Multiple myeloma - lenalidomide: appraisal consultation document

NATIONAL INSTITUTE FOR HEALTH AND CLINICAL EXCELLENCE

Appraisal Consultation Document

Lenalidomide for the treatment of multiple myeloma in people who have received at least one prior therapy

The Department of Health has asked the National Institute for Health and Clinical Excellence (NICE or the Institute) to conduct a single technology appraisal (STA) of lenalidomide and provide guidance on its use to the NHS in England and Wales. The Appraisal Committee has had its first meeting to consider both the evidence submitted by the manufacturer and the views put forward by non-manufacturer consultees and commentators, and by the clinical specialist and patient expert representatives nominated for this appraisal by non-manufacturer consultees and commentators. The Committee has developed preliminary recommendations on the use of lenalidomide.

This document has been prepared for consultation with the formal consultees. It summarises the evidence and views that have been considered and sets out the preliminary recommendations developed by the Committee. The Institute is now inviting comments from the formal consultees in the appraisal process (the consultees for this appraisal are listed on the NICE website, www.nice.org.uk). This document should be read in conjunction with the evidence base for this appraisal (the evaluation report) which is available from www.nice.org.uk

Note that this document does not constitute the Institute's formal guidance on this technology. The recommendations made in section 1 are preliminary and may change after consultation .

The process the Institute will follow after the consultation period is summarised below. For further details, see the ‘Guide to the single technology appraisal process' (this document is available on the Institute's website, www.nice.org.uk).

The Appraisal Committee will meet again to consider the original evidence and this appraisal consultation document in the light of the views of the formal consultees.

At that meeting, the Committee will also consider comments made on the document by people who are not formal consultees in the appraisal process.

After considering feedback from the consultation process, the Committee will prepare the final appraisal determination (FAD) and submit it to the Institute.

Subject to any appeal by consultees, the FAD may be used as the basis for the Institute's guidance on the use of the appraised technology in the NHS in England and Wales.

The key dates for this appraisal are:

Closing date for comments: 18 November 2008

Second Appraisal Committee meeting: 6 January 2009

Details of membership of the Appraisal Committee are given in appendix A, and a list of the sources of evidence used in the preparation of this document is given in appendix B.

Note that this document does not constitute the Institute's formal guidance on this technology. The recommendations made in Section 1 are preliminary and may change after consultation.
   
1 Appraisal Committee's preliminary recommendations
1.1 Lenalidomide in combination with dexamethasone is not recommended for the treatment of multiple myeloma in people who have received at least one prior therapy.
1.2 People currently receiving lenalidomide for the treatment of multiple myeloma should have the option to continue therapy until they and their clinicians consider it appropriate to stop.
   
2 The technology
2.1 Lenalidomide (Revlimid, Celgene) is an immunomodulating agent. It belongs to a class of agents often referred to as immunomodulatory derivatives (IMiDs), which are all structural derivates of thalidomide. The exact mechanism of action of lenalidomide is not understood but it has anti-neoplastic, anti-angiogenic and pro-erythropoietic properties. Lenalidomide in combination with dexamethasone is indicated for the treatment of multiple myeloma patients who have received at least one prior therapy. The recommended starting dose of lenalidomide for adults over 18 years is 25 mg orally once daily on days 1 to 21 of repeated 28-day cycles. Treatment with lenalidomide is continued until disease progression or unacceptable adverse effects occur.
2.2 The most serious adverse effects of lenalidomide treatment are grade 4 neutropenia and venous thromboembolism. The most frequently observed adverse reactions which occurred significantly more frequently in the lenalidomide/dexamethasone group compared to the placebo/dexamethasone group in clinical trials were neutropenia, fatigue, asthenia, constipation, muscle cramp, thrombocytopenia, anaemia, diarrhoea and rash. Lenalidomide is structurally related to thalidomide and there is a risk of teratogenesis. Pregnancy must be ruled out before starting treatment in women of child-bearing age and these women must use effective contraception while on lenalidomide. For full details of adverse effects and contraindications, see the summary of product characteristics.
2.3 Lenalidomide 25 mg capsules cost £4368 per 21 capsules (excluding VAT; ‘British national formulary' [BNF] edition 55). Dosage is continued or modified based upon clinical and laboratory findings.  For example, if lenalidomide is continued for ten 28-day cycles without dose reduction, the cost would be £43,680. Costs may vary in different settings because of negotiated procurement discounts.

