Europe PMC
Do data resources managed by EMBL-EBI and our collaborators make a difference to your work?
If so, please take 10 minutes to fill in our survey, and help us make the case for why sustaining open data resources is critical for life sciences research.

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.

Abstract 


Background

Intensive treatment of multiple cardiovascular risk factors can halve mortality among people with established type 2 diabetes. We investigated the effect of early multifactorial treatment after diagnosis by screening.

Methods

In a pragmatic, cluster-randomised, parallel-group trial done in Denmark, the Netherlands, and the UK, 343 general practices were randomly assigned screening of registered patients aged 40-69 years without known diabetes followed by routine care of diabetes or screening followed by intensive treatment of multiple risk factors. The primary endpoint was first cardiovascular event, including cardiovascular mortality and morbidity, revascularisation, and non-traumatic amputation within 5 years. Patients and staff assessing outcomes were unaware of the practice's study group assignment. Analysis was done by intention to treat. This study is registered with ClinicalTrials.gov, number NCT00237549.

Findings

Primary endpoint data were available for 3055 (99·9%) of 3057 screen-detected patients. The mean age was 60·3 (SD 6·9) years and the mean duration of follow-up was 5·3 (SD 1·6) years. Improvements in cardiovascular risk factors (HbA(1c) and cholesterol concentrations and blood pressure) were slightly but significantly better in the intensive treatment group. The incidence of first cardiovascular event was 7·2% (13·5 per 1000 person-years) in the intensive treatment group and 8·5% (15·9 per 1000 person-years) in the routine care group (hazard ratio 0·83, 95% CI 0·65-1·05), and of all-cause mortality 6·2% (11·6 per 1000 person-years) and 6·7% (12·5 per 1000 person-years; 0·91, 0·69-1·21), respectively.

Interpretation

An intervention to promote early intensive management of patients with type 2 diabetes was associated with a small, non-significant reduction in the incidence of cardiovascular events and death.

Funding

National Health Service Denmark, Danish Council for Strategic Research, Danish Research Foundation for General Practice, Danish Centre for Evaluation and Health Technology Assessment, Danish National Board of Health, Danish Medical Research Council, Aarhus University Research Foundation, Wellcome Trust, UK Medical Research Council, UK NIHR Health Technology Assessment Programme, UK National Health Service R&D, UK National Institute for Health Research, Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, Novo Nordisk, Astra, Pfizer, GlaxoSmithKline, Servier, HemoCue, Merck.

Free full text 


Lancet. 2011 Jul 9; 378(9786): 156–167.
PMCID: PMC3136726
PMID: 21705063

Effect of early intensive multifactorial therapy on 5-year cardiovascular outcomes in individuals with type 2 diabetes detected by screening (ADDITION-Europe): a cluster-randomised trial

Simon J Griffin, Dr, DM,a,* Knut Borch-Johnsen, Prof, DMSc,b,c,d Melanie J Davies, Prof, MD,e Kamlesh Khunti, Prof, MD,f Guy EHM Rutten, PhD,g Annelli Sandbæk, Prof, PhD,d Stephen J Sharp, MSc,a Rebecca K Simmons, PhD,a Maureen van den Donk, PhD,g Nicholas J Wareham, Prof, PhD,a and Torsten Lauritzen, Prof, DMScd

Associated Data

Supplementary Materials

Summary

Background

Intensive treatment of multiple cardiovascular risk factors can halve mortality among people with established type 2 diabetes. We investigated the effect of early multifactorial treatment after diagnosis by screening.

Methods

In a pragmatic, cluster-randomised, parallel-group trial done in Denmark, the Netherlands, and the UK, 343 general practices were randomly assigned screening of registered patients aged 40–69 years without known diabetes followed by routine care of diabetes or screening followed by intensive treatment of multiple risk factors. The primary endpoint was first cardiovascular event, including cardiovascular mortality and morbidity, revascularisation, and non-traumatic amputation within 5 years. Patients and staff assessing outcomes were unaware of the practice's study group assignment. Analysis was done by intention to treat. This study is registered with ClinicalTrials.gov, number NCT00237549.

Findings

Primary endpoint data were available for 3055 (99·9%) of 3057 screen-detected patients. The mean age was 60·3 (SD 6·9) years and the mean duration of follow-up was 5·3 (SD 1·6) years. Improvements in cardiovascular risk factors (HbA1c and cholesterol concentrations and blood pressure) were slightly but significantly better in the intensive treatment group. The incidence of first cardiovascular event was 7·2% (13·5 per 1000 person-years) in the intensive treatment group and 8·5% (15·9 per 1000 person-years) in the routine care group (hazard ratio 0·83, 95% CI 0·65–1·05), and of all-cause mortality 6·2% (11·6 per 1000 person-years) and 6·7% (12·5 per 1000 person-years; 0·91, 0·69–1·21), respectively.

Interpretation

An intervention to promote early intensive management of patients with type 2 diabetes was associated with a small, non-significant reduction in the incidence of cardiovascular events and death.

Funding

National Health Service Denmark, Danish Council for Strategic Research, Danish Research Foundation for General Practice, Danish Centre for Evaluation and Health Technology Assessment, Danish National Board of Health, Danish Medical Research Council, Aarhus University Research Foundation, Wellcome Trust, UK Medical Research Council, UK NIHR Health Technology Assessment Programme, UK National Health Service R&D, UK National Institute for Health Research, Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, Novo Nordisk, Astra, Pfizer, GlaxoSmithKline, Servier, HemoCue, Merck.

Introduction

Type 2 diabetes mellitus is common, expensive to manage, and associated with a substantial burden of morbidity and mortality, particularly owing to cardiovascular complications.1 Risk of cardiovascular events and death can be halved among patients with longstanding diabetes and microalbuminuria by intensive multifactorial treatment.2,3 Treatment of individual risk factors, such as blood pressure,4,5 cholesterol,6 and glucose,7 is effective. Outcomes might be improved if this approach were used early in the course of the disease.8 The effect of starting multifactorial treatment from the time of diagnosis is unknown.

Type 2 diabetes is detectable well before it is clinically diagnosed9 and many patients already have evidence of diabetic complications and potentially modifiable cardiovascular risk factors at the time of diagnosis.10 Early detection by screening is not associated with harmful psychological effects11 and, therefore, diabetes meets many suitability criteria for screening.12 Modelling studies have indicated that screening would be an efficient use of resources,13 but there are several critical uncertainties that have prevented its routine widespread implementation.14 No evidence from trials is available to show whether early intensive multifactorial treatment improves outcomes when started between detection by screening and clinical diagnosis. We did the multicentre Anglo-Danish-Dutch Study of Intensive Treatment In People with Screen Detected Diabetes in Primary Care (ADDITION-Europe) to investigate this issue.

Methods

Design

The study design and rationale have been reported.15–18 Briefly, ADDITION-Europe consisted of two phases: a screening phase and a pragmatic, cluster-randomised, parallel-group trial in Denmark, the Netherlands, and the UK (in Cambridge and Leicester). The study was approved by the ethics committee local to each study centre. All participating patients provided informed consent.

General practices in the four study areas within a maximum of 100 miles of the study centres were invited to participate according to previously reported inclusion criteria.17–20 Between April, 2001, and December, 2006, eligible practices undertook population-based, stepwise screening of registered patients aged 40–69 years (50–69 years in the Netherlands) without known diabetes.17–20 Screening involved calculation of a risk score from information in general practice medical records in Cambridge or calculation of a risk score from self-completed questionnaires in Denmark and the Netherlands, followed by capillary glucose testing, or by invitation to attend for an oral glucose tolerance test without previous risk assessment in Leicester.

