elena nicod, panos kanavos developing an evidence-based...

12
Elena Nicod, Panos Kanavos Developing an evidence-based methodological framework to systematically compare HTA coverage decisions: a mixed methods study Article (Accepted version) (Refereed) Original citation: Nicod, Elena and Kanavos, Panos (2016) Developing an evidence-based methodological framework to systematically compare HTA coverage decisions: a mixed methods study. Health Policy, 120 (1). pp. 35-45. ISSN 0168-8510 DOI: 10.1016/j.healthpol.2015.11.007 Reuse of this item is permitted through licensing under the Creative Commons: © 2015 Elsevier Ireland Ltd. CC BY-NC-ND 4.0 This version available at: http://eprints.lse.ac.uk/64802/ Available in LSE Research Online: Online: February 2016 LSE has developed LSE Research Online so that users may access research output of the School. Copyright © and Moral Rights for the papers on this site are retained by the individual authors and/or other copyright owners. You may freely distribute the URL (http://eprints.lse.ac.uk) of the LSE Research Online website.

Upload: others

Post on 03-Sep-2019

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

Elena Nicod, Panos Kanavos

Developing an evidence-based methodological framework to systematically compare HTA coverage decisions: a mixed methods study Article (Accepted version) (Refereed)

Original citation: Nicod, Elena and Kanavos, Panos (2016) Developing an evidence-based methodological framework to systematically compare HTA coverage decisions: a mixed methods study. Health Policy, 120 (1). pp. 35-45. ISSN 0168-8510 DOI: 10.1016/j.healthpol.2015.11.007 Reuse of this item is permitted through licensing under the Creative Commons:

© 2015 Elsevier Ireland Ltd. CC BY-NC-ND 4.0 This version available at: http://eprints.lse.ac.uk/64802/ Available in LSE Research Online: Online: February 2016

LSE has developed LSE Research Online so that users may access research output of the School. Copyright © and Moral Rights for the papers on this site are retained by the individual authors and/or other copyright owners. You may freely distribute the URL (http://eprints.lse.ac.uk) of the LSE Research Online website.

Page 2: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

H

Dsm

EL

a

ARRA

KHEFSSMOCT

1

tncpc[

0

ARTICLE IN PRESSG ModelEAP-3484; No. of Pages 11

Health Policy xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Health Policy

journa l h om epa ge: www.elsev ier .com/ locate /hea l thpol

eveloping an evidence-based methodological framework toystematically compare HTA coverage decisions: A mixedethods study

lena Nicod ∗, Panos Kanavosondon School of Economics and Political Science, Social Policy, LSE Health, Houghton Street, WC2A 2AE London, UK

r t i c l e i n f o

rticle history:eceived 27 April 2015eceived in revised form 17 October 2015ccepted 24 November 2015

eywords:ealth technology assessmentnglandrancewedencotlandixed methodsrphan drugsomparative analysishematic analysis

a b s t r a c t

Health Technology Assessment (HTA) often results in different coverage recommendationsacross countries for a same medicine despite similar methodological approaches. This paperdevelops and pilots a methodological framework that systematically identifies the reasonsfor these differences using an exploratory sequential mixed methods research design. Thestudy countries were England, Scotland, Sweden and France. The methodological frame-work was built around three stages of the HTA process: (a) evidence, (b) its interpretation,and (c) its influence on the final recommendation; and was applied to two orphan medici-nal products. The criteria accounted for at each stage were qualitatively analyzed throughthematic analysis. Piloting the framework for two medicines, eight trials, 43 clinical end-points and seven economic models were coded 155 times. Eighteen different uncertaintiesabout this evidence were coded 28 times, 56% of which pertained to evidence commonlyappraised and 44% to evidence considered by only some agencies. The poor agreement ininterpreting this evidence (� = 0.183) was partly explained by stakeholder input (ns = 48times), or by agency-specific risk (nu = 28 uncertainties) and value preferences (noc = 62“other considerations”), derived through correspondence analysis. Accounting for variabil-

ity at each stage of the process can be achieved by codifying its existence and quantifying itsimpact through the application of this framework. The transferability of this framework toother disease areas, medicines and countries is ensured by its iterative and flexible nature,and detailed description.

. Introduction

Health technology assessment (HTA) is widely adoptedo inform coverage decisions of medicines or health tech-ologies by healthcare systems. It relies on evidence aboutomparative effectiveness of alternative treatments in a

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

articular clinical setting and aims to ensure that thoseovered provide value for money (or are cost-effective)1], ultimately, improving access to medicines. In practice,

∗ Corresponding author.E-mail address: [email protected] (E. Nicod).

http://dx.doi.org/10.1016/j.healthpol.2015.11.007168-8510/© 2015 Elsevier Ireland Ltd. All rights reserved.

© 2015 Elsevier Ireland Ltd. All rights reserved.

countries frequently issue different coverage recommen-dations despite appraising the same body of clinicalevidence and using similar methodological approaches.These differences are inevitable due to the complexity ofthese processes and the context within which they operate,where each country sets its own objectives for conductingHTA reflecting its values, preferences and constraints [2–4].Implications include uneven access to these medicinesacross (often neighbouring) countries, non-optimal use of

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

healthcare resources, and the unpredictability of the phar-maceutical market. Better understanding the applicationof HTA in different settings and the reasons for diverg-ing recommendations through cross-country learning and

Page 3: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

ING Model

ealth Po

ARTICLEHEAP-3484; No. of Pages 11

2 E. Nicod, P. Kanavos / H

sharing of expertise is high on European and supra-nationalagendas, and may contribute to identify ways to minimizethese differences [5,6] or understand how innovation wasrewarded [7,8]. This is all the more important given therecent appreciation of HTA as a means towards universalhealthcare [9] and the commitment of European Mem-ber States in implementing cross-border HTA collaborationthrough the EUnetHTA Joint Action 2.

