Rapid Relative Effectiveness Assessments (4.2)

LIST OF ABBREVIATIONS

AIDS

Acquired immune deficiency syndrome

API

Active pharmaceutical ingredient

ATC

Anatomical therapeutic chemical

CA

Competent Authority

CHMP

Committee for Medicinal Products for Human Use

CI

Confidence Interval

CINAHL

Cumulative Index to Nursing and Allied Health Literature

CSR

Clinical Study Report

CUR

Health problem and current use domain

DARE

Database of Abstracts of Reviews of Effects

DDD

Defined Daily Dose

EEA

European Economic Area

EFF                      

Clinical effectiveness

EFTA

European Fair Trade Association

EMA

European Medicines Agency

EPAR

European Public Assessment Report

ERIC

Education Resources Information Centre

EUnetHTA

European network for Health Technology Assessment

FDA

Food and Drug Administration

GRADE

Grading of Recommendations Assessment, Development and Evaluation

HIV

Human immunodeficiency virus

HR

Hazard ratio

HRQoL

Health-related quality of life

HTA

Health Technology Assessment

INAHTA

The International Network of Agencies for Health Technology Assessment

IAEA

International Atomic Energy Agency

ICD

International Classification of Diseases

ICRP

Publication of International Commission of Radiological Protection

IEC

International Electrotechnical Commission

ISO

International Organization for Standardization

IVD

In Vitro Diagnostic

MeSH

Medical subject headings

MHRA

Medicines and Healthcare products Regulatory Agency

NHS EED

NHS Economic Evaluation Database

NNH

Number needed to harm

NNT

Number needed to treat

NRS

Non-randomised studies

OTC

Over the counter

PICO

Patient, intervention, comparison, outcome

QALY

Quality-adjusted life years

QoL

Quality of life

RCT

Randomised controlled trial

REA

Relative effectiveness assessment

SAE

Serious adverse events

SAF

Safety

SDOR

Summary Diagnostic Odds Ratio

SPC

Summary of Product Characteristics

SuRe Info

Summarized Research in Information Retrieval for HTA

TEC

Description and technical characteristics of the technology domain

TGA

Therapeutic Goods Administration

WHO

World Health Organisation

WP

Work package

1  INTRODUCTION

1.1. The HTA Core Model for Rapid Relative Effectiveness Assessment

The HTA Core Model for Rapid Relative Effectiveness Assessment (REA) abbreviated as ‘Model for Rapid REA’ is a methodological framework for the collaborative production and sharing of HTA information. The Model for Rapid REA defines the content elements to be considered in an HTA and enables standardized reporting. Because the objective of the framework is sharing of commonly required elements of information, only information that is considered both important and transferable is collected. 

The aim of the Model for Rapid REA is:

  • to improve the applicability of HTA information in other (e.g. national or regional) HTA projects
  • to enable actual collaboration between HTA agencies by providing a common framework for the production of rapid REA
  • to avoid duplication of work.

Resting on the HTA Core Model®, the Model for Rapid REA provides an overview for producers of rapid REAs on the basic steps involved and on important generic research questions which should be considered in a HTA.

Rapid REAs contain an analysis of a health technology in comparison with one or more relevant alternative interventions, which is limited to a subset of domains and performed within a limited timeframe. The Model covers generic research questions (i.e. issues) for four different types of technologies:

  • Pharmaceuticals
  • Diagnostic Technologies
  • Medical and Surgical Interventions
  • Screening Technologies.

For a detailed description of the domains, guidance concerning assessments of specific types of technologies and for further potentially relevant research questions to be considered within a rapid REA the HTA Core Model® should be consulted.

What is relative efficacy/effectiveness?

Two definitions are commonly used in the context of an REA [1]:

  • Relative efficacy can be defined as the extent to which an intervention does more good than harm, under ideal circumstances, compared with one or more alternative interventions.
  • Relative effectiveness can be defined as the extent to which an intervention does more good than harm compared with one or more alternative interventions for achieving the desired results when provided under the usual circumstances of health-care practice.

When assessing relative effectiveness, the focus is on determining the magnitude of the health benefits and harms of a (new) technology compared with other existing technologies. As stated in the principles on relative effectiveness [1], an REA should include a comparison with the most appropriate health-care intervention(s). The REA should focus primarily on data derived from usual circumstances of health-care practice, although these are usually not available right after marketing authorisation or market entry of the technology. Additionally, the REA should present the uncertainties affecting interpretation of reliability and clinical relevance of the results. Rapid REAs may assess a new technology recently introduced to the market, or (re)assess a technology for a new indication or when new relevant data are available [2]. 

1.2. Background  

The HTA Core Model for Rapid REA is based on the HTA Core Model®  which consists of three main components:

  1. The HTA ontology contains an extensive list of generic questions that can be asked in a HTA.
  2. Methodological guidance helps researchers to find answers to the questions defined by the Model.
  3. The common reporting structure provides a standard format for the output of HTA projects.

Figure 1: Domains of the HTA Core Model® and of the HTA Core Model for Rapid Relative Effectiveness Assessments

Rapid Figure 1

Figure 2: An assessment element

Rapid Figure 2 w

The HTA Core Model® organises the information by dividing it first into nine domains (see Figure 1). The purpose of dividing the assessment into specific domains is to facilitate the systematic presentation of information. Each domain is divided into topics, and each topic is further divided into several issues (see Figure 2). The issues are the generic questions that should be considered when assessing a health technology. The combination of a domain, topic, and issue defines an assessment element within the HTA Core Model®.  

Since the Model for Rapid REA is intended for assessments within a limited time frame, it covers only the first four domains of the HTA Core Model® (see Figure 1) and within these domains only a subset of issues. The domains covered in the Model for Rapid REA are briefly described below. 

1.3. Domains 

Description and technical characteristics of technology (TEC)

The information presented in this domain describes the technology under assessment (or a sequence of technologies) and its technical characteristics: the type of device, technique, procedure or therapy; its biological rationale and mode/mechanism of action, how the technology differs from its predecessors, and the various current modifications or different manufacturers’ products, especially if the differences affect performance;  when it was developed, for what purpose(s), who will be using it, in what manner, and at what level of health care. The regulatory and reimbursement status of the technology is listed when applicable within the context of the assessment.

The issues in this domain should be described in sufficient detail to differentiate the technology from its comparator(s). The relevant terms and concepts used should be used in a way that allows those unfamiliar with the technology to get an overall understanding of its use. It is important to distinguish between scientifically proven versus suggested mechanisms of action. Important terms should be defined and a glossary or list of product names provided. The section may include pictures, diagrams, videos or other visual material, in order to facilitate understanding for persons who are not experts in the field.

Health problem and current use of the technology (CUR)

The information presented in this domain describes the target condition, target group, epidemiology and the availability and patterns of use of the technology in question. Furthermore, it describes the burden – both on individuals and on society – caused by the health problem, as well as the alternatives to the technology in question. Some of the topics considered relevant for this domain have generally referred to as ‘background information’ in previous European projects or recommendations for conducting assessments [3-5].

The qualitative description of the target condition, which is covered in this domain, includes the condition’s underlying mechanism (pathophysiology), natural history (i.e. course of disease), available screening and diagnostic methods, prognosis and epidemiology (incidence, prevalence), underlying risk factors for acquiring the condition, as well as available treatments. A description of subgroups or special indications should be included especially when the technology does not target the whole population.

Current management patterns of the condition should be described, including the technology and its alternatives, as well as recommended policies for determining the target population. It should also be specified whether the technology is intended to replace or supplement another technology in the management chain. Anticipated problems with the use of the technology within a health system should be identified, e.g. inappropriate extension of indications (off-label use), participation rate or compliance, over-diagnosis and misuse are to be discussed, as are the alternatives to the technology and the agreed-upon policies regarding the choice of patients or target group for treatment.

For assessing diagnostic technologies, it is crucial to understand the role of the technology in the entire health-care pathway, including diagnostics and treatment and also in relation to existing technologies. Current options for diagnostics and therapy should be described, in particular the reference standard and how good the standard is in classifying the condition. Assessments on screening technologies should include the whole management chain, from the screening test, through the subsequent diagnostic tests, to treatments, and should describe whether it is a screening technology that modifies the existing screening pathway only slightly, or whether it is an assessment of a completely new screening pathway.

Clinical Effectiveness (EFF)

The information presented in this domain discusses the relative benefits of a (new) technology in comparison with one or more alternatives which can be determined under experimental conditions (i.e.  efficacy; within the protocol of a randomised controlled trial (RCT)) or under routine conditions (i.e. effectiveness; by a physician in a community hospital treating outpatients) (adapted from the International Network of Agencies for Health Technology Assessment (INAHTA) glossary [6]).

Key elements of a benefit assessed under routine conditions are:

(a) the most relevant interventions should be directly compared where possible, and,

(b) studies should include patients who are typical of day-to-day health-care settings [7].

The scope of REAs is to determine the relative benefits of a technology under routine conditions, i.e. its effectiveness. Ideally, both types of data would be available from RCTs, allowing the assessment under ideal circumstances underpinned by data obtained under routine conditions.

Effectiveness could as well, as supportive measure of the two data sources mentioned above, be estimated with real world data (non-RCT studies). Once the extent of the effect obtained in experimental designs is known, it can be additionally checked by observational designs to evaluate the external validity or generalizability of the effect  [8].

The assessment of health benefits should primarily consider clinically meaningful endpoints such as mortality, morbidity and quality of life (QoL). The choice of clinical (primary) endpoints will depend upon the target population, main characteristics of the disease of interest (non-life-threatening versus life-threatening) and the aim of treatment. For a life-threatening disease, a mortality or survival endpoint is generally preferred as the primary endpoint, whereas morbidity and/or health-related quality of life (HRQoL) may be preferred as secondary endpoints. In non-life-threatening diseases, morbidity and HRQoL endpoints will be preferred for the primary endpoints. The clinical endpoints used should be measurable for all or most patients within a reasonable time frame [9]. Surrogate endpoints act as substitutes for clinically meaningful endpoints and are expected to predict the effect of the technology (benefit and/or harm). Surrogate endpoints should only be used if they are validated adequately. The level of evidence, the associated uncertainties and the limits of their use should be explained explicitly.

Safety (SAF)

The information presented in this domain describes any unwanted or harmful effects caused by using a health technology. Safety issues should be covered that are important to patients or otherwise likely to be important in guiding the decision of health care providers and policy makers [4]. The harmful effects of a technology are essential in quantifying the net benefit (benefit minus harms) of an intervention and are essential for being able to form a balanced view of the overall diagnostic or therapeutic value of a technology. The harms are identified and quantified in terms of frequency, incidence, severity and seriousness, and are then compared to those of the comparator(s).

Uncertainties due to a restricted knowledge base (small numbers, short follow-up) should be addressed when serious or late harms can be expected foremost if the technology is compared to well-established comparator(s). For screening and diagnostic technologies, further harms including indirect ones, such as false-negative or false-positive test results should be considered.

Checklist for potential ethical, organisational, patient and social and legal aspects

The other five domains of the HTA Core Model® (i.e. on costs, ethical, legal, social and organisational issues) were excluded from the Model for Rapid REA because the information contained therein is highly context dependent and has therefore limited transferability. However, ethical, organisational, patient and social as well as legal aspects that may need to be addressed in-depth are covered by a short checklist (see Appendix 3: Template 3. Checklist for potential ethical, organisational, patient and social and legal aspects). If the response to a question in the checklist is ‘yes’, further analysis may be warranted, otherwise the checklist does not need to be considered further. Since the assessment is comparative in nature, only those issues for which a difference exists between the technology to be assessed and its major comparator(s) should be described.

Further relevant assessment elements from these four domains may be selected from the HTA Core Model®. Pre-established problems/issues, with regard to ethical, organisational, patient and social as well as legal aspects, which are common to the technology to be assessed and its comparator(s) will, as a rule, not be addressed, as it is not to be expected that the addition of a new technology will lead to changes.

2  METHODS

2.1. Setting the general scope of the assessment

Key messages for scoping

  • Scoping should be performed according to the PICO structure (see Appendix 3 Template 1. Format for scoping the assessment).
  • The choice of comparator(s) and outcomes should be justified explicitly in the assessment.
  • Consultation with the sponsors of technologies under assessment regarding the scope of the assessment may be a valuable source.
  • During the scoping phase, the Checklist for potential ethical, organisational, patient and social and legal aspects should be completed.
  • At the end of the scoping phase, a final project plan will have been completed. 

The first step in a rapid REA is to specify what should be assessed (i.e. the scope) following the so-called PICO structure. PICO stands for:

  • Population/patients with the disease of interest
  • Intervention(s), i.e. the technology under assessment
  • Comparison(s) that should serve as reference
  • Outcome(s), which encompass the endpoints for assessing effectiveness and safety.

The PICO structure will drive the evaluation in all four domains. The population, intervention and comparison will generally be the same for all domains. However, it may sometimes be necessary to deviate from the scope because of, e.g. a subpopulation of special interest, or the absence of data for the population defined in the scope. General guidance for deriving a well-defined research question is provided in the Cochrane Handbook [10]. The following considerations are relevant regarding the PICO elements in the context of a rapid REA.

