testing methods had been developed. However, third-generation trials continue to provide the bulk of evidence available to allow relative effectiveness of treatments to be assessed. The inclusion of PEM therapy in such an evidence network requires some confirmation that undifferentiated third-generation trials did not in fact conceal unsuspected important differences originating in different disease histology. A review of the available published trials identified four studies which reported multivariate statistical testing including histology as a candidate explanatory variable: Smit et al. 46 (PAX vs GEM), Gridelli et al. 45 (VNB vs GEM), Fossella et al. 44 (DOC vs VNB) and Scagliotti et al. 43 (GEM vs PAX vs VNB). In none of these trials did the authors report any significant influence of histology (squamous vs non-squamous) in determining effectiveness. On this basis it was considered appropriate to assume that trial evidence from trials of third-generation chemotherapy agents are equally applicable to patients with squamous disease as to those with non-squamous disease.
Inconsistency of third-generation trial data in gefitinib trials
However, the situation is quite different for patients with EGFR M+ disease, who predominantly have non-squamous histology. Only a limited number of trials with modest numbers of such patients have so far reported results. All of these compare EGFR-TKI products with third-generation chemotherapy regimens, but none compare with PEM + CIS which is indicated specifically for non-squamous (adenocarcinoma and large cell) disease. In order to consider the viability of incorporating all available third-generation trial evidence in an evidence network including GEF therapy, the PFS and OS profiles of the comparator arms in three GEF trials were compared with the profiles of the same treatments in the full third-generation network. This revealed that effectiveness of third-generation treatments was consistently far better in the EGFR M+ population than in the mixed populations (squamous and non-squamous disease), indicating that these patients have a better prognosis than other NSCLC patients, independent of the treatment received.
As a result it was considered inappropriate to carry out any meta-analysis involving third-generation trials not restricted to the EGFR M+ population and, therefore, no relative effectiveness estimates could be derived relating GEF to PEM (which would otherwise be a natural comparator for GEF). Instead, a separate analysis was undertaken restricted to the three reported GEF trials involving non-trivial numbers of EGFR M+ patients: IPASS,15,64 North East Japan Gefitinib Study Group (NEJGSG)63 and Western Japan Thoracic Oncology Group (WJTOG). 65 The synthesis method employed was weighted pooling of the PFS and OS Kaplan–Meier results for the GEF arm and for the comparator arm of the trials, and using these profiles directly to inform the model. The base case uses data from all three trials (despite mixing PAX and DOC comparator arms) and testing both PAX and DOC comparators in the model. Two alternative scenarios were also considered: A1 – pooling only the two trials involving a PAX comparator (IPASS15,64 and NEJGSG63); and A2 – using the WJTOG65 trial results directly in the model compared with DOC as comparator.
Other outcome variables
Patient disposition at disease progression
Following a PFS event (i.e. confirmed disease progression or death without prior disease progression) it is important to estimate the proportions of patients likely to receive additional systemic treatment or palliative care only. This requires values to be estimated for two parameters:
-
the proportion of PFS events which are fatal
-
the proportion of patients receiving at least second-line systemic treatment.
From these the proportion of patients receiving only palliative care after failure of first-line treatments can be derived.
Unfortunately, neither of these outcomes are routinely reported in published clinical trials, nor even in clinical study reports. Only two trials were found from which fatality figures could be deduced (Chen et al. 52 and Fukuoka et al. 64) and since these relate to different patient populations and different interventions, pooling these results would be inappropriate. In practice, it was found that the main limitation on the fatality parameter is the need to ensure that the model logic does not generate negative estimates of PPS at any time for any treatment, and this imposes an effective maximum fatality between 20% and 25%. In the base-case analysis, 16% fatality is assumed with sensitivity analysis performed to explore the impact of uncertainty. The logic for the choice of 16% for this parameter was preserve the integrity of the PPS estimates by limiting the upper end of the PSA sampling range to effectively exclude any negative postprogression values being generated, assuming that the standard error of the parameter was 10% of the chosen mean value, and a maximum sampling point corresponded to 4 standard errors above the mean.
Patients receiving second-line systemic therapies were reported in 10 trials,43–45,50,57,59,61,64,126,127 from which was obtained a pooled estimate of 45.2% for all populations, or 34.5% for the squamous disease population, 40.6% for the non-squamous disease population (including Scagliotti et al. 128 with the third-generation trials), 71.3% for DOC or PAX, and 77.5% for GEF in the EGFR M+ population (taken from the supplementary appendix by Mok et al. 15).
Agent-specific adverse events
The costs and disutilities of treatment-related AEs are limited in the model to seven major categories (using the results of a multivariate model by Nafees et al. 116 described in detail below): diarrhoea, fatigue, neutropenia, febrile neutropenia, hair loss, nausea/vomiting and skin rash.
Reported incidence of grade 3/4 AEs in all published trials were pooled to obtain estimates of the proportion of patients suffering each event during chemotherapy. No attempt was made to carry out a more sophisticated meta-analysis as reporting of AEs was often incomplete and lacking in consistency. Table 57 details the incidence rates obtained for each primary chemotherapy agent. No attempt has been made to distinguish between the types of PLAT treatment given in first-line doublets, as there were inadequate data in many cases to obtain meaningful estimates at that level of disaggregation.
Treatment | AE | ||||||
---|---|---|---|---|---|---|---|
Diarrhoea | Fatigue | Febrile neutropenia | Hair loss | Nausea/vomiting | Neutropenia | Skin rash | |
DOC | |||||||
Mean (%) | 6.4 | 9.0 | 2.9 | 0.0 | 20.4 | 62.1 | 0.0 |
95% CI | 5.2 to 7.7 | 7.6 to 10.5 | 2.1 to 3.7 | 0.0 to 0.2 | 18.4 to 22.4 | 59.7 to 64.5 | 0.0 to 0.2 |
VNB | |||||||
Mean (%) | 1.8 | 10.8 | 6.6 | 1.2 | 20.3 | 63.1 | 0.1 |
95% CI | 1.2 to 2.6 | 9.3 to 12.3 | 5.4 to 7.9 | 0.7 to 1.8 | 18.4 to 22.2 | 60.8 to 65.4 | 0.0 to 0.5 |
PAX | |||||||
Mean (%) | 2.3 | 7.1 | 4.9 | 0.0 | 13.5 | 57.4 | 0.4 |
95% CI | 1.7 to 3.0 | 6.1 to 8.3 | 4.1 to 5.8 | 0.0 to 0.2 | 12.2 to 14.9 | 55.4 to 59.3 | 0.2 to 0.8 |
GEM | |||||||
Mean (%) | 1.8 | 11.7 | 2.8 | 1.4 | 19.1 | 37.1 | 0.5 |
95% CI | 1.2 to 2.4 | 10.2 to 13.3 | 2.1 to 3.7 | 0.9 to 1.9 | 17.6 to 20.6 | 35.3 to 39.0 | 0.2 to 0.9 |
PEM | |||||||
Mean (%) | 1.3 | 6.7 | 1.3 | 0.0 | 11.2 | 20.6 | 0.1 |
95% CI | 0.7 to 2.2 | 5.1 to 8.6 | 0.7 to 2.2 | 0.0 to 0.3 | 9.4 to 13.2 | 18.2 to 23.1 | 0.0 to 0.4 |
GEF | |||||||
Mean (%) | 3.1 | 0.9 | 0.1 | 0.0 | 0.6 | 2.8 | 3.3 |
95% CI | 2.0 to 4.4 | 0.4 to 1.6 | 0.0 to 0.5 | 0.0 to 0.3 | 0.2 to 1.3 | 1.8 to 4.1 | 2.2 to 4.7 |
ERL | |||||||
Mean (%) | 1.5 | 3.3 | 0.0 | 0.0 | 0.7 | 0.0 | 8.0 |
95% CI | 0.8 to 2.4 | 2.2 to 4.5 | 0.0 to 0.3 | 0.0 to 0.3 | 0.2 to 1.3 | 0.0 to 0.3 | 6.3 to 9.8 |
Agent-specific response rates
The Nafees et al. 116 multivariate utility model also includes two levels of response to chemotherapy as predictive variables: ‘responder’ (either complete or partial response) and ‘stable disease’ (neither response nor disease progression). Estimates for these variables were obtained by pooling reported responses in published clinical trials in a similar manner to the derivation of AE incidence rates. The parameter values obtained are shown in Table 58.
Treatment | Responders (%) | Stable disease (%) | ||
---|---|---|---|---|
Mean | 95% CI | Mean | 95% CI | |
DOC | 26.7 | 24.5 to 29.0 | 39.1 | 36.7 to 41.6 |
PAX | 27.5 | 25.6 to 29.3 | 34.1 | 32.1 to 36.1 |
VNB | 28.6 | 26.4 to 31.0 | 36.5 | 34.1 to 39.0 |
GEM | 27.3 | 25.4 to 29.3 | 38.5 | 36.4 to 40.6 |
PEM | 30.6 | 27.4 to 33.9 | 41.2 | 37.7 to 44.7 |
PAX + DOC − (EGFR M+) − base case | 38.1 | 32.7 to 43.6 | 44.7 | 39.1 to 50.3 |
PAX (EGFR M+) − A1 | 39.5 | 33.5 to 45.7 | 44.4 | 38.3 to 50.7 |
DOC (EGFR M+) − A2 | 32.2 | 21.0 to 44.5 | 45.8 | 33.3 to 58.5 |
GEF (EGFR M+) − base case | 70.4 | 65.1 to 75.4 | 20.7 | 16.4 to 25.4 |
GEF (EGFR M+) − A1 | 72.4 | 66.6 to 77.8 | 18.3 | 13.7 to 23.3 |
GEF (EGFR M+) − A2 | 62.1 | 49.3 to 74.0 | 31.0 | 19.9 to 43.4 |
Chemotherapy acquisition costs
With the exception of the oral medications (GEF and ERL), all chemotherapy doses are calculated individually on the basis of the patient's body surface area. Calculations are carried out separately for males and females, and a weighted average cost is obtained using the relative proportions of recorded deaths from malignant neoplasm of trachea, bronchus and lung in England and Wales in 2010 (56.1% males, 43.9% females). 124 CIS costs are calculated for a single dose of 75 mg/m2 each cycle. CARB costs are based on a dose of 400 mg/m2 each cycle, with an alternative option based on flat dosing for a target AUC 5 level as described by Ekhart et al. 129 Sensitivity analysis by dosing calculation method should that using the alternative method produced minimal differences in any of the cost-effectiveness results described below.
Two sources are available as options to provide unit costs of purchasing chemotherapy drugs: the prices of generic medicines listed in the BNF (BNF 62,130 September 2011) and the electronic market information tool131 (eMIT) produced by the Commercial Medicines Unit of the Department of Health which provides estimated mean product prices for generic medicines drawn from information from about 95% of NHS trusts. Table 59 summarises the unit cost data employed in the estimation of chemotherapy acquisition costs.
Product | Vial content (mg) | BNF 62130 price, mean (£) | eMIT131 price, mean (£) |
---|---|---|---|
DOCa | 20 | 154.61 | 90.20 |
80 | 508.01 | 287.45 | |
140 | 720.10 | 285.09 | |
GEMa | 200 | 32.00 | 4.81 |
1000 | 162.00 | 22.58 | |
2000 | 324.00 | 41.99 | |
PAXa | 30 | 66.85 | 5.02 |
100 | 200.35 | 13.28 | |
150 | 300.52 | 12.45 | |
300 | 601.03 | 31.13 | |
PEM | 100 | 160.00 | 160.00 |
500 | 800.00 | 800.00 | |
VNB i.v.a | 10 | 29.00 | 5.11 |
50 | 139.00 | 23.09 | |
VNB oral | 20 | 43.98 | 43.98 |
30 | 65.98 | 65.98 | |
80 | 175.92 | 175.92 | |
GEFb CISa |
Per patient | 12,200.00 | 12,200.00 |
10 | 5.85 | 1.69 | |
50 | 17.00 | 3.58 | |
100 | 50.22 | 6.87 | |
CARBa | 50 | 22.04 | 2.03 |
150 | 56.92 | 4.65 | |
450 | 168.85 | 13.50 | |
600 | 260.00 | 17.23 | |
ERL | 30 × 150 mg | 1631.53 | 1631.53 |
NHS discount | 14.50% | 14.50% | |
Dexamethasonea | 50 × 2 mg | 6.77 | 1.99 |
Chlorphenamine i.v.a | 10 | 1.95 | 1.62 |
Ranitidine i.v.a | 50 | 0.54 | 0.31 |
Hydroxocobalamin i.v.a | 1 | 0.68 | 0.31 |
Folic acida | 90 × 400 µg | 2.43 | 2.43 |
Chemotherapy costs are estimated per 21-day cycle for all regimens except GEF, where a fixed price per patient receiving more than two packs of tablets has been negotiated for use in England and Wales. These are shown in Table 60, for both BNF and eMIT prices; the base-case analysis is carried out using the BNF prices but, in general, eMIT prices may be considered more representative of the normal NHS cost environment.
Regimen | Estimated cost − BNF 62130 prices (£) | Estimated cost − eMIT131 prices (£) | |||||
---|---|---|---|---|---|---|---|
Cycle 1 | Cycles 2+ | Per patient | Cycle 1 | Cycles 2+ | Per patient | ||
First-line regimens | |||||||
1A | DOC i.v. + CIS | 852.17 | 852.17 | NA | 367.52 | 367.52 | NA |
1B | DOC i.v. + CARB | 1081.46 | 1081.46 | NA | 377.83 | 377.83 | NA |
2A | GEM i.v. + CIS | 807.19 | 807.19 | NA | 112.16 | 112.16 | NA |
2B | GEM i.v. + CARB | 1036.47 | 1036.47 | NA | 122.48 | 122.48 | NA |
3A | PAX i.v. + CIS | 698.27 | 698.27 | NA | 49.16 | 49.16 | NA |
3B | PAX i.v. + CARB | 927.55 | 927.55 | NA | 59.47 | 59.47 | NA |
4A | VNB i.v. + CIS | 330.97 | 380.55 | NA | 58.55 | 66.78 | NA |
4B | VNB i.v. + CARB | 560.25 | 609.83 | NA | 68.86 | 77.10 | NA |
5A | VNB oral + CIS | 537.26 | 546.10 | NA | 496.50 | 505.34 | NA |
5B | VNB oral + CARB | 766.54 | 775.38 | NA | 506.81 | 515.65 | NA |
6A | PEM i.v. + CIS | 1535.40 | 1535.40 | NA | 1493.11 | 1493.11 | NA |
7 | GEF oral | NA | NA | 12,200.00 | NA | NA | 12,200.00 |
Second-line chemotherapy | |||||||
8 | DOC i.v. monotherapy | 799.66 | 799.66 | NA | 355.77 | 355.77 | NA |
9 | ERL oral | 1394.96 | 1394.96 | NA | 1394.96 | 1394.96 | NA |
Administration cost of chemotherapy regimens
Clinical advisors from three specialist centres provided information on the context within which each regimen is normally delivered. There was general agreement that combination chemotherapy using CARB is always administered as a day-case episode, and that treatments involving only daily self-administered oral medication are prescribed at a monthly outpatient consultation. Combination regimens involving CIS show variation in clinical practice from 100% managed as day cases to up to 80% requiring an inpatient stay.
It was decided to assume 100% of these patients are managed as day cases, but to apply a sensitivity analysis in which 50% of patients require an additional overnight stay following administration.
The unit costs employed for chemotherapy administration, based on NHS Reference Costs 2009–2010,132 are shown in Table 61.
