Scolaris Content Display Scolaris Content Display

Diabetes self‐management education and support delivered by mobile health (m‐health) interventions for adults with type 2 diabetes mellitus

Esta versión no es la más reciente

Contraer todo Desplegar todo

Abstract

This is a protocol for a Cochrane Review (Intervention). The objectives are as follows:

To assess the effects of diabetes self‐management education and support delivered by mobile health interventions in adults with type 2 diabetes mellitus.

Background

One of the most important goals in the treatment of type 2 diabetes mellitus (T2DM) is to prevent complications at both the macrovascular and microvascular level. To do so, diabetes treatment aims to control glycaemia, blood pressure, and cholesterol levels, and if necessary to reduce body weight and achieve smoking cessation. More than 20% of all individuals with T2DM in primary care need insulin treatment (Sharma 2016), and this group needs to be especially capable of adequate self‐management, which means they should make healthy food choices, exercise frequently, monitor their blood glucose regularly, administer insulin properly, and adjust both dietary intake and insulin dose in relation to physical activity. However, optimal self‐management of T2DM requires sufficient knowledge on relevant topics and often behavioural change. Diabetes self‐management education (DSME) can provide people with this knowledge, as well as the abilities and skills necessary to apply it (Funnel 2010), but to implement and sustain the targeted behaviour, they should also receive diabetes self‐management support (DSMS), defined as "activities that assist the person with diabetes in implementing and sustaining the behaviours needed to manage his or her condition on an ongoing basis" (Powers 2015). DSME and DSMS together are referred to as diabetes self‐management education and support (DSME/S), which forms the essential basis for self‐management in individuals with T2DM (Powers 2015).

Traditionally, healthcare providers have delivered DSME/S via face‐to‐face contact. However, because the number of diabetes monitoring visits is limited, healthcare providers are only able to provide their patients with DSME/S a few times a year, and the amount of information they give may be overwhelming for their patients. Additionally, in some countries the number of healthcare providers cannot keep up with the increasing number of individuals with T2DM. These shortcomings have prompted the need for innovative and (cost‐)effective solutions to deliver DSME/S.

New technologies have the potential to deliver DSME/S and consequently improve diabetes self‐management. A potential low‐cost and easily accessible way to deliver DSME/S may be by using mobile health (m‐health): health care supported by mobile devices. A Cochrane Review on computer‐based diabetes self‐management interventions, published in 2013, found that these interventions had small benefits on glycaemic control (low‐ to moderate‐quality evidence); the effect size was larger in the mobile phone group (Pal 2011). Since then, m‐health interventions have continued to gain popularity, but their effects remain uncertain. In 2013 over 40,000 application software (apps) regarding health and fitness were available (Aitken 2013); the number of diabetes apps in the iTunes store increased from 60 in July 2009 to 260 in February 2011 (Chomutare 2011). Although randomised controlled trials (RCTs) have evaluated a few of these interventions (Hunt 2015), most apps are not supported by evidence, making it hard to choose one that is suitable. Evidence on the effectiveness of m‐health interventions may help people with T2DM and their healthcare providers to make better decisions regarding their use.

Description of the condition

T2DM is a chronic metabolic condition with a multifactorial aetiology. As a result of a decreased insulin secretion and increased insulin resistance, plasma glucose levels rise, resulting in chronic hyperglycaemia. Eventually this may lead to complications at both the microvascular (retinopathy, nephropathy, neuropathy) and macrovascular level (i.e. cardiovascular diseases).

Description of the intervention

In this review we will focus on both DSME and DSMS delivered via m‐health interventions. The ubiquitous spread of mobile phones and technology, combined with limited human and financial resources forhealth care, have made m‐health increasingly popular among different fields of medicine worldwide (WHO 2011). In T2DM, m‐health offers a wide variety of features, which we will roughly divide into: self‐monitoring of blood glucose, blood pressure, weight, carbohydrate intake, physical activity, insulin dose and medication intake; diabetes self‐management education; diabetes self‐management support; and novel communication methods (Chomutare 2011). Self‐monitoring features often enable synchronisation with a personal health record, accessible to both healthcare providers and patients, and they also commonly include automatic feedback functions.

As mentioned earlier, in this review we will focus on DSME/S delivered via m‐health interventions. Eligible interventions are DSME alone, DSMS alone and combination DSME/S (DSME followed by DSMS).

Adverse effects of the intervention

The Cochrane Review on computer‐based diabetes self‐management interventions found only two possible adverse events in 16 included trials (Pal 2011): in one study a participant dropped out due to study‐related anxiety (Wise 1986), in another study there was an increase in hypoglycaemic events in the intervention group, although it was not statistically significant (Lim 2011). Only one study explicitly stated that there were no adverse events (Quinn 2011). These findings suggest that either adverse events almost never happen, or they are hardly reported.