 

3 The manufacturer's submission
  The Appraisal Committee (appendix A) considered evidence submitted by the manufacturer of lenalidomide and a review of this submission by the Evidence Review Group (ERG; appendix B).
3.1 The manufacturer produced an analysis of the clinical and cost effectiveness of lenalidomide for the treatment of multiple myeloma in people who had had at least one prior therapy. This included people at first and subsequent relapse or people who had progressive disease after at least two cycles of anti-myeloma treatment. The trial population was divided in to five subgroups for the economic analysis. For people who had received only one prior therapy the main comparator was bortezomib monotherapy which is recommended by NICE guidance (TA 129). For people in whom bortezomib was contraindicated, for people who had received more than one prior therapy and for people who had received prior thalidomide (only one or more than one prior therapy) the comparator was dexamethasone.
3.2 Two randomised controlled trials (RCTs), of identical design but differing in their locations (MM-009 and MM-010), compared treatment with lenalidomide plus dexamethasone (len/dex) with dexamethasone alone for people with multiple myeloma who had received at least one prior therapy. In both arms, the regimen of dexamethasone was pulsed high-dose dexamethasone in 28-day cycles. The trials enrolled 353 and 351 participants, respectively (n = 704). Participants were stratified according to their serum concentration of b2-microglobulin, previous stem cell transplantation and number of previous anti-myeloma therapies. Treatment was continued until disease progression or the occurrence of unacceptable adverse effects. The primary outcome was time to progression (TTP). Secondary outcomes were overall survival, response rates, adverse effects and time to decrease of performance status. Response was assessed using the European Group for Blood and Marrow Transplantation criteria, and six response categories were defined: complete response, near-complete response, partial response, stable disease, disease progression and ‘response not evaluable'. A number of post-hoc subgroups from the pooled populations were investigated, including people with pre-existing peripheral neuropathy and people who had received prior thalidomide or bortezomib therapy. At disease progression or unblinding, participants in the dexamethasone monotherapy group were allowed to receive lenalidomide.
3.3 The median TTP at unblinding from the pooled trials was 48.3 weeks (95% confidence interval [CI] 41.1 to 60.1 weeks) for the len/dex arms and 20.1 weeks (95% CI 19.9 to 20.7 weeks) in the dexamethasone arms. The pooled hazard ratio for TTP was 0.35 (95% CI 0.29 to 0.43; log-rank p < 0.001). The median overall survival in one trial, analysed 3 years and 3 months after study initiation, was 29.6 months in the len/dex arm and 20.2 months in the dexamethasone arm (hazard ratio 0.44; 95% CI 0.30 to 0.65; p < 0.001). In the second trial the median overall survival was analysed 2 years and 8 months after study initiation; it could not be estimated in the len/dex arm (because of the number of participants still alive), and was 20.6 months in the dexamethasone arm (hazard ratio 0.66; 95% CI 0.45 to 0.96; p = 0.03). For the pooled trials a complete, near-complete or partial response was obtained in 60.6% of participants in the len/dex arms and 21.9% of participants in the dexamethasone arms. The remaining 39.4% of participants in the len/dex arms and 78.1% of participants in the dexamethasone arms had stable or progressive disease, or were not evaluable. The odds ratio for this dichotomised response (complete, near complete or partial response versus stable disease, progressive disease or response not evaluable) was 5.48 (95% CI 3.94 to 7.63; p < 0.001).
3.4 The results for overall survival were affected by crossover of participants at unblinding: 170 of 351 participants in the dexamethasone arm opted to receive lenalidomide at disease progression or unblinding. However, these participants were analysed as remaining in the dexamethasone arm. The TTP was also affected by the crossover but to a lesser degree because most participants (over 75%) had shown disease progression at unblinding.
3.5 In both trials, the differences in TTP and response rates (in favour of len/dex) were observed in all the prespecified subgroups. The post-hoc subgroups in the trial showed that the efficacy of len/dex relative to dexamethasone alone remained statistically significant in subgroups that had received prior treatment with thalidomide or bortezomib and in subgroups specified by the number of previous therapies for multiple myeloma.