  • Population/patients with the disease of interest. The diseases or conditions of interest should be defined using explicit criteria for establishing their presence or not. Second, the broad population and setting of interest should be defined. This involves deciding whether a special population group is of interest, determined by factors such as age, sex, race, educational status or the presence of a particular condition such as angina or shortness of breath. Interest may focus on a particular setting such as a community, hospital, nursing home, chronic care institution, or outpatient setting [10]. For pharmaceuticals, an initial definition of patients who will receive the intervention is generally provided by the marketing authorisation, which in turn is based on the evidence provided by the sponsors of the technology. For other technologies, HTAs, guidelines and reviews and clinical experts are relevant sources that can be used. The purpose of use of the technology should be specified, for example, first- or second- line treatment or whether the intended purpose is treatment or prevention.
  • Intervention(s). Factors usually specified include the precise nature of the intervention (e.g. the method of administration of a drug), the person delivering the intervention (e.g. a community psychiatric nurse versus a non-professional carer) or setting in which the intervention is delivered (e.g. inpatient or outpatient). The dose(s) and frequency of the technologies and their comparators is a crucial issue. This is true for direct, as well as indirect, comparisons. For example, when the comparator (or one of the comparators) is a pharmaceutical administered at low doses, this will lead to over-estimation of the technology’s efficacy or effectiveness and estimation of safety will be compromised. For pharmaceuticals, dose schedules and the route of administration should be presented either with those recommended in the marketing authorisation or representative of those used in standard clinical practice in Europe (if European guidelines recommend a difference to the marketing authorisation). Knowledge of their dose–response relationships are a prerequisite for interpreting the results of the comparisons. For more complex interventions, it should be considered what is delivered, at what intensity and at which frequency and whether people involved need to be trained [10].
  • Comparison(s). The comparator(s) should be chosen carefully, preferably based on up to date high-quality clinical practice guidelines at European or international level with good quality evidence on the efficacy and safety profile from published scientific literature. In the context of a rapid REA, the number of comparators should be limited. The comparator(s) may be another procedure, a drug or, for medical devices, quite often a sham device or procedure. The choice of comparator(s) should be justified explicitly in the report.
  • Outcome(s). For the assessment of relative effectiveness, consideration must be given to the appropriateness of the outcome variables on which information on the intervention’s effect is available. When surrogate variables (e.g. low-density lipoprotein cholesterol concentration; blood pressure) are used as outcome measures, the clinical validity of these measures needs to be considered. Composite endpoints should generally not be used if a suitable single primary endpoint is available. If a single primary endpoint is not available, or if a composite endpoint can be justified to be more suitable (e.g. rare disease/event), it may be chosen instead. When possible, adverse events relevant for the assessment should be identified in advance and should be listed in the scope. The choice of outcomes should be justified explicitly in the report.

A template for reporting the scope is included in Appendix 3 (Template 1. Format for scoping the assessment).

2.2. How to work with the assessment elements

Assessment elements are the standardised parts of HTA information. Every answer to the issues defined by the assessment elements is recorded as a structured piece of information.

Key messages for the assessment elements table

  • Assessment elements are standardised pieces of HTA information.
  • Each domain has specific assessment elements that contain issues, i.e. generic research questions that can be answered for that domain.
  • In all domains, each issue should be considered individually for its relevance for the rapid REA.
  • The selected issues should be translated into actual research questions (answerable questions).

Selecting relevant issues from the model

In this phase the research team(s) should go through all the domains they are interested in and consider each issue (i.e. the generic questions in the relevant assessment element table) one by one. Each issue should be defined either as relevant or irrelevant.

The issues defined as relevant will be studied in the assessment or they can be tagged as "consider later" to allow flexibility in the working process. The relevance is based on whether the issue presented is relevant in the context of the particular technology that is being assessed. One should be practical: not to try to find "artificial" relevance, but not to reject issues too easily as irrelevant either.

If an issue is considered relevant, but no data are available to answer the question, it should be reported in the assessment. Thus, issues should not be excluded based on a lack of data, but the gap in evidence should be identified and reported. In these cases, further studies can be recommended, after their feasibility has been confirmed.

Further assessment elements from the HTA Core Model® application for medical and surgical interventions, screening, pharmaceuticals or diagnostic technologies, which are not contained in the Model for Rapid REA, could also be screened and included in rapid REAs when deemed relevant.

Formulating research questions

In this phase, the authors should translate the issues into actual research questions. One issue usually translates into one research question, but it is sometimes necessary to translate a single issue into two or more research questions. It is important that this phase results in a set of pragmatic and answerable questions with which the authors can proceed. 

2.3. Collecting and analysing data

Methods for finding, selecting, analysing, synthesizing and interpreting evidence on clinical effectiveness recommended for conducting systematic reviews such as the Cochrane Handbook or the CRD handbook [10, 11] are in principle applicable to all health technologies.

Potential information sources:

The following information sources can be used:

  • Bibliographical databases: e.g. Medline, Embase
  • Specialised databases: e.g. CINAHL, ERIC
    • Administrative databases: e.g. Emerald Library, Pub Med Central
    • Incident reporting databases: e.g. US Manufacturer and User Facility Device Experience Database [MAUDE])
  • Trial registers: e.g. Clinical Trials, WHO International Clinical Trials Registries Platform portal
  • Databases on specific study designs: e.g. DARE, NHS EED
  • Useful other sources:
    • Surveys, epidemiological research, national and regional guidelines, routine statistics and administrative databases, conference proceedings (Web of Science Database), expert opinions
    • Additional information can be collected also from contacts with the sponsors of the technology e.g. Submission Files.
  • For pharmaceuticals:
    • EPAR including the Summary of Product Characteristics (SPC), of the pharmaceutical of interest. The availability of the EPAR and SPC depends greatly on the timing of the assessment. In case of an early assessment (before marketing authorisation), the documents may not yet be available. Therefore, the Committee for Medicinal Products for Human Use (CHMP) report, or a draft of the EPAR can be used initially for drafting first versions of the rapid REA. However in the final stages of the assessment preparation, information should be checked against the EPAR and SPC for inconsistencies.
    • EPARs, including SPCs of comparators
    • Original unpublished studies that are relevant for the rapid REA in the format of Clinical Study Reports (CSRs). Unpublished data should only be included in the assessment if the authors are allowed to present the data in the report.
  • For medical devices:
    • Instructions for Use
    • CE mark
    • Orienting/initial information on safety may also be retrieved from device registries, incident reporting databases (e.g. US Manufacturer and User Facility Device Experience Database [MAUDE]) and administrative databases. For details refer to the EUnetHTA Safety Guideline (chapter 2.3.5) and to the Summarized Research in Information Retrieval for HTA (SuRE Info) on the HTAi webpage (http://vortal.htai.org/?q=sure-info).
    • Information on FDA-approved devices, including data used for approval, is available via http://www.accessdata.fda.gov/scripts/cdrh/devicesatfda/index.cfm.

Literature search

Information retrieval for systematic reviews needs to be performed in a thorough, transparent and reproducible manner. The aim is to identify all relevant studies and study results on the question of interest (within resource limits) [10]. This requires both searches in several information sources and the use of comprehensive search strategies [3-5]. This approach is a key factor in minimizing bias in the review process [12]. This literature search can be provided either within a Submission File completed by the sponsors or it should be conducted by the authors.

All final search strategies should be included in the rapid REA, including searches for ongoing trials in clinical trial registries, e.g. ClinicalTrials.gov.

For guidance on domain specific information sources please refer to the SuRe Info (available from: http://vortal.htai.org/?q=sure-info).

Appropriate study types

Health problem and current use and Description and technical characteristics of the technology

There is no single methodological approach that can be applied to all issues in these domains. Descriptive and observational study designs, narrative reviews, surveys, observational and qualitative research, registry analyses and market research reports, as well as guidelines and consensus statements, can be used for compiling the domains.

 

Efficacy and effectiveness data

Following the hierarchy of study designs [13], reviews on efficacy/effectiveness are generally limited to randomised designs. To assess the generalizability to routine clinical practice it might be relevant to distinguish between efficacy (explanatory) and effectiveness (pragmatic) RCTs. A set of criteria has been suggested to differentiate between them [14].

Therefore, as a general rule, RCTs should be considered for assessing the health benefits of a technology and ideally, for a rapid REA most of the data should be retrieved from RCTs [15]. A (well-conducted) meta-analysis of the results of more than one RCT provides the highest level of evidence. Although data about the relative benefits under routine conditions are preferred for a relative effectiveness assessment, they are rarely available soon after marketing authorisation or at start of usage. Where sufficient good quality head-to-head studies are available, direct comparisons are preferred as the level of evidence is high. Should substantial indirect evidence be available, then it can act to validate the direct evidence [16, 17].

Although RCTs provide the most robust evidence, other types of studies may provide additional information on the relative efficacy or effectiveness. Non-randomised intervention studies or observational studies can be considered where an RCT has not been conducted, published yet or is not feasible, or complementary data are presented to RCTs. Since the regulatory approval process for medical devices in Europe does not necessarily require conducting RCTs, literature search can be broadened to include all types of study designs, including case series and even case reports. For the study of long-term outcomes such as revision rates and quality assurance of medical devices, comprehensive disease-based registries are preferable. Observational studies and randomised trials can be nested within these registries [18]. In addition, registry data reflecting clinical routine care help judging whether study populations, interventions and outcomes in RCTs are comparable to clinical practice.

For diagnostics specifically, other study types may provide evidence about test safety, accuracy, impact on management and the effectiveness of the treatment when direct trial evidence is not available. Evidence from these studies can be linked so as to yield an estimate of the diagnostic technology’s effectiveness (linked evidence).

Within a rapid REA, guidance on how to deal with studies with a high or unclear risk of bias should be specified in advance. There are three main options:

  • Rely only on studies with a low risk of bias;
  • Perform sensitivity analyses according to the different risk of bias categories; or
  • Describe the uncertainty with regard to the different levels of risk of bias, so that subsequent decisions can be made considering this uncertainty [19].

No general recommendation can be made as to which alternative is preferable, because the decision depends on topic-specific circumstances, regulatory context, resources and time expenditure. Inclusion of non-comparative studies such as case series poses additional difficulties to researchers, because the lack of between-group comparisons precludes assessment of relative effectiveness [20]. Due to risk of bias such evidence usually does not allow drawing definite conclusions on treatment effects. Possible reasons favouring the inclusion of non-randomised studies (NRS) include:

  • The research question cannot (or only with the greatest difficulty) be answered in RCTs. This may be the case because of organisational reasons (e.g. in public health interventions) or epidemiologic circumstances (e.g. very rare diseases).
  • The research question can probably be answered with NRS evidence, because very large effects are likely (or at least possible).
  • There is an external need to offer a ‘best guess’ rather than no answer at all. Such a situation may be present early in the life cycle of a new intervention or when HTA is used to make only a temporary decision which is followed by an early reassessment (e.g. in a coverage with evidence development model) [20] (further information on non-randomised study designs can be found in the Cochrane Handbook Chapter 13 [10]).

 

Safety data

A broad range of evidence sources may be considered to identify adverse effects relevant for the assessment. These sources may include regulatory sources (e.g. EPAR, SPC and RMP), manufacturer dossiers, randomised clinical trials, observational studies, country registries and case reports. Various sources can bring different and complementary information; randomised clinical trials may inform on common risks, whereas other data sources, although at higher risk of bias, (e.g. observational studies, country registries and case reports) can give insight on less frequent risks, long-term risks, and risks in populations not being part of randomised clinical trials [21].

Quality appraisal

Health problem and current use and Description and technical characteristics of the technology

Quality assessment of the information retrieved may be difficult, as there is often no standard way of assessing it and because many aspects and facets must be taken into account when information is evaluated in terms of its quality. The validity of the information may differ considerably depending on the type and source of information requested (quantitative or qualitative; registries, administrative data, etc.). Appropriate methods for appraising the available evidence should be selected considering the target level of detail and precision in providing information on these domains.

 

Clinical effectiveness data

Assessing the methodological quality of the included studies is crucial. Tools for critical appraisal can comprise different quality aspects of studies or publications. The risk of bias tool of the Cochrane Collaboration examines internal validity (risk of bias) of studies and endpoints, whereas other checklists combine questions to assess precision and external validity as well (see Cochrane Handbook Chapter 8) [10]. Internal validity describes the extent to which the (treatment) difference observed in a trial (or a meta-analysis) is likely to reflect the ‘true’ effect within the trial (or in the trial population) by considering methodological quality criteria. Because the ‘truth’ can never be assessed, it is more appropriate to speak of the potential for risk of bias. The risk of bias concept should be used to assess the internal validity of clinical studies within a rapid REA. The risk of bias should be assessed on two levels, i.e. first, on a (general) study level, and secondly, on an outcome level. For example, selection and performance bias threaten the validity of the entire study, while the other types of bias may be outcome specific.

If an REA is to be performed on the basis of systematic reviews rather than on primary studies, the methodological quality of the underlying reviews can be assessed either by the Oxman and Guyatt index [22], or by ‘A Measurement Tool to Assess Systematic Reviews’ (AMSTAR) [19, 23]. For non-randomised studies the ACROBAT-NRSI (A Cochrane Risk of Bias Assessment Tool) and RoBANS (Risk of Bias Assessment Tool for Nonrandomised Studies) can be used [20]. Further information on possible tools can be found in the References.

 

Safety data

Methods used to assess bias should be described clearly, and the risk of bias regarding the information sources and how the data was collected should be reported. The way risk of bias information was used in the rapid REA should be explained clearly.