Based on our experience and the findings of the earlier Cochrane Review (Pal 2011), we hypothesise that the following adverse events will be the most common in m‐health interventions that provide DSME/S.

  • Hypoglycaemic events, especially in those relating to insulin use (due to increased physical activity, decreased carbohydrate intake, incorrect advice or misinterpretation of advice).

  • Weight gain (due to higher insulin dosages, incorrect advice or misinterpretation of advice).

  • Frustration due to technical problems and feelings of failure due to computer illiteracy.

How the intervention might work

DSME delivered via m‐health interventions may improve people's knowledge and understanding of diabetes and increase their self‐management abilities and skills (Pal 2011). Eventually, this may lead to increased physical activity, a healthier diet, improved therapy adherence and even the ability to self‐titrate insulin.

Besides, m‐health interventions may also have the potential to deliver DSMS because the intervention may function as a behavioural trigger, also known as prompt, stimulus or cue to action (Fogg 2009). Triggers stimulate people to engage in healthy behaviour and are contemplated in the Health Belief Model (Janz 1984), Leventhal's self‐regulation model (Leventhal 1998), the Transtheoretical Model of Behaviour Change (Prochaska 2002), and the Fogg Behavior Model (Fogg 2009). Via this mechanism of ongoing stimuli, the intervention delivered via m‐health may reinforce people's awareness of the importance of self‐management and their sense of control over their disease (Hunt 2015). Since DSMS can also help people implement the targeted behaviour, it can also increase their abilities and skills. Moreover, DSME can also function as a behavioural trigger. See Figure 1 for a conceptual framework of how the intervention might work.


Conceptual framework of how the intervention might workFPG: fasting plasma glucose; HbA1c: glycosylated haemoglobin.

Conceptual framework of how the intervention might work

FPG: fasting plasma glucose; HbA1c: glycosylated haemoglobin.

Why it is important to do this review

Previous systematic reviews (to June 2016) showed a beneficial effect of m‐health on glycaemic control in individuals with T2DM (Cui 2016; Pal 2011). However, the rapid and continuous development of the m‐health field necessitates a new up‐to‐date systematic review and meta‐analysis. The Cochrane Review on computer‐based diabetes self‐management interventions reviewed a broader topic than m‐health, namely electronic health or e‐health (which includes m‐health), and in 2011, review authors identified only four studies with an m‐health intervention (Pal 2011). Moreover, previous systematic reviews did not report on important topics such as cost‐effectiveness, adverse events or subgroup effects (if certain subgroups benefit more that others).

Objectives

To assess the effects of diabetes self‐management education and support delivered by mobile health interventions in adults with type 2 diabetes mellitus.

Methods

Criteria for considering studies for this review

Types of studies

We will include RCTs regardless of their publication status or language of publication.

Types of participants

Adults (aged 18 or older) with T2DM.

Diagnostic criteria for type 2 diabetes mellitus

In order to be consistent with changes in the classification of and diagnostic criteria for diabetes mellitus over the years, the diagnosis should be established using the standard criteria valid at the time of the trial commencing (for example, ADA 2003; ADA 2008; WHO 1998). Ideally, trial authors should have described the diagnostic criteria. We will use the trial authors' definition of diabetes mellitus if necessary.

Because changes in diagnostic criteria did not appear after 2003, and the vast majority of RCTs on m‐health have taken place after 2003, we do not expect that diagnostic criteria will produce significant variability in the clinical characteristics of the participants included as well as in the results obtained. Trials involving participants with comorbid disorders will be eligible for inclusion as long as the primary focus of the intervention is DSME/S. Trials involving a broader population (e.g. in individuals with a chronic illness) can only be included when they present results for T2DM patients separately. When these data are not available we will request separate data from the authors.

Types of interventions

Definition of m‐health interventions

Mobile health (m‐health) interventions that provide diabetes self‐management education (DSME) and/or diabetes self‐management support (DSMS). An m‐health intervention is eligible when DSME/S is provided either via short message service (SMS), text messages, voice messages or via a smartphone application, and studied as the main intervention. We include SMS because from a conceptual point of view, we think that messages delivered by SMS and by a smartphone app will have the same effect, differing only in technical aspects. So although the SMS technique may soon become obsolete, the results of studies on DSME/S delivered by SMS still contribute to the body of evidence on m‐health. All mobile devices are eligible vehicles for the intervention: mobile phones, smartphones, tablets and other mobile devices. Wearables will be only included when DSME/S is delivered directly to the wearable, or when DSME/S is delivered to a mobile phone, smartphone or other mobile device that is connected to the wearable.

