Scolaris Content Display Scolaris Content Display

Secretagogos de insulina para la prevención o el retraso de la diabetes mellitus tipo 2 y las complicaciones asociadas en los pacientes en mayor riesgo para el desarrollo de diabetes mellitus tipo 2

Collapse all Expand all

Resumen

Antecedentes

El aumento proyectado en la incidencia de diabetes mellitus tipo 2 (DMT2) se podría convertir en un problema significativo de salud en todo el mundo. Se desconoce si los secretagogos de insulina (sulfonilureas y análogos de la meglitinida) pueden prevenir o retrasar la DMT2 y las complicaciones asociadas en los pacientes en riesgo de desarrollar DMT2.

Objetivos

Evaluar los efectos de los secretagogos de insulina sobre la prevención o el retraso de la DMT2 y las complicaciones asociadas en los pacientes con intolerancia a la glucosa, deterioro de la glucemia en ayunas, hemoglobina glucosilada A1c (HbA1c) moderadamente elevada o cualquier combinación de estos.

Métodos de búsqueda

Se hicieron búsquedas en el Registro Cochrane Central de Ensayos Controlados (Cochrane Central Register of Controlled Trials), MEDLINE, PubMed, Embase, ClinicalTrials.gov, la World Health Organization International Clinical Trials Registry Platform, y en las listas de referencias de revisiones sistemáticas, artículos y en informes de evaluación de tecnología sanitaria. Se preguntó a los investigadores de los ensayos incluidos para obtener información acerca de ensayos adicionales. La fecha de la última búsqueda para todas las bases de datos fue abril 2016.

Criterios de selección

Se incluyeron los ensayos controlados aleatorios (ECA) con una duración de 12 semanas o más que compararon secretagogos de insulina con cualquier intervención farmacológica de disminución de la glucosa, intervención de cambio del comportamiento, placebo o ninguna intervención en pacientes con deterioro de la glucosa en ayunas, intolerancia a la glucosa, HbA1c moderadamente elevada o combinaciones de estos.

Obtención y análisis de los datos

Dos revisores leyeron todos los resúmenes y el texto completo de los artículos / registros, evaluaron la calidad y extrajeron los datos de resultado de forma independiente. Un autor de la revisión extrajo los datos que fueron verificados por un segundo autor. Las discrepancias se resolvieron mediante consenso o la intervención de un tercer autor de la revisión. Para los metanálisis se utilizó un modelo de efectos aleatorios y se investigaron los cocientes de riesgo (CR) de los resultados dicotómicos y las diferencias de medias (DM) de los resultados continuos, con el uso de intervalos de confianza (IC) del 95% para las estimaciones del efecto. Se realizaron análisis secuenciales de ensayos (ASE) de todos los resultados con los que fue posible realizar un metanálisis. Se evaluó la calidad general de las pruebas mediante el instrumento GRADE.

Resultados principales

Se incluyeron seis ECA con 10 018 participantes; 4791 participantes con datos sobre la asignación a los grupos de intervención se asignaron al azar a una sulfonilurea de segunda o tercera generación o un análogo de la meglitinida como monoterapia y 29 participantes se asignaron al azar a una sulfonilurea de segunda generación más metformina. Tres ensayos investigaron una sulfonilurea de segunda generación, dos ensayos investigaron una sulfonilurea de tercera generación y un ensayo un análogo de la meglitinida. Un total de 4873 participantes con datos sobre la asignación a los grupos control se asignaron al azar a un grupo comparador; 4820 participantes se asignaron al azar a placebo, 23 a dieta y ejercicios, y 30 participantes a monoterapia con metformina. Un ECA de nateglinida contribuyó con el 95% de todos los participantes. La duración de la intervención varió de seis meses a cinco años. Se consideró que ninguno de los ensayos incluidos tuvo bajo riesgo de sesgo en todos los dominios del "Riesgo de sesgo".

Con muy poca frecuencia se observó mortalidad por todas las causas y mortalidad cardiovascular después del tratamiento con sulfonilurea (glimepirida) (pruebas de muy baja calidad). el CR para la incidencia de DMT2 al comparar la monoterapia con glimepirida con placebo fue 0,75; IC del 95%: 0,54 a 1,04; P = 0,08; dos ensayos; 307 participantes; pruebas de muy baja calidad. Uno de los ensayos que informó la incidencia de DMT2 no definió los criterios diagnósticos utilizados. El otro ensayo diagnosticó la DMT2 como dos valores de glucemia en ayunas consecutivos ≥ 6,1 mmol/l. El ASE indicó que hasta el presente sólo se ha alcanzado el 4,5% del tamaño de información necesario ajustado por la diversidad. Ningún ensayo informó datos sobre los eventos adversos graves, el infarto de miocardio (IM) no mortal, el accidente cerebrovascular no mortal, la insuficiencia cardíaca (IC) congestiva, la calidad de vida relacionada con la salud o los efectos socioeconómicos.

Un ensayo con un seguimiento de cinco años comparó un análogo de la meglitinida (nateglinida) con placebo. Un total de 310/4645 participantes (6,7%) asignados a nateglinida murieron en comparación con 312/4661 (6,7%) participantes asignados a placebo (cociente de riesgos instantáneos [CRI] 1,00; IC del 95%: 0,85 a 1,17; P = 0,98; pruebas de calidad moderada). Los dos criterios principales para el diagnóstico de DMT2 fueron un nivel de glucosa en plasma en ayunas ≥ 7,0 mmol/l o una prueba de tolerancia a la glucosa a las dos horas ≥ 11,1 mmol/l. La DMT2 se desarrolló en 1674/4645 (36,0%) participantes del grupo de nateglinida y en 1580/4661 (33,9%) del grupo placebo (CRI 1,07; IC del 95%: 1,00 a 1,15; P = 0,05; pruebas de calidad moderada). Se informaron uno o más eventos adversos graves en 2066/4602 (44,9%) participantes asignados a nateglinida en comparación con 2089/4599 (45,6%) participantes asignados a placebo. Un total de 126/4645 participantes (2,7%) asignados a nateglinida murieron debido a enfermedades cardiovasculares en comparación con 118/4661 (2,5%) participantes asignados a placebo (CRI 1,07; IC del 95%: 0,83 a 1,38; P = 0,60; pruebas de calidad moderada). La comparación de los participantes que recibieron nateglinida con los que recibieron placebo para los resultados IM, accidente cerebrovascular no mortal e IC proporcionó las siguiente tasas de eventos: IM 116/4645 (2,5%) versus 122/4661 (2,6%), accidente cerebrovascular 100/4645 (2,2%) versus 110/4661 (2,4%) y número de hospitalizados por IC 85/4645 (1,8%) versus 100/4661 (2,1%) ‐ (CRI 0,85; IC del 95%: 0,64 a 1,14; P = 0,27). La calidad de las pruebas era moderado para todos estos resultados. No se informó la calidad de vida relacionada con la salud ni los efectos socioeconómicos.

Conclusiones de los autores

No hay pruebas suficientes para demostrar si los secretagogos de insulina comparados principalmente con placebo reducen el riesgo de desarrollar DMT2 y las complicaciones asociadas en los pacientes en mayor riesgo de desarrollar DMT2. La mayoría de los ensayos no investigaron resultados importantes para los pacientes.

PICOs

Population
Intervention
Comparison
Outcome

The PICO model is widely used and taught in evidence-based health care as a strategy for formulating questions and search strategies and for characterizing clinical studies or meta-analyses. PICO stands for four different potential components of a clinical question: Patient, Population or Problem; Intervention; Comparison; Outcome.

See more on using PICO in the Cochrane Handbook.

Resumen en términos sencillos

¿Los fármacos secretagogos de insulina que disminuyen la glucosa pueden prevenir o retrasar la diabetes mellitus tipo 2 y las complicaciones asociadas en los pacientes en mayor riesgo de esta enfermedad?

Pregunta de la revisión

¿El grupo de fármacos que disminuyen la glucosa llamados secretagogos de insulina pueden prevenir o retrasar el desarrollo de la diabetes mellitus tipo 2 y las complicaciones asociadas en los pacientes en riesgo de desarrollar diabetes mellitus tipo 2?

Antecedentes

Los secretagogos de insulina se utilizan ampliamente para tratar a los pacientes con diabetes mellitus tipo 2. Los secretagogos de insulina se pueden fraccionar en dos clases principales de fármacos para la disminución de la glucosa, a saber, las sulfonilureas (p.ej. glibenclamida / gliburida, glipizida y gliclazida) y los análogos de la meglitinida (nateglinida y repaglinida). Los secretagogos de insulina disminuyen la glucemia al estimular la secreción de insulina en el cuerpo, por lo que aumentan los niveles de insulina en sangre. A menudo se dice que las personas con niveles de glucosa moderadamente elevados tienen un mayor riesgo de desarrollar diabetes tipo 2 (a menudo llamado "prediabetes"). Por lo tanto, a los pacientes con niveles de glucosa moderadamente elevados se les recomienda con frecuencia aumentar el ejercicio y reducir la ingesta de calorías (intervenciones de cambio de comportamientos o en el "estilo de vida") para prevenir el desarrollo de diabetes tipo 2. Actualmente no se sabe si los secretagogos de insulina se deben prescribir a los pacientes con niveles elevados de glucosa en sangre que no cumplen los criterios diagnósticos para presentar diabetes mellitus tipo 2. Se deseaba determinar si los secretagogos de insulina podrían prevenir o retrasarían el desarrollo de la diabetes mellitus tipo 2 en los pacientes con niveles de glucosa moderadamente elevados. Además, se deseaba analizar los efectos de los secretagogos de insulina en resultados importantes para los pacientes como las complicaciones de la diabetes (por ejemplo, la enfermedad renal y ocular, los ataques cardíacos, los accidentes cerebrovasculares), la muerte por cualquier causa, la calidad de vida relacionada con la salud y los efectos secundarios de los fármacos.

Características de los estudios

En la bibliografía médica y los registros de los ensayos en curso se buscaron ensayos controlados aleatorios de al menos 12 semanas de duración que compararan los secretagogos de insulina con otro fármaco para la disminución de la glucosa, placebo o ninguna intervención. Los ensayos controlados aleatorios son estudios clínicos que asignan al azar a las personas a uno de dos o más grupos para poder comparar de forma directa los efectos de diferentes intervenciones. Los participantes incluidos en los estudios tenían que tener niveles de glucosa mayores de los que se consideran normales, pero por debajo de los niveles de glucosa que se utilizan para diagnosticar la diabetes mellitus tipo 2. Se combinaron los hallazgos de varios estudios para responder a la pregunta de la revisión. Se encontraron seis ensayos controlados aleatorios. Se incluyó un total de 10 018 participantes. La duración de las intervenciones varió de seis meses a cinco años.

Estas pruebas se actualizaron hasta abril de 2016.

Resultados clave

Pocos participantes murieron después del tratamiento con sulfonilureas. Las sulfonilureas (la mayoría de las pruebas estuvieron disponibles para la glimepirida) no redujeron el riesgo de desarrollar diabetes mellitus tipo 2 en comparación con placebo. Ningún estudio de sulfonilureas informó sobre efectos secundarios graves, ataques cardíacos no mortales, accidente cerebrovascular no mortal, insuficiencia cardíaca, calidad de vida relacionada con la salud o efectos socioeconómicos.

Solamente un estudio informó datos sobre un análogo de la meglitinida (nateglinida). Este gran estudio contribuyó con el 95% de todos los participantes de esta revisión. No es posible establecer pruebas firmes sobre los resultados muerte por cualquier causa, riesgo de desarrollar diabetes mellitus tipo 2 o efectos secundarios graves. Este estudio no informó sobre la calidad de vida relacionada con la salud ni los efectos socioeconómicos.

Los estudios futuros deben investigar resultados importantes para los pacientes y, especialmente, los efectos secundarios de los fármacos, porque no se sabe con seguridad si la "prediabetes" es sólo una afección arbitrariamente definida por una medición de laboratorio o en realidad es un factor de riesgo real de diabetes mellitus tipo 2, que podría ser tratable.

Calidad de la evidencia

Todos los ensayos incluidos tuvieron deficiencias en la forma en la que se realizaron o en cómo informaron los ítems clave. En las comparaciones individuales el número de participantes fue pequeño, lo que dio lugar a un alto riesgo de errores aleatorios (influencia del azar).

Authors' conclusions

Implications for practice

There is insufficient evidence to demonstrate whether insulin secretagogues compared with pharmacological glucose‐lowering interventions, placebo, behaviour‐changing interventions or no intervention influence the risk of type 2 diabetes mellitus and its associated complications. The evidence on patient‐important outcomes such as mortality, macro‐ and microvascular complications is sporadic and sparsely addressed in the existing trials. We are currently not able to provide a reliable benefit:risk ratio for this type of intervention in preventing or delaying the development of type 2 diabetes mellitus.

Implications for research

Even though it remains to be clarified whether there are any beneficial or harmful effects of insulin secretagogues in people at high risk of type 2 diabetes mellitus, no ongoing trials are investigating this issue. If new randomised controlled trials are to be performed in the future, they should focus on patient‐important outcomes. In addition, future trials should be reported according to the CONSORT (CONsolidated Standards of Reporting Trials) statement.

Summary of findings

Open in table viewer
Summary of findings for the main comparison. Summary of findings (sulphonylureas)

Insulin secretagogues for prevention or delay of type 2 diabetes mellitus and its associated complications in persons at risk for the development of type 2 diabetes mellitus

Population: people at risk for the development of type 2 diabetes mellitus

Settings: outpatient

Intervention: sulphonylureas (data available for glimepiride only)

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(trials)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo

Glimepiride

All‐cause mortality

Follow‐up: mean 3.7 years

See comment

See comment

See comment

274
(1)

⊕⊝⊝⊝
very lowa

5/136 (3.7%) participants in the glimepiride group versus 2/138 (1.4%) in the placebo group

Incidence of type 2 diabetes mellitus

Measured as 2 consecutive fasting blood glucose values ≥ 6.1 mmol/L (NANSY 2011b) or no definition provided (Eriksson 2006)
Follow‐up: 6 months and a mean of 3.7 years

361 per 1000

271 per 1000 (195 to 376)

RR 0.75 (0.54 to 1.04)

307
(2)

⊕⊝⊝⊝
very lowc

Serious adverse events

See comment

See comment

See comment

See comment

See comment

Not reported

Cardiovascular mortality

Follow‐up: mean 3.7 years

See comment

See comment

See comment

274
(1)

⊕⊝⊝⊝
very lowa

1/136 (0.7%) participants died due to cardiovascular disease in the sulphonylurea monotherapy group and 2/138 (1.4%) participants died in the placebo group

Non‐fatal myocardial infarction, non‐fatal stroke, congestive heart failure

See comment

See comment

See comment

See comment

See comment

Not reported

Health‐related quality of life

See comment

See comment

See comment

See comment

See comment

Not reported

Socioeconomic effects

See comment

See comment

See comment

See comment

See comment

Not reported

*The basis for the assumed risk (e.g. the median control group risk across trials) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; RR: risk ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

*Assumed risk was derived from the event rates in the comparator groups
aDowngraded by three levels because of serious imprecision and possible publication bias
bDiagnostic criterion for trial entry was impaired fasting glucose in the NANSY trial (baseline glycosylated haemoglobin A1c was 4.9% for both groups) and impaired glucose tolerance in Eriksson 2006. In the NANSY trial participants took glimepiride on the days when glycaemic variables were measured
cDowngraded by three levels because of indirectness, serious imprecision and possible publication bias. Trial sequential analysis showed that only 4.5% of the diversity‐adjusted information size was accrued so far to detect or reject a 10% relative risk reduction

Open in table viewer
Summary of findings 2. Summary of findings (meglitinide analogues)

Insulin secretagogues for prevention or delay of type 2 diabetes mellitus and its associated complications in persons at risk for the development of type 2 diabetes mellitus

Population: people at risk for the development of type 2 diabetes mellitus

Settings: outpatients

Intervention: meglitinide analogues (nateglinide)

Comparison: placebo

Outcomes

Placebo

Nateglinide

Relative effect
(95% CI)

No of participants
(trials)

Quality of the evidence
(GRADE)

Comments

All‐cause mortality

Follow‐up: median 6.5 years

See comment

See comment

See comment

9306 (1)

⊕⊕⊕⊝
moderatea

310/4645 (6.7%) participants died in the nateglinide group versus 312/4661 (6.7%) participants in the placebo group. Vital status was available for 95.7% of participants at the end of follow‐up. The HR was 1.00; 95% CI 0.85 to 1.17; P = 0.98