Timelines of literature and registration data should be evaluated, as well as their applicability in vulnerable patient groups, such as elderly people with polypharmacy, people with comorbidities, neonates and children, pregnant women and immunosuppressed patients.

Effect measures and confidence intervals

A number of different types of data and corresponding measures of the intervention’s effect are in use. For dichotomous outcome data, relative effect measures, such as risk ratio (relative risk), odds ratio, and relative risk reduction, or absolute effect measures, such as risk difference (absolute risk reduction), are commonly used. The latter is often converted into number needed to treat (NNT) or events per thousand patients, to allow comparison across studies and to facilitate interpretation [10]. Both relative and absolute effect measures convey important complementary information, and therefore, presentation of both measures is encouraged by recent approaches such as the Grading of Recommendations Assessment, Development and Evaluation (GRADE) profiler (see www.gradeworkinggroup.org).

Continuous data are often more difficult to summarise. Commonly used effect measures that allow the summary of treatment effects are “standardised mean difference” or “weighted mean difference”. Unfortunately, both measures are difficult to interpret in a clinical context. A more recent statistic, the ratio of means, reports the percentage reduction for continuous data such as proteinuria. This measure allows a meaningful interpretation to clinicians. Ordinal outcome data and measurement scales may be either analysed as continuous data or made into dichotomous data by combining categories, and thus, according effect measures described above are used [10, 24].

A measure of the precision of the effect estimate (standard error or confidence interval (CI)) is required for the interpretation of the data. The absence of this essential information should be reported.

For safety data, it is recommended that, whenever possible, the frequency of adverse events should be quantified, and information on the frequency of occurrence, relative risk or number needed to harm (NNH) should be obtained [4]. For the analyses of rare events, the rate ratio (RR) is commonly used, comparing the rate of events in two groups. Time-to-event data (survival data) should be summarised expressing the intervention effect as a hazard ratio, describing how many times more (or less) likely an event occurs when receiving the intervention [10]. Randomised trials are methodologically most solid, and may alone be an appropriate source of evidence for some review questions about harm. However, safety reporting in randomised trials is heterogeneous and often inadequate. Rare adverse effects are not usually detected in randomised trials, and even relatively frequent harms with a longer latency period cannot be quantified easily. Information about new, serious, rare or long-term adverse effects are thus typically found in observational studies (cohort, case-control and cross-sectional studies). Risk of late-onset harms (e.g. number of radiation-induced cancers) can be estimated based on analogies and assumptions from epidemiological studies. In cases where adverse events are incorporated in utility values of QoL, the source of the quantification should be accessible.

The values of ratio effect measures (e.g., the odds ratio, risk ratio, rate ratio and hazard ratio) usually undergo log transformations before analysis. For more details about types of data, effect measures and their calculations, please refer to the comprehensive, user-friendly description of common measures in the Cochrane handbook. The handbook also includes guidance on Bayesian approaches to analysing data [10].

Extrapolation of efficacy to give relative effectiveness data

Ideally, for a rapid REA most of the data is retrieved from high-quality RCTs. As these trials were conducted in a specific setting, it is relevant to consider the applicability of the results to the intended population for treatment [25]. Exploring effect modifiers and critical factors for implementation may enhance the value of a review of clinical effectiveness for users [26]. If heterogeneity within and between studies can be explained by effect modification, these factors should be considered in clinical practice. For interventions with therapeutic MD implying surgery or other procedures, individual and institutional expertise (including infrastructure) and learning effects/curve have to be taken into account as potentially effect modifying factors.

In the case of surrogate outcomes, transformation into patient-relevant final outcomes of treatment should be considered [25].

It may be relevant for a rapid REA to include data from indirect comparisons. Where sufficient good-quality head-to-head studies are available, direct comparisons are preferred as the level of evidence is high. If substantial indirect evidence is available, then it can act to validate the direct evidence. However, when there is limited head-to-head evidence, or more than two treatments are being considered simultaneously, the use of indirect methods may be helpful.

Interpreting evidence

At this stage, the authors of a rapid REA should check that the data extracted are relevant to the research questions formulated at the start of the process, and that analysing and synthesising the data continues to answer the questions. Often, the evidence available is not quite as useful as anticipated; in such cases, a clear report should be made on how well the evidence answers the original research question. Cases in which no data were available should also be reported.

The reader should be given an idea of the nature and magnitude or frequency of the event, and the overall robustness of the evidence behind this assessment. There are several ways to provide this information. In many cases, plain text is sufficient; in others, an evidence table would be helpful.

Evidence tables

Comprehensive and informative evidence tables about the methodology and the content of the individual studies:

  • foster transparency and reliability, which are prerequisites for the transfer of a rapid REA from one setting to another;
  • allow a judgment of the similarities and differences of the studies included; and
  • provide the basis for the conclusions of the review.

Evidence tables, therefore, should be part of each rapid REA.

The majority of HTA organisations produce tabulated evidence summaries that follow the PICO structure, ideally with an additional cell for comments on issues that are not captured by the PICO elements but that could have an impact on the results. Although the items reported in each cell will be driven by the review questions, they follow some core considerations [27]. A description of the data extraction process, including the number of reviewers involved, assures objectivity and reliability of the results.

To interpret the evidence, the following aspects should be discussed in the assessment:

  • The strength/uncertainties of the evidence available. This should include the internal validity of the body of evidence as well as the applicability of the evidence.
  • The clinical relevance of the findings:
    • Statistical significance is not a sufficient precondition because numerically small differences can be statistically significant, but clinically meaningless. Consider the magnitude (i.e. relevance) of the treatment effect (independent of its statistical significance) and compare this with the minimal clinically important effect size. One approach is to compare the lower limit of the 95% CI of an estimated treatment effect with a ‘maximal clinically unimportant effect size’.
    • Consider the relevance of the outcomes for clinical decision making (distinguishing between primary and secondary outcomes as is done when developing the project plan).
    • Identify knowledge gaps by comparing the research questions (including the predefined outcome) with the available evidence.

To allow transfer of data across countries, rapid REAs have to be sufficiently transparent and distinguish between evidence (‘facts’) and judgements (including values and preferences). Value judgements and preferences (of individuals or of health-care systems) have to be labelled as such, but rapid REAs should not contain recommendations for or against technologies assessed.

2.4. Reporting

In order to assess relative effectiveness, a synthesis of both effectiveness (benefits) and safety (harms) data is needed. The benefits and harms of the intervention(s) should be presented in comparison with the comparator(s). The following, at least, should be included:

  • Scope: description of the technology; description of comparator(s); description of the health problem; description of the current treatment of targeted health conditions according to guidelines and standard clinical practice.
  • Results: description of available evidence and ongoing trials; description of relative effectiveness results; description of relative safety results.
  • Summary table of relative effectiveness (a template for writing the summary can be found in Appendix 3 (Template 2: Summary of relative effectiveness).  
  • Discussion: discussion of potential limitations, including internal validity and applicability, of available evidence and identification of evidence gaps.
  • Conclusion: conclusion for each comparator as to whether the technology is less, similarly, or more effective and safe; conclusion as to whether further research is required.

References

[1]  European Commission, Enterprise and Industry - Working Group on Relative Effectiveness. Core principles on relative effectiveness.  2008  [cited 09.04.2015; Available from: http://ec.europa.eu/DocsRoom/documents/7581?locale=en 

[2]  Kleijnen S, George E, Goulden S, d'Andon A, Vitre P, Osinska B, et al. Relative effectiveness assessment of pharmaceuticals: similarities and differences in 29 jurisdictions. Value Health. 2012 Sep-Oct;15(6):954-60. 

[3]  Burls A, Cummins C, Fry-Smith A, Gold L, Hyde C, Jordan R, et al. West Midlands Development and Evaluation Service Handbook. Birmingham: West Midlands Development and Evaluation Service; 2000. 

[4]  Velasco M, Perleth M, Drummond M, Gurtner F, Jorgensen T, Jovell A, et al. Best practice in undertaking and reporting health technology assessments. Working group 4 report. Int J Technol Assess Health Care. 2002;18(2):361-422. 

[5]  Liberati A, Sheldon TA, Banta HD. EUR-ASSESS Project Subgroup report on Methodology. Methodological guidance for the conduct of health technology assessment. Int J Technol Assess Health Care. 1997;13(2):186-219. 

[6]  Facey K, Topfer L-A, Chan L, on behalf of the International Network of Agencies for Health Technology Assessment (INAHTA). Health Technology Assessment (HTA) Glossary.  2006  [cited 09.04.2015; Available from: http://inahta.episerverhotell.net/upload/HTA_resources/Edu_INAHTA_glossary_July_2006_final.pdf 

[7]  Sox HC, Greenfield S. Comparative effectiveness research: a report from the Institute of Medicine. Ann Intern Med. 2009 Aug 4;151(3):203-5. 

[8]  Ashcroft R. What is clinical effectiveness. Studies in History and Philosophy of Science Part C: Studies in History and Philosophy of Biological and Biomedical Sciences. 2002;33(2):219-33. 

[9]  European Network for Health Technology Assessment. Endpoints used in relative effectiveness assessment of pharmaceuticals - Surrogate Endpoints 2013. 

[10]  Higgins JP, Green S, eds. Cochrane Handbook for Systematic Reviews of Interventions 2011. 

[11]  Centre for Reviews and Dissemination. Systematic Reviews.; 2009. 

[12]  Institute for Quality and Efficiency in Health Care. Process of information retrieval for systematic reviews and health technology assessments on clinical effectiveness; 2015. 

[13]  Guyatt G, Gutterman D, Baumann MH, Addrizzo-Harris D, Hylek EM, Phillips B, et al. Grading strength of recommendations and quality of evidence in clinical guidelines: report from an american college of chest physicians task force. Chest. 2006 Jan;129(1):174-81. 

[14]  Gartlehner G, Hansen RA, Nissman D, Lohr KN, TS. C. Criteria for Distinguishing Effectiveness From Efficacy Trials in Systematic Reviews. Rockville: AHRQ Technical reviews and Summaries; 2006. 

[15]  Tice JA, Helfand M, Feldman MD. Clinical evidence for medical devices: regulatory processes focussing on Europe and the United States of America: World Health Organization (WHO); 2010. 

[16]  European Network for Health Technology Assessment. Comparators and Comparisons: Direct and indirect comparisons. 2013. 

[17]  Jansen JP, Fleurence R, Devine B, Itzler R, Barrett A, Hawkins N, et al. Interpreting indirect treatment comparisons and network meta-analysis for health-care decision making: report of the ISPOR Task Force on Indirect Treatment Comparisons Good Research Practices: part 1. Value Health. 2011 Jun;14(4):417-28. 

[18]  Lauer MS, D'Agostino RB, Sr. The randomized registry trial--the next disruptive technology in clinical research? The New England journal of medicine. 2013 Oct 24;369(17):1579-81. 

[19]  European Network for Health Technology Assessment. Internal validity of randomized controlled trials EUnetHTA; 2013. 

[20]  European Network for Health Technology Assessment. Internal validity of non-randomised studies (NRS) on interventions; 2015. 

[21]  European Network for Health Technology Assessment. Endpoints used in relative effectiveness assessment of pharmaceuticals - Safety 2013. 

[22]  Oxman AD, Guyatt GH. Validation of an index of the quality of review articles. J Clin Epidemiol. 1991;44(11):1271-8. 

[23]  Shea BJ, Grimshaw JM, Wells GA, Boers M, Andersson N, Hamel C, et al. Development of AMSTAR: a measurement tool to assess the methodological quality of systematic reviews. BMC Med Res Methodol. 2007;7:10. 

[24]  Higgins JP, Green S. Cochrane Handbook for Systematic Reviews of Interventions: The Cochrane Collaboration,. 

[25]  Australian Government Department of Health and Ageing (AGDH). Guidelines for preparing submissions to the Pharmaceutical Benefits Advisory Committee. Version 4.4.  2013  [cited 09.04.2015; Available from: http://pbac.pbs.gov.au/ 

[26]  Squires JE, Valentine JC, Grimshaw JM. Systematic reviews of complex interventions: framing the review question. J Clin Epidemiol. 2013 Nov;66(11):1215-22. 

[27]  Malmivaara A, Koes BW, Bouter LM, van Tulder MW. Applicability and clinical relevance of results in randomized controlled trials: the Cochrane review on exercise therapy for low back pain as an example. Spine. 2006 Jun 1;31(13):1405-9. 

[28]  Weinstein MC, O'Brien B, Hornberger J, Jackson J, Johannesson M, McCabe C, et al. Principles of good practice for decision analytic modeling in health-care evaluation: report of the ISPOR Task Force on Good Research Practices--Modeling Studies. Value Health. 2003 Jan-Feb;6(1):9-17. 

Health Problem and Current Use of the Technology

Assessment elements

A0002 Assessment element card

Issue: What is the disease or health condition in the scope of this assessment?

Topic: Target Condition

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes1
Clarification
Common to all used applications

Use the target condition and International Classification of Diseases (ICD) codes defined in the scope of the project and consider adding details such as: description of anatomical site, disease aetiology and pathophysiology, types of disease or classification according to origin, subtype, severity, stages, or risk level and different manifestations of the condition. The following properties of the target condition are defined in separate assessment elements: risk factors (A0003), natural course (A0004), symptoms (A0005) and burden of disease for the society (A0006).