Nine studies [10–18] compared HTA coverage recom-mendations for medicines in more than one country andidentified important variations, where agreement rangedfrom poor to moderate [10,11,13]. The countries comparedincluded Canada, Australia, England, Scotland, France,New Zealand, and other European countries. One studyconcluded that the most common reasons for differing rec-ommendations related to the HTA process and context [10].Another study highlighted cross-country variations for sev-enteen of the most expensive medicines, but the extent of,and reasons for these differences were not explored [12].A more recent study investigated oncologic medicines,where negative recommendations were largely due to thehigh costs outweighing the marginal benefits [14]. Possiblereasons for variations included differences in interpre-ting the clinical endpoints or in levels of patient input,or issues around appropriate comparators [14]. Anotherstudy highlighted differences across therapy areas andcountries, suggesting that preferences varied according tothe therapy area being appraised [13]. These studies have incommon the qualitative approach adopted (retrospectivedescriptive or cohort analyses) to identify these cross-country variations, highlighting possible reasons for thesethrough single case study analyses. None, however, haveattempted to scrutinize these variations and query whythey occur in a systematic manner. This is likely due todecision-making processes being complex with many fac-tors being accounted for, which may also be inter-relatedand thus challenging to compare. Comparing these deci-sion processes systematically could contribute to betterunderstanding the full range of factors accounted for anddetermining the extent to which they explain differencesin coverage recommendations. Doing so would require amethodological approach that decomposes these processesto identify the key drivers contributing to decision-makingin a systematic way. While this approach may not neces-sarily eliminate the variation observed in the criteria usedto arrive at decisions, reducing it considerably would alsobe beneficial.

The aim of this study is to develop and pilot such amethodological framework that allows for a compre-hensive and systematic identification and comparisonof the key factors that influence coverage decisions indifferent stages of HTA processes. A better understandingof value assessment processes may help address someof the methodological challenges in conducting HTA and,potentially, minimize cross-country differences whenthese were a consequence of the review or interpretationof the evidence.

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

The framework proposed in this study is informedby evidence from medicines with a European MedicinesAgency (EMA) orphan medicinal designation [19], whichhave undergone an HTA in different settings in Europe.

PRESSlicy xxx (2015) xxx–xxx

Orphan medicinal products are often characterized by sig-nificant inequalities in access [20] and are not alwayscost-effective [21]. In this context, a broader range of fac-tors are likely to be accounted for during the HTA process,which are to be captured by the proposed framework.

2. Methods

2.1. Study design

A sequential exploratory mixed methods researchapproach was used to develop and pilot the methodolog-ical framework in the form of an instrument developmentdesign (Fig. 1) [23]. Both the depth and breadth of theHTA decision process were captured within the qualitative(stages I and II) and quantitative strands (stage III) [22,23].A key characteristic of mixed methods design is the “itera-tive and cyclic approach used in the research” [24], wherean inductive logic was used in the qualitative strand inexploring and identifying the decision-making criteria, anda deductive position was used to test the hypothesis madeby means of this framework in order to draw inferencesfrom the findings in the qualitative strand [25]. Prioritywas given to outline specifically the steps achieved indesigning and piloting this methodological framework,while showcasing how the data collected can be analyzedquantitatively without drawing any conclusions due tothe small sample size.

2.2. Sampling

Purposeful sampling was used to select the studycountries with [27]: (a) well-established HTA agenciesand processes, (b) similar decision-making criteria (clin-ical and/or cost-effectiveness), (c) adopting differentapproaches in HTA (e.g. clinical benefit versus clinicalcost-effectiveness assessment, health service versus soci-etal approach), and (d) publicly available HTA reports. Thecountries included were England, Scotland, Sweden andFrance (Box 1).

Medicine and indication pairs were the unit of analysis.The two case studies used to develop the proposed method-ological framework were selected from all EMA approvedorphan medicinal products – until December 2012 – andappraised in the four study countries. Excluded were thosemedicines that: (a) did not undergo the single technologyassessment process at NICE, or the full submission processat SMC, and (b) did not receive diverging coverage rec-ommendations. Coverage recommendations were either tolist, restrict or reject the medicine under review, or in thecase of France, to issue a ranking of clinical benefit (ServiceMédical Rendu, SMR) defining the coverage decision andrate, and one of improvement in clinical benefit (Amélio-ration du Service Médical Rendu, ASMR) providing a basisfor the price fixing regime applicable, ranging from majorto insufficient. For example, a medicine receiving an ASMRV is considered not to provide any additional benefit and

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

is covered only if its price is inferior or equal to the othertreatments available.

This resulted in the selection of two compounds:eltrombopag (REVOLADE®) for the treatment idiopathic

Page 4: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

ARTICLE IN PRESSG ModelHEAP-3484; No. of Pages 11

E. Nicod, P. Kanavos / Health Policy xxx (2015) xxx–xxx 3

F rates thi data soufi

t(c

2

“icbtcvbspS

oieE

ig. 1. Visual model of the mixed methods research design used. Illustnterrelationships by distinguishing between the different methods and

nal stage.

hrombocytopenic purpura (ITP) and everolimusAFINITOR®) as second-line treatment for advanced renal-ell carcinoma (RCC) after failure of alternative therapies.

.3. Data sources and analysis

Stage I (qualitative strand) involved case studies used asan intensive study of a single unit for the purpose of defin-ng a larger class of similar units” [29], to determine theiromparability across countries. Previous research aiming atetter understanding HTA decision-making was also usedo outline the structure of the process [13]. Data sourcesomprised HTA recommendation reports and other rele-ant material published in the study countries and accessedy the authors. Although these materials adopt similartructures and outline the rationale for the decision, theirurposes may differ (e.g. legal document and memo inweden, summary of advice to the NHS in Scotland).

A case study template and coding manual were devel-

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

ped. The case study template provides tables for thenformation to be extracted and coded, to ensure it isasily understandable and homogeneous across countries.ach line item represents one criterion and the countries

e stages of this mixed methods study aiming to simplify the complexrces used to address the research questions and their integration in the

considering it, and includes identifiers to ensure cross-country comparability. The coding manual exhaustivelylists all the criteria included in the case study templates,organized into hierarchical levels clustered into commonthemes, referred to as first-order, second-order (clusteringfirst-order themes) and third-order (clustering second-order themes) themes (Appendix A).

