3 The company's submission

The Appraisal Committee (section 7) considered evidence submitted by Gilead Sciences and a review of this submission by the Evidence Review Group (ERG; section 8).

Clinical effectiveness evidence

Systematic review

3.1 The company's systematic literature review identified 6 randomised controlled trials that were relevant to the decision problem because they included patients whose disease was relapsed or refractory. The company noted that none of the trials identified in the literature review compared idelalisib plus rituximab directly with the appropriate comparators identified in the NICE scope.

3.2 One single arm study was identified that investigated the effectiveness of idelalisib with rituximab in untreated chronic lymphocytic leukaemia in patients with a 17p deletion or TP53 mutation.

Previously treated chronic lymphocytic leukaemia (Study 116)

3.3 The company presented the results of Study 116, which was a phase III, double‑blind, randomised controlled trial across 90 centres in the US and Europe (including the UK). The study evaluated idelalisib plus rituximab compared with rituximab plus placebo in people with chronic lymphocytic leukaemia. A total of 220 patients were randomised to either idelalisib (150 mg oral tablets, twice daily) plus rituximab (375 mg/m2 at week 0, then 500 mg/m2 at weeks 2, 4, 6, 8, 12, 16 and 20) or rituximab (same dose) plus placebo (matching tablet, twice daily, until progression or withdrawal due to tolerability issues). Patients were included if they were aged 18 years or older, had previously had at least 1 treatment line (either an anti‑CD20 or 2 or more cytotoxic regimens) and had a reported Karnofsky performance score of 40 or more. Patients were excluded if their disease had a known histological transformation from chronic lymphocytic leukaemia to a more aggressive lymphoma.

3.4 The primary outcome of Study 116 was progression‑free survival, defined as the interval from randomisation to first documentation of definitive disease progression or death from any cause (whichever was sooner). Definitive disease progression was defined using the criteria from the International Workshop on Chronic Lymphocytic Leukaemia (IWCLL). Secondary outcomes included rates of overall response (complete and partial), lymph node response (defined as a decrease of 50% or more in lymphadenopathy) and overall survival. The primary and secondary end points were examined in pre‑specified subgroups, including: the presence or absence of a 17p deletion, TP53 mutation or both; sex (male or female); and age (less than 65 years or more than 65 years old). Health‑related quality of life was assessed using a change in domain and symptom scores from the Functional Assessment of Cancer Therapy: Leukaemia (FACT‑Leu) instrument, and using the EQ‑5D instrument. These were administered at baseline and at each study visit.

3.5 Patients could cross over from the rituximab plus placebo group to having idelalisib monotherapy in an extension study if their disease progression were confirmed by an independent review committee. The intention‑to‑treat analysis was done according to the treatment to which patients were randomised, and this included patients who had crossed over to the idelalisib monotherapy group. Progression‑free survival was calculated using the Kaplan–Meier method. A Cox proportional hazards model with adjustment for stratification was used to calculate hazard ratios.

Results of Study 116

3.6 The company stopped the blinded phase of the trial after the first interim analysis, because the 2‑sided p‑value for the primary progression‑free survival analysis crossed the prespecified alpha boundary of 0.001. All 110 patients in the idelalisib plus rituximab group and 107 of 110 in the rituximab plus placebo group had the assigned treatment. Of the 3 patients in the rituximab plus placebo group who did not have the study treatment, 2 withdrew from the study because of an adverse event before study treatment was started and 1 had not had the study treatment before the data cut‑off. The mean age of patients in the trial was 71 years. Between 41.8% and 44.5% of patients had a 17p deletion or TP53 mutation (or both), and 82.7% to 84.5% had an immunoglobulin variable region heavy chain non‑mutation.

3.7 The results showed a statistically significant improvement in median progression‑free survival for idelalisib plus rituximab compared with rituximab plus placebo of 19.4 months (95% confidence interval [CI] 12.3, not reported) compared with 6.5 months (95% CI 4.0 to 7.3). The reported hazard ratio (HR) was 0.15 (95% CI 0.09 to 0.24, p=<0.001). The intention‑to‑treat analysis for median overall survival showed a statistically significant difference for idelalisib plus rituximab compared with rituximab plus placebo (HR 0.34, 95% CI 0.19 to 0.60, p=<0.001).