Incidence of type 2 diabetes mellitus

Defined as: fasting plasma glucose ≥ 7.0 mmol/L (126 mg/dL) or a 2‐hour blood glucose after a glucose‐load test ≥ 11.1 mmol/L (200 mg/dL) or by an adjudication committeeb

Follow‐up: median 5 years

See comment

See comment

See comment

9306 (1)

⊕⊕⊕⊝
moderatea

Type 2 diabetes mellitus developed in 1674/4645 (36.0%) participants in the nateglinide group and in 1580/4661 (33.9%) in the placebo group. The HR was 1.07; 95% CI 1.00 to 1.15; P = 0.05

Serious adverse events

Follow‐up: median 5 years

See comment

See comment

See comment

9306 (1)

⊕⊕⊕⊝
moderatea

The number of participants who experienced a serious adverse events was 2066/4602 (44.9%) participants in the nateglinide group versus 2089/4599 (45.6%) participants in the placebo group

Cardiovascular mortality

Follow‐up: 6.5 years

See comment

See comment

See comment

9306 (1)

⊕⊕⊕⊝
moderatea

The number of participants who died due to cardiovascular disease was 126/4645 (2.7%) participants in the nateglinide group versus 118/4661 (2.5%) participants in the placebo group. The HR was 1.07; 95% CI 0.83 to 1.38; P = 0.60

(a) Non‐fatal myocardial infarction

(b) Non‐fatal stroke

(c) Congestive heart failure

Follow‐up: median 6.3 years

See comment

See comment

See comment

9306 (1)

(a), (b), (c):

⊕⊕⊕⊝
moderatea

(a) The number of participants who experienced a non‐fatal myocardial infarction during the trial was 116/4645 (2.5%) participants in the nateglinide group versus 122/4661 (2.6%) participants in the placebo group

(b) The number of participants who experienced a non‐fatal stroke during the trial was 100/4645 (2.2%) participants in the nateglinide group versus 110/4661 (2.4%) participants in the placebo group

(c) The number of participants developing congestive heart failure was not reported. However, the number of participants hospitalised for congestive heart failure was 85/4645 (1.8%) participants in the nateglinide group versus 100/4661 (2.1%) participants in the placebo group. The HR was 0.85; 95% CI 0.64 to 1.14; P = 0.27

Health‐related quality of life

See comment

See comment

See comment

See comment

See comment

Not reported

Socioeconomic effects

See comment

See comment

See comment

9306 (1)

See comment

One trial specified the assessment of health economics (NAVIGATOR 2010). However, trial authors did not provide data

*The basis for the assumed risk (e.g. the median control group risk across trials) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; HR: hazard ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

aDowngraded by one level because of imprecision, high risk of selective reporting and possible publication bias (see Appendix 18)
bDiagnostic criterion for the NAVIGATOR trial entry was impaired glucose tolerance; baseline glycosylated haemoglobin A1c was 5.8% for both groups. Progression to diabetes was confirmed by laboratory measurements in 1587 participants in the nateglinide group (34.2%) and 1495 participants in the placebo group (32.1%). Progression to diabetes was determined by the adjudication committee in the case of 87 participants assigned to nateglinide (1.9%) and 85 assigned to placebo (1.8%)

Background

Description of the condition

'Prediabetes', 'borderline diabetes', the 'prediabetic stage', 'high risk of diabetes', 'dysglycaemia' or 'intermediate hyperglycaemia' are often characterised by various measurements of elevated blood glucose concentrations, such as isolated impaired fasting glucose (IFG), isolated impaired glucose tolerance (IGT), isolated elevated glycosylated haemoglobin A1c (HbA1c) or combinations thereof (WHO/IDF 2006). These elevated blood glucose levels, which are indicative of hyperglycaemia, are too high to be considered normal but are below the diagnostic threshold for type 2 diabetes mellitus (T2DM). Because of this continuous glycaemic spectrum from the normal to the diabetic stage, a sound evidence base is needed so that glycaemic thresholds for people at high risk of diabetes can be defined. The different terms used to describe various stages of hyperglycaemia may give rise to differing emotional reactions in affected persons. For example, a person told s/he has 'prediabetes' may take this to imply that diabetes is unavoidable, whereas someone told they are at (high) risk of diabetes may take this as meaning that they may possibly be able to avoid the disease altogether. In addition to the disputable construct of intermediate health states termed 'prediseases' (Viera 2011), many people may associate the label 'prediabetes' with dire consequences. Alternatively, any diagnosis of 'prediabetes' may be an opportunity to review, for example, eating habits and physical activity levels, thus enabling affected individuals to actively change their way of life.

The American Diabetes Association (ADA) and the World Health Organization (WHO) have established the criteria that are most commonly used today to define people with a high risk of developing T2DM. IGT was the first glycaemic measurement used by the US National Diabetes Data Group to define the prediabetic stage (NDDG 1979). It is based on the measurement of plasma glucose 2 hours after ingestion of 75 g of glucose (glucose load). The dysglycaemic range is defined as a plasma glucose level between 7.8 and 11.1 mmol/L (140 and 200 mg/dL) 2 hours after the glucose load. Studies indicate that IGT is caused by insulin resistance and defective insulin secretion (Abdul‐Ghani 2006; Jensen 2002). In 1997, the ADA, and later the WHO, introduced the IFG concept to define 'prediabetes' and intermediate hyperglycaemia (ADA 1997; WHO 1999). The initial definition of IFG was a blood glucose level of 6.1 to 6.9 mmol/L (110 to 125 mg/dL). Later, the ADA reduced the lower threshold for defining IFG to 5.6 mmol/L (100 mg/dL) (ADA 2003). However, the WHO did not endorse this lower cut‐off point for IFG for the definition of 'prediabetes' (WHO/IDF 2006). IFG seems to be associated with β‐cell dysfunction (impaired insulin secretion) and an increase in the hepatic glucose output (DeFronzo 1989). More recently, HbA1c levels have been used to identify people at high risk of developing T2DM. In 2009, the International Expert Committee (IEC) suggested that HbA1c levels ranging from 6.0% to 6.4% can be used to identify people at high risk of T2DM (IEC 2009). Shortly afterwards, the ADA redefined this HbA1c range as 5.7% to 6.4% (ADA 2010). Unlike IFG and IGT, HbA1c levels reflect longer‐term glycaemic control (i.e. a person's blood glucose levels during the preceding two to three months) (IEC 2009).

The International Diabetes Federation (IDF) estimated that, in 2010, the prevalence of IGT was 343 million people, and this is predicted to increase to 471 million people by 2035 (IDF 2013). Studies have shown poor correlations between HbA1c levels and IFG/IGT (Gosmanov 2014; Selvin 2011). Notably, the various glycaemic tests do not seem to identify the same people as there is imperfect overlap among the glycaemic modalities available to define dysglycaemia (Gosmanov 2014; Selvin 2011). A person's risk of progressing to T2DM depends on the diagnostic criteria used to identify that risk. Some people with dysglycaemia will never develop T2DM, and some people will return to normoglycaemia. IGT is often accepted as the best glycaemic variable predicting the risk of progression to T2DM (Morris 2013). However, studies indicate that less than half of the people defined as 'prediabetic' by means of IGT will develop T2DM in the following 10 years (Morris 2013). Both IFG and HbA1c levels are thought to predict a different risk spectrum for developing T2DM (Cheng 2006; Morris 2013). Most importantly, dysglycaemia is commonly an asymptomatic condition and, naturally, often remains 'undiagnosed' (CDC 2015).

It has yet to be clarified whether or not any particular intervention, especially glucose‐lowering drugs, should be recommended for people at risk for T2DM (Yudkin 2014). Trials have indicated that the progression to T2DM is reduced, or possibly just delayed, with behavioural interventions (increased physical activity, dietary changes or both) (Diabetes Prevention Program 2002; Diabetes Prevention Program FU 2009; Finnish Diabetes Prevention Study Group 2001). A recent meta‐analysis of 22 trials with interventions that changed behaviour in people at high risk of T2DM concluded that the effect of these interventions on longer‐term diabetes prevention is not clear (Dunkley 2014). Hence, more research is needed to establish optimal strategies for reducing the risk of T2DM with behavioural approaches (Dunkley 2014).

International diabetes associations and clinicians do not generally accept the prescription of pharmacological glucose‐lowering interventions for the prevention of T2DM. Several groups of pharmacological glucose‐lowering interventions have been investigated in people at risk of T2DM. Some findings indicate that the progression to T2DM is reduced or may only be delayed by such agents (Diabetes Prevention Program 2002; Diabetes Prevention Program FU 2009). However, the ADA recommends metformin in people at risk of T2DM with a body mass index (BMI) above 35 kg/m² who are aged less than 60 years, and women with prior gestational diabetes mellitus (ADA 2015).

Description of the intervention

Since the introduction of the sulphonylureas in the 1950s, this class of glucose‐lowering intervention has been a mainstay in the treatment of people with T2DM. The first of these agents to be introduced to the market were first‐generation sulphonylureas (acetohexamide, carbutamide, chlorpropamide, tolazamide and tolbutamide). Later the second‐ and third‐generations of sulphonylureas were introduced, and have now almost completely replaced the first‐generation sulphonylureas (Harrower 2000). Second‐generation sulphonylureas (e.g. glibenclamide (in the US: glybyride), glipizide and gliclazide) and third‐generation sulphonylureas (gliclazide modified release (MR), glipizide gastrointestinal therapeutic system (GITS) and glimepiride) are thought to have a better safety profile than first‐generation agents (Harrower 2000).

Another class of insulin secretagogues, meglitinide analogues, was introduced to the market in the 1990s (Black 2007). Two meglitinide analogues are currently available for clinical use in people with T2DM in Europe and the USA: repaglinide and nateglinide (ADA 2015). Another meglitinide analogue, mitiglinide, is approved for clinical use in people with T2DM in Japan (Phillippe 2013).

Sulphonylureas and meglitinide analogues can be prescribed as monotherapy in people with T2DM, usually if diet and exercise alone are not sufficient in controlling T2DM or if metformin is not tolerated or contraindicated. However, they can also be combined with other existing glucose‐lowering interventions (ADA 2015).

All sulphonylureas and meglitinide analogues are orally administered. The daily dose recommended in people with T2DM varies according to the different types of sulphonylurea or meglitinide analogue. Due to the varying half‐life of the sulphonylureas, some have to be taken once daily and others are taken twice or three times daily. The meglitinide analogues have a short half‐life and are administered in relation to meals (Blickle 2006).

For glimepiride, the recommended dose is up to 4 mg/day (Drugs.com 2016a). For gliclazide, the recommended starting dose is between 40 mg/day and 80 mg/day, but can be increased to 320 mg/day (Drugs.com 2016b).

Adverse effects of the intervention

All sulphonylureas and meglitinide analogues have the potential to cause hypoglycaemia. The risk of hypoglycaemia varies according to the type of sulphonylurea. Some sulphonylureas, such as glibenclamide, are more prone to causing prolonged hypoglycaemia than others (Harrower 2000). The risk of hypoglycaemia appears more pronounced for the first‐generation sulphonylureas compared with newer generations (Harrower 2000). Because of their short half‐life, meglitinide analogues do not cause prolonged hypoglycaemia (Scott 2012).

In 1976, the University Group Diabetes Program (UGDP) suggested that the sulphonylurea tolbutamide was associated with adverse cardiovascular effects compared with placebo and insulin in people with T2DM (UGDP 1976). More recent randomised clinical trials (RCTs) have not shown a significant increased risk of cardiovascular disease with sulphonylureas compared with other glucose‐lowering interventions in people with T2DM (ADOPT 2006; UKPDS 33 1998). Several observational studies have indicated increased risks of mortality and cardiovascular disease with sulphonylurea monotherapy compared with metformin monotherapy in people with T2DM (Roumie 2012; Schramm 2011). However, risk may vary among the different sulphonylureas (Pantalone 2012; Schramm 2011). No association between the use of meglitinide analogues and an increase in cardiovascular risk was reported in one observational study (Schramm 2011); however, some confounding factors may not have been detected in this study (Deeks 2003).

A substudy of the UK Prospective Diabetes Study (UKPDS) showed that, in participants receiving a sulphonylurea, the early addition of metformin was associated with an increased risk of mortality compared with continuation on a sulphonylurea alone (UKPDS 34 1998). The debate about the potential adverse effects of this combination therapy is ongoing.

How the intervention might work

The primary mechanism of action of the sulphonylureas and meglitinide analogues is to stimulate insulin release from the insulin‐secreting pancreatic β‐cells; hence, the term 'insulin secretagogues'. Sulphonylureas and meglitinide analogues increase pancreatic insulin release by closing the potassium‐sensitive adenosine triphosphate channels in β‐cells (Harrower 2000; Scott 2012).

The pharmacokinetic and pharmacodynamic properties of different insulin secretagogues vary, mainly as a result of differing binding affinities for sulphonylurea receptors on the β‐cell, and differing half‐lives. The meglitinide analogues exhibit a fast association/dissociation to/from the sulphonylurea receptor, and therefore mimic physiological early‐phase insulin secretion. With regard to sulphonylureas, half lives range from around 5 hours (glimepiride) to 36 hours (chlorpropamide) (McCall 2001). The half‐life of the meglitinide analogues is relatively short (1 to 1.5 hours) (Scott 2012).

It has been hypothesised that postprandial hyperglycaemia rather than fasting glucose levels is associated with cardiovascular disease (Meigs 2002). Due to the short‐acting mechanism of action of the meglitinide analogues, which primarily reduces postprandial hyperglycaemia, it has been hypothesised that meglitinide analogues could be effective in decreasing the risk of T2DM and cardiovascular disease in individuals with IGT (NAVIGATOR 2010). However, a large‐scale RCT failed to show any beneficial effect of nateglinide compared with placebo in individuals with IGT and established cardiovascular disease (or cardiovascular risk factors) after five years of intervention (NAVIGATOR 2010).

The glucagon‐like peptide‐1 (GLP‐1) and the dipeptidyl peptidase‐4 (DPP‐4) inhibitors stimulate insulin secretion by a glucose‐dependent mechanism, and inhibit glucagon secretion. These drugs increase insulin secretion indirectly by means of GLP‐1 and the glucose‐dependent insulinotropic polypeptide, two hormones that are secreted by endocrine cells located in the epithelium of the small intestine. The effects of the DPP‐4 inhibitors and the GLP‐1 receptor agonists in individuals at increased risk of developing T2DM will be evaluated in a separate Cochrane review (Hemmingsen 2016a).

Why it is important to do this review

This review is part of a series of reviews on interventions that may prevent or delay the development of T2DM and its associated complications in persons at increased risk of T2DM, which is funded by the WHO (Hemmingsen 2016a; Hemmingsen 2016b). The protocol for this review has previously been published (Hemmingsen 2016c). There has been an increased focus on the prevention or delay of T2DM with non‐pharmacological interventions and glucose‐lowering medications. Currently, several trials are ongoing to clarify whether the progression from an at‐risk status to T2DM can be stopped or postponed with glucose‐lowering compounds (ClinicalTrials.gov). However, a more important issue for people with dysglycaemia is whether or not these interventions reduce the risk of death and the complications ‐ especially cardiovascular disease ‐ related to T2DM.

Objectives

To assess the effects of insulin secretagogues on the prevention or delay of T2DM and its associated complications in people with impaired glucose tolerance, impaired fasting blood glucose, moderately elevated glycosylated haemoglobin A1c (HbA1c) or any combination of these.

Methods

Criteria for considering studies for this review

Types of studies

We included RCTs in participants at increased risk of type 2 diabetes mellitus (T2DM) comparing a second‐ or third‐generation sulphonylurea or a meglitinide analogue with another pharmacological glucose‐lowering interventions, behaviour changing intervention, placebo or no intervention, with a duration of 12 weeks or more (Hemmingsen 2016c).

Types of participants

We included individuals without a diagnosis of diabetes who were at increased risk of T2DM.

We included trials in obese people or in participants with previous gestational diabetes, provided trial investigators stated that the participants had intermediate hyperglycaemia.

Diagnostic criteria for people at risk of developing T2DM

To be consistent with changes to the classification of, and diagnostic criteria for dysglycaemia (impaired fasting glucose (IFG), impaired glucose tolerance (IGT) or elevated glycosylated haemoglobin A1c (HbA1c)) that have occurred over the years, a diagnosis should have been established using the standard criteria valid at the trial start (e.g. ADA 1997; ADA 2010; NDDG 1979; or WHO 1999). Ideally, the diagnostic criteria used in each study should have been described. We used the trial authors' definition of risk, but we contacted trial authors for additional information, if necessary. As differences in the glycaemic measurements used to define risk may introduce substantial heterogeneity, we planned to subject the diagnostic criteria used to subgroup analysis.

