3 Evidence

The appraisal committee considered evidence from a number of sources. See the committee papers for full details of the evidence.

Clinical need and practice

3.1 Ovarian cancer is a common gynaecological cancer which represents a group of different tumours arising from diverse types of ovarian tissue. The most common type arises from epithelial cells (the outside layer of cells), and can often spread from the ovary to any surface within the abdominal cavity, including the fallopian tubes and peritoneal cavity. Symptoms of ovarian cancer tend to be non‑specific and include persistent pelvic and abdominal pain, abdominal bloating, urinary frequency or urgency, loss of appetite, and abnormal or postmenopausal bleeding. Most women are diagnosed with advanced stage disease.

3.2 Recurrent ovarian cancer may be categorised according to the response to first‑line platinum chemotherapy as follows: platinum‑sensitive (disease that responds to first‑line platinum‑based therapy but relapses after 6 months or more – can be further subdivided into partially platinum‑sensitive disease that relapses between 6 and 12 months and fully platinum sensitive disease that relapses after 12 months or more); platinum‑resistant (disease that relapses within 6 months of completion of initial platinum‑based chemotherapy); and platinum‑refractory (disease that does not respond to initial platinum‑based chemotherapy). However, the 'partially platinum‑sensitive' and 'platinum‑resistant' categories should not necessarily be defined rigidly.

3.3 Ovarian cancer predominantly occurs in older women, with over 80% of cases being diagnosed in women over 50 years. In 2010, around 7,000 new cases of ovarian cancer were diagnosed and there were approximately 4,300 deaths from ovarian cancer in the UK. The overall 5‑year survival rate for ovarian cancer is approximately 43%. Although a significant percentage of women have ovarian cancer that responds to initial chemotherapy, between 55% and 75% relapse within 2 years of completing treatment with chemotherapy.

3.4 Fear of recurrence and subsequent treatment, particularly for women with platinum‑refractory disease, has an emotional impact. Recurrence of disease is associated with poorer prognosis and treatment options are limited. Treatment for recurrent ovarian cancer is also likely to diminish people's physical and emotional wellbeing to a point where they can no longer work, or need ongoing support with day‑to‑day activities. In NICE technology appraisal guidance 91 (to be replaced by the recommendations in this guidance), the recommended options for the second‑line (or subsequent) treatment of women with platinum‑sensitive or partially platinum‑sensitive advanced ovarian cancer were paclitaxel in combination with a platinum‑based compound (carboplatin or cisplatin), or pegylated liposomal doxorubicin hydrochloride (PLDH) for partially platinum‑sensitive ovarian cancer and for women allergic to platinum‑based compounds. The recommended second‑line (or subsequent) treatment options for women with platinum‑resistant or platinum‑refractory ovarian cancer, and for women allergic to platinum based compounds, were single‑agent paclitaxel, PLDH, or topotecan for women for whom PLDH and paclitaxel were considered inappropriate. NICE technology appraisal guidance 222 (also to be replaced by the recommendations in this guidance) did not recommend trabectedin in combination with PLDH for treating relapsed platinum‑sensitive ovarian cancer.

Clinical effectiveness

3.5 The assessment group carried out a systematic review and identified 16 randomised controlled trials, evaluating 14 different pairwise comparisons that met the inclusion criteria. Eleven of these trials were open label, and the masking technique was unclear in the remaining studies. The size of the trial populations ranged from 61 to 976 patients. The interventions in the trials were paclitaxel (6 trials), pegylated liposomal doxorubicin hydrochloride (PLDH; 5 trials), topotecan (3 trials), gemcitabine (1 trial) and trabectedin (1 trial). The assessment group stated that 5 of these trials evaluated the interventions and comparators within their licensed indications, dosage and routes of administration. The remaining 11 trials included dosages or routes of administration different from the relevant marketing authorisations. The population in 9 of the 16 trials was restricted to women experiencing their first recurrence. This included the main trials available for gemcitabine plus carboplatin and trabectedin plus PLDH, and 3 of the 4 trials for topotecan. One trial comparing weekly topotecan with 3‑weekly topotecan included women with platinum resistance who were experiencing subsequent recurrences. The assessment group stated that in general the trials were well designed and conducted. It expressed some concerns about the difference in baseline characteristics between trials, that masking and independent reviewer assessment in some trials was unclear, that some trials were not sufficiently powered to detect differences in overall survival and progression‑free survival, and that the results of some trials may have been confounded because patients crossed over between the intervention and control groups.