Definition of diabetes self‐management education

Diabetes self‐management is "the ongoing process of facilitating the knowledge, skill, and ability necessary for diabetes self‐care" (Powers 2015). The ultimate goal of DSME is to improve clinical outcomes, health status and quality of life by means of activating patients towards informed decision making, self‐care behaviours, problem solving, and active collaboration with the healthcare team (Powers 2015).

Definition of diabetes self‐management support

Diabetes self‐management supports are "activities that assist the person with diabetes in implementing and sustaining the behaviours needed to manage his or her condition on an ongoing basis" (Powers 2015).This can be behavioural, educational, psychosocial or clinical support.

Definition of usual care

Usual care is defined as standard care that individuals with T2DM should receive according to national guidelines.

We plan to investigate the following comparisons of intervention versus control/comparator.

Intervention

  • M‐health intervention that provides DSME

  • M‐health intervention that provides DSMS

  • M‐health intervention that provides DSME/S

Comparator

  • Attention placebo control (APC) compared with any eligible intervention. Since the m‐health intervention may have a psychosocial component, the classical design of an active treatment versus a control group might not be appropriate. To test the effect of the psychosocial intervention, an APC can be included. This APC group does not receive the actual psychosocial intervention but receives an intervention that covers the same amount of time and attention as the experimental group receives (Popp 2015).

  • Usual care (no intervention) compared with any intervention.

Note: we will perform a subgroup analysis to investigate any differences in effect estimates between studies with an APC group and studies with a usual care control group (see the Subgroup analysis and investigation of heterogeneity section).

Concomitant interventions will have to be the same in both the intervention and comparator groups to establish fair comparisons.

Minimum duration of intervention

The clinically meaningful minimal duration of the intervention will be two months.

Minimum duration of follow‐up

Minimal duration of follow‐up will be two months after start of the intervention, that is, we will include effects measured immediately after the end of the intervention.

We will define extended follow‐up periods (also called open‐label extension studies) for outcomes measured once the original trial, as specified in the trial protocol, has been terminated. However, such studies are frequently of an observational nature, and we will primarily evaluate them for adverse events (Buch 2011; Megan 2012).

Summary of specific exclusion criteria

  • Trials in individuals aged less than 18 years.

  • Trials including pregnant women.

  • Trials investigating a mixture of type 1 and type 2 diabetes participants, when the results for type 1 and type 2 diabetes participants are not presented separately (when these data are not available we will request separate data from the authors) or when fewer than 75% of the participants have T2DM.

  • Trials investigating personal communication by mobile phones only, such as telephone calls with healthcare professionals.

  • Trials investigating non‐automated interventions, such as tailored feedback on glucose values from healthcare professionals instead of an automated algorithm.

  • Trials investigating personal records, data entries or diaries.

  • Trials investigating m‐health interventions targeted at healthcare workers.

Types of outcome measures

We will not exclude trials that do not report on one or several of our primary or secondary outcome measures. We will exclude trials failing to report on any our primary or secondary outcomes, but we will provide some basic information in an additional table.

We will consider most outcomes measured at the end of the intervention or within 30 days after the end of the intervention (short‐term follow‐up), between one month and six months after the end of the intervention (medium‐term follow‐up), or more than six months after the end of the intervention (long‐term follow‐up). However, we will consider adverse events and all‐cause mortality at any time after randomisation.

Primary outcomes

  • Glycosylated haemoglobin A1c (HbA1c).

  • Change in body weight.

  • Hypoglycaemic episodes.

Secondary outcomes

  • Adverse events other than hypoglycaemic episodes.

  • All‐cause mortality.

  • Health‐related quality of life.

  • Diabetes treatment satisfaction.

  • Self‐care behaviours.

  • Blood pressure.

  • Lipids.

  • Fasting plasma glucose (FPG).

  • Healthcare related costs.

Method of outcome measurement

  • HbA1c: measured in % (mmol/mol).

  • Change in body weight: measured in kilograms (kg).

  • Hypoglycaemic episodes: classified as mild (self‐managed), moderate (daily activities interrupted but self‐management) and severe (requiring assistance from others).

  • Adverse events other than hypoglycaemic episodes: such as anxiety and depression.

  • All‐cause mortality: defined as death from any cause.

  • Health‐related quality of life: evaluated by a validated instrument such as the diabetes‐dependent quality of life (ADDQoL) questionnaire.

  • Treatment satisfaction with diabetes care: evaluated by a validated instrument such as the diabetes treatment satisfaction questionnaire (DTSQ). When satisfaction with the intervention is specifically measured, data will be extracted for this outcome too.