Individuals at risk were assessed in general practice, and those diagnosed as having type 2 diabetes according to WHO criteria,21 including the requirement for confirmatory testing on a separate occasion, were included in the study. Exclusion criteria for patients were assessed by family physicians. They were illness with a life expectancy of less than 12 months or psychological or psychiatric disorders that might invalidate informed consent, or being housebound or pregnant, or lactation.

Randomisation

The practices were randomly assigned to provide routine diabetes care or intensive multifactorial treatment in a 1:1 ratio, by statisticians in each centre, according to a computer-generated list, independent of measurement teams.

In Denmark, randomisation included stratification by county and number of full-time family physicians. In the Netherlands, practices were stratified by single-handed or group status. In the UK, randomisation included minimisation for the local district hospital, the number of patients per practice with diabetes in Cambridge, and for practice demographics, deprivation status, and prevalence of type 2 diabetes in Leicester. Patients were unaware of their general practice's group assignment throughout the study.

Intervention

The characteristics of the intensive treatment intervention, which involved the addition of several features to existing diabetes care, have been described previously,15–18 and the methods used to educate and support staff in providing intensive treatment are summarised in table 1 and on the study website. Target-driven and guideline-driven management of hyperglycaemia, blood pressure, and cholesterol levels by medical treatment and promotion of healthy lifestyles, based on the stepwise regimen used in the Steno-2 study2 and other trial data obtained from people with type 2 diabetes,4–6,26 was added to routine care (table 2). The same approach was used across all centres, although final decisions about prescriptions, including choice of individual drugs, were made by family physicians and patients.

Table 1

Characteristics of screening programmes, intervention delivery, and outcome ascertainment, by study centre

Screening programmeIntervention deliveryOutcome ascertainment
Cambridge, UKElectronic medical records of patients aged 40–69 years searched for routinely collected information to enable calculation of Cambridge diabetes risk scores22
Individuals with scores ≥0·17 invited by their family physcian to participate in a stepwise screening programme, including capillary RBG, FBG, and HbA1c tests
Practice-based educational meetings held with family physicians and nurses to discuss treatment targets, algorithms, and lifestyle advice
Audit and feedback via follow-up practice-based meetings up to twice per year
Practice staff provided with educational materials for patients
Small financial incentives given to family physicians*
Participants tagged for mortality with the Office for National Statistics
Sensitive electronic searches of general practice records undertaken between March, 2009, and February, 2010, in which possible events highlighted and copies made of medical records
Additional information obtained from hospital medical records and coroners' offices, as required
DenmarkPatients aged 40–69 years sent letters that included questions from the Danish Diabetes Risk Score Questionnaire23 and advised recipients with scores ≥5 (high risk) to arrange an appointment with family physician for assessment, including RBG, FBG, and HbA1c tests, or patients attending family physician practice asked to complete risk score questionnaire and those with scores ≥5 underwent blood glucose testsSmall group or practice-based educational meetings with family physicians and nurses to discuss treatment targets, algorithms, and lifestyle advice
Audit and feedback included in follow-up group meetings up to twice per year or coordinated by post
Practice staff provided with educational materials for patients
Small financial incentives given to family physicians*
Patients sent reminders if annual assessments overdue
National patient register searched on Dec 31, 2009, for deaths, ICD-10 codes for cardiovascular events (I08–77), and surgical amputations and revascularisations.
For possible events information obtained from hospital medical records and coroners' offices, as required
Leicester, UKAll patients aged 40–69 years invited directly for an OGTT in a local testing facilityPatients referred to the DESMOND structured education programme24
Patients offered appointments every 2 months with a diabetes nurse or family physician, in a community clinic, for 1 year, and every 4 months thereafter
Clinic staff prompted to contact patients who missed appointments
Small financial incentives given to family physicians*
Participants tagged for mortality with the Office for National Statistics
Sensitive electronic searches of general practice records undertaken between March, 2009, and February, 2010, in which possible events highlighted and copies made of medical records
Additional information obtained from hospital medical records and coroners' offices, as required
NetherlandsPatients aged 50–69 years sent letters from family physicians that included the Hoorn study Symptom Risk Questionnaire25 and advised recipients with scores ≥4 (41 practices closest to study centre) or ≥6 (38 practices furthest from study centre) to arrange an appointment with their family physician for assessment
Attending patients assessed with RGB, FBG, and OGTT (41 practices closest to study centre) or a FBG and OGTT (38 practices furthest from study centre)
Small group or practice-based educational meetings with family physicians and nurses to discuss treatment targets, algorithms, and lifestyle advice
Audit and feedback included in follow-up meetings up to twice per year or coordinated by post
Patients seen by diabetes nurses authorised to prescribe medication and adjust doses under supervision by family physicians
Patients sent reminders if annual assessments overdue
Small financial incentives given to family physicians*
General practice records searched manually and extracted endpoint and vital status information recorded on standard forms
For patients who had moved practice, endpoint data obtained by telephone interview with current family physician

RBG=random blood glucose. FBG=fasting blood glucose. HbA1c=glycated haemoglobin A1c. ICD-10=International Classification of Diseases, version ten. OGTT=oral glucose tolerance test. DESMOND=diabetes education and self management for ongoing and newly diagnosed programme.

*Payment up to equivalent of three 10 min consultations with a family physician and three 15 min consultations wit a nurse, per patient, per year, for 3 years.

Table 2

Treatment recommendations and targets in the intensive treatment group

Treatment targetTreatment thresholdApproach at baseline if threshold passedAction at review
Review 1Review 2Review 3
HbA1c<7·0%>6·5%DietValue >threshold, prescribe metforminValue >threshold, increase metformin dose or add a PGR, sulphonylurea, or TZDValue >threshold, add second or third medication (PGR or sulphonurea, or TZD) and consider adding insulin
Blood pressure≤135/85 mm Hg*≥120/80 mm HgIf CVD+, prescribe an ACE inhibitor titrated to maximum doseValue >target, add thiazide diuretic or calcium-channel blockerValue >target, add calcium-channel blocker or thiazide diureticValue >target, add β blocker or α blocker
Total cholesterol level without IHD<5·0 mmol/L≥3·5 mmol/LPrescribe a statinValue >target, increase statin dose up to maximumValue >target, increase statin dose up to maximumValue >target, consider adding a fibrate
Cholesterol level with IHD<4·5 mmol/L≥3·5 mmol/LPrescribe a statinValue >target, increase statin up to maximum doseValue >target, continue statin titration if maximum dose not reachedValue >target, consider adding a fibrate
AspirinNoneNone75–80 mg daily75–80 mg daily75–80 mg daily75–80 mg daily

HbA1c=glycated haemoglobin A1c. PGR=prandial glucose regulator. TZD=thiazolidinedione. ACE=angiotensin-converting enzyme. IHD ischaemic heart disease, CVD+=cardiovascular event or presence of cardiovascular risk factor other than diabetes. BP=blood pressure.

*≤130/80 mm Hg in Leicester.
All patients receiving antihypertensive medication and without specific contraindications.

Intensive treatment was promoted to the participating general practices by small group or practice-based educational meetings attended by family physicians and nurses to discuss the treatment targets and algorithms and lifestyle advice and provide supporting evidence. Audit and feedback were done in follow-up meetings held up to twice per year or were coordinated by post. All participating practices received additional funding to support the delivery of care (up to the equivalent of three 10 min consultations with a family physician and three 15 min consultations with a nurse, per patient, per year for 3 years).

Family physicians were advised to consider prescribing angiotensin-converting-enzyme inhibitors for patients who had blood pressure of 120/80 mm Hg or higher, a cardiovascular risk factor other than diabetes, or who had had a previous cardiovascular event,5 and 75 mg aspirin daily for patients who had no specific contraindications. After publication of the Heart Protection Study27 we altered the treatment algorithm to recommend prescription of a statin to all patients with a cholesterol concentration of 3·5 mmol/L or higher at any time.

