An adaptable implementation package targeting evidence-based indicators in primary care: A pragmatic cluster-randomised evaluation. - Abstract - Europe PMC Europe PMC Europe PMC

Abstract 


BACKGROUND:In primary care, multiple priorities and system pressures make closing the gap between evidence and practice challenging. Most implementation studies focus on single conditions, limiting generalisability. We compared an adaptable implementation package against an implementation control and assessed effects on adherence to four different evidence-based quality indicators. METHODS AND FINDINGS:We undertook two parallel, pragmatic cluster-randomised trials using balanced incomplete block designs in general practices in West Yorkshire, England. We used 'opt-out' recruitment, and we randomly assigned practices that did not opt out to an implementation package targeting either diabetes control or risky prescribing (Trial 1); or blood pressure (BP) control or anticoagulation in atrial fibrillation (AF) (Trial 2). Within trials, each arm acted as the implementation control comparison for the other targeted indicator. For example, practices assigned to the diabetes control package acted as the comparison for practices assigned to the risky prescribing package. The implementation package embedded behaviour change techniques within audit and feedback, educational outreach, and computerised support, with content tailored to each indicator. Respective patient-level primary endpoints at 11 months comprised the following: achievement of all recommended levels of haemoglobin A1c (HbA1c), BP, and cholesterol; risky prescribing levels; achievement of recommended BP; and anticoagulation prescribing. Between February and March 2015, we recruited 144 general practices collectively serving over 1 million patients. We stratified computer-generated randomisation by area, list size, and pre-intervention outcome achievement. In April 2015, we randomised 80 practices to Trial 1 (40 per arm) and 64 to Trial 2 (32 per arm). Practices and trial personnel were not blind to allocation. Two practices were lost to follow-up but provided some outcome data. We analysed the intention-to-treat (ITT) population, adjusted for potential confounders at patient level (sex, age) and practice level (list size, locality, pre-intervention achievement against primary outcomes, total quality scores, and levels of patient co-morbidity), and analysed cost-effectiveness. The implementation package reduced risky prescribing (odds ratio [OR] 0.82; 97.5% confidence interval [CI] 0.67-0.99, p = 0.017) with an incremental cost-effectiveness ratio of £1,359 per quality-adjusted life year (QALY), but there was insufficient evidence of effect on other primary endpoints (diabetes control OR 1.03, 97.5% CI 0.89-1.18, p = 0.693; BP control OR 1.05, 97.5% CI 0.96-1.16, p = 0.215; anticoagulation prescribing OR 0.90, 97.5% CI 0.75-1.09, p = 0.214). No statistically significant effects were observed in any secondary outcome except for reduced co-prescription of aspirin and clopidogrel without gastro-protection in patients aged 65 and over (adjusted OR 0.62; 97.5% CI 0.39-0.99; p = 0.021). Main study limitations concern our inability to make any inferences about the relative effects of individual intervention components, given the multifaceted nature of the implementation package, and that the composite endpoint for diabetes control may have been too challenging to achieve. CONCLUSIONS:In this study, we observed that a multifaceted implementation package was clinically and cost-effective for targeting prescribing behaviours within the control of clinicians but not for more complex behaviours that also required patient engagement. TRIAL REGISTRATION:The study is registered with the ISRCTN registry (ISRCTN91989345).

Free full text 


Logo of plosmedLink to Publisher's site
PLoS Med. 2020 Feb; 17(2): e1003045.
Published online 2020 Feb 28. doi: 10.1371/journal.pmed.1003045
PMCID: PMC7048270
PMID: 32109257

An adaptable implementation package targeting evidence-based indicators in primary care: A pragmatic cluster-randomised evaluation

Thomas A. Willis, Conceptualization, Investigation, Methodology, Project administration, Writing – original draft, Writing – review & editing,1,* Michelle Collinson, Data curation, Formal analysis, Methodology, Writing – original draft, Writing – review & editing,2 Liz Glidewell, Conceptualization, Funding acquisition, Investigation, Methodology, Project administration, Writing – review & editing,3 Amanda J. Farrin, Conceptualization, Formal analysis, Funding acquisition, Investigation, Methodology, Supervision, Writing – review & editing,2 Michael Holland, Data curation, Formal analysis, Writing – original draft, Writing – review & editing,2 David Meads, Formal analysis, Methodology, Writing – original draft, Writing – review & editing,1 Claire Hulme, Conceptualization, Funding acquisition, Writing – review & editing,4 Duncan Petty, Investigation, Methodology, Project administration, Writing – review & editing,5 Sarah Alderson, Investigation, Methodology, Writing – review & editing,1 Suzanne Hartley, Conceptualization, Funding acquisition, Methodology, Project administration, Writing – review & editing,2 Armando Vargas-Palacios, Formal analysis, Writing – review & editing,1 Paul Carder, Methodology, Project administration, Resources, Writing – review & editing,6 Stella Johnson, Methodology, Project administration, Writing – review & editing,6 Robbie Foy, Conceptualization, Funding acquisition, Investigation, Methodology, Supervision, Writing – original draft, Writing – review & editing,1 and on behalf of the ASPIRE programme team
Sanjay Basu, Academic Editor

Associated Data

Supplementary Materials
Data Availability Statement

Abstract

Background

In primary care, multiple priorities and system pressures make closing the gap between evidence and practice challenging. Most implementation studies focus on single conditions, limiting generalisability. We compared an adaptable implementation package against an implementation control and assessed effects on adherence to four different evidence-based quality indicators.

Methods and findings

We undertook two parallel, pragmatic cluster-randomised trials using balanced incomplete block designs in general practices in West Yorkshire, England. We used ‘opt-out’ recruitment, and we randomly assigned practices that did not opt out to an implementation package targeting either diabetes control or risky prescribing (Trial 1); or blood pressure (BP) control or anticoagulation in atrial fibrillation (AF) (Trial 2). Within trials, each arm acted as the implementation control comparison for the other targeted indicator. For example, practices assigned to the diabetes control package acted as the comparison for practices assigned to the risky prescribing package. The implementation package embedded behaviour change techniques within audit and feedback, educational outreach, and computerised support, with content tailored to each indicator. Respective patient-level primary endpoints at 11 months comprised the following: achievement of all recommended levels of haemoglobin A1c (HbA1c), BP, and cholesterol; risky prescribing levels; achievement of recommended BP; and anticoagulation prescribing. Between February and March 2015, we recruited 144 general practices collectively serving over 1 million patients. We stratified computer-generated randomisation by area, list size, and pre-intervention outcome achievement. In April 2015, we randomised 80 practices to Trial 1 (40 per arm) and 64 to Trial 2 (32 per arm). Practices and trial personnel were not blind to allocation. Two practices were lost to follow-up but provided some outcome data. We analysed the intention-to-treat (ITT) population, adjusted for potential confounders at patient level (sex, age) and practice level (list size, locality, pre-intervention achievement against primary outcomes, total quality scores, and levels of patient co-morbidity), and analysed cost-effectiveness. The implementation package reduced risky prescribing (odds ratio [OR] 0.82; 97.5% confidence interval [CI] 0.67–0.99, p = 0.017) with an incremental cost-effectiveness ratio of £1,359 per quality-adjusted life year (QALY), but there was insufficient evidence of effect on other primary endpoints (diabetes control OR 1.03, 97.5% CI 0.89–1.18, p = 0.693; BP control OR 1.05, 97.5% CI 0.96–1.16, p = 0.215; anticoagulation prescribing OR 0.90, 97.5% CI 0.75–1.09, p = 0.214). No statistically significant effects were observed in any secondary outcome except for reduced co-prescription of aspirin and clopidogrel without gastro-protection in patients aged 65 and over (adjusted OR 0.62; 97.5% CI 0.39–0.99; p = 0.021). Main study limitations concern our inability to make any inferences about the relative effects of individual intervention components, given the multifaceted nature of the implementation package, and that the composite endpoint for diabetes control may have been too challenging to achieve.

Conclusions

In this study, we observed that a multifaceted implementation package was clinically and cost-effective for targeting prescribing behaviours within the control of clinicians but not for more complex behaviours that also required patient engagement.

Trial registration

The study is registered with the ISRCTN registry (ISRCTN91989345).

Author summary

Why was this study done?

  • Commonly used interventions to implement evidence-based practice, e.g., audit and feedback, educational outreach, and computerised prompts, generally have modest if variable effects on clinical performance.

  • The effects of such interventions may be enhanced by tailoring them to identified needs and barriers.

  • Trials of implementation interventions typically address single conditions; it is difficult to judge whether an intervention that works for one condition will work for another.

What did the researchers do and find?

  • We conducted two parallel, pragmatic trials to evaluate an implementation package for primary care that was adapted to overcome barriers for different clinical priorities.

  • General practices were randomly assigned to receive an implementation package targeting diabetes control or risky prescribing (Trial 1); blood pressure control or anticoagulation in atrial fibrillation (Trial 2). Respective primary endpoints assessed were as follows: achievement of all recommended levels of haemoglobin A1c, BP, and cholesterol; risky prescribing levels; achievement of recommended BP; and anticoagulation prescribing.

  • The implementation package produced a significant clinically and cost-effective reduction in one target only: risky prescribing.

What do these findings mean?

  • In this study, we found that an adaptable implementation package was cost-effective for targeting prescribing behaviours within the control of clinicians, but not for more complex behaviours that also required patient engagement.

  • Given known associations between risky prescribing combinations and increased morbidity, mortality, and health service use, a scaled-up risky prescribing implementation package could have an important population impact.

Introduction

Clinical research can only benefit patient and population health if findings are incorporated into routine care. There are delays and inappropriate variations in uptake of effective treatments and withdrawal of less effective or harmful treatments [1]. This translation gap is important to policy makers, healthcare systems, and research funders because it limits the health, social, and economic impacts of clinical research [2].

United Kingdom primary care presents particular implementation challenges: growing demand, increasing complexity of care, and limited workforce capacity, against a background of frequent organisational reconfigurations [3,4]. We identified 107 clinical guidelines produced by the National Institute for Health and Care Excellence (NICE) relevant to UK general practice [5]. Many implementation studies focus on a single clinical condition or problem (e.g., diabetes, antibiotic stewardship), limiting generalisability, as it is uncertain whether an implementation strategy developed for one condition will work for another. It is impracticable and inefficient to devise an implementation strategy for every new guideline. Furthermore, the clinical significance of behaviours often targeted in implementation studies, such as receipt of processes of care or investigations, is doubtful [6]. Implementation strategies are required, capable of integration into primary care systems and adaptable to different clinical priorities.

We earlier derived evidence-based indicators that could be measured using routinely collected data [5]. We found marked variations in indicator achievement and scope for improvement in a sample of 89 general practices [7]. We subsequently focused our efforts on four ‘high impact’ indicators that would benefit population health if implemented more consistently: achievement of recommended treatment targets for all of haemoglobin A1c (HbA1c), blood pressure (BP), and cholesterol in type 2 diabetes [8]; avoidance of risky prescribing of nonsteroidal anti-inflammatory drugs (NSAIDs) and anti-platelet drugs [9]; achievement of recommended BP levels in patients at high risk of cardiovascular events [10]; and anticoagulant prescribing for stroke prevention in atrial fibrillation (AF) [11]. We drew upon systematic reviews of implementation strategies [6,1214], theory-guided interviews with primary care staff [15], and workshops with health professionals and patients to develop a multifaceted implementation package [16]. We compared the effects of the adaptable implementation package against implementation control on adherence to four different evidence-based high impact indicators.