3.6 The assessment group determined that it was appropriate to analyse the results from patients with platinum‑sensitive disease and patients with platinum‑resistant or ‑refractory disease separately. Patients with platinum allergy were assumed to have the same probability of response to therapy as patients without an allergy for the same non‑platinum treatments, and therefore treatments for patients with platinum allergy were not analysed separately. The assessment group stated that there were insufficient data for most comparisons to carry out a standard pairwise meta‑analysis. Consequently, a series of network meta‑analyses were conducted for platinum‑sensitive disease, and platinum‑resistant or ‑refractory disease using a Bayesian Markov Chain Monte Carlo simulation. In the absence of individual patient data for all trials, the network meta‑analysis synthesised data on relative treatment effect from the whole study populations.

3.7 For patients with platinum‑sensitive disease, it was not possible to construct a complete network based on the trials identified, and therefore it was necessary to generate 2 discrete networks. Platinum‑sensitive network 1 evaluated platinum‑based treatments and platinum‑sensitive network 2 evaluated non‑platinum‑based treatments. The assessment group emphasised that these networks cannot be compared directly. For trials not limited to patients with platinum‑sensitive, ‑resistant or ‑refractory disease, results for the full trial population were presented, but the assessment group stated that these results were not synthesised in a network meta‑analysis because, in practice, patients with platinum‑sensitive, ‑resistant or ‑refractory disease would not have the same range of treatments and therefore the results of this analysis would not be clinically meaningful.

3.8 In general, the assessment group stated that treatment groups within trials were well matched. Some differences in baseline characteristics between trials were identified, in particular with respect to length of the platinum‑free interval, number of previous lines of chemotherapy and the method used to diagnose recurrence. However, the assessment group considered that the magnitude of these differences was unlikely to affect estimates of the relative effect of treatment and the trials were sufficiently clinically homogeneous to compare the clinical effectiveness of treatments. The assessment group clarified that the assessment of clinical homogeneity was limited to platinum‑sensitive network 1, which evaluated platinum‑based therapies. For this network, the assessment group considered that as a result of the imbalance in Eastern Cooperative Oncology Group (ECOG) status at baseline, the treatment effect associated with platinum may have been underestimated. Baseline characteristics were not reported for subgroups, and therefore an assessment of clinical heterogeneity was not possible for platinum‑sensitive network 2 evaluating non‑platinum‑based regimens, or for platinum‑resistant or ‑refractory groups, because both were informed by subgroup analyses. The assessment group also expressed concern that the subgroup data may not have been sufficiently powered to detect differences in overall survival or progression‑free survival. In addition, it was noted that statistical assessment of heterogeneity was not possible for either network, primarily because of the low number of trials identified.

3.9 The assessment group specified that unadjusted hazard ratios were used for progression‑free survival and overall survival in the network meta‑analysis. It acknowledged that adjusting for baseline characteristics may be important because certain characteristics are considered to influence prognosis. However, in the absence of a consistent dataset for all comparisons, the assessment group did not consider it appropriate to analyse a blend of unadjusted and adjusted hazard ratios.

Progression‑free survival

3.10 For platinum‑sensitive network 1 evaluating platinum‑based regimens, the assessment group included 5 trials evaluating progression‑free survival in the network meta‑analysis. Results from the network meta‑analysis found that paclitaxel plus carboplatin, gemcitabine plus carboplatin, and PLDH plus carboplatin statistically significantly improved progression‑free survival compared with platinum alone with hazard ratios (HR) of 0.73 (95% confidence interval [CI] 0.64 to 0.84), 0.71 (95% CI 0.57 to 0.90) and 0.59 (95% 0.50 to 0.71) respectively. PLDH plus carboplatin was found to be statistically significantly more effective at prolonging progression‑free survival than paclitaxel plus carboplatin (HR=0.81, 95% CI 0.71 to 0.92). No other statistically significant differences were identified between platinum‑combination regimens.

3.11 For the platinum‑sensitive network 2 evaluating non‑platinum‑based regimens, the assessment group included 3 trials evaluating progression‑free survival in the network meta‑analysis. Results found that trabectedin plus PLDH statistically significantly improved progression‑free survival compared with PLDH alone, paclitaxel alone and topotecan alone, with hazard ratios of 0.73 (95% CI 0.56 to 0.94), 0.44 (95% CI 0.26 to 0.82) and 0.55 (95% CI 0.38 to 0.82) respectively. No statistically significant differences were identified among the monotherapies evaluated (that is, PLDH, topotecan, and paclitaxel).