Types of interventions

We included trials in which a fraction of the included participants were explicitly described as having intermediate hyperglycaemia. We contacted the investigators in order to obtain separate data on the participants with intermediate hyperglycaemia.

We included a trial even if one or more of our primary or secondary outcome measures were not reported in a publication. In this case, we contacted the corresponding author for supplementary data. If no additional data were available, we present data from this trial in a supplementary table. We also list information about trials with a duration of the intervention shorter than 12 weeks in Appendix 1.

We planned to investigate the following comparisons of insulin secretagogues versus all pharmacological glucose‐lowering interventions, behaviour‐changing interventions, placebo or no intervention.

Intervention

(a) Second‐ or third‐generation sulphonylureas as monotherapy.

(b) Second‐ or third‐generation sulphonylureas as a part of combination therapy.

(c) Meglitinide analogues as monotherapy.

(d) Meglitinide analogues as a part of combination therapy.

Comparator

  • Any pharmacological glucose‐lowering intervention (e.g. acarbose, metformin, sodium–glucose cotransporter 2 inhibitors) compared with (a) or (c).

  • Any pharmacological glucose‐lowering agent (e.g. acarbose, metformin, sodium–glucose cotransporter 2 inhibitors) compared with (b) or (d) if this glucose‐lowering agent was the same in both the intervention and comparator groups (e.g. meglitinide analogue + metformin versus metformin).

  • Behaviour‐changing interventions (e.g. diet, exercise, diet and exercise) compared with (a) or (c).

  • Placebo compared with (a) or (c).

  • No intervention compared with (a) or (c).

Other concomitant interventions (e.g. educational programmes or additional pharmacotherapy) had to be the same in both the intervention and comparator groups in order to establish a fair comparison.

Minimum duration of intervention

We included trials that investigated the intervention for a duration of 12 weeks or more.

Specific exclusion criteria

  • We excluded trials in people diagnosed with the 'metabolic syndrome' as this is a special population which is not representative of people with only intermediate hyperglycaemia. Also, the composite of risk indicators, such as elevated blood lipids, insulin resistance, obesity and high blood pressure, which is termed metabolic syndrome, is of doubtful clinical usefulness and uncertain distinct disease entity. However, if we identified trials investigating participants with any definition of the metabolic syndrome, we intended to summarise some basic trial information in an additional table.

  • We excluded trials evaluating participants with intermediate hyperglycaemia in combination with another condition (e.g. cystic fibrosis).

  • We excluded trials evaluating participants with intermediate hyperglycaemia due to other medical interventions (e.g. glucocorticoids).

Types of outcome measures

Primary outcomes

  • All‐cause mortality

  • Incidence of T2DM

  • Serious adverse events

Secondary outcomes

  • Cardiovascular mortality

  • Non‐fatal myocardial infarction

  • Non‐fatal stroke

  • Congestive heart failure

  • Amputation of lower extremity

  • Blindness or severe vision loss

  • End‐stage renal disease

  • Non‐serious adverse events

  • Hypoglycaemia

  • Health‐related quality of life

  • Time to progression to T2DM

  • Measures of blood glucose control

  • Socioeconomic effects

Method and timing of outcome measurement

  • All‐cause mortality: defined as death from any cause. Measured at any time of the intervention and during follow‐up.

  • Incidence of T2DM and time to progression to T2DM: defined according to diagnostic criteria valid at the time the diagnosis was established using the standard criteria valid at the time the trial commenced (e.g. ADA 2008; WHO 1998). If necessary, we used the trial authors' definition of T2DM. Measured at the end of the intervention and the end of follow‐up.

  • Serious adverse events: defined according to the International Conference on Harmonization Guidelines as any event that lead to death, was life‐threatening, required inpatient hospitalisation or prolongation of existing hospitalisation, resulted in persistent or significant disability; or any important medical event which may have jeopardised the participant or required intervention to prevent it (ICH 1997); or as reported in trials. Measured at any time of the intervention and during follow‐up.

  • Cardiovascular mortality, non‐fatal myocardial infarction, non‐fatal stroke, amputation of lower extremity, blindness or severe vision loss, congestive heart failure, hypoglycaemia (mild, moderate, severe/serious): defined as reported in trials. Measured at the end of the intervention and at the end of follow‐up.

  • End‐stage renal disease: defined as dialysis, renal transplantation or death due to renal disease. Measured at the end of the intervention and at the end of follow‐up.

  • Non‐serious adverse events: defined as the number of participants with any untoward medical occurrence not necessarily having a causal relationship with the intervention. Measured at any time of the intervention and during follow‐up.

  • Health‐related quality of life: defined as mental and physical health‐related quality of life, assessed separately or combined using a validated instrument such as Short‐Form 36. Measured at the end of the intervention and at the end of follow‐up.

  • Measures of blood glucose control: fasting blood glucose (FBG), blood glucose 2 hours after ingestion of 75 g glucose and HbA1c measurements. Measured at the end of the intervention and at the end of follow‐up.

  • Socioeconomic effects: for example costs of the intervention, absence from work, medication consumption. Measured at the end of the intervention and at the end of follow‐up.

Specification of key prognostic variables

  • Age

  • Gender

  • Equity issues (access to health care, social determinants)

  • Ethnicity

  • Hypertension

  • Cardiovascular disease

  • Obesity

  • Previous gestational diabetes

Summary of findings table

We present a 'Summary of findings' table to report the following outcomes, listed according to priority.

  1. All‐cause mortality.

  2. Incidence of T2DM.

  3. Serious adverse events.

  4. Cardiovascular mortality.

  5. Non‐fatal myocardial infarction/stroke and congestive heart failure.

  6. Health‐related quality of life.

  7. Socioeconomic effects.

Search methods for identification of studies

Electronic searches

We searched the following sources from inception to the specified date, and placed no restrictions on the language of publication.

  • Cochrane Central Register of Controlled Trials (CENTRAL) (4 April 2016).

  • MEDLINE (1946 to present) (4 April 2016).

  • Embase (1974 to 5 April 2016) (4 April 2016).

  • ClinicalTrials.gov (4 April 2016).

  • WHO International Clinical Trials Registry Platform (ICTRP) Search Portal (http://apps.who.int/trialsearch/) (4 April 2016).

We continuously applied a MEDLINE (via Ovid SP) email alert service, established by the Cochrane Metabolic and Endocrine Disorders (CMED) Group, to identify newly published trials using the same search strategy as described for MEDLINE (for details on search strategies, see Appendix 2). If we identified new trials for inclusion, we intended to evaluate them, incorporate the findings into our review and resubmit another review draft (Beller 2013).

If we had detected any additional key words of relevance during any of the electronic or other searches, we intended to modify the electronic search strategies to incorporate these terms.

We obtained evaluations of all relevant non‐English articles.

Searching other resources

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

As none of the existing insulin secretagogues is currently approved for the treatment of persons with intermediate hyperglycaemia we did not search databases of the regulatory agencies (European Medicines Agency, US Food and Drug Administration).

Data collection and analysis

Selection of studies

Two review authors (BH and DS) independently scanned the abstract or title, or both, of every record retrieved in order to determine which trials should be assessed further. We investigated the full‐text articles of all potentially relevant articles. We resolved discrepancies through consensus or by recourse to a third review author (BR). We prepared a flow diagram of the number of trials identified and excluded at each stage, in accordance with PRISMA guidelines (Liberati 2009).

Data extraction and management

For trials that fulfilled our inclusion criteria, two review authors (BH and DS) independently extracted outcome data. Key characteristics of participants and interventions were extracted by one author (BH) and checked by another (DS). We reported data on efficacy outcomes and adverse events using standard data extraction sheets from the CMED Group. We resolved disagreements by discussion or, if required, by consultation with a third review author (BR) (for details, see Characteristics of included studies; Table 1; Appendix 1; Appendix 3; Appendix 4; Appendix 5; Appendix 6; Appendix 7; Appendix 8; Appendix 9; Appendix 10; Appendix 11; Appendix 12; Appendix 13; Appendix 14; Appendix 15).

Open in table viewer
Table 1. Overview of trial populations

Intervention(s) and comparator(s)

Description of power and sample size calculation

Screened/eligible
(N)

Randomised
(N)

ITT
(N)

Analysed
(N)

Finishing trial
(N)

Randomised finishing trial
(%)

Follow‐up
(extended follow‐up)a

Eriksson 2006

I: glipizide 2.5 mg

b

‐c

17

16

16

6 months (18 months)

C: placebo

c

17

17

16

total:

37

34

33

32

NANSY 2011

I: glimepiride 1.0 mg

Quote: "...assuming 3% conversion rate per year and 33% reduction of diabetes development with 5% significance and 80% statistical power"

d

136

136

e

5 years or until diabetes developed, average follow‐up period 3.7 years

C: placebo

d

138

138

e

total:

288d

274

274

203

74.1

NAVIGATOR 2010

I: nateglinide 60 mg, three times daily

Quote: "The sample size calculation was therefore based on a 'subadditivity / 75% additivity of effects' approach, assuming an effect size of 32% on cardiovascular outcome of the two drugs in combination. The treatment discontinuation rate was assumed to be 30% over five years, corresponding to approximately 6.9% per annum. While patients on treatment were assumed to have the full effect (i.e. 20% reduction of hazard rate if in the monotherapy group), it was assumed that patients who discontinued treatment would have only ¼ of the treatment effect remaining as carry‐over effect. Furthermore, it was expected that 75% of the patients who discontinued treatment could be followed up for events. The remaining 25% would comprise patients completely lost to follow‐up, patients who die (without reaching a primary endpoint), and those for whom events are unintentionally not reported by the investigator. Based on these assumptions, a total of 9152 patients will provide 90% testwise power to detect a treatment difference in the extended cardiovascular endpoint"

43 502

4748

4645

4645

3726

78.5

Quote: "The median follow‐up time for data on vital status was 6.5 years, and the median follow‐up times for data on the diabetes, extended cardiovascular, and core cardiovascular outcomes were 5.0, 6.3, and 6.4 years, respectively"f

C: placebo

4770

4661

4661

3747

78.6

total:

9518

9306

9306

7473

78.5

Osei 2004

I: GITS 5 mg

9

9

9

24 months (26 months)

C: placebo

9

9

9

total:

18

18

18

Page 1993

I: gliclazide 40 mg twice daily

6

6

6

6

100

6 months (7 months)

C1: placebo

8

7

7

7

87.5

C2: diet + exercise

23

18

18

18

78.2

total:

37

31

31

31

83.8

Papoz 1978

I1: glibenclamide 2.0 mg twice daily + metformin 850 mg twice daily

29

22

22

22

75.9

2 years (2 years)

I2: glibenclamide 2.0 mg twice daily + placebo

28

22

22

22

78.6

C1: placebo + metformin 850 mg twice daily

30

23

23

23

76.7

C2: placebo

33

19

19

19

57.6

total:

120

86

86

86

71.7

Grand total

All interventionsh

4820

3792

All comparatorsh

4873

3830

All interventions and comparatorsi

10,018

7825 j

‐ denotes not reported

aFollow‐up under randomised conditions until end of trial or, if not available, duration of intervention; extended follow‐up refers to follow‐up of participants once the original study was terminated as specified in the power calculation
bParticpants identified through screening of another trial (Botnia Study 1996). Quote: "The subjects included in the present study represented the first consecutive 37 subjects who maintained their IGT status on repeated OGTT testing during 1 year"
cThe investigators described that they randomised 37 participants, and three dropped out shortly after. However, they do not describe how these three participants were allocated, but only describe that after the three participants had left 17 were allocated to each intervention group
dThe investigators described that 14 randomised participants withdrew before the first occasion to establish the conversion to type 2 diabetes mellitus. All except one dropped out for administrative reasons. However, it was not specified to which intervention group these participants were allocated
e71 individuals interrupted participation prematurely, however it was not described to which groups they belonged
fThe trial was predefined to stop and the final analysis performed when 1374 participants have had an adjudication committee confirmed extended cardiovascular endpoint
hNot all trials described the number of participants randomised to each intervention group
iTwo trials did not report the number of randomised participants per intervention group. Therefore, numbers do not add up accurately
jNot all trials reported the number of participants finishing the trial

C: comparator; GITS: glipizide gastrointestinal therapeutic system; I: intervention; ITT: intention‐to‐treat; NANSY: The Nepi ANtidiabetes StudY

We planned to include information about potentially relevant ongoing trials, including the trial identifier, in a table of characteristics of ongoing studies.

For each included trial we tried to retrieve the protocol. If not available from a search of the databases, reference screening or Internet searches, we asked authors to provide a copy of the protocol. We entered predefined outcomes in a 'Matrix of trial endpoint (publications and trial documents)' (see Appendix 7).

We emailed all authors of the included trials to enquire whether they were willing to answer questions regarding their trials. We present the results of this survey in Appendix 16. We sought relevant missing information on the trials from the primary author(s) of the articles, if possible.

Dealing with duplicate and companion publications

In the event of duplicate publications, companion documents or multiple reports of a primary trial, we maximised the information by collating all available data and used the most complete data set aggregated across all known publications. We list duplicate publications, companion documents or multiple reports of a primary trial as secondary references under the primary reference of the included or excluded trial.

Assessment of risk of bias in included studies

Two review authors (BH and DS) independently assessed the risk of bias of each included trial. We resolved any disagreements by consensus, or by consultation with a third review author (BR). If adequate information was not available from the trial publication, trial protocol or both, we contacted trial authors for missing data on 'Risk of bias' items.

We used the Cochrane 'Risk of bias' assessment tool (Higgins 2011a; Higgins 2011b) and judged 'Risk of bias' criteria as being 'low', 'high', or 'unclear', evaluating individual bias items as described in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011a).

Random sequence generation (selection bias due to inadequate generation of a randomised sequence) ‐ assessment at trial level

We assessed for each included trial whether the method used to generate the allocation sequence was described in sufficient detail to allow an assessment of whether it should produce comparable groups.

  • Low risk of bias: sequence generation was achieved using computer random number generation or a random number table. Drawing of lots, tossing a coin, shuffling cards or envelopes and throwing dice were adequate if performed by an independent person not otherwise involved in the trial. Use of the minimisation technique was considered as equivalent to being random.

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

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

Allocation concealment (selection bias due to inadequate concealment of allocations prior to assignment) ‐ assessment at trial level

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

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

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

  • High risk of bias: use of: an open random allocation schedule (e.g. a list of random numbers); assignment envelopes without appropriate safeguards; alternation or rotation; date of birth; case record number; any other explicitly unconcealed procedure. We excluded such trials from our review.

We also evaluated trial baseline data so as to incorporate an assessment of baseline imbalance into the 'Risk of bias' judgement for selection bias (Corbett 2014; Egbewale 2014; Riley 2013). Chance imbalances might also affect judgements on the risk of attrition bias. In case of unadjusted analyses we distinguished between trials rated as at low risk of bias on the basis of both randomisation methods and baseline similarity, and trials rated as at low risk of bias on the basis of baseline similarity alone (Corbett 2014). We reclassified judgements of unclear, low or high risk of selection bias as specified in Appendix 15.

Blinding of participants and study personnel (performance bias due to knowledge of the allocated interventions by participants and personnel during the trial) ‐ assessment at outcome level

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

  • Low risk of bias: blinding of participants and key study personnel ensured, and unlikely that the blinding could have been broken; no blinding or incomplete blinding, but the review authors judged that the outcome was not likely to be influenced by lack of blinding.

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

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

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

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

  • Low risk of bias: blinding of outcome assessment ensured, and unlikely that the blinding could have been broken; no blinding of outcome assessment, but the review authors judged that the outcome measurement was not likely to be influenced by lack of blinding.

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

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

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

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

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

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

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

Selective reporting (reporting bias due to selective outcome reporting) ‐ assessment at trial level

We assessed outcome reporting bias by integrating Appendix 7 (Matrix of trial endpoints (publications and trial documents) (Boutron 2014; Mathieu 2009) with Appendix 8 (High risk of outcome reporting bias according to ORBIT [Outcome Reporting Bias In Trials]) classification) (Kirkham 2010). This analysis formed the basis for the judgement of selective reporting.

  • Low risk of bias: the trial protocol was available and all of the trial’s prespecified (primary and secondary) outcomes that were of interest in the review have been reported in the prespecified way; the study protocol was not available but it was clear that the published reports included all expected outcomes (ORBIT classification).