Treatment setting | HRG code | Description | Mean (£) | Standard error (£) |
---|---|---|---|---|
Day case | SB14Z | Complex chemotherapy at first attendance | 309.17 | 14.73 |
Day case | SB15Z | Subsequent doses of chemotherapy | 284.45 | 8.95 |
Inpatient (short stay) | DZ17A | Respiratory neoplasms with complicating conditions | 462.88 | 12.88 |
Outpatient | TCLFUSFF 370 | Medical oncology | 128.69 | 3.92 |
Health state costs
Costs have been estimated relating to patient monitoring and supportive care in three health states: in PFS (either during and following first-line chemotherapy or subsequently related to second-line chemotherapy), post progression when no active treatment is received, and for terminal care assumed to last on average for 14 days.
In both PFS and PPS, patients are expected to receive regular consultant-led outpatient consultations, and periodic diagnostic tests [chest radiography, CT scan and electrocardiogram (ECG)]. In addition, community-based supportive care is provided by the patient's general practitioner (GP) (in surgery, or at home) and community nursing staff. In the terminal phase, care is likely to be more intensive, with the package varying by the chosen setting.
Table 62 details the mean volumes of each resource assumed and Table 63 summarises the unit costs employed together with the relevant sources.
Resource | PFS | PPS | Terminal care | Source |
---|---|---|---|---|
Outpatient visit | 9.61 pa | 7.91 pa | – | Big Lung Trial133 |
Chest radiography | 6.79 pa | 6.50 pa | – | Big Lung Trial133 |
CT scan (chest) | 0.62 pa | 0.24 pa | – | Big Lung Trial133 |
CT scan (other) | 0.36 pa | 0.42 pa | – | Big Lung Trial133 |
ECG | 1.04 pa | 0.88 pa | – | Big Lung Trial133 |
Hospital/hospice episode | – | – | 9.66 days | Average stay for non-elective long-stay inpatient episode plus average inpatient excess days for HRG DZ17A – NHS Reference Costs 2009–2010132 |
Community nurse visit | 8.70 visits (20 minutes) pa | 8.70 visits (20 minutes) pa | 28 hours (2 hours per day) | Appendix 1 of NICE Guideline CG81134 |
Marie Curie report135 | ||||
Clinical nurse specialist | 12 hours contact time pa | 12 hours contact time pa | Appendix 1 of NICE Guideline CG81134 | |
GP surgery | 12 consultations pa | – | – | Appendix 1 of NICE Guideline CG81134 |
GP home visit | – | 26.09 pa (fortnightly) | Seven visits (alternate days) | Marie Curie report135 |
Therapist visit | – | 26.09 pa (fortnightly) | – | Appendix 1 of NICE Guideline CG81134 |
Macmillan nurse | – | 50 hours | Marie Curie report135 | |
Drugs/equipment | – | – | As required | Marie Curie report135 |
Location of terminal care | – | – | Hospital 55.8% | Office for National Statistics death tables 5.2 and 12124 |
Hospice 16.9% | ||||
Home 27.3% |
Resource | Unit cost | Source |
---|---|---|
Outpatient follow-up visit | £101.43 | NHS Reference Costs 2009–2010, HRG code TCLFUSFF 800 clinical oncology132 |
Chest radiography | £24.04 | NICE technology appraisal TA199; TAG report, p. 328136 |
CT scan (chest) | £145.83 | NHS Reference Costs 2009–2010, HRG code RA12Z (two areas with contrast)132 |
CT scan (other) | £162.25 | NHS Reference Costs 2009–2010, HRG code RA13Z (three areas with contrast)132 |
ECG | £32.69 | NHS Reference Costs 2009–2010, code DA01 – direct access ECG (12 lead)132 |
Community nurse | £78.00 per hour | PSSRU Unit Costs of Health and Social Care 2010, p. 159 cost per hour spent on home visits (including qualification)137 |
Clinical nurse specialist | £91.00 per contact hour | PSSRU Unit Costs of Health and Social Care 2010, p. 162 cost per contact hour (including qualification)137 |
GP surgery visit | £36.00 | PSSRU Unit Costs of Health and Social Care 2010, p. 167 cost per surgery visit (11.7 minutes, including direct care staff)137 |
GP home visit | £120.00 | PSSRU Unit Costs of Health and Social Care 2010, p. 167 cost per home visit (23.4 minutes, including travel time)137 |
Therapist | £42.00 | PSSRU Unit Costs of Health and Social Care 2010, p. 177 cost per hour (including training)137 |
Terminal care inpatient care | £2655.55 + 0.92 excess days at £196.61 per day | NHS Reference Costs 2009–2010, code DZ17A (respiratory neoplasms with major CC), non-elective inpatient (long stay – episode/excess days)132 |
Terminal care in hospice | 25% increase on hospital IP care | Assumption |
Macmillan nurse | 66.7% of community nurse cost | Assumption |
Drugs and equipment | £500 | Marie Curie report figure of £240 increased for inflation135 |
Adverse event costs
The costs of treating grade 3/4 chemotherapy-related AEs are spread over 12 weeks (four cycles) and estimated using NHS Reference Costs for 2009–2010,132 as follows.
Diarrhoea
It is assumed that a typical patient will have two hospital admissions during chemotherapy, corresponding to Healthcare Research Group (HRG) code FZ48C (malignant general abdominal disorders of length of stay ≤ 1 day) as a non-elective short-stay episode, each costing £443.54.
Fatigue
It is assumed that a typical patient will have one hospital admission during chemotherapy, corresponding to HRG code WA17X (other admissions related to neoplasms with intermediate complicating conditions) as a non-elective long-stay episode of 8–9 days costing £2536.95.
Hair loss
It is assumed that there are no hospital episodes related to the AE and no direct costs are incurred.
Nausea/vomiting
It is assumed that a typical patient will have two hospital admissions during chemotherapy, corresponding to HRG code FZ48C (malignant general abdominal disorders of length of stay ≤ 1 day) as a non-elective short-stay episode, each costing £443.54.
Skin rash
It is assumed that a typical patient will have one additional outpatient consultation during chemotherapy for this condition. A weighted average reference cost of £113.03 is used, based on codes 370 (medical oncology) and 800 (clinical oncology).
Neutropenia (non-febrile)
It is assumed that 10% of patients require hospital treatment, each requiring two episodes during chemotherapy. The cost per episode is £537.52 and is estimated from the weighted average of mean costs for HRG code WA02W (disorders of immunity without HIV/AIDS with complicating condition) across non-elective long- and short-stay episodes and day-case admissions.
Febrile neutropenia
The NICE Decision Support Unit report on the cost of febrile neutropenia has been updated for current NHS Reference Costs 2009–2010. 132 This assumes 1.4 episodes per patient during the four cycles (12 weeks) of chemotherapy. The estimated cost per patient suffering febrile neutropenia is £6260.
In the model, the estimated cost per patient of chemotherapy-related AEs is shown in Table 64.
AE | Product cost (£) | ||||||
---|---|---|---|---|---|---|---|
DOC | VNB | PAX | GEM | PEM | GEF | ERL | |
Diarrhoea | 57 | 16 | 20 | 16 | 12 | 27 | 14 |
Fatigue | 229 | 273 | 181 | 297 | 171 | 22 | 83 |
Febrile neutropenia | 179 | 411 | 310 | 172 | 83 | 8 | 0 |
Hair loss | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Nausea/vomiting | 181 | 180 | 120 | 170 | 100 | 5 | 6 |
Neutropenia | 129 | 131 | 119 | 77 | 43 | 6 | 0 |
Skin rash | 0 | 0 | 2 | 2 | 0 | 11 | 27 |
Total AE cost | 773 | 1011 | 751 | 733 | 409 | 80 | 129 |
Health valuation estimation
Ideally, the utility of NSCLC patients should be informed by data obtained directly from the relevant patient population relating to their perceived condition at all phases of the treatment pathway covered by the economic model. Unfortunately, this is practically and ethically impractical for patients suffering advanced disease with severe symptoms (arising from either the natural course of the disease or related to treatments received) and who have generally very limited life expectancy. A recent study in the Netherlands33 attempted to obtain such data (using the EQ-5D instrument) from an observational study of NSCLC patients treated between 2004 and 2007, and surviving to 2008. Unfortunately, this patient sample is not representative of the populations considered in this model (locally advanced and metastatic NSCLC) since only 44% of patients had received any chemotherapy, only 41% had stage III/IV disease and only 14% had local/regional or metastatic recurrent disease at the time of the survey. Clearly, the results obtained are dominated by patients who were diagnosed at an early stage and had successful surgery, potentially biasing numeric estimates of utility toward higher values.
The only alternative to direct measurement of patient symptoms for estimating utility is via a structure sample of the general public valuing a set of typical patient scenarios, representing the range of likely conditions experienced by NSCLC patients during their remaining lifetime. Two such recent studies have been identified. Doyle et al. 138 recruited 101 volunteers from the general public in the London (UK) area, who were asked to value six typical health states experienced by advanced NSCLC patients, using the standard gamble method. This allowed estimation of a mean utility value for patients with stable disease on treatment, as well as the incremental effect of response to treatment, and also the incremental disutility of three common symptoms (cough, dyspnoea and pain). Although promising, this study provides only limited results which are insufficient to populate all the health states and important AEs which feature in the current model.
The utility scheme which has been adopted for use in the current model is that described in a paper published in 2008 by Nafees et al. 116 This also uses the standard gamble method and employed 100 volunteers from the UK general population. In this case a more extensive set of scenarios were used (17 specific disease health states plus two ‘anchor’ states), developed with the help of a panel of oncologists and designed specifically to address a range of the most common severe AEs experienced by advanced NSCLC patients. A mixed-model analysis yielded simultaneous utility estimates for three health states (responding to treatment, stable disease and progressive disease) together with incremental disutility values for seven common serious (grade 3/4) AEs – neutropenia, febrile neutropenia, fatigue, diarrhoea, nausea and vomiting, hair loss (alopecia) and rash.
Applying the treatment-specific AE incidence rates (see Table 57) and treatment response rates (see Table 58) to the Nafees et al. 116 utility model yields a full set of health state utilities for each treatment option as shown in Table 65. The utility for the terminal period (last 2 weeks of life) was obtained by use of results reported for average EQ-5D scores relative to the time prior to death (figure 3 of the van der Hout et al. 2006 study139 of palliative radiotherapy in patients with NSCLC) and the utility estimate for PPS 2 was adjusted to reflect progressive disease prior to the terminal period.
First-line chemotherapy | PFS 1 on treatment | PFS 1 post treatment | PPS 1 following first progression |
---|---|---|---|
DOC | 0.5833 | 0.6610 | 0.4896 |
PAX | 0.5929 | 0.6618 | 0.4896 |
VNB | 0.5801 | 0.6617 | 0.4896 |
GEM | 0.6060 | 0.6612 | 0.4896 |
PEM | 0.6307 | 0.6614 | 0.4896 |
GEF (EGFR+) | 0.6625 | 0.6686a | 0.4896 |
PAX (EGFR+) | 0.5934 | 0.6623 | 0.4896 |
Second-line chemotherapy | PFS 2 | PPS 2 | Terminal period (2 weeks) |
DOC | 0.5927 (on chemotherapy) 0.6559 (post chemotherapy) |
0.4275 | 0.0686 |
ERL | 0.6524 | 0.4275 | 0.0686 |
Discounting
In the base-case analysis both costs and outcomes are discounted at 3.5% per annum in line with NICE guidance. 140 Sensitivity analyses are reported for discount rates between 0% and 6%.
Time horizon
A lifetime perspective is taken in the model, which projects all costs, patient events and costs to a maximum of 10 years, at which time it is assumed all patients will have died.
Modelling assumptions
First-line chemotherapy regimens with the same primary agent but different PLAT therapy (A vs B) differ only in terms of treatment costs. Although meta-analyses84,85 found some minor differences in outcomes favouring CIS over CARB, and in AEs (more thrombocytopenia with CARB, and more nausea/vomiting and nephrotoxicity with CIS), on balance it was concluded that the evidence suggested only a limited net difference in patient benefit, unlikely to influence the results of any comparisons.
Results for population 1 (patients with squamous disease)
Deterministic analysis
Base case (British National Formulary prices)
Summary model results for the base-case analysis using BNF drug acquisition prices are shown in Tables 66 and 67 (costs and QALYs, respectively). For all primary chemotherapy agents, use of CARB is associated with slightly higher costs than use of CIS. Outcomes vary between regimens, between DOC (best) and VNB (worst). Figure 30 indicates that two CIS regimens lie on the efficiency frontier: PAX (3A) and DOC (1A), with a pair-wise ICER of £27,159 per QALY gained for 1A (DOC + CIS) compared with 3A (PAX + CIS). VNB is more expensive and less effective than PAX and is therefore dominated, whereas GEM is more expensive and less effective than DOC.
Regimen code | Drug acquisition (£) | Drug admin. (£) | AEs (£) | Supportive care (£) | Terminal care (£) | Total cost (£) | |
---|---|---|---|---|---|---|---|
1 | A | 4876 | 968 | 661 | 6542 | 3812 | 16,859 |
B | 5636 | 968 | 661 | 6542 | 3812 | 17,619 | |
2 | A | 4250 | 2966 | 738 | 5288 | 3829 | 17,070 |
B | 5067 | 2167 | 738 | 5288 | 3829 | 17,088 | |
3 | A | 3715 | 1387 | 690 | 5325 | 3833 | 14,950 |
B | 4471 | 1105 | 690 | 5325 | 3833 | 15,424 | |
4 | A | 3076 | 2465 | 896 | 5023 | 3841 | 15,302 |
B | 3836 | 2012 | 896 | 5023 | 3841 | 15,609 | |
5 | A | 3180 | 2465 | 896 | 5023 | 3841 | 15,405 |
B | 3939 | 2012 | 896 | 5023 | 3841 | 15,712 |
Regimen code | Time in PFS 1 | Time after PD 1 | OS | QALYs in PFS 1 | QALYs after PD 1 | Total QALYs | |
---|---|---|---|---|---|---|---|
1 | A/B | 0.4338 | 0.7261 | 1.1599 | 0.2729 | 0.3288 | 0.6017 |
2 | A/B | 0.5341 | 0.5007 | 1.0348 | 0.3423 | 0.2267 | 0.5690 |
3 | A/B | 0.4439 | 0.5517 | 0.9956 | 0.2815 | 0.2498 | 0.5314 |
4 | A/B | 0.4392 | 0.5121 | 0.9512 | 0.2760 | 0.2319 | 0.5079 |
5 | A/B | 0.4392 | 0.5121 | 0.9512 | 0.2760 | 0.2319 | 0.5079 |
Alternative scenario (electronic market information tool prices)
Applying mean NHS negotiated prices in place of published list prices leads to substantial reductions in acquisition costs, but has no other effects on costs or outcomes. The revised cost estimates are shown in Table 68.
Regimen code | Drug acquisition (£) | Total cost (£) | |
---|---|---|---|
1 | A | 3268 | 15,251 |
B | 3302 | 15,285 | |
2 | A | 1590 | 14,410 |
B | 1627 | 13,648 | |
3 | A | 1359 | 12,594 |
B | 1394 | 12,346 | |
4 | A | 2609 | 14,835 |
B | 2644 | 14,416 | |
5 | A | 2828 | 15,054 |
B | 2863 | 14,635 |
The corresponding efficiency frontier (Figure 31) now features three regimens, two using CARB as the PLAT component. The estimated ICER for GEM + CARB compared with PAX + CARB is £34,605 per QALY gained, and for DOC + CIS compared with GEM + CARB is £49,065 per QALY gained. However, there is minimal difference between the PLAT compounds when used in combination with DOC. VNB remains dominated because of its inferior outcomes. The general change to preferring CARB doublets in this scenario arises because with heavy price discounting the importance of NHS administration costs to the overall cost is increased relative to acquisition costs, so that the less demanding CARB regimens which are more likely to be deliverable in a day-case setting incur lower delivery costs.