3.12 For the platinum‑resistant or platinum‑refractory ovarian cancer group, the assessment group included 3 trials evaluating progression‑free survival for inclusion in the network meta‑analysis. The assessment group also highlighted that trabectedin plus PLDH is outside of the scope for this subgroup, and although the data were included in the network to capture all the available evidence, they were not included in the economic analysis. Results from the network meta‑analysis found no statistically significant differences in progression‑free survival between PLDH, paclitaxel and topotecan alone, and these results were in line with results from the individual trials.

3.13 For the fully platinum‑sensitive ovarian cancer subgroup, the assessment group stated that although 3 trials (OVA‑301, ICON4/AGO‑OVAR, and a study by Pfisterer et al.) included subgroups with fully platinum‑sensitive ovarian cancer, only the OVA‑301 trial reported data, so it was not possible to perform an indirect comparison. In addition, 4 trials (OVA‑301, CALYPSO, ICON4/AGO‑OVAR and the study by Pfisterer et al.) included subgroups with partially platinum‑sensitive recurrent ovarian cancer, but only the OVA‑301 and CALYPSO trials reported data, and as they did not contain a common comparator it was not possible to make an indirect comparison. The OVA‑301 trial reported a statistically significant improvement in progression‑free survival with trabectedin plus PLDH compared with PLDH alone (HR=0.65, 95% CI 0.45 to 0.92; p=0.015). The CALYPSO trial reported a statistically significant improvement in progression‑free survival with PLDH plus carboplatin compared with paclitaxel plus carboplatin (HR=0.73, 95% CI 0.58 to 0.90; p=0.004).

Overall survival

3.14 For platinum‑sensitive network 1 evaluating platinum‑based regimens, the assessment group included 6 trials evaluating overall survival in the network meta‑analysis. Results indicated that PLDH plus carboplatin statistically significantly improved overall survival compared with platinum therapy alone (HR=0.79, 95% CI 0.64 to 0.97). Paclitaxel plus carboplatin was also found to statistically significantly improve overall survival compared with platinum alone (HR=0.77, 95% CI 0.66 to 0.91). No other statistically significant differences in overall survival were identified between platinum‑combination regimens.

3.15 For platinum‑sensitive network 2 evaluating non‑platinum‑based regimens, the assessment group included 4 trials evaluating overall survival in the network meta‑analysis. Results indicated that PLDH alone statistically significantly improved overall survival compared with topotecan alone (HR=0.73, 95% CI 0.56 to 0.97). Trabectedin plus PLDH was also found to statistically significantly improve overall survival compared with topotecan alone (HR=0.60, 95% CI 0.43 to 0.86). No other statistically significant differences were identified between platinum‑combination regimens.

3.16 For the platinum‑resistant or platinum‑refractory ovarian cancer group, the assessment group included 4 trials evaluating overall survival in the network meta‑analysis. The assessment group stated that trabectedin plus PLDH is outside of the scope for this subgroup and although the data were included in the network to capture all the available evidence, the data were not included in the economic analysis. Results from the network meta‑analysis found no statistically significant differences in overall survival among the treatments evaluated. This was in line with results from the individual trials.

3.17 For the fully platinum‑sensitive ovarian cancer group, the assessment group identified 4 trials evaluating overall survival. The assessment group stated that it was not possible to perform a network meta‑analysis because only 2 of the trials reported the necessary data for analysis and these trials did not have a common comparator.

3.18 For the partially platinum‑sensitive ovarian cancer group, the assessment group identified the same 4 trials evaluating overall survival. As before, the assessment group constructed 2 networks. Network 1, evaluating platinum‑based regimens, included only 1 trial (CALYPSO). No statistically significant difference in overall survival was identified for PLDH plus carboplatin compared with paclitaxel plus carboplatin in this trial. For network 2, evaluating non‑platinum‑based regimens, results indicated that trabectedin plus PLDH statistically significantly improved overall survival compared with PLDH alone (HR=0.84, 95% CI 0.667 to 1.032). Trabectedin plus PLDH was also found to statistically significantly improve overall survival compared with topotecan alone (HR=0.60, 95% CI 0.43 to 0.86).