3.6 A meta-analysis was also performed to combine the results of the trials and to confirm the results obtained by the pooling of trials. This resulted in a median difference in TTP of 28.24 weeks (95% CI 18.39 to 38.08 weeks) and an odds ratio for overall survival of 1.44 (95% CI 1.34 to 1.56). The hazard ratio was not calculated. There was no evidence of heterogeneity between the trials.
3.7 An indirect comparison was undertaken to compare len/dex with bortezomib monotherapy because there were no head-to-head trials. The results of the trials for len/dex were compared with the results of the Assessment of Proteasome Inhibition for Extending Remissions (APEX) RCT. The APEX study compared bortezomib with high-dose dexamethasone. For median TTP, len/dex had a 34-week advantage over bortezomib for people who had one prior therapy only, and there were no statistically significant differences for the secondary outcomes of complete response, partial response and progressive disease. However, this analysis is limited by few data points. In addition, the common comparator (high dose dexamethasone) was an active treatment and was not used in the same dose across the trials, and the definition of response differed between the trials.
3.8 The economic evaluation in the manufacturer's submission used a discrete-event simulation model. This model used two separate prediction equations to calculate TTP and post-progression survival values, which were then added together to give overall survival. A cohort was created by randomly sampling (with replacement) participants from the pooled trial populations. For subgroups within the model, the cohort was created from the relevant population. The model attempted to capture the variability between individuals in the trial and allow correlation between observed parameters to be retained within the model.
3.9 The model divided participants from both arms of both trials into four groups according to their level of response. In building a cohort for the study population or any of the subgroups, the model ensured that the proportion of participants achieving a particular response in the trial was replicated in the cohort. To calculate TTP, the model assumed a Weibull distribution. For bortezomib, the response rates were taken from the APEX trial and the equation for TTP was calibrated such that the median TTP was the same as that within the trial.
3.10 The equation for post-progression survival was assumed to take an exponential form. However, the trial results were affected by the crossover of participants at unblinding from the dexamethasone arm to receive lenalidomide. Therefore the equation included an adjustment factor that calibrated the modelled median overall survival in the dexamethasone group post progression to be equal to that observed in UK Medical Research Council (MRC) multiple myeloma trials. This assumed that survival of people with multiple myeloma in this cohort was the same when treated with dexamethasone as with all other regimens used in the MRC trials. The participant profiles from the RCTs in the manufacturer's submission were applied to the predictors in the survival equations derived from the MRC trial data to predict survival in the dexamethasone arm if crossover had not occurred.
3.11 The model considered subgroups of participants who had received one prior therapy (with this group divided further into those who did and did not have peripheral neuropathy), participants who had received two or more prior therapies, and participants who had received thalidomide (divided further into those who had received only one prior therapy and those who had received more than one prior therapy). For participants who had received only one prior therapy, len/dex was compared with bortezomib monotherapy. For participants with peripheral neuropathy and for those who had received two or more prior therapies, the comparator was dexamethasone alone.
3.12 The utility values were based on a study that evaluated intensive chemotherapy followed by myeloablation and autologous stem cell transplantation in people with multiple myeloma. For the complete response, partial response and stable disease states, a utility value of 0.81 was used. This value was based on the utility of the general public at an age (median 54 years) corresponding to that of the participants in the study. A utility value of 0.64 was applied to the progressive disease state. After 2 years a utility value of 0.77 was applied to those participants whose disease had not progressed.