Sensitivity analysis
A full univariate sensitivity analysis was carried out to explore the relative importance of uncertainty in each parameter to the estimated ICER of DOC + CIS compared with PAX + CIS using the base-case scenario, and varying parameter values across the 95% CI. The main exceptions are the proportions of patients receiving chemotherapy in a day-case setting, and the proportions of second-line patients receiving ERL (rather than DOC). In these cases an absolute variation of ± 10% was applied, equivalent to a relative variation of about 20%. The only parameter where a 10% relative variation was applied is the proportion of PFS events which are fatal; as previously mentioned, this parameter is not amenable to much wider variation if it is to avoid taking both invalid lower and upper values.
The results for 20 variables which most affect the estimated ICER are shown in Figure 32.
The estimated (correlated) HRs for PFS and OS for the comparator versus PAX are clearly the dominant variables in the model. Next most influential are the estimated utility parameters for progressive and stable disease in the Nafees et al. utility model. 116 Thereafter, uncertainty in type of second-line therapy, and in the mode and cost of chemotherapy administration are influential.
Probabilistic sensitivity analysis
A PSA was performed for the base-case scenario including all parameters for which uncertainty could be characterised statistically (details are shown in Appendices 30 and 31). CIS was assumed as the PLAT component in all regimens, and i.v. VNB was preferred to the oral formulation. The PSA was repeated for the alternative scenario (eMIT prices), using CARB throughout. The summary results of both scenarios for the four treatments are shown in Table 69, and the cost-effectiveness acceptability plots are displayed in Figures 33 and 34. The PSA repeated the favourable result for GEM suggested in the deterministic analysis when eMIT drug prices are assumed, indicating that only VNB doublets do not lie on the efficiency frontier.
Regimen | PSA result | Base-case scenario | Alternative scenario | ||
---|---|---|---|---|---|
Mean | Incremental | Mean | Incremental | ||
DOC | Total cost (£) | 17,112 | 1796 | 15,244 | 2877 |
Total QALYs | 0.6017 | +0.0704 | 0.6017 | +0.0704 | |
GEM | Total cost (£) | 17,572 | 2257 | 13,713 | 1347 |
Total QALYs | 0.5691 | +0.0378 | 0.5691 | +0.0378 | |
PAX | Total cost (£) | 15,315 | 0 | 12,367 | 0 |
Total QALYs | 0.5313 | 0 | 0.5313 | 0 | |
VNB | Total cost (£) | 15,619 | 304 | 14,666 | 2299 |
Total QALYs | 0.5103 | −0.0211 | 0.5103 | −0.0211 | |
ICER | £25,533/QALY DOC vs PAX | £35,664/QALY GEM vs PAX £46,939/QALY DOC vs GEM |
Summary of results for population 1 (patients with squamous disease)
-
In both deterministic and probabilistic analyses for both the base-case and alternative pricing scenarios, VNB doublets yield the least patient benefit (as measured by expected discounted QALYs), and are not the least expensive option. As a result, VNB cannot be considered to provide either optimal effective or cost-effective chemotherapy treatment.
-
PAX doublets are consistently minimum cost options and therefore represent the initial ‘good value’ treatment, only to be supplanted if an alternative option yields greater benefit at an acceptable ‘willingness-to-pay’ (WTP) threshold.
-
The choice of preferred alternative main agent to PAX generally favours DOC over GEM as its greater effectiveness appears to outweigh the additional acquisition cost, although both lie on the efficiency frontier.
Three more general observations may also be made on the basis of these results.
-
The difference in incremental QALYs gained between the treatments reflect only very marginal differences in benefit.
-
The sensitivity of the results to the general level of drug prices especially relating to the choice of PLAT compound indicates that in a competitive market, which has driven most generic prices down to very low levels, the price of drugs becomes less important than differences in the cost of drug administration and in the relative cost of AEs. Thus, achieving increased efficiency under these circumstances involves maximising the likelihood that patients can receive chemotherapy without recourse to inpatient admission.
-
The differences in estimated ICERs between the deterministic and probabilistic analyses is predominantly attributable to the fact that the greatest source of parameter uncertainty relates to estimated HRs which are subject to non-linear (logarithmic) distributions, leading to asymmetric cost-effectiveness results. Under these circumstances, the probabilistic results should be considered more reliable.
Results for population 2 (patients with non-squamous disease)
Deterministic analysis
Base case (British National Formulary prices)
The summary model results for the base-case analysis using BNF drug acquisition prices are shown in Tables 70 (costs) and 71 (costs and QALYs, respectively). For all primary chemotherapy agents, use of CARB is associated with slightly higher costs than use of CIS. Outcomes vary between regimens, between PEM (best) and VNB (worst). Figure 35 indicates that two CIS regimens lie on the efficiency frontier: PAX (3A) and PEM (6A), with a pair-wise ICER of £26,175 per QALY gained for 6A (PEM + CIS) compared with 3A (PAX + CIS). However, it is apparent that DOC + CIS lies very close to the frontier and should be considered of similar cost-effectiveness. VNB is more expensive and less effective than PAX and is therefore dominated, whereas GEM is less effective than DOC and with similar net incremental cost per patient.
Regimen code | Drug acquisition (£) | Drug admin. (£) | AEs (£) | Supportive care (£) | Terminal care (£) | Total cost (£) | |
---|---|---|---|---|---|---|---|
1 | A | 4876 | 968 | 661 | 6548 | 3812 | 16,865 |
B | 5637 | 968 | 661 | 6548 | 3812 | 17,626 | |
2 | A | 4251 | 2966 | 738 | 5281 | 3829 | 17,065 |
B | 5067 | 2167 | 738 | 5281 | 3829 | 17,083 | |
3 | A | 3715 | 1387 | 690 | 5325 | 3833 | 14,950 |
B | 4471 | 1105 | 690 | 5325 | 3833 | 15,424 | |
4 | A | 3076 | 2465 | 896 | 5028 | 3841 | 15,306 |
B | 3836 | 2012 | 896 | 5028 | 3841 | 15,613 | |
5 | A | 3179 | 2465 | 896 | 5028 | 3841 | 15,409 |
B | 3939 | 2012 | 896 | 5028 | 3841 | 15,716 | |
6 | A | 7434 | 1522 | 505 | 6980 | 3790 | 20,231 |
B | 8297 | 1522 | 505 | 6980 | 3790 | 21,094 |
Regimen code | Time in PFS 1 | Time after PD 1 | OS | QALYs in PFS 1 | QALYs after PD 1 | Total QALYs | |
---|---|---|---|---|---|---|---|
1 | A/B | 0.4341 | 0.7268 | 1.1609 | 0.2730 | 0.3291 | 0.6022 |
2 | A/B | 0.5348 | 0.4995 | 1.0343 | 0.3427 | 0.2262 | 0.5689 |
3 | A/B | 0.4439 | 0.5517 | 0.9956 | 0.2815 | 0.2498 | 0.5314 |
4 | A/B | 0.4389 | 0.5128 | 0.9517 | 0.2759 | 0.2322 | 0.5081 |
5 | A/B | 0.4389 | 0.5128 | 0.9517 | 0.2759 | 0.2322 | 0.5081 |
6 | A/B | 0.6496 | 0.6777 | 1.3274 | 0.4231 | 0.3100 | 0.7331 |
Alternative scenario (electronic market information tool prices)
Applying mean NHS negotiated prices in place of published list prices, leads to substantial reductions in acquisition costs, but has no other effects on costs or outcomes. The revised cost estimates are shown in Table 72.
Regimen code | Drug acquisition (£) | Total cost (£) | |
---|---|---|---|
1 | A | 3268 | 15,257 |
B | 3302 | 15,292 | |
2 | A | 1590 | 14,405 |
B | 1627 | 13,642 | |
3 | A | 1359 | 12,594 |
B | 1394 | 12,346 | |
4 | A | 2609 | 14,839 |
B | 2644 | 14,420 | |
5 | A | 2828 | 15,058 |
B | 2862 | 14,639 | |
6 | A | 7022 | 19,819 |
B | 7061 | 19,857 |
The corresponding efficiency frontier (Figure 36) now features three regimens: PAX + CARB, GEM + CARB and PEM + CIS. The estimated ICER for GEM + CARB compared with PAX + CARB is £34,542 per QALY gained and for PEM + CIS compared with GEM + CARB is £37,608 per QALY gained. VNB remains dominated because of its inferior outcomes. As in the squamous disease population results, the general change to preferring CARB doublets in this scenario arises because with heavy price discounting the importance of NHS administration costs to the overall cost is increased relative to acquisition costs.
Sensitivity analysis
A full univariate sensitivity analysis was carried out to explore the relative importance of uncertainty in each model parameter to the estimated ICER of PEM + CIS compared with PAX + CIS using the base-case scenario, and varying most parameter values across the 95% CI. In other variables a notional absolute range of ± 10% of the estimated value was used. The results for 20 variables which most affect the estimated ICER are shown in Figure 37.
The estimated (correlated) HRs for PFS and OS for PEM compared with PAX are clearly the dominant variables in the model. Next most influential are the estimated utility parameters for progressive and stable disease in the Nafees et al. 116 utility model. Other parameters make only minor contributions to uncertainty in the estimated ICER.
Probabilistic sensitivity analysis
A PSA was performed for the base-case scenario including all parameters for which uncertainty could be characterised statistically. CIS was assumed as the PLAT component in all regimens, and i.v. VNB was preferred to the oral formulation. The PSA was repeated for the alternative scenario (eMIT prices), using CARB throughout, except for PEM.
The summary results of both scenarios for the four treatments are shown in Table 73 and the cost-effectiveness acceptability plots are displayed in Figures 38 and 39.
Regimen | PSA result | Base-case scenario | Alternative scenario | ||
---|---|---|---|---|---|
Mean | Incremental | Mean | Incremental | ||
DOC | Total cost (£) | 17,153 | 1838 | 15,285 | 2918 |
Total QALYs | 0.6044 | +0.0731 | 0.6044 | +0.0731 | |
GEM | Total cost (£) | 17,561 | 2246 | 13,702 | 1335 |
Total QALYs | 0.5687 | +0.0373 | 0.5687 | +0.0373 | |
PAX | Total cost (£) | 15,315 | 0 | 12,367 | 0 |
Total QALYs | 0.5313 | 0 | 0.5313 | 0 | |
VNB | Total cost (£) | 15,617 | 302 | 14,664 | 2297 |
Total QALYs | 0.5101 | −0.0212 | 0.5101 | −0.0212 | |
PEM | Total cost (£) | 21,284 | 5968 | 20,803 | 8436 |
Total QALYs | 0.7137 | +0.1824 | 0.7137 | +0.1824 | |
ICER | £25,155/QALY DOC vs PAX £37,779/QALY PEM vs DOC |
£35,776/QALY GEM vs PAX £44,293/QALY DOC vs GEM £50,470/QALY PEM vs DOC |
Summary of results for population 2 (patients with non-squamous disease)
The addition of a PEM doublet to the four third-generation chemotherapy agents changes the relationship between the regimens, owing to the clear outcome advantage of PEM therapy in terms of improved expected survival for patients with non-squamous disease. However, the high price of branded PEM compared with the other drugs (in most cases available generically) means that PEM is only preferred on cost-effectiveness grounds if the WTP threshold is set > £37,000 per QALY (or £50,000 per QALY if sampled NHS contract prices are assumed). This means that PAX remains a viable treatment (and possibly GEM and DOC). However, VNB is clearly not cost-effective in either scenario.
Results for population 3 (epidermal growth factor receptor mutation positive)
Deterministic analysis
Base case (British National Formulary prices)
The summary model results for the base-case analysis using BNF drug acquisition prices are shown in Tables 74 and 75 (costs and QALYs, respectively). For both primary chemotherapy agents (DOC and PAX), use of CARB is associated with slightly higher costs than use of CIS. Outcomes vary between regimens: in terms of expected survival chemotherapy appears to have a small advantage over GEF (about 2 weeks) but GEF provides a modest improvement in expected QALYs compared with chemotherapy (0.0786), owing to the extended period prior to disease progression. The slightly higher cost per patient and poorer outcomes of DOC regimens compared with PAX excludes them from consideration for cost-effectiveness. The estimated deterministic ICER for GEF compared with PAX + CIS is £57,440 per QALY gained.
Regimen code | Drug acquisition (£) | Drug admin. (£) | AEs (£) | Supportive care (£) | Terminal care (£) | Total cost (£) | |
---|---|---|---|---|---|---|---|
1 | A | 7459 | 1102 | 843 | 18,064 | 3531 | 30,998 |
B | 8327 | 1102 | 843 | 18,064 | 3531 | 29,812 | |
3 | A | 5566 | 1722 | 929 | 18,064 | 3552 | 34,325 |
B | 6434 | 1397 | 929 | 18,064 | 3531 | 31,866 | |
7 | 13,261 | 733 | 507 | 16,272 | 3531 | 30,355 |
Regimen code | Time in PFS 1 | Time after PD 1 | OS | QALYs in PFS 1 | QALYs after PD 1 | Total QALYs | |
---|---|---|---|---|---|---|---|
1 | A/B | 0.5264 | 2.2859 | 2.8123 | 0.3338 | 1.0833 | 1.4171 |
3 | A/B | 0.5264 | 2.2859 | 2.8123 | 0.3338 | 1.0833 | 1.4171 |
7 | 0.9406 | 1.8266 | 2.7673 | 0.6226 | 0.8731 | 1.4957 |
Base case (electronic market information tool prices)
Applying mean NHS negotiated prices in place of published list prices, leads to substantial reductions in acquisition costs for chemotherapy treatments, but has no other effects on costs or outcomes. The revised cost estimates are shown in Table 76. Deterministic estimated cost per patient for base-case analysis using eMIT prices.
Regimen code | Drug acquisition (£) | Total cost (£) | |
---|---|---|---|
1 | A | 5624 | 29,164 |
B | 5663 | 29,203 | |
3 | A | 2661 | 26,908 |
B | 2700 | 26,621 | |
7 | 12,302 | 33,366 |
Using PAX in combination with CARB now offers the minimum cost regimen for comparison with GEF. The estimated ICER for GEF compared with PAX + CARB is £85,848 per QALY gained
Sensitivity analysis
A full univariate sensitivity analysis was carried out to explore the relative importance of uncertainty in each model parameter to the estimated ICER of PAX + CIS compared with GEF using the base-case scenario with BNF prices, and varying most parameter values across the 95% CI. In other variables, a notional absolute range of ± 10% of the estimated value was used. The results for 20 variables which most affect the estimated ICER are shown in Figure 40. The most model parameters contributing most to uncertainty in the ICER are the utility model parameter values and unit costs of community health services.
The model assumes that AEs increase costs and result in disutilities for the whole duration of treatment. This is a reasonable approximation for chemotherapy, given for a limited number of cycles, but could be considered excessive for a continuous oral medication given throughout the progression-free period. To test the importance of this assumption to the estimated ICER, an additional sensitivity analysis was conducted in which the incidences of all GEF-related AEs were reduced by 50%. This resulted in a small reduction in incremental cost and a small increase in incremental QALYs gained, and reducing the base-case ICER (GEF vs PAX) from £57,440 to £53,401 per QALY gained.
Probabilistic sensitivity analysis
A PSA was performed for the base-case scenario including all parameters for which uncertainty could be characterised statistically. CIS was assumed as the PLAT component in chemotherapy regimens. The PSA was carried out using both BNF and eMIT prices (using CARB in place of CIS).
The summary results of both scenarios for the three treatments are shown in Table 77, and the cost-effectiveness acceptability plots are displayed in Figures 41 and 42.
