Scolaris Content Display Scolaris Content Display

Tratamiento combinado con metformina y sulfonilurea de segunda o tercera generación para pacientes adultos con diabetes mellitus tipo 2

Contraer todo Desplegar todo

Antecedentes

La cantidad de pacientes con diabetes mellitus tipo 2 (DMT2) está aumentando a nivel mundial. La combinación de metformina y sulfonilurea (M+S) es un tratamiento utilizado ampliamente. Si la M+S muestra mejores o peores efectos en comparación con otros fármacos antidiabéticos en pacientes con DMT2 todavía es polémico.

Objetivos

Evaluar los efectos del tratamiento combinado con metformina y sulfonilureas (de segunda o tercera generación) en pacientes adultos con diabetes mellitus tipo 2.

Métodos de búsqueda

Se actualizó la búsqueda de una revisión sistemática reciente del Agency for Healthcare Research and Quality (AHRQ). La búsqueda actualizada incluyó CENTRAL, MEDLINE, Embase, ClinicalTrials.gov y WHO ICTRP. La fecha de la última búsqueda fue marzo 2018. Se realizaron búsquedas en los sitios web de los fabricantes y las listas de referencias de los ensayos incluidos, las revisiones sistemáticas, los metanálisis y los informes de evaluación de tecnología de la salud. Se solicitó a los investigadores de los ensayos incluidos información acerca de ensayos adicionales.

Criterios de selección

Se incluyeron ensayos controlados aleatorios (ECA) que habían asignado al azar a participantes a partir de los 18 años de edad con DMT2 a la M+S en comparación con metformina más otra intervención hipoglucemiante o monoterapia con metformina con una duración del tratamiento de 52 semanas o más.

Obtención y análisis de los datos

Dos autores de la revisión leyeron todos los resúmenes y artículos de texto completo o registros, evaluaron el riesgo de sesgo y extrajeron los datos de resultado de forma independiente. Se utilizó un modelo de efectos aleatorios para realizar el metanálisis, y se calcularon los cocientes de riesgos (CR) para los resultados dicotómicos y las diferencias de medias (DM) para los resultados continuos, mediante intervalos de confianza (IC) del 95% para los cálculos del efecto. La certeza de la evidencia se evaluó mediante los criterios GRADE.

Resultados principales

Se incluyeron 32 ECA que asignaron al azar a 28 746 pacientes. La duración del tratamiento osciló entre uno y cuatro años. Se consideró que ninguno de estos ensayos presentó un riesgo de sesgo bajo en todos los dominios del "Riesgo de sesgo". Los eventos más importantes por persona fueron la mortalidad por todas las causas y cardiovascular, los eventos adversos graves (EAG), el accidente cerebrovascular no mortal (ACNM), el infarto de miocardio (IM) no mortal y las complicaciones microvasculares. Las comparaciones más importantes fueron las siguientes:

Cinco ensayos compararon M+S (N = 1194) con metformina más un análogo del péptido similar al glucagón tipo 1 (N = 1675): la mortalidad por todas las causas fue de 11/1057 (1%) versus 11/1537 (0,7%), cociente de riesgos (CR) 1,15 (intervalo de confianza [IC] del 95%: 0,49 a 2,67); tres ensayos; 2594 participantes; evidencia de certeza baja; la mortalidad cardiovascular de 1/307 (0,3%) versus 1/302 (0,3%), evidencia de certeza baja; los eventos adversos graves (EAG) de 128/1057 (12,1%) versus 194/1537 (12,6%), CR 0,90 (IC del 95%: 0,73 a 1,11); tres ensayos; 2594 participantes; evidencia de certeza muy baja; el infarto de miocardio (IM) no mortal de 2/549 (0,4%) versus 6/1026 (0,6%), CR 0,57 (IC del 95%: 0,12 a 2,82); dos ensayos; 1575 participantes; evidencia de certeza muy baja.

Nueve ensayos compararon M+S (N = 5414) con metformina más un inhibidor de dipeptidil peptidasa tipo 4 (N = 6346): la mortalidad por todas las causas fue de 33/5387 (0,6%) versus 26/6307 (0,4%), CR 1,32 (IC del 95%: 0,76 a 2,28); nueve ensayos; 11 694 participantes; evidencia de certeza baja; la mortalidad cardiovascular de 11/2989 (0,4%) versus 9/3885 (0,2%), CR 1,54 (IC del 95%: 0,63 a 3,79); seis ensayos; 6874 participantes; evidencia de certeza baja; los EAG de 735/5387 (13,6%) versus 779/6307 (12,4%), CR 1,07 (IC del 95%: 0,97 a 1,18); nueve ensayos; 11 694 participantes; evidencia de certeza muy baja; el ACNM de 14/2098 (0,7%) versus 8/2995 (0,3%), CR 2,21 (IC del 95%: 0,74 a 6,58); cuatro ensayos; 5093 participantes; evidencia de certeza muy baja; el IM no mortal de 15/2989 (0,5%) versus 13/3885 (0,3%), CR 1,45 (IC del 95%: 0,69 a 3,07); seis ensayos; 6874 participantes; evidencia de certeza muy baja; un ensayo en 64 participantes informó que no se observó ninguna complicación microvascular (evidencia de muy baja certeza).

Once ensayos compararon M+S (N = 3626) con metformina más una tiazolidinediona (N = 3685): la mortalidad por todas las causas fue de 123/3300 (3,7%) versus 114/3354 (3,4%), CR 1,09 (IC del 95%: 0,85 a 1,40); seis ensayos; 6654 participantes; evidencia de certeza baja; la mortalidad cardiovascular de 37/2946 (1,3%) versus 41/2994 (1,4%), CR 0,78 (IC del 95%: 0,36 a 1,67); cuatro ensayos; 5940 participantes; evidencia de certeza baja; los EAG 666/3300 (20,2%) versus 671/3354 (20%), CR 1,01 (IC del 95%: 0,93 a 1,11); seis ensayos; 6654 participantes; evidencia de certeza muy baja; el ACNM de 20/1540 (1,3%) versus 16/1583 (1%), CR 1,29 (IC del 95%: 0,67 a 2,47); P = 0,45; dos ensayos; 3123 participantes; evidencia de certeza muy baja; el IM no mortal de 25/1841 (1,4%) versus 21/1877 (1,1%), CR 1,21 (IC del 95%: 0,68 a 2,14); P = 0,51; tres ensayos; 3718 participantes; evidencia de certeza muy baja; tres ensayos (3123 participantes) no informaron ninguna complicación microvascular (evidencia de muy baja certeza).

Tres ensayos compararon M+S (N = 462) con metformina más una glinida (N = 476): un paciente murió en cada grupo de intervención (3 ensayos; 874 participantes; evidencia de certeza baja); ninguna mortalidad cardiovascular (dos ensayos; 446 participantes; evidencia de certeza baja); EAG 34/424 (8%) versus 27/450 (6%), CR 1,68 (IC del 95%: 0,54 a 5,21); P = 0,37; tres ensayos; 874 participantes; evidencia de certeza baja; ningún ACNM (un ensayo; 233 participantes; evidencia de certeza muy baja); IM no mortal 2/215 (0,9%), participantes en el grupo de M+S; dos ensayos; 446 participantes; evidencia de certeza baja; ninguna complicación microvascular (un ensayo; 233 participantes; evidencia de certeza baja).

Cuatro ensayos compararon M+S (N = 2109) con metformina más un inhibidor del cotransportador de sodio‐glucosa tipo 2 (N = 3032): la mortalidad por todas las causas fue de 13/2107 (0,6%) versus 19/3027 (0,6%), CR 0,96 (IC del 95%: 0,44 a 2,09); cuatro ensayos; 5134 participantes; evidencia de certeza muy baja; la mortalidad cardiovascular de 4/1327 (0,3%) versus 6/2262 (0,3%), CR 1,22 (IC del 95%: 0,33 a 4,41); tres ensayos; 3589 participantes; evidencia de certeza muy baja; los EAG de 315/2107 (15,5%) versus 375/3027 (12,4%), CR 1,02 (IC del 95%: 0,76 a 1,37); cuatro ensayos; 5134 participantes; evidencia de certeza muy baja; el ACNM de 3/919 (0,3%) versus 7/1856 (0,4%), CR 0,87 (IC del 95%: 0,22 a 3,34); dos ensayos; 2775 participantes; evidencia de certeza muy baja; el IM no mortal de 7/890 (0,8%) versus 8/1374 (0,6%), CR 1,43 (IC del 95%: 0,49 a 4,18; dos ensayos); 2264 participantes; evidencia de certeza muy baja; la amputación de la extremidad inferior de 1/437 (0,2%) versus 1/888 (0,1%); evidencia de certeza muy baja.

Los ensayos informaron más episodios hipoglucémicos con la combinación de M+S en comparación con todas las otras combinaciones de metformina con agentes antidiabéticos. Los resultados para la M+S versus monoterapia con metformina fueron no concluyentes. No hubo ningún ECA que comparara la M+S con metformina más insulina. Se identificaron nueve ensayos en curso y dos ensayos están en espera de evaluación. En conjunto, estos ensayos incluirán aproximadamente 16 631 participantes.

Conclusiones de los autores

Hay evidencia no concluyente sobre si el tratamiento combinado con M+S en comparación con metformina más otra intervención hipoglucemiante resulta en efectos beneficiosos o perjudiciales para los resultados más importantes para los pacientes (mortalidad, EAG, complicaciones macrovasculares y microvasculares) con la excepción de la hipoglucemia (más efectos perjudiciales para la combinación de M+S). Ningún ECA informó sobre la calidad de vida relacionada con la salud.

PICO

Population
Intervention
Comparison
Outcome

El uso y la enseñanza del modelo PICO están muy extendidos en el ámbito de la atención sanitaria basada en la evidencia para formular preguntas y estrategias de búsqueda y para caracterizar estudios o metanálisis clínicos. PICO son las siglas en inglés de cuatro posibles componentes de una pregunta de investigación: paciente, población o problema; intervención; comparación; desenlace (outcome).

Para saber más sobre el uso del modelo PICO, puede consultar el Manual Cochrane.

Tratamiento combinado con metformina y sulfonilurea para pacientes adultos con diabetes mellitus tipo 2

Pregunta de la revisión

Se pretendía investigar los efectos de la combinación de los fármacos antidiabéticos metformina más sulfonilurea en comparación con otras intervenciones antidiabéticas en pacientes con diabetes tipo 2.

Antecedentes

Muchos pacientes con diabetes tipo 2 son tratados con varios tipos de fármacos hipoglucemiantes como “sulfonilureas” (por ejemplo glibenclamida o gliburida, glipizida y gliclazida). Estos fármacos disminuyen la glucemia al estimular la secreción de insulina en el organismo, por lo que aumentan los niveles de insulina en sangre. Otro agente antidiabético, la metformina, reduce el nivel de glucemia al mejorar la capacidad del cuerpo para lograr que la insulina funcione mejor (sensibilidad a la insulina). La combinación de metformina más sulfonilurea se usa ampliamente. Además, se buscó analizar los efectos de la metformina más sulfonilurea sobre resultados importantes para el paciente como las complicaciones de la diabetes (por ejemplo, nefropatía y enfermedades oculares, ataques cardíacos, 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

Se encontraron 32 estudios controlados aleatorios (ensayos clínicos en los que los pacientes son asignados al azar a uno de dos o más grupos de tratamiento), que asignaron a 28 746 pacientes con diabetes tipo 2 a la metformina más sulfonilurea o a un grupo comparador. Los grupos comparadores constaban de los siguientes tipos de fármacos antidiabéticos además de metformina: cinco estudios con análogos del péptido similar al glucagón 1; nueve estudios con inhibidores de dipeptidil peptidasa tipo 4; 11 estudios con tiazolidinedionas, tres estudios con glinidas y cuatro estudios con inhibidores del cotransportador de sodio‐glucosa tipo 2.

Los participantes de los estudios fueron tratados durante un periodo de entre uno y cuatro años. Hubo diferencias grandes entre los pacientes que participaron en los estudios, en especial con respecto a la edad, por cuánto tiempo los pacientes habían tenido diabetes y si había complicaciones de la diabetes presentes al comienzo del estudio.

Esta evidencia está actualizada hasta marzo 2018.

Resultados clave

Los datos sobre los resultados importantes para los pacientes fueron pocos, y los datos fueron dispersos para todas las comparaciones de la metformina más sulfonilurea con otros fármacos antidiabéticos. Los datos disponibles no mostraron diferencias notables entre la metformina más sulfonilurea y otras combinaciones de metformina con fármacos antidiabéticos o metformina sola para los resultados más importantes para los pacientes. Hubo más eventos de niveles bajos de azúcar sanguíneo (episodios hipoglucémicos) con el tratamiento combinado con metformina más sulfonilurea en comparación con todas las otras combinaciones de metformina con otro compuesto antidiabético.

No se identificaron estudios que informaran sobre la calidad de vida relacionada con la salud. Se identificaron nueve estudios en curso y dos estudios aún no publicados están en espera de evaluación. En conjunto, estos estudios incluirán alrededor de 16 631 participantes. Una vez que se publiquen los resultados, estos estudios podrían influir de manera significativa en los resultados de esta revisión.

Certeza de la evidencia

Todos los estudios incluidos tuvieron deficiencias en la manera en que se realizaron o en cómo los autores del estudio presentaron los resultados. Para las comparaciones individuales de los fármacos antidiabéticos la cantidad de participantes a menudo fue pequeña, lo cual dio lugar a un alto riesgo de errores aleatorios (intervención del azar).

Conclusiones de los autores

disponible en

Implicaciones para la práctica

No existe evidencia sólida sobre si la combinación de metformina más sulfonilurea en comparación con metformina más otro agente hipoglucemiante o la monoterapia con metformina aumenta los efectos beneficiosos o perjudiciales para los resultados más importantes para los pacientes (mortalidad por todas las causas, eventos adversos graves, complicaciones macrovasculares [mortalidad cardiovascular, infarto de miocardio no mortal, accidente cerebrovascular no mortal] y complicaciones microvasculares [amputación de la extremidad inferior, ceguera o pérdida de visión grave, nefropatía terminal]). Hubo más episodios hipoglucémicos informados con la combinación de metformina más sulfonilurea en comparación con todas las otras combinaciones de agentes antidiabéticos y metformina. El riesgo de hipoglucemia aumenta con las metas de niveles bajos de glucosa lo cual puede no aplicar a la mayoría de los pacientes con diabetes de edad muy avanzada. No hubo ningún ensayo que comparara metformina más sulfonilurea con metformina más insulina. Se identificaron nueve ensayos en curso y dos ensayos están en espera de evaluación. En conjunto, estos ensayos incluirán a alrededor de 16 631 participantes y podrían tener una repercusión sobre los resultados de esta revisión.

Implicaciones para la investigación

Aún debe aclararse si hay efectos beneficiosos o perjudiciales considerables de la metformina más sulfonilurea en pacientes con diabetes mellitus tipo 2. Varios ensayos en curso con alrededor de 15 147 participantes están investigando este tema y cada ensayo proporcionará datos sobre uno o más resultados de interés para la revisión. Las fechas de finalización de los ensayos se calculan entre 2018 y 2021. Además dos ensayos con 1484 participantes que investigan la metformina más sulfonilurea en comparación con metformina más inhibidores de SGLT‐2 están en espera de evaluación. Los ensayos controlados aleatorios a largo plazo futuros deben centrarse en los resultados importantes para los pacientes (en especial la mortalidad, la calidad de vida relacionada con la salud, los eventos adversos graves, y las complicaciones macrovasculares y microvasculares).

Summary of findings

Open in table viewer
Summary of findings for the main comparison. Metformin‐sulphonylurea (second‐ or third‐generation) combination therapy compared with metformin plus another antidiabetic drug for adults with type 2 diabetes mellitus

Metformin‐sulphonylurea (second‐ or third‐generation) combination therapy compared with metformin plus another antidiabetic drug for adults with type 2 diabetes mellitus

Patient: people with type 2 diabetes mellitus

Settings: outpatients

Intervention: metformin + sulphonylurea

Comparison: metformin plus another antidiabetic drug

Outcomes

Metformin + antidiabetic drug

Metformin + sulphonylurea

Relative effect
(95% CI)

No. of participants
(trials)

Certainty of the evidence
(GRADE)

Comments

All‐cause mortality (N)

M + GLP1‐A
Follow‐up: 2‐3 years

7 per 1000

8 per 1000 (4 to 19)

RR 1.15 (0.49 to 2.67)

2594 (3)

⊕⊕⊝⊝a1

Low

M + DPP4‐I
Follow‐up: 1‐3 years

4 per 1000

5 per 1000 (3 to 9)

RR 1.32 (0.76 to 2.28)

11,694 (9)

⊕⊕⊝⊝
Lowb1

M + thiazolidinedione
Follow‐up: 1‐5.5 years

34 per 1000

37 per 1000 (29 to 48)

RR 1.09 (0.85 to 1.40)

6654 (6)

⊕⊕⊝⊝
Lowc1

M + nateglinide

Follow‐up: 1‐2 years

See comment

874 (3)

⊕⊕⊝⊝
Lowd1

1 participant died in each intervention group

M + SGLT2‐I
Follow‐up: 2‐4 years

6 per 1000

6 per 1000 (3 to 13)

RR 0.96 (0.44 to 2.09)

5134 (4)

⊕⊝⊝⊝
Very lowe1

Cardiovascular mortality (N)

M + GLP1‐A
Follow‐up: 2‐3 years

See comment

609 (1)

⊕⊕⊝⊝
Lowa2

1/307 (0.3%) participants died due to cardiovascular disease in the M+S group compared with 1/302 (0.3%) participants in the M + GLP1‐A group

M + DPP4‐I
Follow‐up: 1‐3 years

2 per 1000

4 per 1000 (1 to 9)

RR 1.54 (0.63 to 3.79)

6874 (6)

⊕⊕⊝⊝
Lowb2

M + thiazolidinedione
Follow‐up: 1‐5.5 years

14 per 1000

11 per 1000 (5 to 23)

RR 0.78 (0.36 to 1.67)

5940 (4)

⊕⊕⊝⊝
Lowc2

M + nateglinide

Follow‐up: 1 year

See comment

446 (2)

⊕⊕⊝⊝
Lowd2

No cardiovascular death was reported

M + SGLT2‐I
Follow‐up: 2‐4 years

3 per 1000

3 per 1000 (1 to 12)

RR 1.22 (0.33 to 4.41)

3589 (3)

⊕⊝⊝⊝
very lowe2

Serious adverse events (N)

M + GLP1‐A
Follow‐up: 2‐3 years

126 per 1000

114 per 1000 (92 to 140)

RR 0.90 (0.73 to 1.11)

2594 (3)

⊕⊝⊝⊝
Very lowa3

M + DPP4‐I
Follow‐up: 1‐3 years

124 per 1000

132 per 1000 (120 to 146)

RR 1.07 (0.97 to 1.18)

11,694 (9)

⊕⊝⊝⊝
Very lowb3

M + thiazolidinedione
Follow‐up: 1‐5.5 years

200 per 1000

202 per 1000 (186 to 222)

RR 1.01 (0.93 to 1.11)

6654 (6)

⊕⊝⊝⊝
Very lowc3

M + nateglinide

Follow‐up:

60 per 1000

101 per 1000 (32 to 313)

RR 1.68 (0.54 to 5.21)

874 (3)

⊕⊕⊝⊝
Lowd3

M + SGLT2‐I
Follow‐up: 2‐4 years

124 per 1000

126 per 1000 (94 to 170)

RR 1.02 (0.76 to 1.37)

5134 (4)

⊕⊝⊝⊝
Very lowe3

Non‐fatal stroke (N)

M + GLP1‐A

Not reporteda4

M + DPP4‐I
Follow‐up: 1‐2 years

3 per 1000

6 per 1000 (2 to 18)

RR 2.21 (0.74 to 6.58)

5093 (4)

⊕⊝⊝⊝
Very lowb4

M + thiazolidinedione
Follow‐up: 1‐4.8 years

10 per 1000

13 per 1000 (7 to 25)

RR 1.29 (0.67 to 2.47)

3123 (2)

⊕⊝⊝⊝
Very lowc4

M + nateglinide

Follow‐up: 52 weeks

See comment

233 (1)

⊕⊝⊝⊝
Very lowd4

No non‐fatal stroke was reported

M + SGLT2‐I
Follow‐up: 2 years

4 per 1000

3 per 1000 (1 to 13)