3.13 Only grade 3 and 4 adverse effects were included in the model. Utility decrements for adverse effects were not included. Resource-use data associated with, for example, adverse effects, routine follow-up and laboratory tests were collected to build up a profile of resource use, depending on disease state and treatment. Resource-use profiles were developed for people during relapse and/or on treatment, and for people in remission on maintenance therapy or off therapy. Resource use was estimated by interviewing 15 specialists across England and Wales who specialised in the management of multiple myeloma.
3.14 The economic analysis did not produce cost effectiveness estimates for the whole trial population. In the base case, for the subgroup with one prior therapy for which the comparator was bortezomib, the model resulted in an incremental cost-effectiveness ratio (ICER) per quality-adjusted life year (QALY) gained that the manufacturer stated was not cost effective. For this one prior therapy subgroup when compared with dexamethasone the ICER was £46,865 per QALY gained. For participants who had received more than one prior therapy the ICER was £24,584 per QALY gained. In the subgroup of participants who had received prior thalidomide, the ICERs were £38,861 per QALY gained for participants with only one prior therapy and £22,589 per QALY gained for participants who had received more than one prior therapy.
3.15 The ERG repeated the indirect comparison of len/dex with bortezomib with methods that it considered to be more appropriate. This resulted in a hazard ratio of 0.557 (95% CI 0.337 to 0.912). The ERG pointed out that this comparison was with bortezomib as monotherapy and that bortezomib was commonly used in combination with dexamethasone in routine clinical practice. The economic analysis for the comparison of len/dex with bortezomib also assumed a maximum of eight cycles of bortezomib instead of the 11 allowed in the trial, and did not model the response-based rebate scheme recommended in ‘Bortezomib monotherapy for relapsed multiple myeloma' (NICE technology appraisal guidance 129). The ERG also suggested that the administration costs associated with bortezomib may have been overestimated in the manufacturer's model. In addition, the dose intensity for bortezomib was assumed to be 100%; that is, the analysis did not allow for dose reductions and treatment interruptions, which had been included for lenalidomide. All of the above issues would have had the effect of increasing the ICERs for the comparison of len/dex with bortezomib in the one prior therapy only subgroup.
3.16 The ERG explored the precision with which the fitted curve for len/dex matched the actual trial data used in the model. It observed that the fitted curve overestimated overall survival. It also noted that the curve for the dexamethasone arm had been adjusted to predict the median overall survival in the MRC trials. However, the ERG stated that it was more methodologically correct to fit the dexamethasone curve to predict the mean overall survival in the MRC trials, because the ICERs calculated from the model were a ratio of means and not medians. The ERG conducted an exploratory analysis with an improved fit of the len/dex overall survival curve and also with the dexamethasone curve adjusted to the mean overall survival in the MRC trials. For the one prior therapy only subgroup where len/dex was compared with bortezomib, the ICER increased more than thirty-fold from the manufacturer's base case. For the one prior therapy only subgroup where the comparator was dexamethasone, the ICER increased from £46,865 to £69,500 per QALY gained. For the subgroup of participants who had received more than one prior therapy, the ICER increased from £24,584 to £47,100 per QALY gained. For participants who had received prior thalidomide, the ICER increased from £38,861 to £56,500 per QALY gained if they had received only one prior therapy and from £22,589 to £43,600 per QALY gained if they had received more than one prior therapy.
3.17 In addition, the ERG noted that the cost associated with routine medical management (non-drug costs) assumed in the model were lower than the figures that were accepted in the appraisal of bortezomib and may therefore have been underestimated. It also noted that the model also had no disutility attached to the occurrence of adverse effects and did not include the costs of granulocyte colony-stimulating factor used for management of the adverse effect of bone marrow suppression. Finally, it noted that the cost of anti-thrombotic prophylaxis that was routinely used with lenalidomide was not included in the model. The ERG stated that the inclusion of the above considerations in the model would increase the ICERs for all subgroups further from the exploratory reanalyses quoted above. Full details of all the evidence are in the manufacturer's submission and the ERG report, which are available from www.nice.org.uk/TAxxx