Regimen | PSA result | Base case (BNF prices) | Base case (eMIT prices) | ||
---|---|---|---|---|---|
Mean | Incremental | Mean | Incremental | ||
DOC | Total cost (£) | 31,184 | 978 | 29,004 | 2149 |
Total QALYs | 1.4183 | 0 | 1.4183 | 0 | |
PAX | Total cost (£) | 30,205 | 0 | 26,855 | 0 |
Total QALYs | 1.4183 | 0 | 1.4183 | 0 | |
GEF | Total cost (£) | 34,485 | 4280 | 33,341 | 6485 |
Total QALYs | 1.4956 | +0.0773 | 1.4956 | +0.0773 | |
ICER | £55,364/QALY GEF vs PAX | £83,899/QALY GEF vs PAX |
Alternative scenario 1 (British National Formulary prices): pooling results from two trials
Summary model results for an alternative analysis based on pooling results from the two PAX trials (IPASS15,64 and NEJGSG63) using BNF drug acquisition prices are shown in Tables 78 and 79 (cost and QALYs, respectively). For PAX doublet therapy, use of CARB is associated with slightly higher costs than use of CIS. Expected survival with GEF appears to be a little better than PAX (1 month) and a corresponding benefit in terms of discounted QALYs (+0.1398). As a result, the estimated deterministic ICER for GEF vs PAX + CIS is reduced, compared with the base-case analysis, to £39,015 per QALY gained.
Regimen code | Drug acquisition (£) | Drug admin. (£) | AEs (£) | Supportive care (£) | Terminal care (£) | Total cost (£) | |
---|---|---|---|---|---|---|---|
3 | A | 5559 | 1718 | 926 | 15,406 | 3627 | 27,236 |
B | 6424 | 1393 | 926 | 15,406 | 3627 | 32,688 | |
7 | 13,193 | 734 | 493 | 14,656 | 3612 | 27,776 |
Regimen code | Time in PFS 1 | Time after PD 1 | OS | QALYs in PFS 1 | QALYs after PD 1 | Total QALYs | |
---|---|---|---|---|---|---|---|
3 | A/B | 0.5018 | 1.9287 | 2.4305 | 0.3175 | 0.9141 | 1.2316 |
7 | 0.8994 | 1.6228 | 2.5222 | 0.5957 | 0.7757 | 1.3714 |
Alternative scenario 1 (electronic market information tool prices): pooling results from two trials
Applying mean NHS negotiated prices in place of published list prices, leads to substantial reductions in acquisition costs for chemotherapy treatments, but has no other effects on costs or outcomes. Revised cost estimates are shown in Table 80.
Regimen code | Drug acquisition (£) | Total cost (£) | |
---|---|---|---|
3 | A | 2663 | 24,340 |
B | 2702 | 24,054 | |
7 | 12,234 | 31,729 |
Using PAX in combination with CARB now offers the minimum cost regimen for comparison with GEF. The estimated ICER for GEF compared with PAX + CARB is £54,911 per QALY gained.
Alternative scenario 2 (British National Formulary prices): WJTOG trial only
The summary model results for an alternative analysis based on only the WJTOG trial65 using BNF drug acquisition prices are shown in Tables 81 and 82 (QALYs). For DOC doublet therapy, use of CARB is associated with slightly higher costs than use of CIS. Expected survival with GEF appears to be better than PAX (3.5 months) and a corresponding benefit in terms of discounted QALYs (+0.2325). As a result, the estimated deterministic ICER for GEF vs DOC + CIS is reduced, compared with the base-case analysis, to £25,705 per QALY gained.
Regimen code | Drug acquisition (£) | Drug admin. (£) | AEs (£) | Supportive care (£) | Terminal care (£) | Total cost (£) | |
---|---|---|---|---|---|---|---|
1 | A | 7477 | 1113 | 852 | 17,627 | 3552 | 30,621 |
B | 8354 | 1113 | 852 | 17,627 | 3552 | 31,498 | |
7 | 13,458 | 733 | 534 | 18,401 | 3472 | 36,598 |
Regimen code | Time in PFS 1 | Time after PD 1 | OS | QALYs in PFS 1 | QALYs after PD 1 | Total QALYs | |
---|---|---|---|---|---|---|---|
1 | A/B | 0.6480 | 2.1635 | 2.8116 | 0.4116 | 1.0253 | 1.4369 |
7 | 1.0176 | 2.0837 | 3.1013 | 0.6736 | 0.9959 | 1.6694 |
Alternative scenario 2 (electronic market information tool prices): WJTOG trial only
Applying mean NHS negotiated prices in place of published list prices, leads to substantial reductions in acquisition costs for chemotherapy treatments, but has no other effects on costs or outcomes. Revised cost estimates are shown in Table 83.
Regimen code | Drug acquisition (£) | Total cost (£) | |
---|---|---|---|
1 | A | 5623 | 28,767 |
B | 5663 | 28,807 | |
7 | 12,499 | 35,639 |
Using DOC in combination with CARB remains slightly more expensive than DOC + CIS for comparison with GEF. The estimated ICER for GEF compared with PAX + CIS is £29,553 per QALY gained.
Probabilistic sensitivity analysis
Probabilistic sensitivity analyses for all scenarios and prices yielded ICERs which were closely similar to the corresponding deterministic ICERs (Table 84).
Scenario | Comparator | Analysis | Estimated ICER (£) |
---|---|---|---|
Base case (BNF prices) | PAX + CIS | Deterministic | 57,440 |
Probabilistic | 55,364 | ||
Base case (eMIT prices) | PAX + CARB | Deterministic | 85,849 |
Probabilistic | 83,899 | ||
A1 (BNF prices) | PAX + CIS | Deterministic | 39,015 |
Probabilistic | 37,749 | ||
A1 (eMIT prices) | PAX + CARB | Deterministic | 54,911 |
Probabilistic | 55,605 | ||
A2 (BNF prices) | DOC + CIS | Deterministic | 25,705 |
Probabilistic | 25,841 | ||
A2 (eMIT prices) | DOC + CIS | Deterministic | 29,553 |
Probabilistic | 30,438 |
Summary of results for population 3 (epidermal growth factor receptor mutation positive)
The base-case analyses for GEF compared with the two chemotherapy doublets for which evidence is available show poor cost-effectiveness for GEF. Results are improved somewhat by disaggregating the three trials, but even then cost-effective ICERs (< £30,000 per QALY gained) are only obtained for the second alternative scenario based on the smallest RCT comparing GEF with the DOC + CIS doublet.
Discussion
Summary of key results
Decision analysis results for population 1 (patients with squamous disease) and population 2 (patients with non-squamous disease) consistently show VNB to be the least efficacious of the four third-generation chemotherapy agents. Although the HRs of the four treatments estimated at 12 months after randomisation appear similar, differences in long-term modelled trends, especially for OS, suggest that estimated lifetime OS is likely to be worse for VNB than for the regimens involving DOC, PAX or GEM. Moreover, VNB is consistently more expensive than the PAX options regardless of the price source used, so that VNB regimens are always dominated by other options. DOC consistently outperforms the other third-generation chemotherapy drugs primarily because of its superior long-term trend for OS. Although its long-term standardised disease risk trend is poor, this is outweighed by a favourable HR for OS at 12 months. When the more realistic NHS contract prices are considered, GEM-based regimens come into consideration alongside PAX and DOC, and CARB doublets appear preferable as a result of the less demanding administration requirements so that fewer patients need to be admitted overnight.
For population 2 (patients with non-squamous disease), PEM + CIS is clearly superior to all the third-generation chemotherapy regimens in terms of outcomes. When BNF list prices are used, PEM + CIS appears to be the most cost-effective treatment. However, when NHS-discounted contract prices for generic third-generation drugs are considered, the situation is less clear-cut, with the ICER for PEM + CIS compared with DOC exceeding £40,000 per QALY gained when assessed probabilistically.
In population 3 (patients who are EGFR M+), the trial evidence indicates that these patients have a far better prognosis than other patients with NSCLC when treated with third-generation drugs. This finding prevented the use in meta-analysis of most of the published clinical trials on patients with mixed NSCLC. Only three trials of GEF compared with either PAX or DOC doublets were found to be suitable. There was no evidence in similar populations to link PEM + CIS to the evidence network, so no economic comparison is currently possible between GEF and PEM; despite this being the most clinically relevant candidate comparator for GEF. The cost-effectiveness results are not generally favourable for GEF, which generates base-case ICERs in excess of £50,000 per QALY gained, and achieves ICERs < £30,000 per QALY only when clinical evidence is restricted to the smallest of the three RCTs.
Generalisability of results
The clinical effectiveness evidence is drawn from a comprehensive international review of RCTs undertaken to assess active systemic first-line treatments for NSCLC patients and is, therefore, of general applicability. The perspective of the economic assessment is that of the UK NHS, and draws on UK unit costs, clinical practices and guidelines to furnish model parameters. As a result, conclusions on relative cost-effectiveness may vary in other national environments.
Strengths and limitations of analysis
A novel approach to modelling trial outcomes was developed and implemented with the objective of capturing contrasting patterns of patient outcomes over time between the various treatments available. It is frequently observed that the four third-generation chemotherapy agents are considered ‘clinically equivalent’, but this assessment may merely mean that estimated HRs do not differ according to conventional standards. When probabilistic analysis is undertaken covering uncertainty in multiple parameters important differences in cost-effectiveness may be revealed, notwithstanding the absence of individual parameter differences normally considered significant (in this case HRs for OS and PFS). However, as there are important differences among the drugs in their mode of action, it should not be surprising that these lead to more subtle but important differences in long-term prognosis. The analysis of PFS and OS profiles for each drug pooled across all available trials indicated this to be the case, as particularly exemplified by a comparison of PEM with the third-generation drugs. Of course, this requires pooling individual trial arms and thus ‘breaks randomisation’. To counter this problem, standardised profiles were developed and then conventional HRs preserving randomisation were applied to adjust the unique profiles to represent faithfully the expected PFS and OS outcomes of each regimen. A particular strength of this method is that it avoids recourse to modelling time trends on the basis of selecting from a small number of conventional statistical parametric functions, without any obvious or explicit supporting logic.
The analysis undertaken on population 3 (patients who are EGFR M+) could not be applied to include PEM + CIS as was originally intended, owing to the lack of evidence of PEM efficacy in patients with EGFR-activating mutations. With only three modestly sized trials available and two different comparators, it was not possible to carry out any sort of indirect comparison. Therefore, the assessment is based solely on using the trial data directly – pooling all three trials for the base case and assuming equivalent effectiveness in the comparators. The results obtained are necessarily tentative, rest on limited data and are subject to question.
In particular, authors of all three trials have drawn attention to the high levels of crossover of patients randomised to chemotherapy choosing to switch to EGFR-TKI therapy on disease progression, and this is considered sufficient to explain why in none of these trials has any difference in OS been observed. However, the authors of a recent meta-analysis141 have concluded that ‘the lack of an OS benefit for initial GEF in these studies – in the overall population or even exclusively in patients with EGFR mutations – is a robust finding of this meta-analysis and apparent across all four studies’. To consider the strength of the argument for OS benefit from use of GEF obscured by high levels of crossover, a simple comparison was made of OS HRs and the proportion of chemotherapy patients switching to GEF treatment on disease progression, intended to detect a trend away from a HR of 1.0 in favour of GEF as the extent of crossover diminishes. The results (Figure 43) show no evidence of such a trend and, therefore, in the absence of any evidence to the contrary, the analysis shown here is based on unadjusted trial data without any alteration for crossover.
There is clearly a need for further clinical trials to be undertaken in patients with EGFR-activating mutations, which should include PEM + CIS as an important potential comparator to GEF, and should be designed to resolve the issue of crossover confounding.
Chapter 5 Assessment of factors relevant to the NHS and other parties
This review highlights that histology and EGFR mutation status are important clinical factors in determining optimal chemotherapy regimens for patients and preventing the use of ineffective treatments. The recommended use of PEM and GEF require improvements in the standardisation of histology and EGFR testing within the UK. Histocytology and genetic testing need to become commonplace, standardised and routinely carried out within clinical practice in order for patients to receive optimal care. Testing for EGFR mutation status is crucial for determining which patients are eligible for EGFR-TK inhibitor drug treatment. All centres can now access EGFR mutation testing and genetic testing is becoming commonplace in the UK. However, different organisations differ in their approach to genetic testing and uptake is variable across regions, but most centres will send samples of all adenocarcinomas of lung origin to be tested for EGFR status.
There is a relatively large number of chemotherapy drugs for the first-line treatment of NSCLC that are currently being tested within Phase III trials or are filed for approval and these are shown in Table 85. Trials of PLAT resistance, chemotherapy in elderly patients with NSCLC and assessment of any added value of maintenance chemotherapy to first-line chemotherapy; these are all current areas of research. The proportion of squamous patients is currently decreasing in the UK and, although there are no chemotherapy agents on the immediate horizon, this is an obvious research area to explore having demonstrated different responses in patients with non-squamous NSCLC with different chemotherapy drugs.
Drug | Possible indication | Regulatory prediction | Clinical trials | Company |
---|---|---|---|---|
Afatinib (Tomtovok®) Oral | First-line monotherapy for patients with EGFR+ mutations | EU filing expected 2012: market 2013? (Confidential) |
NCT01121393 (LUX-Lung 6): single agent afatinib vs GEM + CIS for lung adenocarcinoma with tumour harbouring an EGFR-activating mutation NCT00949650 (LUX-Lung 3): single agent afatinib vs PEM + CIS for lung adenocarcinoma with tumour harbouring an EGFR-activating mutation |
Boehringer Ingelheim |
Cediranib (Recentin™) Oral | First-line combination therapy for advanced/metastatic disease | 2016 (Confidential) | NCT00795340: cediranib plus PAX/CARB vs PAX/CARB for the treatment of advanced or metastatic NSCLC | AstraZeneca |
Cetuximab (Erbitux®) i.v. | First-line combination therapy in patients with high EGFR expression | Filed in EU March 2011. Withdrawn September 2012142 Previously filed in 2008, but CHMP issued negative opinion |
NCT00112294: taxane/CARB + centuximab vs taxane/CARB as first-line treatment for patients with advanced/metastatic NSCLC NCT00148798 (FLEX): CIS/VNB + centuximab vs CIS/VNB as first-line treatment for patients with EGFR-expressing advanced NSCLC |
Merck Serono |
Crizotinib (Xalkori®) Oral | First-line locally advanced or metastatic; non-squamous cell NSCLC positive for ALK fusion gene | Likely to be filed shortly for second-/third-line treatment (already filed in US). Phase III first-line treatment study started January 2011 | NCT01154140 (PROFILE 1014) crizotinib vs standard chemotherapy (PEM + CIS or CARB) in patients with non-squamous carcinoma of the lung harbouring a translocation or inversion event involving the ALK gene locus | Pfizer |
ERL (Tarceva®) Oral | First-line monotherapy in EGFR mutation-positive disease | Filed in EU June 2010. 2011 – positive opinion | NCT01342965: ERL vs GEM/CIS in patients with mutations in the tyrosine kinase domain of the EGFR NCT00446225 (EUTRAC): ERL vs chemotherapy (CARB + GEM or doxetaxol/CIS) in patients with advanced NSCLC with mutations in the tyrosine kinase domain of the EGFR. This study was stopped early as it had met its primary end point |
Genentech, Roche |
Iniparib i.v. | First-line combination therapy in advanced (stage IV) squamous cell NSCLC | EU approval possibly third quarter 2012 (Confidential) |
NCT01082549 (ECLIPSE): GEM/CARB with or without iniparib in patients with previously untreated stage IV squamous NSCLC | Sanofi-aventis |
Ipilimumab (Yervoy®) i.v. | First-line combination therapy in squamous cell, stage IV or recurrent NSCLC | Unknown. Pivotal study due to complete 2015 | NCT01285609: ipilimumab plus PAX/CARB vs PAX/CARB in subjects with squamous only, stage IV/recurrent NSCLC | Bristol-Myers Squibb |
Motesanib Oral | First-line combination therapy in non-squamous or adenocarcinoma NSCLC | Unknown. Primary end point, OS was not achieved in MONET 1 | NCT00460317 (MONET 1): motesanib + PAX/CARB vs chemotherapy alone in patients with advanced non-squamous NSCLC and in patients with adenocarcinoma histology | Takeda |
Chapter 6 Conclusions
This comprehensive review is unique to the field of NSCLC research in that it compares all of the regimens currently licensed in Europe and approved by NICE for the first-line systemic treatment of patients with advanced NSCLC and is important because the future of NSCLC treatments has reached a crossroads. In summary, this review provides a basis from which to move forward, despite being limited by the published clinical effectiveness and cost-effectiveness evidence available. This review may assist clinicians to make decisions regarding the treatment of patients with advanced NSCLC as new evidence related to the important subgroups of patients becomes available in published form.