3.8 In terms of secondary outcomes, the overall response rate was 83.6% for idelalisib plus rituximab compared with 15.5% for rituximab plus placebo. The odds ratio for overall response for idelalisib with rituximab compared with rituximab was 27.76 (95% CI 13.4 to 57.49). No patients in the trial had a complete response, meaning that the overall response rate was entirely made up of partial responders. The lymph node response rate was 96.2% in the idelalisib plus rituximab group compared with 6.7% in the rituximab plus placebo group.

3.9 The company also presented the results for the prespecified subgroups. In patients without a 17p deletion or TP53 mutation, median progression‑free survival was 19.4 months in the idelalisib plus rituximab group compared with 8.1 months in the rituximab plus placebo group. For people with a 17p deletion or TP53 mutation, progression‑free survival was not reached in the idelalisib plus rituximab group. In the rituximab plus placebo group, median progression‑free survival was 4.0 months.

3.10 Patients having idelalisib plus rituximab showed improvements in health‑related quality of life, with the EQ‑5D analysis showing a statistically significant treatment effect. The results of the FACT‑Leu questionnaire also showed that patients in the idelalisib plus rituximab group had greater symptom improvement than patients in the rituximab group at each time point throughout the trial.

3.11 The company reported that 15 patients had treatment‑emergent adverse events that led to death (4 having idelalisib plus rituximab, 11 having rituximab plus placebo). It noted that the causes of death were consistent with advanced chronic lymphocytic leukaemia and the underlying frailty, age, and poor prognosis of the study population.

3.12 The company reported that 32 patients – 19 (17.3%) of those having idelalisib plus rituximab and 13 (12.0%) of those having rituximab plus placebo – discontinued treatment because of an adverse event. Infections and infestations occurred in 11 of the 32 patients who discontinued (5 having idelalisib plus rituximab and 6 having rituximab plus placebo) and gastrointestinal disorders occurred in 6. Respiratory, thoracic, and mediastinal disorders accounted for a further 6 patients discontinuing because of adverse events (3 having idelalisib plus rituximab and 3 having rituximab plus placebo).

3.13 The company noted that the most commonly reported adverse events in the idelalisib plus rituximab group were: pyrexia (40.0%, 44 patients), fatigue (30.9%, 34 patients), diarrhoea (29.1%, 32 patients), nausea (27.3%, 30 patients) and neutropenia (25.5%, 28 patients). The most commonly reported adverse events in the rituximab plus placebo group were fatigue (33.3%, 36 patients), cough (31.5%, 34 patients) and infusion‑related reactions (30.6%, 33 patients).

Untreated chronic lymphocytic leukaemia with a 17p deletion or TP53 mutation (Study 101‑08)

3.14 Study 101‑08 was a phase II, single‑arm study of idelalisib plus rituximab in patients with untreated chronic lymphocytic leukaemia. A total of 64 patients were enrolled in centres across the US to have idelalisib (150 mg oral tablets, twice daily) and rituximab (375 mg/m2 for 8 weeks of continuous treatment). Only a small subset of patients (n=9) had a 17p deletion or TP53 mutation.

3.15 The primary outcome of the study was overall response rate, which was defined as the proportion of patients who achieved a complete or partial response as defined according to IWCLL criteria. Secondary outcomes included adverse events, progression‑free survival and overall survival.

Results of Study 101‑08

3.16 Of the 64 patients who were enrolled, 43 completed 48 weeks of treatment. The mean age of patients in the trial was 71 years. Only 9 of the 64 patients had a 17p deletion or TP53 mutation, and 37 had immunoglobulin heavy‑chain variable‑region non‑mutated chronic lymphocytic leukaemia.

3.17 At 36 months no patients with a 17p deletion or TP53 mutation had a progression event. Progression‑free survival for the overall population at 36 months was 83%, compared with 100% for the 17p deletion or TP53 mutation patients. Overall survival at 36 months was 100% for the 17p deletion or TP53 mutation patients and 90% for the whole study population.

3.18 The company noted that all 64 patients in the trial had 1 dose or more of the study drug. The most common treatment‑emergent adverse events were diarrhoea or colitis (77%), rash (58%) and pyrexia (42%).