RR 0.87 (0.22 to 3.34)

2775 (2)

⊕⊝⊝⊝
Very lowe4

Non‐fatal myocardial infarction (N)

M + GLP1‐A
Follow‐up: 2‐3 years

6 per 1000

3 per 1000 (1 to 16)

RR 0.57 (0.12 to 2.82)

1575 (2)

⊕⊝⊝⊝
Very lowa5

M + DPP4‐I
Follow‐up: 1‐3 years

3 per 1000

5 per 1000 (2 to 10)

RR 1.45 (0.69 to 3.07)

6874 (6)

⊕⊝⊝⊝
very lowb5

M + thiazolidinedione
Follow‐up: 1‐4.8 years

11 per 1000

14 per 1000 (8 to 24)

RR 1.21 (0.68 to 2.14)

3718 (3)

⊕⊝⊝⊝
Very lowc5

M + nateglinide

Follow‐up: 1 year

See comment

446 (2)

⊕⊕⊝⊝
Lowd5

In 1 trial 2/101 (2%) participants had a non‐fatal myocardial infarction in the M+S group compared with 0/112 participant in the metformin plus nateglinide group

M + SGLT2‐I
Follow‐up: 2‐4 years

6 per 1000

8 per 1000 (3 to 24)

RR 1.43 (0.49 to 4.18)

2264 (2)

⊕⊝⊝⊝
Very lowe5

Microvascular complications (N), definition: end‐stage renal disease, blindness or severe vision loss, amputation of lower extremity

M + GLP1‐A

Not reporteda6

M + DPP4‐I
Follow‐up: 1 year

See comment

64 (1)

⊕⊝⊝⊝
Very lowb6

In 1 trial no participants had a lower‐extremity amputation, developed blindness or severe vision loss, or end‐stage renal disease

M + thiazolidinedione
Follow‐up: 1‐4.8 years

See comment

3123 (2)

⊕⊝⊝⊝
Very lowc6

2 trials (3123 participants) reported that no participants had a lower‐extremity amputation
1 trial (95 participants) reported that no participants developed blindness or severe vision loss, or end‐stage renal disease

M + nateglinide

Follow‐up: 52 weeks

See comment

233 (1)

⊕⊕⊝⊝
Lowd6

No microvascular complications were reported

M + SGLT2‐I
Follow‐up: 2 years

See comment

1325 (1)

⊕⊝⊝⊝
Very lowe6

In 1 trial 1/437 (0.2%) participants had an amputation of the lower extremity in the M+S group compared with 1/888 (0.1%) in the M + SGLT2‐I group

Health‐related quality of life

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; DPP4‐I: dipeptidyl peptidase‐4 inhibitor; GLP1‐A: glucagon‐like peptide 1 analogue; HbA1c: glycosylated haemoglobin A1c; M: metformin; M+S: metformin + sulphonylurea; N: number; N/R: not reported; RR: risk ratio; SGLT2‐I: sodium‐glucose co‐transporter 2 inhibitor; T: thiazolidinedione

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

All‐cause mortality
a1Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 17.
b1Downgraded by one level because of inconsistency (non‐consistent direction of effect) and by one level because of imprecision (CI consistent with both benefit and harm) ‐ see Appendix 18.
c1Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 20.
d1Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 21.
e1Downgraded by one level because of inconsistency (point estimates varied widely) and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Cardiovascular mortality
a2Downgraded by two levels because of serious imprecision (small number of trials, CI consistent with both benefit and harm) ‐ see Appendix 17.
b2Downgraded by one level because of because of inconsistency (non‐consistent direction of effect) and by one level because of imprecision (CI consistent with both benefit and harm) ‐ see Appendix 18.
c2Downgraded by two levels because of serious imprecision (small number of trials, CI consistent with both benefit and harm) ‐ see Appendix 20.
d2Downgraded by two levels because of serious imprecision (small number of trials, unknown event rate) ‐ see Appendix 20.
e2Downgraded by one level because of inconsistency (point estimates varied widely) and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Serious adverse events
a3Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 17.
b3Downgraded by one level because of attrition bias, by one level because of inconsistency (non‐consistent direction of effect) and by one level because of imprecision (CI consistent with both benefit and harm) ‐ see Appendix 18.
c3Downgraded by one level because of attrition bias, by one level because of inconsistency (non‐consistent direction of effect) and by one level because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 20.
d3Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 21.
e3Downgraded by one level because of attrition bias and inconsistency (point estimates varied widely) and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 22.

Non‐fatal stroke
a4No adequate data for analysis.
b4Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 18.
c4Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 20.
d4Downgraded by two levels of evidence because of serious imprecision (low number of trials, unknown event rate) ‐ see Appendix 21.
e4Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Non‐fatal myocardial infarction
a5Downgraded by one level because of attrition bias and by two levels because of serious imprecision (small number of trials, CI consistent with both benefit and harm) ‐ see Appendix 17.
b5Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 18.
c5Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 20.
d5Downgraded by two levels because of serious imprecision (low number of trials, low event rate) ‐ see Appendix 21.
e15Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Microvascular complications
a6No adequate data for analysis.
b6Downgraded by three levels because of very serious imprecision (small number of participants, one trial only, unknown event rate) ‐ see Appendix 18.
c6Downgraded by one level because of attrition bias and reporting bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, unknown event rate) ‐ see Appendix 20.
d6Downgraded by two levels because of serious imprecision (low number of trials, unknown event rate) ‐ see Appendix 21.
e6Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Antecedentes

disponible en

Varias organizaciones médicas han formulado guías o recomendaciones para el tratamiento de la diabetes mellitus tipo 2 (DMT2). A la mayoría de los pacientes con DMT2 se les recomienda inicialmente reducir la ingesta de calorías y aumentar la actividad física para mejorar el control glucémico (ADA 2016). Sin embargo, para lograr y mantener metas glucémicas específicas, la mayoría de los pacientes con DMT2 requerirá intervenciones farmacológicas hipoglucemiantes. En la actualidad la metformina es el fármaco hipoglucemiante de primera línea en pacientes con DMT2 debido a sus beneficios postulados, incluida la ausencia de aumento de peso, o incluso la pérdida de peso, y la ausencia de hipoglucemia (Inzucchi 2012; Nathan 2009). Cuando las intervenciones conductuales como las dietas y el ejercicio y las dosis máximas toleradas de un fármaco oral para la disminución de la glucosa fracasan en el logro del objetivo glucémico, a menudo se agregan otros fármacos para la disminución de la glucosa (ADA 2016). Debido a que la DMT2 es un trastorno progresivo, con el tiempo, una proporción considerable de pacientes con DMT2 requerirá insulina. Algunas guías recomiendan continuar administrando metformina en esta situación (ADA 2016).

Según se describe más adelante, los pacientes con DMT2 tienen un riesgo elevado de desarrollo de complicaciones macrovasculares así como microvasculares (Almdal 2004). En el tratamiento de los pacientes con DMT2 los investigadores han considerado qué meta glucémica es apropiada para disminuir el riesgo de estas complicaciones. Una hipótesis hasta el presente ha sido que los valores bajos de hemoglobina A1c (HbA1c) glucosilada se asocian con complicaciones macrovasculares y microvasculares reducidas. Sin embargo, este paradigma ha sido desafiado por una revisión Cochrane que investiga el control glucémico intensivo en comparación con el control glucémico convencional en pacientes con DMT2 (Hemmingsen 2011). En esta revisión, los autores encontraron información insuficiente para confirmar o excluir una reducción del riesgo de complicaciones macrovasculares así como microvasculares con el control glucémico intensivo en comparación con el convencional.

Descripción de la afección

En todo el mundo, se calculó que la cantidad de pacientes con diabetes fue de 177 000 000 en 2000 y se prevé que aumente a 366 000 000 en 2030 (Wild 2004). La DMT2 comprende un 90% de los pacientes con diabetes y se asocia con un peso corporal excesivo y con la inactividad física (WHO 2015). La DMT2 se caracteriza por hiperglucemia, resistencia a la insulina y alteración de la secreción de insulina (LeRoith 2002). Aunque la definición de DMT2 depende de la glucemia elevada, la DMT2 no ocurre de forma aislada sino como parte de un síndrome metabólico‐cardiovascular complejo que incluye dislipidemia, hipertensión, obesidad, anomalías en la coagulación, microalbuminuria y aterosclerosis acelerada, aunque no todos y cada uno de estos trastornos ocurre en cada paciente con DMT2 (DeFronzo 1999). Los pacientes con DMT2 tienen un riesgo elevado de desarrollo de macrovasculopatía (como muerte cardiovascular, infarto de miocardio, accidente cerebrovascular e isquemia periférica) así como complicaciones microvasculares (como retinopatía, nefropatía y neuropatía) (Almdal 2004).

Descripción de la intervención

Desde la introducción de las sulfonilureas en los años cincuenta estos fármacos que disminuyen los niveles de glucosa han sido la base del tratamiento de la DMT2. Las primeras en ser introducidas en el mercado fueron las sulfonilureas de primera generación (acetohexamida, carbutamida, clorpropamida, tolazamida y tolbutamida). Posteriormente, se introdujeron las sulfonilureas de segunda y tercera generación y ahora fueron reemplazadas casi por completo por las sulfonilureas de primera generación (Harrower 2000). Se cree que las sulfonilureas de segunda generación (p.ej. glibenclamida [en los EE.UU.: glibenclamida], glipizida y gliclazida) y las sulfonilureas de tercera generación (gliclazida de liberación modificada [LM], glipizida de sistema terapéutico gastrointestinal [GITS, por sus siglas en inglés] y glimepirida) tienen un mejor perfil de seguridad (Harrower 2000). A fines de los años cincuenta la metformina de biguanida se introdujo como otro fármaco hipoglucemiante (Bailey 1996).

La metformina por lo general es la primera opción como fármaco hipoglucemiante cuando la dieta y el ejercicio son insuficientes para controlar la DMT2. Sin embargo, en el caso de intolerancia o de contraindicaciones a la metformina, las sulfonilureas podrían prescribirse como monoterapia. Las sulfonilureas son prescritas principalmente como una parte del tratamiento combinado con otros fármacos hipoglucemiantes, en especial metformina (ADA 2016). Las sulfonilureas se administran por vía oral. La dosis diaria recomendada en pacientes con DMT2 depende del compuesto específico de sulfonilurea. Las sulfonilureas tienen diferentes perfiles farmacocinéticos debido a las diferentes uniones al receptor de sulfonilurea en las células β pancreáticas. La clorpropamida tiene una vida media de 36 horas, mientras que la glimepirida tiene una vida media de alrededor de cinco horas (McCall 2001). La metformina tiene una vida media en plasma de 1,5 a 4,9 horas (Bailey 1996). Debido a las variaciones en la vida media de las diferentes sulfonilureas, algunas tienen que ser administradas una vez al día y otras dos o tres veces al día. Para la glimepirida, la dosis recomendada es de hasta 6 mg por día (Langtry 1998). Para la gliclazida la dosis diaria es de 30 mg a 120 mg (Deacon 2015; Harrower 2000a). Asimismo, la metformina es administrada por vía oral. El ajuste de la metformina empieza con una dosis baja (500 mg) administrada una vez o dos veces por día con las comidas. La dosis máxima recomendada es de hasta 1000 mg dos veces al día (Nathan 2009).

Efectos adversos de la intervención

Todas las sulfonilureas tienen el potencial de causar hipoglucemia. El riesgo de hipoglucemia difiere entre los diferentes tipos de sulfonilureas y algunos agentes como la glibenclamida son propensos a causar hipoglucemia prolongada. El riesgo de hipoglucemia es más pronunciado para las sulfonilureas de primera generación que para las generaciones más nuevas de sulfonilureas (Harrower 2000). En pacientes con DMT2 que reciben metformina, los efectos adversos gastrointestinales, incluido el malestar abdominal y la diarrea, son los eventos adversos más frecuentes y ocurren en un 20% a un 30% de los pacientes. Debido a que la metformina no aumenta la secreción de insulina, la hipoglucemia es poco común en pacientes con DMT2 que reciben monoterapia con metformina (DeFronzo 1999). Previamente, la metformina se consideraba contraindicada en muchas afecciones crónicas, debido al mayor riesgo de acidosis láctica. Sin embargo, una revisión Cochrane ha establecido la conclusión de que no existe evidencia de que la metformina se asocie con un mayor riesgo de acidosis láctica y la lista de contraindicaciones para el uso de metformina debe evaluarse nuevamente (Salpeter 2010).

El ensayo del University Group Diabetes Program (UGDP) indicó que la tolbutamida se asoció con efectos adversos cardiovasculares en comparación con placebo e insulina en pacientes con DMT2 (UGDP 1976). Posteriormente, otros ensayos clínicos aleatorios no demostraron evidencia clara de un mayor riesgo de eventos cardiovasculares con la administración de sulfonilurea en comparación con otros fármacos para la disminución de la glucosa en pacientes con DMT2 (ADOPT 2006; UKPDS‐33 1998). Varios estudios observacionales han indicado una mayor mortalidad y riesgo de enfermedades cardiovasculares con la monoterapia con sulfonilurea en comparación con la monoterapia con metformina en pacientes con DMT2 (Morgan 2014; Roumie 2012; Schramm 2011). Sin embargo, el riesgo parece depender del tipo de sulfonilurea (Pantalone 2012; Schramm 2011). Además, debido a los factores de confusión no controlados, o no detectados, o ambos, en los estudios observacionales, los resultados de estos estudios tienen que comprobarse en ensayos controlados aleatorios (ECA; Deeks 2003).

Un subestudio de UKPDS reveló que el agregado temprano de metformina en los participantes tratados con sulfonilurea se asoció con un mayor riesgo de mortalidad en comparación con la continuación de la sulfonilurea sola (UKPDS‐34 1998). Varios estudios observacionales han investigado la asociación entre la combinación de metformina y las sulfonilureas y el riesgo de enfermedades cardiovasculares y mortalidad. En términos generales, estos estudios revelan resultados contradictorios (Evans 2006; Gulliford 2004; Johnson 2002; Kahler 2007).

De qué manera podría funcionar la intervención

El mecanismo primario de acción para las sulfonilureas es estimular la liberación de insulina de las células β pancreáticas. Las sulfonilureas aumentan la liberación de insulina pancreática mediante el cierre de los canales de trifosfato de adenosina sensible al potasio (P‐ATP, por sus siglas en inglés) en las células β (Harrower 2000; Scott 2012). Se cree que la metformina aumenta la sensibilidad a la insulina, lo cual puede dar lugar a diversos efectos metabólicos. La inhibición de la producción de glucosa hepática (mediante la mayor sensibilidad hepática a la insulina) se considera el mecanismo principal mediante el cual la metformina reduce la glucemia (Krentz 2005). La enzima proteinquinasa activada por monofosfato de adenosina 5' (AMPK, por sus siglas en inglés) se ha identificado como una meta del fármaco. Mediante la fosforilación de las proteínas clave que afectan la producción de energía, la AMPK regula y coordina la glucosa celular y el metabolismo de lípidos (Krentz 2005).

Por qué es importante realizar esta revisión

Varios estudios han investigado el tratamiento combinado con metformina y sulfonilureas y el riesgo de enfermedades cardiovasculares y mortalidad (Evans 2006; Gulliford 2004; Johnson 2002; Kahler 2007; UKPDS‐34 1998). Sin embargo, los datos se basan principalmente en estudios observacionales y muestran resultados contradictorios. Por lo tanto, todavía no se conoce si la metformina y la sulfonilurea en combinación aumentan el riesgo de enfermedades cardiovasculares y mortalidad. Las guías indican flexibilidad en la elección del próximo fármaco después del fracaso de la monoterapia con metformina (ADA 2016). Por lo tanto, aún debe aclararse qué clase de fármaco es la segunda línea más apropiada, debido a que la mayoría de los pacientes con DMT2 necesitará un tratamiento combinado con el transcurso del tiempo para lograr las metas glucémicas. Esta revisión sistemática procura evaluar si las sulfonilureas son la mejor opción de tratamiento combinado con metformina.

Objetivos

disponible en

Evaluar los efectos del tratamiento combinado de metformina y sulfonilureas (de segunda o tercera generación) en pacientes adultos con diabetes mellitus tipo 2.

Métodos

disponible en

Criterios de inclusión de estudios para esta revisión

Tipos de estudios

Se incluyeron ensayos controlados aleatorios (ECA).

Tipos de participantes

Adultos a partir de los 18 años de edad con diabetes mellitus tipo 2 (DMT2).

Criterios de diagnóstico para la diabetes mellitus

El diagnóstico debe establecerse con los criterios estándar válidos al comenzar el ensayo para ser compatible con los cambios en la clasificación y los criterios de diagnóstico para la diabetes con el transcurso de los años (p.ej. ADA 2003; ADA 2008; WHO 1998). En condiciones ideales, se deberían haber descrito los criterios diagnósticos. Cuando fue necesario, se utilizó la definición de diabetes mellitus de los autores de los ensayos. Se programó la evaluación de los criterios diagnósticos en un análisis de sensibilidad.

Tipos de intervenciones

Se planificó investigar las siguientes comparaciones de intervención versus control / comparador.

Intervención

  • Tratamiento combinado con metformina y sulfonilurea de segunda o tercera generación (M+S)

Comparador

  • Metformina más otra intervención hipoglucemiante como un tratamiento combinado (p.ej. metformina más inhibidor de dipeptidilpeptidasa‐4, metformina más insulina)

  • Metformina más placebo

  • Monoterapia con metformina

Las intervenciones concomitantes debían ser las mismas en los grupos de intervención y de comparación para realizar comparaciones justas.

Si un ensayo era de grupos múltiples, se incluía cualquier grupo que cumpliera con los criterios de inclusión de la revisión.

Duración mínima de la intervención

Se incluyeron ensayos con una duración mínima de la intervención de 52 semanas. Debido a que principalmente se intentó investigar los resultados importantes para los pacientes, el interés se centró en los ensayos a más largo plazo, debido a que las complicaciones macrovasculares y microvasculares se presentan con el transcurso del tiempo.

Duración mínima del seguimiento

Se incluyeron ensayos con una duración de la intervención de 52 semanas o más. Los períodos de seguimiento prolongados (también llamados estudios de extensión no enmascarados) definidos como un seguimiento de los participantes una vez que el ensayo original haya terminado, según lo especificado en el cálculo del poder estadístico para este ensayo, con frecuencia son de naturaleza observacional y sólo se los evaluó en cuanto a los eventos adversos (Buch 2011; Megan 2012).

Resumen de los criterios de exclusión específicos

  • Se excluyeron las combinaciones de más de dos agentes hipoglucemiantes.

  • Se excluyeron los estudios que investigaban a pacientes mujeres diagnosticadas con diabetes gestacional.

Tipos de medida de resultado

No se excluyeron ensayos sólo sobre la base de que una o varias de las medidas de resultado primarias o secundarias no se informaban en la publicación. Cuando no se informó ninguno de los resultados primarios o secundarios no se incluyó el ensayo pero se proporcionó alguna información básica en una tabla adicional.

Resultados primarios

  • Mortalidad por todas las causas

  • Calidad de vida relacionada con la salud

  • Eventos adversos graves

Resultados secundarios

  • Mortalidad cardiovascular

  • Infarto de miocardio no mortal

  • Insuficiencia cardíaca

  • Accidente cerebrovascular no mortal

  • Amputación del miembro inferior

  • Ceguera o pérdida de visión severa

  • Nefropatía terminal

  • Eventos adversos no graves

  • Hipoglucemia

  • Efectos socioeconómicos

Resultados exploratorios adicionales

  • Peso

  • HbA1c (hemoglobina A1c glucosilada)

Método de medición de resultados

  • Mortalidad por todas las causas: definida como la muerte por cualquier causa

  • Calidad de vida relacionada con la salud: definida como calidad de vida relacionada con la salud mental y física con dominios separados y combinados, evaluada con un instrumento validado como el Short‐Form 36

  • Eventos adversos graves: definidos según las International Conference on Harmonization Guidelines como cualquier evento que dé lugar a la muerte, que sea potencialmente mortal, requiera hospitalización o la prolongación de la hospitalización existente, dé lugar a discapacidad persistente o significativa o cualquier evento médico importante que pueda haber amenazado al paciente o haber requerido una intervención para prevenirlo (ICH 1997), o según lo informado en los ensayos.