A0003 Assessment element card

Issue: What are the known risk factors for the disease or health condition?

Topic: Target Condition

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes2
Clarification
Common to all used applications

Describing risk factors is especially important when they suggest possibilities for primary and secondary prevention. This information may affect the choice of comparator or the appraisal of the overall value of the technology under assessment. The risk factors for acquiring the condition, and the risk factors for relapses or worsening of the condition should be reported here separately. The prevalence of the various risk factors might differ in different geographic areas and among different subpopulations.


A0004 Assessment element card

Issue: What is the natural course of the disease or health condition?

Topic: Target Condition

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes3
Clarification
Common to all used applications

This assessment element should provide information on the prognosis and course of the health condition when untreated. This information is relevant for appraising the overall value of the technology. It may also guide the assessment of the predicted value or effectiveness of the technology, as technologies may work differently at different stages or severity grades of the disease, and there may be a relationship between earlier intervention and better prognosis. This element should also provide information on the time lag between the onset of disease and the symptoms or other findings that eventually trigger the need of diagnostics and care.


A0005 Assessment element card

Issue: What are the symptoms and the burden of disease or health condition for the patient?

Topic: Target Condition

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes4
Clarification
Common to all used applications

This element should describe the patients’ relevant symptoms before intervention with the technology, their severity and whether they are persistent, intermittent or undulating taking into account different stages of the disease. Patients’ perceptions of the burden of the disease are not always in line with the clinical seriousness of the disease or its societal burden.


A0006 Assessment element card

Issue: What are the consequences of the disease or health condition for the society?

Topic: Target Condition

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes5
Clarification
Common to all used applications

Describe consequences and burden of the disease or health condition by providing information on prevalence or incidence of the disease that is prevented or treated by using the technology; disease-specific mortality and disability, life years lost, and/or disability-adjusted life years, QoL, quality-adjusted life years (QALYs).


A0020 Assessment element card

Issue: For which indications has the technology received marketing authorisation or CE marking?

Topic: Regulatory Status

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes6
Clarification
Common to all used applications

There are both international and national market authorisation systems. The systems differ between countries and are more established for pharmaceuticals than for medical devices. An overview of the status with regard to key processes, e.g. CE marking, EMA/US Food and Drug Administration (FDA) approval is recommended. In case the technology is authorised under a different process, e.g. adaptive licensing or conditional reimbursement, information should be presented. Also, information on national data and an analysis of possible discrepancies can be useful.

Specific to diagnostic technologies:

Imaging devices may require approval. Substances needed for obtaining images may require additional approval (e.g. radiotracers). In some cases, the approval for primary screening is different to that for clinical use (FDA recently licensed tests explicitly for screening), but in most cases, approval is obtained for diagnostic use and the test is proposed for screening without any other formal approval.

Specific to screening technologies:

Imaging devices may require approval. Substances needed for obtaining images may require additional approval (e.g. radiotracers). In some cases the approval for primary screening is different to that for clinical use (FDA recently licensed tests explicitly for screening), but in most cases approval is obtained for diagnostic use and the test is proposed for screening without any other formal approval.

Other domainsAlso in: Description and technical characteristics of technology

A0021 Assessment element card

Issue: What is the reimbursement status of the technology?

Topic: Regulatory Status

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes7
Clarification
Common to all used applications

Information on national reimbursement status from different countries for the technology as well as the comparators, including key dates and anticipated licensing time frame. Information on full coverage, co-payments and coverage under special circumstances/conditional coverage is useful.

Other domainsAlso in: Description and technical characteristics of technology

A0024 Assessment element card

Issue: How is the disease or health condition currently diagnosed according to published guidelines and in practice?

Topic: Current Management of the Condition

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes8
Clarification
Common to all used applications

The effectiveness of an intervention may vary in populations which are diagnosed by different diagnostic pathways. A sensitive test tends to have low specificity such that there are several people who do not have the condition among the test-positive population. The effectiveness of an intervention in that population may be lower than in a population examined with a less sensitive test (but with more true-positive cases). It is important to point out possible discrepancies between guidelines and actual practice.


A0025 Assessment element card

Issue: How is the disease or health condition currently managed according to published guidelines and in practice?

Topic: Current Management of the Condition

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes9
Clarification
Common to all used applications

It is important to describe whether the technology is an add-on or a replacement for the existing management options, and what the other evidence-based alternatives are. Are there differences in the treatment of diseases at different disease stages? Deviation from evidence-based guidelines may suggest over- or under-use of the technology. Identification of practice variations due to the differences in the forms, stages or severity of the disease may imply differences in the quality of health care. Different stages of disease may call for different therapeutic procedures (e.g. aortic insufficiency is first treated with medication, and at a certain point of cardiac structural changes, an operation is preferred). Provide an overview of other treatment alternatives. Likewise, diagnostic or monitoring methods used for various diseases may vary depending on the stage of disease.


A0007 Assessment element card

Issue: What is the target population in this assessment?

Topic: Target Population

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes10
Clarification
Common to all used applications

Relevant for all assessments: both safety and effectiveness depend largely on the subpopulation towards which the intervention is targeted. The technology may be used for all patients with the condition, or only those in the early stages, or at a specific severity level or for those at moderate risk of having the condition. Personalised medicine divides the target population into even smaller units when targeting the intervention to specific subgroups based on e.g. genetic profile. Use the target population defined in the scope of the project, and consider adding further details and description of who defined the selected subgroups and why. Point out e.g. if certain populations should be excluded from the analysis. 


A0023 Assessment element card

Issue: How many people belong to the target population?

Topic: Target Population

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes11
Clarification
Common to all used applications

This information can be used to give an idea of the resource requirements in general for implementing the technology. Estimates of incidence and prevalence should be provided. Estimates of likely relevant increases or decreases in the size of the target population in the future should also be included.


A0011 Assessment element card

Issue: How much are the technologies utilised?

Topic: Utilisation

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes12
Clarification
Common to all used applications

Provide national estimates for current and future utilisation rates for the indication under assessment, for both the technology under assessment and its comparators. Variations in utilisation reflect market access, sales figures, actual usage in hospital level and adherence to the use of the technology by professionals and patients. Data on current and previous utilisation reflect the phase of the technology (experimental, emerging, established or obsolete). This also has implications for the availability of evidence and the level of uncertainties.

Specific to screening technologies :

What is the current rate of screening adherence?


Description and technical characteristics of technology

Assessment elements

B0001 Assessment element card

Issue: What is the technology and the comparator(s)?

Topic: Features of the technology

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes1
Clarification
Common to all used applications

This is relevant in all assessments. Use the descriptions of the technology and comparator(s) defined in that scope and elaborate them here in more detail.

Technology may include a single device, a questionnaire, imaging or sequence of technologies. The HTA may address one or several similar technologies. Describe separately for the technology and the comparator: the type of device, technique, procedure or therapy, its biological rationale and mechanism of action; and also, describe how the technology differs from its predecessors, and the various current modifications or different sponsor’s products, especially if the differences affect performance.


A0020 Assessment element card

Issue: For which indications has the technology received marketing authorisation or CE marking?

Topic: Regulatory Status

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes2
Clarification
Common to all used applications

There are both international and national market authorisation systems. The systems differ between countries and are more established for pharmaceuticals than for medical devices. An overview of the status with regard to key processes, e.g. CE marking, EMA/US Food and Drug Administration (FDA) approval is recommended. In case the technology is authorised under a different process, e.g. adaptive licensing or conditional reimbursement, information should be presented. Also, information on national data and an analysis of possible discrepancies can be useful.

Specific to diagnostic technologies:

Imaging devices may require approval. Substances needed for obtaining images may require additional approval (e.g. radiotracers). In some cases, the approval for primary screening is different to that for clinical use (FDA recently licensed tests explicitly for screening), but in most cases, approval is obtained for diagnostic use and the test is proposed for screening without any other formal approval.

Specific to screening technologies:

Imaging devices may require approval. Substances needed for obtaining images may require additional approval (e.g. radiotracers). In some cases the approval for primary screening is different to that for clinical use (FDA recently licensed tests explicitly for screening), but in most cases approval is obtained for diagnostic use and the test is proposed for screening without any other formal approval.

Other domainsAlso in: Health Problem and Current Use of the Technology

B0002 Assessment element card

Issue: What is the claimed benefit of the technology in relation to the comparator(s)?

Topic: Features of the technology

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes3
Clarification
Common to all used applications

This issue is especially relevant in new technologies with uncertain expectations and claims of benefit.

Describe the following aspects:

  • How is it expected to be an improvement over previous/existing technologies used for the same health problem?
  • The expressed objectives for the implementation of the technology in health care; what are the claimed objectives (e.g. increased safety, health benefit, accuracy or patient compliance), and is it intended to replace or to supplement existing technologies?

B0003 Assessment element card

Issue: What is the phase of development and implementation of the technology and the comparator(s)?

Topic: Features of the technology

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes4
Clarification
Common to all used applications

Most technologies will be introduced at approximately the same time in several countries. This information is relevant for the assessment while the evidence base may change rapidly for technologies that are at an earlier stage in their development. It is also important to establish whether new versions of the technology with substantial improvements are expected in the near future. For end-users it is useful to know whether new versions or adaptations of the technology are expected in the near future.

Describe the following aspects:

  • Is the technology an innovation?
  • When was it developed?
  • Is the technology only partially innovative (i.e. a modification of an existing technology), and in that case, is it possible to specify the degree of innovation the technology may represent?
  • When was the technology introduced into health care?
  • Is the technology an already established one, but now used in a different way, for instance for a new indication? This issue may be less relevant for new pharmaceuticals.
  • Is it experimental, emerging, established in use or obsolete (implementation level)?
  • Is the technology field changing rapidly?
  • How does this technology differ from its predecessors (other technologies used for similar purposes)?
  • Are there new aspects that may need to be considered when applying it?
  • Is there evidence that the technology works (or is used) outside its current indication area or produces incidental findings that can have consequences relevant to effectiveness, safety, organisational, social and ethical domains?

B0004 Assessment element card

Issue: Who administers the technology and the comparator(s) and in what context and level of care are they provided?

Topic: Features of the technology

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes5
Clarification
Common to all used applications

This issue should be answered in case there is a relevant difference between the technology and the comparator. Describe the following aspects:

  • Which professionals (nurses, doctors, and other health-care professionals) apply and make decisions about starting or stopping the use of the technology?
  • Do the patients themselves, or their carers, administer the technology?
  • Who can select the patients, make referrals, decide to initiate the use of the technology or interpret the outcome?
  • Are there certain criteria (skills, function, training requirements) for the patients or professionals who will administer the technology?

Describe the level of care in which the technology is used: self-care, primary care, secondary and tertiary care. If secondary or tertiary care, describe whether it is intended to be used in the outpatient or inpatient setting.

Its role in the management pathway can be presented as a replacement, an add-on or for triage.


B0008 Assessment element card

Issue: What kind of special premises are needed to use the technology and the comparator(s)?

Topic: Investments and tools required to use the technology

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes6
Clarification
Common to all used applications

This issue should be answered in case there is a relevant difference between the technology and the comparator.

Many technologies require purpose-built premises, such as radiation-secured areas, Faraday cages, dressing rooms for the patient, or specific premises equipped with fume cupboards for storage and reconstitution of chemotherapy pharmaceuticals. Typical premises in primary or secondary care may differ markedly from country to country. A clear description of necessary facilities is needed instead of a general statement (e.g. to be used in hospitals only).

This issue may be less relevant for pharmaceuticals.


B0009 Assessment element card

Issue: What equipment and supplies are needed to use the technology and the comparator(s)?

Topic: Investments and tools required to use the technology

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes7
Clarification
Common to all used applications

This issue should be answered in case there is a relevant difference between the technology and the comparator.

Examples are syringes, needles, pharmaceuticals and contrast agents, fluids, bandages and tests to identify patients eligible for treatment.


A0021 Assessment element card

Issue: What is the reimbursement status of the technology?

Topic: Regulatory Status

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes8
Clarification
Common to all used applications

Information on national reimbursement status from different countries for the technology as well as the comparators, including key dates and anticipated licensing time frame. Information on full coverage, co-payments and coverage under special circumstances/conditional coverage is useful.

Other domainsAlso in: Health Problem and Current Use of the Technology

Safety

Assessment elements

C0008 Assessment element card

Issue: How safe is the technology in relation to the comparator(s)?

Topic: Patient safety

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes1
Clarification
Common to all used applications

Here, one should identify and describe the direct harms of the use and the administration of the technology and the comparator(s). Highlight the differences in the most important risks (i.e. the most severe and frequent harms) of the technology and its comparator and consider if there are uncertainties with regard to safety because of small numbers and/or short duration of follow-up.

Consider:

  • What is the frequency and what are serious adverse events (SAEs) of the technology in relation to the comparator(s)?
  • What are the most frequent AEs of the technology in relation to the comparator(s)?
  • What is the frequency of discontinuation of treatment due to AEs of the technology in relation to the comparator(s)?
  • What is the frequency of SAEs leading to death for the technology in relation to the comparator(s)?
  • What is the frequency of unexpected AEs in participants and comparison groups?     

C0002 Assessment element card

Issue: Are the harms related to dosage or frequency of applying the technology?