For the routine care group, family physicians were only provided with diagnostic test results. Patients received standard diabetes care, according to the recommendations applicable in each centre.28–30

Outcomes

The primary endpoint was a composite of first cardiovascular event, including cardiovascular mortality, cardiovascular morbidity (non-fatal myocardial infarction and non-fatal stroke), revascularisation, and non-traumatic amputation. The secondary outcomes were the individual components of the primary endpoint and all-cause mortality. Centrally trained staff assessed patients' health at baseline and after 5 years by collection of data on biochemical and anthropometric features and use of questionnaires to assess activities, including use of medication, according to standard operating procedures (see webappendix p 1). Additionally, information on prescribed medication, and data potentially linked to the endpoints were obtained from general practice records or national registers. All staff collecting data from records and undertaking health assessments were unaware of patients' treatment group allocations. For each endpoint possibly met, the relevant clinical information (eg, death certificates, post-mortem reports, medical records, hospital discharge summaries, electrocardiographs, laboratory results, etc) were sent to two members of the expert committee within the country for independent adjudication according to an agreed protocol. Committee members were unaware of treatment allocations. Outcomes were recorded on standard case report forms. Committee members met to reach consensus over discrepancies.

The date of completion of follow-up for the primary endpoint was deemed to be the date of the first primary endpoint event, the 5-year assessment if no event occurred, or the date that the endpoint search was undertaken if the participant did not experience an event or attend follow-up.

Statistical analysis

The analysis plan was finalised before preparation of the endpoint dataset, and analysis was by intention to treat. We summarised baseline characteristics of individuals and general practices within each randomised group, by centre, and overall. We plotted the cumulative probability of the primary endpoint. To assess intervention effects we used Cox's regression to estimate hazard ratios (HRs) and 95% CI within each centre. In Leicester there were few participants and events and, therefore, we combined the endpoint data with those for Cambridge. In view of randomisation being at the practice level, we calculated robust SE that took into account the two-level structure of the data and any potential correlation between the individuals within practices. We calculated the correlation coefficients within practices for the primary endpoint. Centre-specific log HRs and SE were combined with a fixed effects meta-analysis, and we calculated the I2 statistic to represent the proportion of variability (in log HRs) between centres owing to heterogeneity. We tested the proportional hazards assumption by including a parameter for treatment by time interaction in each centre-specific Cox's regression model.

Continuous intermediate endpoints were analysed within each centre with normal errors regression, with adjustment for the endpoint baseline values; individuals who died or who were lost to follow-up were excluded from this analysis. We calculated robust SE. We combined the estimated differences in mean change from baseline across centres with fixed effects meta-analysis. In the regression models, we included individuals with missing outcome values at baseline, according to the missing indicator method,31 and variables with a skewed distribution were log transformed. We estimated the effect of the intervention on prescribing endpoints within each centre by use of logistic regression, and combined the estimated odds ratios across centres with fixed effects meta-analysis. We undertook sensitivity analyses by excluding follow-up clinical data obtained from general practice records, and did prespecified subgroup analyses for the primary endpoint by including interaction terms between intervention group, patient's age, and self-reported history of cardiovascular disease, which were then pooled across centres. The cumulative incidence of the composite cardiovascular endpoint was calculated with the method for competing risks described by Gooley and colleagues,32 the competing events here being the primary endpoint and death from non-cardiovascular causes. For total mortality, Kaplan-Meier estimates of cumulative incidence were calculated. Analyses were done with STATA (version 11).

We calculated that a patient-level randomised trial would have required enrolment of 2700 individuals (1350 per treatment group) to detect a 30% reduction in the risk of the primary endpoint at a 5% significance level, and with 90% power. This calculation allowed for 10% loss to follow-up and assumed an event rate in the routine care group of 3% per year, on the basis of the results of the UK Prospective Diabetes Study Group (UKPDS).26 We expected a minimum effect of clustering within general practice, with the estimated within-cluster correlation coefficient being 0·01. We assumed that the average number of participants per general practice would be 10 and, therefore, the design effect was 1·09. Thus, we inflated the estimated sample size for this cluster trial to 3000 patients in total.

Role of the funding sources

The sponsors of the study had no role in study design, data collection, data analysis, data interpretation, or writing of the report. The corresponding author had full access to all the data in the study and had final responsibility for the decision to submit for publication.

Results

Of 1312 general practices invited, 379 (29%) agreed to participate in the study. 36 (9%) of these were excluded and 343 (91%) were randomised to provide routine diabetes care (n=176) or intensive multifactorial treatment (n=167; figure 1), of which 327 (routine care n=165, intensive treatment n=162) completed screening and 318 (routine care n=157, intensive treatment n=161) included eligible patients. Participating practices in the UK and the Netherlands have been described.17,18,20 The mean patient list size was 7378 in the routine care group and 7160 in the intensive treatment group. The median prevalence of known diabetes was 3·5%. Data for the prevalence of diabetes in the Denmark practices were not available.

Screening identified 3233 patients with type 2 diabetes, of whom 3057 agreed to take part (Denmark, n=1533; Cambridge, UK, n=867; Netherlands, n=498; and Leicester, UK, n=159). The characteristics of these patients did not differ significantly from those of the 176 patients who were eligible but chose not to participate. Baseline demographic, clinical, biochemical, and treatment characteristics of patients in the two treatment groups were well matched overall (table 3). In Denmark, however, more patients were identified in practices assigned intensive treatment than in those assigned routine care (910 vs 623), and more patients in the former group had a history of ischaemic heart disease (International Classification of Diseases [version 10; ICD-10] codes I20–25, 102 [11·2%] vs 53 [8·5%]) or other cardiac diagnosis (ICD10 codes I30–52, 76 [8·4%] vs 28 [4·5%]). As previously described, patients overall exhibited high levels of untreated cardiovascular risk factors at diagnosis.19

Table 3

Clinical, biochemical, and treatment characteristics of patients at baseline and mean follow-up of 5·3 years