Quality of life

3.19 Of the 16 trials identified, 10 reported data on quality of life. The most commonly used scale in the trials was the EORTC QLQ‑C30 questionnaire. However, the assessment group reported that there were considerable differences in the level of reporting of results, the questionnaires used to evaluate quality of life, and the time points for evaluation. Broadly, improvements in quality of life were identified for PLDH plus platinum compared with paclitaxel plus platinum; paclitaxel compared with oxaliplatin; and trabectedin plus PLDH compared with PLDH alone, in a subgroup of patients with partially platinum‑sensitive ovarian cancer.

Adverse reactions

3.20 The assessment group stated that the most frequently reported adverse reactions in the trials reflected those listed in the individual summaries of product characteristics. Consequently, based on advice from clinical experts, the assessment group limited its network meta‑analyses to the following severe grade 3–4 adverse events, which it considered to be the most problematic: allergic reaction, alopecia, anaemia, fatigue, febrile neutropenia, nausea/vomiting and neuropathy. In many cases a network meta‑analysis was not possible due to lack of available data. The majority of results, supplemented by the individual trial results when a network meta‑analysis was not possible, indicated that the likelihoods of adverse events were not statistically significantly different across treatment regimens. However, in some instances, chemotherapies were estimated as having statistically significantly lower risks of 1 or more adverse events but significantly higher risks of other adverse events. For example, when compared with paclitaxel plus platinum, PLDH plus platinum was associated with statistically significantly lower risks of allergic reaction and alopecia but statistically significantly higher risks of anaemia and nausea/vomiting. Overall, no chemotherapy was consistently associated with either a lower risk or a higher risk of the adverse events assessed.

Cost effectiveness

Company's model – trabectedin

3.21 The company for trabectedin submitted cost‑effectiveness evidence as part of its submission. The company developed a decision analytical model comparing trabectedin plus PLDH with PLDH alone in patients with recurrent platinum‑sensitive ovarian cancer for whom platinum‑based chemotherapy was not suitable because of allergy or intolerance or because they have partially platinum‑sensitive disease. The cohort only had 1 previous platinum‑based chemotherapy regimen, and experienced recurrence or progression.

3.22 The structure was identical to the model developed for NICE technology appraisal guidance 91. Disease was classified into 3 distinct periods: stable disease, progressive disease, and death.

3.23 The company stated that because the OVA‑301 trial was not powered for subgroup analysis within the platinum‑sensitive group, data for the entire platinum‑sensitive population were considered appropriate for the cost‑effectiveness analyses. The company fitted 5 parametric survival distributions, adjusting for potential covariates. Based on the Weibull distributions, the mean progression‑free survival for trabectedin plus PLDH and PLDH alone was 11.26 and 8.25 months respectively. Based on the log‑logistic distributions, the mean overall survival for trabectedin plus PLDH and PLDH alone was 44.69 and 34.97 months respectively.

3.24 The company's base‑case deterministic results, incorporating the patient access scheme for trabectedin, indicated incremental costs of £13,397 and incremental quality‑adjusted life years (QALYs) of 0.49 for trabectedin plus PLDH compared with PLDH alone, resulting in an incremental cost‑effectiveness ratio (ICER) of £27,573 per QALY gained. The corresponding probabilistic results indicated an ICER of £27,761 per QALY gained, and the ICER was most sensitive to the estimate of overall survival. Scenario analyses indicated that the results were also sensitive to the adjustment of the platinum‑free interval as an explanatory variable and alternative survival distributions for progression‑free survival and overall survival.

3.25 The company argued that trabectedin was eligible for consideration under the end‑of‑life criteria. It stated that trabectedin plus PLDH was indicated for women with a life expectancy of less than 2 years without treatment: for patients treated with PLDH alone, median overall survival in the platinum‑sensitive and partially platinum‑sensitive populations was 24.1 months and 16.4 months respectively. Accounting for the imbalance in platinum‑free interval and other prognostic factors in the platinum‑sensitive population reduced the median to 19.4 months. For women with platinum‑sensitive and partially platinum‑sensitive recurrent ovarian cancer, trabectedin treatment increased median survival (after correction of prognostic factors including progression‑free interval) by 4 months, and the estimated mean survival suggested this extension of life could be in excess of 9 months. The company also estimated that trabectedin would be indicated for approximately 500 women with relapsed platinum‑sensitive ovarian cancer in 2014.