 

4 Consideration of the evidence
4.1 The Appraisal Committee reviewed the data available on the clinical and cost effectiveness of lenalidomide, having considered evidence on the nature of the condition and the value placed on the benefits of lenalidomide by people with multiple myeloma, those who represent them, and clinical specialists. It was also mindful of the need to take account of the effective use of NHS resources.
4.2 The Committee understood that multiple myeloma is an incurable disease. It was aware that the disease and its course were heterogeneous and that for relapsed multiple myeloma the choice of therapy for an individual patient is influenced by the initial treatment and the response to it, the inherent characteristics of the disease and the patient's performance status and preferences. The Committee heard from clinical specialists and patient experts that lenalidomide is an important advance in the treatment of multiple myeloma and could be considered as an alternative to bortezomib at first relapse and subsequent relapses. The Committee noted the importance that patients, their carers and physicians placed on having effective options to treat multiple myeloma at presentation and at subsequent relapses. However, it understood that the optimal sequence of agents to use was as yet unclear and would depend on several factors, including an individual's treatment history, comorbidities and disease characteristics.
4.3 The Committee understood that, in accordance with current NICE guidance (NICE technology appraisal guidance 129), bortezomib is routinely used in clinical practice for the treatment of progressive multiple myeloma in people who are at first relapse having received only one prior therapy. Therefore it considered bortezomib to be the most appropriate comparator for lenalidomide in people who have had only one prior therapy. The Committee noted that bortezomib is usually used in combination with dexamethasone, but that there is variation in clinical practice and limited formal evidence for the superior efficacy of this combination compared with bortezomib monotherapy. The Committee understood that a variety of regimens could be used at second and subsequent relapses, but that no studies had been identified that demonstrated the superiority of these compared with dexamethasone alone. Therefore it accepted that in people who have received more than one prior therapy, high-dose dexamethasone was a reasonable comparator for lenalidomide.
4.4 The Committee discussed the RCTs comparing len/dex with dexamethasone alone for the management of relapsed multiple myeloma. It noted that TTP (the primary outcome) was statistically significantly increased in the len/dex arm for the whole trial population as well as in subgroups of people who had received prior therapy with bortezomib or thalidomide. It considered that the RCTs provided evidence that overall survival and response rates were also higher with len/dex compared with dexamethasone alone. The Committee concluded that the len/dex combination improved outcomes in people with relapsed multiple myeloma when compared with dexamethasone. This included people who had received either one or more than one prior therapy, and when prior therapies included the use of thalidomide.
4.5 The Committee next discussed the relative effectiveness of len/dex compared with bortezomib. It noted that the evidence for the effectiveness of len/dex compared with bortezomib monotherapy was derived from an indirect comparison via the common comparator of high dose dexamethasone. It considered that there was uncertainty in the results of the indirect comparison due to heterogeneity between the studies used; for example differences in the regimen of dexamethasone and the definition of response. It noted that there was additional uncertainty in interpreting the context of current practice, because it understood that bortezomib was usually used with dexamethasone in clinical practice.
4.6 The Committee discussed adverse effects associated with lenalidomide. It noted that from the patients' viewpoint lenalidomide was associated with a more favourable adverse effect profile compared with most other regimens and agents used in the management of relapsed multiple myeloma. It heard from clinical specialists and patient experts that lenalidomide was particularly useful for people with pre-existing peripheral neuropathy in whom the use of bortezomib at first relapse was restricted. However, the Committee noted that lenalidomide was associated with a statistically significant increased risk of venous thrombosis and embolism. It heard from clinical specialists that this risk was usually managed with prophylaxis in the form of low-dose aspirin. However, in people with a history of venous thromboembolism or other relevant risk factors the use of warfarin or low-molecular-weight heparin would be considered. The Committee heard that with such prophylaxis the risk would return to baseline levels. The additional cost incurred for the management of people with multiple myeloma would be minimal if low-dose aspirin were used, but could have an impact if either low-molecular-weight heparin or warfarin were needed.
4.7 The Committee considered the manufacturer's economic evaluation of the use of lenalidomide and the critique from the ERG. It accepted that the general structure of the manufacturer's model was reasonable. It considered that the subgroups in the model were those relevant to decision-making in routine clinical practice. It discussed the sensitivity and scenario analyses presented by the manufacturer, as well as those explored by the ERG using the manufacturer's model. In particular, the Committee discussed the methods used for adjustment for the crossover effect in the RCTs, the extrapolation of survival data, the costs of medical management and administration of bortezomib therapy and the utility values reflecting health-related quality of life for the pre- and post-progression states and from adverse effects.