Implications for practice
The treatment of patients with NSCLC is complex. In contrast to previous research, recent clinical effectiveness evidence from RCTs demonstrates that patient health outcomes depend not only on the treatment received, but also on the characteristics of the patient population participating in the trial and of the cancer subtypes. However, in addition to the clinical evidence available, clinicians need to take the specific needs and wishes of their patients into consideration when making treatment decisions. Closer examination of clinical effectiveness and cost-effectiveness data means that we have been able to provide a comprehensive framework of information which clinicians can refer to as they attempt to balance patient factors, available treatments, treatment costs and AEs in their daily decision-making.
The results in this report relate solely to first-line systemic therapy for patients with advanced NSCLC. No inference should be drawn from them about chemotherapy in any other context. This includes adjuvant therapy, combination therapy (with radiotherapy or surgery), and second-line and maintenance therapy.
Specific treatment options
Until recently, patients with NSCLC were treated as a homogenous group; the results of previous systematic reviews concluded that, in patients with NSCLC, there were no statistically significant survival differences between DOC, PAX, GEM and VNB. This is no longer the case and increasingly trials are distinguishing between three populations of patients: patients with squamous disease, patients with non-squamous disease and patients who are EGFR M+. Our report discusses the available clinical effectiveness and cost-effectiveness evidence for agents currently approved by NICE for use in England for each of these three patient groups in turn.
However, one finding of our review and economic modelling work that applies equally to all of these patient populations is that VNB (oral or i.v.) is less effective and more costly than at least one of the other options (DOC, PAX and GEM) and, therefore, is not shown to be cost-effective under any circumstances. Clearly, this finding will be of concern to those clinicians who currently favour the use of this treatment.
Given the recent changes in chemotherapy costs (that is the decrease in costs as drugs come off patent) other factors begin to enter into the decision-making process. One important issue identified by this review is the effect these changes may have on the choice of use of the PLAT component of chemotherapy doublet regimens. The use of CIS is more likely than CARB to require an overnight stay in hospital, and with reducing drug costs, additional administration costs begin to impact significantly on the overall cost-effectiveness of the various treatment options, and may potentially lead to greater use of CARB administered in a day-case setting.
Patients with squamous disease
Our report shows that for patients with squamous disease, there is no statistically significant difference in terms of OS between DOC, PAX, GEM and VNB. However, our analyses demonstrate that there are slight differences between these treatments in terms of clinical effectiveness and when these differences are modelled over the longer term (> 12 months) and the costs of the treatments are taken into consideration, then differences in cost-effectiveness begin to appear. For this group of patients, PAX is shown to be the preferred option when the WTP threshold is low. As the WTP threshold is increased GEM and DOC can be considered cost-effective treatments, so that at high WTP thresholds DOC becomes the preferred option.
Patients with non-squamous disease
In terms of OS, the clinical evidence shows that PEM is the preferred option for this group of patients, showing a statistically significant gain in OS over all of the third-generation doublet regimens. For cost-effectiveness, a similar pattern of ranking applies as was found for treatment of patients with squamous disease (PAX → GEM → DOC); however, with PEM added as the final ‘most effective but most costly’ option a high WTP threshold (up to £50,000 per QALY) is required in order for PEM to be considered acceptable. If and when the acquisition cost of PEM is reduced, the case for its wider use will be strengthened.
Patients who are epidermal growth factor receptor mutation positive
Patients with EGFR M+ status are a small subgroup of patients with NSCLC who have predominantly non-squamous disease. Trial evidence indicates that this patient population has a far better clinical prognosis than other patients with NSCLC. However, it is difficult to identify optimal treatments for this group of patients as the available trial evidence indicates that there is a PFS benefit for patients associated with GEF, but that there is no statistically significant OS benefit associated with GEF compared with DOC or PAX. Decision analysis based on the three GEF trials15,63–65 currently published suggests high ICERs when comparing GEF to third-generation chemotherapy doublets (PAX and DOC), greater than would normally be considered acceptable in the UK. The absence of any direct evidence of PEM effectiveness in the small EGFR M+ subgroup currently precludes any comparison between GEF and PEM.
Research recommendations
Future trials of first-line treatments for patients with NSCLC will need to take into consideration many more factors than has historically been the case. NSCLC is no longer considered as a single disease entity and the design of future lung cancer trials needs to reflect the influence of factors such as histology, genetics and any new prognostic biomarkers that are currently being identified. In addition, trials will need to be adequately powered so as to be able to test for statistically significant clinical effectiveness differences within patient populations.
Current standard treatment for patients with advanced and metastatic NSCLC is first-line chemotherapy; second- and third-line treatments are also available for those patients who are fit enough. As more patients become eligible for second- and third-line treatments, more consideration has to be given to the design of trials and how OS can be appropriately measured. Flexibility is required to design trials which not only permit patients to cross over to other treatments but also to design trials where the survival data collected can be meaningfully interpreted. For example, this may lead to more trials being designed with designated sequencing of treatments. It is acknowledged that such trials are unlikely to be funded by the pharmaceutical industry where demonstration of PFS is the accepted marker for obtaining market authorisation.
However, there are other gaps in our knowledge about current treatments and outcomes for patients with NSCLC. The results in this report are based entirely on the analysis of published data from Phase II and Phase III clinical trials. It is well known that patients in such trials are not necessarily representative of patients seen in UK clinical practice. New initiatives to collect data on UK patients and the treatment they receive are now in place through the emergence of the National Cancer Intelligence Network. 87 The National SACT data set became operational on the 1 April 2012 and will provide this detailed information. It will also be feasible to include health economic data into such future analyses. We would strongly endorse the development of initiatives of this kind in the effort to provide data that can more accurately define the true cost–benefit ratio of treatment interventions in this patient population.
A major gap in the literature that has been identified by this review is the lack of published HRQoL data in this patient population in a clinical trial setting. Results of recent research have shown that it is possible to collect reliable HRQoL data in cancer patients during treatment. As clinicians consider the AE profiles of treatments and subsequent effects on HRQoL in their decision-making, all trials should include mechanisms to elicit and report good-quality HRQoL data reflecting patients' experiences of their treatment during trials.
Concluding remarks
The completion of this review has taken a significant length of time and during that period there has been explicit acknowledgement in the published literature of the important differences in the characteristics of patients who previously were identified as having NSCLC. It is anticipated that no further RCTs will be carried out involving patients with NSCLC as a homogeneous group, but that consideration of the important patient subgroups will take precedence and allow for the development of more specialised and targeted treatments which, in turn, will require RCTs of increasingly sophisticated design.
This report offers clinicians informed evidence about all aspects of currently available treatments for patients with lung cancer. Clearly, health-care professionals make daily decisions about what is best for their patients. For instance, individual side-effect profiles may mean that a particular drug is selected that might be assessed as less cost-effective but better suit a particular patient's preference (e.g. the use of alternative drugs to DOC where high-dose steroids, hair loss or neurological side effects need to be avoided). In this context, the short OS gain and significant symptoms experienced by patients with advanced NSCLC need to be considered. However, health-care professionals are also tasked with making difficult decisions with populations in mind and it is hoped that this report will provide up-to-date information that will support clinicians in their discussion with patients regarding the benefits of the various treatment options.
Acknowledgements
Expert Advisory Panel and peer reviewers: David Baldwin, Paul Beckett, Jeremy Braybrooke, Andrew Champion, Jesme Fox, John Green, Matthew Hatton, Noelle O'Rourke, Michael Peake and Robert Rintoul.
Thanks to Juliet Hockenhull for database expertise and Janet Atkinson for administrative support.
Contribution of authors
Adrian Bagust Economic analysis and modelling.
Angela Boland Economic analysis and systematic review of economic studies.
Michaela Blundell Assessment of statistics and data analysis including meta-analyses.
Tamara Brown Project management, data management, systematic review of clinical effectiveness data and preparation of report.
Rumona Dickson Input into all aspects of the clinical component of the review.
Yenal Dundar Development of search strategies, data checking, quality assessment and review of draft report.
Janette Greenhalgh Data extraction and quality assessment of clinical trials.
Monica Lai Economic modelling.
Gerlinde Pilkington Data extraction and quality assessment of clinical trials and preparation of report.
James Oyee Assessment of statistics and data analysis including meta-analyses.
Carlos Martin Saborido Data extraction of clinical and economic studies.
Catrin Tudur Smith Statistical expertise.
All contributors took part in the editing and production of this report.
Disclaimer
This report presents independent research funded by the National Institute for Health Research (NIHR). The views and opinions expressed by authors in this publication are those of the authors and do not necessarily reflect those of the NHS, the NIHR, NETSCC, the HTA programme or the Department of Health.
References
- Cancer Research UK . CancerStats: Lung Cancer and Smoking – Key Facts n.d. http://info.cancerresearchuk.org/cancerstats/types/lung/ (accessed January 2011).
- Mackenbach J, Huisman M, Andersen O, Bopp M, Borgan J, Borrell C, et al. Inequalities in lung cancer mortality by the educational level in 10 European populations. Eur J Cancer 2004;40:126-35. http://dx.doi.org/10.1016/j.ejca.2003.10.018.
- Pugh H, Power C, Goldblatt P, Arber S. Women's lung cancer mortality, socio-economic status and changing smoking patterns. Soc Sci Med 1991;32:1105-10. http://dx.doi.org/10.1016/0277-9536(91)90086-R.
- Pollock AM, Vickers N. Breast, lung and colorectal cancer incidence and survival in South Thames Region, 1987–1992: the effect of social deprivation. J Pub Health Med 1997;19:288-94. http://dx.doi.org/10.1093/oxfordjournals.pubmed.a024632.
- Cancer Research UK . CancerHelp UK, Statistics and Outlook for Lung Cancer 2011. www.cancerhelp.org.uk/type/lung-cancer/treatment/statistics-and-outlook-for-lung-cancer#outcome (accessed February 2011).
- Cancer Research UK . CancerHelp UK, Lung Cancer Risks and Causes n.d. www.cancerhelp.org.uk/type/lung-cancer/about/lung-cancer-risks-and-causes (accessed February 2011).
- National Institute for Health and Care Excellence (NICE) . The Diagnosis and Treatment of Lung Cancer CG 121 2011. www.nice.org.uk/nicemedia/live/13465/54199/54199.pdf (accessed February 2011).
- The NHS Information Centre . National Lung Cancer Audit Information Sheet (audit Period 2008) 2009. www.ic.nhs.uk/canceraudits/lung (accessed July 2012).
- TNM classification of malignant tumours. London: Wiley-Blackwell; 2009.
- Cancer Research UK . CancerHelp UK, Performance Status n.d. www.cancerhelp.org.uk/about-cancer/cancer-questions/performance-status (accessed February 2011).
- Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol 1982;5:649-55. http://dx.doi.org/10.1097/00000421-198212000-00014.
- The Information Centre for Health and Social Care . National Lung Cancer Audit: Key Findings about the Quality of Care for People With Lung Cancer in England and Wales. Report for the Audit Period 2006. 2007. www.ic.nhs.uk/services/national-clinical-audit-support-programme-ncasp/audit-reports/lung-cancer (accessed July 2012).
- National Institute for Health and Care Excellence (NICE) . Final Appraisal Determination: Pemetrexed for First-Line Treatment of Non-Small Cell Lung Cancer. 2009. www.nice.org.uk/nicemedia/pdf/FADPemetrexedLungCancer.pdf (accessed July 2012).
- The NHS Information Centre . National Lung Cancer Audit, Report for the Audit Period 2008 2009. www.royalsurrey.nhs.uk/Default.aspx?DN=c29e1a74-6b75-4201-9f9d-9745981b7d7a (accessed July 2012).
- Mok TS, Wu Y-L, Thongprasert S, Yang C-H, Chu D-T, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Eng J Med 2009;361:947-57. http://dx.doi.org/10.1056/NEJMoa0810699.
- National Institute for Health and Care Excellence (NICE) . Final Appraisal Determination: Gefitinib for the First-Line Treatment of Locally Advanced or Metastatic Non-Small-Cell Lung Cancer 2010. http://guidance.nice.org.uk/TA/Wave10/21/FAD/FinalAppraisalDetermination/pdf/English (accessed July 2012).
- Rossell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Eng J Med 2009;361:958-67. http://dx.doi.org/10.1056/NEJMoa0904554.
- Diagnosis and treatment of lung cancer. London: National Institute for Health and Clinical Excellence; 2005.
- National Institute for Health and Care Excellence (NICE) . Lung Cancer (non-Small Cell) – Pemetrexed (maintenance) – Final Appraisal Determination 2010. www.nice.org.uk/guidance/index.jsp?action=download&o=48303 (accessed July 2012).
- National Institute for Health and Care Excellence (NICE) . Lung Cancer (non-Small Cell) – Erlotinib: Guidance 2008. http://guidance.nice.org.uk/TA162 (accessed July 2012).
- National Institute for Health and Care Excellence (NICE) . Lung Cancer (non-Small-Cell, Advanced or Metastatic Maintenance Treatment) – Erlotinib (monotherapy): Appraisal Consultation Document 2011. www.nice.org.uk/guidance/index.jsp?action=article&o=51743 (accessed February 2011).
- National Institute for Health and Care Excellence (NICE) . NICE Guidance by Topic- Cancer 2009. www.nice.org.uk/guidance/index.jsp?action=byTopic&o=7178 (accessed September 2009).
- Greenhalgh J, McLeod C, Bagust A, Boland A, Fleeman N, Dundar Y, et al. Pemetrexed for maintenance treatment of locally advanced or metastatic non-small cell lung cancer (NSCLC): a Single Technology Appraisal. Liverpool: Liverpool Reviews and Implementation Group (LRiG), The University of Liverpool; 2009.
- Cancer Research UK. Lung Cancer – UK Incidence Statistics n.d. http://info.cancerresearchuk.org/cancerstats/types/lung/incidence/ (accessed January 2011).
- Cappuzzo F, Ciuleanu T, Stelmakh L, Cicenas S, Szczésna A, Juhász E. Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: a multicentre, randomised, placebo-controlled phase 3 study. Lancet Oncol 2010;11:521-9. http://dx.doi.org/10.1016/S1470-2045(10)70112-1.
- National Institute for Health and Care Excellence (NICE) . NICE Technology Appraisal Guidance 181. Pemetrexed for the First-Line Treatment of Non-Small-Cell Lung Cancer. 2009. www.nice.org.uk/nicemedia/live/12243/45501/45501.pdf (accessed July 2012).
- National Institute for Health and Care Excellence (NICE) . NICE Technology Appraisal Guidance 192. Gefitinib for the First-Line Treatment of Locally Advanced or Metastatic Non-Small-Cell Lung Cancer 2010. http://guidance.nice.org.uk/TA192/Guidance/pdf/English (accessed July 2012).
- Wang J, Wu N, Cham M, Song Y. Tumor response in patients with advanced non–small cell lung cancer: perfusion CT evaluation of chemotherapy and radiation therapy. Am J Roentgenol 2009;193:1090-6. http://dx.doi.org/10.2214/AJR.08.1367.
- Aaronson N, Ahmedzai S, Bergman B. 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;85:365-76. http://dx.doi.org/10.1093/jnci/85.5.365.
- Bergman B, Aaronson N, Ahmedzai S. The EORTC QLQ-LC13: a modular supplement to the EORTC Core Quality of Life Questionnaire (QLQ-C30) for use in lung cancer clinical trials – EORTC Study Group on Quality of Life. Eur J Cancer 1994;30:635-42. http://dx.doi.org/10.1016/0959-8049(94)90535-5.