Assessment group's model

3.26 The assessment group conducted a systematic review and stated that no cost‑effectiveness analyses including the full range of interventions and comparators were available in the literature. It noted that the majority of analyses available were based on the model developed for NICE technology appraisal guidance 91. The assessment group considered that this model structure, also adopted by the company for trabectedin, was the most appropriate for the decision problem, and therefore used it to develop a de novo model. The model had a lifetime time horizon, which was set as 15 years because at this point over 99.9% of patients in the model would have died. In NICE technology appraisal guidance 91, and other models based on it, the time spent in each health state was based on the estimated mean time to progression (time spent in the stable disease health state) and mean time to death (time spent in the progressed disease health state, after subtracting time spent in the stable disease health state). The assessment group incorporated a similar methodology to estimate the proportion of patients in each health state, but full survival curves rather than mean estimates were derived from the clinical data for each therapy. The assessment group stated that this would appropriately capture time in the economic model, and facilitate the assignment of costs, utilities and discounting.

3.27 The NICE scope for this appraisal specified that the interventions of interest for women with platinum‑sensitive ovarian cancer were paclitaxel alone or paclitaxel plus platinum chemotherapy, PLDH alone or PLDH plus platinum chemotherapy, gemcitabine plus carboplatin, trabectedin plus PLDH, and topotecan. The assessment group explained that although all interventions specified in the scope were considered, 2 independent networks were constructed evaluating platinum and non‑platinum‑based regimens, and therefore interventions were not simultaneously compared with each other.

3.28 The populations with platinum‑sensitive disease and platinum‑resistant or ‑refractory disease were modelled separately and there was no explicit analysis of the full population. The assessment group explained that this was because separation of the results by platinum sensitivity is more clinically relevant because the platinum‑free interval was a key prognostic factor, as confirmed by experts, and this approach was also in line with the data available to inform the analysis. The assessment group stated that data for women with fully or partially platinum‑sensitive disease was insufficient, so these groups were considered in sensitivity rather than base‑case analyses. The assessment group considered that response to non‑platinum‑based therapies would be expected to be consistent between patients with or without an allergy or intolerance to platinum‑based therapy. Therefore, the platinum‑allergic subgroup was included in platinum‑sensitive network 2 and platinum‑resistant and platinum‑refractory subgroups.

3.29 The assessment group noted 3 main concerns with the use of data from the network meta‑analyses in the model. First, due to lack of individual patient data, the network meta‑analyses synthesised data from the whole trial population. Individual patient data would have allowed for differences in baseline characteristics within and between trials to be incorporated. In addition, as discussed in section 3.9, unadjusted hazard ratios were incorporated, which could include potential bias. Second, using hazard ratios based on the literature assumes proportional hazards; that is, the relative treatment effects captured by the hazard ratios hold true across all time points. However, log‑cumulative hazard plots indicated that this assumption may not be appropriate. The assessment group highlighted that when the relative hazard decreases over time for both progression‑free survival and overall survival, the model was likely to overestimate the relative benefit of treatment and vice versa. Third, it was noted that several of the included trials allowed for crossover, which could have confounded overall survival data. The assessment group was unable to assess the degree of crossover bias because of a lack of individual patient data and because none of the trials described the crossover treatment.

3.30 The assessment group included grade 3 and 4 adverse events associated with significant costs in the base‑case analysis – allergic reaction, anaemia, febrile neutropenia, nausea and vomiting. The relative likelihood of an adverse event associated with each therapy was estimated from the network meta‑analysis. Adverse events were not analysed by population because of a lack of data; instead, adverse event data from any population (platinum sensitive or platinum resistant/refractory) were included in the analysis, therefore assuming that the likelihood of an adverse reaction is independent of the platinum‑free interval. Inconsistent reporting between trials led to differences in the networks of treatments available to assess the relative effect of treatment on each adverse event. Consequently, estimates of the impact of treatment on the rates of adverse events were not available for all treatments for all adverse events. Although it was possible to estimate the possibility of each adverse event for the baseline treatment in each network, odds ratios and expert opinion were used to estimate probabilities for the remainder.

3.31 The assessment group conducted a systematic review and identified 22 studies measuring health‑related quality of life. It was noted that the utility values based on the OVA‑301 trial were most relevant, because EQ‑5D utility values in the recurrent ovarian cancer population for the health states needed for the economic model were reported, and were based on a large sample of patients (n=600). The company clarified that these utilities were derived from the platinum‑sensitive population (n=400). The mean estimates of utility in the stable and progressive disease health states were estimated to be 0.718 and 0.649 respectively. These estimates were used in NICE technology appraisal guidance 222, and were identical to the EQ‑5D data identified by the assessment group from the systematic review of the literature. Disutilities associated with adverse events were not included in the base‑case analysis because the estimates identified were based on small samples. This was also to avoid double counting, because the effect of adverse events on quality of life associated with trabectedin plus PLDH and PLDH alone were already included in health state EQ‑5D estimates from NICE technology appraisal guidance 222. This was explored in sensitivity analyses.