  • Mortalidad cardiovascular: definida como la muerte por infarto de miocardio, insuficiencia cardíaca o accidente cerebrovascular

  • Infarto de miocardio no mortal, insuficiencia cardíaca, accidente cerebrovascular no mortal, amputación de la extremidad inferior, ceguera o pérdida de visión severa, hipoglucemia (leve, moderada, grave): definidos según lo informado en los ensayos. Medidos al final de la intervención y al final del seguimiento.

  • Nefropatía terminal: definida como diálisis, trasplante renal o muerte por la nefropatía

  • Eventos adversos no graves: definidos como el número de pacientes con cualquier evento médico indeseable que puede no tener necesariamente una relación causal con la intervención.

  • Peso y HbA1c: medido en kg y %

  • Efectos socioeconómicos: por ejemplo, costes de la intervención, ausentismo laboral, consumo de medicación

Momento de la medición de los resultados

  • Mortalidad por todas las causas, eventos adversos graves y eventos adversos no graves: cualquier momento después de que los participantes fueron asignados al azar a los grupos de intervención/comparadores

  • Para todas las otras medidas de resultado: al final de la intervención y al final del seguimiento.

Especificación de las variables pronósticas clave

  • Origen étnico

  • Obesidad

  • Hipertensión

  • Diabetes gestacional anterior

  • Edad

  • Enfermedades cardiovasculares existentes

  • Nefropatía

Métodos de búsqueda para la identificación de los estudios

Búsquedas electrónicas

In 2016 the Agency for Healthcare Research and Quality (AHRQ) published an updated systematic review with meta‐analyses on the effectiveness and safety of glucose‐lowering interventions for people with T2DM, including metformin‐based combination therapies (Maruthur 2016). This report included search results from several databases up to April 2015 and a further update of MEDLINE up to December 2015.

We based our search on the results of this systematic AHRQ report and added new references identified by a revised search strategy from 2015 onwards, in the following literature databases.

  • Cochrane Central Register of Controlled Trials (CENTRAL; 2018, Issue 3) via the Cochrane Register of Studies Online (CRSO)

  • MEDLINE Ovid (Epub Ahead of Print, In‐Process & Other Non‐Indexed Citations, Ovid MEDLINE(R) Daily and Ovid MEDLINE(R); from 1946 to 5 March 2018)

  • Embase Ovid (from 1974 to 13 July 2016)

Additionally we searched the following trials registers:

We continuously applied a MEDLINE (Ovid SP) email alert service to identify newly published trials using the same search strategy as described for MEDLINE (for details on search strategies, see Appendix 1).

Búsqueda de otros recursos

We searched the reference lists of included trials, systematic reviews, meta‐analyses and health technology assessment reports for other potentially eligible trials or ancillary publications. 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 had missed.

We also searched manufacturers' websites and the databases of regulatory agencies (European Medicines Agency (EMA), US Food and Drugs Administration (FDA); Hart 2012; Schroll 2015).

We did not use abstracts or conference proceedings for data extraction because this information source does not fulfil the CONSORT requirements which is "an evidence‐based, minimum set of recommendations for reporting randomized trials" (CONSORT; Scherer 2007).

Obtención y análisis de los datos

Selección de los estudios

Two review authors (KM and PK, LK or FG) independently scanned the abstract, title, or both, of every record we retrieved in the literature searches, to determine which trials we should assess further. We obtained the full text of all potentially‐relevant records. We resolved any disagreements through consensus or by recourse to an additional review author (BH). If we could not resolve a disagreement, we categorised the trial as a 'study awaiting classification' and contacted the trial authors for clarification. We prepared a flow diagram of the number of trials identified and excluded at each stage in accordance with the PRISMA flow diagram of trial selection (Liberati 2009).

Extracción y manejo de los datos

For trials that fulfilled our inclusion criteria, two review authors (KM and PK, LK or FG) independently extracted key participant and intervention characteristics. We reported data on efficacy outcomes and adverse events using standard data extraction sheets from the Cochrane Metabolic and Endocrine Disorders (CMED) Group. We resolved any disagreements by discussion or, if required, by consultation with an additional review author (BH) (for details see Characteristics of included studies; Table 1; Table 2; Appendix 2; Appendix 3; Appendix 4; Appendix 5; Appendix 6; Appendix 7; Appendix 8; Appendix 9; Appendix 10; Appendix 11; Appendix 12; Appendix 13; Appendix 14). We tried to retrieve the protocol for each included trial.

We provided information about potentially relevant ongoing trials in the Characteristics of ongoing studies table and in Appendix 7 'Matrix of trial outcome (publications and trial endpoints)'.

We emailed all authors of included trials to enquire whether they were willing to answer questions regarding their trials. We presented the results of this survey in Appendix 15 'Survey of trial investigators providing information on studies'. We sought relevant missing information on the trial from the primary author(s) of the article, 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 yield by collating all available data and used the most complete dataset aggregated across all known publications. We listed duplicate publications, companion documents, multiple reports of a primary trial and trial documents of included trials (such as trial registry information) as secondary references under the study identifier (ID) of the included trial. Furthermore, we also listed duplicate publications, companion documents, multiple reports of a trial and trial documents of excluded trials (such as trial registry information) as secondary references under the study ID of the excluded trial.

Data from clinical trial registers

In case data of included trials were available as study results in clinical trials registers such as ClinicalTrials.gov, we made full use of this information and extracted the data. If there was also a full publication of the trial, we collated and critically appraised all available data. If a trial fulfilled the inclusion criteria and was marked as a completed study in the clinical trials register but no additional information was available, we added this trial to the table of 'Studies awaiting classification'.

Evaluación del riesgo de sesgo de los estudios incluidos

Two review authors (KM and PK, LK or FG) independently assessed the risk of bias of each included trial. We resolved disagreements by consensus, or by consultation with an additional review author (BH). In cases of disagreement, we consulted the remainder of the review author team and made a judgement based on consensus. If adequate information was not available from the publications, trial protocols, or other sources, we contacted the trial authors for more detail to request missing data on 'Risk of bias' items.

We used the Cochrane 'Risk of bias' assessment tool (Higgins 2011a; Higgins 2017) assigning assessments of low, high, or unclear risk of bias (for details, see Appendix 2; Appendix 3). We evaluated individual bias items as described in the Cochrane Handbook for Systematic Reviews of Interventions, according to the criteria and associated categorisations contained therein (Higgins 2017).

Summary assessment of risk of bias

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

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

We considered the following outcomes as self‐reported.

  • Health‐related quality of life

  • Non‐serious adverse events

  • Hypoglycaemia

  • Weight

We considered the following outcomes as investigator‐assessed.

  • All‐cause mortality

  • Serious adverse events

  • Cardiovascular mortality

  • Non‐fatal myocardial infarction

  • Heart failure

  • Non‐fatal stroke

  • Amputation of lower extremity

  • Blindness or severe vision loss

  • End‐stage renal disease

  • Hypoglycaemia

  • Socio‐economic effects

  • Weight

  • HbA1c

Risk of bias for a trial across outcomes
Some risk of bias domains, such as selection bias (sequence generation and allocation sequence concealment), affected the risk of bias across all outcome measures in a trial. In case of high risk of selection bias, we marked all outcomes investigated in the associated trial as high risk. Otherwise, we did not perform a summary assessment of the risk of bias across all outcomes for a trial.

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

Risk of bias for an outcome across trials and across domains
These were our main summary assessments that we incorporated into our judgements about the certainty of the evidence in Summary of findings table 1. We defined outcomes as at low risk of bias when most information came from trials at low risk of bias, unclear risk when most information came from trials at low or unclear risk of bias, and high risk when a sufficient proportion of information came from trials at high risk of bias.

Medidas del efecto del tratamiento

When at least two trials were available for a comparison of a given outcome, we expressed dichotomous data as risk ratio (RR) or an odds ratio (OR) with 95% confidence interval (CI). For continuous outcomes measured on the same scale (e.g. weight loss in kg) we estimated the intervention effect using the mean difference (MD) with 95% CI. For continuous outcomes measuring the same underlying concept (e.g. health‐related quality of life) but applying different measurement scales, we calculated the standardised mean difference (SMD). We planned to calculate time‐to‐event data as hazard ratio (HR) with 95% CI with the generic inverse variance method.

The scales measuring health‐related quality of life could go in different directions. Some scales increase in values with improved health‐related quality of life, whereas other scales decrease in values with improved health‐related quality of life. To adjust for the different directions of the scales, we planned to multiply the scales that reported better health‐related quality of life with decreasing values by ‐1.

Cuestiones relativas a la unidad de análisis

We took 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 either combined groups to create a single pair‐wise comparison or we appropriately reduced the sample size so that the same participants did not contribute multiply (splitting the 'shared' group into two or more groups). Although the latter approach offers some solution for adjusting the precision of the comparison, it does not account for correlation arising from inclusion of the same set of participants in multiple comparisons (Higgins 2011b).

We planned to re‐analyse cluster‐RCTs that did not appropriately adjust for potential clustering of participants within clusters in their analyses. Variance of the intervention effects was planned to be inflated by a design effect (DEFF). Calculation of a DEFF involves estimation of an intra‐cluster correlation (ICC). We planned to obtain estimates of ICCs by contacting trial authors, or by imputing ICC values using either estimates from other included trials that reported ICCs or external estimates from empirical research (e.g. Bell 2013). We planned to examine the impact of clustering by performing sensitivity analyses.

Manejo de los datos faltantes

We tried to obtain missing data from trial authors and carefully evaluate important numerical data such as screened, randomly‐assigned participants as well as intention‐to‐treat (ITT), and as‐treated and per‐protocol populations. We investigated attrition rates (e.g. dropouts, losses to follow‐up, withdrawals), and we critically appraised issues concerning missing data and imputation methods (e.g. last observation carried forward (LOCF)).

In trials where the standard deviation of the outcome was not available at follow‐up or could not be recreated, we standardised by the average of the pooled baseline standard deviation (SD) from those trials in which this information was reported.

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

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

Evaluación de la heterogeneidad

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 identified heterogeneity (inconsistency) by visually inspecting the forest plots and by using a standard Chi² test with a significance level of α = 0.1 (Deeks 2017). 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); an I² statistic of 75% or more indicates a considerable level of heterogeneity (Higgins 2011b).

When heterogeneity was present, we attempted to determine possible reasons for it by examining individual trial and subgroup characteristics.

Evaluación de los sesgos de notificación

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 (Sterne 2017). Therefore, we planned to interpret the results carefully (Sterne 2011).

Síntesis de los datos

We undertook meta‐analysis only if we judged participants, interventions, comparisons and outcomes to be sufficiently similar to ensure an answer that was clinically meaningful. 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 with consideration to 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 presented in the Cochrane Handbook for Systematic Reviews of Interventions (Deeks 2017).

Trial sequential analyses

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, that is 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 2011b). Sequential monitoring boundaries for benefit, harm, or futility can be applied to meta‐analyses as well, called trial sequential monitoring boundaries (Higgins 2011; Wetterslev 2008). In a trial sequential analysis (TSA), the addition of each trial in a cumulative meta‐analysis is regarded as an interim meta‐analysis and helps to clarify if significance is reached 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 (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 (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 clinical important effect. We based the required information size on an a priori effect corresponding to a 10% relative risk reduction (RRR).

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

For the heterogeneity adjustment of the required information size we used the diversity (D²) estimated in the meta‐analyses of included trials.

We performed TSA on primary and secondary outcomes.

Análisis de subgrupos e investigación de la heterogeneidad

We expected the following characteristics to introduce clinical heterogeneity and carried out the following subgroup analyses (on our primary and secondary outcomes) including investigation of interactions (Altman 2003).

  • Trials with a long duration (≥ 2 years) versus trials with a short duration (< 2 years).

  • Trials including obese participants versus trials including non‐obese participants.

Análisis de sensibilidad

We performed sensitivity analyses on our primary and secondary outcomes to explore the influence of the following factors (when applicable) on effect sizes, by restricting the analysis to the following.

  • Published trials

  • Effect of 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

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

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

Certainty of the evidence

We presented the overall certainty of the evidence for each outcome specified below, 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 (KM and PK, LK or FG) independently rated the certainty of the evidence for each outcome.

We included an appendix titled 'Checklist to aid consistency and reproducibility of GRADE assessments' to help with the standardisation of the 'Summary of findings' tables (Meader 2014). Alternatively, we would have used the GRADEpro Guideline Development Tool (GDT) software and presented evidence profile tables as an appendix (GRADEproGDT 2015). We presented results for outcomes as described in the Types of outcome measures section. If meta‐analysis was not possible, we presented the results in a narrative format in the 'Summary of findings' table. We justified all decisions to downgrade the certainty of the evidence using footnotes, and we made comments to aid the reader's understanding of the Cochrane Review when necessary.

'Summary of findings' table

We presented a summary of the evidence in Summary of findings table 1. This provides key information about the best estimate of the magnitude of effect, in relative terms and as absolute differences for each relevant comparison of alternative management strategies, 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 using the methods described in the Cochrane Handbook for Systematic Reviews of Interventions (Schünemann 2017), along with Review Manager 5 (RevMan 5) table editor (Review Manager 2014). We reported the following outcomes, listed according to priority.

  • All‐cause mortality

  • Cardiovascular mortality

  • Serious adverse events

  • Non‐fatal stroke

  • Non‐fatal myocardial infarction

  • Microvascular complications (end‐stage renal disease, blindness or severe vision loss, amputation of lower extremity)

  • Health‐related quality of life

Results

Description of studies

For a detailed description of trials, see Table 1, Table 2, 'Characteristics of included studies', 'Characteristics of excluded studies, and 'Characteristics of ongoing studies'.

Results of the search

Our databases search identified 2754 records to be screened. We excluded most of the records on the basis of their titles and abstracts because they clearly did not meet the inclusion criteria. We assessed a total of 296 full‐text articles/records for eligibility. After screening, 32 trials with 41 trial arms, published in 164 publications/records finally met our inclusion criteria.

Handsearching of systematic reviews and reference lists identified two trials described in seven publications (Gerich 2005; Ristic 2007). Handsearching of manufacturers' websites identified eight records.

We excluded a total of 118 full‐text articles/records, 80 of these publications/records described 62 trials and 38 of these publications/records did not describe trials. We identified nine ongoing trials described in 11 publications/records (see 'Ongoing studies'). We identified two trials awaiting assessment (see 'Characteristics of studies awaiting classification'). For an overview of trial selection, please see Figure 1.


Trial flow diagram
 AHRQ: Agency for Healthcare Research and Quality; DPP4‐I: dipeptidyl‐peptidase 4 inhibitor; GLP1‐A: glucagon‐like peptide 1 analogue; M+S: metformin + sulphonylurea; SGLT2‐I: sodium‐glucose co‐transporter 2 inhibitor

Trial flow diagram
AHRQ: Agency for Healthcare Research and Quality; DPP4‐I: dipeptidyl‐peptidase 4 inhibitor; GLP1‐A: glucagon‐like peptide 1 analogue; M+S: metformin + sulphonylurea; SGLT2‐I: sodium‐glucose co‐transporter 2 inhibitor

Included studies

A detailed description of the characteristics of included trials is presented elsewhere (see Characteristics of included studies and Appendix 4; Appendix 5; Appendix 6; Appendix 7; Appendix 8; Appendix 9; Appendix 10; Appendix 11; Appendix 12; Appendix 13; Appendix 14). The following is a succinct overview.

Source of data

Twenty‐six trials reported data with relevance for this review, published in medical journals (Charbonnel 2005; Del Prato 2014; Del Prato 2015; Derosa 2005; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Filozof 2010; Gallwitz 2012a; Gallwitz 2012b; Gerich 2005; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Leiter 2015; Maffioli 2013; Matthews 2010; Nauck 2013; Petrica 2009; Petrica 2011; Ridderstråle 2014; Schernthaner 2015; Seck 2010; Vaccaro 2017).

Fourteen trials reported data in trials registers (Ahrén 2014; Del Prato 2014; Del Prato 2015; Gallwitz 2012a; Gallwitz 2012b; Göke 2013; Handelsman 2017; Home 2009; Leiter 2015; Nauck 2013; NCT00367055; Ridderstråle 2014; Schernthaner 2015; Seck 2010).

Six trials reported data on the manufacturer's website (Ahrén 2014; Gerich 2005; Hamann 2008; Matthews 2010; NCT00367055; Ristic 2007).

We contacted all trial authors or investigators by email (see Appendix 15). When important information was lacking on ongoing studies and excluded trials, we contacted investigators for clarification (see Appendix 15). We only received additional data on four included trials through correspondence with authors (Dei Cas 2017; Derosa 2005; Home 2009; Vaccaro 2017).

Comparisons

One trial compared M+S with metformin monotherapy (Derosa 2009a). Two trials compared M+S with metformin plus placebo (Ahrén 2014; Nauck 2013). Five trials compared M+S with metformin plus a GLP‐1 analogue (Ahrén 2014; Derosa 2010; Derosa 2011a; Gallwitz 2012a; Nauck 2013). Nine trials compared M+S with metformin plus a DPP‐4 inhibitor (Ahrén 2014; Dei Cas 2017; Del Prato 2014; Filozof 2010; Gallwitz 2012b; Göke 2013; Matthews 2010; Schernthaner 2015; Seck 2010). One trial compared M+S with metformin plus long‐acting DPP‐4 inhibitor (Handelsman 2017). Eleven trials compared M+S with metformin plus a thiazolidinedione (Charbonnel 2005; Derosa 2005; Derosa 2009a; Derosa 2011b; Hamann 2008; Home 2009; Maffioli 2013; NCT00367055; Petrica 2009; Petrica 2011; Vaccaro 2017). Three trials compared M+S with metformin plus a glinide (Derosa 2009b; Gerich 2005; Ristic 2007). Four trials compared M+S with metformin plus a SGLT‐2 inhibitor (Del Prato 2015; Hollander 2017; Leiter 2015; Ridderstråle 2014).

Several trials implemented more than two comparison groups within their trial designs. For an overview of trials, trial arms, comparators, interventions and number of randomised participants see Table 2.

Overview of trial populations

All, but seven trials provided information on sample size calculations (Derosa 2009b; Derosa 2010; Gerich 2005; NCT00367055; Petrica 2009; Petrica 2011; Seck 2010). All, but 10 trials reported the total number of participants screened (Derosa 2005; Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Filozof 2010; Maffioli 2013; NCT00367055; Ristic 2007). A total of 12,863 participants were randomised to M+S and 15,833 participants were randomised to a comparator. The percentage of participants finishing the trial was approximately 69% in the M+S group and 69% in the comparator groups. Number of randomised participants ranged from 22 to 1556 in the M+S groups and from 22 to 1765 in the comparator groups (Table 1).

Trial design

All of the 32 trials were randomised controlled trials with a parallel design. In 15 trials the primary trial structure had a non‐inferiority design (Ahrén 2014; Del Prato 2014; Del Prato 2015; Filozof 2010; Gallwitz 2012a; Gallwitz 2012b; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Home 2009; Leiter 2015; Matthews 2010; Ridderstråle 2014; Seck 2010).

Two trials had both a placebo group and an active comparator group (Ahrén 2014; Nauck 2013), the rest of the included trials had an active comparator group. Two trials were single‐centre trials (Dei Cas 2017; Maffioli 2013). Two trials did not report number of centres (Petrica 2009; Petrica 2011). The remaining trials were multicentre trials, defined as two or more centres. Seven trials were open‐labelled for participants and personnel (Dei Cas 2017; Gallwitz 2012a; Home 2009; NCT00367055; Petrica 2009; Petrica 2011; Vaccaro 2017). Two trials were single‐blind for investigators but not for the participants (Derosa 2010; Derosa 2011a). The remaining trials were double‐blinded for investigators and participants. Three trials did not report blinding of outcome assessors for any outcome (Dei Cas 2017; Gallwitz 2012a; NCT00367055). The remaining trials reported blinding of outcome assessors for one or more outcomes. Trials were performed between the years 2001 to 2017. The duration of the intervention ranged from 52 weeks to 208 weeks. Twenty trials had a run‐in period (Ahrén 2014; Del Prato 2014; Del Prato 2015; Derosa 2005; Derosa 2009b; Derosa 2010; Filozof 2010; Gallwitz 2012a; Gerich 2005; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Home 2009; Leiter 2015; Maffioli 2013; Nauck 2013; Ridderstråle 2014; Schernthaner 2015; Seck 2010). The remaining trials did not report any run‐in period. Two trials were terminated early (Matthews 2010; Vaccaro 2017). Matthews 2010 had a higher than expected discontinuation rate and fewer participants than expected reached the primary endpoint. Vaccaro 2017 had a lower than expected rate of primary endpoint events during follow‐up.