Methods

Study design and participants

We conducted two parallel, cluster-randomised trials using balanced incomplete block designs. In implementation trials, there may be positive attention or negative demotivation effects from participant knowledge of allocation to an intervention or control group, respectively. Balanced incomplete block designs aim to balance any such nonspecific effects across trial arms, as each arm receives an intervention, thereby increasing confidence that any difference in outcomes is attributable to the intervention [17]. The design is incomplete, as each arm receives only one of the interventions. General practices providing National Health Service (NHS) care were the unit of allocation, as the intervention was delivered at the practice level. We maximised pragmatism in trial design and execution to ensure ‘real-world’ relevance [18].

Practices were recruited from West Yorkshire, England, which covers a diverse population of 2.2 million residents, albeit with deprivation levels above the national average [19]. Around 300 general practices were then organised within 10 clinical commissioning groups (CCGs).

Eligible general practices used SystmOne, the computerised clinical system used by approximately two thirds of West Yorkshire practices (The Phoenix Partnership, http://www.tpp-uk.com). We excluded 31 practices that had contributed to earlier intervention development work. We invited practices via recorded post and email, with reminders at two weeks. The UK National Research Ethics Service granted ethical approval (14/SC/1393). This permitted use of an opt-out approach: all eligible practices were included, unless actively declined within four weeks of invitation. The study protocol has previously been published [20]. The study is registered with the ISRCTN registry (ISRCTN91989345). This study is reported as per the CONSORT guideline for cluster-randomised trials (S1 CONSORT checklist).

Randomisation and masking

The trial statistician performed the two-stage randomisation using a bespoke computer-generated minimisation programme implemented in R, software version 2.15.2 (R Foundation for Statistical Computing, Vienna, Austria) (incorporating a random element). First, practices were stratified by CCG and list size, and randomised to Trial 1 (implementation packages targeting either diabetes control or risky prescribing), Trial 2 (BP control or anticoagulation in AF), or no intervention (80:64:34). As recruitment exceeded expectations, we included a ‘no intervention’ arm to allow us to evaluate any nonspecific effects of research participation (this paper presents comparisons within Trials 1 and 2; analysis of the no intervention group will be reported elsewhere). Secondly, within each trial we stratified practices by CCG, list size, and pre-intervention adherence to the two relevant targeted indicators and randomised them 1:1 to individual trial arms (Fig 1). Within each trial, we assumed that any clinical effects of either implementation package would be independent of one another; each intervention arm therefore acted as the control arm for the other intervention arm in the same trial. For example, practices assigned to the diabetes control package acted as the implementation control comparison for practices assigned to the risky prescribing package. Practices from one CCG (29 practices) involved in a concurrent initiative targeting one of our study priorities, anticoagulation in AF, were excluded from Trial 2. Practices and trial personnel were not blind to allocation.

An external file that holds a picture, illustration, etc.
Object name is pmed.1003045.g001.jpg
Trial profile.

aSafe haven practices are for those patients who have demonstrated violent tendencies, who have been removed from usual general practice, or for ex-offenders. bThe flow diagram presents the whole randomisation process, but this paper reports the planned comparisons within Trials 1 and 2. The no intervention arm is shaded to indicate that it was not included in the analyses described here; analysis of this group will be reported elsewhere. cOne practice in the risky prescribing arm merged with a non-ASPIRE practice in advance of the fourth feedback report. However, as they received the first three feedback reports, some outcome data are available, and they are included in the final analyses. dOne practice in the BP control arm closed in advance of the third feedback report. However, as they received the first two feedback reports, some outcome data are available and they are included in the final analyses. As an example, for the diabetes control practices: there were 14,522 patients acting as the intervention group for the diabetes control implementation package and 18,131 patients acting as the control group for the risky prescribing implementation package. A&F follow-up: delivery of audit and feedback reports during the intervention period; AF, atrial fibrillation; BP, blood pressure; RP, risky prescribing.

General practice and trial personnel involved in intervention delivery and trial statisticians responsible for analysis were, of necessity, aware of allocation assignment. Data collection for all endpoints was masked to allocation.

Procedures

The implementation package (detailed elsewhere [16]) embedded behaviour change techniques within typical primary care interventions: audit and feedback, educational outreach, and computerised support. Implementation package content, specifically the clinical content and embedded behaviour change techniques, was adapted for each of the four targeted indicators and delivered to practices May 2015 to March 2016.

Audit and feedback aimed to give comparative feedback on achievement, inform and prompt recall of clinical goals, highlight consequences of changing or not changing practice, suggest strategies for change, and encourage goal setting and reflection on progress towards goals. Quarterly electronic and paper practice–specific reports presented achievement ranked by practice and compared over time for relevant trial indicators in graphical and numerical forms, using remotely gathered, individualised practice data. Reports also contained brief, evidence-based clinical messages; responses to common queries; and action-planning templates. Practices received computerised search tools to identify relevant patients for review. We encouraged practices to use the feedback reports as supporting materials for professional appraisal and revalidation.

Educational outreach aimed to enhance feedback reports by facilitating individual and group reflection, discussing barriers to action, sharing models of good practice, enhancing motivation, and action planning. We trained pharmacists over two days to deliver 30-minute sessions, designed to fit with existing practice meetings and offered to all staff involved in patient and practice management. We identified a key clinical contact to support practice engagement. We offered a second, follow-up session to review progress and refine action plans, as well as two days of pharmacist support for patient identification and review.

Prompts and reminders reinforced clinical messages and indicator adherence. Computerised prompts for risky prescribing were offered to practices and had to be accepted before they became active on their system. If accepted, they were triggered during consultations and repeat prescribing through an algorithm for patient age, diagnosis, drug, and duration. A one-click justification (ignore, or add or stop medication) was required before users could proceed. Other targeted indicators had existing prompts on practice computer systems, usually to support the Quality Outcomes Framework (QOF), a performance management system whereby general practices are remunerated according to achievement of targets reflecting quality of care [21]. We provided laminated reminders to convey key clinical information (e.g., management pathways) for BP control, anticoagulation for AF, and risky prescribing. We also developed checklists to facilitate shared decision-making with patients for managing BP and diabetes, but we could not make them available in a satisfactory format within the study time lines [16].

We deliberately aligned intervention delivery with the QOF year to fit with existing practice schedules. We assumed that any actions prompted by the implementation package would occur within this period.

Outcomes

Anonymised patient-level outcome data were obtained remotely from general practices from SystmOne. We planned to measure all outcomes at 12 months post-randomisation. However, a one-month delay in randomisation effectively meant that we were only able to assess outcomes at 11 rather than 12 months.

The primary outcomes for each intervention arm comprised the following:

Trial 1

  • Proportion of patients with type 2 diabetes achieving all three treatment targets:

    1. BP below 140/80 mmHg (or 130/80 mmHg if kidney, eye, or cerebrovascular damage);

    2. HbA1c value below or equal to 59 mmol/mol;

    3. cholesterol level below or equal to 5.0 mmol/L.

  • Proportion of patients meeting at least one of nine indicators of high-risk NSAID and anti-platelet prescribing:

    1. prescription of a traditional oral NSAID or low-dose aspirin in patients with a history of peptic ulceration without co-prescription of gastro-protection;

    2. traditional oral NSAID in patients aged 75 years or over without co-prescription of gastro-protection;

    3. traditional oral NSAID and aspirin in patients aged 65 years or over without co-prescription of gastro-protection;

    4. aspirin and clopidogrel in patients aged 65 years or over without co-prescription of gastro-protection;

    5. warfarin and traditional oral NSAID;

    6. warfarin and low-dose aspirin or clopidogrel without co-prescription of gastro-protection;

    7. oral NSAID in patients with heart failure;

    8. oral NSAID in patients prescribed both a diuretic and an angiotensin-converting-enzyme inhibitor (ACE-I) or angiotensin receptor blocker (ARB);

    9. oral NSAID in patients with chronic kidney disease (CKD).

Trial 2

  • Proportion of patients achieving the lowest appropriate BP target:

    1. under 140/90 mmHg if aged under 80 years with hypertension, coronary heart disease, peripheral arterial disease, a history of stroke or transient ischemic attack, or a 10-year cardiovascular disease risk of 20% or higher;

    2. under 150/90 mmHg if aged 80 years and over with hypertension;

    3. under 140/80 mmHg if aged under 80 years with diabetes, under 130/80 mmHg if complications of diabetes or aged under 80 years with CKD and proteinuria.

  • Combined proportion of men with AF and a congestive heart failure, hypertension, age ≥75, diabetes, stroke, vascular disease, age between 65 and 74, and female sex (CHA2DS2-VASc) score of 1 and women with a CHA2DS2-VASc score of 2 or above prescribed anticoagulation therapy.

Secondary outcomes (S1S4 Tables) included individual indicators contributing to composite primary outcomes, processes of care, and continuous intermediate clinical outcomes (last recorded BP, HbA1c, cholesterol). We assessed QOF indicators relevant to implementation packages (diabetes control, BP control, and anticoagulation for AF) to examine any effects on existing routine indicators. We assessed QOF indicators not directly targeted by the implementation packages to assess any unintended wider impacts. These included coronary heart disease and asthma as example long-term physical conditions, mental health (long-term nonphysical condition), and smoking (health promotion).

We obtained practice characteristic data from publicly available sources (Health Education England, Health and Social Care Information Centre): practice list size (number of registered patients); number of general practitioner (GP) partners and salaried GPs; practice training status; practice level Index of Multiple Deprivation (IMD); ethnic profile of practice register; achievement of QOF indicators (2014–2015); patient satisfaction (proportion who would recommend the practice to others); patient-rated practice accessibility (proportion able to speak with GP or nurse within 48 hours of approach); and practice prescribing costs. Patient characteristics (age, sex, comorbidity [number of QOF disease registers on which the patient appeared]) and outcome data were extracted from SystmOne by the local primary care commissioning support unit.

We monitored intervention delivery and fidelity via standardised report forms completed by outreach facilitators following each visit. We recorded reasons for declining visits. We monitored which practices accepted the invitation to join indicator-specific SystmOne organisational groups to access patient searches and any related computerised prompts.

Sample size

Median effect sizes on processes and outcomes of care for a range of guideline implementation studies are around 4%–9% [6]. Given our enhanced interventions and targeting of indicators with scope for improvement [12], we judged an absolute difference of 15% for outcomes related to diabetes, BP control, and anticoagulation for AF as realistic and clinically relevant. Baseline achievement rates in risky prescribing were considerably higher and, considering a potential ceiling effect, a 5% decrease was realistic and clinically relevant.

We aimed to recruit 144 practices: 80 in Trial 1 (diabetes and risky prescribing) and 64 in Trial 2 (BP control and AF), giving 90% power at 2.5% significance (adjusting for two comparisons per trial) to detect the specified effect sizes, allowing for 10% attrition. The mean cluster size (number of patients per practice by indicator), coefficient of variation, intra-cluster correlation coefficient (ICC), and control group achievement rates were estimated from previously collected data [7,20] (S5 Table).

Statistical analysis

Analyses, conducted in SAS software version 9.4 (SAS Institute Inc., Cary, NC) according to the prespecified statistical analysis plan (S1 statistical analysis plan), were based on intention-to-treat (ITT), with two-sided significance testing at the 2.5% level to preserve an overall 5% Type I error rate per trial. We analysed primary outcomes using two-level binary logistic regression models, with patients (level 1) nested within randomised practices (level 2). We analysed binary secondary outcomes for individual indicators within the composite primary outcomes, and recorded processes of care using similar multilevel logistic models. We analysed continuous intermediate clinical outcomes using two-level linear models. All analyses adjusted for patient-level (sex, age) and practice-level covariates (baseline practice list size, CCG, pre-intervention achievement against primary outcomes, total QOF score 2014–2015 [covering clinical and management domains], proportion of patients with 0–3 comorbidities). Sensitivity analyses were conducted when model assumptions were violated. Data completeness for these analyses depended on the completeness of SystmOne medical records and could not be assessed within the trial data set. Missing data in a patient’s record could have led to their incorrect inclusion or exclusion from the denominator of a particular quality indicator or resulted in the patient being incorrectly classified as achieving that indicator. For the primary and secondary analyses, we assumed that data were missing at random. Imputation was not performed for any outcome variables. Complete case analyses were performed for the continuous clinical outcomes. No adjustment for multiple comparisons was made in the secondary endpoint analyses.