Routine care
Intensive treatment
Change from baseline to follow-up (β/OR [95% CI])
Baseline (n=1379)
Follow-up (n=1285)
Baseline (n=1678)
Follow up (n=1574)
Total with data available (%)ValueTotal with data available (%)ValueTotal with data available (%)ValueTotal with data available (%)Value
Demographic variables
Male sex1379 (100%)790 (57·3%)NANA1678 (100%)981 (58·5%)NANANA
Mean (SD) age at diagnosis (years)1379 (100%)60·2 (6·8)NANA1678 (100%)60·3 (6·9)NANANA
White ethnic origin1334 (96·7%)1246 (93·4%)NANA1607 (95·8%)1539 (95·8%)NANANA
Employed1013 (73·5%)425 (42·0%)NANA1197 (71·3%)482 (40·3%)NANANA
Clinical variables
History of myocardial infarction1286 (93·3%)79 (6·1%)NANA1593 (94·9%)109 (6·8%)NANA..
History of stroke1270 (92·1%)24 (1·9%)NANA1558 (92·8%)45 (2·9%)NANA..
Current smoker1347 (97·7%)375 (27·8%)1014 (78·9%)187 (18·4%)1649 (98·3%)444 (26·9%)1293 (82·1%)261 (20·2%)1·06 (0·77 to 1·45)
Median (IQR) units of alcohol per week1183 (85·8%)4 (1–13)1010 (78·6%)3 (0–10)1492 (88·9%)4 (1–13)1280 (81·3%)3 (0–10)−0·24 (−0·77 to 0·29)
Mean (SD) BMI (kg/m2)1342 (97·3%)31·6 (5·6)1112 (86·5%)31·0 (5·6)1615 (96·2%)31·6 (5·6)1405 (89·3%)31·1 (5·7)0·03 (−0·17 to 0·22)
Mean (SD) weight (kg)1344 (97·5%)90·3 (17·6)1192 (92·8%)88·4 (17·8)1615 (96·2%)90·9 (17·5)1490 (94·7%)89·1 (18·2)−0·02 (−0·58 to 0·55)
Mean (SD) waist circumference (cm)1346 (97·6%)106·8 (13·5)1064 (82·8%)105·3 (13·6)1612 (96·1%)107·1 (13·5)1363 (86·6%)105·4 (13·6)−0·30 (−1·01 to 0·42)
Median (IQR) HbA1c (%)1298 (94·1%)6·6 (6·1–7·3)1226 (95·4%)6·5 (6·1–7·1)1591 (94·8%)6·5 (6·1–7·3)1513 (96·1%)6·4 (6·0–6·9)−0·08 (−0·14 to −0·02)
Mean (SD) HbA1c (%)1298 (94·1%)7·0 (1·5)1226 (95·4%)6·7 (0·95)1591 (94·8%)7·0 (1·6)1513 (96·1%)6·6 (0·95)−0·08 (−0·14 to −0·02)
Mean (SD) systolic blood pressure (mm Hg)1346 (97·6%)149·8 (21·3)1205 (93·8%)138·1 (17·6)1617 (96·4%)148·5 (22·1)1517 (96·4%)134·8 (16·8)−2·86 (−4·51 to −1·20)
Mean (SD) diastolic blood pressure (mm Hg)1346 (97·6%)86·5 (11·3)1203 (93·6%)80·7 (10·8)1618 (96·4%)86·1 (11·1)1517 (96·4%)79·5 (10·7)−1·44 (−2·30 to −0·58)
Mean (SD) total cholesterol (mmol/L)1300 (96·3%)5·6 (1·2)1226 (95·4%)4·4 (0·9)1593 (94·9%)5·5 (1·1)1523 (96·8%)4·2 (0·9)−0·27 (−0·34 to −0·19)
Median (IQR) HDL cholesterol (mmol/L)1289 (93·5%)1·2 (1·0–1·5)1200 (93·3%)1·3 (1·1–1·6)1568 (93·4%)1·2 (1·0–1·5)1517 (96·4%)1·2 (1·0–1·5)0 (−0·03 to 0·02)
Mean (SD) LDL cholesterol (mmol/L)1238 (89·8%)3·5 (1·0)1173 (91·3%)2·3 (0·8)1511 (90·0%)3·4 (1·0)1477 (93·8%)2·1 (0·8)−0·20 (−0·26 to −0·13)
Median (IQR) triglycerides (mmol/L)1295 (93·9%)1·7 (1·2–2·4)1213 (94·4%)1·6 (1·1–2·3)1579 (94·1%)1·6 (1·2–2·3)1512 (96·1%)1·5 (1·0–2·1)−0·05 (−0·12 to 0·01)
Mean (SD) creatinine (μmol/L)1266 (91·8%)84·9 (18·6)1208 (94·0%)79·8 (29·9)1565 (93·3%)83·4 (17·1)1513 (95·5%)81·0 (30·5)1·81 (0·10 to 3·53)
Self-reported drug use
Any glucose-lowering drug1340 (97·2%)7 (0·5%)1208 (94·0%)681 (56·4%)1609 (95·9%)8 (0·5%)1524 (96·8%)990 (65·0%)1·53 (1·25 to 1·89)
Median (IQR) total number of glucose-lowering drugs1340 (97·2%)0 (0–0)1208 (94·0%)1 (0–1)1609 (95·9%)0 (0–0)1524 (96·8%)1 (0–1)..
Metformin1340 (97·2%)5 (0·4%)1208 (94·0%)583 (48·3%)1609 (95·9%)6 (0·4%)1524 (96·8%)835 (54·8%)..
Sulphonylurea1340 (97·2%)2 (0·1%)1208 (94·0%)215 (17·8%)1609 (95·9%)2 (0·1%)1524 (96·8%)291 (19·1%)..
Thiazolidinedione1340 (97·2%)01208 (94·0)50 (4·1%)1609 (95·9%)01524 (96·8%)69 (4·5%)..
Insulin1340 (97·2%)01208 (94·0%)43 (3·6%)1609 (95·9%)01524 (96·8%)96 (6·3%)..
Other1340 (97·2%)01208 (94·0%)31 (2·6%)1609 (95·9%)01524 (96·8%)81 (5·3%)..
Any antihypertensive drugs1340 (97·2%)585 (43·7%)1208 (94·0%)911 (75·4%)1609 (95·9%)752 (46·7%)1524 (96·8%)1274 (83·6%)1·61 (1·27 to 2·04)
Median (IQR) total number of antihypertensive drugs1340 (97·2%)0 (0–1)1208 (94·0%)2 (1–3)1609 (95·9%)0 (0–2)1524 (96·8%)2 (1–3)..
ACE inhibitor or ARB1340 (97·2%)248 (18·5%)1208 (94·0%)721 (59·7%)1609 (95·9%)345 (21·4%)1524 (96·8%)1126 (73·9%)1·84 (1·52 to 2·22)
β-blocker1340 (97·2%)252 (18·8%)1208 (94·0%)285 (23·6%)1609 (95·9%)366 (22·7%)1524 (96·8%)462 (30·3%)..
Calcium-channel blocker1340 (97·2%)166 (12·4%)1208 (94·0%)326 (27·0%)1609 (95·9%)202 (12·6%)1524 (96·8%)446 (29·3%)..
Diuretic1340 (97·2%)330 (24·6%)1208 (94·0%)529 (43·8%)1609 (95·9%)415 (25·8%)1524 (96·8%)767 (50·3%)..
Other1340 (97·2%)23 (1·7%)1208 (94·0%)49 (4·1%)1609 (95·9%)32 (2·0%)1524 (96·8%)70 (4·6%)..
Any cholesterol-lowering drugs1340 (97·2%)206 (15·4%)1208 (94·0%)889 (73·6%)1609 (95·9%)274 (17·0%)1524 (96·8%)1241 (81·4%)..
Statins1340 (97·2%)200 (14·9%)1208 (93·9%)864 (71·5%)1609 (95·9%)271 (16·8%)1524 (96·8%)1217 (79·9%)1·46 (1·20 to 1·78)
Aspirin1340 (97·2%)169 (12·6%)1208 (93·9%)504 (41·7%)1609 (95·9%)249 (15·5%)1524 (96·8%)1078 (70·7%)..

The denominators at follow-up exclude 92 patients in the routine care group and 104 in the intensive treatment group who died between baseline and follow-up, and two others in the routine care group who withdrew from the study and for whom primary endpoint data were not available. The denominators used to calculate the percentages of individuals with a particular characteristic are the number of individuals with values for that characteristic. OR=odds ratio. NA=not applicable. BMI=body-mass index. HbA1c=glycated haemoglobin A1c. ACE-angiotensin-converting enzyme. ARB=angiotensin-receptor blocker.

Screening programmes varied by centre (table 1). The median number of follow-up meetings for audit and feedback was four (range two to ten). Primary endpoint data were available for 3055 (99·9%) of 3057 participants. The mean follow-up period was 5·3 (SD 1·6) years, during which 238 first cardiovascular events occurred, with similar numbers and risk in each group (table 4). The cumulative probability plot for the primary endpoint seemed to diverge after 4 years of follow-up (figure 2). In the predefined subgroup analyses, no interactions were seen between the intervention and age or previous cardiovascular event (p>0·1). Estimated HRs, however, were 1·12 (95% CI 0·70–1·79) in patients younger than 60 years and 0·70 (95% CI 0·52–0·95) in those aged 60 years and older. HRs for individual components of the composite endpoint all favoured the intensive treatment group (table 4), although none achieved significance (figure 2). No patient underwent amputation as a first event. The correlation coefficient within clusters for the primary endpoint was 0·002 (Denmark: 0·014, UK [combined] 0·0000016, and the Netherlands 0·025), which suggests that the cluster design had little effect on study power.