Eleven trials had an extended follow‐up period (Ahrén 2014; Charbonnel 2005; Del Prato 2015; Göke 2013; Hollander 2017; Home 2009; Leiter 2015; Nauck 2013; Ridderstråle 2014; Ristic 2007; Seck 2010). With the exception of Ristic 2007 all had high attrition rates between the original time of follow‐up and the extended follow‐up. Four trials reported that they were able to keep the double‐blinding conditions (Del Prato 2015; Göke 2013; Leiter 2015; Ristic 2007).

Settings

All trials were conducted in outpatient clinics.

Participants

All the trials included people with T2DM and with HbA1c ranging from 7% to 9% (Dei Cas 2017; Hollander 2017; Home 2009; Schernthaner 2015; Vaccaro 2017); 7% to 9.5% (Leiter 2015); 6.5% to 10% (Del Prato 2015; Gallwitz 2012b; Göke 2013; Seck 2010); 7% to 10% (Ahrén 2014; Del Prato 2014; Hamann 2008; Ridderstråle 2014); 7% to 11% (Gerich 2005; Nauck 2013); 6.8% to 9% (Ristic 2007); 7.5% to 11% (Charbonnel 2005; Filozof 2010); 6.5% to 8.5% (Matthews 2010; NCT00367055); 6.5% to 9% (Gallwitz 2012a; Handelsman 2017); more than 6.5% (Derosa 2009a); more than 7% (Derosa 2005; Derosa 2009b; Derosa 2011b; Petrica 2009; Petrica 2011) and more than 8% (Derosa 2010; Derosa 2011a; Maffioli 2013).

One trial did not report the duration of T2DM at baseline (Derosa 2009a). For the rest of the trials the mean duration of T2DM ranged from newly diagnosed to more than 10 years.

All trials included both genders. The percentage of women ranged from 29% to 65%.

The mean age of the participants ranged from 52 years to 73 years. One trial only included participants aged 65 years and more (Schernthaner 2015).

Mean HbA1c at baseline ranged from 7.3% to 9.3%.

Ten trials did not report any comorbidities, cointerventions or comedications (Ahrén 2014; Del Prato 2014; Filozof 2010; Gerich 2005; Handelsman 2017; Hollander 2017; Home 2009; Nauck 2013; NCT00367055; Ristic 2007).

Major exclusion criteria were type 1 diabetes mellitus or secondary forms of diabetes, diabetic complications (nephropathy, retinopathy, neuropathy), history of ketoacidosis, organ failure (renal, hepatic, heart), history of pancreatitis, new cardiovascular event or pregnancy.

Diagnosis

Five trials applied the diagnostic criteria of T2DM as defined by the European Association for the Study of Diabetes (EASD) 2007 guideline (Derosa 2009a; Derosa 2010; Derosa 2011a; Derosa 2011b; Maffioli 2013), four trials used the definition of the World Health Organization (WHO) 1999/2006 criteria (Gallwitz 2012a; Hamann 2008; Home 2009; NCT00367055), two trials used the American Diabetes Association (ADA) 2001 criteria (Derosa 2005; Derosa 2009b), one trial applied the ADA 1997 criteria (Dei Cas 2017), and one trial applied the ADA 2017 criteria (Hollander 2017). The remaining trials did not report diagnostic criteria for T2DM.

Interventions

Metformin was administered in all intervention and comparator arms and was mostly given in doses of 500 mg/day to 3000 mg/day (Appendix 4).

Second‐generation sulphonylurea was administered either as glibenclamide, in doses of 5 mg/day to 15 mg/day (Dei Cas 2017; Derosa 2009b; Derosa 2010; Derosa 2011b; Gerich 2005; Maffioli 2013); gliclazide, in doses of 30 to 320 mg/day (Charbonnel 2005; Filozof 2010; NCT00367055; Ristic 2007); or glipizide, in doses of 5 to 20 mg/day (Del Prato 2014; Del Prato 2015; Göke 2013; Seck 2010). Third‐generation sulphonylurea was administered as glimepiride, in doses of 1 mg/day to 8 mg/day (Ahrén 2014; Derosa 2005; Derosa 2009a; Derosa 2011a; Gallwitz 2012a; Gallwitz 2012b; Handelsman 2017; Hollander 2017; Home 2009; Leiter 2015; Matthews 2010; Nauck 2013; Petrica 2009; Petrica 2011; Ridderstråle 2014; Schernthaner 2015). Two trials administered more than one kind of sulphonylurea, that is, glibenclamide or gliclazide (Hamann 2008), or glibenclamide, gliclazide or glimepiride (Vaccaro 2017).

GLP‐1 analogues were administered as albiglutide, in doses of 30 mg to 50 mg once weekly (Ahrén 2014); exenatide, in doses of 10 μg/day to 20 μg/day (Derosa 2010; Derosa 2011a; Gallwitz 2012a); or liraglutide, in doses of 0.6 mg/day to 1.8 mg/day (Nauck 2013).

DPP‐4 inhibitors were administered as either sitagliptin, in doses of 100 mg/day (Ahrén 2014; Seck 2010); vildagliptin (in doses of 100 mg/day (Dei Cas 2017; Filozof 2010; Matthews 2010); alogliptin, in doses of 12.5 mg/day to 25 mg/day (Del Prato 2014); linagliptin (in doses of 5 mg/day (Gallwitz 2012b); or saxagliptin, in doses of 5 mg/day (Göke 2013; Schernthaner 2015).

Thiazolidinediones were administered as either rosiglitazone, in doses of 4 mg/day to 8 mg/day (Derosa 2005; Hamann 2008; Home 2009; NCT00367055; Petrica 2009); or pioglitazone, in doses of 15 mg/day to 45 mg/day (Derosa 2011b; Maffioli 2013; Vaccaro 2017).

Glinides were administered as nateglinide, in doses of 180 mg/day to 540 mg/day (Derosa 2009b; Gerich 2005; Ristic 2007).

SGLT‐2 inhibitors were administered as either dapagliflozin, in doses of 2.5 mg/day to 10 mg/day (Del Prato 2015); canagliflozin, in doses of 100 mg/day to 300 mg/day (Leiter 2015); empagliflozin, in doses of 25 mg/day (Ridderstråle 2014); or ertugliflozin, in doses of 5 mg/day to 15 mg/day (Hollander 2017).

Long‐acting DPP‐4 inhibitors were administered as omarigliptin, in doses of 25 mg/week (Handelsman 2017).

Two trials compared M+S with metformin plus placebo (Ahrén 2014; Nauck 2013).

Twelve trials reported counselling for diet and exercise (Del Prato 2015; Derosa 2005; Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Göke 2013; Maffioli 2013; Ridderstråle 2014; Schernthaner 2015; Seck 2010). One trial counselled on diet only (Charbonnel 2005). The remaining trials did not provide information about counselling.

Outcomes

Six trials did not define a primary outcome (Derosa 2009a; Derosa 2009b; Derosa 2010; Maffioli 2013; Petrica 2009; Petrica 2011). None of these trials was registered in ClinicalTrials.gov or had a design paper published.

Twenty trials were registered at ClinicalTrials.gov (Ahrén 2014; Dei Cas 2017; Del Prato 2014; Del Prato 2015; Filozof 2010; Gallwitz 2012a; Gallwitz 2012b; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Home 2009; Leiter 2015; Matthews 2010; Nauck 2013; NCT00367055; Ridderstråle 2014; Schernthaner 2015; Seck 2010; Vaccaro 2017).

Most trials used HbA1c as the primary outcome measure.

We included 32 trials. All the included trials but eight reported one or more of the primary outcomes of relevance for this review (Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Maffioli 2013; Petrica 2009; Petrica 2011). All the included trials but nine reported on all‐cause mortality (Dei Cas 2017; Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Maffioli 2013; Petrica 2009; Petrica 2011). All the included trials but nine reported on serious adverse events and non‐serious adverse events (Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Maffioli 2013; Petrica 2009; Petrica 2011; Ristic 2007), see Appendix 11. Three trials only reported adverse events leading to discontinuation (Derosa 2009a; Derosa 2010; Maffioli 2013). In three trials, the authors provided safety‐data without the number of participants included in the safety‐analysis (Derosa 2009b; Derosa 2011a; Derosa 2011b). One trial reported on adverse events for up to six months and 6 to 12 months separately (Ristic 2007).

One trial reported on health‐related quality of life (Nauck 2013). This trial measured the outcome 'impact of weight on quality of life' (IWQOL). Authors did not report results per intervention groups.

Four trials reported on 'amputation of lower extremity' (Dei Cas 2017; Derosa 2005; Hollander 2017; Vaccaro 2017). Three trials reported on 'end‐stage renal disease' (Nauck 2013; Dei Cas 2017; Derosa 2005). Two trials reported on 'blindness or severe vision loss' (Dei Cas 2017; Derosa 2005).

Three trials reported cost effectiveness data (Del Prato 2014; Del Prato 2015; Göke 2013).

For definitions of outcomes see Appendix 9 and Appendix 10.

Excluded studies

We excluded 118 publications/records after full‐text evaluation (see Characteristics of excluded studies) mainly for the following reasons. The trial did not compare interventions of interest (N = 45). Two of these trials investigated an investigational drug (EUCTR2004‐002549‐11‐FI (tesaglitazar); NCT01481116 (fasiglifam)) and one trial investigated a drug no longer approved for use (Rubin 2008 (muraglitazar)). Other reasons for exclusion included: duration of intervention less than 52 weeks (N = 15), not an RCT (N = 7), trial cancelled or withdrawn (N = 7) and different duration of the intervention between the intervention groups (N = 4). One trial compared interventions of interest (metformin compared with M+S), but the trial only reported on metformin compared with usual care (Cryer 2005). We contacted the trial authors to request data, but did not receive a reply. One trial ended prematurely and had no study results (EUCTR2009‐014727‐23‐IT). Six publications were expert reviews (Albarran 2013; Fleming 2015; Nishio 2015; Odawara 2015; Scheen 2016; Seufert 2014), one was a correspondence (Kannan 2015), one was a commentary on a systematic review (Bellary 2011) and two were analysing pooled data from multiple RCTs (Reid 2016; Rosenstock 2015). We identified 26 systematic reviews (Amate 2015; Aylsworth 2014; Belsey 2008; Chan 2015; Dai 2014; Foroutan 2016; Geng 2015; Goring 2014; Gu 2015; Guthrie 2015; Hershon 2016; Hou 2015; Kuecker 2016; Lim 2015; Liu 2014; Maruthur 2016; Mearns 2015; Mishriky 2015; Monami 2008; Phung 2010; Phung 2014; Rosenstock 2013; Varvaki 2016; Whalen 2015; Zhou 2015; Zintzaras 2014), and one conference abstract (Alvares 2015). This publication is listed in Additional references.

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 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 particular outcome was not measured in some trials)

Allocation

We judged 24 trials at low risk of selection bias regarding the method of randomisation and allocation concealment (Charbonnel 2005; Dei Cas 2017; Del Prato 2015; Derosa 2005; Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Gallwitz 2012a; Gallwitz 2012b; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Home 2009; Leiter 2015; Maffioli 2013; Nauck 2013; Ridderstråle 2014; Ristic 2007; Schernthaner 2015; Seck 2010; Vaccaro 2017). The remaining trials only reported that the participants were randomised but did not provide any further description (Ahrén 2014; Del Prato 2014; Filozof 2010; Gerich 2005; Matthews 2010; NCT00367055; Petrica 2009; Petrica 2011). Therefore, we judged these trials at unclear risk of bias regarding randomisation and allocation concealment.

We evaluated trial baseline data for our predefined prognostic baseline variables. None of the included trials reported data on all our key prognostic variables. However, all trials reported some variables of interest. None of the trials reporting prognostic variables showed important differences between the intervention groups.

Blinding

Blinding of participants and investigators for all outcomes was adequate in 21 trials (Ahrén 2014; Charbonnel 2005; Del Prato 2014; Del Prato 2015; Derosa 2005; Derosa 2009b; Derosa 2011b; Gallwitz 2012b; Gerich 2005; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Leiter 2015; Maffioli 2013; Matthews 2010; Nauck 2013; Ridderstråle 2014; Ristic 2007; Schernthaner 2015; Seck 2010). Trials ensured blinding of participants and investigators by using identical placebo tablets or injections. Seven trials were open‐label (Dei Cas 2017; Gallwitz 2012a; Home 2009; NCT00367055; Petrica 2009; Petrica 2011; Vaccaro 2017). Two trials were single‐blinded (Derosa 2010; Derosa 2011a).

In one trial the participants and investigators were blinded in the first 26 weeks of the intervention period followed by a 78‐week open‐label extension phase (Nauck 2013).

Eight trials described a blinded outcome committee evaluating cardiovascular and cerebrovascular events (Ahrén 2014; Del Prato 2014; Filozof 2010; Gallwitz 2012b; Home 2009; Matthews 2010; Ridderstråle 2014; Vaccaro 2017).

Where measured, all primary outcomes of this review were investigator‐assessed and we judged these at low risk of performance and detection bias. The trials reporting blood glucose measurements were all performed by the investigators and we judged these outcomes measures at low risk of performance and detection bias. Overall, the risk of performance bias and detection bias was low or unclear for our secondary outcomes.

Incomplete outcome data

All trials reported the complete number of participants randomised and finishing the trial. Two trials adequately addressed incomplete data for all outcomes (Dei Cas 2017; Derosa 2005). We judged the following outcomes to be at unclear or high risk of attrition bias in one or more trials: amputation of lower extremity, blindness or severe vision loss, end‐stage renal disease (Hollander 2017; Nauck 2013; Vaccaro 2017); hypoglycaemia (Ahrén 2014; Charbonnel 2005; Del Prato 2014; Gallwitz 2012b; Gerich 2005; Göke 2013; Hollander 2017; Home 2009; Matthews 2010; Nauck 2013; NCT00367055; Schernthaner 2015; Seck 2010); non‐fatal myocardial infarction, heart failure, non‐fatal stroke (Ahrén 2014; Charbonnel 2005; Del Prato 2014; Del Prato 2015; Gallwitz 2012a; Gallwitz 2012b; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Leiter 2015; Matthews 2010; Nauck 2013; Ridderstråle 2014; Schernthaner 2015; Seck 2010); non‐serious adverse events (Filozof 2010; Göke 2013; Nauck 2013); serious adverse events (Ahrén 2014; Charbonnel 2005; Del Prato 2014; Del Prato 2015; Filozof 2010; Gallwitz 2012a; Gallwitz 2012b; Gerich 2005; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Leiter 2015; Matthews 2010; Nauck 2013; NCT00367055; Ridderstråle 2014; Schernthaner 2015; Seck 2010; Vaccaro 2017); weight (Ahrén 2014; Charbonnel 2005; Del Prato 2014; Del Prato 2015; Filozof 2010; Gallwitz 2012a; Gallwitz 2012b; Gerich 2005; Göke 2013; Hamann 2008; Handelsman 2017; Home 2009; Leiter 2015; Matthews 2010; Nauck 2013; NCT00367055; Ridderstråle 2014; Ristic 2007; Schernthaner 2015; Seck 2010; Vaccaro 2017) and HbA1c (Ahrén 2014; Charbonnel 2005; Del Prato 2014; Del Prato 2015; Filozof 2010; Gallwitz 2012a; Gallwitz 2012b; Gerich 2005; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Home 2009; Leiter 2015; Matthews 2010; Nauck 2013; NCT00367055; Ridderstråle 2014; Ristic 2007; Schernthaner 2015; Seck 2010; Vaccaro 2017).

The reasons for a high risk of attrition bias for dichotomous outcomes were: high dropout rate; dropout rate not balanced between intervention groups; reason for dropout not balanced between intervention groups; no information on imputation method; missing data imputed with 'last observation carried forward' technique; proportion of missing outcomes compared with the observed event risk was substantial enough to result in potentially clinically relevant bias for the intervention effect estimate.

The reasons for an unclear or high risk of attrition bias for continuous outcomes were: high dropout rate; dropout rate not balanced between intervention groups; reason for dropout not balanced between intervention groups; no information on imputation method; missing data imputed with 'last observation carried forward' technique; only participants with a value at baseline and at a specified visit were analysed; only participants who completed the study (excluding participants receiving rescue therapy) were included in the analysis; analyses based on per protocol population; and substantial amount of missing outcomes to potentially result in clinically relevant bias for the observed effect size.

Selective reporting

Twenty‐two of the included trials had a published protocol (Ahrén 2014; Dei Cas 2017; Del Prato 2014; Del Prato 2015; Filozof 2010; Gallwitz 2012a; Gallwitz 2012b; Gerich 2005; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Home 2009; Leiter 2015; Matthews 2010; Nauck 2013; NCT00367055; Ridderstråle 2014; Ristic 2007; Schernthaner 2015; Seck 2010; Vaccaro 2017).

We judged 11 of the included trials at high risk of reporting bias for one or more of our outcomes (Derosa 2005; Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Filozof 2010; Gerich 2005; Maffioli 2013; Petrica 2009; Petrica 2011). For more details, see Appendix 8.

We judged nine trials as at high risk of selective outcome reporting regarding one or more of the primary outcomes with relevance to this review (Derosa 2005; Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Maffioli 2013; Petrica 2009; Petrica 2011); we judged 10 trials as high risk of selective outcome reporting regarding one or more of the secondary outcomes with relevance to this review (Derosa 2005; Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Gerich 2005; Maffioli 2013; Petrica 2009; Petrica 2011).

We judged two trials as high risk of selective outcome reporting regarding one or more of the additional explorative outcomes of relevance to this review (Derosa 2009a; Filozof 2010).

Other potential sources of bias

Twenty‐two trials received support or funding from a pharmaceutical company (Ahrén 2014; Charbonnel 2005; Dei Cas 2017; Del Prato 2014; Del Prato 2015; Filozof 2010; Gallwitz 2012a; Gallwitz 2012b; Gerich 2005; Göke 2013; Hamann 2008; Handelsman 2017; Hollander 2017; Home 2009; Leiter 2015; Matthews 2010; Nauck 2013; NCT00367055; Ridderstråle 2014; Ristic 2007; Schernthaner 2015; Seck 2010). For some comparisons, the same author had performed similar trials (Derosa 2005; Derosa 2009a; Derosa 2010; Derosa 2011a; Derosa 2011b; Maffioli 2013).

Effects of interventions

See: Summary of findings for the main comparison Metformin‐sulphonylurea (second‐ or third‐generation) combination therapy compared with metformin plus another antidiabetic drug for adults with type 2 diabetes mellitus

See summary of findings Table for the main comparison for details on the most important comparisons of this review (M+S compared with metformin plus GLP‐1 analogue, metformin plus DPP‐4 inhibitor, metformin plus thiazolidinedione, metformin plus glinide and metformin plus SGLt‐2 inhibitor).

Baseline characteristics

For details on baseline characteristics, see Appendix 5 and Appendix 6.

Metformin‐sulfonylurea combination therapy versus metformin plus insulin

We identified no trials comparing M+S with metformin plus insulin.

Metformin‐sulfonylurea combination therapy versus metformin monotherapy

One trial compared M+S combination therapy with metformin monotherapy (Derosa 2009a). The intervention group received metformin in doses of 850 mg/day and glimepiride in doses of 2 mg/day to 6 mg/day. The comparator group received metformin in doses of 1000 mg/day to 3000 mg/day.

Primary outcomes

The included trial did not report on all‐cause mortality, health‐related quality of life or serious adverse events.

Secondary outcomes

The included trial did not report on cardiovascular mortality, non‐fatal myocardial infarction, heart failure, non‐fatal stroke, amputation of lower extremity, blindness or severe vision loss, end‐stage renal disease, non‐serious adverse events, hypoglycaemia or socioeconomic effects.

Additional explorative outcomes
Weight

The included trial did not report on weight.

HbA1c

The included trial reported final HbA1c measurements of 7.8% (SD 0.4) and 7.9% (SD 0.5) in the M+S group compared with the metformin monotherapy, respectively. They did not report the number of participants included in the analysis.