- Hollen PJ, Gralla RJ. Comparison of instruments for measuring quality of life in patients with lung cancer. Semin Oncol 1996;23:31-40.
- Cella D, Bonomi A, Lloyd S, Tulsky D, Kaplan E, Bonomi P. Reliability and validity of the Functional Assessment of Cancer Therapy-Lung (FACT-L) quality of life instrument. Lung Cancer 1995;12:199-220. http://dx.doi.org/10.1016/0169-5002(95)00450-F.
- Grutters J, Joore M, Wiegman E, Langendijk J, de Ruysscher D, Hochstenbag M. Health-related quality of life in patients surviving non-small cell lung cancer. Thorax 2010;65:903-7. http://dx.doi.org/10.1136/thx.2010.136390.
- Ranson M, Davidson N, Nicolson M, Falk S, Carmichael J, Loper P, et al. Randomized trial of paclitaxel plus supportive care versus supportive care for patients with advanced non-small-cell lung cancer. J Natl Cancer Inst 2000;92:1074-80. http://dx.doi.org/10.1093/jnci/92.13.1074.
- Bischoff HG, Anderson H, Arellano J, van den Borne B, Langer F, Leschinger MI, et al. Advanced Non-Small Cell Lung Cancer (NSCLC): Observation of Chemotherapy in 5 European Countries 2007. http://ers-education.org/media/2007/eposters/blogs-3571.html (accessed July 2011).
- Eli Lilly . Lilly Reports Third-Quarter 2010 Results n.d. https://investor.lilly.com/releasedetail2.cfm?ReleaseID=521145 (accessed February 2011).
- Espicom Business Intelligence Ltd . Cancer Drug Discoveries: What the Future Holds: Non-Small Cell Lung Cancer Chapter 2009. www.researchandmarkets.com/reportinfo.asp?report_id=568643&t=t&cat_id= (accessed February 2011).
- British National Formulary n.d. www.bnf.org/bnf/index.htm (accessed May 2011).
- Clegg A, Scott DA, Sidhu M, Hewitson P, Waugh N. A rapid and systematic review of the clinical effectiveness and cost-effectiveness of paclitaxel, docetaxel, gemcitabine and vinorelbine in non-small-cell lung cancer. Health Technol Assess 2001;5.
- Moher D, Liberati A, Tetzlaff J, Altman D, Group TP. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: the PRISMA Statement. BMJ 2009;339:332-9. http://dx.doi.org/10.1136/bmj.b2535.
- Royle P, Waugh N. Literature searching for clinical and cost-effectiveness studies used in health technology assessment reports carried out for the National Institute for Clinical Excellence appraisal system. Health Technol Assess 2003;7.
- Centre for Reviews and Dissemination . Centre for Reviews and Dissemination'S Guidance for Undertaking Reviews in Health Care 2009. www.york.ac.uk/inst/crd/pdf/Systematic_Reviews.pdf (accessed July 2012).
- Scagliotti GV, De Marinis F, Rinaldi M, Crino L, Gridelli C, Ricci S, et al. Phase III randomized trial comparing three platinum-based doublets in advanced non-small-cell lung cancer. J Clin Oncol 2002;20:4285-91. http://dx.doi.org/10.1200/JCO.2002.02.068.
- Fossella F, Pereira JR, von Pawel J, Pluzanska A, Gorbounova V, Kaukel E, et al. Randomized, multinational, phase III study of docetaxel plus platinum combinations versus vinorelbine plus cisplatin for advanced non-small-cell lung cancer: the TAX 326 study group. J Clin Oncol 2003;21:3016-24. http://dx.doi.org/10.1200/JCO.2003.12.046.
- Gridelli C, Gallo C, Shepherd FA, Illiano A, Piantedosi F, Robbiati SF, et al. Gemcitabine plus vinorelbine compared with cisplatin plus vinorelbine or cisplatin plus gemcitabine for advanced non-small-cell lung cancer: a phase III trial of the Italian GEMVIN Investigators and the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 2003;21:3025-34. http://dx.doi.org/10.1200/JCO.2003.06.099.
- Smit EF, van Meerbeeck JPAM, Lianes P, Debruyne C, Legrand C, Schramel F, et al. Three-arm randomized study of two cisplatin-based regimens and paclitaxel plus gemcitabine in advanced non-small-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;21:3909-17. http://dx.doi.org/10.1200/JCO.2003.03.195.
- Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Eng J Med 2002;346:92-8. http://dx.doi.org/10.1056/NEJMoa011954.
- Kelly K, Crowley J, Bunn PA, Presant CA, Grevstad PK, Moinpour CM, et al. Randomized phase III trial of paclitaxel plus carboplatin versus vinorelbine plus cisplatin in the treatment of patients with advanced non-small-cell lung cancer: a Southwest Oncology Group trial. J Clin Oncol 2001;19:3210-18.
- Gebbia V, Galetta D, Caruso M, Verderame F, Pezzella G, Valdesi M, et al. Gemcitabine and cisplatin versus vinorelbine and cisplatin versus ifosfamide + gemcitabine followed by vinorelbine and cisplatin versus vinorelbine and cisplatin followed by ifosfamide and gemcitabine in stage IIIB-IV non small cell lung carcinoma: a prospective randomized phase III trial of the Gruppo Oncologico Italia Meridionale. Lung Cancer 2003;39:179-89. http://dx.doi.org/10.1016/S0169-5002(02)00444-0.
- Chang J, Tsao T, Yang C, Lin M, Cheung Y, Liaw C, et al. A randomized study of gemcitabine plus cisplatin and vinorelbine plus cisplatin in patients with advanced non-small-cell lung cancer. Chang Gung Med J 2008;31:559-66.
- Chen YM, Perng RP, Shih JF, Lee YC, Lee CS, Tsai CM, et al. A randomised phase II study of weekly paclitaxel or vinorelbine in combination with cisplatin against inoperable non-small-cell lung cancer previously untreated. Br J Cancer 2004;90:359-65. http://dx.doi.org/10.1038/sj.bjc.6601526.
- Chen YM, Perng RP, Shih JF, Tsai CM, Whang-Peng J. A randomized phase II study of docetaxel or vinorelbine in combination with cisplatin against inoperable, chemo-naive non-small-cell lung cancer in Taiwan. Lung Cancer 2007;56:363-9. http://dx.doi.org/10.1016/j.lungcan.2007.01.011.
- Douillard JY, Gervais R, Dabouis G, Le Groumellec A, D’Arlhac M, Spaeth D, et al. Sequential two-line strategy for stage IV non-small-cell lung cancer: docetaxel-cisplatin versus vinorelbine-cisplatin followed by cross-over to single-agent docetaxel or vinorelbine at progression: final results of a randomised phase II study. Ann Oncol 2005;16:81-9. http://dx.doi.org/10.1093/annonc/mdi013.
- Martoni A, Marino A, Sperandi F, Giaquinta S, Di Fabio F, Melotti B, et al. Multicentre randomised phase III study comparing the same dose and schedule of cisplatin plus the same schedule of vinorelbine or gemcitabine in advanced non-small cell lung cancer. Eur J Cancer 2005;41:81-92. http://dx.doi.org/10.1016/j.ejca.2004.08.029.
- Helbekkmo N, Sundstrom SH, Aasebo U, Fr Brunsvig P, Von Plessen C, Hjelde HH, et al. Vinorelbine/carboplatin vs gemcitabine/carboplatin in advanced NSCLC shows similar efficacy, but different impact of toxicity. Br J Cancer 2007;97:283-9. http://dx.doi.org/10.1038/sj.bjc.6603869.
- Langer CJ, Li S, Schiller J, Tester W, Rapoport BL, Johnson DH. Randomized phase II trial of paclitaxel plus carboplatin or gemcitabine plus cisplatin in Eastern Cooperative Oncology Group performance status 2 non-small-cell lung cancer patients: ECOG. J Clin Oncol 1599;25:418-23. http://dx.doi.org/10.1200/JCO.2005.04.9452.
- Ohe Y, Ohashi Y, Kubota K, Tamura T, Nakagawa K, Negoro S, et al. Randomized phase III study of cisplatin plus irinotecan versus carboplatin plus paclitaxel, cisplatin plus gemcitabine, and cisplatin plus vinorelbine for advanced non-small-cell lung cancer: four-arm cooperative study in Japan. Ann Oncol 2007;18:317-23. http://dx.doi.org/10.1093/annonc/mdl377.
- Thomas P, Robinet G, Gouva S, Fournel P, Lena H, Le Caer H, et al. Randomized multicentric phase II study of carboplatin/gemcitabine and cisplatin/vinorelbine in advanced non-small cell lung cancer. GFPC 99–01 study (Groupe Francais de Pneumo-cancerologie). Lung Cancer 2006;51:105-14. http://dx.doi.org/10.1016/j.lungcan.2005.10.004.
- Tan EH, Rolski J, Grodzki T, Schneider CP, Gatzemeier U, Zatloukal P, et al. Global lung oncology branch trial 3 (GLOB3): final results of a randomised multinational phase III study alternating oral and i.v. vinorelbine plus cisplatin versus docetaxel plus cisplatin as first-line treatment of advanced non-small-cell lung cancer. Ann Oncol 2009;20:1249-56. http://dx.doi.org/10.1093/annonc/mdn774.
- Treat JA, Gonin R, Socinski MA, Edelman MJ, Catalano RB, Marinucci DM, et al. A randomized, phase III multicenter trial of gemcitabine in combination with carboplatin or paclitaxel versus paclitaxel plus carboplatin in patients with advanced or metastatic non-small-cell lung cancer. Ann Oncol 2010;21:540-7. http://dx.doi.org/10.1093/annonc/mdp352.
- Scagliotti GV, Parikh P, Von Pawel J, Biesma B, Vansteenkiste J, Manegold C, et al. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol 2008;26:3543-51. http://dx.doi.org/10.1200/JCO.2007.15.0375.
- Gronberg BH, Bremnes RM, Flotten O, Amundsen T, Brunsvig PF, Hjelde HH, et al. Phase III study by the Norwegian lung cancer study group: pemetrexed plus carboplatin compared with gemcitabine plus carboplatin as first-line chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol 2009;27:3217-24. http://dx.doi.org/10.1200/JCO.2008.20.9114.
- Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Eng J Med 2010;362:2380-8. http://dx.doi.org/10.1056/NEJMoa0909530.
- Fukuoka M, Wu Y-L, Thongprasert S, Sunpaweravong P, Leong SS, Sriuranpong V, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non–small-cell lung cancer in Asia (IPASS). J Clin Oncol 2011;29:2866-74. http://dx.doi.org/10.1200/JCO.2010.33.4235.
- Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 2010;11:121-8. http://dx.doi.org/10.1016/S1470-2045(09)70364-X.
- Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints. Stat Med 1998;17:2815-34. http://dx.doi.org/10.1002/(SICI)1097-0258(19981230)17:24<2815::AID-SIM110>3.0.CO;2-8.
- Williamson P, Smith C, Hutton J, Marson A. Aggregate data meta-analysis with time-to-event outcomes. Stat Med 2002;21:3337-51. http://dx.doi.org/10.1002/sim.1303.
- Higgins J, Thompson S. Quantifying heterogeneity in a meta-analysis. Stat Med 2002;21:1539-58. http://dx.doi.org/10.1002/sim.1186.
- Higgins J, Thompson S, Deeks J, Altman D. Measuring inconsistency in meta-analyses. BMJ 2003;327:557-60. http://dx.doi.org/10.1136/bmj.327.7414.557.
- Lu G, Ades A. Combination of direct and indirect evidence in mixed treatment comparisons. Stat Med 2004;23:3105-24. http://dx.doi.org/10.1002/sim.1875.
- Caldwell D, Ades A, Higgins J. Simultaneous comparison of multiple treatments: combining direct and indirect evidence. BMJ 2005;331:897-900. http://dx.doi.org/10.1136/bmj.331.7521.897.
- Multi-parameter Evidence Synthesis Research Group . Formal Methods for Multi-Parameter Evidence Synthesis and Their Role in Epidemiology and Economic Evaluation n.d. www.bristol.ac.uk/cobm/research/mpes (accessed July 2012).
- Brooks SP, Gelman A. Alternative methods for monitoring convergence of iterative simulations. J Comput Graph Stat 1998;7:434-55.
- Tsai CMWG, Su WJ, Chen YM, Chiu CH, Chiang SC, Perng RP. Randomized phase II study of cisplatin/vinorelbine vs. cisplatin/gemcitabine vs. vinorelbine/gemcitabine in patients with advanced non-small cell lung cancer. Proc Am Soc Clin Oncol 2003;22.
- Kawahara MKK, Yoshioka H, Kawasaki M, Fujita Y, Yonei T, Ogushi F, et al. Randomized phase II study of carboplatin plus either docetaxel (CbD) or paclitaxel (CbP) in Japanese patients with advanced non-small cell lung cancer (NSCLC): JMTO LC07–01. J Clin Oncol 2010;28.
- Huang CH, Langer CJ, Minniti JC, Nahum K, Seldomridge J, Hutter B, et al. Phase III toxicity trial of carboplatin (Cb) plus either docetaxel (D) or paclitaxel (P) in advanced non-small cell lung cancer (NSCLC): preliminary findings of OPN-001. Proc Am Soc Clin Oncol 2002;21.
- McNeil C. Negative data from lung cancer trial may change practice guidelines, study designs. J Natl Cancer Inst 2006;98:1518-20. http://dx.doi.org/10.1093/jnci/djj459.
- Office for National Statistics . Cancer Statistics Registrations. Office for National Statistics; 2008. MB1 39 n.d. www.statistics.gov.uk/downloads/theme_health/mb1–39/mb1-no39–2008.pdf (accessed July 2011).
- Le Chevalier T, Scagliotti G, Natalec R, Dansond S, Roselle R, Stahelf R, et al. Efficacy of gemcitabine plus platinum chemotherapy compared with other platinum containing regimens in advanced non-small-cell lung cancer: a meta-analysis of survival outcomes. Lung Cancer 2005;47:69-80. http://dx.doi.org/10.1016/j.lungcan.2004.10.014.
- US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research, Center for Biologics Evaluation and Research . Guidance for Industry: Clinical Trial Endpoints for the Approval of Cancer Dugs and Biologics 2007. www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm071590.pdf (accessed July 2011).
- Eisenhauer E, Therasse P, Bogaerts J, Schwartz L, Sargent D, Ford R, et al. New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline (version 1.1) 2009. www.recist.com/ (accessed February 2011).
- Ou S, Ziogas A, Zell J. Asian ethnicity is a favorable prognostic factor for overall survival in non-small cell lung cancer (NSCLC) and is independent of smoking status. J Thorac Oncol 2009;4:1083-93. http://dx.doi.org/10.1097/JTO.0b013e3181b27b15.
- Billingham L, Gaunt P, Jarrett HW, Dunlop D, Thompson J, O’Byrne KJ, et al. Quality of life in advanced non-small cell lung cancer, effects of cisplatin dose and carboplatin in combination with gemcitabine: results from BTOG2, a British Thoracic Oncology Group phase III trial in 1363 patients. Thorax 2011;66. http://dx.doi.org/10.1136/thoraxjnl-2011-201054b.86.
- Ardizzoni AL, Boni L, Tiseo M, Fossella F, Schiller J, Paesmans M, et al. Cisplatin- versus carboplatin-based chemotherapy in first-line treatment of advanced non – small-cell lung cancer: an individual patient data meta-analysis. J Natl Cancer Inst 2007;99:847-57. http://dx.doi.org/10.1093/jnci/djk196.
- Jiang J, Liang X, Zhou X, Huang R, Chu Z. A meta-analysis of randomized controlled trials comparing carboplatin-based to cisplatin-based chemotherapy in advanced non-small cell lung cancer. Lung Cancer 2007;57:348-58. http://dx.doi.org/10.1016/j.lungcan.2007.03.014.