Stage II (qualitative strand) aimed to systematicallycapture the criteria accounted for during the HTA processthrough thematic analysis [25] and determine the similar-ities and differences across countries. Bottom-up codingwas undertaken for each HTA report [30], where codeswere created while examining the data to summarize andcategorize the criteria identified within the case studies.The unit of coding, which is the section of text coded rep-resenting one criterion, was defined and illustrated withexamples to avoid confusion or duplication in the results[31]. Double-coding was performed to capture additionalinformation such as those cases where differences across

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

countries were seen, how these were dealt with, andwhether this influenced the final decision. For exam-ple, each uncertainty was double-coded with: (a) thoseagencies that raised the same concern, (b) whether the

Page 5: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

ING Model

ealth Po

ing an important medical benefit and providing an impor-

ARTICLEHEAP-3484; No. of Pages 11

4 E. Nicod, P. Kanavos / H

uncertainty was addressed and by what means (e.g.stakeholder input), and (c) whether it was one of themain reasons for the final outcome. Similar double-codingwas performed for the “other considerations” identified(Appendix A).

Coding and clustering of codes were performed by thelead author. Intra-coding reliability was tested to minimizecoding bias. Reliability of the clustering was tested by anexternal person, who re-categorised each individual codeinto one of these. Where differences were observed, adjust-ments were made. An iterative approach was adoptedthroughout the coding process to ensure that the iden-tified criteria captured the numerous dimensions of thedecision-making process. At the end of the coding period,all the information coded was reviewed to ensure thatthe codes reflect what was meant to be coded (withincase-comparison) across all codes (cross-case comparison).Primary data collection took place by means of the HTAreports summarizing the recommendations and solicitinginput from HTA experts and HTA body representatives toobtain additional insights about the decisions and ensurethat the criteria were coded accurately. The analysis wasperformed using QSR International’s NVivo10 [32].

In stage III, codes were quantitatively analyzed bothvertically and horizontally through descriptive exploratoryanalyses [30] to study their interrelationships. Thematic-matrixes summarizing the codes per medicine and countrywere exported from NVivo10 into Excel and transformedinto nominal variables. The statistical software STATA13was used for the analysis [33]. The vertical dimensionprovided findings about agency-specific risk and valuepreferences. “Risk” was derived from the concerns of theHTA bodies pertaining to uncertainty, and “value” fromthe “other considerations” relating to the disease andtreatment characteristics accounted for. Preferences wereexplored through correspondence analysis, where theassociations between the variables (HTA bodies versusuncertainty or “other considerations”) were measured andillustrated in correspondence analysis biplots [35,36]. Thehorizontal dimension provided a measure of agreementbetween the HTA bodies in interpreting the same evidenceusing Cohen’s kappa scores [37], allowing for a morerobust evaluation of qualitative findings by comparingobserved frequency of agreement with the probability ofagreement occurring by chance.

2.4. Study limitations

Whereas the objective of this study is to develop andpilot a framework, which would then be applicable to awider sample of medicines because of its iterative nature,it is not without limitations. One limitation is whether spe-cific aspects of the decision-making process, particularlythe context within which a decision was made, were cap-tured; these contextual considerations, however, were notwithin the scope of this study. A second limitation relates tothe purpose and level of detail provided in the HTA reports,

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

which varies by country. This is unlikely to have affectedthe results given that the key determinants, defined asthe main reasons for the final recommendations, wereincluded in all the reports and provide a good overview

PRESSlicy xxx (2015) xxx–xxx

of the decision criteria; data triangulation ensured suffi-cient detail was captured in each case, and comprised theHTA reports, other material and case studies, input fromHTA experts (e.g. Advance-HTA consortium, conferences),and interviews with HTA bodies where findings were pre-sented and feedback collected. A third limitation is that theframework relies on two case studies. However, these wereselected to proxy decision frameworks in orphan oncologyand non-oncology treatments because the ways of valuingthese may differ. The two cases are very different in termsof both disease and treatment characteristics, and thereforeare considered to appropriately cover different dimensionsof decision processes. Finally, the transferability of thisframework to other countries and therapy areas is limitedto those cases where similar decision-making criteria areaccounted for, from HTA entities that are arm’s length,responsible for issuing coverage recommendations, andhave a transparent process where sufficient detail aboutthe appraisal process and reasons for the final decision arerecorded in their decision reports.

3. Findings

The first two sections (qualitative strand) outline theinformation collected and coded based on the case studytemplate, showcasing how the proposed structure wasused to set up and pilot the methodological framework.Section 3.3 showcases how the data collected can be quan-titatively analyzed, where the case studies were used asillustrative examples and results are by no means general-izable due to the small sample.

3.1. Qualitative strand: Developing the methodologicalframework

The structure of the methodological framework wasbased on the three key stages of the decision-makingprocess identified: (a) the clinical and cost-effectivenessevidence, comprising clinical trials and endpoints, safety,economic models, and comparators, (b) the interpretationof this evidence, relating to uncertainty, “other consid-erations” and stakeholder input, and (c) the final HTArecommendation, and how the previous two stages influ-enced the recommendation formulation (Fig. 2). Thesecriteria formed the basis for the case study template andcoding manual, both tools forming the methodologicalframework (Appendix A).

3.2. Qualitative strand: Testing the methodologicalframework

Eltrombopag and everolimus received diverging HTArecommendations for the treatment of ITP and RCC, respec-tively. Eltrombopag was rejected in England, restricted inScotland to patients with severe symptomatic ITP or ahigh risk of bleeding, and listed in Sweden until its re-assessment in 2 years’ time. In France, it was valued as hav-

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

tant improvement in medical benefit (ASMR II). Everolimuswas rejected in England and Scotland, listed as applied forin Sweden, and considered to provide an important medicalbenefit in France with a low added benefit (ASMR IV).