An external file that holds a picture, illustration, etc.
Object name is gr2.jpg

Cumulative incidence and relative risk of composite cardiovascular endpoint

(A) Cumulative incidence curves by treatment group. The p value was calculated with Cox's regression and fixed-effects meta-analysis. (B) HRs of development of cardiovascular death, non-fatal myocardial infarction, non-fatal stroke, and revascularisation as a first event (secondary endpoints), and these cardiovascular events combined (primary endpoint), by country and overall. HR=hazard ratio. CVD=cardiovascular disease.

Table 4

Cardiovascular events and all-cause mortality in the two treatment groups

Routine care (n=1377)Intensive treatment (n=1678)Hazard ratio(95% CI)I2 (%)p for routine care vs intensive treatment
Primary endpoint (n [%])
Composite cardiovascular events*117 (8·5%)121 (7·2%)0·83 (0·65–1·05)0%0·12
Secondary endpoints (n [%])
Cardiovascular death22 (1·6%)26 (1·5%)0·88 (0·51–1·51)52%..
Myocardial infarction32 (2·3%)29 (1·7%)0·7 (0·41–1·21)0%..
Stroke19 (1·4%)22 (1·3%)0·98 (0·57–1·71)0%..
Revascularisation44 (3·2%)44 (2·6%)0·79 (0·53–1·18)0%..
Amputation00......
Total mortality92 (6·7%)104 (6·2%)0·91 (0·69–1·21)55%..

Hazard ratios were first estimated within each country with Cox's regression and Huber-White adjustment of SE for clustering within practice, then combined across countries with fixed-effects meta-analysis. The I2 statistic estimates heterogeneity between countries. A p value was calculated for primary endpoint only.

*Any of cardiovascular death, myocardial infarction, stroke, revascularisation, and amputation.
Component of the primary endpoint as a first event.

196 patients died overall (60 [30·6%] cardiovascular deaths, including six patients with first events classified as myocardial infarction, two as stroke, and four as revascularisation, 97 [49·5%] cancer deaths, and 39 [19·9%] from other causes; table 4). The combined HR for death in the intensive treatment group compared with the routine care group was 0·91 (95% CI 0·69–1·21). Heterogeneity of results between countries was not significant (figure 3). In the UK, significantly fewer patients died in the intensive treatment group than in the routine care group, but in Denmark the risk in the routine care group was lower, although not significantly so. All results were unchanged after sensitivity analysis that excluded participants in the two practices for which endpoint data were obtained while aware of treatment allocation (webappendix p 2).

An external file that holds a picture, illustration, etc.
Object name is gr3.jpg

Cumulative incidence and relative risk of all-cause mortality

(A) Kaplan-Meier survival estimates by treatment group. (B) HRs of all-cause mortality, by country and overall. HR=hazard ratio. CVD=cardiovascular disease.

Of the 2859 patients still alive at 5 years, 2400 (84%) returned to a clinical research facility for follow-up health assessments. Clinical and biochemical outcomes could be obtained from general practice records for a further 328 (11·5%) participants. Compared with patients for whom follow-up data were available, the 131 with missing data were more likely to be from an ethnic minority group (10·2% vs 5·7%, p=0·04) and have higher baseline total cholesterol (5·9 mmol/L vs 5·6 mmol/L, p=0·004) and LDL cholesterol values (3·7 mmol/L vs 3·4 mmol/L, p=0·009). Changes in clinical and biochemical values and prescribed medications in the two groups are shown in table 3. By 5 years of follow-up improvements were seen for cardiovascular risk factors in both groups. Small but significant differences between groups were seen in change from baseline for glycated haemoglobin A1c (HbA1c), systolic and diastolic blood pressure and total and LDL cholesterol, in favour of the intensive treatment group. Prescription of glucose-lowering, antihypertensive, and lipid-lowering drugs increased in both groups. At follow-up more patients in the intensive treatment group were prescribed aspirin, angiotensin-converting-enzyme inhibitors or angiotensin-receptor blockers, glucose-lowering drugs, antihypertensive drugs, and lipid-lowering drugs than were those in the routine care group (table 3).

In both groups, more patients had values below target thresholds for HbA1c concentrations, blood pressure, and cholesterol concentrations at follow-up than at baseline (figure 4). The proportion of patients meeting the targets was higher in the intensive treatment group than in the routine care group. The proportions of individuals reporting hypoglycaemia, as assessed by the diabetes treatment satisfaction questionnaire, did not differ (χ2 4·44, p=0·62).33 All results were unchanged after sensitivity analysis was done that excluded participants with follow-up clinical data obtained from general practice records.

An external file that holds a picture, illustration, etc.
Object name is gr4.jpg

Proportion of patients for whom cardiovascular risk factor values were below the intensive treatment intervention target thresholds at baseline and after 5 years of follow-up

1 SE are shown. CVD=cardiovascular disease. HbA1c=glycated haemoglobin A1c.

Discussion

An intervention to promote target-driven, intensive management of patients with type 2 diabetes detected by screening was associated with slightly, but significantly, increased prescription of treatments and improvements in cardiovascular risk factors, and with a non-significant relative reduction in the incidence of cardiovascular events at 5 years. Differences between study groups for all components of the primary endpoint favoured the intensive treatment group. Differences were greatest for myocardial infarction and smallest for stroke.

We cannot rule out the possibility that these findings were due to chance. Rates of cardiovascular events seemed to diverge after 4 years of follow-up, although the risk of adverse outcomes, including death or self-reported hypoglycaemia, was not increased. The incidence of cardiovascular events in the routine care group (8·5%) was lower than expected compared with that in newly diagnosed patients in the UKPDS (12·1%).34 Mortality in this group (6·7%) was lower than that in patients with type 2 diabetes detected by screening in Hoorn35 (25% over 10 years) and newly diagnosed patients in a study in Denmark (33% over 7·4 years).36 In our study, mortality was similar to that reported for people of the same age without diabetes in the general population of Denmark in 1995–2006.37 This lack of difference is likely to be due to the quality of care delivered to patients early in the course of their disease in both groups.

This trial shows that screening for type 2 diabetes and early intensive multifactorial treatment of the detected patients are feasible in general practice. In both study groups, cardiovascular risk factors, such as blood pressure and cholesterol concentrations, improved notably after diagnosis and glycaemia and weight did not increase. Absolute values for risk factors at follow-up and changes from baseline compare favourably with those in other trials in which patients were recruited at clinical diagnosis and followed up for 6 years.34,35 Clinically important differences in risk factors between groups at 1 year,18 however, were not maintained and were considerably smaller than those achieved in similar studies.2,34

Adherence to treatment algorithms might have been suboptimum in this pragmatic trial. In three centres the screening programme used risk scores, including treatment for hypertension as one of the factors to predict the risk of type 2 diabetes, which could have limited the achievable differences between groups in blood pressure. Furthermore, the trial was undertaken against a background of improvements in the delivery of diabetes care in general practice, such as that associated with the introduction of the Quality and Outcomes Framework for primary care in the UK,38 and evidence-based guidelines in Denmark39 and the Netherlands,40 which might also have lowered the achievable differences in treatment between groups.

Participants were drawn from a large, representative, population-based sample in three different European countries. They were identified by a range of different screening programmes and all were diagnosed according to WHO criteria.21 The intensive treatment intervention incorporated various methods that encouraged changes in practitioner behaviour, and treatment algorithms and targets that were based on robust trial data.2,4,6,26 Because the intervention was delivered mainly via family physicians and nurses, we randomised general practices rather than individuals to minimise the risk of contamination. The analyses were appropriate to the cluster design. We achieved high participant retention and independently adjudicated endpoint ascertainment in both trial groups. We assessed clinically important outcomes with standard equipment and protocols by especially trained staff members who were unaware of study group allocation. Patients with and without follow-up data differed little.