4.8 The Committee noted that the trial results included a crossover effect and considered whether it was appropriate to use data from historical MRC studies to account for the effects of crossover and to predict survival for people treated with dexamethasone in this population. The Committee was aware that the MRC data were derived from trials of agents in first-line therapy of multiple myeloma. Despite this, it accepted that these data represented the best available survival data for people with multiple myeloma to be used in extrapolation of overall survival in the current analysis. The Committee also noted that use of this data assumed that dexamethasone monotherapy was a suitable proxy (in the absence of more specific evidence) for all anti-myeloma therapies used in relapse. The Committee also considered that there was no evidence to indicate that the effectiveness of dexamethasone in relation to survival had changed over time since the MRC trial. It accepted the statements from the clinical specialists indicating that where improvements were noticed these were likely to be attributable to the use of the newer agents and stem cell transplantation.
4.9 The Committee considered the ERG's exploratory reanalysis with an improved fit of the len/dex overall survival curve to the trial data and the fitting of the dexamethasone overall survival curve to the mean (and not the median) overall survival in the MRC trials. The Committee considered that the ERG's approach to modelling overall survival in both the len/dex and dexamethasone arms was valid and resulted in more plausible estimates of cost effectiveness than those presented by the manufacturer. The Committee noted that these adjustments to the modelling of survival may have different effects in different subgroups and that the ERG's adjustments had been made separately to subgroups according to number of prior therapies.
4.10 The Committee considered the base-case ICERs resulting from the manufacturer's economic analysis, as well as the results of the ERG's exploratory analysis using the alternative approach to the modelling of overall survival. It noted that in the manufacturer's base case, none of the ICERs for the subgroups with only one prior therapy were within the range that would normally be considered a cost-effective use of NHS resources. The Committee noted that the comparison of len/dex with bortezomib in the one prior therapy only subgroup resulted in a particularly high ICER. The Committee also considered that the ICER for the comparison with bortezomib would increase further if the model took into account: the bortezomib response scheme (as described in NICE technology appraisal guidance 129); the lower costs of bortezomib administration suggested by the ERG; the higher maximum number of cycles of bortezomib; and the likely dosage reduction for bortezomib. When the ERG's approach to modelling overall survival was used, for the comparison of lenalidomide with dexamethasone in people who has received one prior therapy only, the ICER was more than £69,000 per QALY gained. For the comparisons with dexamethasone in people who had received one prior therapy, and where that therapy was thalidomide, the ICERs were above £56,000 per QALY gained.
4.11 The Committee considered the subgroups of people who had received more than one prior therapy, including the subgroup of people who had received prior thalidomide. When the ERG's approach to modelling overall survival was used, the ICERs for these subgroups increased to above £40,000 per QALY gained.
4.12 The Committee considered other issues with the base case analysis. The model did not fully include costs and utility decrements due to adverse effects, and the Committee considered that if appropriate costs and disutilities for adverse effects and anti-thrombosis prophylaxis were used in the model, the ICERs would increase. It also noted that the utility in the model for the pre-progression state was that of the normal population at age 54 and this was considerably younger than the average age of people who usually developed multiple myeloma. The Committee noted the results of the exploratory analysis by the ERG that using lower administration costs for bortezomib, using higher costs for routine medical management and modelling the bortezomib rebate scheme all had the effect of increasing the ICERs for the one prior therapy subgroup. The Committee concluded that in light of these additional issues the most plausible ICERs in all subgroups would be greater than those stated in 4.10 and 4.11.
4.13 The Committee discussed whether there were any further factors that would have a bearing on its considerations about the cost effectiveness of lenalidomide. These included the degree of certainty in the ICERs, the illness experienced by people with multiple myeloma who have received at least one prior therapy and the innovative nature of lenalidomide. It did not identify any factors that would alter its conclusions based on evidence currently available. Overall, the Committee concluded that the use of lenalidomide for the treatment of multiple myeloma in people who had received at least one prior therapy was not a cost-effective use of NHS resources.