Cost-effectiveness

We conducted economic evaluations of three implementation packages using a decision-analytic modelling approach. Given resource constraints, we were unable to evaluate the anticoagulation for AF package. Implementation package costs included researcher and healthcare professional time and materials in delivery and resulting from change in practice. Trial estimates of effectiveness were inputted into decision models, with the main output being cost (UK £, 2017 prices) per incremental quality-adjusted life year (QALY) over a lifetime horizon from the perspective of the UK health and social care provider. Where possible we used models, parameters, and assumptions that had informed the original NICE guidance. For diabetes control, we used the UK Prospective Diabetes Study Outcomes Model (UKPDS-OM) version 2 [22]. For BP control, we recreated and adapted the model used to inform NICE guidance [23], identifying additional parameters through targeted searches and risk engines such as QRISK. We based the risky prescribing model on a previous model of NSAID prescribing used in a NICE osteoarthritis guideline (CG177). We adapted this model and populated it using results reported for three of the most commonly prescribed NSAIDs in the UK (diclofenac, naproxen, and ibuprofen) and aspirin. We created an additional model based on that used to inform the NICE Acute Kidney Injury guideline (CG169), covering NSAID prescribing in patients with CKD. Individual indicators were evaluated and their cost-effectiveness aggregated within each implementation package. We conducted a series of deterministic sensitivity analyses and a probabilistic sensitivity analysis. We used a willingness to pay threshold of £20,000 per QALY gained to indicate cost-effectiveness. We applied a 3.5% discount rate to costs and benefits post one year.

Results

We assessed 278 practices for eligibility in January 2015, excluding 35 because of closure or earlier participation in the programme (Fig 1). We invited 243 (87%) practices to participate in February 2015; 56 (23%) opted out, largely because of workload pressures, and nine were excluded for other reasons.

In April 2015, we randomised 178 (73%) practices to Trial 1 (80 practices), Trial 2 (64 practices), or no intervention (34 practices). Analyses of the no intervention arm will be reported elsewhere. Within each trial, practices were randomised 1:1 to intervention arms.

Baseline characteristics for general practices and patients were well balanced by trial and intervention (Tables (Tables11 and and2).2). The mean practice IMD was 30.22 (SD 13.86) within the top quarter of UK social deprivation [24].

Table 1

General practice characteristics at baseline by trial arm.
Practice characteristicsTrial interventionTotal (n = 144)a
Diabetes control (n = 40)Risky prescribing (n = 40)BP control (n = 32)Anticoagulation in AF (n = 32)
List size
Mean (SD)7,084.4 (3,786.5)7,175.8 (3,857.0)7,538.9 (4,932.9)7,421.3 (4,171.2)7,285.6 (4,128.9)
Overall QOF score 2014/2015b
Mean (SD)535.4 (29.9)531.1 (35.8)527.2 (27.0)533.0 (21.7)531.9 (29.4)
Pre-intervention achievement (%) on primary outcome
Diabetes control
Mean (SD)32.9 (6.9)34.3 (7.7)32.5 (7.1)33.4 (5.5)33.3 (6.9)
Risky prescribing
Mean (SD)7.9 (5.1)7.9 (3.6)7.3 (3.6)7.9 (2.5)7.8 (3.9)
BP control
Mean (SD)66.5 (6.4)66.4 (7.0)65.9 (7.5)65.3 (6.2)66.1 (6.8)
Anticoagulation in AF
Mean (SD)66.5 (14.4)67.3 (8.4)66.5 (10.8)66.3 (8.3)66.7 (10.8)
Deprivation score (IMD 2015)
Mean (SD)30.3(13.0)32.4 (13.7)28.3 (14.6)29.3 (14.7)30.2 (13.9)
Number of GPs (FTE)
Mean (SD)4.0 (3.0)4.1 (2.6)4.2 (3.1)4.0 (2.8)4.1 (2.8)
Number of GP partners (FTE)
Mean (SD)3.3 (2.6)3.5 (2.4)3.6 (2.8)3.4 (2.5)3.4 (2.5)
Percentage of patients who would recommend practice to others
Mean (SD)75.4 (14.5)73.9 (14.0)74.7 (13.9)76.0 (13.5)75.0 (13.9)
Percentage of patients who saw/spoke to nurse or GP within 48 hours of approach
Mean (SD)51.7 (17.3)50.7 (15.6)50.8 (12.9)52.7 (16.1)51.5 (15.5)
Teaching practice?
Yes14 (35%)15 (38%)13 (41%)15 (47%)57 (40%)
No26 (65%)25 (63%)19 (59%)17 (53%)87 (60%)

aThe practices randomised to the no intervention arm are not included here.

bThere was one practice with a missing value for overall QOF score in Trial 2. The 2014/2015 QOF measured achievement against 81 indicators; practices scored points on the basis of achievement against each indicator, up to a maximum of 559.

Abbreviations: AF, atrial fibrillation; BP, blood pressure; FTE, full-time equivalent; GP, general practitioner; IMD, Index of Multiple Deprivation; QOF, Quality and Outcomes Framework, total score 2014/2015

Table 2

Patient characteristics at baseline by trial arm (all patients from all practices in each intervention arm).
Patient characteristicsTrial interventionTotal (n = 1,067,402)a
Diabetes control (n = 288,130)Risky prescribing (n = 290,407)BP control (n = 249,571)Anticoagulation in AF (n = 239,294)
Age (years)
Mean (SD)38.0 (22.9)37.6 (23.1)39.4 (23.2)39.0 (23.2)38.4 (23.1)
Sexb
Female141,328 (49%)144,426 (50%)124,824 (50%)120,289 (50%)530,867 (50%)
Comorbidityc
0–3276,280 (96%)277,184 (95%)239,455 (96%)229,329 (96%)1,022,248 (96%)
4+11,850 (4%)13,223 (5%)10,116 (4%)9,965 (4%)45,154 (4%)

aThe practices randomised to the no intervention arm are not included here.

bEach arm included <0.001% of patients defined as indeterminate or unknown.

cMeasured as the number of QOF registers on which a patient appears.

Abbreviations: AF, atrial fibrillation; BP, blood pressure; QOF, Quality Outcomes Framework

No practices actively withdrew from the trial. One practice closed (BP intervention arm), another merged (risky prescribing) with a non-trial practice during the intervention period, and two trial practices merged (diabetes and risky prescribing). As outcome data for these practices were available up to the times of the second, third, and fourth feedback reports, respectively, all were included in the analysis using their most recent data.

Between May 2015 and April 2016, all intervention practices received feedback reports as intended, including an end-of-study report (S6 Table summarises intervention delivery). Educational outreach visits were delivered to 67 (47%) of 144 practices; 52 (68%) of the 77 practices declining visits gave no reason. Sixteen practices (24% of those receiving educational outreach visits) utilised additional pharmacist support. One hundred twenty-six (88%) practices joined the organisational groups, allowing them to access searches and computerised prompts (risky prescribing practices only). Second educational visits were delivered to eight (6%) practices.

Primary analyses demonstrated varying results across the implementation packages at 11 months post-randomisation (Table 3).

Table 3

Primary outcome achievement: Baseline rates, outcome rates, and ORs adjusted for baseline achievement and covariates.
Primary outcomesBaseline achievementa (%)Unadjusted model estimatesAdjusted model estimatesc
Outcome achievementb (%)OR (97.5% CI)p-valueOutcome achievementb (%)OR (97.5% CI)p-valueICC
Trial 1: Diabetes control
Intervention33.724.31.016 (0.86–1.20)0.83724.21.025 (0.89–1.18)0.6930.015
Control (risky prescribing)34.424.023.7
Trial 1: Risky prescribing
Intervention7.25.20.783 (0.59–1.04)0.0524.90.815 (0.67–0.99)0.0170.022
Control (diabetes control)7.46.56.0
Trial 2: BP control
Intervention66.752.91.067 (0.94–1.22)0.26653.61.053 (0.96–1.16)0.2150.006
Control (anticoagulation in AF)65.551.252.3
Trial 2: Anticoagulation in AF
Intervention66.472.80.866 (0.68–1.10)0.17573.20.902 (0.75–1.09)0.2140.009
Control (BP control)67.575.575.2

aCalculation of achievement for diabetes control and BP control at baseline uses any BP measurement taken in the previous 12 months.

bCalculation of achievement for diabetes control and BP control at outcome uses the most recent BP measurement taken.

cVariables controlled for in the adjusted analyses were as follows: patient-level sex and age, and practice-level baseline list size, CCG, pre-intervention achievement against primary outcomes, total QOF score 2014–2015, and proportion of patients with 0–3 comorbidities.

Abbreviations: AF, atrial fibrillation; BP, blood pressure; CCG, clinical commissioning group; CI, confidence interval; ICC, intra-cluster correlation coefficient; OR, odds ratio; QOF, Quality Outcomes Framework

The diabetes implementation package had no observed effect on diabetes treatment targets. Achievement was 24.2% in intervention practices; 23.7% in control practices (adjusted odds ratio [OR] 1.03; 97.5% confidence interval [CI] 0.89–1.18; p = 0.693; ICC = 0.015).

The risky prescribing implementation package reduced high-risk NSAID and anti-platelet prescribing. In intervention practices, 4.9% of patients had a record of risky prescribing; 6.0% in control practices (adjusted OR 0.82; 97.5% CI 0.67–0.99; p = 0.017; ICC = 0.022).

The BP implementation package had no observed effect on BP control. Achievement was 53.6% in intervention practices; 52.3% in control practices (adjusted OR 1.05, 97.5% CI 0.96–1.16; p = 0.215; ICC = 0.006).

The AF implementation package had no observed effect on anticoagulant prescribing. Achievement was 73.2% in intervention practices; 75.2% in control practices (adjusted OR 0.90; 97.5% CI 0.75–1.09; p = 0.214, ICC = 0.009).

There was little evidence of any intervention effects for any secondary outcome (S3S6 Tables) except the risky prescribing implementation package, which showed some evidence of improvement against an individual indicator: patients aged 65 years or over prescribed aspirin and clopidogrel without co-prescription of gastro-protection. Adjusted prescribing levels were 25.3% in intervention practices, compared to 35.2% in control practices (adjusted OR 0.62; 97.5% CI 0.39–0.99; p = 0.021). The conclusions for total serum cholesterol and HbA1c were robust to log transformation of the outcomes in a sensitivity analysis conducted following model diagnostic checks. We observed no intervention effects on any QOF indicators, including those not targeted by the implementation packages.

In estimating cost-effectiveness, we assumed equal costs for all four implementation packages. We conservatively doubled the cost per package. The total cost of package delivery was £175,592, and cost per practice was £2,439 (£175,592/144*2). Costs per patient (not only those eligible for indicator criteria) for an average practice list size of 7,130 were negligible (£0.28). No interventions were cost-saving. The risky prescribing package is highly likely to be cost-effective with an ICER of £1,359 and 79% chance of cost-effectiveness (Table 4), with results largely driven by two indicators. While the deterministic ICER for the BP package indicates cost-effectiveness, this is highly uncertain, with the probabilistic results indicating only a 52% chance of cost-effectiveness. We have not presented model results for the diabetes package as incremental costs and benefits were negligible, making model results unreliable. Results were robust to deterministic sensitivity analyses when, for example, we increased and reduced intervention costs. S1S4 Figs present cost-effectiveness planes and acceptability curves for the risky prescribing and BP control implementation packages.