  • Self‐care behaviours: evaluated with a validated instrument such as Summary of Diabetes Self‐Care Activities (SDSCA).

  • Blood pressure: systolic and diastolic blood pressure in mmHg.

  • Lipids: serum cholesterol (total cholesterol, HDL‐cholesterol and LDL‐cholesterol).

  • FPG: measured in mg/dL.

  • Healthcare related costs: such as direct costs defined as admission/readmission rates, average length of stay, visits to general practitioner, accident/emergency visits; medication consumption; indirect costs defined as resources lost due to illness by the participant or their family member.

Specification of key prognostic variables

  • Age

  • Sex

  • Glycaemic control

  • Diabetes duration

Search methods for identification of studies

Electronic searches

We will search the following sources from the year 2000 to the specified date and will place no restrictions on the language of publication. In 2000, approximately one in three inhabitants in the United States had a mobile phone subscription (Little 2012).

  • Cochrane Central Register of Controlled Trials (CENTRAL) via the Cochrane Register of Studies Online (CRSO).

  • MEDLINE Ovid (Epub Ahead of Print, In‐Process & Other Non‐Indexed Citations, Ovid MEDLINE(R) Daily and Ovid MEDLINE(R)).

  • Embase Ovid.

  • PsycINFO Ovid.

  • ClinicalTrials.gov (www.clinicaltrials.gov).

  • World Health Organization International Clinical Trials Registry Platform (ICTRP) (www.who.int/trialsearch/).

We will continuously apply a MEDLINE (via Ovid SP) email alert service to identify newly published trials using the same search strategy as described for MEDLINE (for details on search strategies, see Appendix 1). Should we identify new trials for inclusion, we will evaluate these, incorporate the findings into our review and resubmit another review draft (Beller 2013).

Searching other resources

We will try to identify other potentially eligible trials or ancillary publications by searching the reference lists of included trials, systematic reviews, meta‐analyses and health technology assessment reports. In addition, we will contact authors of included trials to identify any additional information on the retrieved trials and further trials that we may have missed.

We will not use abstracts or conference proceedings for data extraction, because this information source does not fulfil the Consolidated Standards of Reporting Trials (CONSORT) requirements, which is "an evidence‐based, minimum set of recommendations for reporting randomized trials" (CONSORT 2010; Scherer 2007), unless we can obtain full data from trial authors. We will list key data of abstracts in an appendix. We will show key information on abstracts or conference proceedings in the 'Characteristics of studies awaiting classification' table.

Data collection and analysis

Selection of studies

Two review authors (AMB, RV) will independently screen the abstract, title or both, of every record we retrieve in the literature searches, to determine which trials we should assess further. We will obtain the full‐text of all potentially relevant records. We will resolve any disagreements through consensus or by recourse to a third review author (GR). If we cannot resolve a disagreement, we will categorise the trial as a 'study awaiting classification' and will contact the trial authors for clarification. We will present an adapted PRISMA flow diagram to show the process of trial selection (Liberati 2009). We will list all articles excluded after full‐text assessment in a 'Characteristics of excluded studies' table and will provide the reasons for exclusion.

Data extraction and management

For trials that fulfil our inclusion criteria, two review authors (AMB, RV) will independently extract key participant and intervention characteristics. We will describe interventions using the 'template for intervention description and replication' (TIDieR) checklist (Hoffmann 2014; Hoffmann 2017). We will report data on efficacy outcomes and adverse events using standardised data extraction sheets from the Cochrane Metabolic and Endocrine Disorders Group. We will resolve any disagreements by discussion or, if required, we will consult a third review author (GR).

We will provide information about potentially relevant ongoing trials, including the trial identifiers, in the 'Characteristics of ongoing trials' table and in a joint appendix 'Matrix of trial endpoint (publications and trial documents)'. We will try to find the protocol for each included trial and will compare primary, secondary and other outcomes described there and in the full‐text study reports in a joint appendix.

We will email all authors of included trials to enquire whether they would be willing to answer questions regarding their trials. We will present the results of this survey in an appendix. We will thereafter seek relevant missing information on the trial from the primary trial author(s), if required.

Dealing with duplicate and companion publications

In the event of duplicate publications, companion documents or multiple reports of a primary trial, we will maximise the information yield by collating all available data, and we will use the most complete data set aggregated across all known publications. We will list duplicate publications, companion documents, multiple reports of a primary trial and trial documents of included trials (such as trial registry information) as secondary references under the study identifier (ID) of the included trial. Furthermore, we will also list duplicate publications, companion documents, multiple reports of a trial and trial documents of excluded trials (such as trial registry information) as secondary references under the study ID of the excluded trial.