Metformin‐sulfonylurea combination therapy versus metformin plus placebo

Two trials compared M+S combination therapy with metformin plus placebo (Ahrén 2014; Nauck 2013). Both trials administered glimepiride, given in doses of 1 mg/day to 4 mg/day; and metformin in doses of ≥ 1500 mg/day. Both trials had multiple intervention arms with glimepiride, GLP‐1 analogue, DPP‐4 inhibitor or placebo in combination with metformin (Ahrén 2014), as well as glimepiride, GLP‐1 analogue or placebo in combination with metformin (Nauck 2013). For details of the certainty of the evidence see Appendix 16.

Primary outcomes
All‐cause mortality

Both included trials reported data on all‐cause mortality (low‐certainty evidence because of serious imprecision; Analysis 1.1). One trial reported 6 deaths out of 307 participants (2%) and 1 death out of 101 participants (1%) in the M+S combination group compared with the metformin plus placebo group, respectively (Ahrén 2014). However, the number of deaths was unclear due to varied reporting. We contacted the trial authors for clarification but did not receive a reply. To be sure to account for all deaths, we extracted data from the publication that reported the highest number. One trial reported data after an 18‐month, open‐label extension: there were no deaths in 242 participants and no deaths in 121 participants in the M+S combination group compared with the metformin plus placebo group, respectively (Nauck 2013).

Health‐related quality of life

None of the included trials reported on this outcome.

Serious adverse effects

Both included trials reported that a total of 84 participants experienced a serious adverse event; in the M+S group 60/549 (10.9%) participants had a serious adverse event compared with 24/222 (10.8%) participants in the metformin plus placebo group (RR 0.97, 95% CI 0.59 to 1.61; P = 0.91; 2 trials; 771 participants; very low‐certainty evidence because of attrition bias and serious imprecision; Analysis 1.2). One trial reported data after an 18‐month, open‐label extension phase (Nauck 2013).

TSA showed that 2.45% of the diversity‐adjusted required information size to detect or reject a 10% relative risk reduction (RRR) had been accrued. Diversity was 21%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Secondary outcomes
Cardiovascular mortality

Both included trials reported data on cardiovascular mortality (low‐certainty evidence because of serious imprecision; Analysis 1.3). One trial reported 1 cardiovascular death in 307 (0.3%) participants in the M+S group compared with 1 cardiovascular death in 101 (1%) participants in the metformin plus placebo group (Ahrén 2014). One trial reported that no participants died in either intervention group after an 18‐month, open‐label extension phase (Nauck 2013).

Non‐fatal myocardial infarction

Both included trials reported that a total of three participants experienced a non‐fatal myocardial infarction; in the M+S group 2/549 (0.4%) participants had a myocardial infarction compared with 1/222 (0.5%) participants in the metformin plus placebo group (RR 0.63, 95% CI 0.08 to 5.10; P = 0.67; 2 trials; 771 participants; very low‐certainty evidence because of attrition bias and serious imprecision; Analysis 1.4). One trial reported data after an 18‐month, open‐label extension phase (Nauck 2013).

TSA showed that 0.13% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was zero. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Heart failure

Both included trials reported data on heart failure (Analysis 1.5). One trial reported one heart failure in 307 participants (0.3%) and no heart failure in 101 participants in the M+S group compared with the metformin plus placebo group, respectively (Ahrén 2014). One trial reported that no participants experienced a heart failure in either intervention group after an 18‐month, open‐label extension phase (Nauck 2013).

Non‐fatal stroke, amputation of lower extremity, blindness or severe vision loss

None of the included trials reported on this outcome.

End‐stage renal disease

One trial reported that no participants had end‐stage renal disease in either intervention group after an 18‐month, open‐label extension phase (very low‐certainty evidence because of attrition bias and serious imprecision; Nauck 2013).

Non‐serious adverse events

Both included trials reported that a total of 505 participants experienced a non‐serious adverse event: in the M+S group 386/549 (70.3%) participants had a non‐serious adverse event compared with 119/222 (53.6%) participants in the metformin plus placebo group (random RR 1.25, 95% CI 0.96 to 1.64; P = 0.10; fixed RR 1.24, 95% CI 1.10 to 1.41; P < 0.001; 2 trials; 771 participants; Analysis 1.6). None of the trials provided a detailed definition of the outcome. One trial reported data after an 18‐month, open‐label extension phase (Nauck 2013).

TSA showed that 5.8% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 78%. The TSA‐adjusted 95% CI was 0.47 to 3.32.

Hypoglycaemia

Both included trials reported that a total of 181 participants experienced a mild or moderate hypoglycaemic episode; in the M+S group there were 160/549 (29.1%) participants with hypoglycaemic episodes compared with 21/222 (9.5%) participants in the metformin plus placebo group (random RR 3.93, 95% CI 0.71 to 21.88; P = 0.12; fixed RR 2.87, 95% CI 1.90 to 4.34; P < 0.001; 2 trials; 771 participants; Analysis 1.7 in favour of metformin plus placebo). Ahrén 2014 defined mild or moderate hypoglycaemia by blood glucose levels 3.9 mmol/L or lower and Nauck 2013 used blood glucose values 3.1 mmol/L or lower. One trial reported data after an 18‐month, open‐label extension phase (Nauck 2013).

TSA showed that 0.18% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 93%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Both included trials reported data on serious hypoglycaemia (Analysis 1.8). One trial reported one participant with serious hypoglycaemia out of 307 participants (0.3%) and no participants with serious hypoglycaemia out of 101 participants in the M+S group compared with the metformin plus placebo group, respectively (Ahrén 2014). One trial reported that no participants had serious hypoglycaemia in either intervention group after an 18‐month, open‐label extension phase (Nauck 2013).

Socioeconomic effects

None of the included trials reported on this outcome.

Additional explorative outcomes
Weight

Both included trials reported weight change in favour of metformin plus placebo (mean difference (MD) 3.4 kg, 95% CI 1.4 to 5.4; P = 0.001; 2 trials; 476 participants; Analysis 1.9). Nauck 2013 reported data after an 18‐month, open‐label extension phase.

HbA1c

Both included trials reported change in HbA1c in favour of M+S (random MD −0.5%, 95% CI −1.1 to 0.1; P = 0.13; fixed MD −0.6%, 95% CI −0.8 to −0.4; P < 0.001; 2 trials; 472 participants; Analysis 1.10). One trial reported data after an 18‐month, open‐label extension phase (Nauck 2013).

Metformin‐sulfonylurea combination therapy versus metformin plus GLP‐1 analogue

Five trials compared M+S with metformin plus a GLP‐1 analogue (Ahrén 2014; Derosa 2010; Derosa 2011a; Gallwitz 2012a; Nauck 2013). Four trials administered glimepiride in doses of 2 mg/day to 6 mg/day (Ahrén 2014; Derosa 2011a; Gallwitz 2012a; Nauck 2013), and one trial administered glibenclamide in doses of 15 mg/day (Derosa 2010). Three trials administered exenatide in doses of 10 μg/day to 20 μg/day (Derosa 2010; Derosa 2011a; Gallwitz 2012a), one trial administered albiglutide in doses of 30 mg/week to 50 mg/week (Ahrén 2014), and one trial administered liraglutide in doses of 0.6 mg/day to 1.8 mg/day (Nauck 2013). Metformin was given in doses of 1000 mg/day to 1500 mg/day or more.

Primary outcomes
All‐cause mortality

Three trials reported that a total of 22 participants died: in the M+S group 11/1057 participants (1.0%) died compared with 11/1537 participants (0.7%) in the metformin plus GLP‐1 analogue group (RR 1.15, 95% CI 0.49 to 2.67; P = 0.75; 3 trials; 2594 participants; low‐certainty evidence; Analysis 2.1). Calculation of the 95% prediction interval did not provide a meaningful estimate. One trial reported data after an 18‐month, open‐label extension phase (Nauck 2013).

Derosa 2011a reported that no participants died but did not provide the number of participants included in the analysis.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to trials with low risk of selection bias did not change the direction of the effect estimate (RR 0.93, 95% CI 0.30 to 2.93; P = 0.91; 2 trials; 1985 participants). We could not perform sensitivity analysis excluding large trials because none of the included trials for this comparison had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company since all trials received funding from a pharmaceutical company.

TSA showed that 0.61% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was zero. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Health‐related quality of life

We did not identify trials with data on health‐related quality of life for this comparison. One trial reported on 'impact of weight on quality of life' (IWQOL; Nauck 2013). They reported the result separately for each intervention group.

Serious adverse effects

Three trials reported that a total of 322 participants experienced a serious adverse event: in the M+S group 128/1057 participants (12.1%) had a serious adverse event compared with 194/1537 participants (12.6%) in the metformin plus GLP‐1 analogue group (RR 0.90, 95% CI 0.73 to 1.11; P = 0.32; 3 trials; 2594 participants; very low‐certainty evidence; Analysis 2.2). The 95% prediction interval ranged between 0.23 and 3.51. One of the trials reported data after an 18‐month, open‐label extension (Nauck 2013). Derosa 2011a reported that no participants experienced serious adverse events but did not provide the number of participants included in the analysis.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis according to trials with low risk of selection bias and duration of intervention (excluding trials with duration of intervention longer than 104 weeks) contained the same two trials (Gallwitz 2012a; Nauck 2013). The effect estimate did not change substantially (RR 0.94, 95% CI 0.73 to 1.20; P = 0.60; 2 trials; 1985 participants). We could not perform sensitivity analysis excluding large trials because no trials had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 12.4% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was zero. The TSA‐adjusted 95% CI was 0.38 to 2.15.

Secondary outcomes
Cardiovascular mortality

Three trials reported on cardiovascular disease (Ahrén 2014; Derosa 2011a; Nauck 2013). Derosa 2011a reported that no participants died due to cardiovascular disease but did not provide the number of participants included in the analysis. Nauck 2013 reported that no participants died due to cardiovascular disease in any of the intervention groups referring to the 18‐month, open‐label extension phase.

Ahrén 2014 reported that in the M+S group 1/307 (0.3%) participants died of cardiovascular disease compared with 1/302 (0.3%) participants in the metformin plus GLP‐1 analogue group (Analysis 2.3; low‐certainty evidence).

Non‐fatal myocardial infarction

Two trials reported that a total of eight participants experienced a non‐fatal myocardial infarction: in the M+S group 2/549 (0.4%) participants had a non‐fatal myocardial infarction compared with 6/1026 (0.6%) participants in the metformin plus GLP‐1 analogue group (RR 0.57, 95% CI 0.12 to 2.82; P = 0.49; 2 trials; 1575 participants; very low‐certainty evidence; Analysis 2.4).

Nauck 2013 reported data after an 18‐month, open‐label extension phase. Derosa 2011a reported that no participants experienced a non‐fatal myocardial infarction but did not provide the number of participants included in the analysis.

We could not perform sensitivity analyses due to lack of data.

TSA showed that 0.4% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was zero. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Heart failure

Four trials reported that a total of five participants developed heart failure: in the M+S group 1/1057 (0.1%) participants developed heart failure compared with 4/1537 (0.3%) participants in the metformin plus GLP‐1 analogue group (RR 0.54, 95% CI 0.10 to 2.77; P = 0.46; 3 trials; 2594 participants; Analysis 2.5). Calculation of the 95% prediction interval did not provide a meaningful estimate.

Nauck 2013 reported data after an 18‐month, open‐label extension phase. Derosa 2011a reported that no participants developed heart failure but did not provide the number of participants included in the analysis.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to trials with low risk of selection bias and duration of intervention (excluding trials with duration of intervention more than 104 weeks) contained the same two trials (Gallwitz 2012a; Nauck 2013). The effect estimate did not change substantially (RR 0.58, 95% CI 0.06 to 5.54; P = 0.63; 2 trials; 1985 participants). We could not perform sensitivity analysis excluding large trials because no trials had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 0.26% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was zero. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Non‐fatal stroke

Derosa 2011a reported that no participants experienced a stroke during the intervention period but did not provide the number of participants included in the analysis.

Amputation of lower extremity

Derosa 2011a reported that no participants had amputation of lower extremity during the intervention period but did not provide the number of participants included in the analysis.

Blindness or severe vision loss

Derosa 2011a reported that no participants had blindness or severe vision loss during the intervention period but did not provide the number of participants included in the analysis.

End‐stage renal disease

One trial provided data on end‐stage renal disease, however only after the 18‐month, open‐label extension phase (Nauck 2013). In the M+S group no participants out of 242 participants had end‐stage renal disease compared with 1 participant out of 724 (0.1%) participants in the metformin plus GLP‐1 analogue group (Analysis 2.6).

Derosa 2011a reported that no participants had end‐stage renal disease during the intervention period but did not provide the number of participants included in the analysis.

Non‐serious adverse events

Three trials reported that a total of 1621 participants experienced a non‐serious adverse event: in the M+S group 634/1057 (60.0%) participants had a non‐serious adverse event compared with 987/1537 (64.2%) participants in the metformin plus GLP‐1 analogue group (Analysis 2.7). Due to substantial heterogeneity, aggregating the trials in a meta‐analysis was not appropriate. With regard to the M+S groups, the trial with most of the non‐serious adverse events had two placebo interventions (GLP‐1 analogue as subcutaneous injection and placebo DPP‐4 inhibitor as tablet) in addition to M+S (Ahrén 2014).

One trial was open‐label and did not apply placebo (Gallwitz 2012a), and one trial (Nauck 2013), reported data after an 18‐month, open‐label extension. With regard to the metformin plus GLP‐1 analogue treatment arms, each trial used a different GLP‐1 analogue: one administered albiglutide (Ahrén 2014), and one administered exenatide (Gallwitz 2012a), or liraglutide (Nauck 2013).

Derosa 2011a reported that no participants experienced non‐serious adverse events but did not provide the number of participants included in the analysis.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analyses restricted to trials with low risk of selection bias and duration of intervention (excluding trials with duration of intervention longer than 104 weeks) contained the same two trials (Gallwitz 2012a; Nauck 2013). The effect estimate was in favour of M+S combination therapy (RR 0.84, 95% CI 0.76 to 0.92; P = 0.0001; 2 trials; 1985 participants). We could not perform sensitivity analysis excluding large trials because no trials had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

Hypoglycaemia

Three trials reported that a total of 569 participants experienced a mild or moderate hypoglycaemic episode: in the M+S group 400/1057 (37.8%) participants had a hypoglycaemic episode compared with 169/1537 (11.0%) participants in the metformin plus GLP‐1 analogue group (RR 3.24, 95% CI 2.05 to 5.13; P < 0.001; 3 trials; 2594 participants; Analysis 2.8; in favour of metformin plus GLP‐1 analogue). Calculation of the 95% prediction interval did not provide a meaningful estimate. The trial with mostly mild or moderate hypoglycaemic events defined hypoglycaemia as any sign or symptom of hypoglycaemia or blood glucose 3.9 mmol/L or less (Gallwitz 2012a), whereas the other trials defined hypoglycaemia as symptoms of hypoglycaemia plus blood glucose values 3.9 mmol/L or less, or blood glucose values 3.9 mmol/L or less without symptoms (Ahrén 2014), or symptoms of hypoglycaemia or blood glucose values 3.1 mmol/L or less (Nauck 2013). Nauck 2013 reported data after an 18‐month, open‐label extension phase.

Derosa 2011a reported two events of mild or moderate hypoglycaemia in the glimepiride group and one in the exenatide group but did not provide the number of participants included in the analysis.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analyses restricted to trials with low risk of selection bias and duration of intervention (excluding trials with duration of intervention longer than 104 weeks) contained the same two trials (Gallwitz 2012a; Nauck 2013). We could not perform sensitivity analysis excluding large trials because no trials had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

Only three participants reported serious hypoglycaemia: in the M+S group 1/1057 (0.1%) participants had a serious hypoglycaemic event compared with 2/1537 (0.1%) participants in the metformin plus GLP‐1 analogue group (RR 1.00, 95% CI 0.16 to 6.30; P = 1.00; 3 trials; 2594 participants; Analysis 2.9).

Nauck 2013 reported data after an 18‐month, open‐label extension phase. Derosa 2011a reported that no event of serious hypoglycaemia was observed but did not provide the number of participants included in the analysis.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analyses restricted to trials with low risk of selection bias and duration of intervention (excluding trials with duration of intervention longer than 104 weeks) contained the same two trials (Gallwitz 2012a; Nauck 2013). The effect estimate did not change substantially (RR 0.58, 95% CI 0.06 to 5.54; P = 0.63; 2 trials; 1985 participants). We could not perform sensitivity analysis excluding large trials because no trial had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA for severe hypoglycaemia showed that 0.09% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was zero. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Socioeconomic effects

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

Additional explorative outcomes
Weight

Five trials reported weight change in favour of metformin plus GLP‐1 analogue (MD 5.5 kg, 95% CI 3.6 to 7.5; P < 0.001; 5 trials; 1777 participants; Analysis 2.10). Calculation of the 95% prediction interval did not provide a meaningful estimate. Heterogeneity in the findings could have been caused by the various durations of follow‐up, ranging from 52 weeks to 156 weeks, and various doses of glimepiride ranging from 1 mg/day to 6 mg/day. Hollander 2017 did not report the number of participants included in the analysis.

Nauck 2013 reported data after an 18‐month, open‐label extension phase.

HbA1c

Five trials reported change in HbA1c (MD 0.01%, 95% CI ‐0.2 to 0.2; P = 0.91; 5 trials; 2346 participants; Analysis 2.11). The 95% prediction interval ranged between 0% and 1.0%.

Nauck 2013 reported data after an 18‐month, open‐label extension phase. Trials with 52 weeks of treatment reported at greater decrease in Hba1c with sulphonylurea treatment, whereas trials with 104 weeks of treatment or more reported a greater decrease in HbA1c with GLP‐1 analogue treatment.

TSA showed that 0.61% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was 91%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Metformin‐sulfonylurea combination therapy versus metformin plus DPP‐4 inhibitor

Nine trials compared M+S combination therapy with metformin plus a DPP‐4 inhibitor (Ahrén 2014; Dei Cas 2017; Del Prato 2014; Filozof 2010; Gallwitz 2012b; Göke 2013; Matthews 2010; Schernthaner 2015; Seck 2010). Four trials administered glimepiride in doses of 1 mg/day to 6 mg/day (Ahrén 2014; Gallwitz 2012b; Matthews 2010; Schernthaner 2015), three trials administered glipizide in doses of 5 mg/day to 20 mg/day (Del Prato 2014; Göke 2013; Seck 2010), one trial administered glibenclamide in doses of 10 mg/day (Dei Cas 2017), and one trial administered gliclazide in doses of 80 mg/day to 320 mg/day (Filozof 2010). Three trials administered vildagliptin in doses of 100 mg/day (Dei Cas 2017; Filozof 2010; Matthews 2010), two trials administered sitagliptin in doses of 100 mg/day (Ahrén 2014; Seck 2010), two trials administered saxagliptin in doses of 5 mg/day ( Göke 2013; Schernthaner 2015), one trial administered alogliptin (in doses of 12.5 mg/day to 25 mg/day (Del Prato 2014), and one trial administered linagliptin in doses of 5 mg/day (Gallwitz 2012b). One trial administered metformin at any dose (Schernthaner 2015), the remaining trials administered metformin in doses of 1500 mg/day or more.

Primary outcomes
All‐cause mortality

Nine trials reported that a total of 59 participants died: in the M+S group 33 participants died out of 5387 (0.6%) participants, compared with 26 deaths in 6307 (0.4%) participants in the metformin plus DPP‐4 inhibitor group (RR 1.32, 95% CI 0.76 to 2.28; P = 0.32; 9 trials; 11,694 participants; low‐certainty evidence; Analysis 3.1). The 95% prediction interval ranged between 0.68 and 2.55.

A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.77).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to trials with low risk of selection bias did not substantially change the effect estimate (RR 1.29, 95% CI 0.41 to 4.10; P = 0.66; 5 trials; 4363 participants). Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 1.52, 95% CI 0.81 to 2.88; P = 0.19; 8 trials; 8595 participants). We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials had received funding from a pharmaceutical company.

TSA showed that 2.4% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was zero. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Health‐related quality of life

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

Serious adverse effects

Nine trials reported that a total of 1514 participants experienced a serious adverse event: in the M+S group 735/5387 (13.6%) participants had a serious adverse event compared with 779/6307 (12.4%) participants in the metformin plus DPP‐4 inhibitor group (RR 1.07, 95% CI 0.97 to 1.18; P = 0.17; 9 trials; 11,694 participants; very low‐certainty evidence; Analysis 3.2). The 95% prediction interval ranged between 0.95 and 1.20.