The generalisability of our findings to other settings should be considered in light of the non-random recruitment of general practices. We did, however, cover a large geographical area in each country, and the 26% of invited practices that were randomised were nationally representative for key descriptive characteristics.17,18,20 Assessment of hypoglycaemia was imprecise, as it was limited to one question, but we believe this factor is unlikely to be biased because the questionnaire was administered in the same way to participants in the two study groups by staff unaware of study group allocations. We feel that the size of the study means that even with a potentially imprecise instrument the power was sufficient to quantify the effects of the intervention on the frequency of hypoglycaemic events. In two centres (Denmark and the Netherlands), assays used to assess biochemical factors differed slightly between baseline and follow-up, but not between groups (webappendix p 1). By necessity cluster randomisation was done before screening and recruitment of patients, a design feature that can introduce differences between groups in relation to participant characteristics. Overall, patients in the two study groups were well matched, although the intervention could have influenced the number and type of patients recruited to the intensive treatment group. This effect might have been particularly relevant in Denmark, where staff in the intensive treatment group might have been especially alert to the potential benefits of early detection and treatment and, therefore, have been more active testing in individuals at high cardiovascular risk, which could have led to the observed differences in baseline characteristics between study groups in this centre. These differences between groups might have lessened the apparent effects of the intervention and, therefore, could explain, at least partly, differences between centres, which seemed to be present but were not necessarily significant owing to the limited power of heterogeneity test.

Methods of screening, outcome assessment, and laboratory testing were standard across patient groups within centres but did differ between centres. These differences might have contributed to heterogeneity, even though interlaboratory validation suggested that methods were consistent between centres and all potential primary endpoints were adjudicated in the same way. Other sources of heterogeneity include the characteristics of patients, which varied by centre, presumably because of differences in the screening programmes and underlying populations, the delivery of the intensive treatment intervention (table 1), and other unmeasured factors.

The lower-than-expected event rate suggests that 5 years of follow-up is insufficient, and the apparent divergence from 4 years onwards suggests that further follow-up is justified to test whether early intensive multifactorial treatment reduces cardiovascular risk in the long term, as seen in the UKPDS.8 Finally, the complex nature of the intensive treatment intervention and the pragmatic design of the trial make it difficult to say whether any components are individually associated with a reduction in cardiovascular risk.

The observed absolute and relative risks for cardiovascular events and mortality among participants with a mean HbA1c concentration of around 6·5% 5 years after diagnosis, should allay concerns about early intensive treatment of hyperglycaemia.

We saw no interaction between intensive treatment and age or history of cardiovascular events, although benefits associated with this intervention seemed to be greatest in patients aged 60 years or older at diagnosis. Multifactorial treatment of screen-detected patients in both study groups was associated with improvements in cardiovascular risk factors and lower than expected rates of cardiovascular events and mortality. The small differences in treatment between groups at 5 years were associated with a non-significant 17% reduction in the incidence of cardiovascular events, with no obvious adverse consequences. We are undertaking analyses of trial data on microvascular endpoints, quality of life, functional status, and health service costs. Although there is no evidence of harm associated with screening11 and intensive therapy,41 the extent to which the complications of diabetes can be reduced by earlier detection and treatment remains uncertain (panel).

Panel

Research in context

Systematic review

Studies have shown cardiovascular benefits with treatment to lower blood pressure and glucose and cholesterol concentrations in patients with type 2 diabetes mellitus.4–7 Intensive multifactorial treatment halved risk of cardiovascular events and death in patients with longstanding diabetes and microalbuminuria in the STENO-2 trial.2 We searched PubMed for relevant articles by use of “diabetes mellitus, type 2” and “cardiovascular disease” as MeSH headings and the term “randomised controlled trial” in any heading, in combination with the terms “multifactorial” or “screen*”. We placed no restriction on language, year of publication, or study quality. We found no published trial evidence of the effects of multifactorial treatment on cardiovascular outcomes in individuals with screen-detected type 2 diabetes.

Interpretation

Population screening for type 2 diabetes and subsequent intensive treatment is feasible. Cardiovascular risk factors improved in the 5 years after detection by screening, and rates of first cardiovascular events and mortality were lower than expected. The small differences in prescribed treatment, and levels of cardiovascular risk factors, between groups at 5 years were associated with a non-significant 17% reduction in the incidence of cardiovascular events, with no obvious adverse consequences. The extent to which the complications of diabetes can be reduced by earlier detection and treatment remains uncertain.

When compared with routine care, an intervention to promote target-driven, intensive management of patients with type 2 diabetes detected by screening was associated with small increases in the prescription of drugs and improvements in cardiovascular risk factors, but was not associated with significant reductions in the incidence of cardiovascular events or death over 5 years.

Acknowledgments

We thank all patients, practice nurses, and family physicians for their contributions to the ADDITION-Europe study. We thank Ronald P Stolk, and Bruce H R Wolffenbuttel, University Medical Centre Groningen, University of Groningen, Netherlands, for participation on the steering committee that designed the study. ADDITION-Denmark was supported by the National Health Services in the counties of Copenhagen, Aarhus, Ringkøbing, Ribe and South Jutland in Denmark, the Danish Council for Strategic Research, the Danish Research Foundation for General Practice, Novo Nordisk Foundation, the Danish Centre for Evaluation and Health Technology Assessment, the diabetes fund of the National Board of Health, the Danish Medical Research Council, and the Aarhus University Research Foundation. The trial has been given unrestricted grants from Novo Nordisk AS, Novo Nordisk Scandinavia AB, Novo Nordisk UK, ASTRA Denmark, Pfizer Denmark, GlaxoSmithKline Pharma Denmark, Servier Denmark, and HemoCue Denmark. Parts of the grants from Novo Nordisk Foundation, Danish Council for Strategic Research and Novo Nordisk were transferred to the other centres. We thank the Biochemistry Department at Steno Diabetes Centre for biochemical testing, and the following hospital departments for doing clinical assessments: Steno Diabetes Centre Research Laboratory, Esbjerg Hospital Diabetes Clinic, Holstebro Hospital Research department, Aabenraa Hospital Outpatient Clinic and Aarhus University Hospital Eye Department. ADDITION-Cambridge was supported by the Wellcome Trust (grant reference no: G061895), the Medical Research Council (grant reference no: G0001164), the NIHR Health Technology Assessment Programme (grant reference no: 08/116/300), National Health Service R&D support funding (including the Primary Care Research and Diabetes Research Networks), and the National Institute for Health Research under its Programme Grants for Applied Research scheme [RP-PG-0606-1259]. The views expressed in this publication are those of the authors and not necessarily those of the National Health Service, the National Institute for Health Research, or the UK Department of Health. Bio-Rad provided equipment for haemoglobin A1c testing during the screening phase. We thank the Cambridge University Hospitals NHS Foundation Trust Department of Clinical Biochemistry and the NIHR Cambridge Biomedical Research Centre, Core Biochemical Assay Laboratory for carrying out the biochemical assays. ADDITION-Netherlands was supported by unrestricted grants from Novo Nordisk, Glaxo Smith Kline and Merck, and by the Julius Centre for Health Sciences and Primary Care, University Medical Centre, Utrecht. ADDITION-Leicester was supported by Department of Health and ad hoc Support Sciences, the NIHR Health Technology Assessment Programme (grant reference no: 08/116/300), National Health Service research and development support funding (including the Primary Care Research and Diabetes Research Network, and LNR CLAHRC) and the National Institute for Health Research. MJD and KK receive support from the Department of Health NIHR Programme Grant funding scheme [RP-PG-0606-1272]. We thank the University Hospitals of Leicester NHS Trust Department of Clinical Biochemistry for carrying out biochemical assays. Permission to use the DTSQ questionnaire was granted by Professor Clare Bradley, Health Psychology Research, Royal Holloway University of London, Surrey, UK. These results were presented at the European Association for the Study of Diabetes (EASD) Annual Scientific Meeting, Stockholm, Sweden, Sept 20–24, 2010.