Data from clinical trial registers

If data from included trials are available as study results in clinical trial registers such as ClinicalTrials.gov or similar sources, we will make full use of this information and extract the data. If there is also a full publication of the trial, we will collate and critically appraise all available data. If an included trial is marked as a completed study in a clinical trial register but no additional information (study results, publication or both) is available, we will add this trial to the table 'Characteristics of studies awaiting classification'.

Assessment of risk of bias in included studies

Two review authors (AMB, RV) will independently assess the risk of bias for each included trial. We will resolve any disagreements by consensus or by consulting a third review author (GR). In case of disagreement, we will consult the rest of the review author team and make a judgement based on consensus. If adequate information is unavailable from the trials or trial protocols, we will contact the trial authors to recover missing data on 'Risk of bias' items.

We will use the Cochrane 'Risk of bias' assessment tool and assign judgments of low, high or unclear risk of bias (Higgins 2011a; Higgins 2011b). We will evaluate individual bias items as described in the Cochrane Handbook for Systematic Reviews of Interventions according to the criteria and associated categorisations contained therein(Higgins 2011b).

Random sequence generation (selection bias due to inadequate generation of a randomised sequence)

For each included trial we will describe the method used to generate the allocation sequence in sufficient detail to enable assessment of whether it should produce comparable groups.

  • Low risk of bias: the trial authors achieved sequence generation using computer‐generated random numbers or a random numbers table. Drawing of lots, tossing a coin, shuffling cards or envelopes, and throwing dice are adequate if an independent person performed this who was not otherwise involved in the trial. We will consider the use of the minimisation technique as equivalent to being random.

  • Unclear risk of bias: insufficient information about the sequence generation process.

  • High risk of bias: the sequence generation method was non‐random or quasi‐random (e.g. sequence generated by odd or even date of birth; sequence generated by some rule based on date (or day) of admission; sequence generated by some rule based on hospital or clinic record number; allocation by judgement of the clinician; allocation by preference of the participant; allocation based on the results of a laboratory test or a series of tests; or allocation by availability of the intervention).

Allocation concealment (selection bias due to inadequate concealment of allocation prior to assignment)

We will describe for each included trial the method used to conceal allocation to interventions prior to assignment, and we will assess whether intervention allocation could have been foreseen in advance of or during recruitment, or changed after assignment.

  • Low risk of bias: central allocation (including telephone, interactive voice‐recorder, Internet‐based and pharmacy‐controlled randomisation); sequentially numbered drug containers of identical appearance; sequentially numbered, opaque, sealed envelopes.

  • Unclear risk of bias: insufficient information about the allocation concealment.

  • High risk of bias: using an open random allocation schedule (e.g. a list of random numbers); assignment envelopes were used without appropriate safeguards; alternation or rotation; date of birth; case record number; any other explicitly unconcealed procedure.

We will also evaluate trial baseline data to incorporate assessment of baseline imbalance into the 'Risk of bias' judgement for selection bias (Corbett 2014). Chance imbalances may also affect judgements on the risk of attrition bias. In the case of unadjusted analyses, we will distinguish between trials we rate as being at low risk of bias on the basis of both randomisation methods and baseline similarity, and trials we judge as being at low risk of bias on the basis of baseline similarity alone (Corbett 2014). We will re‐classify judgements of unclear, low or high risk of selection bias as specified in Appendix 2.

Blinding of participants and study personnel (performance bias due to knowledge of the allocated interventions by participants and personnel during the trial)

We will evaluate the risk of detection bias separately for each outcome (Hróbjartsson 2013). We will note whether endpoints were self‐reported, investigator‐assessed or adjudicated outcome measures (see below).

  • Low risk of bias: blinding of participants and key study personnel is ensured, and it is unlikely that the blinding could have been broken; no blinding or incomplete blinding, but we judge that the outcome is unlikely to have been influenced by lack of blinding.

  • Unclear risk of bias: insufficient information about the blinding of participants and study personnel; the trial does not address this outcome.

  • High risk of bias: no blinding or incomplete blinding, and the outcome is likely to have been influenced by lack of blinding; blinding of trial participants and key personnel attempted, but likely that the blinding could have been broken, and the outcome is likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias due to knowledge of the allocated interventions by outcome assessment)

We will evaluate the risk of detection bias separately for each outcome (Hróbjartsson 2013). We will note whether endpoints were self‐reported, investigator‐assessed or adjudicated outcome measures (see below).

  • Low risk of bias: blinding of outcome assessment is ensured, and it is unlikely that the blinding could have been broken; no blinding of outcome assessment, but we judge that the outcome measurement is unlikely to have been influenced by lack of blinding.

  • Unclear risk of bias: insufficient information about the blinding of outcome assessors; the trial did not address this outcome.