Evidence for other comparators listed in the NICE scope

3.19 In addition to the comparison of idelalisib plus rituximab with rituximab alone, best supportive care and ofatumumab, the company also submitted evidence for the other comparator technologies listed in the NICE scope, namely fludarabine cyclophosphamide plus rituximab, bendamustine with or without rituximab, chlorambucil with or without rituximab, and steroids plus rituximab. The company's systematic review did not identify any evidence directly comparing idelalisib with these comparators, but identified 5 randomised controlled trials and 13 non‑randomised controlled trials in the relapsed or refractory population. One randomised controlled trial was identified in the untreated population.

ERG comments

3.20 The ERG commented on the population of Study 116 and its applicability to people with chronic lymphocytic leukaemia in the UK (of the 220 patients enrolled, 32 were UK patients). It noted that the trial included some patients (43.2%) with a 17p deletion or TP53 mutation; this type of disease does not respond to standard chemotherapy. The ERG noted that the baseline characteristics of patients in the trial represented a high‑risk patient cohort. The ERG also noted that the use of rituximab as a comparator was limited in its relevance to a UK population, because it is neither recommended by NICE nor by the British Committee on Standards in Haematology guidance.

3.21 The ERG noted that the results of Study 116 should be interpreted with caution because the trial was stopped early for benefit, and this type of stopping can lead to an overestimation of treatment effect.

Cost‑effectiveness evidence

Economic model structure

3.22 The company submitted a de novo economic model for the relapsed or refractory chronic lymphocytic leukaemia population only. The company did not submit an economic model for the untreated group because of the limited evidence available from Study 101‑08. The base‑case analysis modelled the following:

  • idelalisib plus rituximab compared with rituximab alone

  • idelalisib plus rituximab compared with best supportive care

  • idelalisib plus rituximab compared with ofatumumab.

3.23 The company included an additional exploratory analysis of idelalisib plus rituximab compared with the other comparators listed in the NICE scope:

  • fludarabine, cyclophosphamide and rituximab

  • bendamustine

  • bendamustine plus rituximab

  • chlorambucil

  • chlorambucil plus rituximab

  • steroids plus rituximab.

3.24 The company used a Markov model with time‑dependent transition probabilities. It used weekly cycle lengths (with half‑cycle corrections) with a time horizon of 25 years. The mean age of patients entering the model was 71 years. A discount rate of 3.5% was applied to costs and health benefits and the analysis was done from an NHS and personal social services perspective.

3.25 The base‑case analysis used data from Study 116. The model consisted of 5 health states, namely pre‑progression on treatment, pre‑progression off treatment, post progression, terminal care and death. The on treatment state was determined by the area under parametric time‑on‑treatment curves. The pre‑progression off treatment and post‑progression states were informed by the parametric survival curve analysis of progression‑free and overall survival data.

Clinical parameters and assumptions

3.26 To inform the clinical parameters for idelalisib plus rituximab compared with ofatumumab, the company assumed equal efficacy for rituximab and ofatumumab. The company explained that this assumption reflected the results of the ORCHARRD study, a network meta‑analysis in patients with diffuse large B‑cell lymphoma which found no difference in efficacy between ofatumumab and rituximab. The company also assumed equal efficacy for rituximab and best supportive care because there were insufficient data available to model this comparator.

3.27 To inform the model transition probabilities, the company extrapolated the overall survival data beyond the trial cut‑off. The company used the crossover‑adjusted overall survival data from Study 116 (patients could crossover from the rituximab plus placebo group to instead have idelalisib plus rituximab). The company used the Akaike Information Criterion statistic (an indication of the statistical fit between the observed Kaplan–Meier data and the parametric model estimates) to assess the most appropriate functional form. The results of the analyses showed that the exponential model provided the most appropriate fit, followed by the Weibull. The company explained that, on inspection of the model, 5% of patients were still alive after 20 years which it deemed inappropriate. The company therefore chose the Weibull model, as the next best fit, to extrapolate the overall survival data.

3.28 The company explained that progression‑free survival data did not need to be adjusted for crossover before extrapolation, because disease progression in the trial was the main reason for patients crossing over to the idelalisib plus rituximab group. The Akaike Information Criterion statistic suggested that the Weibull model was the most appropriate curve to use to extrapolate beyond the trial data.