Topic: Patient safety

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes2
Clarification
Common to all used applications

This is usually relevant with pharmaceuticals but may also be relevant with medical devices and procedures. Before marketing authorisation, it is relevant to report harms at any dose. After market access, the harms at doses normally used in practice are most relevant for HTAs. Information should be included if safe use of the technology is sensitive to even small changes of the dose because this may have implications for the training and organisation of care. The potential for accumulated harm due to repeated dosage or testing should also be considered.

Specific to pharmaceuticals:

For further information, see guideline on Endpoints used for REA  – Safety.


C0004 Assessment element card

Issue: How does the frequency or severity of harms change over time or in different settings?

Topic: Patient safety

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes3
Clarification
Common to all used applications

This issue is especially relevant for new or evolving technologies where there are considerable uncertainties in the safety evidence, and in technologies with steep learning curves. How does the safety profile of the technology vary between different generations, approved versions or products? Is there evidence that harms increase or decrease in different organisational settings?


C0005 Assessment element card

Issue: What are the susceptible patient groups that are more likely to be harmed through the use of the technology?

Topic: Patient safety

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes4
Clarification
Common to all used applications

Typically, people with comorbidities and co-medication, pregnancy, intolerances, specific genetic profiles, elderly people, children and immunosuppressed patients. Are there any relevant contraindications or interactions with other technologies?


C0007 Assessment element card

Issue: Are the technology and comparator(s) associated with user-dependent harms?

Topic: Patient safety

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes5
Clarification
Common to all used applications

Describe here what is known of the harms caused by the properties or behaviour of professionals, patients or other individuals who apply or maintain the technology. Is there e.g. a noteworthy risk of malfunction of a device, due to deficient user training or personal attitude; or a risk of errors related to reconstitution, dosage, administration or storage of medicines, that may have serious consequences; or, is there a risk of addiction? Describe what is known of the learning curve, intra- or inter-observer variation in interpretation of outcomes, errors or other user-dependent concerns in the quality of care. For further information, see guideline on Endpoint used for REA  – Safety.


B0010 Assessment element card

Issue: What kind of data/records and/or registry is needed to monitor the use of the technology and the comparator(s)?

Topic: Safety risk management

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes6
Clarification
Common to all used applications

Describe the data that needs to be collected about the care process, professionals involved, patients and their health outcomes. These include clinical indications, specified populations, prescriber information, inpatient or outpatient use, test results, review period and health outcomes. In case of new technologies, consult the EVIDENT database.

Specific to pharmaceuticals: refer to the SPC and EPAR.

Describe the general importance of having a registry to monitor the use of this particular technology and the comparator. Are there existing registries that should be used, or should a registry be established, to collect the necessary data to monitor safety or true life effectiveness? Provide national examples. Sometimes registries are connected with the risk-sharing scheme that innovative pharmaceuticals require in some countries. Notice also the requirements of pharmacovigilance monitoring.


C0006 Assessment element card

Issue: What are the consequences of false-positive, false-negative and incidental findings generated by using the technology from the viewpoint of patient safety?

Topic: Patient safety

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes7
Clarification
Common to all used applications

What are the consequences of false-positive, false-negative and incidental findings generated by using the technology?

False-negative test results (Type II error) identify sick people incorrectly as healthy with the possible consequence of incorrectly rejected or delayed treatment. The volume of false-negative test results can be estimated to be 1-sensitivity of the test.

False-positive test results (Type I error) identify healthy people incorrectly as sick with the possible consequence of over-treatment. The volume of false-positive test results can be estimated to be 1-specificity of the test. Incidental findings in tests carry major risk of over-diagnosis and over-treatment.

Specific to screening technologies :

In screening programmes, one should consider separately the false-negative screening test results and the subsequent false-negative diagnostic test results.


Clinical Effectiveness

Assessment elements

D0001 Assessment element card

Issue: What is the expected beneficial effect of the technology on mortality?

Topic: Mortality

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes1
Clarification
Common to all used applications

Report the results both in absolute terms and relative to the comparator. Mortality is the preferred, objective endpoint for assessments of life-threatening conditions. Overall mortality, disease-specific mortality and mortality due to causes other than the target disease are distinguished. Several methods are used to adjust mortality rates and survival curves, e.g. relative survival (observed versus expected survival), which can be quite misleading; and HR (derived from a statistical method comparing the median survivals in the two groups). Note that progression-free survival is not a mortality endpoint; it describes the time from the beginning of an intervention until a patient shows signs of disease progression.

Overall mortality refers to all-cause mortality. It is expressed either as mortality rates (incidence in given population, at given time point and usually risk standardised), or survival (number of people alive for a given period after an intervention).

Disease-specific mortality is a proportion of all-cause mortality. It should be noted that even if a given treatment reduces one type of death, it could increase the risk of dying from another cause, to an equal or greater extent. Disease-specific mortality is typically presented as rates and as age- and risk-adjusted measures such as HR. It is a frequently used endpoint in screening trials, where it is considered to be subject to bias. Consider separately, absolute mortality (compared with placebo or waiting list) and mortality relative to the comparator.

Mortality due to causes other than the target disease includes all unintended, either positive or negative effects of the technology on mortality. There may be e.g. a decrease of mortality of another disease observed or suspected; or increased mortality due to accidents or hazardous medical interventions after false-positive or incidental test results. Supplement with relevant data if differences can be expected for specific subgroups.

Disease-specific mortality is a proportion of the all-cause mortality. It should be noted that even if a given treatment reduces one type of death, it could increase the risk of dying from another cause, to an equal or greater extent. Disease-specific mortality is typically presented as rates and as age- and risk- adjusted measures such as HR. It is a frequently used endpoint in screening trials, where it is considered to be subject to bias. Supplement with relevant data if differences can be expected for specific subgroups.

Specific to diagnostic technologies:

In diagnostic and screening technologies, this issue refers to the expected beneficial effect of the test-treatment chain.

Specific to screening technologies:

In diagnostic and screening technologies, this issue refers to the expected beneficial effect of the test-treatment chain. With screening tests, one should consider the effects of lead-time bias, length-time bias and selection bias to the mortality.


D0005 Assessment element card

Issue: How does the technology affect symptoms and findings (severity, frequency) of the disease or health condition?

Topic: Morbidity

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes2
Clarification
Common to all used applications

Report the results both in absolute terms and relative to the comparator. Describe the efficacy and effectiveness of the technology on relevant disease outcomes (symptoms and findings). Outcomes such as function, QoL and patient satisfaction are reported in other assessment elements of this domain. Report changes in severity, frequency and recurrence of symptoms and findings.

Supplement with relevant data if differences can be expected for specific subgroups. (see guideline on Endpoints used for REA  – Clinical endpoints).


D0006 Assessment element card

Issue: How does the technology affect progression (or recurrence) of the disease or health condition?

Topic: Morbidity

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes3
Clarification
Common to all used applications

Report the results both in absolute terms and relative to the comparator. Report here efficacy and effectiveness outcomes such as complete cure, progression-free survival, time-to-event, next stage of disease, relapse. Describe here the duration of treatment effect on symptoms and findings: permanent, short-term, long-term, intermittent, undulating.

Supplement with relevant data if differences can be expected for specific subgroups.


D0011 Assessment element card

Issue: What is the effect of the technology on patients’ body functions?

Topic: Function

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes4
Clarification
Common to all used applications

Report the results both in absolute terms and relative to the comparator. International classification of function proposes the following categories for body functions: mental, sensory and pain, voice and speech, cardiac, respiratory and immune functions, genitourinary and reproductive functions, movement-related, and skin functions. Report the results both in absolute terms and relative to the comparator. Supplement with relevant data if differences can be expected for specific subgroups.


D0016 Assessment element card

Issue: How does the use of the technology affect activities of daily living?

Topic: Function

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes5
Clarification
Common to all used applications

Report the results both in absolute terms and relative to the comparator. Supplement with relevant data if differences can be expected for specific subgroups. Activities of Daily Living (ADL) is used in rehabilitation as an umbrella term relating to self-care, comprising those activities or tasks that people undertake routinely in their everyday life. The activities can be subdivided into personal care and domestic and community activities.


D0012 Assessment element card

Issue: What is the effect of the technology on generic health-related quality of life?

Topic: Health-related Quality of life

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes6
Clarification
Common to all used applications

Report the results both in absolute terms and relative to the comparator. Supplement with relevant data if differences can be expected for specific subgroups.

Health related quality of life (HRQoL) is typically measured with self- or interviewer-administered questionnaires to measure cross-sectional differences in QoL between patients at a point in time (discriminative instruments) or longitudinal changes in HRQoL within patients during a period of time (evaluative instruments). Two basic approaches to HRQoL measurement are available: generic instruments that provide a summary of HRQoL; and specific instruments that focus on problems associated with single disease states, patient groups, or areas of function. Generic instruments include health profiles and instruments that generate health utilities. Each approach has its strengths and weaknesses and may be suitable for different circumstances.


D0013 Assessment element card

Issue: What is the effect of the technology on disease-specific quality of life?

Topic: Health-related Quality of life

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes7
Clarification
Common to all used applications

Report the results both in absolute terms and relative to the comparator. Supplement with relevant data if differences can be expected for specific subgroups. HRQoL is typically measured with self- or interviewer-administered questionnaires to measure cross-sectional differences in QoL between patients at a point in time (discriminative instruments) or longitudinal changes in HRQoL within patients during a period of time (evaluative instruments). Two basic approaches to HRQoL measurement are available: generic instruments that provide a summary of HRQoL; and specific instruments that focus on problems associated with single disease states, patient groups, or areas of function. Generic instruments include health profiles and instruments that generate health utilities. Each approach has its strengths and weaknesses and may be suitable for different circumstances.


D0017 Assessment element card

Issue: Were patients satisfied with the technology?

Topic: Patient satisfaction

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes8
Clarification
Common to all used applications

Describe patients’ overall perception of the value of the intervention and their satisfaction with the treatment. For further information, see guideline on Endpoints used for REA – Clinical endpoints.


D0032 Assessment element card

Issue: How does the test-treatment intervention modify the magnitude and frequency of morbidity?

Topic: Morbidity

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes9
Clarification
Common to all used applications

A more accurate replacement test could improve treatment and effectiveness. A satisfactory triage test may decrease the number of adverse outcomes from another test. An add-on test may increase sensitivity so that more patients receive proper treatment and thus improved outcomes.


D1001 Assessment element card

Issue: What is the accuracy of the test against reference standard?

Topic: Test accuracy

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes10
Clarification
Common to all used applications

Accuracy in terms of sensitivity and specificity, and other measures such as likelihood ratios, pre-test probabilities, SDORs, area under the curve (AUC) or Q.


D1005 Assessment element card

Issue: What is the optimal threshold value in this context?

Topic: Test accuracy

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes11
Clarification
Common to all used applications

Sensitivity and specificity vary according to the threshold value. Optimal combination of sensitivity and specificity defines optimal threshold value. The optimum depends on the consequences of the test results, e.g. whether it does more harm to overlook a case or to treat someone unnecessarily.

Specific to screening technologies:

In screening programmes, one should consider separately the screening test and the subsequent diagnostic tests.


Other aspects

Assessment elements

F0010 Assessment element card

Issue: What are the known and estimated benefits and harms for patients when implementing or not implementing the technology?

Topic: ETH Benefit-harm balance

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes1
Clarification
Common to all used applications

Decisions concerning the implementation of new technologies generally require carefully considering the balance between benefits and harms. Examples of questions that can be answered are:

Who is the right candidate for the technology? What is the balance between benefits and harms? For instance, is the technology estimated to improve health, health-related quality of life, quality of life and/or survival compared to alternative technologies? Can the technology harm individual patients, or any other stakeholder, in any way? How many patients might face harm in order for the technology to have a benefit for one patient? What is the extent of these benefits and harms?

What are the perceived benefits and harms of the technology in the eyes of the patients/users themselves? It might be useful to note that the patient is often the best judge of benefits and harms for themselves.


F0011 Assessment element card

Issue: What are the benefits and harms of the technology for relatives, other patients, organisations, commercial entities, society, etc.?

Topic: ETH Benefit-harm balance

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes2
Clarification
Common to all used applications

Examine the following: Can the technology have positive effects for others apart from the patients in question? Can the technology harm relatives, other patient groups, organisations, commercial entities, society, etc.? Some technologies have the potential to unfold unwanted or harmful effects not only on the patients that the technology is directly applied to but also indirectly on others. For example results of genetic tests may negatively interfere with the family planning and social life of not only the individual being tested but also of his or her relatives. Another example is how the caregivers’ burden and well-being will be affected by the technology.

Benefits and harms to individuals must be balanced with benefits and harms that can have impact on society as a whole (social utility, maximizing public health). These harmful effects may manifest themselves in the physical, social, financial or even other domains of life.

Changes in the availability of new, more effective technologies may significantly alter the requirements placed on the health care system. Is the symbolic value of the technology of any moral relevance?

Another relevant question is how the assessed technology relates to more general challenges of modern medicine (over-diagnosis, medicalization)?

Table 1 in the process description can be used to describe benefits and harms.


F0003 Assessment element card

Issue: Are there any other hidden or unintended consequences of the technology and its applications for patients, relatives, other patients, organisations, commercial entities, society etc.?

Topic: ETH Benefit-harm balance

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes3
Clarification
Common to all used applications

The technology may be used for other indications (extended use) or other purposes, e.g., in combination with other technologies (unintended use). It may have side-effects in addition to those following from the intended use.