  • High risk of bias: no blinding of outcome assessment, and the outcome measurement is likely to have been influenced by lack of blinding; blinding of outcome assessment, but likely that the blinding could have been broken, and the outcome measurement is likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias due to amount, nature or handling of incomplete outcome data)

For each included trial and or each outcome, we will describe the completeness of data, including attrition and exclusions from the analyses. We will state whether the trial reported attrition and exclusions, and the number of participants included in the analysis at each stage (compared with the number of randomised participants per intervention/comparator groups). We will also note if the trial reported the reasons for attrition or exclusion and whether missing data were balanced across groups or were related to outcomes. We will consider the implications of missing outcome data per outcome such as high dropout rates (e.g. above 15%) or disparate attrition rates (e.g. difference of 10% or more between trial arms).

  • Low risk of bias: no missing outcome data; reasons for missing outcome data unlikely to be related to true outcome (for survival data, censoring unlikely to introduce bias); missing outcome data balanced in numbers across intervention groups, with similar reasons for missing data across groups; for dichotomous outcome data, the proportion of missing outcomes compared with observed event risk is not enough to have a clinically relevant impact on the intervention effect estimate; for continuous outcome data, plausible effect size (mean difference or standardised mean difference) among missing outcomes is not enough to have a clinically relevant impact on observed effect size; trial authors used appropriate methods, such as multiple imputation, to handle missing data.

  • Unclear risk of bias: insufficient information to assess whether missing data in combination with the method used to handle missing data were likely to induce bias; the trial did not address this outcome.

  • High risk of bias: reason for missing outcome data is likely to be related to true outcome, with either imbalance in numbers or reasons for missing data across intervention groups; for dichotomous outcome data, the proportion of missing outcomes compared with observed event risk enough to induce clinically relevant bias in the intervention effect estimate; for continuous outcome data, plausible effect size (mean difference or standardised mean difference) among missing outcomes enough to induce clinically relevant bias in observed effect size; 'as‐treated' or similar analysis done with substantial departure of the intervention received from that assigned at randomisation; potentially inappropriate application of simple imputation.

Selective reporting (reporting bias due to selective outcome reporting)

We will assess outcome reporting bias by integrating the results of the appendix 'Matrix of trial endpoints (publications and trial documents)' with those of the appendix 'High risk of outcome reporting bias according to ORBIT classification' (Boutron 2014; Jones 2015; Kirkham 2010; Mathieu 2009). This analysis will form the basis for the judgement of selective reporting.

  • Low risk of bias: the trial protocol is available, and authors have reported all of the trial's pre‐specified (primary and secondary) outcomes that are of interest in the review in the pre‐specified way; the study protocol is unavailable, but it is clear that the published reports include all expected outcomes (ORBIT classification).

  • Unclear risk of bias: insufficient information about selective reporting.

  • High risk of bias: not all of the trial's pre‐specified primary outcomes are reported; one or more primary outcomes are reported using measurements, analysis methods or subsets of the data (e.g. subscales) that were not pre‐specified; one or more reported primary outcomes were not pre‐specified (unless clear justification for their reporting is provided, such as an unexpected adverse effect); one or more outcomes of interest to the Cochrane Review are reported incompletely so that we cannot enter them in a meta‐analysis; the trial report fails to include results for a key outcome that we would expect to have been reported for such a trial (ORBIT classification).

Other bias

  • Low risk of bias: the trial appears to be free from other sources of bias.

  • Unclear risk of bias: there is insufficient information to assess whether an important risk of bias exists or insufficient rationale or evidence that an identified problem introduced bias.

  • High risk of bias: the trial has a potential source of bias related to the specific trial design used; the trial has been claimed to have been fraudulent; or the trial had some other serious problem.

Summary assessment of risk of bias

We will present a 'Risk of bias' graph and a 'Risk of bias' summary figure.

We will distinguish between self‐reported, investigator‐assessed and adjudicated outcome measures.

We will accept the following outcomes that were self‐reported by participants.

  • Hypoglycaemic episodes.

  • Adverse events other than hypoglycaemic episodes.

  • Health‐related quality of life.

  • Diabetes treatment satisfaction (including satisfaction with the intervention).

  • Self‐care behaviours.

  • Changes in weight.

  • Blood pressure.

  • Fasting plasma glucose.

Investigator‐assessed outcomes will include the following.

  • HbA1c.

  • Changes in weight.

  • Hypoglycaemic episodes.

  • Adverse events other than hypoglycaemic episodes.

  • All‐cause mortality.

  • Blood pressure.

  • Lipids.

  • Fasting plasma glucose.

  • Health‐related costs.