Table 4

Cost-effectiveness analysis: Mean probability sensitivity analysis outcomes at the practice level for the primary risky prescribing and BP control outcomes.
Probability sensitivity analysis outcomesRisky prescribingBP control
Mean incremental QALY0.9013.00
Mean incremental cost£1,225£42,192
ICER£1,359£3,246
Incremental Net Monetary Benefit*£16,810£217,730
Probability cost-effective0.790.52

*Assumes willingness to pay threshold of £20,000.

Abbreviations: BP, blood pressure; ICER Incremental cost-effectiveness ratio; QALY, quality-adjusted life year

Discussion

Our pragmatic, randomised trials found that an adaptable, multifaceted implementation package improved clinical care for only one of four high-impact indicators in general practices serving relatively socially deprived populations. The odds of risky prescribing for a patient in an intervention practice were 18.5% lower than for a patient with the same characteristics in a control practice, which could ultimately be associated with changes in mortality, morbidity, and health service use, depending on generalisability to the general population. There was insufficient evidence of an effect upon diabetes control, BP control, and anticoagulation for AF. Our findings suggest that the design and delivery of implementation strategies need to account for differences in the nature of targeted clinical behaviours and go further than the kinds of adaptations in content that we applied.

Commonly used interventions to implement evidence-based practice, such as audit and feedback, educational outreach, and computerised prompts generally have modest if variable effects on clinical performance [1214]. Tailoring such interventions to identified needs and barriers offers a means to enhance their effects, but how best to do this and improve patient outcomes remains uncertain [25].

Our findings confirm the effectiveness, generalisability, and value of interventions incorporating audit and feedback to improve prescribing safety in UK primary care [2628]. The PINCER trial compared one-off feedback with pharmacist outreach visits to one-off feedback alone, whilst the D-QIP study provided financial incentives in addition to weekly feedback. Given our pragmatic approach to practice recruitment, our intervention and findings compare most closely to those of the EFIPPS trial, which found that quarterly feedback, with or without embedded behaviour change techniques, reduced risky prescribing by a similar magnitude [28]. Our modelling also indicated relative cost-effectiveness, mainly driven by improved levels of gastro-protection. It is noteworthy that the cost of the risky prescribing package was only £0.28 per patient.

We found no benefit of the implementation package on the management and outcomes of our targeted long-term conditions. Systematic review evidence suggests that interventions targeting systems of patient management along with patient-mediated interventions (which we did not include) are likely to be important components of strategies in this context [29].

The balanced incomplete block designs permitted comparison of intervention effects while mitigating any potential nonspecific performance effects of trial participation [30]. Our trials were pragmatic in three ways. Opt-out recruitment ensured that participating practices were generally representative of the wider population [19]. Data collection was minimally intrusive. For intervention delivery, all practices received but were not obliged to act on feedback reports, whilst outreach visits were optional. Hence, the implementation packages were tested under real-world conditions, increasing confidence in wider applicability to routine general practice settings. We further demonstrated no adverse impacts on incentivised indicators of care not targeted by the implementation package.

Our evaluation had five main limitations. First, the use of routinely collected data may have compromised the precision of trial outcomes and hence ability to demonstrate effects. However, we extracted structured data that are reasonably reliably coded in general practice, partly incentivised by QOF. Second, given the multifaceted nature of the implementation package, we cannot make any inferences about the relative effects of individual intervention components. Third, educational outreach visits were delivered by facilitators not allocated to specific trial arms, thereby risking contamination between arms. We had instructed facilitators to focus only on delivering the implementation package to which each practice was assigned. Fourth, an 11-month follow-up period may have been too short to detect changes in patient outcomes, such as those related to diabetes or BP control, especially if a number of general practices only received educational outreach visits later in this period. This explanation is unlikely, as other trials have demonstrated changed clinical outcomes within similar durations of follow-up [29]; we also did not detect any improvements in processes of care for diabetes and BP control, respectively. Fifth, our composite endpoint for diabetes control requiring achievement of all HbA1c, BP, and cholesterol treatment goals may have been too demanding. Our clinical and patient advisors had considered this endpoint fair, if challenging.

Our work highlights three methodological issues. First, the implementation package effect on risky prescribing was modest but important at a population level. Foregoing a randomised design, as has been suggested for quality improvement research [31], would have reduced confidence in the validity of our findings and risked false positive conclusions. For example, the trial design accounted for temporal changes, including improvements in risky prescribing and anticoagulation achievement and declines in BP and diabetes achievement (Table 3), which would otherwise be difficult to interpret. Second, whilst we see the pragmatic design as a strength, a more explanatory approach could have made full engagement with our implementation package a condition of trial participation. Such mandating is seldom possible or even desirable in quality improvement programmes dependent upon professional engagement, particularly if they encourage ‘gaming’ behaviours to achieve goals whilst circumventing real action. Similarly, our opt-out approach to recruitment may have reduced self-selection of more enthusiastic practices, as well as administrative burden. Those responsible for leading quality improvement initiatives often specifically wish to include less enthusiastic or poorer performing practices. Third, a critical challenge prior to pragmatic evaluations is to develop interventions that are sufficiently durable to withstand the relatively hard-pressed and evolving environments of clinical practice. Whilst we followed the UK Medical Research Council framework for the development and evaluation of complex interventions, practice engagement with our implementation package was highly variable [32]. We would now recommend more intensive field work involving iterative cycles of testing and refining interventions prior to scaling up for definitive evaluation.

We had set out to develop an implementation package that could be adapted for different clinical priorities in primary care. We offer four interrelated explanations for the observed differences in intervention effects. First, the targeted clinical behaviour(s) and associated endpoints varied according to the extent of control held over them by clinical staff. Clinicians could make relatively straightforward changes to reduce risky prescribing, such as adding gastro-protection for prescribed aspirin, with limited input from patients. The observed significant difference in achievement of the associated secondary outcome on gastro-protection provides evidence to support such a mechanism (although we would still advise cautious interpretation, given that these analyses were not formally adjusted as we tested across multiple secondary endpoints). In contrast, improving BP control can require at least two consultations and changes in patient behaviour, as well as finding a pharmacological agent that is acceptable to and effective for an individual patient. This is consistent with evidence that adherence to clinical recommendations that are more complex or disruptive to routine practice is lower compared with simpler recommendations [33]. Interventions involving audit and feedback have also been shown to reduce other discouraged prescribing behaviours, namely of antibiotics [34], and offer a means to address urgent priorities such as rising opioid prescribing [35]. Second, the risky prescribing package included an automated computerised prompt requiring a one-step justification for continued prescribing. Such prompts are generally effective in changing prescribing behaviour [14]. However, a recent UK trial found that on-screen reminders did not improve adherence to the other prescribing behaviour we targeted, anticoagulation prescribing for AF [36]. With the benefit of hindsight, this is unsurprising. Qualitative work we undertook before the trials revealed that anticoagulation prescribing comprises a series of deceptively complex considerations and behaviours, which include balancing benefits and risks with patients [15]. Clinicians sometimes lacked confidence in starting treatment, given that they encountered it relatively infrequently in routine practice and felt frustrated by complicated guidance that made treatment difficult to explain to patients. Third, the number of patients requiring action to achieve indicators varied by trial arm, with far more in the diabetes and BP control arms relative to risky prescribing and anticoagulation for AF (see S5 Table). Hence, the larger numbers of patients requiring action may have undermined clinicians’ perceived feasibility and motivation. Moreover, making changes for even a small number of patients within the risky prescribing indicator set would have had a comparatively larger effect upon achievement relative to other indicators. Fourth, the indicators for three targeted long-term conditions were incentivised in the QOF. As well as aligning the implementation package with existing quality improvement schemes, our intention had been to encourage practices to move beyond the QOF by adopting more challenging, evidence-based goals. Within the present constraints of UK primary care, it was hard for practices to take on additional, unrewarded work. Clinicians may, nevertheless, have been motivated to address risky prescribing because it concerned patient safety.

In conclusion, an adaptable implementation package improved prescribing safety in general practice. However, we observed no benefits of the package within the context of an existing financial incentive system targeting similar aspects of care for three long-term conditions. Improving patient outcomes for long-term conditions requiring relatively complex management may require systemised approaches that target patient as well as professional behaviour.

Supporting information

S1 Table

Secondary outcomes from Trial 1: Achievement of individual indicators that contributed to composite outcomes; processes of care; and continuous intermediate clinical outcomes.

All adjusted for covariates and baseline achievement of primary outcomes. Table presents mean percentage achievement, unless otherwise stated. Variables controlled for in the adjusted analyses were as follows: patient-level sex and age, and practice-level baseline list size, CCG, pre-intervention achievement against primary outcomes, total QOF score 2014–2015, and proportion of patients with 0–3 comorbidities. ACE-I, angiotensin-converting-enzyme inhibitor; ACR: albumin:creatinine ratio; ARB, angiotensin receptor blocker; BMI, body mass index; BP, blood pressure; CCG, clinical commissioning group; CI, confidence interval; CKD, chronic kidney disease; eGFR, estimated glomerular filtration rate; HbA1c, haemoglobin A1c; NSAID, non-steroidal anti-inflammatory drug; PCR, protein:creatinine ratio; QOF, Quality Outcomes Framework.

(DOCX)

S2 Table

Secondary outcomes from Trial 1: Achievement of QOF indicators relating to the implementation packages; and non-trial related QOF indicators.

All adjusted for covariates and baseline achievement of primary outcomes. Note: Formal statistical testing was inappropriate due to violation of the modelling assumptions for the following trial-related indicators: DM006, DM014, DM018; and the following non-trial related indicators: CHD005, CHD007, MH002, MH003, SMOK004, and SMOK005. Summary statistics only are presented for these indicators. The HbA1c and total serum cholesterol continuous intermediate clinical outcomes were analysed using a log transformation in order to satisfy the modelling assumptions. The predicted means presented are on the untransformed (original) scale, but the estimated intervention effect (and 97.5% CI) are on the log scale. *If urine albumin:creatinine ratio ≥3, or retinopathy, or record of cerebrovascular accident or transient ischemic attack. Variables controlled for in the adjusted analyses were as follows: practice-level baseline list size, CCG, pre-intervention achievement against primary outcomes, and total QOF score 2014–2015. ACE-I, angiotensin-converting-enzyme inhibitor; ARB, angiotensin receptor blocker; CCG, clinical commissioning group; CHD, coronary heart disease; CI, confidence interval; CKD, chronic kidney disease (stage 3–5); COPD, chronic obstructive pulmonary disease; HbA1c, haemoglobin A1c; IFCC, International Federation of Clinical Chemistry and Laboratory Medicine; PAD, peripheral arterial disease; QOF, quality and outcomes framework; RCP, Royal College of Physicians; TIA, transient ischemic attack.

(DOCX)

S3 Table

Secondary outcomes from Trial 2: Achievement of individual indicators that contributed to composite outcomes; processes of care; and continuous intermediate clinical outcomes.

All adjusted for covariates and baseline achievement of primary outcomes. Values are percentage achievement, unless otherwise stated. Variables controlled for in the adjusted analyses were as follows: patient-level sex and age, and practice-level baseline list size, CCG, pre-intervention achievement against primary outcomes, total QOF score 2014–2015, and proportion of patients with 0–3 comorbidities. *If urine albumin:creatinine ratio ≥3, or retinopathy, or record of cerebrovascular accident or transient ischemic attack. AF, atrial fibrillation; BP, blood pressure; CCG, clinical commissioning group; CHA2DS2-VASc, congestive heart failure, hypertension, age>75, diabetes, stroke, vascular disease, age between 65 and 74, and female sex; CHD, coronary heart disease; CI, confidence interval; CKD, chronic kidney disease; CVD, cardiovascular disease; HTN, hypertension; PAD, peripheral arterial disease; QOF, Quality Outcomes Framework; TIA, transient ischemic attack.