3.29 The company used time‑on‑treatment data from Study 116 to estimate the drug resource use for idelalisib in the pre‑progression on treatment state. In the trial, idelalisib was indicated to be taken until disease progression or unacceptable toxicity, so the company noted that time on treatment followed a similar course to progression‑free survival. To extrapolate the data, it determined that a Cox proportional hazards model should be used to calculate a hazard ratio for time on treatment compared with progression‑free survival. This produced a hazard ratio of 1.31. It then applied this to the progression‑free survival curve for idelalisib plus rituximab to derive a time‑on‑treatment curve. For consistency, the company used the same hazard ratio to estimate a time‑on‑treatment curve for the rituximab plus placebo group of Study 116.

3.30 The company used the overall response rates reported in Study 116 to inform resource use assumptions in the model. The overall response rates were 84% in the idelalisib plus rituximab group and 15% in the rituximab plus placebo group. For the comparison with best supportive care, patients were assumed to have no overall response. For the comparison with ofatumumab, the overall response rate for rituximab plus placebo was applied.

Other comparators listed in the NICE scope

3.31 The company identified additional evidence from the literature for the other comparators listed in the NICE scope (table 1). It selected those studies which reported overall survival and progression‑free survival so it could extrapolate the data over the model time horizon. No studies were identified for the comparison with chlorambucil or chlorambucil plus rituximab, so the company used data from a published phase III trial (Knauf et al.) in patients with untreated chronic lymphocytic leukaemia and applied the reported hazard ratios to the bendamustine and bendamustine plus rituximab survival curves. The company assumed a shape parameter equal to that estimated for treatment arm data from Study 116, in which patients had idelalisib plus rituximab, to allow estimation of survival curves for the exploratory comparisons to external trial data. Treatment arm data were chosen because, unlike comparator arm data, these had not been confounded by crossover. The company validated the curves by visual inspection against the Kaplan–Meier data reported in the studies. It also adjusted for differences in baseline characteristics between the different studies.

Table 1 Results of the studies used to inform the company's additional comparator analysis

Treatment regimen

Median overall survival (months)

Median progression‑free survival (months)

Number of patients

Study design

Fludarabine, cyclophosphamide and rituximab

47

21

284

Open‑label, phase II study (Badoux 2011)

Bendamustine

44

20

49

Open‑label randomised controlled trial (Niederle 2013)

Bendamustine plus rituximab

34

15

78

Open‑label, phase II study (Niederle 2013)

Steroids plus rituximab

31

12

29

Single‑arm open‑label study (Pileckelyte 2011)

Ofatumumab

15

6

79

Single‑arm open‑label study (Wierda 2010)

Utility values and adverse events

3.32 The company used EQ‑5D data collected alongside Study 116 to inform the utility values for patients in the pre‑progression on treatment state. It used a generalised estimation equation regression to determine whether there was a difference in quality of life between the study groups. The company also assumed that utility values in the terminal care state were equal to those in the post‑progression state. However, no EQ‑5D trial data were collected for patients in the post‑progression or post‑treatment discontinuation states, so the company conducted a systematic literature review to identify studies reporting utility values for different chronic lymphocytic leukaemia health states. The company identified a range of studies, and chose to use Dretzke et al. (2010) for the post‑progression and pre‑progression off treatment states.

3.33 The company derived adverse event frequencies directly from Study 116. Those considered in the model were grade 3 or 4 events which occurred in at least 3% of patients in either treatment groups of Study 116.

Resource use and costs

3.34 The company used the time‑on‑treatment curves from Study 116 to estimate the length of time patients would have idelalisib plus rituximab and rituximab alone in the pre‑progression on treatment state. The company used the same dosing regimen from Study 116 for idelalisib plus rituximab and rituximab alone. For the other comparators, treatment costs were applied to the proportion of patients remaining in the progression‑free health states in the model, up to maximum treatment durations. The proportion of modelled patients who progressed before maximum treatment durations were not assumed to complete the full course of treatment.