Risk of bias for a trial across outcomes

Some 'Risk of bias' domains, such as selection bias (sequence generation and allocation sequence concealment), affect the risk of bias across all outcome measures in a trial. In case of high risk of selection bias, we will mark all endpoints investigated in the associated trial as being at high risk. Otherwise, we will not perform a summary assessment of the risk of bias across all outcomes for a trial.

Risk of bias for an outcome within a trial and across domains

We will assess the risk of bias for an outcome measure by including all entries relevant to that outcome (i.e. both trial‐level entries and outcome‐specific entries). We consider low risk of bias to denote a low risk of bias for all key domains, unclear risk to denote an unclear risk of bias for one or more key domains and high risk to denote a high risk of bias for one or more key domains.

Risk of bias for an outcome across trials and across domains

These are the main summary assessments that we will incorporate into our judgements about the quality of evidence in the 'Summary of findings' tables. We will define outcomes as being at low risk of bias when most information comes from trials at low risk of bias, unclear risk when most information comes from trials at low or unclear risk of bias, and high risk when a sufficient proportion of information comes from trials at high risk of bias.

Measures of treatment effect

When at least two included trials are available for a comparison and a given outcome, we will try to express dichotomous data as a risk ratio (RR) or odds ratio (OR) with 95% confidence interval (CI). For continuous outcomes measured on the same scale (e.g. weight loss in kg) we will estimate the intervention effect using the mean difference with 95% CI. For continuous outcomes that measure the same underlying concept (e.g. health‐related quality of life) but use different measurement scales, we will calculate the standardised mean difference (SMD). We will express time‐to‐event data as a hazard ratio (HR) with 95% CI.

Unit of analysis issues

We will take into account the level at which randomisation occurred, such as cross‐over trials, cluster‐randomised trials and multiple observations for the same outcome. If more than one comparison from the same trial is eligible for inclusion in the same meta‐analysis, we will either combine groups to create a single pair‐wise comparison or appropriately reduce the sample size so that the same participants do not contribute more than once (splitting the 'shared' group into two or more groups). While the latter approach offers some solution to adjusting the precision of the comparison, it does not account for correlation arising from the same set of participants being in multiple comparisons (Higgins 2011c).

We will attempt to reanalyse cluster‐RCTs that have not appropriately adjusted for potential clustering of participants within clusters in their analyses. The variance of the intervention effects will be inflated by a design effect. Calculation of a design effect involves estimation of an intra‐cluster correlation (ICC). We will obtain estimates of ICCs through contact with the trial authors, impute them using estimates from other included trials that report ICCs, or use external estimates from empirical research (e.g. Bell 2013). We plan to examine the impact of clustering using sensitivity analyses.

Dealing with missing data

If possible, we will obtain missing data from the authors of the included trials. We will carefully evaluate important numerical data such as screened, randomly assigned participants as well as intention‐to‐treat, and as‐treated and per‐protocol populations. We will investigate attrition rates (e.g. dropouts, losses to follow‐up, withdrawals), and we will critically appraise issues concerning missing data and use of imputation methods (e.g. last observation carried forward).

In trials where the standard deviation (SD) of the outcome is not available at follow‐up or cannot be recreated, we will standardise by the average of the pooled baseline SD from those trials that reported this information.

Where included trials do not report means and SDs for outcomes and we do not receive the necessary information from trial authors, we will impute these values by estimating the mean and variance from the median, range, and the size of the sample (Hozo 2005).

We will investigate the impact of imputation on meta‐analyses by performing sensitivity analyses, and we will report which trials were included with imputed SDs for each outcome.

Assessment of heterogeneity

In the event of substantial clinical or methodological heterogeneity, we will not report trial results as the pooled effect estimate in a meta‐analysis.

We will identify heterogeneity (inconsistency) by visually inspecting the forest plots and by using a standard Chi² test with a significance level of α = 0.1. In view of the low power of this test, we will also consider the I² statistic, which quantifies inconsistency across trials to assess the impact of heterogeneity on the meta‐analysis (Higgins 2002; Higgins 2003).

When we find heterogeneity, we will attempt to determine the possible reasons for it by examining individual trial and subgroup characteristics.

Assessment of reporting biases

If we include 10 or more trials that investigate a particular outcome, we will use funnel plots to assess small‐trial effects. Several explanations may account for funnel plot asymmetry, including true heterogeneity of effect with respect to trial size, poor methodological design (and hence bias of small trials) and publication bias. Therefore we will interpret the results carefully (Sterne 2011).