A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.82).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to only trials with low risk of selection bias did not substantially change the effect estimate (RR 1.09, 95% CI 0.93 to 1.28; P = 0.27; 5 trials; 4363 participants). Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 1.06, 95% CI 0.94 to 1.20; P = 0.33; 8 trials; 8595 participants). We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 61% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was zero. The TSA‐adjusted 95% CI was 0.93 to 1.23.

Secondary outcomes
Cardiovascular mortality

Six trials reported that a total of 20 participants died due to cardiovascular disease: in the M+S group 11 participants died out of 2989 (0.4%) participants compared with 9 out of 3885 (0.2%) participants in the metformin plus DPP‐4 inhibitor group (RR 1.54, 95% CI 0.63 to 3.79; P = 0.34; 6 trials; 6874 participants; low‐certainty evidence; Analysis 3.3). The 95% prediction interval ranged between 0.43 and 5.52. Two trials reported a composite outcome of cardiovascular and cerebrovascular events (Filozof 2010: M+S 12/493 (2.4%) participants, metformin plus DPP‐4 inhibitor 7/510 (1.4%) participants; Matthews 2010: M+S 60/1546 (3.9%) participants, metformin plus DPP‐4 inhibitor 59/1553 (3.8%) participants).

We could not perform a test for subgroup differences according to duration of follow‐up due to lack of data.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to only trials with low risk of selection bias did not substantially change the effect estimate (RR 1.72, 95% CI 0.44 to 6.69; P = 0.43; 4 trials; 3645 participants).

We could not perform sensitivity analysis excluding large trials because no trials had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 0.92% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was zero. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Non‐fatal myocardial infarction

Six trials reported that a total of 28 participants experienced a non‐fatal myocardial infarction: in the M+S group 15/2989 (0.5%) participants had a myocardial infarction compared with 13/3885 (0.3%) participants in the metformin plus DPP‐4 inhibitor group (RR 1.45, 95% CI 0.69 to 3.07; P = 0.33; 6 trials; 6874 participants; very low‐certainty evidence; Analysis 3.4). The 95% prediction interval ranged between 0.50 and 4.20.

We could not perform a test for subgroup differences according to duration of follow‐up due to lack of data.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to trials with low risk of selection bias did not substantially change the effect estimate (RR 1.44, 95% CI 0.55 to 3.77; P = 0.45; 4 trials; 3645 participants). We could not perform sensitivity analysis excluding large trials because no trials had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 0.95% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was zero. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Heart failure

Eight trials reported that a total of 29 participants developed heart failure: in the M+S group 15/4894 (0.3%) participants developed heart failure compared with 14/5797 (0.2%) participants in the metformin plus DPP‐4 inhibitor group (RR 1.05, 95% CI 0.47 to 2.34; P = 0.90; 8 trials; 10,691 participants; Analysis 3.5). The 95% prediction interval ranged between 0.39 and 2.86.

A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.08).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to only trials with low risk of selection bias did not substantially change the direction of the effect estimate (RR 1.21, 95% CI 0.38 to 3.78; P = 0.75; 5 trials; 4363 participants). Sensitivity analysis excluding large trials did not substantially change the direction of the effect estimate (RR 1.07, 95% CI 0.42 to 2.69; P = 0.89; 7 trials; 7592 participants). We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

Non‐fatal stroke

Four trials reported that a total of 22 participants experienced a non‐fatal stroke: in the M+S group 14/2098 (0.7%) participants had a non‐fatal stroke compared with 8/2995 (0.3%) participants in the metformin plus DPP‐4 inhibitor group (RR 2.21, 95% CI 0.74 to 6.58; P = 0.15; 4 trials; 5093 participants; very low‐certainty evidence; Analysis 3.6). The 95% prediction interval ranged between 0.12 and 40.89.

We could not perform subgroup analysis due to lack of data.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis restricted to trials with low risk of selection bias due to lack of data. We could not perform sensitivity analysis excluding long trials because all trials had a follow‐up of 104 weeks or less. We could not perform sensitivity analysis excluding large trials because no trials had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 0.76% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was 23%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Amputation of lower extremity

Dei Cas 2017 did not observe amputation of the lower extremity in either intervention group (64 participants; very low‐certainty evidence).

Blindness or severe vision loss

Dei Cas 2017 did not observe blindness or severe vision loss in either intervention group (64 participants; very low‐certainty evidence).

End‐stage renal disease

Dei Cas 2017 did not observe end‐stage renal disease in either intervention group (64 participants; very low‐certainty evidence).

Non‐serious adverse events

Seven trials reported that a total of 4751 participants experienced a non‐serious adverse event: in the M+S group 2156/3348 (64.4%) participants had a non‐serious adverse event compared with 2595/4244 (61.1%) participants in the metformin plus DPP‐4 inhibitor group (RR 1.18, 95% CI 1.03 to 1.35; P = 0.02; 7 trials; 7592 participants; Analysis 3.7 in favour of metformin plus DPP‐4 inhibitor). The 95% prediction interval ranged between 0.73 and 1.91. There was substantial heterogeneity, which could be caused by various definitions of the outcome (most trials did not adequately specify this outcome measure).

A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.11).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to trials with low risk of selection bias did not substantially change the effect estimate (RR 1.27, 95% CI 1.06 to 1.52; P = 0.01; 5 trials; 4363 participants). We could not perform sensitivity analysis excluding large trials because no trials had more than 1000 participants randomised to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 21.5% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 94%. The TSA‐adjusted 95% CI was 0.88 to 1.58.

Hypoglycaemia

Seven trials reported that a total of 1544 participants experienced a mild or moderate hypoglycaemic event: in the M+S group 1359/4535 (30.0%) participants had a mild or moderate hypoglycaemic episode compared with 185/5438 (3.4%) participants in the metformin plus DPP‐4 inhibitor group (RR 7.42, 95% CI 4.77 to 11.53; P < 0.001; 7 trials; 9973 participants; Analysis 3.8 in favour of metformin plus DPP‐4 inhibitor). The 95% prediction interval ranged between 1.86 and 29.66. There was substantial heterogeneity, probably caused by various definitions of the outcome. All trials that stated a definition of outcome defined mild/moderate hypoglycaemia in different ways (Ahrén 2014; Del Prato 2014; Göke 2013; Matthews 2010).

The test for subgroup differences analysing trials according to duration of intervention showed a statistically significant difference between subgroups (P = 0.04; Analysis 3.8). However, CIs overlap indicating that in fact there was no true interaction.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to trials with low risk of selection bias did not substantially change the effect estimate (RR 6.71, 95% CI 3.94 to 11.44; P < 0.001; 4 trials; 3645 participants). Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 6.00, 95% CI 4.33 to 8.32; P < 0.001; 6 trials; 6874 participants). We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 1.4% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was 88%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Eight trials reported that a total of 55 participants experienced a serious hypoglycaemic event: in the M+S group 51/4894 (1.0%) participants had a serious hypoglycaemic episode compared with 4/5797 (0.1%) participants in the metformin plus DPP‐4 inhibitor group (RR 8.04, 95% CI 3.31 to 19.53; P < 0.01; 8 trials; 10,691 participants; Analysis 3.9 in favour of metformin plus DPP‐4 inhibitor). The 95% prediction interval ranged between 2.66 and 24.35.

A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.78).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis restricted to trials with low risk of selection bias did not substantially change the effect estimate (RR 8.62, 95% CI 2.81 to 26.43; P = 0.0002; 5 trials; 4363 participants). Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 6.93, 95% CI 2.72 to 17.65; P < 0.001; 7 trials; 7592 participants). We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 0.62% of the diversity‐adjusted information size had been accrued to detect or reject a 10% RRR. Diversity was 0%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Socioeconomic effects

Two trials performed economic analyses of trial data (Del Prato 2014; Göke 2013).

One trial presented an economic analysis using the IMS Core Diabetes Model (Del Prato 2014). Treatment with alogliptin 12.5 mg or 25 mg compared with sulphonylurea was associated with an incremental cost‐effectiveness ratio of GBP 10,959 or GBP 7217 per quality adjusted life year, respectively.

One trial presented an economic analysis using the Cardiff Stochastic Simulation Cost‐utility Model (Göke 2013). The overall mean cost per quality adjusted life year gained with saxagliptin plus metformin compared with M+S was GBP 7888.

Additional explorative outcomes
Weight

Nine trials reported weight change. Metformin plus DPP‐4 inhibitor compared with M+S combination therapy resulted in a weight loss of 2.2 kg (95% CI 1.7 to 2.6; P < 0.001; 9 trials; 10,228 participants; Analysis 3.10). The 95% prediction interval ranged between 0.8 kg and 3.5 kg.

HbA1c

Nine trials reported HbA1c (random MD −0.1%, 95% CI −0.1 to 0.03; P = 0.25; 9 trials; 9320 participants; fixed MD −0.1%, 95% CI −0.1 to −0.04; P < 0.001; Analysis 3.11 in favour of M+S). The 95% prediction interval ranged between −0.3% and 0.2%.

Metformin‐sulfonylurea combination therapy versus metformin plus a long‐acting DPP‐4 inhibitor

One trial compared M+S with metformin plus a long‐acting DPP‐4 inhibitor (Handelsman 2017). Glimepiride was given in doses of 1 mg/day to 6 mg/day, omarigliptin in doses of 25 mg/week and metformin in doses of 1500 mg/day or more. For details of the certainty of the evidence see Appendix 19.

Primary outcomes
All‐cause mortality

In the M+S group no participants died out of 375 participants, compared with 2 deaths in 375 (0.5%) participants in the metformin plus long‐acting DPP‐4 inhibitor group (RR 0.20, 95% CI 0.01 to 4.15; P = 0.30; very low‐certainty evidence because of indirectness and very serious imprecision; Analysis 4.1).

Health‐related quality of life

Handelsman 2017 did not report on this outcome.

Serious adverse effects

In the M+S group 18/375 (4.8%) participants experienced a serious adverse event compared with 24/375 (6.4%) participants in the metformin plus long‐acting DPP‐4 inhibitor group (RR 0.75, 95% CI 0.41 to 1.36; P = 0.34; very low‐certainty evidence because of very serious imprecision, Analysis 4.2).

Secondary outcomes
Cardiovascular mortality

In the M+S group no participants out of 375 died compared with 1 death in 375 (0.3%) participants in the metformin plus long‐acting DPP‐4 inhibitor (RR 0.33, 95% CI 0.01 to 8.16; P = 0.50; very low‐certainty evidence because of indirectness and very serious imprecision; Analysis 4.3).

Non‐fatal myocardial infarction

In the M+S group 1/375 (0.3%) participants experienced a non‐fatal myocardial infarction compared with 0/375 participants in the metformin plus long‐acting DPP‐4 inhibitor (RR 3.00, 95% CI 0.12 to 73.41; P = 0.50; very low‐certainty evidence because of indirectness and very serious imprecision, Analysis 4.4).

Heart failure, non‐fatal stroke, amputation of lower extremity, blindness or severe vision loss, end‐stage renal disease

Handelsman 2017 did not report on these outcomes.

Non‐serious adverse events

In the M+S group 125/375 (33.3%) participants experienced a non‐serious adverse event compared with 43/375 (11.5%) participants in the metformin plus long‐acting DPP‐4 inhibitor (RR 2.91, 95% CI 2.12 to 3.99; P < 0.001; Analysis 4.5), favouring metformin plus long‐acting DPP‐4 inhibitor.

Hypoglycaemia

In the M+S group 110/375 (29.3%) participants experienced a mild or moderate hypoglycaemic episode compared with 21/375 (5.6%) participants in the metformin plus long‐acting DPP‐4 inhibitor (RR 5.24, 95% CI 3.36 to 8.17; P < 0.001; Analysis 4.6 in favour of metformin plus long‐acting DPP‐4 inhibitor).

In the M+S group 6/375 (1.6%) participants experienced a serious hypoglycaemic episode compared with 1/375 (0.3%) participant in the metformin plus long‐acting DPP‐4 inhibitor (RR 6.00, 95% CI 0.73 to 49.60; P = 0.10; Analysis 4.7).

Socioeconomic effects

Handelsman 2017 did not report on this outcome.

Additional explorative outcomes
Weight

Change from baseline in body weight increased by 1.5 kg (SD 4.0) in 375 participants in the M+S group compared with a weight reduction of 0.4 kg in 375 participants in the metformin plus long‐acting DPP‐4 inhibitor group (MD 1.9 kg, 95% CI 1.3 to 2.5; P < 0.001; Analysis 4.8).

HbA1c

In the M+S group there was a MD in HbA1c change of −0.5% in 375 participants compared with −0.3% in 375 participants in the metformin plus long‐acting DPP‐4 inhibitor group (MD of −0.2%, 95% CI −0.3 to −0.1; P = 0.006; Analysis 4.9).

Metformin‐sulfonylurea combination therapy versus metformin plus thiazolidinedione

Eleven trials compared M+S combination therapy with metformin plus a thiazolidinedione (Charbonnel 2005; Derosa 2005; Derosa 2009a; Derosa 2011b; Hamann 2008; Home 2009; Maffioli 2013; NCT00367055; Petrica 2009; Petrica 2011; Vaccaro 2017). Four trials administered glimepiride in doses of 2 mg/day to 4 mg/day (Derosa 2005; Derosa 2009a; Petrica 2009; Petrica 2011), two trials administered glibenclamide in doses of 5 mg/day to 15 mg/day (Derosa 2011b; Maffioli 2013), and two trials administered gliclazide in doses of 80 mg/day to 320 mg/day (Charbonnel 2005; NCT00367055). Three trials treated participants with various sulphonylureas: glibenclamide and gliclazide in doses of 5 mg/day to 15 mg/day and 80 mg/day to 320 mg/day, respectively (Hamann 2008); glibenclamide, gliclazide and glimepiride in doses up to 15 mg/day, 240 mg/day and 4 mg/day, respectively (Home 2009); glibenclamide, gliclazide and glimepiride in doses of 5 mg/day to 15 mg/day, 30 mg/day to 120 mg/day and 2 mg/day to 6 mg/day, respectively (Vaccaro 2017). Five trials administered rosiglitazone in doses of 4 mg/day to 8 mg/day (Derosa 2005; Hamann 2008; Home 2009; NCT00367055; Petrica 2009), and six trials administered pioglitazone in doses of 15 mg/day to 45 mg/day (Charbonnel 2005; Derosa 2009a; Derosa 2011b; Maffioli 2013; Petrica 2011; Vaccaro 2017). Metformin was given in doses of 850 mg/day to 2550 mg/day.

Primary outcomes
All‐cause mortality

Six trials reported that a total of 237 participants died: in the M+S group 123/3300 (3.7%) participants died compared with 114/3354 (3.4%) participants in the metformin plus thiazolidinedione group (RR 1.09, 95% CI 0.85 to 1.40; P = 0.51; 6 trials; 6654 participants; low‐certainty evidence; Analysis 5.1). The 95% prediction interval ranged between 0.75 and 1.55. One of the trials provided data from a time‐to‐event analysis on all‐cause mortality: the HR for metformin plus pioglitazone versus M+S was 1.10 (95% CI 0.75 to 1.61, P = 0.63; Vaccaro 2017).

In one trial, comparing M+S with metformin plus pioglitazone, the investigators reported that no participants died but did not provide the number of participants included in the analysis (Derosa 2011b).

A test for subgroup differences comparing rosiglitazone with pioglitazone did not indicate interaction (P = 0.84; Analysis 5.1). A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.91; Analysis 6.1).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis according to trials with low risk of selection bias did not substantially change the effect estimate (RR 1.08, 95% CI 0.84 to 1.38; P = 0.57; 5 trials; 6570 participants). Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 2.08, 95% CI 0.49 to 8.80; P = 0.32; 4 trials; 1404 participants). We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company due to lack of data.

TSA showed that 7.7% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. The TSA‐adjusted 95% CI was 0.39 to 3.01.

Health‐related quality of life

None of the included trials reported on this outcome.

Serious adverse effects

Six trials reported that a total of 1337 participants experienced a serious adverse event: in the M+S group 666/3300 (20.2%) participants had a serious adverse event compared with 671/3354 (20.0%) participants in the metformin plus thiazolidinedione group (RR 1.01, 95% CI 0.93 to 1.11; P = 0.80; 6 trials; 6654 participants; very low‐certainty evidence; Analysis 5.2). The 95% prediction interval ranged between 0.88 and 1.16.

Derosa 2011b compared M+S with metformin plus pioglitazone and reported that no participants experienced a serious adverse event but did not provide the number of participants included in the analysis.

A test for subgroup differences comparing rosiglitazone with pioglitazone did not indicate interaction (P = 0.84; Analysis 5.2). A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.28; Analysis 6.2).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis according to trials with low risk of selection bias did not substantially change the effect estimate (RR 1.01, 95% CI 0.92 to 1.10; P = 0.90; 5 trials; 6570 participants). Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 1.13, 95% CI 0.69 to 1.86; P = 0.63; 4 trials; 1404 participants). We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company due to lack of data.

TSA showed that 10.6% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 81%. The TSA‐adjusted 95% CI was 0.41 to 2.22.

Secondary outcomes
Cardiovascular mortality

Four trials reported that a total of 78 participants died due to cardiovascular disease: in the M+S group 37/2946 (1.3%) participants died compared with 41/2994 (1.4%) participants in the metformin plus thiazolidinedione group (RR 0.78, 95% CI 0.36 to 1.67; P = 0.52; 4 trials; 5940 participants; low‐certainty evidence; Analysis 5.3). The 95% prediction interval ranged between 0.07 and 8.92. Vaccaro 2017 provided data from a time‐to‐event analysis on cardiovascular mortality: metformin plus pioglitazone versus M+S had a HR of 2.24 (95% CI 0.69 to 7.28, P = 0.18). Derosa 2011b, compared M+S with metformin plus pioglitazone and reported that no participants died due to cardiovascular disease but did not provide the number of participants included in the analysis.

A test for subgroup differences comparing rosiglitazone with pioglitazone did not indicate interaction (P = 0.40; Analysis 5.3). A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.36; Analysis 6.3).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias since all trials were evaluated as low risk of selection bias. We could not perform sensitivity analyses excluding large trials and trials funded by a pharmaceutical company due to lack of data.

TSA showed that 1.3% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 66%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Non‐fatal myocardial infarction

Three trials reported that a total of 46 participants experienced a non‐fatal myocardial infarction: in the M+S group 25/1841 (1.4%) participants had a non‐fatal myocardial infarction compared with 21/1877 (1.1%) participants in the metformin plus thiazolidinedione group (RR 1.21, 95% CI 0.68 to 2.14; P = 0.51; 3 trials; 3718 participants; very low‐certainty evidence; Analysis 5.4). The 95% prediction interval ranged between 0.03 and 48.76.

Vaccaro 2017 provided data from a time‐to‐event analysis on non‐fatal myocardial infarction: M+S versus metformin plus pioglitazone had a HR of 0.87 (95% CI 0.48 to 1.55; P = 0.63).

Derosa 2011b compared M+S with metformin plus pioglitazone and reported that no participants experienced a non‐fatal myocardial infarction but did not provide the number of participants included in the analysis.

A test for subgroup differences comparing rosiglitazone with pioglitazone did not indicate interaction (P = 0.58; Analysis 5.4). A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.58; Analysis 6.4).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. Sensitivity analyses excluding large trials and trials funded by a pharmaceutical company could not be performed due to lack of data.

TSA showed that 1.72% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Heart failure

Five trials reported that a total of 83 participants developed heart failure: in the M+S group 33/3259 (1.0%) participants developed heart failure compared with 50/3311 (1.5%) participants in the metformin plus thiazolidinedione group (RR 0.67, 95% CI 0.43 to 1.04; P = 0.08; 5 trials; 6570 participants; Analysis 5.5). The 95% prediction interval ranged between 0.33 and 1.37.

Vaccaro 2017 provided data from a time‐to‐event analysis on heart failure: the HR comparing participants in the sulphonylureas group with participants in the pioglitazone group was 1.57 (95% CI 0.76 to 3.24; P = 0.22).

Derosa 2011b compared M+S with metformin plus pioglitazone and reported that no participants developed heart failure but did not report the number of participants included in the analysis.