Consider not only the consequences of the formal intended use of the technology, but also the ethical consequences of unintended and extended use. If unintended consequences are not well-known, they should be speculated and elaborated upon. Evaluate the intended purpose and uses of the technology against the likely uses and consequences of the technology in reality.

The mode of delivery, the need for laboratory tests or clinical follow-up to ensure safe and effective dosage and the way of delivery (at home, outpatient or in-patient) may have a large impact on the health care processes, systems and on individuals. They may also change the concepts of disease and normality (e.g. change an untreatable cancer into a chronic disorder or changing the border values when the concept of normality also changes).

New technologies tend to lead to new areas of inventions and give rise to new ethical questions (e.g., in vitro fertilisation (IVF) and development of genetic testing has led to questions of preimplantation genetic diagnostics (PGD)). As pre-symptomatic screening tests have become available, the healthcare system has to be prepared to handle moral issues raised by true positive and false negative findings.

Another relevant question is whether or not there will be a moral obligation related to the implementation, withdrawal, or use of the technology (e.g. check-ups or alternative procedures).

Specific to Diagnostic Technologies

Diagnostic technologies may also have effects on relatives. Not only genetic tests, but all diagnoses of hereditary disorders, also provide knowledge about relatives. Diagnostic information may also affect social relations (e.g. STD)’.

Specific to Pharmaceuticals

Pharmaceuticals have usually been designed and studied for a specific and defined group of patients, but they may be used for a larger group (variation in age and severity of the disorder and persons with comorbidities and/or need for other pharmaceuticals). Expensive pharmaceuticals (orphan disorders, new cancer treatments) and the prescription of pharmaceuticals according to genetic profiles challenge the equal and just use of health care resources. The health care system has to be prepared to handle moral issues raised by the new, expensive possibilities to treat rare, otherwise non-treatable disorders and to prolong life in chronic disorders.

Specific to Screening Technologies

Screening positive and being diagnosed with the disease may have effects on relatives as all diagnoses of hereditary disorders also provide knowledge about relatives. Screening results may also affect social relations.


F0104 Assessment element card

Issue: Are there any ethical obstacles for evidence generation regarding the benefits and harms of the intervention?

Topic: ETH Benefit-harm balance

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes4
Clarification
Common to all used applications

When assessing benefits and harms of an intervention there may be ethical obstacles to the conduct of further research in order to strengthen the scientific basis. This concerns issues like the following:

• When clinical experience shows that the intervention has an effect on a group for whom there are no treatment alternatives and it would thus be ethically unacceptable to conduct a study in which the comparative group would be denied the procedure,

• In the case of a vulnerable group of subjects who are difficult to study,

• Where specific integrity problems would arise if research were to be conducted.


F0005 Assessment element card

Issue: Is the technology used for individuals that are especially vulnerable?

Topic: ETH Autonomy

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes5
Clarification
Common to all used applications

Clarify the right and justification to use the technology on vulnerable persons. Persons who are vulnerable could, for example, be pregnant women (in which case their unborn child needs to be protected), critically ill patients or individuals that have reduced decision-making capacity (children, persons with cognitive disabilities or patients that due to their illness/state have limited decision making capacity). Who has the right to balance the benefit against possible harm in these situations? On what grounds can these decisions be made? Is the technology so valuable, as to justify its use on people who cannot give informed consent?


F0004 Assessment element card

Issue: Does the implementation or use of the technology affect the patient´s capability and possibility to exercise autonomy?

Topic: ETH Autonomy

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes6
Clarification
Common to all used applications

Many technologies can alter a person´s self-determination. The technology may interfere with a patient’s right to autonomy, directly or indirectly, by influencing/subtracting their decisional capacity. However, patients have, in most cases, a right to autonomy, i.e. a right to be self-governing agents. This means they possess both the right to decide (not) to use/participate, and the right to receive relevant information. Drugs for sedation and surgical treatment of severely ill patients are examples where patient autonomy may be reduced.

Technology may require users/patients to behave in a certain way (e.g. dietary restrictions for faecal blood test). In order to be able to decide autonomously, the user/receiver of the technology should understand all alternative treatments or different therapeutic paths following test results. They should be able to make informed consent at every step.

The practical challenge with treatment technologies is that, in order to be fully autonomous, the patient should understand not just direct risks of the treatment, but also all alternatives, whether side-effects take place, and how these can affect the living quality or choices (e.g. car driving, nutrition).


F0006 Assessment element card

Issue: Is there a need for any specific interventions or supportive actions concerning information in order to respect patient autonomy when the technology is used?

Topic: ETH Autonomy

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes7
Clarification
Common to all used applications

Focus on the following: Is the common professional practice of discussing the technology with patients enough, or is special information needed to decide on this technology? Can the technology entail special challenges/risks that the patient/person needs to be informed of? Should the patient be explicitly informed, for example, that false positive results of a test may lead to unnecessary further investigations and treatments, sometimes with serious harms? An example is screening programmes for early identification of life-threatening situations that may have life-threatening side effects, such as invasive surgery with risk of death. Technology used for off-label use may have unexpected severe side-effects (e.g. patients with comorbidities or children).

The information should enable the user/receiver of the technology to understand the technology and its associated risks/challenges. It should be in accordance to their personal values and intellectual capacity, thereby enabling users to decide accordingly. The patient should be explicitly informed, for example, that the treatment may have serious side effects, may have an effect on personality or lead to increased need of sleep or serious weight gain. They should also be informed of when the mode of delivery or action may affect their daily life (e.g. no car driving allowed, restricted travelling).


F0007 Assessment element card

Issue: Does the implementation or withdrawal of the technology challenge or change professional values, ethics or traditional roles?

Topic: ETH Autonomy

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes8
Clarification
Common to all used applications

Technologies may change the patient-physician relationship, challenge professional autonomy or otherwise interfere with professional ethics and values. The patient-physician relationship is traditionally based on mutual trust, confidentiality and professional autonomy so that individual treatment decisions can be made in the best interest of the patient. Technologies that interfere with core values and principles of medical and professional ethics challenge the professional integrity of the physicians or other healthcare professionals (e.g. screening for drug abuse when use is denied). Technologies that are aligned with professional ethics are more likely to be implemented successfully. For example, people may ask for the technology for many reasons, while the professionals may see them as unnecessary and even potentially harmful (e.g. antibiotics, sleep medicine, antidepressants, whole body MRI scans).


F0008 Assessment element card

Issue: Does the implementation or use of the technology affect human dignity?

Topic: ETH Respect for persons

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes9
Clarification
Common to all used applications

Especially those technologies that are applied to persons with reduced autonomy (children, mentally impaired, severely ill) may violate a person's dignity, i.e. challenge the idea that all human beings have intrinsic value, and should thus not be seen as means to others ends. Labelling people as result of using the technology may also threaten their dignity.

Some technologies may cause healthy people to be labelled as sick (e.g. PSA for prostate cancer) or otherwise less worthy, abnormal, less clean, etc. For instance labelling people as needing psychiatric medication for their behavioural difficulties may threaten their dignity. People with physical disabilities may be labelled by prenatal screening programmes, which imply that their handicap is an indication for abortion.


F0009 Assessment element card

Issue: Does the implementation or use of the technology affect the patient’s moral, religious or cultural integrity?

Topic: ETH Respect for persons

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes10
Clarification
Common to all used applications

A technology may challenge integrity by preventing (or having the possibility to prevent) patients to live according to their moral convictions, values, preferences or commitments. It may also interfere with the coherent image or identity of the user’s self. This is especially important to analyse for vulnerable patient groups.

The technology may challenge religious, cultural or moral convictions or beliefs of some groups (e.g. pharmaceuticals produced from human blood given to cultural groups that do not accept blood transfusion, pharmaceuticals used for abortion in cultural groups that do not accept abortion, and assisted reproductive technologies that have separated the concept of genetic, biological and social motherhood).

The technology may change generally or locally accepted social arrangements by challenging traditional conceptions or social roles. For instance, ADHD medication might challenge the integrity of people who value personality, and cochlear implants may be problematic for those who do not see deafness as a disability.

Identifying the conceptions behind the beliefs and values may help put them in perspective when considering the ethical consequences of use and the overall acceptability of the technology. When possible, considering other acceptable alternatives for the affected groups of users is important. Use of the technology can also be detrimental to integrity if it is associated with discouraging honesty or ethical conduct, e.g., systems that encourages users to lie about their health state in order to get better service/treatment.


F0101 Assessment element card

Issue: Does the technology invade the sphere of privacy of the patient/user?

Topic: ETH Respect for persons

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes11
Clarification
Common to all used applications

The sphere of privacy can be invaded both virtually and physically. Describe, e.g., these issues: Does the technology affect the population’s possibility to have control over personal information? Is dissemination or gathering of information regarding the individual patient or the population justified? Is cooperation and sharing of information with professional groups outside the health services needed? Is the handling of personal information reasonable, given the purpose of using the technology? Is the technology more or less invasive than the alternatives, regarding the physical body and/or the spatial sphere? Is a violation of the privacy of the patient or population necessary and reasonable to achieve desired outcomes?


F0012 Assessment element card

Issue: How does implementation or withdrawal of the technology affect the distribution of health care resources?

Topic: ETH Justice and Equity

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes12
Clarification
Common to all used applications

Many technologies imply substantial costs, sometimes covered with resources from other areas. A new technology may require re-allocation of human resources, funding and training. A large re-allocation of resources may seriously jeopardise other patient groups (e.g. new technology that requires human resources in acute care or new diagnostic technology that uncovers a large pool of unmet needs for treatment). How this reallocation affects the existing health care system has to be studied. Who will gain and who will lose? Is the prioritisation explicit or implicit?

Specific to Diagnostic Technologies

Diagnostic technologies sometimes acquire significant symbolic value (e.g. foetal ultrasound, PSA) that may create demands for tests that are not justified on health grounds.

Specific to Pharmaceuticals

Pharmaceuticals may acquire abstract promise of health benefit that may create demand that is not justified. Some diagnosis may create demands for pharmaceuticals that are not always justified to be prescribed on health grounds (e.g. large variation in prescribing ADHD medication for children by various countries).

Specific to Screening Technologies

Screening technologies sometimes acquire significant symbolic value (e.g. foetal ultrasound, PSA) that may create demands for tests that are not justified on health grounds.


F0013 Assessment element card

Issue: How are technologies with similar ethical issues treated in the health care system?

Topic: ETH Justice and Equity

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes13
Clarification
Common to all used applications

Clearly presenting how technologies with similar ethical issues are treated in a healthcare system may help in adopting coherent and just health policies, either by applying past precedents to current cases, or by showing that past cases need reconsideration. Similarity is to be defined individually for each technology. The idea is to focus only on the similarities relevant for solving the ethical problems considered important for the current HTA project. The similar ethical problems can be related to similarities in the technology’s medical, technological, economic, social, organisational or legal nature.


H0012 Assessment element card

Issue: Are there factors that could prevent a group or person from gaining access to the technology?

Topic: ETH Justice and Equity / SOC Social group aspects

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes14
Clarification
Common to all used applications

This issue concerns inequality in health. Investing in the reduction of health inequalities is a target of the European Commission, as it contributes to social cohesion and breaks the vicious spiral of poor health being a contributor to, and a result of, poverty and exclusion. Can the technology be applied in a way that gives equal access to those in equal need? How can this be guaranteed? Could potential discrimination or other inequalities (geographic, gender, ethnic, religious, employment, insurance) prevent access?


F0014 Assessment element card

Issue: Does the implementation or use of the technology affect the realisation of basic human rights?

Topic: ETH Legislation / LEG Ethical aspects

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes15
Clarification
Common to all used applications

The basic human rights are most notably declared in the United Nations Declaration of Human Rights (http://www.un.org/en/documents/udhr/ ). They are universal and consider the most important goods, protections and freedoms for mankind. For HTA, perhaps the most relevant are the rights to equality, non-discrimination, safety, adequate standard of living, and healthcare.


F0016 Assessment element card

Issue: Can the use of the technology pose ethical challenges that have not been considered in the existing legislations and regulations?

Topic: ETH Legislation / LEG Ethical aspects

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes16
Clarification
Common to all used applications

Describe whether legislation and regulation to use the technology is fair and adequate. Use of the technology may lead to ethical issues that make current regulations inadequate. Screening and diagnostic technologies are commonly regulated differently than treatments, especially medications. Ethical reflection is essential in order to assess what kind of legislation, regulation or amendments are needed.


F0017 Assessment element card

Issue: What are the ethical consequences of the choice of endpoints, cut-off values and comparators/controls in the assessment?

Topic: ETH Ethical consequences of the HTA

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes17
Clarification
Common to all used applications

Address any risks of the chosen end-points, cut-off values or comparators/controls giving a biased description of the results of the technology.

Clinical effectiveness should ideally be directly related to the disease under treatment. This is not always entirely possible, so other end-points may need to be used (e.g. surrogate markers for preventing a life-threatening disease). In addition, the technology may have several aims (e.g. those related to treating the disease and preventing secondary morbidity).

The choice of cut-off values for sensitivity and specificity should be done considering the moral value of different results – for example, high specificity is required if false positives have serious consequences.


F0102 Assessment element card

Issue: Are there any ethical problems related to the data or the assumptions in the economic evaluation?