 

5 Implementation
5.1 The Healthcare Commission assesses the performance of NHS organisations in meeting core and developmental standards set by the Department of Health in ‘Standards for better health' issued in July 2004. The Secretary of State has directed that the NHS provides funding and resources for medicines and treatments that have been recommended by NICE technology appraisals normally within 3 months from the date that NICE publishes the guidance. Core standard C5 states that healthcare organisations should ensure they conform to NICE technology appraisals.
5.2 'Healthcare standards for Wales' was issued by the Welsh Assembly Government in May 2005 and provides a framework both for self-assessment by healthcare organisations and for external review and investigation by Healthcare Inspectorate Wales. Standard 12a requires healthcare organisations to ensure that patients and service users are provided with effective treatment and care that conforms to NICE technology appraisal guidance. The Assembly Minister for Health and Social Services issued a Direction in October 2003 that requires local health boards and NHS trusts to make funding available to enable the implementation of NICE technology appraisal guidance, normally within 3 months.

5.3

 

NICE has developed tools to help organisations implement this guidance (listed below). These are available on our website (www.nice.org.uk/TAXXX). [NICE to amend list as needed at time of publication]

  • Slides highlighting key messages for local discussion.
  • Costing report and costing template to estimate the savings and costs associated with implementation.
  • Implementation advice on how to put the guidance into practice and national initiatives which support this locally.
  • Audit support for monitoring local practice.

 

6 Related NICE guidance
 

Published

  • Guidance on cancer services – improving outcomes in haematological cancers (2003). Available from www.nice.org.uk/Guidance/CSGHO
  • Bortezomib monotherapy for relapsed multiple myeloma. NICE technology appraisal guidance 129 (2007). Available from www.nice.org.uk/TA129

 

7 Proposed date for review of guidance
7.1 The review date for a technology appraisal refers to the month and year in which the Guidance Executive will consider whether the technology should be reviewed. This decision will be taken in the light of information gathered by the Institute, and in consultation with consultees and commentators.
7.2 It is proposed that the guidance on this technology is considered for review in October 2011. The Committee was not aware of any ongoing trials or impending results that would necessitate a review of the topic in less than the routine 3 years. The Institute would particularly welcome comment on this proposed date.

 

David Barnett
Chair, Appraisal Committee
October 2008
Appendix A: Appraisal Committee members and NICE project team

A. Appraisal Committee members

The Appraisal Committee is a standing advisory committee of the Institute. Its members are appointed for a 3-year term. A list of the Committee members who took part in the discussions for this appraisal appears below. The Appraisal Committee meets three times a month except in December, when there are no meetings. The Committee membership is split into three branches, each with a chair and vice chair. Each branch considers its own list of technologies, and ongoing topics are not moved between the branches.

Committee members are asked to declare any interests in the technology to be appraised. If it is considered there is a conflict of interest, the member is excluded from participating further in that appraisal.

The minutes of each Appraisal Committee meeting, which include the names of the members who attended and their declarations of interests, are posted on the NICE website.

Professor Keith Abrams
Professor of Medical Statistics, University of Leicester

Dr Ray Armstrong
Consultant Rheumatologist, Southampton General Hospital

Dr Jeff Aronson
Reader in Clinical Pharmacology, University Department of Primary Health Care, University of Oxford

Dr Darren Ashcroft
Reader in Medicines Usage and Safety, School of Pharmacy and Pharmaceutical Sciences, University of Manchester

Professor David Barnett (Chair)
Professor of Clinical Pharmacology, University of Leicester

Dr Peter Barry
Consultant in Paediatric Intensive Care, Leicester Royal Infirmary

Professor John Cairns
Public Health and Policy, London School of Hygiene and Tropical Medicine

Dr Mark Charkravarty
Director, External Relations, Procter and Gamble Health Care, Europe

Professor Jack Dowie
Health Economist, London School of Hygiene and Tropical Medicine

Dr Fergus Gleeson
Consultant Radiologist, Churchill Hospital, Oxford

Ms Sally Gooch
Independent Nursing and Healthcare Consultant

Mrs Eleanor Grey
Lay member

Professor Gary McVeigh
Professor of Cardiovascular Medicine, Queens University, Belfast

Dr Ruairidh Milne
Senior Lecturer in Public Health, National Coordinating Centre for Health Technology, University of Southampton