A test for subgroup differences comparing rosiglitazone with pioglitazone did not indicate interaction (P = 0.65; Analysis 5.5). A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.79; Analysis 6.5).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 0.51, 95% CI 0.12 to 2.07; P = 0.35; 3 trials; 1320 participants). We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company due to lack of data.

TSA showed that 3.3% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Non‐fatal stroke

Two trials compared M+S with metformin plus thiazolidinedione: in the M+S group 20/1540 (1.3%) participants had a non‐fatal stroke compared with 16/1583 (1%) participants in the metformin plus thiazolidinedione group (RR 1.29, 95% CI 0.67 to 2.47; P = 0.45; 2 trials; 3123 participants; very low‐certainty evidence; Analysis 5.6). Vaccaro 2017 administered pioglitazone combined with metformin and also provided data from a time‐to‐event analysis on non‐fatal stroke: M+S compared with metformin plus pioglitazone showed a HR of 0.79 (95% CI 0.41 to 1.53; P = 0.49; 3028 participants). Only Vaccaro 2017, in his trial of long duration, observed non‐fatal strokes (Analysis 6.6).

Derosa 2011b administered pioglitazone combined with metformin and reported that no participants experienced a non‐fatal stroke but did not provide the number of participants included in the analysis.

Amputation of lower extremity

Two trials with 3123 participants provided data on amputation of lower extremity (very low‐certainty evidence; Analysis 5.7). Neither of the trials reported any events.

Blindness or severe vision loss

One trial with 95 participants provided data on blindness or severe vision loss (very low‐certainty evidence; Analysis 5.8). They did not report any events.

End‐stage renal disease

One trial with 95 participants provided data on end‐stage renal disease (very low‐certainty evidence; Analysis 5.9). They did not report any events.

Non‐serious adverse events

Five trials reported that a total of 3072 participants experienced a non‐serious adverse event: in the M+S group 1510/2987 (50.6%) participants had a non‐serious adverse event compared with 1562/3037 (51.4%) participants in the metformin plus thiazolidinedione group (RR 0.94, 95% CI 0.44 to 2.01; P = 0.87; 5 trials; 6024 participants; Analysis 5.10). The 95% prediction interval ranged between 0.05 and 16.49.

Derosa 2011b compared M+S with metformin plus pioglitazone and reported that two and three participants experienced non‐serious adverse events, respectively. However, they did not provide the number of participants included in the analysis.

A test for subgroup differences comparing rosiglitazone with pioglitazone did not indicate interaction (P = 0.36; Analysis 5.10). A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.94; Analysis 6.7).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis according to trials with low risk of selection bias did not substantially change the effect estimate (RR 0.92, 95% CI 0.36 to 2.35; P = 0.87; 4 trials; 5940 participants). Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 0.97, 95% CI 0.76 to 1.25; P = 0.83; 3 trials; 774 participants). Sensitivity analysis excluding trials funded by a pharmaceutical company did not substantially change the effect estimate (RR 0.93, 95% CI 0.82 to 1.05; P = 0.24; 2 trials; 3123 participants).

TSA showed that the cumulative z‐curve crossed the futility boundaries suggesting that a 10% or greater RRR could be rejected at this point. Diversity was 0%. The TSA‐adjusted 95% CI was 0.74 to 1.19.

Hypoglycaemia

Five trials reported that a total of 926 participants had a mild or moderate hypoglycaemic episode: in the M+S group 721/2999 (24.0%) participants had a mild or moderate hypoglycaemic episode compared with 205/3060 (6.7%) participants in the metformin plus thiazolidinedione group (RR 3.63, 95% CI 2.98 to 4.44; P < 0.001; 5 trials; 6059 participants; Analysis 5.11 in favour of metformin plus thiazolidinedione). The 95% prediction interval ranged between 2.30 and 5.73. Derosa 2011b compared M+S with metformin plus pioglitazone and reported two and one events of mild/moderate hypoglycaemia, respectively. However, they did not provide the number of participants included in the analysis.

A test for subgroup differences comparing rosiglitazone with pioglitazone did not indicate interaction (P = 0.62; Analysis 5.11). A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.48; Analysis 6.8).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. Sensitivity analysis according to trials with low risk of selection bias did not substantially change the effect estimate (RR 3.63, 95% CI 2.92 to 4.52; P < 0.00001; 4 trials; 5975 participants). Sensitivity analysis excluding large trials did not substantially change the effect estimate (RR 5.99, 95% CI 2.43 to 14.76; P = 0.0001; 3 trials; 809 participants). Sensitivity analysis excluding trials funded by a pharmaceutical company did not substantially change the effect estimate (RR 3.37, 95% CI 2.84 to 3.99; P < 0.00001; 2 trials; 3123 participants).

TSA showed that 8.0% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 45%. The TSA‐adjusted 95% CI was 1.63 to 8.09.

Five trials reported that 36 participants experienced serious hypoglycaemia: in the M+S group 30/3259 (0.9%) participants had a serious hypoglycaemic episode compared with 6/3311 (0.2%) participants in the metformin plus thiazolidinedione group (random RR 3.98, 95% CI 0.34 to 46.01; P = 0.27; fixed RR 4.77, 95% CI 2.05 to 11.09; P < 0.001; 5 trials; 6570 participants; Analysis 5.12; in favour of metformin plus thiazolidinedione). However, there was substantial heterogeneity, probably caused by various definitions of serious hypoglycaemia. Only one trial provided a clear description of serious hypoglycaemia (Vaccaro 2017).

Derosa 2011b compared M+S with metformin plus pioglitazone and reported no serious hypoglycaemic events. However, they did not provide the number of participants included in the analysis.

A test for subgroup differences comparing rosiglitazone with pioglitazone showed a statistically significant difference between subgroups (P = 0.009; Analysis 5.12). However, CIs overlap slightly indicating that in fact there was no true interaction. A test for subgroup differences according to duration of follow‐up did not indicate interaction (P = 0.85; Analysis 6.9).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. We could not perform sensitivity analysis of large trials and trials funded by a pharmaceutical company due to lack of data.

TSA showed that 0.08% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 79%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Socioeconomic effects

None of the included trials reported on this outcome.

Additional explorative outcomes
Weight

Seven trials reported weight change (random MD −0.6 kg, 95% CI −2.8 to 1.6; P = 0.62; fixed MD −2.0 kg, 95% CI −2.4 to −1.6; P < 0.001; 7 trials; 6877 participants; Analysis 5.13; in favour of M+S). The 95% prediction interval ranged between −8.3 kg and 7.2 kg.There was substantial heterogeneity probably caused by two of the rosiglitazone trials (Derosa 2005; Home 2009), and by duration of follow‐up (ranging from one year to 5.5 years), various sulphonylureas (glimepiride, glibenclamide, gliclazide) and various doses of rosiglitazone ranging from 4 mg/day to 8 mg/day.

HbA1c

Ten trials reported change in HbA1c (random MD 0.2%, 95% CI 0.04 to 0.3; P = 0.01; fixed MD 0.2%, 95% CI 0.1 to 0.2; P < 0.001; 10 trials; 7020 participants; Analysis 5.14; in favour of metformin plus thiazolidinedione). The 95% prediction interval ranged between −0.2% and 0.5%. There was substantial heterogeneity probably caused by duration of follow‐up (ranging from 1 year to 5.5 years), various sulphonylureas (glimepiride, glibenclamide, gliclazide) and various doses of sulphonylureas and thiazolidinediones.

Derosa 2009a reported HbA1c final values of 7.8% (SD 0.4) in the M+S group and 7.2% (SD 0.3) in the metformin plus pioglitazone group. However, they did not provide the number of participants included in the analysis.

Metformin‐sulfonylurea combination therapy versus metformin plus glinide

Three trials compared M+S combination therapy with metformin plus a glinide (Derosa 2009b; Gerich 2005; Ristic 2007). Two trials administered glibenclamide in doses of 1.25 mg/day to 15 mg/day and one trial administered gliclazide in doses of 80 mg/day to 240 mg/day. All trials administered nateglinide in doses of 180 mg/day to 540 mg/day. The trials administered metformin in doses of 500 mg/day to 3000 mg/day.

Primary outcomes
All‐cause mortality

Three trials with 874 participants reported data on all‐cause mortality and one person died in each intervention group (low‐certainty evidence; Analysis 7.1).

Health‐related quality of life

None of the included trials reported on this outcome.

Serious adverse effects

Three trials reported that a total of 61 participants experienced a serious adverse event: in the M+S group 34/424 (8%) participants had a serious adverse event compared with 27/450 (6%) participants in the metformin plus thiazolidinedione group (RR 1.68, 95% CI 0.54 to 5.21; P = 0.37; 3 trials; 874 participants; low‐certainty evidence; Analysis 7.2).

In one trial serious adverse events were reported for up to six months and six to 12 months separately. For up to six months 5/126 (4.0%) participants in the M+S group had a serious adverse event compared with 7/130 (5.4%) participants in the metformin plus glinide group. For six to 12 months, 7/101 (6.9%) participants and 2/112 (1.8%) participants experienced a serious adverse event in the M+S group compared with the metformin plus glinide group, respectively (Ristic 2007).

Secondary outcomes
Cardiovascular mortality

Two trials with 446 participants provided data on cardiovascular mortality (Derosa 2009b; Ristic 2007). No cardiovascular death was observed in either trial (low‐certainty evidence; Analysis 7.3).

Non‐fatal myocardial infarction

Two trials provided data on non‐fatal myocardial infarction in 446 participants. In total two non‐fatal myocardial infarctions were reported in 2/215 (0.9%) participants in the M+S group compared with 0/231 participants in the metformin plus thiazolidinedione group (2 trials; 446 participants; low‐certainty evidence). Derosa 2009b stated that no participants experienced a non‐fatal myocardial infarction. One trial reported non‐fatal myocardial infarction for up to six months and six to 12 months separately (Ristic 2007). For up to six months no event occurred in either intervention group. For six to 12 months, 2/101 (2%) participants had non‐fatal myocardial infarction in the M+S group compared to 0/112 participant in the metformin plus glinide group (RR 5.54, 95% CI 0.27 to 114.02; P = 0.27).

Heart failure

Two trials provided data on heart failure. Derosa 2009b stated that no heart failure occurred. One trial reported data on heart failure for up to six months and six to 12 months separately. For up to six months no event occurred in either intervention group. For six to 12 months, 0/101 participants and 1/112 (0.9%) participants developed heart failure in the M+S group compared to the metformin plus glinide group, respectively (Ristic 2007).

Non‐fatal stroke

Derosa 2009b stated that no non‐fatal stroke occurred (233 participants; very low‐certainty evidence).

Amputation of lower extremity

Derosa 2009b stated that no amputation of lower extremity occurred (233 participants; low‐certainty evidence).

Blindness or severe vision loss

Derosa 2009b stated that no blindness or severe vision loss occurred (233 participants; low‐certainty evidence).

End‐stage renal disease

Derosa 2009b stated that no end‐stage renal disease occurred (233 participants; low‐certainty evidence).

Non‐serious adverse events

Derosa 2009b stated that no non‐serious adverse events occurred.

Hypoglycaemia

Two trials provided data on mild or moderate hypoglycaemia. Gerich 2005 reported data from a subgroup analysis of participants 65 years and older: in the M+S group 7/40 (17.5%) participants had mild or moderate hypoglycaemia compared with 1/35 (2.9%) participants in the metformin plus glinide group (Analysis 7.4). Derosa 2009b reported two mild or moderate hypoglycaemic episodes in the M+S group compared with three events in the metformin plus glinide group.

Two trials provided data on serious hypoglycaemia. Gerich 2005 reported that 2/209 (1%) participants in the M+S group compared with 0/219 participants in the metformin plus glinide group experienced a serious hypoglycaemic episode (Analysis 7.5). Derosa 2009b stated that no serious hypoglycaemia occurred.

Socioeconomic effects

Neither of the included trials reported on this outcome.

Additional explorative outcomes
Weight

Two trials reported weight change (MD 1.1 kg, 95% CI −0.1 to 2.3; P = 0.06; 2 trials; 619 participants; Analysis 7.6).

HbA1c

Three trials reported change in HbA1c (random MD 0.2%, 95% CI ‐0.6 to 1.0; P = 0.69; fixed MD 0.4%, 95% CI 0.3 to 0.5; P < 0.00001; 3 trials; 852 participants; Analysis 7.7; in favour of metformin plus glinide). Calculation of the 95% prediction interval did not provide a meaningful estimate. There was substantial heterogeneity probably caused by various durations of follow‐up (ranging from 52 weeks to 104 weeks), various sulphonylureas (glibenclamide and gliclazide) and various doses of nateglinide (ranging from 180 mg/day to 540 mg/day).

Metformin‐sulfonylurea combination therapy versus metformin plus sodium‐glucose co‐transporter 2 (SGLT‐2) inhibitor

Four trials compared M+S combination therapy with metformin plus a SGLT‐2 inhibitor (Del Prato 2015; Hollander 2017; Leiter 2015; Ridderstråle 2014). Three trials administered glimepiride in doses of 1 mg/day to 8 mg/day (Hollander 2017; Leiter 2015; Ridderstråle 2014), and one trial administered glipizide in doses of 5 mg/day to 20 mg/day (Del Prato 2015). All trials administered various SGLT‐2 inhibitors: dapagliflozin in doses of 2.5 mg/day to 10 mg/day (Del Prato 2015), ertugliflozin in doses of 15 mg/day (Hollander 2017), canagliflozin in doses of 100 mg/day to 300 mg/day (Leiter 2015), and empagliflozin in doses of 25 mg/day (Ridderstråle 2014). Metformin was given in doses of 1000 mg/day to 1500 mg/day or more.

Primary outcomes
All‐cause mortality

Four trials reported that a total of 32 participants died: in the M+S group 13/2107 (0.6%) participants died compared with 19/3027 (0.6%) participants in the metformin plus SGLT‐2 inhibitor group (RR 0.96, 95% CI 0.44 to 2.09; P = 0.91; 4 trials; 5134 participants; very low‐certainty evidence; Analysis 8.1). The 95% prediction interval ranged between 0.17 and 5.30.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. Sensitivity analysis excluding long trials did not substantially change the effect estimate (RR 0.72, 95% CI 0.30 to 1.76; P = 0.47; 3 trials; 4320 participants). One trial reported data for 104 weeks (Ridderstråle 2014). We could not perform sensitivity analysis excluding large trials because all trials randomised fewer than 1000 participants to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 1.04% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Health‐related quality of life

None of the included trials reported on this outcome.

Serious adverse effects

Four trials reported that a total of 690 participants experienced a serious adverse event: in the M+S group 315/2107 (15.5%) participants had a serious adverse event compared with 375/3027 (12.4%) participants in the metformin plus SGLT‐2 inhibitor group (RR 1.02, 95% CI 0.76 to 1.37; P = 0.90; 4 trials; 5134 participants; very low‐certainty evidence; Analysis 8.2). The 95% prediction interval ranged between 0.30 and 3.51.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. Sensitivity analysis excluding long trials did not substantially change the effect estimate (RR 0.93, 95% CI 0.35 to 2.49; P = 0.88; 2 trials; 2775 participants). We could not perform sensitivity analysis excluding large trials because all trials randomised fewer than 1000 participants to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 12.4% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 78%. The TSA‐adjusted 95% CI was 0.31 to 3.36.

Secondary outcomes
Cardiovascular mortality

Three trials reported that a total of 10 participants died due to cardiovascular disease: in the M+S group 4/1327 (0.3%) participants died compared with 6/2262 (0.3%) participants in the metformin plus SGLT‐2 inhibitor group (RR 1.22, 95% CI 0.33 to 4.41; P = 0.77; 3 trials; 3589 participants; very low‐certainty evidence; Analysis 8.3). Calculation of the 95% prediction interval did not provide a meaningful estimate.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. Sensitivity analysis excluding long trials did not substantially change the effect estimate (RR 1.02, 95% CI 0.25 to 4.18; P = 0.98; 2 trials; 2775 participants). We could not perform sensitivity analysis excluding large trials because all trials randomised fewer than 1000 participants to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 0.36% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Non‐fatal myocardial infarction

Two trials reported that a total of 15 participants experienced a non‐fatal myocardial infarction: in the M+S group 7/890 (0.8%) participants had a non‐fatal myocardial infarction compared with 8/1374 (0.6%) participants in the metformin plus SGLT‐2 inhibitor group (RR 1.43, 95% CI 0.49 to 4.18; P = 0.52; 2 trials; 2264 participants; very low‐certainty evidence; Analysis 8.4).

We could not perform sensitivity analyses due to lack of data.

TSA showed that 0.46% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Heart failure

Three trials reported that a total of four participants developed heart failure: in the M+S group 4/1670 (0.2%) participants developed heart failure compared with 0/2139 participant in the metformin plus SGLT‐2 inhibitor group (Peto OR 9.21, 95% CI 1.26 to 67.24; P = 0.03; 3 trials; 3809 participants; Analysis 8.5).

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. Sensitivity analysis excluding long trials did not substantially change the effect estimate but widened the CI (Peto OR 11.84, 95% CI 0.68 to 205.34; P = 0.09; 2 trials; 2264 participants). We could not perform sensitivity analysis excluding large trials because all trials randomised fewer than 1000 participants to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

None of the participants in the comparator group (metformin plus SGLT‐2 inhibitor) developed heart failure. TSA could therefore not be performed as it was not possible to calculate an event rate in the comparator group. However, applying 0.5 events in each of the trials in the comparator group showed that 10.9% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. The TSA‐adjusted 95% CI did not provide a meaningful estimate.

Non‐fatal stroke

Two trials reported that a total of 10 participants experienced a non‐fatal stroke: in the M+S group 3/919 (0.3%) participants had a non‐fatal stroke compared with 7/1856 (0.4%) participants in the metformin plus SGLT‐2 inhibitor group (RR 0.87, 95% CI 0.22 to 3.34; P = 0.83; 2 trials; 2775 participants; very low‐certainty evidence; Analysis 8.6).

We could not perform sensitivity analyses due to lack of data.

TSA showed that 0.37% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Amputation of lower extremity

Hollander 2017 reported one amputation of lower extremity in 437 (0.2%) participants in the M+S group compared with one amputation in 888 (0.1%) participants in the metformin plus SGLT‐2 inhibitor group (very low‐certainty evidence, Analysis 8.7).

Blindness or severe vision loss

None of the included trials reported on this outcome.

End‐stage renal disease

None of the included trials reported on this outcome.

Non‐serious adverse events

Three trials reported that a total of 2284 participants had non‐serious adverse events: in the M+S group 1139/1670 (68.2%) participants experienced a non‐serious adverse event compared with 1145/2139 (53.5%) participants in the metformin plus SGLT‐2 inhibitor group (RR 1.27, 95% CI 1.01 to 1.59; P = 0.04; 3 trials; 3809 participants; Analysis 8.8; in favour of metformin plus SGLT‐2 inhibitor). The 95% prediction interval ranged between 0.07 and 23.77. None of the trials provided a detailed definition of this outcome measure.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. Sensitivity analysis excluding long trials did not substantially change the direction of the effect estimate (RR 1.38, 95% CI 1.06 to 1.80; P = 0.02; 2 trials; 2264 participants). We included one trial with a duration of intervention longer than 104 weeks of treatment in the analysis because they reported data for non‐serious adverse events after 52 weeks of treatment (Del Prato 2015). We could not perform sensitivity analysis excluding large trials because all trials randomised fewer than 1000 participants to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 6.2% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 96%. The TSA‐adjusted 95% CI was 0.53 to 3.01.

Hypoglycaemia

Three trials reported that a total of 603 participants experienced mild or moderate hypoglycaemia: in the M+S group 514/1670 (30.8%) participants had a mild or moderate hypoglycaemic episode compared with 89/1639 (5.4%) participants in the metformin plus SGLT‐2 inhibitor group (RR 5.60, 95% CI 2.38 to 13.14; P < 0.001; 3 trials; 3309 participants; Analysis 8.9; in favour of metformin plus SGLT‐2 inhibitor). Calculation of the 95% prediction interval did not provide a meaningful estimate.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. We could not perform sensitivity analysis excluding long trials due to lack of data. We could not perform sensitivity analysis excluding large trials because all trials randomised fewer than 1000 participants to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 0.44% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 93%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Four trials reported that a total of 38 participants had serious hypoglycaemia: in the M+S group 30/2107 (1.4%) participants had a serious hypoglycaemic episode compared with 8/3027 (0.3%) participants in the metformin plus SGLT‐2 inhibitor group (RR 6.16, 95% CI 2.92 to 12.97; P < 0.001; 4 trials; 5134 participants; Analysis 8.10; in favour of metformin plus SGLT‐2 inhibitor). The 95% prediction interval ranged between 1.20 and 31.58. For one of the trials included in the meta‐analysis, the number of serious hypoglycaemic events was unclear due to varied reporting (Leiter 2015). We contacted the trial authors for clarification but did not receive a reply. To be sure to have included all serious hypoglycaemic events, we extracted the highest number of serious hypoglycaemic events reported.