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

  • High risk of bias: not all of the trial’s prespecified primary outcomes were reported; one or more primary outcomes was reported using measurements, analysis methods or subsets of the data (e.g. subscales) that were not prespecified; one or more reported primary outcome was not prespecified (unless clear justification for its reporting was provided, such as an unexpected adverse effect); one or more outcomes of interest in the review was reported incompletely so that they could not be entered in a meta‐analysis; the trial report failed to include results for a key outcome that would have been expected to have been reported for such a trial (ORBIT classification).

Other bias (bias due to problems not covered elsewhere) ‐ assessment at trial level

We assessed any other risk of bias that reflected other circumstances that may have threatened the validity of the trial.

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

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

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

We established a 'Risk of bias' graph and a 'Risk of bias' summary figure.

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

We defined the following outcomes as self‐reported.

  • Non‐serious adverse events.

  • Hypoglycaemia, if reported by participants.

  • Health‐related quality of life.

  • Blood glucose control, if measured by trial participants.

We required the following outcomes to be investigator‐assessed.

  • All‐cause mortality.

  • Incidence of T2DM.

  • Time to progression to T2DM.

  • Serious adverse events.

  • Cardiovascular mortality.

  • Non‐fatal myocardial infarction.

  • Non‐fatal stroke.

  • Congestive heart failure.

  • Amputation of lower extremity.

  • Blindness or severe vision loss.

  • End‐stage renal disease.

  • Hypoglycaemia, if measured by trial personnel.

  • Blood glucose control, if measured by trial personnel.

  • Socioeconomic effects.

Summary assessment of risk of bias

Risk of bias for a trial across outcomes: some 'Risk of bias' domains such as selection bias (sequence generation and allocation sequence concealment) may affect the risk of bias across all outcome measures in a trial. Otherwise, we did not perform a summary assessment of the risk of bias across all outcomes for a trial. If we identified a high risk of selection bias, we excluded the trial.

Risk of bias for an outcome within a trial and across domains: we assessed the risk of bias for an outcome measure including all of the entries relevant to that outcome (i.e. both trial‐level entries and outcome‐specific entries). We defined 'low' risk of bias as low risk of bias for all key domains, 'unclear' risk of bias as unclear risk of bias for one or more key domains, and 'high' risk of bias as high risk of bias for one or more key domains.

Risk of bias for an outcome across trials and across domains: these were the main summary assessments that we incorporated in our judgements about the quality of evidence in the 'Summary of findings' table(s). We defined 'low' risk of bias as most information coming from trials at low risk of bias, 'unclear' risk of bias as most information coming from trials at low or unclear risk of bias and 'high' risk of bias as a sufficient proportion of information coming from trials at high risk of bias.

Measures of treatment effect

When at least two trials were available for comparison of a given outcome, we expressed dichotomous data as risk ratios (RRs) with 95% confidence intervals (CIs) and with Trial Sequential Analysis (TSA)‐adjusted 95% CIs if the diversity‐adjusted required information size was not reached. We expressed continuous data reported using the same scale as mean differences (MDs) with 95% CIs and with TSA‐adjusted CIs if the diversity‐adjusted required information size was not reached. For trials addressing the same outcome but using different outcome measure scales, we intended to use standardised mean differences (SMDs) with 95% CIs. We planned to calculate time‐to‐event data as hazard ratios (HRs) with 95% CIs using the generic inverse variance method. Our preference would have been to use unadjusted HRs, as adjustment may have differed among the included trials. For outcomes meta‐analysed as SMDs and the generic inverse variance method, we were unable to conduct TSA and adjust the 95% CIs.

Some scales measuring health‐related quality of life increase in value with improved health‐related quality of life, whereas other scales decrease in value with improved health‐related quality of life. To adjust for this, we planned to multiply by –1 the scales that report better health‐related quality of life with decreasing values.

Unit of analysis issues

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

We planned to reanalyse cluster‐randomised trials that did not appropriately adjust for potential clustering of participants within clusters in their analyses. We intended to inflate the variance of the intervention effects using a design effect (DEFF). Calculation of a DEFF involves estimation of an intra‐cluster correlation (ICC). We planned to obtain estimates of ICCs through contact with authors, or by imputing them either using estimates from other included studies that report ICCs or using external estimates from empirical research (e.g. Bell 2013). We planned to examine the impact of clustering using sensitivity analyses.

Dealing with missing data

We attempted to obtain missing data from trial authors and carefully evaluated important numerical data such as numbers screened and randomised, as well as intention‐to‐treat (ITT), as‐treated and per‐protocol populations.

We investigated attrition rates (e.g. dropouts, losses to follow‐up, withdrawals) and critically appraised issues concerning missing data and imputation methods (e.g. last observation carried forward).

We converted standard errors and CIs to standard deviations (SDs) (Higgins 2011a). When no differences in means and SDs from baseline were reported, we used end of follow‐up values (Higgins 2011a). Where means and SDs for outcomes were not reported and we did not receive the information required from trial authors, we calculated the SDs from standard errors, if possible. Otherwise we planned to impute the values by assuming the SDs of the missing outcome to be the average of the SDs from the trials that reported this information.

We planned to investigate the impact of imputation on meta‐analyses by performing sensitivity analyses.

Assessment of heterogeneity

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

We investigated heterogeneity (inconsistency) by visually inspecting the forest plots and by using a standard Chi² test with a significance level of α = 0.1. In view of the low power of this test, we also considered the I² statistic, which quantifies inconsistency across trials to assess the impact of heterogeneity on the meta‐analysis (Higgins 2002; Higgins 2003), where an I² statistic ≥ 75% indicated a considerable level of heterogeneity (Higgins 2011a).

Assessment of reporting biases

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

Data synthesis

Unless good evidence showed homogeneous effects across trials, we primarily summarised data at low risk of bias using a random‐effects model (Wood 2008). We interpreted random‐effects meta‐analyses taking into consideration the whole distribution of effects, ideally by presenting a prediction interval (Higgins 2009). A prediction interval specifies a predicted range for the true treatment effect in an individual trial (Riley 2011). In addition, we performed statistical analyses according to the statistical guidelines contained in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011a).

TSA

In a single trial sparse data and interim analyses increase the risk of type I and type II errors. To avoid type I errors, group sequential monitoring boundaries are applied to decide whether a trial could be terminated early because of a sufficiently small P value (i.e. the cumulative Z‐curve crosses the monitoring boundaries) (Lan 1983). Likewise, before reaching the planned sample size of a trial, the trial may be stopped due to futility if the cumulative Z‐score crosses the futility monitoring boundaries (Higgins 2011a). Sequential monitoring boundaries for benefit, harm or futility can be applied to meta‐analyses as well (termed trial sequential monitoring boundaries) (Higgins 2010; Wetterslev 2008). In TSA, the addition of each trial in a cumulative meta‐analysis is regarded as an interim meta‐analysis and helps to clarify whether significance or futility is reached, or whether additional trials are needed (Wetterslev 2008).

TSA combines a calculation of the diversity‐adjusted required information size (cumulated meta‐analysis sample size to detect or reject a specific relative intervention effect) for meta‐analysis with the threshold of data associated with statistics. We performed TSA on all outcomes (Brok 2009; Pogue 1997; Wetterslev 2008).

The idea in TSA is that if the cumulative Z‐curve crosses the boundary for benefit or harm before a diversity‐adjusted required information size is reached, a sufficient level of evidence for the anticipated intervention effect has been reached with the assumed type I error and no further trials may be needed. If the cumulative Z‐curve crosses the boundary for futility before a diversity‐adjusted required information size is reached, the assumed intervention effect can be rejected with the assumed type II error and no further trials may be needed. If the Z‐curve does not cross any boundary, then there is insufficient evidence to reach a conclusion. To construct the trial sequential monitoring boundaries, the required information size is needed and is calculated as the least number of participants needed in a well‐powered single trial and subsequently adjusted for diversity among the included trials in the meta‐analysis (Brok 2009; Wetterslev 2008). We applied TSA as it decreases the risk of type I and II errors due to sparse data and multiple updating in a cumulative meta‐analysis, and it provides us with important information in order to estimate the risks of imprecision when the required information size is not reached. Additionally, TSA provides important information regarding the need for additional trials and the required information size of such trials (Wetterslev 2008).

We applied trial sequential monitoring boundaries according to an estimated clinically important effect. We based the required information size on an a priori effect corresponding to a 10% relative risk reduction (RRR) for beneficial effects of the interventions and a 30% relative risk increase for harmful effects of the interventions.

TSA for continuous outcomes was performed with MDs, by using trials applying the same scale to calculate the required sample size. For continuous outcomes we tested the evidence for the achieved differences in cumulative meta‐analyses.

For adjustment of heterogeneity of the required information size we used the diversity (D²) estimated in the meta‐analyses of included trials. When diversity was zero in a meta‐analysis, we performed a sensitivity analysis using an assumed diversity of 20% when future trials are included, possibly changing future heterogeneity among trials.

Quality of evidence

We presented the overall quality of the evidence for each outcome according to the GRADE approach, which takes into account issues relating not only to internal validity (risk of bias, inconsistency, imprecision, publication bias) but also to external validity, such as directness of results. Two review authors (BH and DS) independently rated the quality of evidence for each outcome. We present a summary of the evidence in the summary of findings Table for the main comparison. This provides key information about the best estimate of the magnitude of the effect, in relative terms and as absolute differences, for each relevant comparison of alternative management strategies, the numbers of participants and trials addressing each important outcome, and rates the overall confidence in effect estimates for each outcome. We created the 'Summary of findings' table on the basis of methods described in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011a) by means of the table editor in Review Manager (RevMan 2014), and include two appendices (Appendix 17; Appendix 18) providing checklists as guides to the consistency and reproducibility of GRADE assessments (Meader 2014) to help with the standardisation of the 'Summary of findings' tables. Alternatively, we would have used the GRADEproGDT software (GRADEproGDT 2015) and presented evidence profile tables as an appendix. We present results for the outcomes as described in the Types of outcome measures section. If meta‐analysis was not possible, we present the results in a narrative format in the 'Summary of findings' table. We justify all decisions to downgrade the quality using footnotes, and we make comments to aid the reader's understanding of the review where necessary.

Subgroup analysis and investigation of heterogeneity

We expected the following characteristics to introduce clinical heterogeneity, and planned to carry out subgroup analyses with investigation of interactions.

  • Type of sulphonylurea and type of meglitinide analogue.

  • Trials with long duration (two or more years) versus trials with short duration (less than two years).

  • Diagnostic criteria (IFG, IGT, moderately elevated HbA1c).

  • Age, depending on data.

  • Ethnicity, depending on data.

  • Comorbid conditions, such as hypertension, obesity, or both.

  • Participants with previous gestational diabetes mellitus.

Sensitivity analysis

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

  • Published trials.

  • Taking into account risk of bias, as specified in the 'Assessment of risk of bias in included studies' section.

  • Very long or large trials to establish the extent to which they dominate the results.

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

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

Results

Description of studies

For a detailed description of trials, see Table 1, Characteristics of included studies, Characteristics of excluded studies and Characteristics of ongoing studies.

Results of the search

The initial search of the databases identified 2262 records after duplicates were removed. We excluded most of the references on the basis of their titles and abstracts because they clearly did not meet the inclusion criteria (Figure 1). We evaluated 53 references further. After screening the full texts, six RCTs published in 16 records met our inclusion criteria. We excluded a total of 39 references after full‐text evaluation.


Trial flow diagram.

Trial flow diagram.

We identified no health technology assessment reports, systematic reviews or meta‐analyses focusing on sulphonylureas or meglitinide analogues in people at increased risk for the development of T2DM. However, four systematic reviews published in five records included a sulphonylurea or meglitinide analogue as a comparator in participants with intermediate hyperglycaemia (Anderson 2005; Bhardwaj 2010; Hopper 2011; Phung 2012; Van de Laar 2006). We evaluated all these systematic reviews but did not identify additional trials.

From the main publication of one of the included trials we identified an additional reference describing the same trial (Papoz 1978). We retrieved an additional trial protocol through an Internet search on the Nateglinide+Valsartan to Prevent or Delay Type 2 Diabetes Mellitus and Cardiovascular Complications (NAVIGATOR) trial (NAVIGATOR 2010).

We did not find any ongoing trials investigating our research question.

We sent all trial authors of the included trials a list of references and a request for information on additional trials of relevance. The trial authors did not provide additional trials or any supplementary information on our included trials.

Included studies

See Characteristics of included studies; Table 1 and Appendix 3; Appendix 4; Appendix 5; Appendix 6; Appendix 7; Appendix 8; Appendix 9; Appendix 10; Appendix 11; Appendix 12; Appendix 13; and Appendix 14.

Overview of trial populations

Only one trial reported the number of participants screened (NAVIGATOR 2010). Two trials did not report the number of participants randomised to each intervention group upon trial initiation (Eriksson 2006; NANSY 2011). A total of 4791 participants were randomised to a second‐ or third‐generation sulphonylurea or meglitinide analogue as monotherapy and 29 participants were randomised to a second‐generation sulphonylurea plus metformin (Papoz 1978). Three trials had a second‐generation sulphonylurea in the intervention arm (Eriksson 2006; Page 1993; Papoz 1978), two trials investigated a third‐generation sulphonylurea (NANSY 2011; Osei 2004) and one trial a meglitinide analogue, nateglinide (NAVIGATOR 2010). A total of 4873 participants were randomised to a comparator group; 4820 participants were randomised to placebo (Eriksson 2006; NANSY 2011; NAVIGATOR 2010; Osei 2004; Page 1993; Papoz 1978), 23 participants to diet and exercise (Page 1993) and 30 participants to metformin monotherapy (Papoz 1978).

Two publications provided information about sample size and power calculations (NANSY 2011; NAVIGATOR 2010).

Trial design

All the included trials were parallel randomised controlled clinical trials (Eriksson 2006; NANSY 2011; NAVIGATOR 2010; Osei 2004; Page 1993; Papoz 1978). All trials had performed blinding of the participants and investigators. However, in one trial only the comparison of gliclazide versus placebo was blinded, whereas the comparison of gliclazide versus diet and exercise was not (Page 1993). One RCT had a factorial design (NAVIGATOR 2010). The NAVIGATOR trial assigned participants to receive valsartan plus placebo, nateglinide plus placebo, nateglinide plus valsartan, or placebo plus placebo. Tests of interaction for the factorial allocation were provided for progression to T2DM and the two primary cardiovascular outcomes (NAVIGATOR 2010). None of the tests of interaction showed a relevant impact of the factorial design (NAVIGATOR 2010).

The duration of the intervention in the included trials varied from six months to five years. In four trials the duration of the intervention was two years or more (NANSY 2011; NAVIGATOR 2010; Osei 2004; Papoz 1978). Two trials included an extended follow‐up period after the intervention period had stopped (Eriksson 2006; Page 1993). One trial had a duration of intervention of six months, and thereafter a 12 month follow‐up period (Eriksson 2006). Another trial followed the participants one month after the end of the intervention period (i.e. for a total of seven months) (Page 1993).

The number of participants varied from 18 (Osei 2004) to 9518 (NAVIGATOR 2010). One trial contributed 97.4% of all randomised participants (NAVIGATOR 2010). Two trials were multicentre trials (NANSY 2011; NAVIGATOR 2010), three trials were single‐centre trials (Eriksson 2006; Osei 2004; Papoz 1978) and one trial did not provide any description of the number of centres involved (Page 1993).

All trials were performed in outpatient settings.

Five of the included trials stated that they had received grants from a pharmaceutical company (Eriksson 2006; NANSY 2011; NAVIGATOR 2010; Osei 2004; Page 1993), and one of these explicitly acknowledged several individuals employed by a pharmaceutical company for their contribution to the trial (NAVIGATOR 2010).

Participants

Two trials reported the ethnicity of participants; one trial included mainly white participants (NAVIGATOR 2010) and the other only black Americans (Osei 2004). Only one trial included participants from low‐income countries (NAVIGATOR 2010). In one trial all participants fulfilled the diagnostic criteria for IGT 12 months prior to randomisation and this was confirmed 12 months later after the participants were randomised (Eriksson 2006). One trial had a treatment‐free run‐in period (NAVIGATOR 2010). One trial included only males (Papoz 1978) and one trial did not report the gender of the participants (Osei 2004). For the remaining trials, authors provided gender information. The age of included participants varied from 39 to 60.4 years (Appendix 5).