3.32 The assessment group model included costs associated with the technologies, administration costs, health state‑related costs and adverse event costs. Chemotherapy costs per cycle were estimated using drug costs in the 'British national formulary' (BNF). The regimens used were as per the summaries of product characteristics (see section 2), except for PLDH, which was assumed to be started at a dose of 40 mg/m2 of body surface area in the base case (the licensed dose was considered in a scenario analysis) and paclitaxel, which was assumed to be used in a weekly regimen (80 mg/m2/week) in the platinum‑resistant or refractory subgroup only. These modifications were based on the advice of clinical experts and were intended to reflect the regimens used in clinical practice in the NHS.

3.33 The model assumed that treatments would be administered as infusions in a hospital, and associated administration costs were included in the model. For the base‑case analyses, it was assumed that no vial sharing would happen.

Results of network 1 – platinum‑based regimens in platinum‑sensitive disease

3.34 Both deterministic and probabilistic results indicated that PLDH plus platinum was strictly dominated by (that is, it was more costly and less effective than) paclitaxel plus platinum. Similarly, gemcitabine plus carboplatin was extendedly dominated by paclitaxel plus platinum (that is, its ICER was higher than that of the next, more effective, option when compared with platinum). Therefore, PLDH plus platinum and gemcitabine plus carboplatin were excluded, leaving paclitaxel plus platinum compared with platinum alone as the only relevant comparison for this network. For this comparison, the deterministic ICER was estimated as £24,361 per QALY gained; paclitaxel plus platinum was associated with an estimated incremental cost of £5,694 and an additional 0.23 QALYs when compared with platinum alone. The probabilistic ICER for paclitaxel plus platinum compared with platinum alone was £24,539 per QALY gained. The assessment group also estimated an ICER of £114,410 per QALY gained for gemcitabine plus carboplatin compared with platinum alone and an ICER of £30,188 per QALY gained for PLDH plus platinum compared with platinum alone.

3.35 One‑way sensitivity analyses on various model parameters indicated that the comparisons of paclitaxel plus platinum, and PLDH plus platinum, when compared with platinum alone, were most sensitive to the relative effect of treatment on overall survival. For example:

  • When the lower bounds of the hazard ratio for survival for gemcitabine plus carboplatin compared with paclitaxel plus platinum was used, the ICER for gemcitabine plus carboplatin compared with platinum alone was £23,578 per QALY gained. However when the upper bound was used, gemcitabine plus carboplatin was dominated.

  • When the hazard ratio for survival for platinum alone compared with paclitaxel plus platinum was used, gemcitabine plus carboplatin was dominated by platinum alone. When the lower bound of the hazard ratio for survival for gemcitabine plus carboplatin compared with paclitaxel plus platinum was used, paclitaxel plus platinum was less costly and less effective than gemcitabine plus carboplatin. When the upper bound was used, the ICER was £8,719 per QALY gained.

  • When the lower bound of the hazard ratio for survival for PLDH plus platinum compared with paclitaxel plus platinum was used, the ICER for PLDH plus platinum compared with paclitaxel plus platinum was £20,672 per QALY gained. When the upper bound was used, PLDH plus platinum was less costly and less effective than paclitaxel plus platinum.

    The assessment group stated that the impact of other parameters, such as the relative effect of treatment on progression‑free survival and the utility value associated with each health state, were relatively minimal.

3.36 Probabilistic sensitivity analyses indicated that at a maximum acceptable ICER of £20,000 per QALY gained, the probabilities of paclitaxel plus platinum or PLDH plus platinum being considered cost effective compared with platinum alone were 13% and 3% respectively. Furthermore, PLDH plus platinum was estimated to be almost as likely to result in greater costs and QALYs as to be dominated by paclitaxel plus platinum. The assessment group highlighted that the costs and QALYs accumulated by the addition of paclitaxel or PLDH to platinum therapy were similar, producing cost‑effectiveness estimates that were sensitive to minor changes in parameter estimates.