(DOCX)

S4 Table

Secondary outcomes from Trial 2: Achievement of QOF indicators relating to the implementation packages; and non-trial-related QOF indicators.

All adjusted for covariates and baseline achievement of primary outcomes. Note: Formal statistical testing was inappropriate due to violation of the modelling assumptions for the following trial-related indicators: AF006, AF007; and the following non-trial related indicators: CHD005, CHD007, MH002, MH003, SMOK004, and SMOK005. Summary statistics only are presented for these indicators. Variables controlled for in the adjusted analyses were as follows: practice-level baseline list size, CCG, pre-intervention achievement against primary outcomes and total QOF score 2014–2015. CCG, clinical commissioning group; CHA2DS2-VASc, congestive heart failure, hypertension, age>75, diabetes, stroke, vascular disease, age between 65 and 74, and female sex; CHD, coronary heart disease; CI, confidence interval; CKD, chronic kidney disease; COPD, chronic obstructive pulmonary disease; PAD, peripheral arterial disease; QOF, Quality and outcomes framework; RCP, Royal College of Physicians; TIA, transient ischemic attack.

(DOCX)

S5 Table

Key sample size assumptions.

Data from an earlier work package of the ASPIRE programme were used to inform the trial sample size assumptions. Mean cluster size, cluster size coefficient of variation, ICC, and mean achievement rates were calculated using real data from practices within West Yorkshire for each primary outcome indicator. aMean achievement is the control arm achievement rate for each primary outcome indicator estimated using data available from the earlier work package. ICC, intra-cluster correlation coefficient.

(DOCX)

S6 Table

Intervention delivery across trial practices.

aOne practice in the risky prescribing arm merged with a non-ASPIRE practice in advance of the final feedback report. bOne practice in the BP arm closed in advance of the third feedback report. cOnly practices receiving an initial outreach visit were offered additional support; these practices are used as the denominator in the percentages presented. dThese granted access to the computerised searches (all arms) and prompts (risky prescribing only). BP, blood pressure.

(DOCX)

S1 Fig

Scatterplot of simulated incremental cost and QALY for the composite indicator of risky prescribing.

ICER, incremental cost-effectiveness ratio; PSA, probability sensitivity analysis; QALY, quality-adjusted life year.

(TIF)

S2 Fig

Cost-effectiveness acceptability curve for the composite indicator of risky prescribing.

QALY, quality-adjusted life year; WTP, Willingness to Pay.

(TIF)

S3 Fig

Scatterplot of simulated incremental cost and QALY for the composite indicator of BP control.

BP, blood pressure; ICER, incremental cost-effectiveness ratio; PSA, probability sensitivity analysis; QALY, quality-adjusted life year.

(TIF)

S4 Fig

Cost-effectiveness acceptability curve for the composite indicator of BP control.

BP, blood pressure; QALY, quality-adjusted life year; WTP, Willingness to Pay.

(TIF)

S1 CONSORT Checklist

(DOCX)

S1 Statistical Analysis Plan

(DOCX)

S1 Trial Protocol

(DOC)

Acknowledgments

The ASPIRE programme team comprises Susan Clamp, Rebecca Lawton, Rosie McEachan, Martin Rathfelder, Judith Richardson, Tim Stokes, Vicky Ward, Robert West, and Ian Watt, in addition to the named authors. The ASPIRE programme team can be contacted via Robbie Foy (ku.ca.sdeel@yof.r). We acknowledge the contributions of Andrew Davies, Peter Heudtlass, Chris Jackson, and John Turgoose and thank the members of the ASPIRE Programme Steering Committee for their advice and expertise. We thank Richard Neal, Suzanne Richards, and Noah Ivers for helpful comments on earlier versions of this manuscript.

Disclaimer: The views expressed are those of the authors and not necessarily those of the NIHR or the Department of Health and Social Care.

Abbreviations

ACE-Iangiotensin-converting-enzyme inhibitor
AFatrial fibrillation
ARBangiotensin receptor blocker
BPblood pressure
CCGclinical commissioning group
CHA2DS2-VASccongestive heart failure, hypertension, age ≥75, diabetes, stroke, vascular disease, age between 65 and 74, and female sex
CIconfidence interval
CKDchronic kidney disease
GPgeneral practitioner
HbA1chaemoglobin A1c
ICCintra-cluster correlation coefficient
IMDIndex of Multiple Deprivation
ITTintention-to-treat
NHSNational Health Service
NICENational Institute for Health and Care Excellence
NSAIDnonsteroidal anti-inflammatory drug
ORodds ratio
QALYquality-adjusted life year
QOFQuality Outcomes Framework
UKPDS-OMUK Prospective Diabetes Study Outcomes Model

Funding Statement

This study was funded by the National Institute for Health Research (NIHR) (Programme Grants for Applied Research [Grant Reference Number RP-PG-1209-10040], Principal Investigator = RF), https://www.nihr.ac.uk/. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Data Availability

Data cannot be shared publicly owing to a need to maintain patient confidentiality. Interested researchers may contact ku.ca.sdeel@sseccaatad-urtc to request and obtain relevant data.

References

1. Levine DM, Linder JA, Landon BE. The quality of outpatient care delivered to adults in the United States, 2002 to 2013. JAMA Intern Med. 2016;176(12): 1778–1790. 10.1001/jamainternmed.2016.6217 [Abstract] [CrossRef] [Google Scholar]
2. Cooksey R. A review of UK health research funding. London: HMSO; 2006. [Google Scholar]
3. Baird B, Charles A, Honeyman M, Maguire D, Das P. Understanding pressures in general practice. London: The King’s Fund; 2016. [Google Scholar]
4. Hobbs FDR, Bankhead C, Mukhtar T, Stevens S, Perera-Salazar R, Holt T, et al. Clinical workload in UK primary care: a retrospective analysis of 100 million consultations in England, 2007–14. Lancet. 2016;387(10035): 2323–2330. 10.1016/S0140-6736(16)00620-6 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
5. Rushforth B, Stokes T, Andrews E, Willis TA, McEachan R, Faulkner S, et al. Developing ‘high impact’ guideline-based quality indicators for UK primary care: a multi-stage consensus process. BMC Fam Pract. 2015;16(1): 156. [Europe PMC free article] [Abstract] [Google Scholar]
6. Grimshaw JM, Thomas RE, MacLennan G, Fraser C, Ramsay CR, Vale L, et al. Effectiveness and efficiency of guideline dissemination and implementation strategies. Health Technol Assess. 2004;8(6): iii–iv, 1–72. 10.3310/hta8060 [Abstract] [CrossRef] [Google Scholar]
7. Willis TA, Rushforth B, West R, Faulkner S, Stokes T, Glidewell L, et al. Variations in achievement of evidence-based, high-impact quality indicators in general practice: an observational study. PLoS ONE. 2017;12(7): e0177949 10.1371/journal.pone.0177949 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
8. National Institute for Health and Care Excellence. Type 2 diabetes in adults: management. London: Royal College of Physicians; 2015. [Google Scholar]
9. Howard RL, Avery AJ, Slavenburg S, Royal S, Pipe G, Lucassen P, et al. Which drugs cause preventable admissions to hospital? A systematic review. Brit J Clin Pharmacol. 2007;63(2): 136–147. [Europe PMC free article] [Abstract] [Google Scholar]
10. The Blood Pressure Lowering Treatment Trialists’ Collaboration. Blood pressure-lowering treatment based on cardiovascular risk: a meta-analysis of individual patient data. Lancet. 2014;384(9943): 591–598. 10.1016/S0140-6736(14)61212-5 [Abstract] [CrossRef] [Google Scholar]
11. National Institute for Health and Care Excellence. Atrial fibrillation: the management of atrial fibrillation. London: National Institute for Health and Care Excellence; 2014. [Google Scholar]
12. Ivers N, Jamtvedt G, Flottorp S, Young JM, Odgaard-Jensen J, French SD, et al. Audit and feedback: effects on professional practice and patient outcomes. Cochrane Database Syst Rev. 2012(6). Art. No.: CD000259 10.1002/14651858.CD000259.pub3 [Abstract] [CrossRef] [Google Scholar]
13. O’Brien MA, Rogers S, Jamtvedt G, Oxman AD, Odgaard-Jensen J, Kristoffersen DT, et al. Educational outreach visits: effects on professional practice and health care outcomes. Cochrane Database Syst Rev. 2007(4). Art. No.: CD000409 10.1002/14651858.CD000409.pub2 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
14. Shojania KG, Jennings A, Mayhew A, Ramsay CR, Eccles MP, Grimshaw J. The effects of on-screen, point of care computer reminders on processes and outcomes of care. Cochrane Database Syst Rev. 2009(3). Art.No.: CD001096 10.1002/14651858.CD001096.pub2 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
15. Lawton R, Heyhoe J, Louch G, Ingleson E, Glidewell L, Willis TA, et al. Using the Theoretical Domains Framework (TDF) to understand adherence to multiple evidence-based indicators in primary care: a qualitative study. Implement Sci. 2016;11(113). [Europe PMC free article] [Abstract] [Google Scholar]
16. Glidewell L, Willis TA, Petty D, Lawton R, McEachan RRC, Ingleson E, et al. To what extent can behaviour change techniques be identified within an adaptable implementation package for primary care? A prospective directed content analysis. Implement Sci. 2018;13(32). [Europe PMC free article] [Abstract] [Google Scholar]
17. Eccles M, Grimshaw J, Campbell M, Ramsay C. Research designs for studies evaluating the effectiveness of change and improvement strategies. Qual Saf Health Care. 2003;12(1): 47–52. 10.1136/qhc.12.1.47 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
18. Loudon K, Treweek S, Sullivan F, Donnan P, Thorpe KE, Zwarenstein M. The PRECIS-2 tool: designing trials that are fit for purpose. BMJ. 2015;350: h2147 10.1136/bmj.h2147 [Abstract] [CrossRef] [Google Scholar]
19. Lord PA, Willis TA, Carder P, West RM, Foy R. Optimizing primary care research participation: a comparison of three recruitment methods in data-sharing studies. Fam Pract. 2016;33(2): 200–204. 10.1093/fampra/cmw003 [Abstract] [CrossRef] [Google Scholar]
20. Willis TA, Hartley S, Glidewell L, Farrin AJ, Lawton R, McEachan RRC, et al. Action to Support Practices Implement Research Evidence (ASPIRE): protocol for a cluster-randomised evaluation of adaptable implementation packages targeting ‘high impact’ clinical practice recommendations in general practice. Implement Sci. 2016;11(1): 1–11. [Europe PMC free article] [Abstract] [Google Scholar]
21. Doran T, Fullwood C, Gravelle H, Reeves D, Kontopantelis E, Hiroeh U, et al. Pay-for-performance programs in family practices in the United Kingdom. N Engl J Med. 2006;355(4): 375–384. 10.1056/NEJMsa055505 [Abstract] [CrossRef] [Google Scholar]
22. Hayes AJ, Leal J, Gray AM, Holman RR, Clarke PM. UKPDS Outcomes Model 2: a new version of a model to simulate lifetime health outcomes of patients with type 2 diabetes mellitus using data from the 30 year United Kingdom Prospective Diabetes Study: UKPDS 82. Diabetologia. 2013;56(9): 1925–1933. 10.1007/s00125-013-2940-y [Abstract] [CrossRef] [Google Scholar]
23. Lovibond K, Jowett S, Barton P, Caulfield M, Heneghan C, Hobbs FD, et al. Cost-effectiveness of options for the diagnosis of high blood pressure in primary care: a modelling study. Lancet. 2011;378(9798): 1219–1230. 10.1016/S0140-6736(11)61184-7 [Abstract] [CrossRef] [Google Scholar]
24. Department for Communities and Local Government. English indices of deprivation 2015 [Cited 2019 Nov 12]. https://www.gov.uk/government/statistics/english-indices-of-deprivation-2015
25. Baker R, Camosso-Stefinovic J, Gillies C, Shaw EJ, Cheater F, Flottorp S, et al. Tailored interventions to address determinants of practice. Cochrane Database Syst Rev. 2015(4). Art. No.: CD005470 10.1002/14651858.CD005470.pub3 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
26. Avery AJ, Rodgers S, Cantrill JA, Armstrong S, Cresswell K, Eden M, et al. A pharmacist-led information technology intervention for medication errors (PINCER): a multicentre, cluster randomised, controlled trial and cost-effectiveness analysis. Lancet. 2012;379(9823): 1310–1319. 10.1016/S0140-6736(11)61817-5 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
27. Dreischulte T, Donnan P, Grant A, Hapca A, McCowan C, Guthrie B. Safer prescribing—a trial of education, informatics, and financial incentives. N Engl J Med. 2016;374(11): 1053–1064. 10.1056/NEJMsa1508955 [Abstract] [CrossRef] [Google Scholar]
28. Guthrie B, Kavanagh K, Robertson C, Barnett K, Treweek S, Petrie D, et al. Data feedback and behavioural change intervention to improve primary care prescribing safety (EFIPPS): multicentre, three arm, cluster randomised controlled trial. BMJ. 2016;354: i4079 10.1136/bmj.i4079 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
29. Tricco AC, Ivers NM, Grimshaw JM, Moher D, Turner L, Galipeau J, et al. Effectiveness of quality improvement strategies on the management of diabetes: a systematic review and meta-analysis. Lancet. 2012;379(9833): 2252–2261. 10.1016/S0140-6736(12)60480-2 [Abstract] [CrossRef] [Google Scholar]
30. McCambridge J, Witton J, Elbourne DR. Systematic review of the Hawthorne effect: new concepts are needed to study research participation effects. J Clin Epidemiol. 2014;67(3): 267–277. 10.1016/j.jclinepi.2013.08.015 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
31. Berwick DM. The science of improvement. JAMA. 2008;299(10): 1182–1184. 10.1001/jama.299.10.1182 [Abstract] [CrossRef] [Google Scholar]
32. Craig P, Dieppe P, Macintyre S, Michie S, Nazareth I, Petticrew M. Developing and evaluating complex interventions: the new Medical Research Council guidance. BMJ. 2008;337: a1655 10.1136/bmj.a1655 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
33. Grol R, Dalhuijsen J, Thomas S, Veld C, Rutten G, Mokkink H. Attributes of clinical guidelines that influence use of guidelines in general practice: observational study. BMJ. 1998;317(7162): 858–861. 10.1136/bmj.317.7162.858 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
34. Hallsworth M, Chadborn T, Sallis A, Sanders M, Berry D, Greaves F, et al. Provision of social norm feedback to high prescribers of antibiotics in general practice: a pragmatic national randomised controlled trial. Lancet. 2016;387(10029): 1743–1752. 10.1016/S0140-6736(16)00215-4 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
35. Foy R, Leaman B, McCrorie C, Petty D, House A, Bennett M, et al. Prescribed opioids in primary care: cross-sectional and longitudinal analyses of influence of patient and practice characteristics. BMJ Open. 2016;6(5): e010276 10.1136/bmjopen-2015-010276 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
36. Holt TA, Dalton A, Marshall T, Fay M, Qureshi N, Kirkpatrick S, et al. Automated software system to promote anticoagulation and reduce stroke risk: cluster-randomized controlled trial. Stroke. 2017;48(3): 787–790. 10.1161/STROKEAHA.116.015468 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
2020 Feb; 17(2): e1003045.
Published online 2020 Feb 28. doi: 10.1371/journal.pmed.1003045.r001