All trials reported fasting glucose values at baseline, which reported plasma glucose values from 4.8 mmol/L to 6.1 mmol/L (NAVIGATOR 2010; Osei 2004). Four trials reported 2‐hour glucose values after a glucose‐load test at baseline, which varied from 7.6 mmol/L to 9.2 mmol/L (NAVIGATOR 2010; Papoz 1978). HbA1c values were reported at baseline in two trials (NANSY 2011; NAVIGATOR 2010). One trial did not report BMI at baseline (Papoz 1978). In the other trials all participants had at baseline a mean BMI over 25 kg/m2. Two trials had participants with a mean BMI over 30 kg/m2 at baseline (NAVIGATOR 2010; Osei 2004). Only one trial reported the number of participants with previous cardiovascular diseases at baseline (NAVIGATOR 2010).

Most trials excluded participants with other endocrine conditions, or hepatic or kidney disease.

The diagnostic criteria used for identifying intermediate hyperglycaemia varied in the included trials: in one trial IFG was the only inclusion criterion. This trial defined IFG as two overnight consecutive FBG values ≥ 5.6 mmol/L with a mean between 5.6 and 6.0 mmol/L (NANSY 2011). Five trials included participants with IGT (Eriksson 2006; NAVIGATOR 2010; Osei 2004; Page 1993; Papoz 1978). One trial evaluated intermediate hyperglycaemia by FPG levels < 7 mmol/L and by 2‐hour plasma glucose levels after a glucose‐load test ≥ 7.8 mmol/L and < 11.1 mmol/L (Eriksson 2006). The NAVIGATOR trial required FPG levels between 5.3 and 7.0 mmol/L (NAVIGATOR 2010). One trial required FPG levels < 7.8 mmol/L (Osei 2004). Three trials required 2‐hour plasma glucose values after a glucose‐load test ≥ 7.8 mmol/L and < 11.1 mmol/L (Eriksson 2006; NAVIGATOR 2010; Osei 2004). Two trials explicitly stated that they applied the criteria for IFG as recommended by WHO at the time of screening (Eriksson 2006; Osei 2004). One trial applied FPG levels > 5.6 mmol/L and 60‐minute plasma glucose levels during a continuous infusion of glucose > 9.3 mmol/L, which was stated to be equivalent to the WHO criteria for IGT (Page 1993). One trial applied the European Diabetes Epidemiology Study Group 1970 criteria and required two separate tests. Particiants had to have 2‐hour blood glucose values after a glucose‐load test ≥ 6.6 mmol/L but < 8.3 mmol/L or FBG levels ≥ 5.5 mmol/L up to 7.2 mmol/L on the second test (Papoz 1978).

Interventions

All the participants in the included trials were treatment‐naïve with regard to pharmacological glucose‐lowering interventions. Three trials included a second‐generation sulphonylurea in the sulphonylurea‐intervention arms (Eriksson 2006; Page 1993; Papoz 1978). None of the trials investigated the same second‐generation sulphonylurea; one trial investigated glipizide 2.5 mg once daily (Eriksson 2006); one trial investigated gliclazide 40 mg twice daily (Page 1993); and one trial investigated glibenclamide 2.0 mg twice daily (Papoz 1978). One trial combined a second‐generation sulphonylurea (glibenclamide 2.0 mg twice daily) with metformin (850 mg twice daily) (Papoz 1978). Two trials investigated a third‐generation sulphonylurea (NANSY 2011; Osei 2004), one trial investigated glipizide GITS 5 mg once daily (Osei 2004) and one trial investigated glimepiride 1.0 mg once daily (NANSY 2011). One trial investigated the meglitinide analogue, nateglinide, 60 mg three times daily (NAVIGATOR 2010). All the included trials were placebo controlled. In addition, one trial included a comparator arm with an active pharmacological glucose‐lowering agent (metformin) (Papoz 1978), and one had diet and exercise as a comparator (Page 1993). We judged placebo as well as diet and exercise as adequate comparators to establish fair comparisons (Appendix 3).

In two trials participants did not take the study drug the day glycaemic tests were performed (Osei 2004; Page 1993). In one trial the participants took the study drug after the glycaemic test had been performed (NAVIGATOR 2010). In two trials the participants took the study drug on the morning before testing (NANSY 2011; Papoz 1978). In one trial participants had stopped the study drug 15 days prior to the last assessment of glycaemic variables (Papoz 1978). One trial did not specify whether the study drug was taken on the day that glycaemic variables were measured (Eriksson 2006); however, an additional glucose assessment was performed 12 months after the study drug was stopped (Eriksson 2006).

None of the included trials, except one (NAVIGATOR 2010), described the intervention strategy for the participants that progressed to type 2 diabetes mellitus (T2DM). The first phase of the NAVIGATOR trial investigated the impact of intensified lifestyle interventions with diet and exercise. If these were insufficient metformin could be added. Finally, a second non‐insulin secretagogue could be added or bedtime insulin was started (NAVIGATOR 2010).

Outcomes

Three trials explicitly specified primary outcomes but did not define secondary outcomes (NANSY 2011; NAVIGATOR 2010; Papoz 1978). The remaining trials did not specify primary or secondary outcomes (Eriksson 2006; Osei 2004; Page 1993) (see Appendix 7). Only one trial was registered on ClinicalTrials.gov (NAVIGATOR 2010), where 14 documented changes were tracked, the last change date being 28 June 2011 (NAVIGATOR 2010). Two coprimary outcomes were predefined for the NAVIGATOR trial: incident T2DM and an extended composite cardiovascular outcome (death from a cardiovascular cause, non‐fatal myocardial infarction, non‐fatal stroke, hospitalisation for heart failure, arterial revascularisation, hospitalisation for unstable angina). One of these primary outcomes (death from a cardiovascular cause, non‐fatal myocardial infarction, non‐fatal stroke or hospitalisation for heart failure) was initially designed to be assessed as a secondary outcome (NAVIGATOR 2010).

Three trials reported one or more of the primary outcomes of relevance for this review (Eriksson 2006; NANSY 2011; NAVIGATOR 2010). Three assessed the incidence of T2DM as an outcome (Eriksson 2006; NANSY 2011; NAVIGATOR 2010). One trial defined T2DM as FPG ≥ 7.0 mmol/L or a 2‐hour blood glucose after a glucose‐load test ≥ 11.1 mmol/L, confirmed by two separate measurements (NAVIGATOR 2010). One trial reported T2DM as FBG values ≥ 6.1 mmol/L on two separate measurements (NANSY 2011). One trial did not report the diagnostic criteria for T2DM, but defined IGT according to WHO 1999 criteria (Eriksson 2006). It is therefore likely, that the WHO recommendation was also applied to the diagnosis of T2DM. One trial predefined the assessment of mortality and cardiovascular complications (NAVIGATOR 2010).

The reporting of adverse events was lacking in most trials. Only one trial reported all non‐serious and serious adverse events experienced during the trial (NAVIGATOR 2010) (see Appendix 11; Appendix 12; Appendix 13; Appendix 14).

All the included trials reported on one or more of the glycaemic variables that we had predefined to assess in our review. None of the included trials reported on microvascular outcomes, health‐related quality of life or socioeconomic effects.

Source of data

We attempted to contact all authors or investigators via email; however, no additional data were provided (see Appendix 16).

Excluded studies

We excluded a total of 39 articles after full‐text evaluation (Figure 1). These references are listed in Characteristics of excluded studies and some are detailed in 'Trials with a duration less than 12 weeks' (Appendix 1).

We excluded 11 trials published in 10 references as they did not allocate participants to sulphonylureas or meglitinide analogues by randomisation. One of the trials was an RCT in which participants were randomised to diet and exercise versus control (Cederholm 1985). The participants randomised to diet and exercise were offered glipizide and were therefore not randomised to this intervention.

We excluded six trials as they did not include participants of relevance to this review. For one of the trials we were unable to judge whether participants with intermediate hyperglycaemia were included after full‐text evaluation (Gudipaty 2014). We made contact with the trial authors and they confirmed that all participants had T2DM (Gudipaty 2014). Two trials included pregnant participants with gestational diabetes (NCT00744965; NCT01563120).

We excluded four trials published in 14 records as it was not possible to obtain separate data on the participants of interest for our review, neither from the publication nor through correspondence with the investigators. We contacted two authors of one trial but did not receive a reply (Igata 2014). We also did not receive a reply from the primary investigator of the DIAbetes and diffuse coronary NArrowing study (DIANA 2012). The corresponding author of another trial responded and asked which additional data we needed (Major‐Pedersen 2008). We sent the requested information but did not receive additional data, even after sending a reminder. We were also unable to obtain additional data on the participants with intermediate hyperglycaemia in the Fasting Hyperglycaemia Study (The Fasting Hyperglycaemia Study 1997a).

We excluded three trials because of the trial duration was less than 12 weeks (Lindblad 2001; Saloranta 2002; Schmoelzer 2006) and four systematic reviews published in five references (Anderson 2005; Bhardwaj 2010; Hopper 2011; Phung 2012; Van de Laar 2006).

Risk of bias in included studies

For details on the risk of bias of the included trials, see Characteristics of included studies.

For an overview of review authors' judgements about each risk of bias item for individual trials and across all trials, see Figure 2 and Figure 3.


Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included trials (blank cells indicate that the particular outcome was not measured in some trials).

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included trials (blank cells indicate that the particular outcome was not measured in some trials).


Risk of bias summary: review authors' judgements about each risk of bias item for each included trial (blank cells indicate that the trial did not measure that particular outcome).

Risk of bias summary: review authors' judgements about each risk of bias item for each included trial (blank cells indicate that the trial did not measure that particular outcome).

None of the included trials reported on microvascular outcomes, health‐related quality of life or socioeconomic effects.

Allocation

We judged only one trial to be at low risk of selection bias with regard to the methods of randomisation and allocation concealment (NAVIGATOR 2010). The remaining trials reported that participants were randomised but provided no any further description (Eriksson 2006; NANSY 2011; Osei 2004; Page 1993; Papoz 1978). We therefore judged these trials as at unclear risk of bias regarding randomisation and allocation concealment.

We evaluated trial baseline data for our predefined prognostic baseline variables. Only one trial reported all the prognostic baseline variables of interest, which were all balanced between the intervention groups (NAVIGATOR 2010). The remaining trials reported only some of our predefined key prognostic variables of interest (Eriksson 2006; NANSY 2011; Osei 2004; Page 1993; Papoz 1978). One of the trials reporting key prognostic variables showed important differences between the intervention groups for several variables (Page 1993) (see Appendix 4; Appendix 5; Appendix 6). However, the uneven distribution of these key prognostic variables was not in favour of any particular intervention group.

Blinding

All trials explicitly reported the blinding of participants and investigators. The blinding of participants was ensured by the use of placebo tablets. However, one trial had an addition to the double‐blinded placebo arm ‐ a diet and exercise arm ‐ for which no blinding of the investigators or participants was described (Page 1993). One trial mentioned that a blinded outcome committee evaluated mortality, incidence of T2DM, cardiovascular outcomes, serious adverse events and severe hypoglycaemia (NAVIGATOR 2010). None of the remaining trials reported that a blinded outcome committee was instituted to assess any of the reported outcomes.

Where measured, all primary outcomes of this review were to be investigator assessed, and we judged these to be at low risk of performance and detection bias. All the included trials reported blood glucose measurements performed by the investigators and we judged these outcomes measures to be at low risk of performance and detection bias.

Non‐serious adverse events and mild hypoglycaemia were partly or exclusively self‐reported in all trials. Only one trial reported non‐serious adverse events other than mild hypoglycaemia (NAVIGATOR 2010). One trial reported that the only adverse events observed were hypoglycaemic symptoms (Eriksson 2006). The number of participants with mild hypoglycaemia was reported in four trials (Eriksson 2006; NAVIGATOR 2010; Osei 2004; Page 1993). However, two trials reported that no participants experienced mild hypoglycaemia (Osei 2004; Page 1993). In one of these trials one of the intervention arms (diet and exercise) was not blinded (Page 1993). Overall, we considered the risk of performance bias and detection bias to be low or unclear for our secondary outcomes.

Incomplete outcome data

We considered overall risk of attrition bias to be unclear for most of our outcomes.

Only three trials reported the numbers of participants randomised and finishing the trial (NAVIGATOR 2010; Page 1993; Papoz 1978). The percentages of randomised participants completing the trials varied from 58% to 100%. Two trials did not describe how many participants were originally randomised but reported the number analysed (Eriksson 2006; NANSY 2011). In the NANSY trial, the authors stated that 71 participants interrupted participation prematurely, the reason being death for seven of these participants. Unfortunately, the trial authors did not describe numbers per allocated group. In another trial, three participants dropped out early but the trial authors did not specify to which intervention these participants belonged to (Eriksson 2006). The NAVIGATOR trial reported that 163 participants in the nateglinide group and 143 participants in the placebo group withdrew, but did not provide details (NAVIGATOR 2010). One trial gave a detailed description of the participants who did not complete the trial (Page 1993). Another trial reported on how many participants were lost to follow‐up but did not provide reasons (Papoz 1978). One trial did not mention whether any participants withdrew or were lost to follow‐up (Osei 2004).

In one trial, two participants in the glipizide group withdrew due to hypoglycaemia (Eriksson 2006). As these two dropouts could have had a substantial impact on the effect estimate for hypoglycaemia, we therefore judged the risk of attrition bias to be high for this outcome.

Selective reporting

Only one trial had a published protocol (NAVIGATOR 2010). We judged five of the included trials to be at high risk of reporting bias on one or more of the outcomes of relevance for our review (Eriksson 2006; NANSY 2011; NAVIGATOR 2010; Page 1993; Papoz 1978). One trial had an unclear risk of reporting bias (Osei 2004). For more details see Appendix 7 and Appendix 8.

Other potential sources of bias

Five of the included trials stated that they had received grants from a pharmaceutical company (Eriksson 2006; NANSY 2011; NAVIGATOR 2010; Osei 2004; Page 1993) and one explicitly acknowledged several individuals employed by a pharmaceutical company for their contribution to the trial (NAVIGATOR 2010). It is known that trials receiving funding or provision of free drug or devices from a pharmaceutical company lead to more favourable results and conclusions than trials sponsored by other sources (Lundh 2012). Therefore, we judged only one trial to be free of other sources of bias (Papoz 1978); for the rest we judge other sources of bias to be unclear.

Effects of interventions

See: Summary of findings for the main comparison Summary of findings (sulphonylureas); Summary of findings 2 Summary of findings (meglitinide analogues)

Baseline characteristics

For details of baseline characteristics, see Appendix 4; Appendix 5; Appendix 6

Sulphonylureas as monotherapy versus any pharmacological glucose‐lowering intervention

One trial compared sulphonylurea monotherapy with another pharmacological glucose‐lowering intervention. This trial had four intervention arms: glibenclamide plus metformin, glibenclamide plus placebo, metformin plus placebo and placebo only (Papoz 1978). This trial reported FBG and blood glucose values 2 hours after an oral glucose load. The mean fasting glucose value at the end of intervention in the glibenclamide plus placebo group was 5.0 (SD 0.5) mmol/L measured in 22 participants versus 5.3 (SD 0.5) mmol/L in 23 participants in the metformin plus placebo group. The 2‐hour blood glucose values showed a mean of 6.3 (SD 1.6) mmol/L in 22 participants versus 6.4 (SD 1.3) mmol/L in 23 participants, respectively. For both glycaemic variables we converted data from mg/dL into mmol/L and calculated SDs from reported standard errors (Papoz 1978).

Sulphonylureas as monotherapy versus behaviour‐changing interventions

One trial compared sulphonylurea monotherapy with diet and exercise (Page 1993). This trial reported fasting glucose. The mean fasting glucose value at the end of intervention in the gliclazide group showed a mean of 5.1 (SD 0.8) mmol/L measured in six participants versus 5.6 (SD 0.7) mmol/L in 18 participants in the diet and exercise group. None of the participants experienced mild or severe hypoglycaemia.

Sulphonylureas as monotherapy versus placebo

Five trials compared a sulphonylurea monotherapy with placebo (Eriksson 2006; NANSY 2011; Osei 2004; Page 1993; Papoz 1978). Two trials included a third‐generation sulphonylurea (NANSY 2011; Osei 2004), the others included a second‐generation sulphonylurea (Eriksson 2006; Page 1993; Papoz 1978). Two trials had a follow‐up period without any pharmacological intervention after the intervention period was stopped (Eriksson 2006; Page 1993).

A description of the outcomes for this comparison is listed in the summary of findings Table for the main comparison.