Dr Neil Milner
General Practitioner, Tramways Medical Centre, Sheffield

Dr John Pounsford
Consultant Physician, Frenchay Hospital, Bristol

Dr Rosalind Ramsay
Consultant Psychiatrist, Adult Mental Health Services, Maudsley Hospital, London

Dr Lindsay Smith
General Practitioner, East Somerset Research Consortium

Mr Roderick Smith
Finance Director, West Kent Primary Care Trust

Mr Cliff Snelling
Lay member

Professor Andrew Stevens
Professor of Public Health, Department of Public Health and Epidemiology, University of Birmingham

Dr Rod Taylor
Associate Professor in Health Services Research, Peninsula Medical School, Universities of Exeter and Plymouth

Ms Nathalie Verin
Health Economics Manager, Boston Scientific UK and Ireland

Dr Colin Watts
Consultant Neurosurgeon, Addenbrooke's Hospital

Mr Thomas Wilson
Director of Contracts and Information Management and Technology, Milton Keynes Primary Care Trust

B. NICE project team

Each technology appraisal is assigned to a team consisting of one or more health technology analysts (who act as technical leads for the appraisal), a technical adviser and a project manager.

Elangovan Gajraj
Technical Lead

Helen Chung
Technical Adviser

Shaun Minehan
Project Manager

Appendix B: Sources of evidence considered by the Committee
A

The Evidence Review Group (ERG) report for this appraisal was prepared by Peninsula Technology Assessment Group (PenTAG):

  • Hoyle M, Rogers G, Garside R et al, The clinical and cost-effectiveness of lenalidomide for multiple myeloma in people who have received at least one prior therapy: an evidence review of the submission from Celgene, September 2008.
B

The following organisations accepted the invitation to participate in this appraisal. They were invited to comment on the draft scope, the ERG report and the appraisal consultation document (ACD). Organisations listed in I were also invited to make written submissions. Organisations listed in II and III had the opportunity to give their expert views. Organisations listed in I, II and III also have the opportunity to appeal against the final appraisal determination.

I. Manufacturer/sponsor:

  • Celgene

II. Professional/specialist and patient/carer groups:

  • Leukaemia CARE
  • Leukaemia Research Fund
  • Macmillan Cancer Support
  • Myeloma UK
  • Royal College of Nursing
  • Royal College of Pathologists
  • Royal College of Physicians, Medical Oncology Joint Special Committee
  • Royal College of Radiologists
  • UK Myeloma Forum

III. Other consultees

  • Department of Health
  • Rotherham Primary Care Trust
  • Sandwell Primary Care Trust
  • Welsh Assembly Government

IV. Commentator organisations (did not provide written evidence and without the right of appeal)

  • Celgene/Pharmion
  • Department of Health, Social Services and Public Safety for Northern Ireland
  • GlaxoSmithKline
  • Janssen-Cilag
  • MRC Clinical Trials Unit
  • National Collaborating Centre for Cancer
  • National Coordinating Centre for Health Technology Assessment
  • NHS Quality Improvement Scotland
  • Peninsula Technology Assessment Group
  • Pfizer
  • Schering-Plough
C

The following individuals were selected from clinical specialist and patient advocate nominations from the non-manufacturer/sponsor consultees and commentators. They gave their expert personal view on lenalidomide by attending the initial Committee discussion and providing written evidence to the Committee. They are invited to comment on the ACD.

  • Mr Michael Brown, nominated by Myeloma UK – patient expert
  • Dr Jamie Cavenagh, Consultant Haematologist, Barts and the London NHS Trust, nominated by the UK Myeloma Forum –clinical specialist
  • Mr Eric Low, Chief Executive, Myeloma UK, nominated by Myeloma UK – patient expert
  • Dr Steve Schey, Consultant Haematologist, Kings College Hospital, nominated by National Cancer Research Institute (NCRI)/Royal College of Physicians (RCP)/Royal College of Radiologists(RCR)/Association of Cancer Physicians (ACP)/ Joint Collegiate Council on Oncology (JCCO) – clinical specialist

 

This page was last updated: 30 March 2010