Topic: ETH Ethical consequences of the HTA

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes18
Clarification
Common to all used applications

Consider whether there are any ethical problems related to the data or assumptions that have been used in the economic evaluation. An example is whether or not indirect costs have been valued in a fair and adequate way.


F0103 Assessment element card

Issue: What are the ethical consequences of conducting the technology assessment at this point of time?

Topic: ETH Ethical consequences of the HTA

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes19
Clarification
Common to all used applications

At what time of the technology’s lifetime is the technology assessed? What are the consequences of assessing the technology with respect to prioritisation?

Who would (not) get access to the new technology, as result of conducting HTA at this point of time? If there are methodological and ethical obstacles to fill a knowledge gap, what are the consequences for the patient group if the knowledge gap cannot be filled in the (near) future? Should the technology be made available to patients despite the inadequate scientific basis at the time of assessment?


G0001 Assessment element card

Issue: How does the technology affect the current work processes?

Topic: ORG Health delivery process

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes20
Clarification
Common to all used applications

Describe current tasks and work processes. This helps illustrate the whole process as well as the continuity of care across professional and organisational boundaries. Who is doing what in the process?

There are many actors at different levels (intra-organisational, inter-organisational and health care system level) in the process. Continuity should be ensured so that there will be no gaps between the steps of the process.

Explain what kind of changes a new technology could have: it might replace or reduce some activities. In addition, the new technology may have an impact on current pathways of care (e.g. shift towards community care or inpatient care).

Specific to Diagnostic Technologies

The implementation of a new diagnostic test can substantially increase (or decrease) the number of patients in need of treatment thus changing the relationships between different organizations and influencing the health care system as a whole.

Specific to Pharmaceuticals 

Specify the differences in work processes between the new medicine and the comparator. For example, the new medicine does not need routine laboratory unlike the comparator.

Specific to Screening Technologies

Describe how the screening process has been organised, e.g. (1) how the target population is chosen; (2) how and by whom the invitation is carried out (open/fixed invitation, announcement/personal invitation letter); (3) how and by whom the information for consent is given; (4) how, where and by whom the test is executed, 5) how, where and by whom the further investigations and treatment are carried out; (6) how, when, and by whom the follow up services are carried out (e.g. notification of results, recalls, reminders).

It’s important to describe all steps needed in the screening process for the economic evaluation.


G0100 Assessment element card

Issue: What kind of patient/participant flow is associated with the new technology?

Topic: ORG Health delivery process

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes21
Clarification
Common to all used applications

This issue deals with the path of the patient/participant from their own point of view. Describe the patient’s path step by step. This also includes waiting times for diagnosis and/or treatment and waiting times for the analysis of the technology.

Take into account all preparations that patients/participants need to make before and after (e.g. diet before bariatric surgery), as well as the need for self/home monitoring.

In addition, take into account the impact of the technology on current pathways of care. It may e.g. shift towards community care or inpatient care.


G0002 Assessment element card

Issue: What kind of involvement has to be mobilized for patients/participants and important others and/or caregivers?

Topic: ORG Health delivery process

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes22
Clarification
Common to all used applications

This issue concerns the role of patients/participants. A new technology may require task distribution among the people involved in treatment and care. Patients/participants and their important others and/or caregivers may be more actively involved in their own care and treatment, or otherwise, tasks they used to carry out may be taken over by health professionals.

Specific to Diagnostic Technologies 

Some diagnostic tests are used by patients at home, and patients should be taught how to use them.

Specific to Pharmaceuticals 

The way in which the patient administers the medicine and how he is involved in the follow-up (monitoring by patients/participants or by their important others and/or caregivers).

Specific to Screening Technologies 

The screening needs to be organised in such a way that the test and further investigations are easily attainable; e.g. mobile mammography.


G0003 Assessment element card

Issue: What kind of process ensures proper education and training of staff?

Topic: ORG Health delivery process

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes23
Clarification
Common to all used applications

A new technology may require new kinds of professionals or new tasks for existing personnel. This issue deals with how the organisation can ensure proper education. Take into account the effect of training on the management and effectiveness

Implementing a technology can change the nature of the work and thus have influence on job satisfaction.

Specific to Screening Technologies

When implementing new screening technologies, proper staff education has to be ensured. For example, when implementing a screening for foetal abnormalities, it takes time to educate nurses and develop their competence in operating the ultrasound.


G0004 Assessment element card

Issue: What kind of co-operation and communication of activities have to be mobilised?

Topic: ORG Health delivery process

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes24
Clarification
Common to all used applications

Co-operation and communication is crucial in order to achieve a fluent patient pathway. Implementing a technology can demand new co-operation and communication in and outside the organization, e.g. with other hospitals, pharmacies and manufactures. Therefore structure of co-ordination is important. Also, interaction and communication with patients/participants and their important others and/or caregivers could change. Adaptation of self/home monitoring needs close co-operation and fluent communication.

Specific to Screening Technologies

Screening needs close co-operation and fluent communication between all actors involved in the screening process in all steps (e.g. screening unit, laboratory, hospital, registry, participants). There are actors at different levels which make the communication and co-operation challenging, especially when developing a new screening. The information must be fluent, and electronic communication (software) is crucial. Adequate communication with participants and their important others and/or caregivers must be taken into account.

Different kinds of "patient information" could be defined for screening. For example: (1) "promotional/educational information" with the aim of involving the target population and promoting participation; (2) "screening related information" to communicate with participant the "phase related information" in the different phases of the process (e.g. sending invitation; communicating the test results etc.).

Information strategies should be tailored to the specific subgroup of the target population (depending on socio-economic status, cultural background, epidemiological features, etc.). Risk families need special information.


G0012 Assessment element card

Issue: In what way is the quality assurance and monitoring system of the new technology organised?

Topic: ORG Health delivery process

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes25
Clarification
Common to all used applications

A new technology usually affects current quality assurance not only inside the organization but also outside in different health care levels. To assure quality, a monitoring system with standards and indicators is needed; it is also possible for there to be variation in how the quality assurance and monitoring system is implemented. Take into account who the responsible person for quality assurance and for the monitoring system is, and how any follow up has been arranged.

Additionally, consider how quality assurance and the monitoring system affect management and effectiveness.

Other international, national, regional and/or (cross) organisational demands for quality assurance (e.g. quality standards and monitoring) and for registration could also be in place, and this is another thing to keep in mind.

Specific to Pharmaceuticals

Describe what information has to be gathered (clinical indicators, special patient groups, laboratory results).

There are national standards for Pharmacovigilance of pharmaceuticals. Some countries legally oblige physicians to report the adverse events. In most countries, manufacturers are required to submit all the reports of adverse events they receive from healthcare providers to the national authority. A specific monitoring system may be necessary for innovative pharmaceuticals.

Specific to Screening Technologies

Screening involves asymptomatic participants and quality control is therefore crucial. Quality control needs to be systematic at every step of the screening process and throughout the screening programme. Specify the acceptable delay between screening test to test positive result and finally to treatment. Pay special attention to the control in cases where the programme is provided by several entities (e.g. a combination of private and public health care organisations) and when test and further investigations are separated.


G0005 Assessment element card

Issue: How do de-centralisation or centralisation requirements influence the implementation of the technology?

Topic: ORG Structure of health care system

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes26
Clarification
Common to all used applications

The setting (primary - secondary - tertiary care) can vary between different countries depending on the health care system. (De)centralisation could have some economic and qualitative benefits. Centralisation could make the technology more difficult to access. Usually, expensive technologies are centralised to tertiary care units with special educated staff.

Specific to Pharmaceuticals

Report in what health care level the medicine is implemented.

Specific to Screening Technologies

Sometimes, a screening test (for example, a maternal ultrasound) needs specifically trained personnel; this is possible after education/training and a sufficient amount of patients/participants. Centralisation could make screening or further investigation more difficult to access. For example, in foetal screening, timing is important. Decentralisation makes screening more attainable but its quality can weaken.


G0101 Assessment element card

Issue: What are the processes ensuring access to the new technology for patients/participants?

Topic: ORG Structure of health care system

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes27
Clarification
Common to all used applications

Access to care is often measured in terms of utilisation. There are different viewpoints: individual, population-specific and health system factors. Access to care is related to e.g. social, cultural, economic, organisational, relational or geographical factors.

Access to care by wide definition includes availability, accessibility, accommodation, affordability and acceptability.

This issue is related to the issue of acceptability of new technology (G0010)


G0006 Assessment element card

Issue: What are the costs of processes related to acquisition and setting up the new technology?

Topic: ORG Process-related costs

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes28
Clarification
Common to all used applications

Implementing the required changes in e.g. hospital premises may be costly for organisations. Starting costs or running costs of a new technology could be very high. High costs can influence the decision on whether to introduce the new technology. Costs can be divided if some organisation(s) is(are) responsible for the acquisition costs and others for the running costs. Take into consideration any investments at all stages of the process.

Specific to Pharmaceuticals

This includes e.g. devices, special room and software needed for the new medicine.

Specific to Screening Technologies

When constructing a new screening programme, there is a need for many investments (e.g. equipment, education and implementation support, training). Take into account e.g. screening program management requirements and sample types which could have critical economic implications. In addition, patient’s preferences and social aspects can influence the compliance rate and thus may have an impact on ECO domain.


D0023 Assessment element card

Issue: How does the technology modify the need for other technologies and use of resources?

Topic: ORG Process-related costs

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes29
Clarification
Common to all used applications

New (less invasive) interventions may reduce the need for surgical interventions. Some treatments require ongoing monitoring and healthcare visits, including hospitalisation.

Specific to Screening Technologies

Screening tests may cause further diagnostic testing and different treatment due to having detected the disease at an earlier stage.


G0007 Assessment element card

Issue: What are the likely budget impacts of implementing the technologies being compared?

Topic: ORG Process-related costs

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes30
Clarification
Common to all used applications

Whenever a technology is introduced, there will be an impact on health care budgets. It is possible to undertake a budget impact analysis which attempts to examine the likely impact of introducing a technology on finances or budgets from e.g. the perspective of different payers. Different payers include: government-level institutions; regions; municipalities; employers; insurance companies and patients/participants. The relevant perspective from which to estimate budget impact may change during different phases of the management process, and incentives are connected to this issue.

For example: What kind of incentives does the budget impact impose on different actors? How might this potentially impact on each organisation? What is the estimated net financial (e.g. annual) cost of introducing the technology? Budget impact analysis provides data to inform an assessment of the affordability of a technology. It also provides a service planning tool to inform decisions about taking the technology into use.

Specific to Screening Technologies

The relevant ‘payer’ can change during the screening process (e.g. a municipality pays for the screening test but then a hospital district pays for further investigations). Screening is usually free of charge for people, but sometimes participants have to pay e.g. a hospital fee for further investigations. Note that when initiating a new screening programme, initial cost outlays may be necessary.


G0008 Assessment element card

Issue: What management problems and opportunities are attached to the technology?

Topic: ORG Management

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes31
Clarification
Common to all used applications

The issue concerns the administrative/managerial questions of technology: management of resources (e.g. investments), co-ordination (in relation to different levels and different steps of the process), establishment of objectives, monitoring and control (how quality assurance affects management or effectiveness), evaluation and sanctioning. Take into account the relevant data/information management systems connected to each of these points.

This issue also includes risk management and safety issues (e.g. safety of personnel).


G0009 Assessment element card

Issue: Who decides which people are eligible for the technology and on what basis?

Topic: ORG Management / CUR Utilisation

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes32
Clarification
Common to all used applications

Provide information on the key actors who decide on the use of the technology. Do most important decisions take place on the national level (e.g. population screening) or are they, for example, made by individual professionals (e.g. surgical method for a specific disease)? How is the decision made – are there some documented criteria?

Information about the possible variations on the decision level and decision criteria has ethical implications.

This issue may be especially important in the context of rare diseases.

This issue is related to the issue of work processes (G0001).

Specific to Pharmaceuticals

Companion diagnostics (tests or measurements) assist physicians in making treatment decisions for their patients by elucidating the efficacy and/or safety of a specific pharmaceutical or a class of pharmaceuticals for a targeted patient group or sub-groups. Specify and explain how companion diagnostics should be used to identify eligible patients.

Specify the criteria for higher risk groups of patients such as the elderly and children.

Specific to Screening Technologies 

Decisions about people eligible for screening are made in the beginning of the screening. Usually, the decisions have been made nationally or regionally (in municipalities) but also locally (by employers). In systematic screening, the screening unit does not make decisions about who is eligible for screening. The management of positive test results needs systems to guarantee proper follow-up and, sometimes, case specific evaluation. In this topic responsibilities should be identified.


G0010 Assessment element card

Issue: How is the technology accepted?

Topic: ORG Culture

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes33
Clarification
Common to all used applications

Look at acceptance from the perspectives of organisation, personnel, and patients/participants. The organisational view can be separated into the intra-organisational (primary care), inter-organisational (secondary care) and health care system level. Acceptance can vary on different levels and with different actors. Alternative ways to introduce a new technology into an organisation could cause problems such as resistance among staff and dysfunction of processes.

Acceptability is related to access to care.

Specific to Screening Technologies

Acceptance may vary even within one specific screening process; for example, in foetal screening someone might accept an ultrasound but not a chromosomal (serum) test. When describing organisational acceptance, an example would be how sometimes screening may consist of elements which are not suitable for the image of the organisation.