Primary outcomes
All‐cause mortality

One trial reported all‐cause mortality (NANSY 2011). The participants of this trial were allocated to glimepiride or placebo. A total of 5/136 (3.7%) participants in the glimepiride group died compared with 2/138 (1.4%) in the placebo group. In the glimepiride group, one person died from cardiovascular disease, two persons from cancer, one person from suicide and one person from drowning. In the placebo group two participants died of cardiovascular causes.

Incidence of type 2 diabetes mellitus

Two trials comparing a sulphonylurea monotherapy with placebo reported data on the incidence of T2DM (Eriksson 2006; NANSY 2011). In the NANSY trial, participants took the trial drug (glimepiride) on the days when glycaemic variables were measured. A diagnosis of T2DM was defined as two consecutive FBG values ≥ 6.1 mmol/L (NANSY 2011). The other trial reporting the incidence of T2DM did not state whether the participants took any study drug (glipizide) on the days when glycaemic measurements were performed but retested glycaemic variables 12 months after the trial drug was stopped (Eriksson 2006). The trial authors did not provide any specific glycaemic value for the diagnosis of T2DM but stated that IGT was defined according to WHO recommendations. It is therefore likely that WHO 1999 criteria were also used to define T2DM (Eriksson 2006).

One of the trials had an intervention period of six months and thereafter an extended follow‐up period of 12 additional months (Eriksson 2006). The other trial had a duration of five years or until T2DM developed (average follow‐up 3.7 years) (NANSY 2011). One trial contributed 96/97 events (NANSY 2011) and only one participant developed T2DM during the intervention period in the other trial (Eriksson 2006). The RR for the incidence of T2DM comparing glimepiride monotherapy with placebo was 0.75 (95% CI 0.54 to 1.04; P = 0.08; 2 trials; 307 participants; very low‐quality evidence; Analysis 1.1).

TSA showed that 4.5% of the diversity‐adjusted information size was accrued so far to detect or reject a 10% RRR. Diversity was zero, but we applied a diversity of 20% when calculating the diversity‐required information size as heterogeneity is likely to increase when future trials are included. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR was accrued so far, the TSA‐adjusted 95% CIs could not be calculated with a diversity at 20%. However, when we set diversity to 0%, the TSA‐adjusted 95% CIs were 0.20 to 2.84.

Serious adverse events

We did not identify trials with data on serious adverse events for this comparison. However, one trial described that all reported adverse effects, with the exception of hypoglycaemia, were mild (Eriksson 2006). It is therefore likely that this trial collected data on serious adverse events but did not identify any events (Eriksson 2006).

Secondary outcomes
Cardiovascular mortality

One trial reported that in the glimepiride group 1/136 (0.7%) participant died of cardiovascular reasons compared with 2/138 (1.4%) participants in the placebo group (NANSY 2011).

Non‐fatal myocardial infarction

We did not identify trials with data on non‐fatal myocardial infarction for this comparison.

Non‐fatal stroke

We did not identify trials with data on non‐fatal stroke for this comparison.

Congestive heart failure

We did not identify trials with data on congestive heart failure for this comparison.

Amputation of lower extremity

We did not identify trials with data on amputation of lower extremity for this comparison.

Blindness or severe vision loss

We did not identify trials with data on blindness or severe vision for this comparison.

End‐stage renal disease

We did not identify trials with data on end‐stage renal disease for this comparison.

Non‐serious adverse events

We did not identify trials with data on non‐serious adverse events for this comparison. However, we considered one trial at high risk of selective outcome reporting for non‐serious adverse events (Eriksson 2006). The publication stated that side effects were mild (Eriksson 2006). However, no details were published.

Hypoglycaemia

Three trials reported data on mild hypoglycaemia (Eriksson 2006; Osei 2004; Page 1993). Two of the trials reported that no participants experienced hypoglycaemia (Osei 2004; Page 1993). Eriksson 2006 reported that 7/16 participants in the sulphonylurea group compared with 5/17 participants in the placebo group experienced mild hypoglycaemic events (Analysis 1.2; Analysis 1.3).

Two participants withdrew from one of the trials because of hypoglycaemia (Eriksson 2006). Whether this was due to repetitive mild hypoglycaemia or severe hypoglycaemia could not be determined from the publication (Eriksson 2006).

Health‐related quality of life

We did not identify trials with data on health‐related quality of life for this comparison.

Time to progression to T2DM

We did not identify trials with data on time to progression to T2DM for this comparison.

Measures of blood glucose control

Fasting blood glucose

Four trials comparing a sulphonylurea monotherapy with placebo reported fasting glucose values at the end of the intervention (Eriksson 2006; Osei 2004; Page 1993; Papoz 1978). In two trials the participants did not take the trial drug on the days glycaemic variables were measured (Osei 2004; Page 1993). One trial applied the trial drug on the day of glycaemic testing after 2 and 14 months; however, the last glycaemic measurements at 26 months were performed 15 days after the trial drug was stopped (Papoz 1978). One trial did not report whether the participants took any study drug on the days of glycaemic measurements, but performed a retesting of glycaemic variables 12 months after the trial drug was stopped (Eriksson 2006). Comparing sulphonylurea monotherapy with placebo showed a MD in FBG of ‐0.31 mmol/L (95% CI ‐0.59 to ‐0.02; P = 0.03; 4 trials; 105 participants; overall low or unclear risk of bias; Analysis 1.4). The termination of the trial drug 15 days before measurement at the end of the intervention in Papoz 1978 may explain why this trial had the highest fasting glucose level at the end of intervention. Unfortunately, the largest trial comparing a sulphonylurea monotherapy with placebo (274 participants) did not report fasting glucose values, even though it was obviously measured (NANSY 2011). One trial measured glucose in whole blood (Papoz 1978). In one trial it was not clearly reported how glucose was measured, but according to the diagnostic criteria for intermediate hyperglycaemia it might have been plasma glucose (Eriksson 2006). In the remaining trials it was clearly stated that glucose was measured in plasma (Osei 2004; Page 1993). Whole blood glucose values were converted to plasma glucose values (diabetes.co.uk 2016a).

Subgroup analyses for FBG: a subgroup analysis according to the type of sulphonylurea (third‐generation versus second‐generation) showed no interaction between these subgroups (Analysis 1.5 and Appendix 19). All trials comparing a sulphonylurea monotherapy with placebo included participants with IGT. Two trials had a duration of two years or more (Osei 2004; Papoz 1978). The subgroup analysis according to the duration of the intervention showed no interaction between these subgroups (Analysis 1.6 and Appendix 19). Two trials applied diagnostic criteria for IGT as recommended by WHO (Eriksson 2006; Osei 2004). Subgroup analysis according to diagnostic criteria showed no interaction between these subgroups (Analysis 1.7 and Appendix 19). The participants included in Eriksson 2006 were about 10 to 15 years older (mean age 56.5 years) than the participants in the other trials comparing sulphonylurea monotherapy with placebo (Osei 2004; Page 1993; Papoz 1978). Subgroup analysis for age showed no marked interaction (Analysis 1.8 and Appendix 19). We could not perform subgroup analyses according to ethnicity, comorbidity or previous gestational diabetes mellitus because of lack of reporting in the included trials.

Sensitivity analyses for FBG: a sensitivity analysis excluding trials with a duration of less than two years (Osei 2004; Papoz 1978) showed a MD in fasting glucose of ‐0.28 mmol/L (95% CI ‐0.95 to 0.39). In comparison, trials with a duration of two years or longer showed a MD in fasting glucose of ‐0.35 mmol/L (95% CI ‐0.69 to 0.00) (Analysis 1.6). Only one trial did not receive funding from a pharmaceutical company (Papoz 1978). Excluding this trial resulted in a MD in fasting glucose of ‐0.34 mmol/L (95% CI ‐0.84 to 0.15). The predefined sensitivity analyses regarding publication status, risk of bias, language of publication, imputation or country could not be performed. TSA showed that 23.3% of the diversity‐adjusted information size has been accrued so far to detect or reject a mean difference of ‐0.31 mmol/L. Diversity was zero, but we applied a diversity of 20% as heterogeneity might increase when future trials are added. The TSA‐adjusted 95% CI was ‐0.94 to 0.33.

Two trials reported fasting glucose values after the intervention periods were stopped and observed participants without any intervention (Eriksson 2006; Page 1993). Comparing sulphonylurea monotherapy with placebo, the MD in FBG was ‐0.08 mmol/L (95% CI ‐1.04 to 0.89; Analysis 1.9). TSA showed that 0.17% of the diversity‐adjusted information size had been accrued so far to detect or reject a mean difference of ‐0.08 mmol/L. Diversity was 70.7%. As only a minor fraction of the diversity‐adjusted required information size was accrued so far, the TSA‐adjusted 95% CI could not be calculated.

Blood glucose 2 hours after an oral glucose load

Glucose values 2 hours after an oral glucose load at the end of the intervention period were reported in three trials (Eriksson 2006; Osei 2004; Papoz 1978). Sulphonylurea monotherapy compared with placebo showed a MD in 2‐hour blood glucose of ‐0.42 mmol/L (‐1.28 to 0.43; P = 0.33; 3 trials; 92 participants; overall low or unclear risk of bias; Analysis 1.10). None of the subgroup analyses showed an interaction between the subgroups (Appendix 19).

Subgroup analyses for type of sulphonylurea (Analysis 1.11), duration of intervention (Analysis 1.12) and diagnostic criteria (Analysis 1.13) showed no interaction between subgroups. We could not perform subgroup analyses according to ethnicity, comorbidity and previous gestational diabetes mellitus because of lack of reporting in the included trials.

One trial reported glucose values 2 hours after an oral glucose load one month after the intervention period had ended (Eriksson 2006). The glucose values 2 hours after oral glucose load had a mean of 7.0 mmol/L (SD 1.6) in 16 participants originally allocated to sulphonylurea monotherapy and 8.6 mmol/L (SD 2.4) in 16 participants originally allocated placebo.

HbA1c

One trial clearly stated that HbA1c was measured but reported only that no statistically significant changes were found (NANSY 2011).

Socioeconomic effects

We did not identify trials with data on socioeconomic effects for this comparison.

Sulphonylureas as monotherapy versus no intervention

We did not identify trials comparing sulphonylurea monotherapy with no intervention.

Sulphonylureas as a part of combination therapy versus any pharmacological glucose‐lowering agent

One trial compared sulphonylureas as a part of combination therapy with another pharmacological glucose‐lowering agent. This trial had four intervention arms: glibenclamide plus metformin, glibenclamide plus placebo, metformin plus placebo and placebo only (Papoz 1978). This trial reported fasting glucose and glucose values 2 hours after an oral glucose load. Glucose values were reported as blood glucose values. The FBG value at the end of intervention in the glibenclamide plus metformin group was 5.2 (SD 0.8) mmol/L measured in 22 participants versus 5.3 (SD 0.5) mmol/L in 23 participants in the metformin plus placebo group. The blood glucose value 2 hours after an oral glucose load was 6.4 (SD 1.0) mmol/L in 22 participants in the glibenclamide plus metformin group versus 6.4 (SD 1.3) mmol/L in 23 participants in the metformin plus placebo group. For both glycaemic variables we converted data from mg/dL into mmol/L and calculated SDs from reported standard errors (Papoz 1978).

Meglitinide analogues as monotherapy versus any pharmacological glucose‐lowering intervention

We did not identify trials comparing meglitinide analogues as monotherapy with other pharmacological glucose‐lowering interventions.

Meglitinide analogues as monotherapy versus behaviour‐changing interventions

We did not identify trials comparing meglitinide analogues as monotherapy with behaviour changing interventions.

Meglitinide analogues as monotherapy versus placebo

One trial compared a meglitinide analogue as monotherapy (nateglinide) with placebo (NAVIGATOR 2010). The trial had a factorial design, as the participants were also randomised to valsartan or placebo (NAVIGATOR 2010). A narrative description of the outcomes is listed in the summary of findings Table 2. Because we identified only one trial with meglitinide analogues we could not perform meta‐analyses or TSA.

Primary outcomes
All‐cause mortality

A total of 310 of 4645 (6.7%) participants allocated to nateglinide versus 312 of 4661 (6.7%) participants allocated to placebo died during the trial. Participants who were considered lost to follow‐up remained in the trial until trial end or until death, if known. We determined vital status at each visit and at trial end by searching public records. Vital status was available for 95.7% of the possible participants at the end of follow‐up. HR for all‐cause mortality was 1.00 (95% CI 0.85 to 1.17; P = 0.98).

Incidence of T2DM

The two main criteria for defining T2DM in NAVIGATOR trial were a FPG level ≥ 7.0 mmol/L or a 2‐hour post challenge glucose ≥ 11.1 mmol/L. A confirmatory oral glucose tolerance test had to be performed within 12 weeks after measurement of an elevated glucose level. The date of onset of T2DM was specified as the date of the first elevated glucose measurement. Furthermore, an independent committee adjudicated cases where diabetes was diagnosed by other means (e.g. cases suggestive of diabetes where the glycaemic test‐based definition was not available because of missing central laboratory measurements or repeat tests outside the 12‐week time limit). The committee also adjudicated cases where diabetes was diagnosed by a primary care physician (possibly based on local laboratory assessments), initiation of glucose‐lowering interventions or both. On testing days the participants were told to take the trial drug after the glucose tests.

T2DM developed in 1674 of 4645 (36.0%) participants in the nateglinide group and in 1580 of 4661 (33.9%) in the placebo group. The test of interaction for the factorial allocation to valsartan and placebo did not show any important influence on the effect estimate (P = 0.5). The HR for the incidence of T2DM was 1.07 (95% CI 1.00 to 1.15; P = 0.05). The incidence of T2DM was confirmed by laboratory measurements in 1587 participants in the nateglinide group and 1495 participants in the placebo group. The incidence of T2DM was determined by the adjudication committee in 87 participants in the nateglinide group and 85 participants in the placebo group.

The trial authors also investigated the influence of several factors on the risk of developing T2DM by means of subgroup analyses, such as age, sex, ethnicity, region, fasting plasma glucose, 2‐hour postprandial glucose, BMI, waist circumference, blood pressure control, hypertension, history of cardiovascular disease and angiotensin‐converting enzyme (ACE) inhibitor treatment. All reported HRs refer to the development of T2DM.

  • A diagnosis on the basis of FPG resulted in a HR of 0.87 (95% CI 0.79 to 0.96; P = 0.005) in favour of nateglinide. A diagnosis on the basis of plasma glucose levels 2 hours after a glucose challenge resulted in a HR of 1.24 (95% CI 1.13 to 1.36; P < 0.001) in favour of placebo.

  • Participants aged < 60 years had a HR of 0.99 (95% CI 0.88 to 1.12), participants aged 60 to 67 years showed a HR of 1.19 (95% CI 1.06 to 1.35) and participants aged ≥ 67 years had a HR of 1.04 (95% CI 0.91 to 1.18) in favour of placebo.

  • Male participants had a HR of 0.96 (95% CI 0.87 to 1.06) and female participants a HR of 1.21 (95% CI 1.10 to 1.34) in favour of placebo.

  • Participants from Asian regions had a HR of 1.20 (95% CI 0.90 to 1.61), participants from European regions had a HR of 1.08 (95% CI 0.98 to 1.19), participants from Latin America had a HR of 1.02 (95% CI 0.86 to 1.21) and participants from North America had a HR of 1.05 (95% CI 0.91 to 1.22). Participants from various other regions had a HR of 1.26 (95% CI 0.84 to 1.91).

  • Participants with a FPG ≤ 6.1 mmol/L had a HR of 1.17 (95% CI 1.04 to 1.30) in favour of placebo. Participants with a FPG > 6.1 mmol/L had a HR of 1.01 (95% CI 0.92 to 1.11).

  • Participants with a 2‐hour plasma glucose value ≤ 9.0 mmol/L had a HR of 1.07 (95% CI 0.95 to 1.19). Participants with 2‐hour plasma glucose > 9 mmol/L had a HR of 1.07 (95% CI 0.97 to 1.17).

  • Participants with a BMI ≤ 25 kg/m2 had a HR of 1.14 (95% CI 0.92 to 1.42). Participants with a BMI 25 kg/m2 to 30 kg/m2 had a HR of 1.16 (95% CI 1.03 to 1.30) in favour of placebo. Participants with a BMI 30 kg/m2 to 35 kg/m2 had a HR of 1.00 (95% CI 0.88 to 1.13). Participants with a BMI > 35 kg/m2 had a HR of 1.01 (95% CI 0.86 to 1.18).