Page 6: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

ARTICLE IN PRESSG ModelHEAP-3484; No. of Pages 11

E. Nicod, P. Kanavos / Health Policy xxx (2015) xxx–xxx 5

F procesm ely anala ined diff

3

fesegws

r3btorsn(fafAbocmHpt

cfic

ig. 2. Methodological framework for the systematic comparison of HTAethodological framework, together with the outcomes from quantitativ

nd horizontally (e.g. when differences at each stage of the process expla

.2.1. Clinical and cost effectiveness evidenceThe same phase III primary trials were considered

or both medicines. Additional trials were considered forltrombopag, one of which was an indirect compari-on with romiplostim appraised by all except HAS. Forverolimus, subgroup analyses of the primary trial by pro-nostic categories were also considered by SMC and HAS,hereas NICE additionally conducted a meta-analysis of 28

tudies (Table 1).The clinical endpoints from the same trials were

eported in a variety of ways (Table 1). For example, WHO–4 bleeding events were only recorded for eltrombopagy NICE, and health-related quality of life (HRQoL) relied onhe number of domains reported, where it was significantver four (as reported by SMC) but not over six domains (aseported by NICE). Results from indirect comparisons weretatistically significant across the whole population, butot significant when considering only patient subgroupsas reported only by NICE). A similar scenario was seenor everolimus, where HRQoL was not reported by TLVnd neither were objective response rate or progression-ree survival from the subgroup analysis by NICE or TLV.dverse events were reported by all agencies except TLV,ut this can be explained by the difference in purposef the TLV reports, which are of legal nature. The mostommon and clinically significant adverse events and treat-ent discontinuation rates were reported homogeneously.AS usually provided more detail about the percentage ofatients affected and deaths (even if not associated withhe treatment).

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

The clinical evidence identified included: (a) the clini-al trials, comprising eight trials (three primary trials) andve subgroup analyses with their respective comparators,oded 51 times across countries; (b) 43 different clinical

ses. Illustrates the three key stages identified and used as a basis for theysing the data collected both vertically (e.g. agency-specific preferences)erences in HTA recommendations across countries).

endpoints coded 68 times, and (c) the assessment of safety,recorded in a variety of ways and coded 22 times. Thisresulted in a total of 141 codes each representing anindividual criterion. For example, each trial was codedaccording to the type of trial (e.g. phase III, phase II),the type of comparator (e.g. placebo, standard care), andwhether it was a primary trial.

Economic models were considered by all HTA agen-cies except HAS. Those considered for eltrombopag differedfrom the comparator and/or type of model considered. NICEappraised three models with different comparators: (1)“watch and rescue”, (2) romiplostim, and (3) a sequenceof treatments. The watch and rescue model was consid-ered the most appropriate (reflecting clinical practice), andthe other two were rejected on the basis that they donot represent clinical practice (romiplostim at the timewas under review at NICE), or were not valid, respectively.In contrast, SMC considered a cost-utility model compar-ing eltrombopag to romiplostim in splenectomised andnon-splenectomised patients, and TLV a cost-minimizationanalysis with the same comparator based on a non-inferiority claim. For everolimus, the same cost-utilitymodels were considered. This resulted in a cost per QALYranging from £49,000 to £51,700 for NICE, £61,330 forSMC, and was not specified in TLV’s HTA report. The cost-effectiveness evidence consisted in the economic modeland its comparator, both coded 7 times (Appendix A).

3.2.2. Interpretation of the evidenceA total cumulative number of 18 clinical uncertainties

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

were raised by the agencies (10 for eltrombopag and 8 foreverolimus), coded 28 times (as some may have been raisedby more than one agency) (Table 2). 56% (of 18 uncertain-ties) were based on evidence commonly appraised by all,

Page 7: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

ARTICLE IN PRESSG ModelHEAP-3484; No. of Pages 11

6 E. Nicod, P. Kanavos / Health Policy xxx (2015) xxx–xxx

Table 1Clinical trials and their endpoints considered for eltrombopag and everolimus (non-exhaustive list).

Eltrombopag England NICE Scotland SMC Sweden TLV France HAS

RAISE Phase III, 6-month,placebo-controlled(N = 197)

Platelet response√ √ √ √

Primary endpointRescue treatment

√ √ √Bleeding (WHO1-4)

√ √ √ √Bleeding (WHO2-4)

√ √ √Clinically significant bleeding

Bleeding (WHO3-4) × Gross (grade 3) and debilitating(grade 4) blood loss

Main reduction in bleeding × √Seen in grade WHO2

HRQOL (SF36)√

4×6

√4 Significant over 4 domains, and

not over 6KUTER Indirect comparison

with RAISEPlatelet response

√NR NR NA Primary endpoint

Platelet response × Splenectomised patientsPlatelet response × Non-splenectomised patients

EverolimusRECORD-1 Phase III,

placebo-controlled(N = 416)

Progression-free survival√ √ √ √

Primary endpointOverall survival × × × × Blinded phaseHRQOL R* ×* ×** *EORTC, FKSI-DRS

**QLQ-C30Objective response × ×

Progression-free survival(subgroup analysis)

√ √Per risk stratification group

NICE: National Institute for Health and Care Excellence; SMC: Scottish Medicines Consortium; TLV: Dental and Pharmaceutical Benefits Board; HAS:Haute Autorité de Santé;

√: Statistically significant; ×: Non-statistically significant; R: reported; NR: not reported; NA: not applicable; EORTC: European

ssessm the dis

Organisation for Research and Treatment of Cancer; FKSI-DRS: Functional AQLQ-C30: quality of life questionnaire and the symptoms associated with

while the remaining 44% on evidence included in only someof the appraisal reports. 38% (of 18 uncertainties) were alsoput forward as one of the main reasons for the final recom-mendation.

Table 2 illustrates the different phases in interpretingthe clinical evidence. The first column (e.g. evidence, con-sidered by) reports, per agency, the evidence interpretedand whether it was primary evidence. The second col-umn (e.g. interpretation, raised by) reports cases when thisevidence was considered uncertain, and whether it wasnevertheless deemed acceptable or not (e.g. addressed ornot). The last column (e.g. outcome, main reason for rec-ommendation) reports cases when this issue was also oneof the main reasons put forward for the final recommen-dation.

This allowed to understand how these issues were dealtwith across settings. For example in the case of eltrom-bopag, the lack of direct comparative data was generallya concern and one of the main reasons for the final rec-ommendation for TLV and HAS, and the primary trial’ssmall sample size was a concern only for TLV, but con-sidered acceptable given the treatment’s orphan status.Another example is NICE’s concern that no improvementwas seen in the lower incidence of the most severe bleed-

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

ing events (WHO grades 3 and 4); this issue was not raisednor recorded by the other agencies either because the end-point was not specifically appraised or was not identifiedas being relevant to the decision. In the case of everolimus,

ent of Cancer Therapy Kidney Symptom Index-Disease Related Symptoms;ease; Primary endpoint in bold.

the primary trial was terminated early due to the superiorefficacy stopping rule, and results were biased due tocross-overs (81% of patients). Nevertheless, overall survivalestimates were deemed plausible for NICE based on clini-cal expertise and results from a meta-analysis, and for SMCand TLV based on a specific tool used to derive these esti-mates. In contrast for HAS, no benefit was demonstrated(Table 2).