Decision Letter 0

Caitlin Moyer, Senior Editor

29 Oct 2019

Dear Dr. Willis,

Thank you very much for submitting your manuscript "An adaptable implementation package targeting evidence-based indicators in primary care: a pragmatic cluster-randomised evaluation" (PMEDICINE-D-19-02682) for consideration at PLOS Medicine.

Your paper was evaluated by a senior editor and discussed among all the editors here. It was also discussed with an academic editor with relevant expertise, and sent to three independent reviewers, including a statistical reviewer. The reviews are appended at the bottom of this email and any accompanying reviewer attachments can be seen via the link below:

[LINK]

In light of these reviews, I am afraid that we will not be able to accept the manuscript for publication in the journal in its current form, but we would like to consider a revised version that addresses the reviewers' and editors' comments. Obviously we cannot make any decision about publication until we have seen the revised manuscript and your response, and we plan to seek re-review by one or more of the reviewers.

In revising the manuscript for further consideration, your revisions should address the specific points made by each reviewer and the editors. Please also check the guidelines for revised papers at http://journals.plos.org/plosmedicine/s/revising-your-manuscript for any that apply to your paper. In your rebuttal letter you should indicate your response to the reviewers' and editors' comments, the changes you have made in the manuscript, and include either an excerpt of the revised text or the location (eg: page and line number) where each change can be found. Please submit a clean version of the paper as the main article file; a version with changes marked should be uploaded as a marked up manuscript.

In addition, we request that you upload any figures associated with your paper as individual TIF or EPS files with 300dpi resolution at resubmission; please read our figure guidelines for more information on our requirements: http://journals.plos.org/plosmedicine/s/figures. While revising your submission, please upload your figure files to the PACE digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at gro.solp@enicideMSOLP.

We expect to receive your revised manuscript by Nov 19 2019 11:59PM. Please email us (gro.solp@enicidemsolp) if you have any questions or concerns.

***Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.***

We ask every co-author listed on the manuscript to fill in a contributing author statement, making sure to declare all competing interests. If any of the co-authors have not filled in the statement, we will remind them to do so when the paper is revised. If all statements are not completed in a timely fashion this could hold up the re-review process. If new competing interests are declared later in the revision process, this may also hold up the submission. Should there be a problem getting one of your co-authors to fill in a statement we will be in contact. YOU MUST NOT ADD OR REMOVE AUTHORS UNLESS YOU HAVE ALERTED THE EDITOR HANDLING THE MANUSCRIPT TO THE CHANGE AND THEY SPECIFICALLY HAVE AGREED TO IT. You can see our competing interests policy here: http://journals.plos.org/plosmedicine/s/competing-interests.

Please use the following link to submit the revised manuscript:

https://www.editorialmanager.com/pmedicine/

Your article can be found in the "Submissions Needing Revision" folder.

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see http://journals.plos.org/plosmedicine/s/submission-guidelines#loc-methods.

Please ensure that the paper adheres to the PLOS Data Availability Policy (see http://journals.plos.org/plosmedicine/s/data-availability), which requires that all data underlying the study's findings be provided in a repository or as Supporting Information. For data residing with a third party, authors are required to provide instructions with contact information for obtaining the data. PLOS journals do not allow statements supported by "data not shown" or "unpublished results." For such statements, authors must provide supporting data or cite public sources that include it.

We look forward to receiving your revised manuscript.

Sincerely,

Caitlin Moyer, Ph.D.

Associate Editor

PLOS Medicine

plosmedicine.org

-----------------------------------------------------------

Requests from the editors:

1. Competing Interest Statement: Please clarify the role that DP, PC and SJ had in the implementation and analyses of the study, in light of their associations with NHS Bradford Districts CCG, and Prescribing Support Services, Ltd.

2. Data Availability Statement: PLOS Medicine requires that the de-identified data underlying the specific results in a published article be made available, without restrictions on access, in a public repository or as Supporting Information at the time of article publication, provided it is legal and ethical to do so. Please see the policy at

http://journals.plos.org/plosmedicine/s/data-availability

and FAQs at http://journals.plos.org/plosmedicine/s/data-availability#loc-faqs-for-data-policy.

Please include an appropriate contact (web and/or email address) for inquiries to obtain access to the data (please note: this cannot be a study author).

3. Abstract Background: The final sentence should clearly state the study question/hypothesis to be tested.

4. Abstract: Methods and Findings: Please define the control for the intervention.

5. Abstract: Methods and Findings (Line 120): You mention here that 34 practices were recruited as non-intervention controls. This could be misleading because you do not include any analyses of these controls in this manuscript, and comparisons are made to within-arm controls. Please remove the mention of these non-intervention controls here, and clarify the control for these analyses.

6. Abstract: Methods and Findings: In the last sentence of the Abstract Methods and Findings section, please describe the main limitation(s) of the study's methodology.

7. Abstract: Conclusions: Please interpret the study based on the results presented in the abstract, emphasizing what is new without overstating your conclusions; the phrase "In this study, we observed ..." may be useful. Please avoid vague statements such as "these results have major implications for policy/clinical care". Mention only specific implications substantiated by the results. Specifically, both sentences of the conclusion are vague and do not clearly reflect what can be concluded from the study findings.

8. Author Summary: Thank you for including an Author Summary. The Author Summary should immediately follow the Abstract in your revised manuscript.

9. Author Summary: “What do these findings mean?”: Please revise the final point to: “In this study, we found that a ‘one-size-fits-all’ strategy did not work…” Please also revise this point to clarify what is meant by “did not work” and “the nature of clinical behaviors targeted”.

10.Introduction: Please conclude the Introduction with a clear description of the study question or hypothesis.

11. Methods: The following points in the Methods differ from those described in the trial registry (ISRCTN registry ISRCTN91989345): In manuscript: “Audit and Feedback”, “Educational Outreach”, and “Prompts and Reminders” are described (Methods: Procedures: Lines 224-249). In the registered trial protocol, “GP Appraisal and Revalidation” and “Patient Mediated Intervention” are also described. Please explain, including in the manuscript.

12. Methods: The following points in the Methods differ from those described in the trial registry: There are a number of primary and secondary outcomes described in the manuscript (Methods: Outcomes: Lines 253-312). However, the registered protocol describes the primary outcome as: “Adherence; Timepoint(s): Adherence will be measures up to 12 mth post randomisation”, with no secondary outcomes described. Please clarify and explain the discrepancy. If the outcomes were not pre-specified in the protocol, please indicate that they were post hoc and explain why they were added. Post hoc comparisons should be presented as hypothesis generating rather than conclusive. Please explain, including in the manuscript.

13. Methods: Line 328: Please provide a link to the pre-specified statistical analysis plan referred to here, and include as supplementary information.

14. Results: Line 347: Please provide statistical evidence (in the appropriate tables) for the statement that: “Baseline characteristics for general practices and patients were well balanced by trial and intervention (Tables 1 and 2).”

15. Results: Line 375: Please provide a reference for the statement: “The mean practice IMD was 30.22 (SD 13.86), within the top quarter of UK social deprivation.”

16. Results: Lines 429-441: For the following adjusted analyses (the main analyses describing effects of diabetes, risky prescribing, BP, and AF implementation package on outcomes) please also provide the unadjusted analyses.

17. Results: Lines 445-447: For the following adjusted analyses (the analysis of the effect of risky prescribing implementation on adjusted prescribing levels) please also provide the unadjusted analyses.

18. Results: Lines 462-463: Please revise to: “Supplementary Figures 1-4 present cost-effectiveness planes and acceptability curves for the risky prescribing and blood pressure control implementation packages.” or similar.