  • Participants with a waist circumference < 88 cm (women) and < 102 cm (men) had a HR of 1.17 (95% CI 1.03 to 1.33) in favour of placebo. Participants with a waist circumference ≥ 88 cm (women) and ≥ 102 cm (men) had a HR of 1.03 (95% CI 0.95 to 1.13).

  • Participants with systolic blood pressure < 140 mmHg or diastolic blood pressure < 90 mmHg had a HR of 1.11 (95% CI 1.00 to 1.23). Participants with a systolic blood pressure > 140 mmHg or a diastolic blood pressure > 90 mmHg or taking antihypertensive drugs had a HR of 1.05 (95% CI 0.95 to 1.15).

  • Participants with a history of cardiovascular disease had a HR of 1.04 (95% CI 0.91 to 1.20). Participants without a history of cardiovascular disease had a HR of 1.09 (95% CI 1.00 to 1.18).

  • Participants with ACE inhibitor treatment had a HR of 0.90 (95% CI 0.70 to 1.15) and participants without ACE inhibitor treatment a HR of 0.90 (95% CI 0.70 to 1.15).

Serious adverse events

Serious adverse events were reported in 2066/4602 (44.9%) participants in the nateglinide group versus 2089/4599 (45.4%) participants in the placebo group

Secondary outcomes
Cardiovascular mortality

The numbers of participants who died due to cardiovascular disease during the trial were 126/4645 (2.7%) allocated to nateglinide versus 118/4661 (2.5%) allocated to placebo. The HR for death due to cardiovascular disease was 1.07 (95% CI 0.83 to 1.38; P = 0.60).

Non‐fatal myocardial infarction

The numbers of participants who experienced a non‐fatal myocardial infarction during the trial were 116/4645 (2.5%) in the nateglinide group versus 122/4661 (2.6%) in the placebo group.

Non‐fatal stroke

The numbers of participants who experienced a non‐fatal stroke during the trial were 100/4645 (2.2%) in the nateglinide group versus 110/4661 (2.4%) in the placebo group.

Congestive heart failure

The numbers of participants developing congestive heart failure were not reported. However, the numbers of participants hospitalised for congestive heart failure were 85/4645 (1.8%) in the nateglinide group versus 100/4661 (2.1%) allocated to placebo. The HR was 0.85 (95% CI 0.64 to 1.14; P = 0.27).

Amputation of lower extremity

No data on amputation of the lower extremity were reported.

Blindness or severe vision loss

One participant developed blindness during the trial. This participant was allocated to the placebo group.

End‐stage renal disease

No data on end‐stage renal disease were reported. However, it is very likely that data on this outcome had been collected.

Non‐serious adverse events

The numbers of participants who experienced a non‐serious adverse event were 3921/4602 (85.2%) allocated to nateglinide group versus 3866/4599 (84.1%) allocated to placebo.

Hypoglycaemia

A mild hypoglycaemic event was experienced by 676/4645 (14.6%) participants allocated to nateglinide versus 411/4661 (8.8%) participants allocated to placebo. Severe hypoglycaemia was experienced by 21/4645 (0.5%) allocated to nateglinide versus 12/4661 (0.3%) allocated to placebo.

Health‐related quality of life

No data on health‐related quality of life were reported.

Time to progression to T2DM

Please see section 'Incidence of T2DM' above.

Measures of blood glucose control

Fasting glucose values were lower in participants in the nateglinide group than in those in the placebo group during the trial (MD 0.03 mmol/L; 95% CI 0.003 to 0.05; P = 0.03). However, no statistically significant difference between the intervention groups was apparent at the end of follow‐up. FBG values and SDs at the end of follow‐up were not reported, but we estimated these from the published figure. We estimated FPG measurements at the end of follow‐up of 6.2 mmol/L (SD 1.7) in the nateglinide group versus 6.3 mmol/L (SD 2.6) in the placebo group. We were not able to estimate how many participants were included in the analyses of glucose measurements at the end of follow‐up from the figure in the publication.

Two hours after an oral glucose load glucose values were higher in the nateglinide group than in the placebo group (MD 0.24 mmol/L; 95% CI 0.16 to 0.33; P < 0.001). According to the figure in the publication, glucose values 2 hours after an oral glucose load were 9.5 mmol/L (SD 3.4) in the nateglinide group and 9.2 mmol/L (SD 3.4) in the placebo group. However, we were not able to estimate how many participants were included in the analyses of glucose measurements at the end of follow‐up from the figure in the publication.

It was clearly stated in the publication that HbA1c was measured. However, trial authors reported HbA1c values only at the time the diagnosis of T2DM was established (nateglinide 6.1% (SD 0.6) versus placebo 6.3% (SD 0.6)).

Socioeconomic effects

No data on socioeconomic effects were reported. However, trial authors stated that health economics assessments would be performed.

Meglitinide analogues as monotherapy versus no intervention

We did not identify trials comparing meglitinide analogues as monotherapy with other glucose‐lowering interventions.

Meglitinide analogues as a part of combination therapy versus any pharmacological glucose‐lowering agent

We did not identify trials comparing meglitinide analogues as monotherapy with other glucose‐lowering interventions.

Subgroup analyses

We did not perform subgroups analyses for most comparisons because there were insufficient trials to estimate effects in various subgroups. However, we performed subgroup analyses for the comparison sulphonylureas as monotherapy versus placebo (Appendix 19).

Sensitivity analyses

We did not perform sensitivity analyses for most comparisons because there were insufficient trials to explore the influence of our predefined factors on effect sizes. However, we performed sensitivity analyses for the comparison sulphonylureas as monotherapy versus placebo (Appendix 19).

Assessment of reporting bias

We did not draw funnel plots due to limited number of trials.

Ongoing trials

We did not identify any ongoing RCTs.

Discussion

Summary of main results

This Cochrane review is the first systematic review investigating the effects of insulin secretagogues versus other pharmacological glucose‐lowering interventions, placebo, diet and exercise or no intervention in people at increased risk for the development of type 2 diabetes mellitus (T2DM). We included six trials with a total of 10,018 participants. We judged all trials as at unclear or high risk of bias in one or more 'Risk of bias' domains. The amount of evidence on patient‐important outcomes was limited. The single meta‐analysis comparing sulphonylurea monotherapy (glimepiride) with placebo on the incidence of T2DM after the end of the intervention established neither an advantage nor a disadvantage for glimepiride treatment on the development of T2DM. Here, as well as for all‐cause mortality and cardiovascular mortality, we judged the quality of evidence as very low. All of the included trials reported one or more glycaemic variables. However, for all variables the diversity‐adjusted required information size to confirm the findings from the meta‐analyses was not reached.

One large trial investigated a meglitinide analogue (nateglinide). There was moderate‐quality evidence for the outcomes all‐cause and cardiovascular mortality, incidence of T2DM, serious adverse events, non‐fatal myocardial infarction or stroke and congestive heart failure. Overall, we observed firm evidence neither for or against nateglinide treatment.

Overall completeness and applicability of evidence

We conducted an extensive search for trials, including publications in all languages, and tried to obtain additional data on all trials. However, no additional data were provided. We looked for additional trials and cross‐checked our data with the data from other meta‐analyses and Cochrane reviews of relevance (Anderson 2005; Bhardwaj 2010; Hopper 2011; Phung 2012; Van de Laar 2006).

The diagnosis of intermediate hyperglycaemia varied among trials and some trials used a definition which may have included participants judged to be euglycaemic or having T2DM. Detailed information about the participants was lacking in most trials, and only one trial reported the number of participants with previous cardiovascular disease at baseline (NAVIGATOR 2010). The included trials applied different types of sulphonylurea at varying doses (Eriksson 2006; NANSY 2011; Osei 2004; Page 1993; Papoz 1978). Only one trial included a meglitinide analogue (NAVIGATOR 2010).

A potential selection bias exists as more healthy and motivated people may participate in a clinical trial. However, a Cochrane systematic review observed that clinical outcomes in people participating in RCTs are comparable to similar individuals outside trials (Vist 2008).

Quality of the evidence

None of the six included trials in our review was classified as at low risk of bias on all 'Risk of bias' domains. In general, the description of randomisation and allocation in the included studies was insufficient. Most trials had insufficient reporting of one or more outcomes of relevance to our review and were classified as at high risk of bias for selective outcome reporting bias. We were able to assess one or more of our predefined outcomes in all included trials.

For the sulphonylureas (glimepiride) we judged the quality of evidence to be very low because of the risk of bias and very limited data resulting in imprecision. For the meglitinide analogues (nateglinide), we judged the quality of evidence to be moderate, mainly due to imprecision.

Certain potential limitations of this review warrant special consideration, one being that we were dealing with a heterogeneous group of trials. Our meta‐analyses are limited by an inability to use individual patient data to assess whether distinct clinical characteristics may have influenced the effect estimates of the interventions. We would have explored heterogeneity using sensitivity analyses for our patient‐important outcomes, if possible. However, only two meta‐analyses (both on glycaemic variables) provided sufficient data to perform subgroup and sensitivity analyses. Many of the included trials were not designed or powered to detect our predefined patient‐important outcomes.

Some trials required the participants to take the study drug on the days the glycaemic variables were measured, whereas others did not. This may have influenced the glucose measurements in these trials, as well as the incidence of T2DM (which is based on glycaemic measurements) making it difficult to compare incidence rates.

Some of the trials reported glucose values in whole blood whereas other reported glucose values in plasma. We converted values of whole blood glucose to plasma glucose values in order to make the data comparable. However, it is well known that such conversions might be inaccurate (WHO/IDF 2006).

Most of the included trials had a relatively small number of participants and the information sizes in the meta‐analyses were equally small. This increases the risk of unrealistic estimates of the intervention effects due to bias (systematic errors) and chance (random errors) (Wetterslev 2008; Wood 2008). We have attempted to clarify systematic errors. We contacted all trial authors for clarification if one of the bias domains was not adequately reported. However, trial authors provided no additional information. To reduce the risk of random errors, we conducted trial sequential analyses on all predefined outcomes, whenever possible.

All trials except one (Papoz 1978) received funding from the pharmaceutical industry. It is known that trials receiving funding or provision of free drug or devices from a pharmaceutical company lead to more favourable results and conclusions than trials sponsored by other sources (Lundh 2012).

Potential biases in the review process

Despite an extensive search and attempts to contact the authors of the included trials we did not retrieve any additional trials. We were unable to draw funnel plots in order to assess small study bias due to lack of data. If more data had been available and more meta‐analyses could have been performed we would have tried to investigate heterogeneity and the potential reasons for it. We excluded several trials as they did not provide separate data on participants with intermediate hyperglycaemia (DIANA 2012; Igata 2014; Major‐Pedersen 2008; The Fasting Hyperglycaemia Study 1997a). In all cases we approached the investigators in order to request separate data. None of the investigators provided additional data.

We excluded studies investigating first‐generation sulphonylureas because of the current very limited use of these drugs,

Several trials were published in more than one publication, which for some trials made it difficult to separate the primary publication from companion papers (for details see Included studies).

We made a concerted effort to obtain additional data from trial authors. As most of the included trials were relatively old, we found it difficult to identify contact information for some trials. However, if we were unable to retrieve contact information for the corresponding author, we attempted to contact one of the other coauthors. For all trials we identified contact information for one or more authors.

We excluded trials including participants with IGT due to other conditions (e.g. cystic fibrosis or glucocorticoid treatment).

We included trials with a minimum duration of 12 weeks in order to be able to detect clinically relevant differences for the predefined outcomes. We identified three trials with a duration of less than 12 weeks. Unfortunately, the reporting of long‐term data in the trials included in our review was poor.

We excluded studies assessing composite macrovascular outcomes from our review because composite outcomes are often problematic due to varying definitions in composite outcome measures. One of the trials (NAVIGATOR 2010) reported two composite cardiovascular outcomes (an extended composite cardiovascular outcome; HR 0.93; 95% CI 0.83 to 1.03; P = 0.16) and a core composite cardiovascular outcome (HR 0.94; 95% CI 0.82 to 1.09; P = 0.43).

Data extraction was carried out by two review authors. However, the review authors extracting the data were not blinded as to which trial they were extracting data from.

Agreements and disagreements with other studies or reviews

Several RCTs have assessed the effects of different pharmacological glucose‐lowering interventions for the prevention of T2DM (ACT NOW 2011; Diabetes Prevention Program FU 2009; DREAM 2008). The first RCT to investigate whether an insulin secretagogue could be effective in people at high risk of T2DM was the Bedford trial (Bedford 1982). The trial randomised participants to tolbutamide versus placebo. The trial found no statistically significant difference in the incidence of T2DM between the intervention groups after 8.5 years of follow‐up (Bedford 1982). Later, the Malmö trial randomised participants with IGT to tolbutamide versus diet, placebo, both, or no intervention and followed participants for 10 years (Sartor 1980). More than 50% of the participants in the tolbutamide group dropped out and data on T2DM prevention were inconclusive (Sartor 1980).

A pharmacological approach to the prevention of T2DM is appealing to both the clinician and the pharmaceutical industry. However, although a reduction in the incidence of T2DM is important, the major public health impact of prevention trials will be determined by whether the prevention ‐ or a delay ‐ in the development of T2DM will translate into a reduction in diabetes‐specific macro‐ and microvascular complications.

The results of the factorial NAVIGATOR RCT were largely inconclusive. Neither nateglinide (nor the combination of nateglinide and valsartan) definitely reduced the incidence of T2DM or the events of the two coprimary cardiovascular disease outcomes. The only positive result was a reduction in the incidence of T2DM with valsartan (HR 0.86; 95% CI 0.80 to 0.92; P < 0.001).

Unfortunately, our review could not clarify whether insulin secretagogues reduce the incidence of T2DM and its associated complications in individuals at high risk of T2DM. None of the trials showed statistically significant reductions in blood glucose values 2 hours after an oral glucose load at the end of the intervention compared with placebo. This was also the case for the NAVIGATOR trial, which might seem surprising, taking the mechanism of action of the meglitinide analogues into account (NAVIGATOR 2010). The authors of the NAVIGATOR trial describe this paradoxical finding as a rebound effect, since nateglinide was not administered on the mornings when the oral glucose tolerance tests were performed (NAVIGATOR 2010). Another trial did also not administer the trial drug (GITS) on the mornings when the glycaemic measurements were performed (Osei 2004). However, in this trial both fasting blood glucose (FBG) and 2‐hour glucose levels were higher in the placebo group than in the GITS group. Nevertheless, no statistically significant difference between the groups was seen. If such a rebound effect really exists, it remains to be proven.

When all the trials investigating sulphonylureas were combined FBG was reduced compared with placebo. In the NAVIGATOR trial, FBG was lower in participants allocated to nateglinide than in those receiving placebo. None of the trials reported end‐of study HbA1c levels, which can partly be explained by the age of some of the trials. However, although the NAVIGATOR trial measured HbA1c it reported this measurement only for participants who developed T2DM. For the participants who developed T2DM in the NAVIGATOR trial, HbA1c was lower in those allocated to nateglinide than in those receiving placebo. No increased risk of hypoglycaemia was reported in the trials comparing a sulphonylurea with placebo. However, the trials rarely reported hypoglycaemia as an outcome; when they did, such reporting was often insufficient (Eriksson 2006; Osei 2004; Page 1993). The only outcome which seemed to be influenced by nateglinide in the NAVIGATOR trial was hypoglycaemia; there was an increased risk with nateglinide versus placebo (NAVIGATOR 2010).

We did not identify any ongoing trials investigating the effects of an insulin secretagogue in people at increased risk of the development of T2DM. This reflects a lack of interest from the scientific community and the pharmaceutical companies as their focus now is towards newer and more expensive pharmacological glucose‐lowering interventions (Hemmingsen 2016a).

Trial flow diagram.
Figures and Tables -
Figure 1

Trial flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included trials (blank cells indicate that the particular outcome was not measured in some trials).
Figures and Tables -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included trials (blank cells indicate that the particular outcome was not measured in some trials).

Risk of bias summary: review authors' judgements about each risk of bias item for each included trial (blank cells indicate that the trial did not measure that particular outcome).
Figures and Tables -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included trial (blank cells indicate that the trial did not measure that particular outcome).