A similar analysis about the interpretation of the eco-nomic models is possible in order to understand the typesof concerns raised by each HTA body, how these are compa-rable and dealt with. For example, the model appraised byNICE was considered highly uncertain and driven by costsrather than benefit. One of the main concerns was aboutthe assumption that differences in treatment arise becauseof bleeding events, which was used as the main effective-ness endpoint despite platelet response being the trial’sprimary endpoint. SMC was also concerned about the rela-tive risk of bleeding events, but with a different comparator(romiplostim).

A number of “other considerations” were identified inthe HTA reports and coded 62 times, differentiated bywhether they pertained to the treatment (e.g. type of ben-efit, innovativeness) or to the disease (e.g. severity, unmet

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

need) (Table 3). These may have been put forward as partof the reasoning for the final recommendation and/or mayhave been raised by different stakeholders (e.g. patients,clinicians). For instance, the oral administration benefit of

Page 8: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

Please

cite

this

article

in

p

ress

as:

Nicod

E,

K

anavos

P.

D

evelopin

g

an

eviden

ce-based

meth

odological

fram

e-w

ork

to

systematically

com

pare

H

TA

coverage

decision

s:

A

mixed

m

ethod

s

stud

y.

Health

Policy

(2015),

http

://dx.d

oi.org/10.1016/j.health

pol.2015.11.007

AR

TIC

LE

IN P

RE

SS

G M

odelH

EAP-3484;

N

o. of

Pages 11

E. N

icod, P.

Kanavos

/ H

ealth Policy

xxx (2015)

xxx–xxx

7

Table 2Differences and similarities in the interpretation of the clinical evidence and main reasons for recommendation.

Evidence considered by Interpretation uncertainty raised by Outcome main reason for recommendation√

Evidence considered√

*evidenceconsidered within the pivotal trial

√Positive influence

(addressed)× Negative influence(not addressed)

√Positive influence

(addressed)× Negative influence(not addressed)

Clinical uncertainties EnglandNICE

ScotlandSMC

SwedenTLV

FranceHAS

EnglandNICE

ScotlandSMC

SwedenTLV

FranceHAS

EnglandNICE

ScotlandSMC

SwedenTLV

FranceHAS

Eltrombopag Lack of comparator√

*√

*√

*√

* × × × × × ×Short duration of trial

√*

√*

√*

√* × × × ×

Sample size√

*√

*√

*√

*√ √

Trial population, indication underreview

√*

√*

√*

√* × ×

Trial population, generalizability√

*√

*√

*√

*√ ×

Trial population, low platelet countpatients instead of those with severerisk of bleeding

√*

√*

√*

√*

Significant bleeding events (WHO3-4)√

* × ×Quality of life estimate

√ √ × ×Liver function monitoring

√ ×Uncertain nature of the indirectcomparison

√ √√ √ × ×

Everolimus Bias in overall survival (cross-overs)√

*√

*√

*√

*√ √ √ √ √ √ √ ×

Weak overall and partial response√

*√

* ×Lack of comparative safety evidence

√ √ √ √ ×Trial population, patients withco-morbidities excluded

√*

√*

√*

√* ×

Trial population, generalizability√

*√

*√

*√

*√

Risk of pneumonitis,immunosuppression

√ √ √ √ √

Quality of life estimate√ √ √ ×

Risk stratification method notapplicable (subgroup analysis)

√ √ √ ×

NICE: National Institute for Health and Care Excellence; SMC: Scottish Medicines Consortium; TLV: Dental and Pharmaceutical Benefits Board; HAS: Haute Autorité de Santé.

Page 9: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

Please

cite

this

article

in

p

ress

as:

Nicod

E,

K

anavos

P.

D

evelopin

g

an

eviden

ce-based

meth

odological

fram

e-w

ork

to

systematically

com

pare

H

TA

coverage

decision

s:

A

mixed

m

ethod

s

stud

y.

Health

Policy

(2015),

http

://dx.d

oi.org/10.1016/j.health

pol.2015.11.007

AR

TIC

LE

IN P

RE

SS

G M

odelH

EAP-3484;

N

o. of

Pages 11

8

E. N

icod, P.

Kanavos

/ H

ealth Policy

xxx (2015)

xxx–xxx

Table 3“Other considerations” identified in the HTA reports.

Eltrombopag Everolimus

Subcategory Illustrative quotations/code England NICE ScotlandSMC

SwedenTLV

FranceHAS

England NICE ScotlandSMC

SwedenTLV

FranceHAS

Treatmentcharacteristics

Type of benefit Curative, life-extending√ √

End-of-life√

Innovativeness Innovative, new class of drugs√

Clinicians√

Main√

Clinicians/patientsAdverse effects Similar across treatment arms

√Patients willing to cope, manageable, tolerated,transient, reversible

√Patients

Administration Oral administration√

Main√

MainDisease

characteristicsUnmet need Unmet need, no treatment alternatives, few

alternatives, need for treatment options

√Clinicians

√Main

√Main

√ √Clinicians/patients

Nature of thecondition

Disease severity, serious condition√

Main√ √

MainLife-threatening

√Clinicians/patients

√Main

√Clinicians/patients/End-of-life

√ √

Short life-expectancy√

End-of-life√

Impact on quality of life, functional capacity,impact on daily activities

√Clinicians/patients

√Main

Social stigma, limiting of life-style choices,ability to work, travel or undertake leisureactivities, limiting life-style choices

√Clinicians/patients

Rare disease Orphan status, small population, rarity√

Main√

Main√ √

Clinicalpractice

No routine standard pathway, complex clinicalpractice, tailored to the patient

√Clinicians

√Clini-

ciansComparator unlicensed for indication, andassociated with important adverse events andanxiety

√Clinicians/patients

No long term evidence and unknown correctdosage of comparator

√Clinicians

Later diagnosis when disease advanced√

Licensed over non-licensed preferred√

CliniciansNationalpriority

Plan Maladies Rares (2004)√

NICE: National Institute for Health and Care Excellence; SMC: Scottish Medicines Consortium; TLV: Dental and Pharmaceutical Benefits Board; HAS: Haute Autorité de Santé; Main: considered one of the mainreason for the final recommendation; X: considered during the assessment; Clinicians: put forward by clinicians; Patients: put forward by patients; End-of-life: eligible as “end-of-life treatment” for NICE.