3.35 Based on expert advice, the company assumed that intravenous immunoglobulin therapy would be used in 45% of non‑responders and 0% of responders in the pre‑progression health states. The company estimated that the cost of intravenous immunoglobulin therapy, incorporating the acquisition cost and administration of 5 weekly infusions, was £13,706. Active treatment was assumed to be every 3.5 weeks, based on British Committee for Standards in Haematology guidelines.

Company's base‑case results and sensitivity analyses

3.36 When using the simple discount agreement price for idelalisib, treatment with idelalisib plus rituximab was associated with higher costs and greater quality‑adjusted life year (QALY) gains compared with rituximab alone. The deterministic incremental cost‑effectiveness ratio (ICER) for idelalisib plus rituximab compared with rituximab alone was £13,634 per QALY gained (incremental costs £26,128; incremental QALYs 1.92). Compared with best supportive care, the ICER for idelalisib plus rituximab was £20,461 per QALY gained (incremental costs £39,211; incremental QALYs 1.92). Compared with ofatumumab, the ICER was £1527 per QALY gained (incremental costs £2926; incremental QALYs 1.92).

3.37 The company conducted a range of deterministic sensitivity analyses on the base‑case parameters. The results showed that the survival curve parameter estimates had the greatest influence on the results.

3.38 The company conducted a probabilistic sensitivity analysis for the base‑case parameters, presenting scatter plots and cost‑effectiveness acceptability curves for the 3 base‑case comparisons. The results showed that idelalisib plus rituximab was cost effective with a 90% probability compared with rituximab alone, an 80% probability compared with best supportive care, and a 100% probability compared with ofatumumab (at a maximum acceptable ICER of £30,000 per QALY gained). The mean probabilistic ICER for the comparison of idelalisib plus rituximab with rituximab alone was £13,680 per QALY gained. For idelalisib plus rituximab compared with ofatumumab, the mean probabilistic ICER was £1692 per QALY gained; for the comparison with best supportive care, it was £20,021 per QALY gained. The mean probabilistic ICER for the comparison of idelalisib plus rituximab with rituximab alone was £13,680 per QALY gained. For the comparison with ofatumumab, the mean probabilistic ICER was £1692 per QALY gained; for the comparison with best supportive care, it was £20,021 per QALY gained.

Company's exploratory scenarios

3.39 When using the simple discount agreement price for idelalisib, treatment with idelalisib plus rituximab was associated with both higher costs and greater QALY gains compared with:

  • fludarabine, cyclophosphamide and rituximab: £26,215 per QALY gained (incremental costs £63,232; incremental QALYs 2.41)

  • bendamustine: £36,424 per QALY gained (incremental costs £49,677; incremental QALYs 1.36)

  • bendamustine plus rituximab: £21,910 per QALY gained (incremental costs £35,910; incremental QALYs 1.64)

  • chlorambucil: £33,224 per QALY gained (incremental costs £55,471; incremental QALYs 1.67)

  • chlorambucil plus rituximab: £35,082 per QALY gained (incremental costs £66,267; incremental QALYs 1.89)

  • steroids plus rituximab: £17,106 per QALY gained (incremental costs £23,689; incremental QALYs 1.38)

  • ofatumumab (clinical effectiveness from literature): £4,254 per QALY gained (incremental costs £8232; incremental QALYs 1.96).

3.40 The company presented a subgroup analysis for patients in Study 116 with a 17p deletion or TP53 mutation. The results showed an increase in the ICERs for all 3 base‑case comparators: £20,200 per QALY gained for the comparison with rituximab alone, £27,543 per QALY gained for the comparison with best supportive care and £7066 per QALY gained for the comparison with ofatumumab.

ERG comments

3.41 The ERG noted the company's assumption that treatment effects continue beyond the trial. The ERG acknowledged that the treatment benefits of idelalisib may continue beyond the time horizon of the trial, but would be unlikely to continue for the rest of a patient's life. It noted that any reduction in treatment benefit following a discontinuation could result in a smaller treatment benefit for idelalisib with rituximab compared with the other regimens.

3.42 The ERG highlighted a possible issue with the company's methodology to adjust for crossover in the trial, noting that the company's analysis relies on the assumption that idelalisib alone has equal efficacy to idelalisib plus rituximab. It stated that a lower efficacy for idelalisib alone would result in a lower treatment benefit for idelalisib plus rituximab compared with rituximab and the magnitude of this change would only be substantial if idelalisib alone is considered inferior to idelalisib plus rituximab.