- Ferry D, Billingham L, Jarrett H, Dunlop D, Thompson J, Kumar M, et al. British Thoracic Oncology Group Trial, BTOG2: Randomised Phase III Clinical Trial of Gemcitabine (1250 Mg m2) Combined With Cisplatin 50 Mg M2 (GC50) Versus Cisplatin 80 Mg M2 (GC80) Versus Carboplatin AUC 6 (GCb6) in Advanced NSCLC 2011.
- National Cancer Intelligence Network . Using Information to Improve Quality & Choice 2010. www.ncin.org.uk/home.aspx (accessed July 2011).
- Gridelli C. The ELVIS trial: a phase III study of single-agent vinorelbine as first-line treatment in elderly patients with advanced non-small cell lung cancer. Oncologist 2001;6:4-7. http://dx.doi.org/10.1634/theoncologist.6-suppl_1-4.
- Gridelli C, Perrone F, Gallo C, Cigolari S, Rossi A, Piantedosi F, et al. Chemotherapy for elderly patients with advanced non-small-cell lung cancer: The multicenter Italian lung cancer in the elderly study (MILES) phase III randomized trial. J Natl Cancer Inst 2003;95:362-72. http://dx.doi.org/10.1093/jnci/95.5.362.
- Peake M, Thompson S, Lowe D, Pearson M. centres Obotp . Ageism in the management of lung cancer. Age Aging 2003;32:171-7. http://dx.doi.org/10.1093/ageing/32.2.171.
- Beckett P, Woolhouse I, Peake M, Stanley R, Harrison R. P215 The influence of age on management of lung cancer patients in England. Thorax 2010;65:A167-A8. http://dx.doi.org/10.1136/thx.2010.151068.16.
- Chen YM, Perng RP, Lee YC, Shih JF, Lee CS, Tsai CM, et al. Paclitaxel plus carboplatin, compared with paclitaxel plus gemcitabine, shows similar efficacy while more cost-effective: a randomized phase II study of combination chemotherapy against inoperable non-small-cell lung cancer previously untreated. Ann Oncol 2002;13:108-15. http://dx.doi.org/10.1093/annonc/mdf009.
- Clegg A, Scott DA, Hewitson P, Sidhu M, Waugh N. Clinical and cost effectiveness of paclitaxel, docetaxel, gemcitabine, and vinorelbine in non-small cell lung cancer: a systematic review. Thorax 2002;57:20-8. http://dx.doi.org/10.1136/thorax.57.1.20.
- Dooms CA, Lievens YN, Vansteenkiste JF. Cost-utility analysis of chemotherapy symptomatic advanced nonsmall cell cancer. Eur Respir J 2006;27:895-901.
- Klein R, Muehlenbein C, Liepa AM, Babineaux S, Wielage R, Schwartzberg L. Cost-effectiveness of pemetrexed plus cisplatin as first-line therapy for advanced nonsquamous non-small cell lung cancer. J Thorac Oncol 2009;4:1404-14. http://dx.doi.org/10.1097/JTO.0b013e3181ba31e0.
- Le Lay K, Myon E, Hill S, Riou-Franca L, Scott D, Sidhu M, et al. Comparative cost-minimisation of oral and intravenous chemotherapy for first-line treatment of non-small cell lung cancer in the UK NHS system. Eur J Health Econ 2007;8:145-51. http://dx.doi.org/10.1007/s10198-006-0034-1.
- Lees M, Aristides M, Maniadakis N, McKendrick J, Botwood N, Stephenson D. Economic evaluation of gemcitabine alone and in combination with cisplatin in the treatment of non-small cell lung cancer. Pharmacoeconomics 2002;20:325-37. http://dx.doi.org/10.2165/00019053-200220050-00004.
- Lievens Y, Kesteloot K, Van den Bogaert W. CHART in lung cancer: economic evaluation and incentives for implementation. Radiother Oncol 2005;75:171-8. http://dx.doi.org/10.1016/j.radonc.2005.03.011.
- Maniadakis N, Fragoulakis V, Pallis A, Prezerakos P, Georgoulias V. Economic evaluation of docetaxel/gemcitabine versus docetaxel as frontline treatment of patients with advanced/metastatic non-small cell lung cancer in Greece. Lung Cancer 2007;58:275-81. http://dx.doi.org/10.1016/j.lungcan.2007.06.012.
- Neubauer MA, Hoverman JR, Kolodziej M, Reisman L, Gruschkus SK, Hoang S, et al. Cost effectiveness of evidence-based treatment guidelines for the treatment of non-small-cell lung cancer in the community setting. J Oncol Prac 2010;6:12-8. http://dx.doi.org/10.1200/JOP.091058.
- Neymark N, Lianes P, Smit EF, Van Meerbeeck JP. Economic evaluation of three two-drug chemotherapy regimens in advanced non-small-cell lung cancer. Pharmacoeconomics 2005;23:1155-66. http://dx.doi.org/10.2165/00019053-200523110-00007.
- Novello S, Kielhorn A, Stynes G, Selvaggi G, De Marinis F, Maestri A, et al. Cost-minimisation analysis comparing gemcitabine/cisplatin, paclitaxel/carboplatin and vinorelbine/cisplatin in the treatment of advanced non-small cell lung cancer in Italy. Lung Cancer 2005;48:379-87. http://dx.doi.org/10.1016/j.lungcan.2004.11.014.
- Pimentel FL, Bhalla S, Laranjeira L, Guerreiro M. Cost-minimization analysis for Portugal of five doublet chemotherapy regimens from two phase III trials in the treatment of advanced non-small cell lung cancer. Lung Cancer 2006;52:365-71. http://dx.doi.org/10.1016/j.lungcan.2006.03.005.
- Rubio-Terres C, Luis Tisaire J, Kobina S, Moyano A. Cost-minimisation analysis of three regimens of chemotherapy (docetaxel-cisplatin, paclitaxel-cisplatin, paclitaxel-carboplatin) for advanced non-small-cell lung cancer. Lung Cancer 2002;35:81-9. http://dx.doi.org/10.1016/S0169-5002(01)00280-X.
- Maniadakis N, Fragoulakis V, Pallis AG, Simou E, Georgoulias V. Economic evaluation of docetaxel-gemcitabine versus vinorelbine–cisplatin combination as front-line treatment of patients with advanced/metastatic nonsmall-cell lung cancer in Greece: A cost-minimization analysis. Ann Oncol 2010;21:1462-7. http://dx.doi.org/10.1093/annonc/mdp551.
- Drummond MF, Jefferson TO. Guidelines for authors and peer reviewers of economic submissions to the BMJ. The BMJ Economic Evaluation Working Party. BMJ 1996;313:275-83. http://dx.doi.org/10.1136/bmj.313.7052.275.
- Medicare – Centers for Medicare & Medicaid Services (CMS) . Payment Allowance Limits for Medicare: Part B Drugs Effective January 1, 2008 through March 31, 2008 n.d. www.cms.hhs.gov/McrPartB DrugAvgSalesPrice/01a_2008aspfiles.asp#TopOfPage (accessed January 8 2008).
- British national formulary. London: BMA and RPS; 2000.
- Department of Health . The New NHS – 2000 Reference Costs n.d. www.doh.gov.uk/nhsexec/refcosts.htm (accessed 28 March 2002).
- Netten A, Curtis L. Unit costs of health and social care 2000. Canterbury: Personal Social Services Research Unit, University of Kent at Canterbury; 2000.
- Bagust A, Barraza-Llorens M, Philips Z. Economic evaluation of gemcitabine in the treatment of advanced non-small cell lung cancer. Stage 3: cost-effectiveness analysis. York Health Economics Consortium, University of York: Eli Lilly and Company Ltd; 1999.
- College Tarieven Gezondheidszorg . Tariefboek Medisch Specialitsen and Bijlage Bij Tarieflijst Instellingen 2002 n.d. www.ctgzorg.nl (accessed September 2003).
- Farmakotherapeutisch Kompas 2002. Amstelveen: Medisch Farmaceutische Voor-lichting; 2002.
- Vansteenkiste JF, Vandebroek JE, Nackaerts KL, Weynants P, Valcke YJ, Verresen DA, et al. Clinical-benefit response in advanced non-small-cell lung cancer: A multicentre prospective randomised phase III study of single agent gemcitabine versus cisplatin-vindesine. Ann Oncol 2001;12:1221-30. http://dx.doi.org/10.1023/A:1012208711013.
- Vansteenkiste J, Edelman MJ, De Marinis F, Douillard J-Y, Stahel RA, Arellano J, et al. Indirect Comparison of Pemetrexed Plus Cisplatin With Other Platinum Based Therapies for First Line Advanced Non Small Cell Lung Cancer 2008.
- Nafees B, Stafford M, Gavriel S, Bhalla S, Watkins J. Health state utilities for non small cell lung cancer. Health Qual Life Outcome 2008;6. http://dx.doi.org/10.1186/1477-7525-6-84.
- Anderson H, Hopwood P, Stephens RJ, Thatcher N, Cottier B, Nicholson M, et al. Gemcitabine plus best supportive care (BSC) vs BSC in inoperable non-small cell lung cancer--a randomized trial with quality of life as the primary outcome. UK NSCLC Gemcitabine Group. Non-Small Cell Lung Cancer. Br J Cancer 2000;83:447-53. http://dx.doi.org/10.1054/bjoc.2000.1307.
- Cardenal F, Lopez-Cabrerizo MP, Anton A, Alberola V, Massuti B, Carrato A, et al. Randomized phase III study of gemcitabine-cisplatin versus etoposide-cisplatin in the treatment of locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 1999;17:12-8.
- Crino L, Scagliotti GV, Ricci S, De Marinis F, Rinaldi M, Gridelli C, et al. Gemcitabine and cisplatin versus mitomycin, ifosfamide, and cisplatin in advanced non-small-cell lung cancer: A randomized phase III study of the Italian Lung Cancer Project. J Clin Oncol 1999;17:3522-30.
- Comella P. Interim analysis of a phase III trial. Triple- vs double-agent chemotherapy for advanced non-small-cell lung cancer. Southern Italy Cooperative Oncology Group. Oncology 2000;14:35-40.
- Berthelot JM, Will BP, Evans WK, Coyle D, Earle CC, Bordeleau L. Decision framework for chemotherapeutic interventions for metastatic non-small-cell lung cancer. J Natl Cancer Inst 2000;92:1321-9. http://dx.doi.org/10.1093/jnci/92.16.1321.
- Pemetrexed for the first-line treatment of locally advanced or metastatic non-small cell lung cancer (NSCLC). London: NICE; 2009.
- Gefitinib for the first-line treatment of locally advanced or metastatic non-small cell lung cancer (NSCLC). London: NICE; 2009.
- Office for National Statistics . Death Registration Summary Tables England and Wales 2010 n.d. www.ons.gov.uk/ons/publications/re-reference-tables.html?edition=tcm%3A77–227638 (accessed September 2011).
- Carlson J, Veenstra D, Ramsey S. Pharmacoeconomic evaluations in the treatment of non-small cell lung cancer. Drugs 2008;68:1105-13. http://dx.doi.org/10.2165/00003495-200868080-00007.
- Crino L, Cappuzzo F, Zatloukal P, Reck M, Pesek M, Thompson JC, et al. Gefitinib versus vinorelbine in chemotherapy-naive elderly patients with advanced non-small-cell lung cancer (invite): A randomized, phase II study. J Clin Oncol 2008;26:4253-60. http://dx.doi.org/10.1200/JCO.2007.15.0672.
- Kudoh S, Takeda K, Nakagawa K, Takada M, Katakami N, Matsui K, et al. Phase III study of docetaxel compared with vinorelbine in elderly patients with advanced non-small-cell lung cancer: results of the West Japan Thoracic Oncology Group Trial (WJTOG 9904). J Clin Oncol 2006;24:3657-63. http://dx.doi.org/10.1200/JCO.2006.06.1044.
- Scagliotti G, Novello S, von Pawel J, Reck M, Pereira JR, Thomas M, et al. Phase III study of carboplatin and paclitaxel alone or with sorafenib in advanced non-small-cell lung cancer. J Clin Oncol 2010;28:1835-42. http://dx.doi.org/10.1200/JCO.2009.26.1321.
- Ekhart C, de Jonge ME, Huitema ADR, Schellens JHM, Rodehuis S, Beijnen JH. Flat dosing of carboplatin is justified in adult patients with normal renal function. Clin Cancer Res 2006;12:6502-8. http://dx.doi.org/10.1158/1078-0432.CCR-05-1076.
- British national formulary. London: BMA and RPS; 2011.
- Electronic market information tool (eMit), Commercial Medicines Unit, Department of Health . Electronic Market Information Tool (eMit) n.d. http://cmu.dh.gov.uk/electronic-market-information-tool-emit/ (accessed September 2011).
- Department of Health . NHS Reference Costs 2009–2010 n.d. www.dh.gov.uk/en/Publicationsandstatistics/PublicationsPolicyAndGuidance/DH_123459 (accessed September 2011).
- Maslove L, Gower NH, Spiro SG, Rudd RM, Stephens R, West P. Estimation of the additional costs of chemotherapy for patients with advanced non-small cell lung cancer. Thorax 2005;60:564-9. http://dx.doi.org/10.1136/thx.2004.039479.
- National Institute for Health and Care Excellence . Advanced Breast Cancer: Diagnosis and Treatment 2009. www.nice.org.uk/nicemedia/live/11778/43414/43414.pdf (accessed July 2011).
- Taylor D, Carter S. Valuing choice – dying at home: a case for the more equitable provision of high quality support for people who wish to die at home. London: School of Pharmacy, University of London; 2004.
- National Institute for Health and Care Excellence (NICE) . Etanercept, Infliximab and Adalimumab for the Treatment of Psoriatic Arthritis 2010. www.nice.org.uk/nicemedia/live/13110/50422/50422.pdf (accessed July 2011).
- Personal Social Services Research Unit . Unit Costs of Health and Social Care 2010 2010. www.pssru.ac.uk/uc/uc2010contents.htm (accessed July 2011).
- Doyle S, Llyod A, Walker M. Health state utility scores in advanced non-small cell lung cancer. Lung Cancer 2008;62:374-80. http://dx.doi.org/10.1016/j.lungcan.2008.03.019.
- van den Hout W, Kramer G, Noordijk E, Leer J-W. Cost-utility analysis of short- vesrus long-course palliative radiotherapy in patients with non-small-cell ling cancer. J Natl Cancer Inst 2006;98:1786-94. http://dx.doi.org/10.1093/jnci/djj496.
- National Institute for Health and Care Excellence (NICE) . Guide to the Methods of Technology Appraisal. 2008. www.nice.org.uk/media/B52/A7/TAMethodsGuideUpdatedJune2008.pdf (accessed 2009 June).
- Ku GY, Haaland BA, Lopes. Gefitinib vs. chemotherapy as first-line therapy in advanced non-small cell lung cancer: meta-analysis of phase III trials. Lung Cancer 2011;74:469-73. http://dx.doi.org/10.1016/j.lungcan.2011.04.008.
- European Medicines Agency . Erbitux n.d. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000558/wapp/Post-authorisation/human_wapp_000145.jsp&mid=WC0b01ac058001d128 (accessed February 2011).
- Fisher MD. Irinotecan/cisplatin versus vindesine/cisplatin versus irinotecan alone in advanced non-small-cell lung cancer. Clin Lung Cancer 2000;2:180-1.
- Georgoulias V, Pallis AG, Kourousis C, Alexopoulos A, Ardavanis A, Agelidou A, et al. Docetaxel versus docetaxel/cisplatin in patients with advanced non-small-cell lung cancer: Preliminary analysis of a multicenter, randomized phase III study. Clin Lung Cancer 2003;4:288-93. http://dx.doi.org/10.3816/CLC.2003.n.008.