Page 10: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

ING ModelH

ealth Pol

edtpmcitt

rtidatn7ppa

3m

3

iwTbcoa

p0(eaacAnpotgsdcctOati

3

c

ARTICLEEAP-3484; No. of Pages 11

E. Nicod, P. Kanavos / H

ltrombopag was one of the main reasons for the finalecision by SMC and TLV, patients and clinicians statedhat adverse events are tolerable and manageable, and thatatients are willing to cope with them in order to get treat-ent. Another example is the life-threatening nature of the

ondition that was put forward by patients and cliniciansn the NICE appraisal for everolimus, and was also one ofhe criteria for recognising the treatment as an end-of-lifereatment.

Stakeholder input was identified 46 times in the NICEeports and twice in the SMC reports; none was iden-ified for TLV or HAS. For NICE, patient input providingnformation about “other considerations” (e.g. impact onaily activities) was seen in 30% of cases (14 out of 46),nd clinical input about “other considerations” (e.g. limitedreatment options) or commenting on clinical and eco-omic uncertainties (e.g. clinical practice) was seen in0% of cases (32 out of 46). For SMC, clinical expertsrovided input about “other considerations” (e.g. clinicalractice) and commented on uncertainty (e.g. generaliz-bility) (Table 3).

.3. Quantitative strand (Stage III): Outcomes from theethodological framework

.3.1. Clinical evidence and its interpretationReasons for differing HTA recommendations include

nstances when different evidence was considered, orhen the same evidence was interpreted differently.

able 2 illustrates cases when uncertainties were raisedased on different clinical evidence. For example, HAS wasoncerned about the lack of HRQoL data given the impactf the disease on the patient. This was not an issue for NICEnd SMC since it was included in the assessment.

When considering the same evidence, agreement wasoor using Cohen’s kappa scores (� = 0.183, 95% CI [0.015;.35]) [38]. These differences may relate to a subjectiveunexplained) component of the decision or to differ-nt risk or value preferences. Risk preferences, when onegency is relatively more likely to raise a concern whenppraising the same evidence compared to the other agen-ies, were identified through correspondence analysis.lthough the chi-squared probability of independence wason-significant given the small sample size (�2 = 22.49;

= 0.550), the results nevertheless provide an indicationf the relationships among these variables as well ashe type of analysis that this framework allows for on areater sample, illustrated in the correspondence analy-is biplot (Appendix B). Similarly, value preferences wereerived from the “other considerations” identified throughorrespondence analysis, and revealed a significant asso-iation in terms of the relative value preferences forhese two medicines (�2 = 30.97; p = 0.029) (Appendix C).ne example illustrating how preferences influenced thecceptability of an uncertainty was for eltrombopag, wherehe small sample size was deemed acceptable for TLV givents orphan status (Tables 2 and 3, Appendixes B and C).

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

.3.2. Criteria driving the decisionsThe criteria that drove the HTA decision-making pro-

ess consist of the main reasons for the recommendation

PRESSicy xxx (2015) xxx–xxx 9

identified at each stage of the process (Tables 2 and 3),potentially influenced by agency-specific risk or valuepreferences or because of the subjectivity in the interpreta-tion of the results (measured by agreement levels), whichresulted into the following decisions.

Eltrombopag was rejected by NICE mainly because ofthe high uncertainty that increased the ICER to a levelgreater than what is considered cost-effective. For SMC,although the clinical evidence was weak, eltrombopagwas significantly more effective than placebo in plateletresponse and considered cost-effective, as greater uncer-tainty in the economic analysis was accepted because itoffers additional treatment options, is an orphan medicinalproduct, and is administered orally. For TLV, eltrombopagwas considered cost-effective because of its similar effect ata lower cost compared to romiplostim, which had alreadybeen considered as cost-effective by the TLV. The orphanstatus, severity of the condition, and impact on the patient’sHRQoL were also put forward, with a follow-up of effective-ness to take place in October 2013. For HAS, while the trialduration was limited and comparative data was lacking,eltrombopag was considered similar to romiplostim untilfurther evidence is provided (risk assessment plan).

Because of the early termination of the trial, the esti-mated clinical benefit of everolimus was considered biased.For NICE, overall survival was considered superior to 3months and the treatment was eligible as “end-of-life treat-ment”. Despite this, sensitivity analysis showed only avery low probability of the medicine being cost-effectiveand was rejected. For SMC, the price was considered toohigh in comparison with the positive benefits provided. InSweden, the high cost per QALY was acceptable given thedisease’s severity. In France, despite being a serious andlife-threatening condition, the evidence presented was notsufficient to demonstrate any improvement in survival orHRQoL relative to alternatives.

4. Discussion and policy implications

4.1. Summary of key results

This empirical study fitted a mixed methods researchdesign to a research question requiring both an in-depthunderstanding of the HTA decision-making process anda systematic approach to comparing cases in order togain a broader understanding of the HTA outcome. Thecase study analyses highlighted the complexity of thesedecisions and identified a structure facilitating the under-standing and comparability of these processes. This wasused to derive and pilot the methodological framework,which divided the decision-making process into threestages within which a set of criteria were identified andcoded. This enabled to identify the criteria driving decisionprocesses and explaining cross-country differences.