3.43 The ERG commented on the company's use of the constant shape parameter for the survival curves for the other treatments. It noted that the company could have fitted survival models directly using the digitised Kaplan–Meier plots that were generated. The ERG noted that this would improve the extrapolation for the fludarabine, cyclophosphamide and rituximab data, where the model (assuming a constant shape) was a poor fit for the data. In addition, the ERG questioned why the company had chosen to use hazard ratios from Badoux et al. to adjust for baseline differences in its extrapolations instead of the other studies identified in the literature review.

3.44 The ERG noted that in the company's base case, costs for idelalisib, rituximab and ofatumumab were accrued until treatment discontinuation, but for other comparators patients were assumed to complete the full maximum dosing indicated for that product. The ERG stated that a more realistic approach would be to use time‑on‑treatment data from Study 116 to estimate the proportion of the maximum number of doses actually administered for rituximab monotherapy, in the same way as was done for idelalisib, and apply the costs to these estimated time‑on‑treatment data.

3.45 The ERG noted that because no attempt was made to account for differences between the study populations and UK patients, the results of the company's analyses may be uncertain. In addition, it noted that the plausibility of the results may be affected by assuming that ofatumumab and best supportive care have equal efficacy to rituximab alone. Finally, the ERG stated that even though the methodology used to conduct the analyses for the other comparators in the NICE scope was less reliable than that used in the formal evidence synthesis, many of the comparators are used in the UK and therefore results from these analyses (with appropriately conservative assumptions) are important to understanding the cost‑effectiveness of idelalisib compared with other available treatments.

3.46 The ERG raised concerns with the assumptions relating to the frequency of resource use parameters, specifically about the number of patients whose disease did not respond to treatment being given intravenous immunoglobulin therapy. The ERG noted that this is important because the biggest difference in clinical outcomes between idelalisib plus rituximab and rituximab alone was the overall response rate. This meant that the clinical assumptions made in the model resulted in considerably higher disease management costs for patients having rituximab alone than those having idelalisib plus rituximab.

3.47 The ERG highlighted that utility values for patients who had discontinued treatment with idelalisib plus rituximab (taken from Dretzke et al.) in the pre‑progression states remained higher than those for patients having rituximab alone. It noted that this difference was more than could be explained by the adverse event disutilities calculated from Study 116, and meant that patients discontinuing idelalisib maintained a higher quality of life than those still having rituximab (an assumption not justified in the company submission)

ERG exploratory analyses

3.48 The ERG conducted an exploratory analysis changing the following parameters:

  • Using the Dretzke et al. utility values to inform both the pre‑progression and post‑progression model states, instead of those collected alongside Study 116 for the pre‑progression state.

  • Using time‑on‑treatment data for rituximab monotherapy to inform estimated drug costs rather than assuming that all patients completed the full course. For treatments outside Study 116, patients were assumed to take the same proportion of the maximum dosing duration as for rituximab alone in Study 116.

  • Using the statistically best fitting survival curve for fludarabine, cyclophosphamide and rituximab, rather than using the constant shape parameter with the curve for rituximab alone, as used in the company model.

  • Changing the length of treatment benefit for agents other than rituximab to 5 years.

3.49 Applying the simple discount agreement price for idelalisib, treatment with idelalisib plus rituximab was associated with higher costs and greater QALY gains compared with:

  • rituximab: £16,947 per QALY gained (incremental costs £24,335; incremental QALYs 1.44)

  • best supportive care: £26,058 per QALY gained (incremental costs £37,418; incremental QALYs 1.44)

  • ofatumumab (base case): £788 per QALY gained (incremental costs £1132; incremental QALYs 1.44)

  • fludarabine, cyclophosphamide and rituximab: £33,795 per QALY gained (incremental costs £71,177; incremental QALYs 2.11)

  • bendamustine: £52,815 per QALY gained (incremental costs £48,821; incremental QALYs 0.92)

  • bendamustine plus rituximab: £29,548 per QALY gained (incremental costs £34,921; incremental QALYs 1.18)

  • chlorambucil: £44,315 per QALY gained (incremental costs £53,779; incremental QALYs 1.21)

  • chlorambucil plus rituximab: £45,445 per QALY gained (incremental costs £64,893; incremental QALYs 1.43)

  • steroids plus rituximab: £24,065 per QALY gained (incremental costs £22,751; incremental QALYs 0.95)

  • ofatumumab (clinical effectiveness from literature): £5355 per QALY gained (incremental costs £8006; incremental QALYs 1.49).