Screening is voluntary, and for persons who are eligible for screening there is no wrong decision, regardless of whether they decide to participate or not. Giving comprehensible information on pros and cons of screening is important, and the staff’s communicational skills may influence a patient to accept screening.

A patient’s/participant’s preferences on screening could influence the compliance rate and thus may have impacts on ECO domain.


G0011 Assessment element card

Issue: How are the other interest groups taken into account in the planning/implementation of the technology?

Topic: ORG Culture

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes34
Clarification
Common to all used applications

It may be useful to know who the possible stakeholders are, as well as what kind of co-operation exists and what kind of interaction is needed. The stakeholders could e.g.be the pharmaceutical industry and companies offering technologies for screening, authorities national or regional, registries, administrative parties, municipalities, policy makers/decision makers, staff groups, GPs/primary care physicians and patient organisations. One may also ask: Has the patient organisation taken part into the evaluation process? Has it been involved from the beginning (in the planning) or in the later stages, for example as commentator?


H0200 Assessment element card

Issue: What are the experiences of living with the condition?

Topic: SOC Patients’ perspectives

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes35
Clarification
Common to all used applications
This issue concerns the patient’s everyday life living with the disease, e.g. familiar, social and work related roles, ability to manage relationships with other people in a socially appropriate manner in major areas of life, ability to take care of one self etc. It includes:
  • Illness and treatment burden
  • Limitations to activities of daily living: work, family, social life, ability to care for oneself, leisure activities
  • Psychological issues: stigma, anxiety, fear, social acceptance
  • Financial implications, aids needed to support daily living

H0100 Assessment element card

Issue: What expectations and wishes do patients have with regard to the technology and what do they expect to gain from the technology?

Topic: SOC Patients’ perspectives

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes36
Clarification
Common to all used applications
This issue concerns what patients and care-givers expect to gain from the technology; it includes e.g.:
  • Improved survival, delayed progression?
  • Improvement of specific symptoms (e.g. fatigue, incontinence, diarrhoea, mobility etc.)?
  • Improvements/changes related to implications of daily living, social and psychological issues by using the current technology
  • What size of effect is important?

H0006 Assessment element card

Issue: How do patients perceive the technology under assessment?

Topic: SOC Patients’ perspectives

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes37
Clarification
Common to all used applications
This issue is about the patients’ attitudes, perceptions, preferences, satisfaction and expectations to the technology. This covers whether any positive or negative issues arise as a consequence of using the technology e.g. feelings of unity or empowerment, existential experiences (e.g. insecurity, worries, hope, anxiety, stigmatisation, social status, courage to face life, satisfaction, changes in self-conception). It includes:
  • What understanding do patients have of the technology?
  • What implications – positive and negative – does the technology have regarding activities of daily living, social life, psychological issues, financial implications, support and resources (practical, physical, emotional) and requirement in order for the patient to use the technology with satisfactory results?
  • Can the technology be used/taken easily?
  • What treatment benefits could be improved?
  • What side effects are most difficult to manage?

H0002 Assessment element card

Issue: What is the burden on care-givers?

Topic: SOC Patients’ perspectives

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes38
Clarification
Common to all used applications

Describe who the important other people are that are involved in the use of the technology, in addition to the patients (parents, children, friends, people at work place etc.). What kind of support (practical, physical, emotional, financial, nurturing, personal) do care-givers mobilize? It includes e.g.:

  • What challenges do care-givers face when supporting patients to manage their condition and receive care?
  • How do care-givers perceive the new technology; what challenges and benefits might it offer?
  • What support and resources need to be mobilised in order for the patient ton use the technology satisfactorily?

Specific to Screening Technologies 

E.g. the results of screening or genetic and prenatal testing, may affect relatives.


H0201 Assessment element card

Issue: Are there groups of patients who currently don’t have good access to available therapies?

Topic: SOC Social group aspects

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes39
Clarification
Common to all used applications
This issue concerns inequality in health. Investing in the reduction of health inequalities is a target of the European Commission, as it contributes to social cohesion and breaks the vicious spiral of poor health being a contributor to, and a result of, poverty and exclusion. It includes e.g.: Do available therapies give rise to inequality in access and use of the health care? Are there special groups discriminated, e.g.:
  • Children, older people, certain age groups
  • People with a specific genetic mutation, people with disabilities
  • People living in remote areas, ethnic groups etc.
  • People with a certain type of the disease

H0202 Assessment element card

Issue: How are treatment choices explained to patients?

Topic: SOC Communication aspects

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes40
Clarification
Common to all used applications
This issue is about patient participation, incl. what support or limit use of the technology in relation to communication aspects. It includes e.g.:
  • Do patients with the condition have good information sources to explain the condition and treatment options to them?
  • Are there good decision aids available to help shared decision making between patients and doctors and/or other health personnel?
  • Do patients feel themselves involved in a sufficient and appropriate way?

H0203 Assessment element card

Issue: What specific issues may need to be communicated to patients to improve adherence?

Topic: SOC Communication aspects

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes41
Clarification
Common to all used applications
This issue is about communication and how it influences the use of the technology, e.g.:
  • Preparation in advance of intervention, dosage instructions, side effects etc.
  • Is there information which patients would need that are not usually available?

I0002 Assessment element card

Issue: What kind of legal requirements are there for providing appropriate information to the user or patient and how should this be addressed when implementing the technology?

Topic: LEG Autonomy of the patient

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes42
Clarification
Common to all used applications

Describe the rules and recommendations for what patients should know about the implications of using or not using the technology. The right of the patient not-to-know may also be important, as well as the patient's right to complain.

These rules are likely to be helpful for the persons involved in implementing the technology to prepare proper information and counselling. This information may be particularly important with technologies carrying high risks of harm, technologies with the potential to provide information that is not directly relevant to the condition being tested, and in emergency situations in which the patient does not usually have sufficient time to consider the treatment decision.

Specific to Screening Technologies

As screening programs target symptom-free and healthy people, it is particularly important that the individuals are aware of the potential benefits and harmful consequences of attending a screening test. The information provided for individuals attending screening should therefore not be persuasive.


I0034 Assessment element card

Issue: Who is allowed to give consent for minors and incompetent persons?

Topic: LEG Autonomy of the patient

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes43
Clarification
Common to all used applications

According to law, a minor is a person under a certain age, usually the age of majority, which legally demarcates childhood from adulthood. The age of majority depends upon jurisdiction and application, but is generally 18. An incompetent person may be defined as one whose mind is unsound, deranged, or impaired in function, such as a slow I.Q., deterioration, illness or psychosis. What do laws/binding rules require when considering informed consent in these groups? See also I0002.


I0007 Assessment element card

Issue: Is there a possibility that the use of the technology produces additional information that is not directly related to the current care of the patient and may violate their right to respect for privacy?

Topic: LEG Privacy of the patient

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes44
Clarification
Common to all used applications

The protection of sensitive personal data is secured at the EU level. Privacy protection is a modern expression of the ancient ethical principle of confidentiality in doctor-patient relationship. The use of computerised patient record databases and modern genetic diagnostics entail certain challenges to this principle. For example, in Z vs. Finland (ECHR February 25, 1997) there was a case of an HIV infected person whose HIV positive test was an incidental finding, not related to her healthcare intervention at the time.


I0008 Assessment element card

Issue: What do laws/binding rules require with regard to informing relatives about the results?

Topic: LEG Privacy of the patient

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes45
Clarification
Common to all used applications

A test result may indicate that the relatives of a patient may have a medical condition that would need to be addressed. If this can be foreseen, appropriate procedures, complying with the existing legislation, must be considered beforehand – is the information to be revealed to, or withheld from the relatives in question? Describe on what conditions (if any) the privacy of the original patient can be broken in order to inform the relatives of their situation.

There may be situations, e.g. when treatment malpractice is suspected after the death of the patient, when (closest) relatives demand the results. Similar cases could occur in sudden, unexpected deaths and in some cases of highly infectious diseases.


I0009 Assessment element card

Issue: What do laws/binding rules require with regard to appropriate measures for securing patient data and how should this be addressed when implementing the technology?

Topic: LEG Privacy of the patient

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes46
Clarification
Common to all used applications

Provide an overview of the legal requirements regarding policies and procedures, as well as examples of: practical local solutions; securing the kind of patient data that will be generated when using of the technology.

Who is allowed to save and store the patient-data, where is it saved, for how long, and who can have access to it? Does the use of the technology produce some additional (i.e. diagnostically or therapeutically irrelevant) information on the patient that could be relevant for, e.g., health insurance, marketing studies, or safety authorities and how should data protection be handled in these cases? Is it possible that legal data protection requirements have adverse consequences to the quality of care, e.g. by complicating the transfer of patient data in a screening process, and how should this be addressed?


I0011 Assessment element card

Issue: What do laws/binding rules require with regard to appropriate processes or resources which would guarantee equal access to the technology?

Topic: LEG Equality in health care

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes47
Clarification
Common to all used applications

In general, equality in health care is stipulated in the EU Charter of Fundamental Rights and it is also one of the central principles of the Biomedicine Convention. Moreover, in many national constitutions, equality of citizens also covers access to health care. However, there may be experiences on a national level of some specific difficulties in equal access to the technology, and there may probably also be proposed solutions, which could be helpful for decision-makers in other countries as well.


I0012 Assessment element card

Issue: What are the consequences of various EU level and national regulations to the equal access to the technology?

Topic: LEG Equality in health care

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes48
Clarification
Common to all used applications

The possible consequences of the EU Directive of cross border health care could be considered here. There may be nationally legally defined processes, including reimbursement and pricing, determining the implementation of and level of access to a technology. This information may give useful insight also beyond one's own country.


I0015 Assessment element card

Issue: What authorisations and register listings does the technology have?

Topic: LEG Authorisation and safety

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes49
Clarification
Common to all used applications

Describe the register listings, both at EU level and national level, which might be relevant when implementing the technology and planning, e.g., local authorisation, monitoring or evaluation functions, as well as qualification and quality control. Examples include technology registers, registers for marketing authorisation, certification of safety and reimbursement. However, some of the registers, e.g. the one for medical devices (EUDAMED), are not open for HTA doers. Register listings information may be particularly relevant for the technologies which can be used off-label or as investigational intervention outside clinical trials (so-called expanded access or compassionate use).


I0017 Assessment element card

Issue: What do laws/binding rules require with regard to the safety of the technology and how should this be addressed when implementing the technology?

Topic: LEG Authorisation and safety

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes50
Clarification
Common to all used applications

List the legal requirements for safety of the technology and quality of care. Does the technology fulfil these requirements, and what should be done to ensure that the legal requirements maintain fulfilled when implementing the technology? Consider the findings of the SAF and ORG domains here, in the light of relevant European or national safety regulations. See also I0015.


I0019 Assessment element card

Issue: What should be known about the intellectual property rights and potential licensing fees?

Topic: LEG Ownership and liability

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes51
Clarification
Common to all used applications

This information is important because infringement of intellectual property rights can reduce the use of the technology and have implications for the wording of the acquisition contract of a new technology, and possibly also licencing fees.


I0021 Assessment element card

Issue: What should be known about the legal or binding rules regarding the width, depth and length of the manufacturers guarantee?

Topic: LEG Ownership and liability

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes52
Clarification
Common to all used applications

This issue may help the decision-maker to be aware of their legal rights when considering the manufacturer’s guarantee. The user guide plays a part in determining the manufacturer's liability.


I0023 Assessment element card

Issue: What kind of legal price control mechanisms are there that are relevant to the technology?

Topic: LEG Regulation of the market

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes53
Clarification
Common to all used applications

Describe the adopted economic measures for controlling public health expenditures when adopting technologies. This information, although not transferable, gives insight to decision-makers in other countries too.


I0024 Assessment element card

Issue: What kind of regulation exists for the acquisition and use of the technology?

Topic: LEG Regulation of the market

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes54
Clarification
Common to all used applications

Expensive technology and dangerous pharmaceuticals are typically subject to acquisition regulation.


I0025 Assessment element card

Issue: What legal restrictions are there for marketing the technology to the patients?

Topic: LEG Regulation of the market

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes55
Clarification
Common to all used applications

Describe general legal principles of the restrictions placed on the marketing of health technologies to lay people.


I0026 Assessment element card

Issue: What should be known about the legal issues in cases of new technologies where the current legislation is not directly applicable?

Topic: LEG Regulation of the market

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes56
Clarification
Common to all used applications

Novel technologies may not always be unambiguously covered by existing legislation. Sometimes, an otherwise restricted technology can be used in clinical trials or as ‘compassionate use’, i.e. in extended use outside clinical trials. Important questions, such as 'How are the liability issues solved according to existing legislation?', or, 'Is the voluntary participation of patients guaranteed properly?' may be important to consider. If the current law does not provide a straightforward answer to the liability issues it may be advisable to consult a legal expert on the interpretation of the existing provisions with regard to the technology in question. Sometimes even new legislative measures are needed.


I0037 Assessment element card

Issue: Are there relevant concerns about conflicts of interest regarding the preparation of binding rules and their implementation?

Topic: LEG Regulation of the market

Application-specific propertiesApplicationUsedOrder
Rapid Relative Effectiveness Assessments (4.2)Yes57
Clarification
Common to all used applications

Relevant concerns of partiality or conflicts of interest with regard to binding guidance may give useful insight to decision-makers about the importance of implementing a technology.


Appendices

Appendix 1. Shared methodologies

Rapid apx1

Appendix 2. Templates

Rapid apx2