- Gridelli C, Butts C, Ciardiello F, Feld R, Gallo C, Perrone F. An international, multicenter, randomized phase III study of first-line erlotinib followed by second-line cisplatin/gemcitabine versus first-line cisplatin/gemcitabine followed by second-line erlotinib in advanced non-small-cell lung cancer: treatment rationale and protocol dynamics of the TORCH trial. Clin Lung Cancer 2008;9:235-8. http://dx.doi.org/10.3816/CLC.2008.n.037.
- Gridelli C, Gallo C, Ceribelli A, Gebbia V, Gamucci T, Ciardiello F, et al. Factorial phase III randomised trial of rofecoxib and prolonged constant infusion of gemcitabine in advanced non-small-cell lung cancer: the GEmcitabine-COxib in NSCLC (GECO) study. Lancet Oncol 2007;8:500-12. http://dx.doi.org/10.1016/S1470-2045(07)70146-8.
- Grigorescu AC, Draghici IN, Nitipir C, Gutulescu N, Corlan E. Gemcitabine (GEM) and carboplatin (CBDCA) versus cisplatin (CDDP) and vinblastine (VLB) in advanced non-small-cell lung cancer (NSCLC) stages III and IV: A phase III randomised trial. Lung Cancer 2002;37:9-14. http://dx.doi.org/10.1016/S0169-5002(01)00493-7.
- Leong SS, Tan EH, Fong KW, Wilder-Smith E, Ong YK, Tai BC, et al. Randomized double-blind trial of combined modality treatment with or without amifostine in unresectable stage III non-small-cell lung cancer. J Clin Oncol 2003;21:1767-74. http://dx.doi.org/10.1200/JCO.2003.11.005.
- Miller VA, Johnson DH, Krug LM, Pizzo B, Tyson L, Perez W, et al. Pilot trial of the epidermal growth factor receptor tyrosine kinase inhibitor gefitinib plus carboplatin and paclitaxel in patients with stage IIIB or IV non-small-cell lung cancer. J Clin Oncol 2003;21:2094-100. http://dx.doi.org/10.1200/JCO.2003.12.008.
- O’Brien MER, Anderson H, Kaukel E, O’Byrne K, Pawlicki M, von Pawel J, et al. SRL 172 (killed Mycobacterium vaccae) in addition to standard chemotherapy improves quality of life without affecting survival, in patients with advanced non-small-cell lung cancer: phase III results. Ann Oncol 2004;15:906-14. http://dx.doi.org/10.1093/annonc/mdh220.
- Ramalingam S, Barstis J, Perry MC, La Rocca RV, Nattam SR, Rinaldi D, et al. Treatment of elderly non-small cell lung cancer patients with three different schedules of weekly paclitaxel in combination with carboplatin: Subanalysis of a randomized trial. J Thorac Oncol 2006;1:240-4.
- Vansteenkiste J, Vandebroek J, Nackaerts K, Dooms C, Galdermans D, Bosquee L, et al. Influence of cisplatin-use, age, performance status and duration of chemotherapy on symptom control in advanced non-small cell lung cancer: detailed symptom analysis of a randomised study comparing cisplatin-vindesine to gemcitabine. Lung Cancer 2003;40:191-9. http://dx.doi.org/10.1016/S0169-5002(02)00515-9.
- Gao Y, Shi ZQ, Cao CW, Zhu CL, Guo J. A randomized trial of docetaxol plus cisplatin versus gemzar plus cisplatin in treating advanced non-small cell lung cancer. Ai Zheng 2005;24:985-9.
- Lin YC, Lin Y, Lin W, Du CW, Wu MY, Li DR. Comparison of effects of three chemotherapy regimens for advanced non-small cell lung cancer. Ai 2002;21:1359-61.
- Liu L, Wang XW, Li L, Zhang X, Zhang WD, Yu XJ. A randomized comparative trial of three combined regimens containing cisplatin for treatment of advanced non-small cell lung cancer. Ai 2006;25:990-4.
- Semrau S, Fietkau R. Randomized phase II study of cisplatin with gemcitabine or paclitaxel or vinorelbine as induction chemotherapy followed by concomitant chemoradiotherapy for stage IIIB non-small-cell lung cancer: cancer and leukemia group B study 9431. Strahlenther Onkol 2003;179:502-3.
- Teng XY, Zhou NN, Su YS, Zhou ZM, Liu DG, He YJ. Comparison of cisplatin combined with gemcitabine versus cisplatin combined with vinorelbine regimen for treatment of patients with advanced non-small cell lung cancer. Ai Zheng 2003;22:404-6.
- Xu S, Wang W, Liu X. Clinical studies on the therapy of advanced non-small cell lung cancer (NSCLC) in the elderly using domestic gemcitabine in combination with carboplatin. Chin J Clin Oncol 2006;33:35-7.
- Zhang S, Wang J, Wang Q, Yang X, Hu F, Li X, et al. A randomized study of docetaxel plus cisplatin versus paclitaxel plus cisplatin in previously untreated advanced non-small cell lung cancer. Chin J Lung Cancer 2008;11:110-14.
Appendix 1 Details of clinical search strategies
Appendix 2 Protocol
Appendix 3 Details of clinical data abstraction
Appendix 4 Details of clinical quality assessment
Appendix 5 Letter to authors of included studies (via e-mail)
Letter to authors of included studies (via e-mail) (PDF download)
Appendix 6 Code from the Multi-parameter Evidence Synthesis Research Group
Code from the Multi-parameter Evidence Synthesis Research Group (PDF download)
Appendix 7 References of excluded clinical studies with reasons for exclusion
References of excluded clinical studies with reasons for exclusion (PDF download)
Appendix 8 Trial characteristics
Appendix 9 Intervention details
Appendix 10 Patient characteristics
Appendix 11 Inclusion/exclusion criteria
Appendix 12 Summary results for the sensitivity analyses for mixed-treatment comparison for overall survival comparing chemotherapy with chemotherapy in population 1
Summary results for the sensitivity analyses for mixed-treatment comparison for overall survival comparing chemotherapy with chemotherapy in population 1 (PDF download)
Appendix 13 Summary results for the sensitivity analyses for mixed-treatment comparison for progression-free survival comparing chemotherapy with chemotherapy in population 1
Summary results for the sensitivity analyses for mixed-treatment comparison for progression-free survival comparing chemotherapy with chemotherapy in population 1 (PDF download)
Appendix 14 Summary results for the sensitivity analyses for mixed-treatment comparison for time to progression comparing chemotherapy with chemotherapy in population 1
Summary results for the sensitivity analyses for mixed-treatment comparison for time to progression comparing chemotherapy with chemotherapy in population 1 (PDF download)
Appendix 15 Summary results for the sensitivity analyses for mixed-treatment comparison for progression-free survival/time to progression comparing chemotherapy with chemotherapy in population 1
Summary results for the sensitivity analyses for mixed-treatment comparison for progression-free survival/time to progression comparing chemotherapy with chemotherapy in population 1 (PDF download)
Appendix 16 Summary results for the direct meta-analysis and results of the mixed-treatment comparison 1-year survival for trials comparing chemotherapy with chemotherapy in population 1
Summary results for the direct meta-analysis and results of the mixed-treatment comparison 1-year survival for trials comparing chemotherapy with chemotherapy in population 1 (PDF download)
Appendix 17 Summary results for the direct meta-analysis and results of the mixed-treatment comparison 2-year survival for trials comparing chemotherapy with chemotherapy in population 1
Summary results for the direct meta-analysis and results of the mixed-treatment comparison 2-year survival for trials comparing chemotherapy with chemotherapy in population 1 (PDF download)
Appendix 18 Summary results for the sensitivity analyses for mixed-treatment comparison for overall survival comparing chemotherapy with chemotherapy in population 2
Summary results for the sensitivity analyses for mixed-treatment comparison for overall survival comparing chemotherapy with chemotherapy in population 2 (PDF download)
Appendix 19 Summary results for the sensitivity analyses for mixed-treatment comparison for progression-free survival comparing chemotherapy with chemotherapy in population 2
Summary results for the sensitivity analyses for mixed-treatment comparison for progression-free survival comparing chemotherapy with chemotherapy in population 2 (PDF download)
Appendix 20 Summary results for the sensitivity analyses for direct meta-analysis for progression-free survival/time to progression comparing chemotherapy with chemotherapy in population 2
Summary results for the sensitivity analyses for direct meta-analysis for progression-free survival/time to progression comparing chemotherapy with chemotherapy in population 2 (PDF download)
Appendix 21 Summary results for the sensitivity analyses for mixed-treatment comparison for progression-free survival/time to progression comparing chemotherapy with chemotherapy in population 2
Summary results for the sensitivity analyses for mixed-treatment comparison for progression-free survival/time to progression comparing chemotherapy with chemotherapy in population 2 (PDF download)
Appendix 22 Summary results for the direct meta-analysis and mixed-treatment comparison for 1-year survival for trials comparing chemotherapy with chemotherapy in population 2
Summary results for the direct meta-analysis and mixed-treatment comparison for 1-year survival for trials comparing chemotherapy with chemotherapy in population 2 (PDF download)
Appendix 23 Summary results for the direct meta-analysis and mixed-treatment comparison for 2-year survival for trials comparing chemotherapy with chemotherapy in population 2
Summary results for the direct meta-analysis and mixed-treatment comparison for 2-year survival for trials comparing chemotherapy with chemotherapy in population 2 (PDF download)
Appendix 24 Adverse events, haematological grades 3–4
Appendix 25 Adverse events, non-haematological grades 3–4
Adverse events, non-haematological grades 3–4 (PDF download)
Appendix 26 Toxic deaths
Appendix 27 Treatment administration
Appendix 28 Quality of life
Appendix 29 Details of economic search strategies
Appendix 30 Details of probabilistic sensitivity analysis: hazard ratios
Details of probabilistic sensitivity analysis: hazard ratios (PDF download)
Appendix 31 Details of probabilistic sensitivity analysis: other variables
Details of probabilistic sensitivity analysis: other variables (PDF download)
Glossary
- Adenocarcinoma
- Cancer that begins in cells that line certain internal organs and that have glandular (secretory) properties.
- Chemo-naive (chemotherapy naive)
- Having received no prior chemotherapy treatment.
- Chemotherapy
- Treatment with anticancer drugs.
- Chemoradiation
- Combined chemotherapy and radiation therapy.
- Cost-effectiveness analysis
- Economic analysis that compares the costs and consequences (effects) of two or more courses of action. The consequences of the alternatives are measured in natural units, such as life-year(s) gained.
- Heterogeneity
- Variability or differences between studies in the estimates of effects.
- Histological diagnosis
- A diagnosis made by taking a sample of tissue or cells.
- Intention to treat
- A method of data analysis in which all patients are analysed in the group they were assigned to at randomisation regardless of treatment adherence.
- Locally advanced disease
- Stages IIIA/IIIB non-small cell lung cancer.
- Large cell carcinoma
- A group of lung cancers in which the abnormal cells are large.
- Meta-analysis
- A quantitative method for combining the results of many trials into one set of conclusions.
- Metastasis
- The spread of cancer from one part of the body to another. Tumours formed from cells that have spread are called ‘secondary tumours’ and contain cells that are like those in the original (primary) tumour.
- Metastatic disease
- Stage IV non-small cell lung cancer.
- Mixed-treatment comparison
- An indirect comparisons of data that allows for the ranking of different treatments in order of efficacy and estimation of the relative treatment effect of competing interventions.
- Non-small cell lung cancer
- A group of lung cancers that includes squamous cell carcinoma, adenocarcinoma and large cell carcinoma.
- Non-squamous cell carcinoma
- Includes adenocarcinoma and large cell carcinoma.
- Quality-adjusted life-year(s)
- An index of survival that is weighted or adjusted by a patient's quality of life during the survival period. Quality-adjusted life-years are calculated by multiplying the number of life-years by an appropriate utility or preference score.
- Relative risk
- The proportion of diseased people among those exposed to the risk factor divided by the proportion of diseased people among those not exposed to the risk factor.
- Relative risk (RR) reduction
- An alternative way of expressing relative risk. It is calculated as relative risk reduction = (1 − RR) × 100%. The relative risk reduction can be interpreted as the proportion of the baseline ‘risk’ which was eliminated by a given treatment or by avoidance of exposure to a risk factor.
- Squamous cell carcinoma
- Cancer that begins in squamous cells, which are found in the tissue that forms the surface of the skin, the lining of the hollow organs of the body, and the passages of the respiratory and digestive tracts. Also called epidermoid carcinoma.
List of abbreviations
- AC
- Appraisal Committee
- AE
- adverse event
- ASCO
- American Society for Clinical Oncology
- AUC
- area under curve
- BEV
- Bevacizumab
- BNF
- British National Formulary
- BSC
- best supportive care
- BTOG2
- British Thoracic Oncology Group Trial 2
- CARB
- Carboplatin
- CEA
- cost-effectiveness analysis
- CI
- confidence interval
- CIS
- Cisplatin
- CMA
- cost-minimisation analysis
- CRD
- Centre for Reviews and Dissemination
- CT
- computerised tomography
- CUA
- cost-utility analysis
- DOC
- Docetaxel
- ECG
- Electrocardiography
- ECOG
- Eastern Cooperative Oncology Group
- EGFR
- epidermal growth factor receptor
- EGFR-TK
- epidermal growth factor receptor-tyrosine kinase
- EMA
- European Medicines Agency
- eMIT
- electronic market information tool
- EORTC
- European Organisation for
- QLQ
- Research and Treatment of Cancer Quality of Life Questionnaire
- EQ-5D
- European Quality of Life-5 Dimensions
- ERG
- Evidence Review Group
- ERL
- Erlotinib
- EU
- European Union
- FACT-L
- Functional Assessment of Cancer Therapy – Lung questionnaire
- FDA
- US Food and Drug Administration
- GEF
- Gefitinib
- GEM
- Gemcitabine
- GP
- general practitioner
- HR
- hazard ratio
- HRG
- Healthcare Research Group
- HRQoL
- health-related quality of life
- ICER
- incremental cost-effectiveness ratio
- IPASS
- Iressa Pan ASian Study
- ITT
- intention to treat
- i.v.
- Intravenous
- KPS
- Karnofsky Performance Status scale
- LCSS
- Lung Cancer Symptom Scale
- LUCADA
- National Lung Cancer Data Audit
- LYG
- life-year gained
- LYS
- life-year saved
- MCMC
- Markov Chain Monte-Carlo
- MS
- manufacturer's submission
- MST
- median survival time
- M+
- mutation positive (EGFR)
- NEJGSG
- North East Japan Gefitinib Study Group
- NICE
- National Institute for Health and Care Excellence
- NSCLC
- non-small cell lung cancer
- ORR
- overall response rate
- OS
- overall survival
- PAX
- Paclitaxel
- PEM
- Pemetrexed
- PFS
- progression-free survival
- PLAT
- platinum (cisplatin or carboplatin)
- PPS
- postprogression survival
- PS
- performance status
- PSA
- probabilistic sensitivity analysis
- QALY
- quality-adjusted life-year
- QoL
- quality of life
- RCT
- randomised controlled trial
- RDI
- relative dose intensity
- RECIST
- Response Evaluation Criteria in Solid Tumours
- RR
- relative risk
- SACT
- Systemic Anti-Cancer Therapy
- STA
- single technology appraisal
- TKI
- tyrosine kinase inhibitor
- TNM
- tumour, node and metastasis
- TOI
- Trial Outcome Index
- TTP
- time to progression
- UICC
- Union for International Cancer Control
- VNB
- Vinorelbine
- WHO
- World Health Organization
- WJTOG
- Western Japan Thoracic Oncology Group
- WTP
- willingness to pay
All abbreviations that have been used in this report are listed here unless the abbreviation is well known (e.g. NHS), or it has been used only once, or it is a non-standard abbreviation used only in figures/tables/appendices, in which case the abbreviation is defined in the figure legend or in the notes at the end of the table.