19. Discussion: Line 541-543: Please provide an in-text reference to Supplementary Table 1 (S1 Table) for the statement that there are fewer eligible patients in the risky prescribing/anticoagulation AF arms.

20. Discussion: Please present and organize the Discussion as follows: a short, clear summary of the article's findings; what the study adds to existing research and where and why the results may differ from previous research; strengths and limitations of the study; implications and next steps for research, clinical practice, and/or public policy; one-paragraph conclusion.

21. Discussion: Thank you for including the link to the ASPIRE protocol, found at the end of the Discussion, after the Author Summary. It may be helpful to refer to any prospective study/analysis protocol in the Methods section of the manuscript. Also, the link does not seem to work. Please include the study protocol document and analysis plan, with any amendments, as Supporting Information to be published with the manuscript if accepted.

22. Figure 1: In the flow diagram, please indicate the number of individuals in each group analyzed in the ITT analysis. Please make it more clear in the diagram that the non-intervention control group (n=34) were not analyzed in the analyses being described here. This is written in the figure legend, but please include some visual indication in the diagram (text and/or shading) to make this point clear to the reader.

23. Figure 1: Please define the abbreviations: A&F (follow up).

24. Supplementary Figures 1, 2, 3, and 4 (S1, S2, S3, S4 Figures): Please define the following abbreviations: ICER, PSA, QALY, WTP.

25. Table 3, and Supplementary Tables 3, 4, 5, and 6: Please provide the unadjusted comparisons as well as the adjusted comparisons.

26. Table 3, and Supplementary Tables 3, 4, 5, and 6: Please specify the variables controlled for in the adjusted analyses in the Table legend.

27. Table 3: Please define the following abbreviations: CI, ICC.

28. Supplementary Table 1: Please clarify sources of data in support of assumptions. Please explain the “mean achievement” measure in the table legend.

29. Supplementary Table 3 :Please define the following abbreviations: CI, BP, HbA1c, ACR, PCR, eGFR, BMI, NSAID, ACE-I, ARB, CKD.

30. Supplementary Table 4: Please define the following abbreviations: CI, QOF, IFCC-HbA1c, RCP, CHD, PAD, TIA, COPD.

31. Supplementary Table 5: Please define the following abbreviations: CI, BP, HTN, CKD, CHD, PAD, TIA, CVD, AF, CHA(2)DS(2)-VASc.

32. Supplementary Table 6: Please define the following abbreviations: CI, QOF, CHA(2)DS(2)-VASc, RCP.

33. Checklist: Please complete the CONSORT checklist and ensure that all components of CONSORT are present in the manuscript. Please include the completed checklist as Supporting Information (e.g. S1 Checklist). When completing the checklist, please use section and paragraph numbers, rather than page numbers.

Comments from the reviewers:

Reviewer #1: Alex McConnachie

Willis et al report on two cluster randomised trials of implementation packages delivered to general practices to improve diabetes control, risky prescribing, blood pressure control, and anticoagulation in AF. This review looks at the use of statistics in the paper.

Overall, the trial design, statistical methods, and presentation of results are very good. My one minor point is that it appears that the primary results were judged at a significance level of 2.5%; this is not explained in the statistical methods, but is mentioned in the results. I presume the reason for this is that each trial involved the assessment of two interventions, so a 2.5% significance level was use to preserve an overall 5% Type I error rate. This should be explained in more detail in the methods section.

However, it could also be argued that the significance level should be set at 1.25%, since the trial as a whole involved testing the effect of four interventions. On that basis, the effect of the risky prescribing intervention would not be considered statistically significant. Either way, with multiple primary analyses, a clearer description of how the authors controlled the false discovery rate should be given.

Was there a statistical analysis plan? Has this been published, or should it be provided as a supplement?

Reviewer #2: This paper presents a trial that evaluates implementation science interventions for multiple evidence based indicators. I agree with the authors that this approach could be useful in the context of multiple morbidity.

However, the paper includes a number of flaws. The main ones are: the aim or hypothesis is not stated; the nature of the 'adaptation' included in the intervention is not sufficiently specified; the statistical results are not correctly interpreted; consequently, the conclusions may not be justified.

Abstract: This could be clearer. In particular the sentence on randomisation would naturally follow-on from the sentence on recruitment.

Introduction: This is clear and succinct. More could have been made of multiple morbidity. The study appears to lack a clear aim or hypothesis. The final paragraph here, explains what was done but not the purpose.

Methods: The study design could be explained more clearly. Initially, it says that this is 'two parallel cluster randomised trials using balanced incomplete block designs'. This should be explained more completely: what are the intervention and comparator trial arms in each trial. Why does this represent a balanced incomplete block design?

Later it refers to 'two stage randomisation' which suggests a single trial , the written description is not clear though Figure 1 is. Looking at Figure 1, the design is clear but the research question is not.

It appears that the no intervention control trial arm was not included in the protocol but was only added because recruitment exceeded expectations. The paper then says that the control group analysis will not be reported in this study. It may be best to focus this paper on what was in the protocol only.

The software for the minimisation could be mentioned. Also, how allocation was concealed as distinct from blinding.

The description of the intervention is generic. It is not made clear how the intervention package differed for each of the four trial arms. What are the comparisons being made? The title refers to an 'adaptable' intervention though the basis of adaptation is not mentioned in the methods.

Under outcomes it refers to 'primary outcomes for each intervention arm'. Normally there is one primary outcome for a trial. Here there are four intervention arms, but the study comprises two trials.

Multiple comparisons require additional discussion.

Missing data: initially it says that missing data could not be assessed, then it says that data were assumed missing at random. Does it mean that not recorded values were assumed to be not performed for process items? Was imputation performed for values like blood pressure? or were complete case analyses performed it is not clear.

Results:

Page 24 lines 429 and 436 and 439, where it says 'no effect', 'absence of evidence is not evidence of absence' as Doug Altman pointed out. https://www.bmj.com/content/311/7003/485

line 434, where it says 'statistically significant improvement. The paper should interpret P values following the ASA recommendations. Cut points such as P<0.05 or P<0.025 should not be used. Reporting should be based on ASA guidelines with not 'bright lines' for interpretation such as P<0.025. https://amstat.tandfonline.com/doi/full/10.1080/00031305.2016.1154108#.XVpvSuhKiUk

Similarly line 442 where it says 'no statistically significant effect'

Line 443 where it says 'significantly improved one of the indicators', was adjustment made for multiple comparisons?

In genereal insufficient details are provided for the cost-effectiveness analysis.

Line 458 'the deterministic ICER for the BP packages indicates cost-effectiveness'. How can this be so when the intervention had 'no effect' (line 436).

Discussion

Again, the paper concludes that the intervention package had no effect for three out of four indicators, but this could be an 'absence of evidence is not evidence of absence error'. Consequently, the conclusion may be too strong. Perhaps more evidence will show that an adaptive approach may be effective.

The paper arrives at the conclusion that an adaptable intervention 'does not work' but it is not made clear from the analysis plan how the various comparisons that were made could lead to such a conclusion.

Reviewer #3: This manuscript describes the findings from a large implementation trial intended to improve quality in primary care for 4 common conditions. The authors went through a careful process of selecting performance measures that matter and designing an implementation package based on the best available evidence at the time. They then implemented this in a large number of GP practices, and assessed the fidelity of the intervention. No differences were seen in their primary outcomes except a decrease in what they term "risky prescribing", meaning a patient safety issue, from 6.0% to 4.9%. A cost effectiveness analysis showed that for this one target, their intervention was cost-effective. The authors conclude that "a one size-fits-all implementation package only worked for 1 in 4 indicators".

The topic area of this manuscript - improving quality and safety in primary care - is high on the agenda for all health care systems. As someone who regularly reviews quality improvement publications as part of their scholarly activities, I can say with authority that this manuscript is a very high outlier in terms of the preparation and planning that went into the intervention and the way it is reported.

Note I am not a cost-effectiveness analyst and so can make no informed critique of their methods or results in this regard.

The results of the QI process must have come as a big disappointment to the investigators, and will be disappointing to readers as well, but these are results that have to be widely disseminated in order for ongoing and planned implementations to be changed now, to avoid continuing ineffective QI initiatives.

I have only a few comments for the authors to consider, all in the discussion sections about "Why?" The first and most obvious is already touched on briefly by the authors, namely that of their primary outcomes, the risky prescribing measure is the easiest for a GP to do. For two other conditions the performance measure is an intermediate outcome (or even a composite of several intermediate outcomes), and therefore much harder for the GP to influence. The AF indicator is a process measure but one that requires 1) that the GP calculate the CHADS-VASC score (maybe the EHR already does this automatically - if so then the manuscript should make this point) AND 2) prescribe a medication known to have a certain risk to it, plus be a pain-in-the-rear-end for patients and practices to monitor: they are signing themselves up to regular monitoring of INR, unless they are going with a new oral agent, which i suspect at the time this trial was going would have been rare in UK GP practices. As opposed to the "risky prescribing" measure, which only required someone to choose paracetamol instead of an NSAID or add omeprazole if they go with the NSAID. So, I wonder if the authors want to discuss a little more the 'easiness' of meeting this measure as opposed to the others. I understand that the process measures for diabetes - also easy things to do - didn't have significant improvements in their intervention group (although they all were a little better in the intervention vs the control), but most of them are topped out, anyway, with not a lot of room for improvement. So I think there is more-than-a-hint that these data are compatible with the conclusion that influencing a process measure is easier than influencing an outcome measure. Perhaps the authors want to expand a little on this text in the discussion, right now it is two sentences and, to me, focuses on the wrong thing- namely that BP control requires 2 visits - instead of the much-more-difficult problem of finding a pharmaceutical that is both effective and acceptable to the patient. The current text also does not deal with the AF performance measure, which like the risky prescribing measure only requires the GP to add a medication. Why isn't adding warfarin as easy as adding omeprazole? Understanding why is key to understanding why their intervention had an effect for one and did not for the other.

Secondly, I wonder whether the "patient safety" angle of the "risky prescribing" measure played a role. The other measures are all clinical quality, whereas the risky prescribing measure is a patient safety issue, making it perhaps more important in the GP's eyes.

Thirdly, I think a little more information about the EHR prompt is warranted. Have I concluded correctly that this was the only measure that had an EHR prompt included? And was the prompt a hard stop, requiring an over-ride, or a soft-stop, something easily blown through by the GP? Judging by the size of the effect, I am assuming a soft stop. These data are also consistent with a conclusion that EHR prompts work for simple actions, which the authors note, but some more information about the prompt, and why there wasn't a prompt for the warfarin measure, would help readers understand when a prompt might work (I am presuming this is because the NSAID prompt was triggered by the e-prescribing of an NSAID, which the EHR can then assess against age, concomitant cytoprotection, GFR, and concomitant warfarin, whereas the AF measures would require an alert to be triggered by something else much more complex. The authors cite another trial in this regard, but rather than make readers go look up that trial they would benefit from a sentence or two about what makes one easy and the other complex).

Lastly, there is the question of additional incentives. When practices are already being paid via QOF for doing a host of things, how much mental energy is left over to participate in something where they aren't getting more payments for doing the "thing"?

So...bottom line, an important study with important (but disappointing) findings, that in my view would benefit from a couple hundred extra words from the authors speculating about why they think some aspects did and did not work as expected.