Contributors

SJG acts as guarantor for this paper. TL is Chair and KB-J Vice-Chair of the steering committee, which also included the following people, who collectively designed the study: SJG, MJD, KK, GEHMR, AS, and NJW. SJG, KB-J, MJD, KK, GEHMR, AS, NJW, and TL are principal investigators for the trial who, along with RKS and MvdD, participated in the acquisition of the data. SJS did the statistical analyses. All authors participated in the analysis and interpretation of data. SJG and RKS drafted the report. All authors participated in the critical revision of the intellectual content of the report.

ADDITION-Denmark

Bendix Carstensen, Merete Frandsen (Steno Diabetes Centre, Gentofte); Else-Marie Dalsgaard, Ynna Nielsen, Søren Bech-Morsing, Mette Vinther Skriver, Helle Terkildsen, Morten Charles (School of Public Health, Aarhus); Toke Bek (Aarhus University Hospital, Aarhus); Henrik Lund Andersen (Glostrup Hospital, Glostrup).

ADDITION-Cambridge

Gisela Baker, Daniel Barnes, Mark Betts, Sandra Bovan, Parinya Chamnan, Sue Emms, Francis Finucane, Susie Hennings, Muriel Hood, Garry King, Christine May Hall, Joanna Mitchell, Emanuella De Lucia Rolfe, Liz White (MRC Epidemiology Unit, Cambridge); Amanda Adler, Sean Dinneen, Mark Evans (Cambridge University Hospitals, NHS Foundation Trust, Cambridge); Judith Argles, Rebecca Bale, Roslyn Barling, Sue Boase, Ryan Butler, Pesheya Doubleday, Tom Fanshawe, Philippa Gash, Julie Grant, Wendy Hardeman, Ann-Louise Kinmonth, Richard Parker, Nicola Popplewell, A Toby Prevost, Megan Smith, Stephen Sutton, Fiona Whittle, Kate Williams, Rebecca Abbott, Georgina Lewis, Lincoln Sargeant (Department of Public Health and Primary Care, University of Cambridge); Robert Henderson (Hinchingbrooke Hospital, Huntingdon).

ADDITION-Netherlands

Kees Gorter, Paul Janssen, Lidian Izeboud, Jacqueline Berends, Marlies Blijleven, Bart Thoolen, Denise de Ridder, Jozien Bensing, Mehmet Akarsubasi, Paula Koekkoek, Carla Ruis, Geert Jan Biessels, Jaap Kappelle, Michiel van der Linden (Utrecht University, Utrecht).

ADDITION-Leicester

Balasubramanian Thiagarajan Srinivasan, David Webb, Mary Quinn, Emma Wilmot, Samiul A Mostafa, Nitin Gholap, Hamid Mani, Winston Crasto, Steve Hiles, Joe Henson, Janet Jarvis, Sukhjit Sehmi, Fiona Ablett, Champa Merry, Emma Healey, Julia Stockman, Sandra Campbell, Janette Barnett, Nil Radia, Mo Radia, Jo Howe, Lesley Bryan, Jane Brela, Jayne Hill, Helen Bray, Rachel Plummer, Zubeir Essat, Francis Pullen, Susan Enright (University Hospitals of Leicester NHS Trust); Laura J Gray, Nick Taub (University of Leicester).

Independent endpoint committee

Kristian Thygesen, Ole Færgeman (Aarhus University Hospital, Aarhus, Denmark); Hans Ibsen (Holbæk Hospital, Holbæk, Denmark); Birger Thorsteinsson (Hillerød Hospital, Hillerød, Denmark); Jane Armitage, Louise Bowman (University of Oxford, Oxford, UK); Cees Tack (Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands) and Jaap Kappelle (University Medical Centre, Utrecht, Netherlands)

Independent endpoint committee Cambridge

Nigel Stott (Chair; University of Cardiff, Cardiff, UK), John Weinman (King's College, London, London, UK), Richard Himsworth (University of Cambridge, Cambridge, UK), Paul Little (University of Southampton, Southampton, UK).

Independent data monitoring and safety committee

Christian Gluud, Per Winkel, Jørn Wetterslev (Copenhagen University, Copenhagen, Denmark).

Additional study support

Daniel Barnes (data cleaning, analysis, and presentation), Adam Dickinson (data management), Iain Morrison (information technology), Matt Sims (technical), Paul Roberts, Kim Mwanza, James Sylvester (field epidemiology; MRC Epidemiology Unit); Wim Rutten, Paulien van Hessen (collaboration), Mardy Eckhardt, Erna Erdtsieck, Leandra Boonman, Annelies van der Smissen (screening, health assessments, and biochemical assays; SHL Centre for Diagnostic Support in Primary Care, Etten-Leur, Netherlands). Nicole Boekema, Robert Veen (data management), Peter Zuithof (statistics), Lizeth Vendrig, Annette Bak (staff recruitment; Julius Centre, Utrecht, Netherlands).

Web Extra Material

Supplementary webappendix:

References

1. IDF . IDF Diabetes Atlas. 4th edn. International Diabetes Federation; Brussels, Belgium: 2009. [Google Scholar]
2. Gaede P, Vedel P, Larsen N, Jensen GV, Parving HH, Pedersen O. Multifactorial intervention and cardiovascular disease in patients with type 2 diabetes. N Engl J Med. 2003;348:383–393. [Abstract] [Google Scholar]
3. Gaede P, Lund-Andersen H, Parving HH, Pedersen O. Effect of a multifactorial intervention on mortality in type 2 diabetes. N Engl J Med. 2008;358:580–591. [Abstract] [Google Scholar]
4. UK Prospective Diabetes Study Group Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38. BMJ. 1998;317:703–713. [Europe PMC free article] [Abstract] [Google Scholar]
5. Heart Outcomes Prevention Evaluation Study Investigators Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE substudy. Lancet. 2000;355:253–259. [Abstract] [Google Scholar]
6. Pyorala K, Pedersen TR, Kjekshus J, Faergeman O, Olsson AG, Thorgeirsson G. Cholesterol lowering with simvastatin improves prognosis of diabetic patients with coronary heart disease. A subgroup analysis of the Scandinavian Simvastatin Survival Study (4S) Diabetes Care. 1997;20:614–620. [Abstract] [Google Scholar]
7. Ray KK, Seshasai SR, Wijesuriya S. Effect of intensive control of glucose on cardiovascular outcomes and death in patients with diabetes mellitus: a meta-analysis of randomised controlled trials. Lancet. 2009;373:1765–1772. [Abstract] [Google Scholar]
8. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359:1577–1589. [Abstract] [Google Scholar]
9. Harris MI, Klein R, Wellborn TA, Knuiman MW. Onset of NIDDM occurs at least 4–7yr before clinical diagnosis. Diabetes Care. 1992;15:815–819. [Abstract] [Google Scholar]
10. Glumer C, Jorgensen T, Borch-Johnsen K. Prevalences of diabetes and impaired glucose regulation in a Danish population: the Inter99 study. Diabetes Care. 2003;26:2335–2340. [Abstract] [Google Scholar]
11. Eborall HC, Griffin SJ, Prevost AT, Kinmonth AL, French DP, Sutton S. Psychological impact of screening for type 2 diabetes: controlled trial and comparative study embedded in the ADDITION (Cambridge) randomised controlled trial. BMJ. 2007;335:486. [Europe PMC free article] [Abstract] [Google Scholar]
12. Wareham NJ, Griffin SJ. Should we screen for type 2 diabetes? Evaluation against national screening committee criteria. BMJ. 2001;322:986–988. [Europe PMC free article] [Abstract] [Google Scholar]
13. Kahn R, Alperin P, Eddy D. Age at initiation and frequency of screening to detect type 2 diabetes: a cost-effectiveness analysis. Lancet. 2010;375:1365–1374. [Abstract] [Google Scholar]
14. Glumer C, Yuyun M, Griffin S. What determines the cost-effectiveness of diabetes screening? Diabetologia. 2006;49:1536–1544. [Abstract] [Google Scholar]
15. Lauritzen T, Griffin S, Borch-Johnsen K, Wareham NJ, Wolffenbuttel BH, Rutten G. The ADDITION study: proposed trial of the cost-effectiveness of an intensive multifactorial intervention on morbidity and mortality among people with Type 2 diabetes detected by screening. Int J Obes Relat Metab Disord. 2000;24(suppl 3):S6–S11. [Abstract] [Google Scholar]
16. Echouffo-Tcheugui JB, Simmons RK, Williams KM. The ADDITION-Cambridge trial protocol: a cluster-randomised controlled trial of screening for type 2 diabetes and intensive treatment for screen-detected patients. BMC Public Health. 2009;9:136. [Europe PMC free article] [Abstract] [Google Scholar]
17. Webb DR, Khunti K, Srinivasan B. Rationale and design of the ADDITION-Leicester study, a systematic screening programme and randomised controlled trial of multi-factorial cardiovascular risk intervention in people with type 2 diabetes mellitus detected by screening. Trials. 2010;11:16. [Europe PMC free article] [Abstract] [Google Scholar]
18. Janssen PG, Gorter KJ, Stolk RP, Rutten GE. Randomised controlled trial of intensive multifactorial treatment for cardiovascular risk in patients with screen-detected type 2 diabetes: 1-year data from the ADDITION Netherlands study. Br J Gen Pract. 2009;59:43–48. [Europe PMC free article] [Abstract] [Google Scholar]
19. Sandbaek A, Griffin SJ, Rutten G. Stepwise screening for diabetes identifies people with high but modifiable coronary heart disease risk. The ADDITION study. Diabetologia. 2008;51:1127–1134. [Europe PMC free article] [Abstract] [Google Scholar]
20. Sargeant LA, Simmons RK, Barling RS. Who attends a UK diabetes screening programme? Findings from the ADDITION-Cambridge study. Diabet Med. 2010;27:995–1003. [Europe PMC free article] [Abstract] [Google Scholar]
21. WHO . Definition, diagnosis, and classification of diabetes mellitus and its complications: report of a WHO consultation. World Health Organization; Geneva, Switzerland: 1999. [Google Scholar]
22. Griffin SJ, Little PS, Hales CN, Kinmonth AL, Wareham NJ. Diabetes risk score: towards earlier detection of Type 2 diabetes in general practice. Diabetes Metab Res Rev. 2000;16:164–171. [Abstract] [Google Scholar]
23. Glumer C, Carstensen B, Sandbaek A, Lauritzen T, Jorgensen T, Borch-Johnsen K. A Danish diabetes risk score for targeted screening: the Inter99 study. Diabetes Care. 2004;27:727–733. [Abstract] [Google Scholar]
24. Davies MJ, Heller S, Skinner TC. Effectiveness of the diabetes education and self management for ongoing and newly diagnosed (DESMOND) programme for people with newly diagnosed type 2 diabetes: cluster randomised controlled trial. BMJ. 2008;336:491–495. [Europe PMC free article] [Abstract] [Google Scholar]
25. Ruige JB, de Neeling JN, Kostense PJ, Bouter LM, Heine RJ. Performance of an NIDDM screening questionnaire based on symptoms and risk factors. Diabetes Care. 1997;20:491–496. [Abstract] [Google Scholar]
26. UK Prospective Diabetes Study Group Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33) Lancet. 1998;352:837–853. [Abstract] [Google Scholar]
27. Collins R, Armitage J, Parish S, Sleigh P, Peto R. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial. Lancet. 2003;361:2005–2016. [Abstract] [Google Scholar]
28. Royal College of General Practitioners in Denmark . Type 2-diabetes in general practice—diagnosis and treatment. Royal College of General Practioners; Copenhagen, Denmark: 1999. [Google Scholar]
29. Royal College of General Practitioners. McIntosh A, Hutchinson A, Home PD. Clinical guidelines and evidence review for type 2 diabetes: management of blood glucose. University of Sheffield; Sheffield, UK: 2001. [Google Scholar]
30. Rutten GEHM, Verhoeven S, Heine RJ, De Grauw WJC, Cromme PVM, Reenders K. Dutch College of General Practitioners. Practice guideline diabetes mellitus type 2. Huisarts Wetensch. 1999;42:67–84. [Google Scholar]
31. White IR, Thompson SG. Adjusting for partially missing baseline measurements in randomized trials. Stat Med. 2005;24:993–1007. [Abstract] [Google Scholar]
32. Gooley TA, Leisenring W, Crowley J, Storer BE. Estimation of failure probabilities in the presence of competing risks: new representations of old estimators. Stat Med. 1999;18:695–706. [Abstract] [Google Scholar]
33. Bradley C, editor. Handbook of psychology and diabetes: a guide to psychological measurement in diabetes research and practice. Psychology Press; London, UK: 1994. [Google Scholar]
34. UK Prospective Diabetes Study Group Overview of 6 years' therapy of type II diabetes: a progressive disease. Diabetes. 1995;44:1249–1258. [Abstract] [Google Scholar]
35. Spijkerman AM, Dekker JM, Nijpels G. Impact of diabetes duration and cardiovascular risk factors on mortality in type 2 diabetes: the Hoorn Study. Eur J Clin Invest. 2002;32:924–930. [Abstract] [Google Scholar]
36. Olivarius NF, Beck-Nielsen H, Andreasen AH, Horder M, Pedersen PA. Randomised controlled trial of structured personal care of type 2 diabetes mellitus. BMJ. 2001;323:970–975. [Europe PMC free article] [Abstract] [Google Scholar]
37. Carstensen B, Kristensen J, Ottosen P, Borch-Johnsen K. The Danish National Diabetes Register: trends in incidence, prevalence and mortality. Diabetologia. 2008;51:2187–2196. [Abstract] [Google Scholar]
38. Doran T, Fullwood C, Gravelle H. Pay-for-performance programs in family practices in the United Kingdom. N Engl J Med. 2006;355:375–384. [Abstract] [Google Scholar]
39. Royal College of General Practitioners in Denmark . Type 2 diabetes in general practice—an evidence based guideline. Royal College of General Practioners; Copenhagen, Denmark: 2004. [Google Scholar]
40. Rutten GEHM, de Grauw WJC, Nijpels G. Dutch Guidelines on Type 2 Diabetes Mellitus. Dutch College of General Practitioners. Huisarts Wetensch. 2006;49:137–152. [Google Scholar]
41. van den Donk M, Gorter KJ, Rutten GE. No negative effects of a multi-factorial, intensified treatment on self-reported health status, treatment satisfaction, and diabetes-related distress in screen-detected type 2 diabetes patients. The ADDITION-Netherlands study. Qual Life Res. 2010;19:509–513. [Europe PMC free article] [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/102366933
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/102366933

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/10.1016/s0140-6736(11)60698-3

Supporting
Mentioning
Contrasting
13
456
0

Article citations


Go to all (251) 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 and ad hoc Support Sciences, the NIHR Health Technology Assessment Programme (1)

Medical Research Council (4)

National Health Service R&amp;D support funding (1)

National Institute for Health Research (NIHR) (3)

Wellcome Trust (1)

the Department of Health NIHR Programme Grant funding scheme (1)

the Medical Research Council (1)

the NIHR Health Technology Assessment Programme (1)