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 1 Incidence of type 2 diabetes.
Figures and Tables -
Analysis 1.1

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 1 Incidence of type 2 diabetes.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 2 Mild hypoglycaemia.
Figures and Tables -
Analysis 1.2

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 2 Mild hypoglycaemia.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 3 Severe hypoglycaemia.
Figures and Tables -
Analysis 1.3

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 3 Severe hypoglycaemia.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 4 Fasting blood glucose control.
Figures and Tables -
Analysis 1.4

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 4 Fasting blood glucose control.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 5 Fasting blood glucose control: type of SU.
Figures and Tables -
Analysis 1.5

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 5 Fasting blood glucose control: type of SU.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 6 Fasting blood glucose control: duration of intervention.
Figures and Tables -
Analysis 1.6

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 6 Fasting blood glucose control: duration of intervention.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 7 Fasting blood glucose control: diagnostic criteria.
Figures and Tables -
Analysis 1.7

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 7 Fasting blood glucose control: diagnostic criteria.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 8 Fasting blood glucose control: age.
Figures and Tables -
Analysis 1.8

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 8 Fasting blood glucose control: age.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 9 Extension period: fasting blood glucose.
Figures and Tables -
Analysis 1.9

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 9 Extension period: fasting blood glucose.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 10 2‐hour glucose [mmol/L].
Figures and Tables -
Analysis 1.10

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 10 2‐hour glucose [mmol/L].

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 11 2‐hour glucose [mmol/L]: type of SU.
Figures and Tables -
Analysis 1.11

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 11 2‐hour glucose [mmol/L]: type of SU.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 12 2‐hour glucose [mmol/L]: duration of intervention.
Figures and Tables -
Analysis 1.12

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 12 2‐hour glucose [mmol/L]: duration of intervention.

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 13 2‐hour glucose [mmol/L]: diagnostic criteria.
Figures and Tables -
Analysis 1.13

Comparison 1 Sulphonylureas as monotherapy vs placebo, Outcome 13 2‐hour glucose [mmol/L]: diagnostic criteria.

Summary of findings for the main comparison. Summary of findings (sulphonylureas)

Insulin secretagogues for prevention or delay of type 2 diabetes mellitus and its associated complications in persons at risk for the development of type 2 diabetes mellitus

Population: people at risk for the development of type 2 diabetes mellitus

Settings: outpatient

Intervention: sulphonylureas (data available for glimepiride only)

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(trials)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo

Glimepiride

All‐cause mortality

Follow‐up: mean 3.7 years

See comment

See comment

See comment

274
(1)

⊕⊝⊝⊝
very lowa

5/136 (3.7%) participants in the glimepiride group versus 2/138 (1.4%) in the placebo group

Incidence of type 2 diabetes mellitus

Measured as 2 consecutive fasting blood glucose values ≥ 6.1 mmol/L (NANSY 2011b) or no definition provided (Eriksson 2006)
Follow‐up: 6 months and a mean of 3.7 years

361 per 1000

271 per 1000 (195 to 376)

RR 0.75 (0.54 to 1.04)

307
(2)

⊕⊝⊝⊝
very lowc

Serious adverse events

See comment

See comment

See comment

See comment

See comment

Not reported

Cardiovascular mortality

Follow‐up: mean 3.7 years

See comment

See comment

See comment

274
(1)

⊕⊝⊝⊝
very lowa

1/136 (0.7%) participants died due to cardiovascular disease in the sulphonylurea monotherapy group and 2/138 (1.4%) participants died in the placebo group

Non‐fatal myocardial infarction, non‐fatal stroke, congestive heart failure

See comment

See comment

See comment

See comment

See comment

Not reported

Health‐related quality of life

See comment

See comment

See comment

See comment

See comment

Not reported

Socioeconomic effects

See comment

See comment

See comment

See comment

See comment

Not reported

*The basis for the assumed risk (e.g. the median control group risk across trials) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; RR: risk ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

*Assumed risk was derived from the event rates in the comparator groups
aDowngraded by three levels because of serious imprecision and possible publication bias
bDiagnostic criterion for trial entry was impaired fasting glucose in the NANSY trial (baseline glycosylated haemoglobin A1c was 4.9% for both groups) and impaired glucose tolerance in Eriksson 2006. In the NANSY trial participants took glimepiride on the days when glycaemic variables were measured
cDowngraded by three levels because of indirectness, serious imprecision and possible publication bias. Trial sequential analysis showed that only 4.5% of the diversity‐adjusted information size was accrued so far to detect or reject a 10% relative risk reduction

Figures and Tables -
Summary of findings for the main comparison. Summary of findings (sulphonylureas)
Summary of findings 2. Summary of findings (meglitinide analogues)

Insulin secretagogues for prevention or delay of type 2 diabetes mellitus and its associated complications in persons at risk for the development of type 2 diabetes mellitus

Population: people at risk for the development of type 2 diabetes mellitus

Settings: outpatients

Intervention: meglitinide analogues (nateglinide)

Comparison: placebo

Outcomes

Placebo

Nateglinide

Relative effect
(95% CI)

No of participants
(trials)

Quality of the evidence
(GRADE)

Comments

All‐cause mortality

Follow‐up: median 6.5 years

See comment

See comment

See comment

9306 (1)

⊕⊕⊕⊝
moderatea

310/4645 (6.7%) participants died in the nateglinide group versus 312/4661 (6.7%) participants in the placebo group. Vital status was available for 95.7% of participants at the end of follow‐up. The HR was 1.00; 95% CI 0.85 to 1.17; P = 0.98

Incidence of type 2 diabetes mellitus

Defined as: fasting plasma glucose ≥ 7.0 mmol/L (126 mg/dL) or a 2‐hour blood glucose after a glucose‐load test ≥ 11.1 mmol/L (200 mg/dL) or by an adjudication committeeb

Follow‐up: median 5 years

See comment

See comment

See comment

9306 (1)

⊕⊕⊕⊝
moderatea

Type 2 diabetes mellitus developed in 1674/4645 (36.0%) participants in the nateglinide group and in 1580/4661 (33.9%) in the placebo group. The HR was 1.07; 95% CI 1.00 to 1.15; P = 0.05

Serious adverse events

Follow‐up: median 5 years

See comment

See comment

See comment

9306 (1)

⊕⊕⊕⊝
moderatea

The number of participants who experienced a serious adverse events was 2066/4602 (44.9%) participants in the nateglinide group versus 2089/4599 (45.6%) participants in the placebo group

Cardiovascular mortality

Follow‐up: 6.5 years

See comment

See comment

See comment

9306 (1)

⊕⊕⊕⊝
moderatea

The number of participants who died due to cardiovascular disease was 126/4645 (2.7%) participants in the nateglinide group versus 118/4661 (2.5%) participants in the placebo group. The HR was 1.07; 95% CI 0.83 to 1.38; P = 0.60

(a) Non‐fatal myocardial infarction

(b) Non‐fatal stroke

(c) Congestive heart failure

Follow‐up: median 6.3 years

See comment

See comment

See comment

9306 (1)

(a), (b), (c):

⊕⊕⊕⊝
moderatea

(a) The number of participants who experienced a non‐fatal myocardial infarction during the trial was 116/4645 (2.5%) participants in the nateglinide group versus 122/4661 (2.6%) participants in the placebo group

(b) The number of participants who experienced a non‐fatal stroke during the trial was 100/4645 (2.2%) participants in the nateglinide group versus 110/4661 (2.4%) participants in the placebo group

(c) The number of participants developing congestive heart failure was not reported. However, the number of participants hospitalised for congestive heart failure was 85/4645 (1.8%) participants in the nateglinide group versus 100/4661 (2.1%) participants in the placebo group. The HR was 0.85; 95% CI 0.64 to 1.14; P = 0.27

Health‐related quality of life

See comment

See comment

See comment

See comment

See comment

Not reported

Socioeconomic effects

See comment

See comment

See comment

9306 (1)

See comment

One trial specified the assessment of health economics (NAVIGATOR 2010). However, trial authors did not provide data

*The basis for the assumed risk (e.g. the median control group risk across trials) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; HR: hazard ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

aDowngraded by one level because of imprecision, high risk of selective reporting and possible publication bias (see Appendix 18)
bDiagnostic criterion for the NAVIGATOR trial entry was impaired glucose tolerance; baseline glycosylated haemoglobin A1c was 5.8% for both groups. Progression to diabetes was confirmed by laboratory measurements in 1587 participants in the nateglinide group (34.2%) and 1495 participants in the placebo group (32.1%). Progression to diabetes was determined by the adjudication committee in the case of 87 participants assigned to nateglinide (1.9%) and 85 assigned to placebo (1.8%)

Figures and Tables -
Summary of findings 2. Summary of findings (meglitinide analogues)
Table 1. Overview of trial populations

Intervention(s) and comparator(s)

Description of power and sample size calculation

Screened/eligible
(N)

Randomised
(N)

ITT
(N)

Analysed
(N)

Finishing trial
(N)

Randomised finishing trial
(%)

Follow‐up
(extended follow‐up)a

Eriksson 2006

I: glipizide 2.5 mg

b

‐c

17

16

16

6 months (18 months)

C: placebo

c

17

17

16

total:

37

34

33

32

NANSY 2011

I: glimepiride 1.0 mg

Quote: "...assuming 3% conversion rate per year and 33% reduction of diabetes development with 5% significance and 80% statistical power"

d

136

136

e

5 years or until diabetes developed, average follow‐up period 3.7 years

C: placebo

d

138

138

e

total:

288d

274

274

203

74.1

NAVIGATOR 2010

I: nateglinide 60 mg, three times daily

Quote: "The sample size calculation was therefore based on a 'subadditivity / 75% additivity of effects' approach, assuming an effect size of 32% on cardiovascular outcome of the two drugs in combination. The treatment discontinuation rate was assumed to be 30% over five years, corresponding to approximately 6.9% per annum. While patients on treatment were assumed to have the full effect (i.e. 20% reduction of hazard rate if in the monotherapy group), it was assumed that patients who discontinued treatment would have only ¼ of the treatment effect remaining as carry‐over effect. Furthermore, it was expected that 75% of the patients who discontinued treatment could be followed up for events. The remaining 25% would comprise patients completely lost to follow‐up, patients who die (without reaching a primary endpoint), and those for whom events are unintentionally not reported by the investigator. Based on these assumptions, a total of 9152 patients will provide 90% testwise power to detect a treatment difference in the extended cardiovascular endpoint"

43 502

4748

4645

4645

3726

78.5

Quote: "The median follow‐up time for data on vital status was 6.5 years, and the median follow‐up times for data on the diabetes, extended cardiovascular, and core cardiovascular outcomes were 5.0, 6.3, and 6.4 years, respectively"f

C: placebo

4770

4661

4661

3747

78.6

total:

9518

9306

9306

7473

78.5

Osei 2004

I: GITS 5 mg

9

9

9

24 months (26 months)

C: placebo

9

9

9

total:

18

18

18

Page 1993

I: gliclazide 40 mg twice daily

6

6

6

6

100

6 months (7 months)

C1: placebo

8

7

7

7

87.5

C2: diet + exercise

23

18

18

18

78.2

total:

37

31

31

31

83.8

Papoz 1978

I1: glibenclamide 2.0 mg twice daily + metformin 850 mg twice daily

29

22

22

22

75.9

2 years (2 years)

I2: glibenclamide 2.0 mg twice daily + placebo

28

22

22

22

78.6

C1: placebo + metformin 850 mg twice daily

30

23

23

23

76.7

C2: placebo

33

19

19

19

57.6

total:

120

86

86

86

71.7

Grand total

All interventionsh

4820

3792

All comparatorsh

4873

3830

All interventions and comparatorsi

10,018

7825 j

‐ denotes not reported

aFollow‐up under randomised conditions until end of trial or, if not available, duration of intervention; extended follow‐up refers to follow‐up of participants once the original study was terminated as specified in the power calculation
bParticpants identified through screening of another trial (Botnia Study 1996). Quote: "The subjects included in the present study represented the first consecutive 37 subjects who maintained their IGT status on repeated OGTT testing during 1 year"
cThe investigators described that they randomised 37 participants, and three dropped out shortly after. However, they do not describe how these three participants were allocated, but only describe that after the three participants had left 17 were allocated to each intervention group
dThe investigators described that 14 randomised participants withdrew before the first occasion to establish the conversion to type 2 diabetes mellitus. All except one dropped out for administrative reasons. However, it was not specified to which intervention group these participants were allocated
e71 individuals interrupted participation prematurely, however it was not described to which groups they belonged
fThe trial was predefined to stop and the final analysis performed when 1374 participants have had an adjudication committee confirmed extended cardiovascular endpoint
hNot all trials described the number of participants randomised to each intervention group
iTwo trials did not report the number of randomised participants per intervention group. Therefore, numbers do not add up accurately
jNot all trials reported the number of participants finishing the trial

C: comparator; GITS: glipizide gastrointestinal therapeutic system; I: intervention; ITT: intention‐to‐treat; NANSY: The Nepi ANtidiabetes StudY

Figures and Tables -
Table 1. Overview of trial populations
Comparison 1. Sulphonylureas as monotherapy vs placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Incidence of type 2 diabetes Show forest plot

2

307

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.54, 1.04]

2 Mild hypoglycaemia Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Severe hypoglycaemia Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

4 Fasting blood glucose control Show forest plot

4

105

Mean Difference (IV, Random, 95% CI)

‐0.31 [‐0.59, ‐0.02]

5 Fasting blood glucose control: type of SU Show forest plot

4

105

Mean Difference (IV, Random, 95% CI)

‐0.31 [‐0.59, ‐0.02]

5.1 Second‐generation SU

3

87

Mean Difference (IV, Random, 95% CI)

‐0.27 [‐0.57, 0.02]

5.2 Third‐generation SU

1

18

Mean Difference (IV, Random, 95% CI)

‐0.70 [‐1.72, 0.32]

6 Fasting blood glucose control: duration of intervention Show forest plot

4

105

Mean Difference (IV, Random, 95% CI)

‐0.31 [‐0.59, ‐0.02]

6.1 duration less than 2 years

2

46

Mean Difference (IV, Random, 95% CI)

‐0.28 [‐0.95, 0.39]

6.2 duration 2 years or more

2

59

Mean Difference (IV, Random, 95% CI)

‐0.35 [‐0.69, 0.00]

7 Fasting blood glucose control: diagnostic criteria Show forest plot

4

105

Mean Difference (IV, Random, 95% CI)

‐0.31 [‐0.59, ‐0.02]

7.1 WHO diagnostic

2

51

Mean Difference (IV, Random, 95% CI)

‐0.22 [‐0.86, 0.42]

7.2 Other criteria

2

54

Mean Difference (IV, Random, 95% CI)

‐0.36 [‐0.70, ‐0.02]

8 Fasting blood glucose control: age Show forest plot

4

105

Mean Difference (IV, Random, 95% CI)

‐0.48 [‐0.79, ‐0.17]

8.1 age less than 50 yrs

3

72

Mean Difference (IV, Random, 95% CI)

‐0.62 [‐0.95, ‐0.30]

8.2 age above 50 years

1

33

Mean Difference (IV, Random, 95% CI)

0.0 [‐0.60, 0.60]

9 Extension period: fasting blood glucose Show forest plot

2

45

Mean Difference (IV, Random, 95% CI)

‐0.08 [‐1.04, 0.89]

10 2‐hour glucose [mmol/L] Show forest plot

3

92

Mean Difference (IV, Random, 95% CI)

‐0.42 [‐1.28, 0.43]

11 2‐hour glucose [mmol/L]: type of SU Show forest plot

3

92

Mean Difference (IV, Random, 95% CI)

‐0.42 [‐1.28, 0.43]

11.1 Second‐generation SU

2

74

Mean Difference (IV, Random, 95% CI)

‐0.16 [‐0.90, 0.57]

11.2 Third‐generation SU

1

18

Mean Difference (IV, Random, 95% CI)

‐2.0 [‐4.20, 0.20]

12 2‐hour glucose [mmol/L]: duration of intervention Show forest plot

3

92

Mean Difference (IV, Random, 95% CI)

‐0.42 [‐1.28, 0.43]

12.1 Duration 2 years or more

2

59

Mean Difference (IV, Random, 95% CI)

‐0.75 [‐2.64, 1.15]

12.2 Duration less than 2 years

1

33

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐1.56, 0.76]

13 2‐hour glucose [mmol/L]: diagnostic criteria Show forest plot

3

92

Mean Difference (IV, Random, 95% CI)

‐0.42 [‐1.28, 0.43]

13.1 WHO criteria

2

51

Mean Difference (IV, Random, 95% CI)

‐0.92 [‐2.38, 0.55]

13.2 Other criteria

1

41

Mean Difference (IV, Random, 95% CI)

0.0 [‐0.95, 0.95]

Figures and Tables -
Comparison 1. Sulphonylureas as monotherapy vs placebo