3.50 The ERG did an additional analysis exploring the frequency of intravenous immunoglobulin therapy for patients whose disease responds to treatment compared with patients whose disease does not respond. In the company's model, 45% of patients whose disease does not respond had 1.24 cycles of intravenous immunoglobulin therapy. Patients whose disease does respond had none at all. The ERG explored increasing the number of cycles to responders and decreasing the number of cycles for non‑responders. The results showed that the ICER was sensitive to the changes; when responders had 0.5 cycles of intravenous immunoglobulin and non‑responders had 1.0 cycle, the ICER for idelalisib with rituximab compared with rituximab alone increased from £16,947 per QALY gained to £52,369 per QALY gained.

New evidence submitted by the company following consultation

3.51 Following a request by the Appraisal Committee in the appraisal consultation document (ACD), the company submitted a revised cost‑effectiveness analysis. The revised analysis compared idelalisib plus rituximab with rituximab alone, best supportive care and ofatumumab and incorporated the following changes to the base‑case analysis:

  • The overall survival benefit for idelalisib plus rituximab, predicted by extrapolating overall survival data from Study 116, was restricted to 5 years.

  • Using utility values from Dretzke et al. (2010) instead of Study 116 EQ‑5D data for all pre‑progression health states.

    In addition, the company provided sensitivity analyses:

  • exploring the effect of reducing treatment benefit with idelalisib from 5 years to 3 years

  • exploring the proportion of patients assumed to have intravenous immunoglobulin therapy

  • using clinical data from the subgroup of patients from Study 116 whose disease was refractory.

3.52 The company submitted 2 more economic analyses. The first, called the 'corrected base‑case analysis', included the changes requested in section 3.51 but made a correction to the dosing of intravenous immunoglobulin therapy. The company explained that in its original base case, the cost of intravenous immunoglobulin therapy was estimated assuming a dose of 0.4 g/kg administered 5 times for 1 active treatment week, and included administration costs for 5 infusions. However, the company noted that this assumption was based on the estimated dosing for diseases other than chronic lyphocytic leukaemia and the appropriate dosing for this condition, according to the Department of Health's clincal guidelines for immunoglobulin use, is 0.4 g/kg of therapy administered once.

3.53 The second additional analysis submitted by the company, called the 'proposed base case', also incorporated the requested analyses in section 3.51 and the changes in the dosage of intravenous immunoglobulin therapy as described in section 3.52, but reverted back to using Study 116 EQ‑5D data for patients in the pre‑progression model state.The company noted the ERG's comments on its original base‑case analysis (see section 3.47) which stated that patients in the pre‑progression model state who discontinued idelalisib treatment were assumed to maintain a permanently higher health related quality of life than those who had rituximab (until disease progression). The company addressed this issue in the proposed base case by assigning the utility from the rituximab arm of Study 116 to those patients discontinuing from idelalisib treatment.

3.54 The results of the proposed base‑case analysis, when using the simple discount agreement price for idelalisib, showed that treatment with idelalisib plus rituximab was associated with higher costs and greater QALY gains compared with rituximab alone. The deterministic ICER for idelalisib plus rituximab compared with rituximab alone was £26,403 per QALY gained (incremental costs £38,933; incremental QALYs 1.47). Compared with best supportive care, the ICER for idelalisib plus rituximab was £35,275 per QALY gained (incremental costs £52,016; incremental QALYs 1.47). Compared with ofatumumab, the ICER was £10,668 per QALY gained (incremental costs £15,731; incremental QALYs 1.47).