Data synthesis

We plan to undertake (or display) a meta‐analysis only if we judge participants, interventions, comparisons and outcomes to be sufficiently similar to ensure an answer that is clinically meaningful. Unless good evidence shows homogeneous effects across trials, we will primarily summarise low risk of bias data using a random‐effects model (Wood 2008). We will interpret random‐effects meta‐analyses with due consideration to the whole distribution of effects and present a prediction interval (Borenstein 2017a; Borenstein 2017b). A prediction interval needs at least three trials to be calculated and specifies a predicted range for the true treatment effect in an individual trial (Riley 2011). For rare events, occurring at rates below 1%, we will use Peto's odds ratio method, provided that there is no substantial imbalance between intervention and comparator group sizes and intervention effects are not exceptionally large. In addition, we will also perform statistical analyses according to the guidelines presented in the Cochrane Handbook for Systematic Reviews of Interventions (Deeks 2011).

Subgroup analysis and investigation of heterogeneity

We expect the following characteristics to introduce clinical heterogeneity, and we plan to carry out subgroup analyses including investigation of interactions (Altman 2003).

  • Sex: we expect men and women to respond differently to DSME delivered via m‐health.

  • Age: because age may be highly correlated to computer literacy, we expect age to be a potential effect modifier. We will use 60 years as the cut‐off age, analysing those under 60 and those aged 60 or older separately.

  • Ethnicities: minorities within a country may have language issues that result in decreased comprehension.

  • Socioeconomic status: people with a low socioeconomic status may have less prior knowledge on DSME.

  • Treatment groups (insulin versus oral glucose‐lowering therapy and behaviour changing advice only): in people who use insulin, baseline HbA1c is likely to be higher, and the risk of weight gain and hypoglycaemia is higher. Besides, their prior DSME knowledge will differ from the DSME knowledge in people not on insulin therapy.

  • Levels of metabolic control: in study populations with higher average baseline HbA1c level (poor metabolic control), the effect size of the intervention might be larger. As a cut‐off point for poor metabolic control we will use: HbA1c of more than 64 mmol/mol (> 8%) versus HbA1c of 64 mmol/mol or less (≤ 8%).

  • Type of control group: studies with an APC group versus studies with a usual care control group (see 'Comparator' within the 'Types of interventions' section).

Sensitivity analysis

We plan to perform sensitivity analyses to explore the influence of the following factors (when applicable) on effect sizes by restricting analysis to the following.

  • Published trials.

  • The effect of risk of bias, as specified in the Assessment of risk of bias in included studies section.

  • Very long (≥ 12 months) or large trials (≥ 200 participants) to establish the extent to which they dominate the results.

  • Using the following filters: imputation, language of publication, source of funding (industry versus other), or country.

We will also test the robustness of results by repeating the analyses using different measures of effect size (RR, OR, etc.) and different statistical models (fixed‐effect and random‐effects models).

Summary of findings table

We will present a summary of the evidence in a 'Summary of findings' table. This will provide key information about the best estimate of the magnitude of the effect, in relative terms and as absolute differences, for each relevant comparison of alternative management strategies, numbers of participants and trials that address each important outcome and a rating of overall confidence in effect estimates for each outcome. We will create the 'Summary of findings' table based on the methods described in the Cochrane Handbook for Systematic Reviews of Interventions (Schünemann 2011), using Review Manager 5 (RevMan 2014). We will present a 'Summary of finding' table to report the following outcomes, listed according to priority.

  1. Health‐related quality of life.

  2. Hypoglycaemic episodes.

  3. Diabetes treatment satisfaction.

  4. Self‐care behaviour.

  5. HbA1c.

  6. Body weight.

  7. Healthcare related costs.

We will present the overall quality of the evidence for each outcome according to the GRADE approach, which takes into account issues related to internal validity (risk of bias, inconsistency, imprecision, publication bias) and also to external validity, such as directness of results. Two review authors (AMB, RV) will independently rate the quality of the evidence for each outcome.

We will include an appendix titled 'Checklist to aid consistency and reproducibility of GRADE assessments' (Meader 2014), to help with standardisation of the 'Summary of findings' tables. Alternatively, we will use the GRADEpro Guideline Development Tool (GDT) software (GRADEpro GDT 2015), presenting evidence profile tables as an appendix. We will present results for the outcomes as described in the Types of outcome measures section. If meta‐analysis is not possible, we will present the results narratively in the 'Summary of findings' table. We will justify all decisions to downgrade the quality of trials using footnotes, and we will make comments to aid the reader's understanding of the Cochrane Review where necessary.

Conceptual framework of how the intervention might workFPG: fasting plasma glucose; HbA1c: glycosylated haemoglobin.
Figuras y tablas -
Figure 1

Conceptual framework of how the intervention might work

FPG: fasting plasma glucose; HbA1c: glycosylated haemoglobin.