-paul shekelle

Any attachments provided with reviews can be seen via the following link:

[LINK]

    2020 Feb; 17(2): e1003045.
    Published online 2020 Feb 28. doi: 10.1371/journal.pmed.1003045.r002

    Author response to Decision Letter 0

    29 Nov 2019

    Attachment

    Submitted filename:

      2020 Feb; 17(2): e1003045.
      Published online 2020 Feb 28. doi: 10.1371/journal.pmed.1003045.r003

      Decision Letter 1

      Caitlin Moyer, Senior Editor

      14 Jan 2020

      Dear Dr. Willis,

      Thank you very much for re-submitting your manuscript "An adaptable implementation package targeting evidence-based indicators in primary care: a pragmatic cluster-randomised evaluation" (PMEDICINE-D-19-02682R1) for review by PLOS Medicine.

      I have discussed the paper with my colleagues and the academic editor and it was also seen again by 3 reviewers. I am pleased to say that provided the remaining editorial and production issues are dealt with we are planning to accept the paper for publication in the journal.

      The remaining issues that need to be addressed are listed at the end of this email. Any accompanying reviewer attachments can be seen via the link below. Please take these into account before resubmitting your manuscript:

      [LINK]

      Our publications team (gro.solp@enicidemsolp) will be in touch shortly about the production requirements for your paper, and the link and deadline for resubmission. DO NOT RESUBMIT BEFORE YOU'VE RECEIVED THE PRODUCTION REQUIREMENTS.

      ***Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.***

      In revising the manuscript for further consideration here, please ensure you address the specific points made by each reviewer and the editors. In your rebuttal letter you should indicate your response to the reviewers' and editors' comments and the changes you have made in the manuscript. Please submit a clean version of the paper as the main article file. A version with changes marked must also be uploaded as a marked up manuscript file.

      Please also check the guidelines for revised papers at http://journals.plos.org/plosmedicine/s/revising-your-manuscript for any that apply to your paper. If you haven't already, we ask that you provide a short, non-technical Author Summary of your research to make findings accessible to a wide audience that includes both scientists and non-scientists. The Author Summary should immediately follow the Abstract in your revised manuscript. This text is subject to editorial change and should be distinct from the scientific abstract.

      We expect to receive your revised manuscript within 1 week. Please email us (gro.solp@enicidemsolp) if you have any questions or concerns.

      We ask every co-author listed on the manuscript to fill in a contributing author statement. If any of the co-authors have not filled in the statement, we will remind them to do so when the paper is revised. If all statements are not completed in a timely fashion this could hold up the re-review process. Should there be a problem getting one of your co-authors to fill in a statement we will be in contact. YOU MUST NOT ADD OR REMOVE AUTHORS UNLESS YOU HAVE ALERTED THE EDITOR HANDLING THE MANUSCRIPT TO THE CHANGE AND THEY SPECIFICALLY HAVE AGREED TO IT.

      Please ensure that the paper adheres to the PLOS Data Availability Policy (see http://journals.plos.org/plosmedicine/s/data-availability), which requires that all data underlying the study's findings be provided in a repository or as Supporting Information. For data residing with a third party, authors are required to provide instructions with contact information for obtaining the data. PLOS journals do not allow statements supported by "data not shown" or "unpublished results." For such statements, authors must provide supporting data or cite public sources that include it.

      If you have any questions in the meantime, please contact me or the journal staff on gro.solp@enicidemsolp.

      We look forward to receiving the revised manuscript by Jan 21 2020 11:59PM.

      Sincerely,

      Caitlin Moyer, Ph.D.

      Associate Editor

      PLOS Medicine

      plosmedicine.org

      ------------------------------------------------------------

      Requests from Editors:

      1. Competing Interest Statement: Thank you for clarifying the roles of the authors in the updated competing interest section in your manuscript. Please update the competing interest statement on the manuscript submission form.

      2. Data Availability Statement: Thank you for providing the contact information for access to your study data. We suggest you shorten the Data Availability Statement to: “Data cannot be shared publicly owing to a need to main patient confidentiality. Interested researchers may contact ku.ca.sdeel@sseccaatad-urtc to request and obtain relevant data.” Please update this in the manuscript submission form with your revised submission.

      3. Throughout: Abstract Lines 104 and 112, Introduction Line 198, and Methods Line 239: Please replace the term “usual practice” with “implementation control”, as your control comparison condition is not accurately described as usual practice.

      4. Throughout:Trademark (™) symbol: Please remove the trademark symbol throughout the manuscript (Lines 218, 309, 363, 367, 396).

      5. Abstract: Line 104-105: In the final sentence, please clarify the nature of the indicators (indicators of what?)

      6. Abstract: Line 108-109: Please revise this sentence to: “We used ‘opt-out’ recruitment, and practices that did not opt out were randomly assigned to to an implementation package…”

      7. Abstract: Line 123-124: Please describe the confounds for which you adjusted.

      8. Abstract: Line 127: Rather than saying that the implementation package “had no effect on other primary endpoints”, please provide the adjusted ORs ( with CIs and p values) for all four primary comparisons from Table 3.

      9. Abstract: Conclusions: Your conclusion only touches on the cost-effectiveness of the intervention. Can you please include your main conclusions regarding the primary endpoint (i.e. adherence is your registered primary endpoint, and the description above in the abstract describes specific patient-level primary endpoints that could be discussed). At Line 135, please avoid using the term “relatively cost-effective”; instead, please use quantitative terms to describe cost-effectiveness.

      10. Author Summary: “What did the researchers do and find?”: Please remove the word “highly” from the first bullet point. Also, please clarify if you are reporting results of one trial or two trials, and mention the outcomes being assessed.

      11. Author Summary: “What do these findings mean?”: In the first point, we suggest you also mention your conclusion regarding the primary outcome of the study, in addition to mentioning the cost-effectiveness outcome.

      12. Methods: Please add the following statement to the Methods section: “This study is reported as per the Consolidated Standards of Reporting Trials (CONSORT) guideline for cluster randomized trials (S1 Checklist).”

      13. Discussion: Lines 532-533: Please clarify the second part of this sentence, we suggest: “...which could ultimately be associated with changes in mortality, morbidity and health service use, depending on generalizability in the general population.” or similar.

      14. Discussion: Lines 550-551: Please clarify this sentence: “The risky prescribing package cost of £0.28 per patient is inexpensive.” Inexpensive relative to what?

      15. Discussion: Line 558: Please remove the word “highly” from the sentence.

      16. References: Please double check that all references are formatted with the "Vancouver" style for reference formatting, and see our website for other reference guidelines https://journals.plos.org/plosmedicine/s/submission-guidelines#loc-references

      For example, reference 7 should be “PLoS ONE”, ref 18, 28, 32, are not complete/have extraneous text; ref 21, 27, should be N Engl J Med.

      17. Link to Trial Protocol: Line 649-650: Thank you for including the trial protocol with your supporting information. The Supporting Information will be available online with the manuscript. As you have included the protocol document, you can remove the extra link here.

      18. Table 2: Please check the “a” “b” “c” superscripts in the table footnote, there appears to be some discrepancy.

      19. Supplementary Figure 2 and 4: Please integrate the “thousands” label for the X axis units with the X axis legend.

      20. Checklist: Thank you for including your CONSORT checklist. For Item 1a, please change the location to “Title” and for item 1b, please change the location to “Abstract”. For item 6b, you could direct the reader to where you discuss changing your primary outcome assessment timepoint to 11 months rather than 12 months. For item 24, please reference your trial protocol included in the supporting information (e.g. S1 text). For item 25, please reference “Funding” as the section for this information.

      Comments from Reviewers:

      Reviewer #1: Having reread the paper, and the authors' responses to my and the other reviewers' comments, I am happy with the changes that have been made. I have no further comments to make.

      Reviewer #2: The revision has generally responded to the review comments.

      In the Abstract where it reads: 'The implementation package reduced risky prescribing (odds ratio 0.82; 97.5% confidence interval (CI) 0.67 to 126 0.99, p = 0.017) with an incremental cost effectiveness ratio of £1,359 per quality-adjusted life year but had no effect on other primary endpoints. No statistically significant effects were observed in any secondary outcome except for reduced co-prescription of aspirin and clopidogrel without gastro-protection in patient aged 65 and over (adjusted OR 0.62; 97.5% 130 CI 0.39 to 0.99; p = 0.021). '

      It appears that the abstract is reporting only those results that are associated with P values less than 0.025, whereas the other outcomes are referred to as having 'no significant effects'. It would be preferable to report each of the primary outcomes in the Abstract.

      The ASA guidelines on P values make the recommendation that 'Scientific conclusions and business or policy decisions should not be based only on whether a p-value passes a specific threshold.' The authors justify their approach with respect to the analysis plan, but it would be preferable to follow the ASA recommendation. Rather than saying there was 'no effect', there was 'insufficient evidence of effect', I expect the authors do not want to add to the catalogue of 'absence of evidence is not evidence of absence' errors.

      Reviewer #3: I don't have any remaining issues with this manuscript and would proceed with accepting it.

      Any attachments provided with reviews can be seen via the following link:

      [LINK]

        2020 Feb; 17(2): e1003045.
        Published online 2020 Feb 28. doi: 10.1371/journal.pmed.1003045.r004

        Author response to Decision Letter 1

        29 Jan 2020

        Attachment

        Submitted filename:

          2020 Feb; 17(2): e1003045.
          Published online 2020 Feb 28. doi: 10.1371/journal.pmed.1003045.r005

          Decision Letter 2

          Caitlin Moyer, Senior Editor

          31 Jan 2020

          Dear Dr Willis,

          On behalf of my colleagues and the academic editor, Dr. Sanjay Basu, I am delighted to inform you that your manuscript entitled "An adaptable implementation package targeting evidence-based indicators in primary care: a pragmatic cluster-randomised evaluation" (PMEDICINE-D-19-02682R2) has been accepted for publication in PLOS Medicine.

          PRODUCTION PROCESS

          Before publication you will see the copyedited word document (in around 1-2 weeks from now) and a PDF galley proof shortly after that. The copyeditor will be in touch shortly before sending you the copyedited Word document. We will make some revisions at the copyediting stage to conform to our general style, and for clarification. When you receive this version you should check and revise it very carefully, including figures, tables, references, and supporting information, because corrections at the next stage (proofs) will be strictly limited to (1) errors in author names or affiliations, (2) errors of scientific fact that would cause misunderstandings to readers, and (3) printer's (introduced) errors.

          If you are likely to be away when either this document or the proof is sent, please ensure we have contact information of a second person, as we will need you to respond quickly at each point.

          PRESS

          A selection of our articles each week are press released by the journal. You will be contacted nearer the time if we are press releasing your article in order to approve the content and check the contact information for journalists is correct. If your institution or institutions have a press office, please notify them about your upcoming paper at this point, to enable them to help maximize its impact.

          PROFILE INFORMATION

          Now that your manuscript has been accepted, please log into EM and update your profile. Go to https://www.editorialmanager.com/pmedicine, log in, and click on the "Update My Information" link at the top of the page. Please update your user information to ensure an efficient production and billing process.

          Thank you again for submitting the manuscript to PLOS Medicine. We look forward to publishing it.

          Best wishes,

          Caitlin Moyer, Ph.D.

          Associate Editor

          PLOS Medicine

          plosmedicine.org


            Articles from PLoS Medicine are provided here courtesy of Public Library of Science

            Citations & impact 


            Impact metrics

            Jump to Citations

            Alternative metrics

            Altmetric
            Discover the attention surrounding your research
            https://www.altmetric.com/details/76812235

            Smart citations by scite.ai
            Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
            Explore citation contexts and check if this article has been supported or disputed.
            https://scite.ai/reports/10.1371/journal.pmed.1003045

            Supporting
            Mentioning
            Disputing
            0
            1
            0

            Article citations

            Data 


            Data behind the article

            This data has been text mined from the article, or deposited into data resources.

            Similar Articles 


            To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


            Funding 


            Funders who supported this work.

            Department of Health (1)

            This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.