Results of network 2 – non‑platinum‑based regimens in platinum‑sensitive disease

3.37 Base‑case results (deterministic and probabilistic) indicated that topotecan was dominated by PLDH. Topotecan was therefore removed from the analysis and the relevant fully incremental comparisons of PLDH compared with paclitaxel and trabectedin plus PLDH compared with PLDH alone were presented. When compared with paclitaxel, PLDH was associated with an incremental cost of approximately £3,900 and approximately 0.16 additional QALYs. This resulted in ICERs of £23,733 and £25,931 per QALY gained in the deterministic and probabilistic analyses, respectively. When compared with PLDH alone, trabectedin plus PLDH was associated with an incremental cost of approximately £13,000 and 0.16 additional QALYs. The resulting ICERs for trabectedin plus PLDH compared with PLDH alone were £85,212 and £81,353 per QALY gained in the deterministic and probabilistic analyses respectively.

3.38 The assessment group did a series of one‑way sensitivity analyses on various model parameters. In network 2, the cost‑effectiveness estimates for all 3 comparisons (PLDH compared with paclitaxel, trabectedin plus PLDH compared with paclitaxel, and trabectedin plus PLDH compared with PLDH alone) were most sensitive to the relative effect of treatment on overall survival:

  • When the lower bound of the hazard ratio for overall survival for paclitaxel compared with PLDH was used, PLDH dominated paclitaxel, but when the upper bound was used the ICER for PLDH compared with paclitaxel was £15,900 per QALY gained.

  • When the lower bound of the hazard ratio for survival for trabectedin plus PLDH compared with PLDH alone was used, the ICER for trabectedin plus PLDH compared with PLDH alone was £44,266 per QALY gained. When the upper bound was used, PLDH alone dominated trabectedin plus PLDH.

  • When the lower bound of the hazard ratio for overall survival for trabectedin plus PLDH compared with PLDH alone was used, the ICER for trabectedin plus PLDH compared with topotecan was £18,437 per QALY gained. When the upper bound was used, the ICER was £30,754 per QALY gained.

  • When the lower bound of the hazard ratio for survival for topotecan compared with PLDH was used, the ICER for trabectedin plus PLDH compared with topotecan was £35,482 per QALY gained. When the upper bound was used, the ICER was £18,478 per QALY gained.

3.39 The assessment group did a series of scenario analyses and noted that the base‑case results were robust in the majority of the scenarios modelled, with the exception of the scenario in which PLDH monotherapy was assumed to be used at its full licensed dose (50 mg/m2 of body surface area). In this scenario topotecan remained dominated, the deterministic ICER for PLDH compared with paclitaxel increased from £23,733 to £31,222 per QALY gained and the deterministic ICER for trabectedin plus PLDH compared with PLDH alone fell from £85,212 to £77,290 per QALY gained. The assessment group stated that apart from this scenario, the ICER for PLDH compared with paclitaxel remained below £30,000 per QALY gained and highlighted that topotecan was dominated by trabectedin plus PLDH in every scenario. The assessment group also carried out an exploratory scenario analysis, using covariate‑adjusted clinical‑effectiveness data from the company for trabectedin's submission, for a head‑to‑head comparison of trabectedin plus PLDH compared with PLDH alone. This resulted in an ICER of £35,646 per QALY gained, compared with ICERs of £85,212 and £27,573 estimated by the assessment group's and the company's base‑case analyses respectively. The assessment group stated that this difference was predominantly a consequence of using adjusted clinical‑effectiveness data.

Results of network 3 – platinum‑resistant and platinum‑refractory group

3.40 The assessment group explained that data for paclitaxel plus platinum were not available from the literature for women with platinum‑resistant or ‑refractory ovarian cancer, so this intervention was not included in the base‑case analysis. Base‑case results showed that paclitaxel was dominated by PLDH alone. In the probabilistic analysis, paclitaxel was associated with an incremental cost of £901 and 0.022 fewer QALYs. Therefore, topotecan compared with PLDH was the only comparison considered in the final cost‑effectiveness analysis. Topotecan was associated with an incremental cost of approximately £7,000 and 0.02 additional QALYs relative to PLDH. The resulting ICERs for topotecan compared with PLDH were £449,553 and £324,188 per QALY gained in the deterministic and probabilistic analyses respectively.

3.41 As with platinum‑sensitive networks 1 and 2, the cost‑effectiveness results were most sensitive to the relative effect of treatment on overall survival:

  • When the lower bound of the hazard ratio for overall survival for paclitaxel compared with PLDH was used, the ICER for paclitaxel compared with PLDH was £17,904 per QALY gained. When the upper bound was used, paclitaxel was less costly and less effective.