3.55 The results of the sensitivity analysis exploring the effect of reducing the proportion of non‑responders having intravenous immunoglobulin therapy to 20% and increasing the proportion of responders having the therapy to 20% showed the following:

  • For the comparison of idelalisib plus rituximab with rituximab alone, the ICERs ranged from £27,773 per QALY gained to £34,869 per QALY gained.

  • For the comparison of idelalisib with best supportive care, the ICERs ranged from £36,645 per QALY gained to £43,742 per QALY gained.

  • For the comparison of idelalisib with ofatumumab, the ICERS ranged from £12,038 per QALY gained to £19,134 per QALY gained.

3.56 The results of the sensitivity analysis exploring the effect on reducing the treatment benefit with idelalisib from 5 years to 3 years showed the following:

  • For the comparison of idelalisib plus rituximab with rituximab alone, the ICER ranged from £26,403 per QALY gained with a 5‑year limit to £33,850 per QALY gained with a 3‑year limit.

  • For the comparison of idelalisib with best supportive care, the ICERs ranged from £35,275 per QALY gained with a 5‑year limit to £45,674 per QALY gained with a 3‑year limit.

  • For the comparison of idelalisib with ofatumumab, the ICERs ranged from £10,668 per QALY gained for a 5‑year limit to £12,880 per QALY gained with a 3‑year limit.

ERG comments

3.57 The ERG reviewed the new evidence submitted by the company. It validated the company's revised base‑case analysis (see 3 .51) against the results it produced (sections 3.48–3.49). Although the results were not identical (£16,977 per QALY gained compared with £16,947 per QALY gained for idelalisib plus rituximab compared with rituximab alone), they were similar and the ERG had no concerns about the validity of the new results submitted.

3.58 The ERG reviewed the results of the proposed base case. It noted the changes to both the assumptions for intravenous immunoglobulin dosing (see section 3.52) and the changes to the utility values (see section 3.53), and considered them appropriate. However, the ERG highlighted that apart from the sensitivity analysis exploring the proportion of patients having intravenous immunoglobulin therapy, the company had continued to assume that 45% of non‑responders and 0% of responders have the therapy in all the analyses.

3.59 The ERG reviewed the results of the probabilistic sensitivity analysis for the comparisons in the proposed base‑case analysis. The results indicated that the proposed base case was highly sensitive to the cost at which treatments were considered to be cost effective. Although idelalisib plus rituximab was cost effective compared with rituximab alone in 97% of simulations at £50,000 per QALY, it was only cost effective in 62% and 12% of simulations at £30,000 and £20,000 per QALY respectively. The equivalent results for idelalisib plus rituximab compared with best supportive care were 88% at £50,000 per QALY, 22% at £30,000 per QALY and 0% at £20,000 per QALY.

3.60 The ERG conducted further analyses on the results of the company's proposed base case to generate ICERs for idelalisib plus rituximab compared with the other comparators listed in the NICE scope. The results of these analyses showed that the ICERs for idelalisib plus rituximab (using the simple discount agreement price for idelalisib) were:

  • £25,106 per QALY gained compared with fludarabine, cyclophosphamide and rituximab

  • £28,284 per QALY gained compared with steroids plus rituximab

  • £32,607 per QALY gained compared with bendamustine plus rituximab

  • £34,922 per QALY gained compared with chlorambucil plus rituximab

  • £44,302 per QALY gained compared with chlorambucil alone

  • £49,523 per QALY gained compared with bendamustine alone.

3.61 The ERG did an exploratory analysis to examine the effect on the ICERs (for the comparisons outside of Study 116) when changing the proportions of patients who had intravenous immunoglobulin therapy. The results of these analyses showed that the ICERs for idelalisib plus rituximab (using the simple discount agreement price for idelalisib) when 20% of non‑responders and 10% of responders had intravenous immunoglobulin therapy were:

  • £26,484 per QALY gained compared with fludarabine, cyclophosphamide and rituximab

  • £32,566 per QALY gained compared with steroids plus rituximab

  • £36,200 per QALY gained compared with bendamustine plus rituximab

  • £36,398 per QALY gained compared with chlorambucil plus rituximab

  • £48,074 per QALY gained compared with chlorambucil alone

  • £51,937 per QALY gained compared with bendamustine alone.

3.62 Full details of all the evidence are available.

  • National Institute for Health and Care Excellence (NICE)