4.2. How do our findings fit with existing evidence?

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

Comparing our results with existing studies that inves-tigated the criteria influencing HTA decisions in at leastone of the study countries (corresponding to the verti-cal dimension of this study), two studies were identified

Page 11: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

ING Model

ealth Po

ARTICLEHEAP-3484; No. of Pages 11

10 E. Nicod, P. Kanavos / H

and their findings are consistent with ours. One review ofquantitative studies identified existing empirical evidenceabout coverage decisions for a range of health technologies[39]. Despite not being directly comparable with our studygiven it included all types of technologies, it is of inter-est to ensure that the components of HTA included in ourstudy are comprehensive. Carroll et al. conducted a the-matic analysis of the assessments of medicines made bythe Evidence Review Group (ERG) at NICE to identify thestrengths and weaknesses in the submissions [40]. Eventhough it is again not directly comparable to our study, itremains of interest since it is accounted for by NICE and cor-responds to the “interpretation of the evidence” stage in ourstudy. Findings from these two studies validate our classi-fication of the decision-making criteria and confirm thatour results are appropriate and comprehensive. Focusingon the horizontal dimension included in our framework,only few comparative studies of HTA decisions exist, asreported in the introduction section. None, however, havecompared the decision-making processes in a systematicmanner.

4.3. The methodological framework

The added-value of this study is by deconstructing HTArecommendations and developing a taxonomy of criteriathat may have contributed to the decision-making process,it enables an enhanced understanding of HTA decision-making [41]. Without a mixed methods study design, wewould not have been able to capture the depth and com-plexity of these decision-making processes, both withinand across countries. The novelty of this methodologicalframework lies, first, in the systematic approach adoptedin analysing the data, and, second, in the inclusion of a hor-izontal dimension to capture additional aspects of the HTAdecisions. The coding and categorization of HTA documentsenabled a systematic identification of the decision-makingcriteria in a homogeneous and comparable manner acrosscountries and medicines. Further, the horizontal dimen-sion also captured, through double-coding, the influence(“positive” or “negative”) of each criterion on the final deci-sion (“main reason for recommendation”) and whether itwas provided through stakeholder input. Finally, this studyexemplifies how this type of design can be implementedto fit a specific research question and disseminated in aclear and transparent manner. It also highlights the inter-disciplinary potential of applying such designs to novelareas, such as HTA.

4.4. Policy implications

Based on its application to two cases, results show thata significant number of additional criteria and consider-ations may be used to inform decisions, which may overridepre-existing rules, such as an ICER threshold. This was aconsequence of the heterogeneity seen in the evidenceand its interpretation, or of additional criteria or input,

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

which may have influenced the decision. This may be due,in part, to the orphan nature of these medicines, whereaccounting for “other considerations” may overcome someof the uncertainty characterising the evidence generated

PRESSlicy xxx (2015) xxx–xxx

from small patient populations. It may also be interpretedeither as a need to examine in greater depth the avail-able evidence on specific medicine-indication pairs ratherthan stick to a yes–no decision based on otherwise inflex-ible rules, or as a recognition of the imperfect nature ofthe HTA process to account for detailed information thatmay matter when making a decision at the margin, or acombination of both. Results from applying this frameworkalso allow to raise awareness on the reasons for cross-country differences. When differences were a consequenceof the review, the interpretation of the evidence and deal-ing with uncertainty, it may contribute to finding solutionsto minimising these differences. When applied across agreater sample of medicines, therapy areas and countries,the application of this framework may be beneficial in avariety of ways: to identify decision-making criteria thatcan feed into other types of models (e.g. MCDA), to iden-tify agency-specific preferences, to understand the type ofstakeholder input meaningful to provide, or to ensure con-sistency in the “other considerations” accounted for (e.g.accountability for reasonableness).

5. Conclusion

Improving the level of access to medicines is a pri-ority at both European and supra-national levels [7,8].The substantial variations seen in HTA recommendationsis one of the causes of differential access to medicinesand possibly reflects weaknesses in HTA methodologies,where the implications are enormous in seeking to obtainvalue for money. The urgent need to better understandthe reasons for variations in decision-making processesand improve the quality of assessments is recognized. Inthis study, we have proposed, developed and piloted amethodological framework aiming to account for (partof) the unexplained heterogeneity in HTA recommen-dations across settings. The framework is detailed andprovides insights into decision-making practices in the casestudies concerned. The framework’s external validity iscurrently being enhanced and applied to a larger sampleof medicines, therapy areas and countries.

Conflict of interest statement

No conflicts of interest arise.

Acknowledgements

This research is funded under the European 7thFramework Programme with Advance-HTA. The resultspresented reflect the authors’ views and not the viewsof the European Commission. The EC is not liable for anyuse of the information communicated. The authors thankall partners from the Advance-HTA consortium for theirreviews, particularly (in alphabetical order) Karl Arnberg,Karen Brigham, Isabelle Durand-Zaleski, Jaime Espin, Karen

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

Facey, Andres Freiburg, Niklas Hedberg, Douglas Lundin,Lise Rochaix, Francis Ruiz, Domenica Taruscio, and ThomasWilkenson. We also thank Aude Bicquelet for her courseson qualitative research. Finally, we thank our colleagues

Page 12: Elena Nicod, Panos Kanavos Developing an evidence-based ...eprints.lse.ac.uk/64802/1/__lse.ac.uk_storage_LIBRARY_Secondary_libfile_shared... · approaches in HTA (e.g. clinical benefit

ING ModelH

ealth Pol

Pr

A

fj

R

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[[

[

[

[

[

[

[analysis of the strengths and weaknesses of manufacturers’ sub-

ARTICLEEAP-3484; No. of Pages 11

E. Nicod, P. Kanavos / H

aul Dolan, Emilie Courtin and Alessandra Ferrario foreviewing this work and providing valuable feedback.

ppendix A. Supplementary data

Supplementary data associated with this article can beound, in the online version, at http://dx.doi.org/10.1016/.healthpol.2015.11.007.

eferences

[1] Luce B, Drummond M, Jonsson B, Neumann P, Schwartz J, Siebert U,et al. HTA, and CER: clearing the confusion. The Milbank Quarterly2010;88:256–76.

[2] Banta D. The development of health technology assessment. HealthPolicy 2003;63:121–32.

[3] Schwarzer R, Siebert U. Methods, procedures, and contextual charac-teristics of health technology assessment and health policy decisionmaking: comparison of health technology assessment agencies inGermany, United Kingdom, France, and Sweden. International Jour-nal of Technology Assessment in Health Care 2009;25:305–14.

[4] Busse R, Orvain J, Velasco M, Perleth M, Drummond M, Gurtner F,et al. Best practice in undertaking and reporting health technologyassessments—working group 4 report. International Journal of Tech-nology Assessment in Health Care 2002;18:361–422.