  • For the incremental comparison of topotecan with paclitaxel, when the lower bound of the hazard ratio for overall survival for topotecan compared with PLDH was used, the ICER was £39,903 per QALY gained. When the upper bound was used, topotecan dominated paclitaxel.

  • When the lower bound of the hazard ratio for overall survival for paclitaxel compared with PLDH was used, topotecan dominated paclitaxel. When the upper bound was used, the ICER for topotecan compared with paclitaxel was £39,485 per QALY gained.

3.42 The assessment group did a series of scenario analyses and noted that the base‑case results were robust in the majority of the scenarios modelled. It highlighted that the ICER for topotecan compared with PLDH ranged from £374,963 to £503,885 per QALY gained across the scenarios. Paclitaxel was dominated in all scenarios except when the cost associated with a 50 mg/m2 dose of PLDH was used and paclitaxel became the least costly treatment, resulting in an ICER of £10,480 per QALY gained for PLDH compared with paclitaxel. In an additional analysis, done after the appraisal had been referred back to the committee after appeal, the assessment group modelled paclitaxel used at its licensed dose of 175 mg/m2 of body surface area every 3 weeks (instead of the weekly regimen used in the base case for the network 3 analysis). In this scenario, the costs associated with paclitaxel were lower and it was no longer dominated. The ICERs for PLDH, relative to paclitaxel, were £69,935 per QALY gained at a dose of 40 mg/m2 of body surface area and £103,810 per QALY gained at a dose of 50 mg/m2 of body surface area.

Additional analyses submitted by the company for trabectedin in response to consultation

3.43 Following consultation on the post‑appeal appraisal consultation document the company submitted an analysis that retrospectively adjusted survival outcomes, treating the platinum‑free interval as a categorical rather than a continuous variable. The analysis categorised patients with platinum‑sensitive disease into a partially platinum‑sensitive disease subgroup (platinum‑free interval 6–12 months) and a fully platinum‑sensitive disease subgroup (platinum‑free interval more than12 months). The company reported that the mean platinum‑free interval was balanced between the two arms of the trial in the partially platinum‑sensitive subgroup but that there was a 3.8 month (17%) difference favouring PLDH in the fully platinum‑sensitive group, which warranted adjusting progression‑free survival and overall survival by categorical platinum‑free interval strata.

3.44 In a Cox regression analysis comparing trabectedin plus PLDH with PLDH alone, adjusting for the platinum‑free interval as a categorical variable and covariates (age, race, ECOG performance score, the antigen CA‑125, prior taxane use, and liver or lung involvement), the hazard ratio for progression‑free survival was 0.68, 95% CI 0.52 to 0.90. Mean progression‑free survival was 11.19 months with trabectedin plus PLDH and 8.28 months with PLDH alone, an incremental improvement of 2.92 months. For overall survival, the hazard ratio was 0.78, 95% CI 0.62 to 0.99, and mean overall survival was 45.03 months with trabectedin plus PLDH and 36.68 months with PLDH alone, an incremental improvement of 8.36 months.

3.45 The company updated its economic model to adjust for the platinum‑free interval as a categorical variable, ECOG performance score and the antigen CA125. Its analyses also incorporated the updated patient access scheme (see section 2.14). This changed the ICER for trabectedin plus PLDH compared with PLDH alone from £27,572 per QALY gained to £28,573 per QALY gained.

Assessment group's comments on the additional evidence submitted by the company for trabectedin

3.46 The assessment group considered that the imbalance between the treatment groups in the platinum‑free interval observed in OVA‑301 did not warrant analysing the data by categorical variable. It also considered that the company's new analysis was another post‑hoc analysis and therefore subject to the same concerns as the analysis of the platinum‑free interval as a continuous variable. The assessment group commented that results from a second, independent study would be needed to demonstrate an association between duration of the platinum‑free interval and response to non‑platinum based treatments. It maintained that unadjusted hazard ratios should be used for survival outcomes because a consistent dataset of adjusted hazard ratios was not available to inform the network meta‑analysis.

3.47 The assessment group incorporated the updated patient access scheme (see section 2.14) into its model. The ICER for trabectedin plus PLDH compared with PLDH alone remained above £70,000 per QALY gained. When the assessment group also incorporated the company's hazard ratios that were adjusted for the platinum‑free interval as a categorical variable and covariates (see section 3.44), the ICER fell to £59,772 per QALY gained. Changing the dose of PLDH monotherapy from 40 mg/m2 to 50 mg/m2 further reduced the ICER to £54,059 per QALY gained.

  • National Institute for Health and Care Excellence (NICE)