[5] Commission E. Directive 2011/24/EU on patients’ rights in cross-border healthcare; 2011.

[6] European Commission, DG Health and Consumers, Public Health.Pharmaceutical pricing and reimbursement information sheet; 2014.

[7] European Commission, Enterprise and Industry, PharmaceuticalForum. Final conclusions and recommendations of the Pharmaceuti-cal Forum: improving access to orphan medicines for all affected EUcitizens; 2013.

[8] Bouvy J, Vogler S. Update on 2004 background paper, BP 8.3 pricingand reimbursement policies: priority medicines for Europe and theworld a public health approach to innovation. WHO CollaboratingCentre for Pharmaceutical Policy and Regulation; 2013.

[9] World Health Organisation. Health intervention and technol-ogy assessment in support of universal health coverage; 2014,http://apps.who.int/gb/ebwha/pdf files/WHA67/A67 R23-en.pdf.

10] Clement F, Harris A, Li J, Yong K, Lee K, Manns B. Using effectivenessand cost-effectiveness to make drug coverage decisions a compar-ison of Britain, Australia, and Canada. JAMA—The Journal of theAmerican Medical Association 2009;302:1437–43.

11] Lexchin J, Mintzes B. Medicine reimbursement recommendations inCanada, Australia, and Scotland. American Journal of Managed Care2008;14:581–8.

12] Morgan S, McMahon M, Mitton C, Roughead E, Kirk R, Kanavos P,et al. Centralized drug review processes in Australia, Canada, NewZealand, and the United Kingdom. Health Affairs 2006;25:337–47.

13] Nicod E, Kanavos P. Commonalities and differences in HTA outcomes:a comparative analysis of five countries and implications for coveragedecisions. Health Policy 2012;108:167–77.

14] Shah K, Mestre-Ferrandiz J, Towse A, Smyth E. A review of healthtechnology appraisals: case studies in oncology. International Journalof Technology Assessment in Health Care 2013;29:101–9.

Please cite this article in press as: Nicod E, Kanavos P. Dwork to systematically compare HTA coverage decisionhttp://dx.doi.org/10.1016/j.healthpol.2015.11.007

15] Kanavos P, Nicod E, Van den Aardweg S, Pomedli S. The impactof health technology assessments: an international comparison.EuroObserver 2010;12:1–6.

16] Pomedli S. HTA and access to cancer medicines. EuroObserver2010;12:7–8.

[

PRESSicy xxx (2015) xxx–xxx 11

17] Nicod E. Harmonizing the outcomes of HTA: an application to CentralNervous System treatments. EuroObserver 2010;12:9–13.

18] Van den Aardweg S. HTA of orphan drugs across six countries: fair,flawed or failing? Euro Observer 2010;12:14–9.

19] European Medicines Agency. Human medicines, rare disease(orphan) designations. London, UK: European Medicines Agency;2013, http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/landing/orphan search.jsp&mid=WC0b01ac058001d12b.

20] Le Cam Y. Inventory of access and prices of orphan drugs acrossEurope: a collaborative work between national alliances on rare dis-eases & Eurordis. Eurordis Rare Diseases Europe; 2010.

21] Dupont A, Van Wilder P. Access to orphan drugs despite poor qual-ity of clinical evidence. British Journal of Clinical Pharmacology2011;71:488–96.

22] Creswell J. The selection of a research approach. In: Oaks T, edi-tor. Research design: qualitative, quantitative and mixed methodsapproaches. Los Angeles, CA: Sage; 2003. p. 1–24.

23] Creswell J. Plano Clark V. Designing and conducting mixed methodsresearch. Los Angeles, CA: Sage Publications; 2011.

24] Teddlie C, Tashakkori A. Overview of contemporary issues in mixedmethods research. In: Tashakkori A, Teddlie C, editors. Sage hand-book of mixed methods in social & behavioral research. Los Angeles,CA: Sage Publications; 2010. p. 1–42.

25] Bryman A. Social research methods. Oxford: Oxford University Press;2004.

27] Creswell J, Plano Clark V. Collecting data in mixed methods research.In: Creswell J, Plano Clark V, editors. Designing and conductingmixed methods research. Los Angeles, CA: Sage Publications; 2011.p. 110–27.

29] Gerring J ,. What is a case study good for? Case study versus large-ncross-case analysis. In: Gerring J, editor. Case study research: Princi-ples and practices. New York, NY: Cambridge University Press; 2007.p. 37–63.

30] Onwuegbuzie A, Combs J. Emergent data analysis techniques inmixed methods research: a synthesis. In: Tashakkori A, Teddlie C,editors. Sage handbook of mixed methods in social & behavioralresearch. Los Angeles, CA: Sage Publications; 2010. p. 397–430.

31] Boyatzis RE. Developing themes and codes. In: Boyatzis RE, editor.Transforming qualitative information: thematic analysis and codedevelopment, vol xvi. Thousand Oaks, CA: Sage Publications; 1998.p. 184.

32] Ltd QIP. NVivo qualitative data software; 2012.33] StataCorp. In: Station C, editor. Stata statistical software: release 13.

TX: StataCorp LP; 2013.35] Dickenson W. Visual displays for mixed methods findings. In:

Tashakkori A, Teddlie C, editors. Sage handbook of mixed methodsin social & behavioral research. Los Angeles, CA: Sage Publications;2010. p. 469–504.

36] Bartholomew D, Steele F, Moustaki I, Galbraith J. Analysis of multi-variate social sciences data, 2nd ed. Florida: Taylor & Francis; 2008.

37] Cohen J. Coefficient of agreement for nominal scales. Educational andPsychological Measurement 1960;20:37–46.

38] Altman D. Practical statistics for medical research. London, England:Chapman and Hall; 1991.

39] Fischer K. A systematic review of coverage decision-making onhealth technologies-Evidence from the real world. Health Policy2012;107:218–30.

40] Carroll C, Kaltenthaler E, FitzGerald P, Boland A, Dickson R. A thematic

eveloping an evidence-based methodological frame-s: A mixed methods study. Health Policy (2015),

missions to the NICE Single Technology Assessment (STA) process.Health Policy 2011;102:136–44.

41] Creswell J, Tashakkori A. Developing publishable mixed methodsmanuscripts. Journal of Mixed Methods Research 2007;1:107–11.