Because all trials were published in English, we could not perform sensitivity analyses according to publication status or language of publication. We could not perform sensitivity analysis according to trials with low risk of selection bias because all trials were evaluated as low risk of selection bias. Sensitivity analysis excluding long trials did not substantially change the effect estimate (RR 6.39, 95% CI 2.89 to 14.12; P < 0.001; 2 trials; 2775 participants). We could not perform sensitivity analysis excluding large trials because all trials randomised fewer than 1000 participants to each intervention group. We could not perform sensitivity analysis excluding trials funded by a pharmaceutical company because all trials received funding from a pharmaceutical company.

TSA showed that 0.56% of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued. Diversity was 0%. As only a minor fraction of the diversity‐adjusted required information size to detect or reject a 10% RRR had been accrued, we could not calculate the TSA‐adjusted 95% CI.

Socioeconomic effects

One trial performed three economic analyses of trial data using the Cardiff Diabetes Model (Del Prato 2015). One economic analysis aimed to assess the cost‐effectiveness of SGLT‐2 inhibitor compared with sulphonylurea when added to metformin for treatment of people in the UK with diabetes mellitus inadequately controlled on metformin alone. There was a mean incremental benefit of 0.47 quality‐adjusted life years (95% CI 0.42 to 0.67), calculated for dapagliflozin plus metformin, the incremental cost‐effectiveness ratio point estimate was GBP 2671 per quality‐adjusted life year.

Another economic analysis aimed to assess the cost‐effectiveness of SGLT‐2 inhibitor compared with sulphonylurea when added to metformin for treatment of Nordic people with diabetes mellitus inadequately controlled on metformin alone. The mean lifetime gain in quality‐adjusted life years for metformin plus dapagliflozin compared to M+S was 0.25 in Denmark, 0.27 in Finland, 0.24 in Norway and 0.28 in Sweden. The cost per quality‐adjusted life year gained was EUR 7944 in Denmark, EUR 5424 in Finland, EUR 4769 in Norway and EUR 6093 in Sweden.

The third economic analysis aimed to assess the cost‐effectiveness of SGLT‐2 inhibitor compared with sulphonylurea when added to metformin for treatment of Spanish people with diabetes mellitus inadequately controlled on metformin alone. Dapagliflozin was a cost‐effective option compared with sulphonylureas and resulted in a cost per quality‐adjusted life year gained of EUR 3560.

Additional explorative outcomes
Weight

Three trials reported weight change (MD 4.4 kg, 95% CI 4.1 to 4.8; P < 0.001; 3 trials; 3294 participants; Analysis 8.11; in favour of metformin plus SGLT‐2 inhibitor). The 95% prediction interval ranged between 2.7 kg and 7.3 kg.

HbA1c

Four trials reported HbA1c (random MD 0.1%, 95% CI −0.1 to 0.2; P = 0.26; fixed MD 0.1%, 95% CI 0.02 to 0.1; P = 0.005; 4 trials; 4182 participants; Analysis 8.12; in favour of metformin plus SGLT‐2 inhibitor). There was substantial heterogeneity, probably caused by various durations of intervention. For one trial (Hollander 2017), we extracted data after 52 weeks of treatment showing benefit of sulphonylurea treatment, whereas for the remaining trials we extracted data after 104 weeks or more showing benefit of SGLT‐2 inhibitor treatment. Furthermore, heterogeneity could have been caused by various sulphonylureas (glipizide and glimepiride) administered in various doses (1 mg/day to 8 mg/day) and various SGLT‐2 inhibitors (dapagliflozin, ertugliflozin, canagliflozin, empagliflozin).

Subgroup analyses

We only performed subgroup analyses on M+S combination therapy versus the combination of metformin plus DPP‐4 inhibitor and metformin plus thiazolidinediones (see above). The remaining combination comparators did not include enough trials to perform subgroup analyses.

We performed subgroup analyses for the comparison of M+S versus metformin plus thiazolidinediones dividing trials into studies investigating rosiglitazone or pioglitazone; see Analysis 5.1 to Analysis 5.14.

We performed subgroup analyses on trials with a long duration (two years and more) versus trials with a short duration (shorter than two years); see Analysis 6.1 to Analysis 6.11.

We did not perform a subgroup analysis on trials including obese participants (BMI ≥ 30) versus trials including non‐obese participants (BMI < 30) due to similar BMIs among the included trials.

Sensitivity analyses

We planned to perform sensitivity analyses for the following factors.

  • Published trials

  • Language of publication

  • Analysis restricted to trials with low risk of selection bias

  • Long trials (trials with duration of intervention longer than 104 weeks excluded)

  • Large trials (trials with more than 1000 participants randomised to each intervention group excluded).

  • Source of funding (trials funded by a pharmaceutical company excluded).

We did not perform sensitivity analysis on diagnostic criteria (often not reported), country (mostly performed in multiple countries) and imputation.

Assessment of reporting bias

We did not draw funnel plots due to limited number of trials for a particular outcome (maximum N = 8).

Ongoing trials

We identified nine ongoing RCTs, which potentially will provide data of interest for this review ( EUCTR2011‐003335‐63‐IT; EUCTR2012‐000152‐34‐IT; JPRN‐UMIN000008815; NCT01243424; NCT01794143; NCT02142309; NCT02730377; NCT02769481; NCT03332771). The ongoing trials will include about 15,147 participants. All ongoing trials assessed one or more outcomes of interest for our review.

Studies awaiting classification

We classified two trials as 'studies awaiting classification'. One trial (Müller‐Wieland 2018), was only published shortly before the publication of this review and one trial (NCT02564926), was submitted in January 2019 but results are not yet publicly available. Both trials compared M+S with metformin plus SGLT‐2 inhibitors. The trials included 1484 participants. Neither trial reports outcomes relevant for summary of findings Table for the main comparison.

Discusión

disponible en

Resumen de los resultados principales

Esta revisión Cochrane investigó los efectos del tratamiento combinado con M+S en comparación con metformina más otra intervención farmacológica hipoglucemiante, placebo o monoterapia con metformina en pacientes con DMT2. Se incluyeron 32 ensayos con un total de 28 746 participantes. Todos los ensayos se consideraron en riesgo poco claro o alto de sesgo en uno o más dominios del “riesgo de sesgo”. La cantidad de evidencia sobre los resultados importantes para los pacientes fue limitada. La administración de M+S no reveló ventajas claras ni desventajas para las medidas de resultado especificadas en la tabla de “resumen de resultados” de esta revisión. Sin embargo, hubo menos episodios hipoglucémicos al comparar M+S con todos los otros tratamientos combinados con metformina más otros fármacos hipoglucemiantes. En dos casos (M+S versus metformina más placebo, M+S versus metformina más inhibidor de DPP‐4), la hipoglucemia se redujo en los grupos comparadores y al mismo tiempo la HbA1c mejoró en el grupo de M+S (Tabla 3). En tres casos (M+S versus metformina más tiazolidinediona, M+S versus metformina más glinida, M+S versus metformina más inhibidor de SGLT‐2), hubo mejores valores de HbA1c y también menos episodios hipoglucémicos en los grupos comparadores (Tabla 3). Debe tenerse en cuenta que el riesgo de hipoglucemia aumenta con las metas de niveles bajos de glucosa lo cual puede no aplicar a la mayoría de los pacientes con diabetes de edad muy avanzada (ADA 2019).

Compleción y aplicabilidad general de las pruebas

Se realizó una búsqueda exhaustiva de ensayos, incluidas publicaciones en todos los idiomas, y se trató de obtener datos adicionales de todos los ensayos. La búsqueda manual de revisiones sistemáticas y listas de referencias identificó dos ensayos adicionales para incluir (Gerich 2005; Ristic 2007). La búsqueda manual en los sitios web de los fabricantes identificó ocho referencias adicionales para incluir en la revisión. Para todos los ensayos, se estableció contacto con uno o más autores de ensayos para obtener información suplementaria sobre los dominios del “riesgo de sesgo” y los resultados. Además, se solicitó a los autores de los ensayos cualquier información adicional acerca del ensayo recuperado y para especificar si existían ensayos adicionales que se pudiesen haber omitido. Seis investigadores de ensayos de diez ensayos acaban de confirmar una pregunta o proporcionaron datos adicionales que podrían implementarse para la evaluación del “riesgo de sesgo” o los metanálisis de los resultados (Dei Cas 2017; Derosa 2005; Derosa 2009b; Derosa 2011a; Derosa 2011b; Gallwitz 2012a; Gallwitz 2012b; Göke 2013; Home 2009; Vaccaro 2017). Para todos los ensayos, se identificó la información de contacto de uno o más autores.

El diagnóstico de la DMT2 fue establecido principalmente mediante las definiciones de la European Association for the Study of Diabetes (EASD), la Organización Mundial de la Salud (OMS) y la American Diabetes Association (ADA). Diecinueve ensayos no especificaron cómo establecieron el diagnóstico de la DMT2 (Ahrén 2014; Charbonnel 2005; Del Prato 2014; Del Prato 2015; Filozof 2010; Gallwitz 2012b; Gerich 2005; Göke 2013; Handelsman 2017; Leiter 2015; Matthews 2010; Nauck 2013; Petrica 2009; Petrica 2011; Ridderstråle 2014; Ristic 2007; Schernthaner 2015; Seck 2010; Vaccaro 2017). Los ensayos incluidos usaron diferentes tipos de sulfonilureas; 16 ensayos administraron una sulfonilurea de segunda generación y 16 ensayos administraron una sulfonilurea de tercera generación.

Puede existir sesgo de selección potencial dentro de los ensayos debido a que se espera que haya personas más saludables y motivadas que participen en un ensayo clínico. Sin embargo, una revisión sistemática Cochrane observó que los resultados clínicos en los pacientes que participaron en ECA son comparables a los resultados en individuos comparables fuera de los ECA (Vist 2008).

Calidad de la evidencia

Ninguno de los 32 ensayos incluidos en la revisión se clasificó como en riesgo bajo de sesgo en todos los dominios del “riesgo de sesgo”. La descripción de la asignación al azar y la asignación en los ensayos incluidos fue insuficiente en ocho ensayos (Ahrén 2014; Del Prato 2014; Filozof 2010; Gerich 2005; Matthews 2010; NCT00367055; Petrica 2009; Petrica 2011). Once ensayos tuvieron un informe insuficiente de uno o más resultados de relevancia para la revisión y, por lo tanto, se los clasificó como en alto riesgo de sesgo para el sesgo de informe de resultado selectivo (Derosa 2005; Derosa 2009a; Derosa 2009b; Derosa 2010; Derosa 2011a; Derosa 2011b; Filozof 2010; Gerich 2005; Maffioli 2013; Petrica 2009; Petrica 2011). Se pudieron evaluar uno o más de los resultados predefinidos en todos los ensayos incluidos.

Para todas las comparaciones, la certeza de la evidencia se juzgó como baja o muy baja principalmente debido a los datos muy limitados, los diversos riesgos de sesgo y la imprecisión.

La mayoría de los ensayos recibieron financiación económica de la industria farmacéutica. Se sabe que los ensayos que reciben financiamiento o provisión de fármacos o dispositivos gratis de una compañía farmacéutica muestran resultados y conclusiones más favorables en comparación con los ensayos patrocinados por otras fuentes (Lundh 2017).

Sesgos potenciales en el proceso de revisión

No fue posible extraer gráficos en embudo para evaluar el sesgo de estudio pequeño debido a la ausencia de datos. Si hubiese habido más datos disponibles y se hubiesen podido realizar más metanálisis, se habría podido investigar la heterogeneidad de forma más detallada.

Se consideró un grupo significativamente heterogéneo de ensayos. Los metanálisis, cuando se realizaron, fueron limitados por la incapacidad para usar los datos individuales de los participantes para evaluar si las características clínicas diferenciadas podrían haber influido en los cálculos del efecto de las intervenciones. Se exploró la heterogeneidad al realizar análisis de sensibilidad y de subgrupos. Sin embargo, debido al número limitado de ensayos sólo se realizaron análisis de subgrupos sobre la M+S versus metformina más inhibidor de la DPP‐4 y de la M+S versus metformina más tiazolidinediona. Muchos de los ensayos incluidos no estuvieron diseñados ni tuvieron el poder estadístico adecuado para detectar los resultados predefinidos importantes para los pacientes.

La mayoría de los ensayos incluidos tuvo un número relativamente pequeño de participantes y los tamaños de información en los metanálisis fueron igual de pequeños. Este hecho aumenta el riesgo de cálculos poco realistas de los efectos de la intervención debido al sesgo (errores sistemáticos) y al azar (errores aleatorios) (Wetterslev 2008; Wood 2008). Se intentó considerar los errores sistemáticos. Se estableció contacto con todos los autores de los ensayos para obtener aclaraciones cuando uno de los dominios del sesgo no se había informado de manera adecuada. Para reducir el riesgo de errores aleatorios, se realizaron ASE de todos los resultados predefinidos, siempre que fue posible.

Varios ensayos se presentaron en más de una publicación, lo cual dificultó la posibilidad de separar la publicación primaria de los artículos complementarios para algunos ensayos (para obtener detalles, ver Estudios incluidos).

Se incluyeron ensayos con una duración mínima de 52 semanas para detectar diferencias clínicamente relevantes para los resultados predefinidos. Lamentablemente, el informe de los resultados relevantes para los pacientes en los ensayos incluidos fue deficiente.

Dos autores de la revisión realizaron la extracción de datos. Sin embargo, los autores de la revisión que extrajeron los datos no fueron cegados en cuanto a los ensayos de los cuales estaban extrayendo datos.

Acuerdos y desacuerdos con otros estudios o revisiones

En la búsqueda de ensayos adicionales se examinaron otras revisiones, revisiones sistemáticas y metanálisis (Amate 2015; Andersen 2016; Aylsworth 2014; Belsey 2008; Chan 2015; Dai 2014; Foroutan 2016; Geng 2015; Goring 2014; Gu 2015; Guthrie 2015; Hershon 2016; Hou 2015; Kuecker 2016; Lim 2015; Liu 2014; Maruthur 2016; Mearns 2015; Mishriky 2015; Monami 2008; Phung 2010; Phung 2014; Rosenstock 2013; Sharma 2017; Varvaki 2016; Wang 2017; Whalen 2015; Zhou 2015; Zintzaras 2014). Debido a que principalmente se intentó investigar los resultados importantes para los pacientes sólo se incluyeron ensayos con una duración mínima de la intervención de 52 semanas. Sin embargo, todas excepto tres revisiones (Goring 2014; Rosenstock 2013; Varvaki 2016), incluyeron ensayos con una duración de la intervención menor que 52 semanas o no describieron la duración de la intervención en los ensayos incluidos. Además, todas excepto siete revisiones (Amate 2015; Foroutan 2016; Hershon 2016; Maruthur 2016; Rosenstock 2013; Varvaki 2016; Wang 2017), se centraron en resultados como el control glucémico, el peso, los lípidos etc. Una revisión (Rosenstock 2013), y una revisión sistemática (Varvaki 2016), sólo incluyeron ensayos con una duración de la intervención de 52 semanas y más, y se centraron en los resultados importantes para los pacientes. Estas revisiones no indicaron un mayor riesgo de eventos cardiovasculares, mortalidad por todas las causas o mortalidad cardiovascular, al comparar M+S con metformina más otro fármaco hipoglucemiante. Varias de las revisiones y revisiones sistemáticas investigaron exclusivamente la M+S versus metformina más un inhibidor de la DPP‐4 (Amate 2015; Foroutan 2016; Gu 2015; Hou 2015; Mishriky 2015; Sharma 2017; Wang 2017; Zhou 2015). Una revisión sistemática y metanálisis compararon la M+S con otros tratamientos combinados con metformina como un grupo (Varvaki 2016). Hasta donde se conoce, la revisión es la primera en investigar los resultados importantes para los pacientes en los ensayos a largo plazo (definido como 52 semanas y más) que consideran la M+S en comparación con metformina más otras intervenciones hipoglucemiantes por separado.

Trial flow diagram
 AHRQ: Agency for Healthcare Research and Quality; DPP4‐I: dipeptidyl‐peptidase 4 inhibitor; GLP1‐A: glucagon‐like peptide 1 analogue; M+S: metformin + sulphonylurea; SGLT2‐I: sodium‐glucose co‐transporter 2 inhibitor
Figuras y tablas -
Figure 1

Trial flow diagram
AHRQ: Agency for Healthcare Research and Quality; DPP4‐I: dipeptidyl‐peptidase 4 inhibitor; GLP1‐A: glucagon‐like peptide 1 analogue; M+S: metformin + sulphonylurea; SGLT2‐I: sodium‐glucose co‐transporter 2 inhibitor

'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).
Figuras y tablas -
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 particular outcome was not measured in some trials)
Figuras y tablas -
Figure 3

'Risk of bias' summary: review authors' judgements about each 'Risk of bias' item for each included trial ((blank cells indicate that the particular outcome was not measured in some trials)

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 1.1

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 1 All‐cause mortality.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 1.2

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 2 Serious adverse events.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 3 Cardiovascular mortality.
Figuras y tablas -
Analysis 1.3

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 3 Cardiovascular mortality.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 4 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 1.4

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 4 Non‐fatal myocardial infarction.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 5 Heart failure.
Figuras y tablas -
Analysis 1.5

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 5 Heart failure.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 6 Non‐serious adverse events.
Figuras y tablas -
Analysis 1.6

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 6 Non‐serious adverse events.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 7 Mild/moderate hypoglycaemia.
Figuras y tablas -
Analysis 1.7

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 7 Mild/moderate hypoglycaemia.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 8 Serious hypoglycaemia.
Figuras y tablas -
Analysis 1.8

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 8 Serious hypoglycaemia.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 9 Weight change.
Figuras y tablas -
Analysis 1.9

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 9 Weight change.

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 10 Change in HbA1c.
Figuras y tablas -
Analysis 1.10

Comparison 1 Metformin plus sulphonylurea vs metformin plus placebo, Outcome 10 Change in HbA1c.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 2.1

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 1 All‐cause mortality.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 2.2

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 2 Serious adverse events.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 3 Cardiovascular mortality.
Figuras y tablas -
Analysis 2.3

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 3 Cardiovascular mortality.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 4 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 2.4

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 4 Non‐fatal myocardial infarction.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 5 Heart failure.
Figuras y tablas -
Analysis 2.5

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 5 Heart failure.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 6 End‐stage renal disease.
Figuras y tablas -
Analysis 2.6

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 6 End‐stage renal disease.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 7 Non‐serious adverse events.
Figuras y tablas -
Analysis 2.7

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 7 Non‐serious adverse events.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 8 Mild/moderate hypoglycaemia.
Figuras y tablas -
Analysis 2.8

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 8 Mild/moderate hypoglycaemia.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 9 Serious hypoglycaemia.
Figuras y tablas -
Analysis 2.9

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 9 Serious hypoglycaemia.

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 10 Weight (change).
Figuras y tablas -
Analysis 2.10

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 10 Weight (change).

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 11 Change in HbA1c.
Figuras y tablas -
Analysis 2.11

Comparison 2 Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue, Outcome 11 Change in HbA1c.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 3.1

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 1 All‐cause mortality.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 3.2

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 2 Serious adverse events.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 3 Cardiovascular mortality.
Figuras y tablas -
Analysis 3.3

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 3 Cardiovascular mortality.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 4 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 3.4

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 4 Non‐fatal myocardial infarction.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 5 Heart failure.
Figuras y tablas -
Analysis 3.5

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 5 Heart failure.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 6 Non‐fatal stroke.
Figuras y tablas -
Analysis 3.6

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 6 Non‐fatal stroke.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 7 Non‐serious adverse events.
Figuras y tablas -
Analysis 3.7

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 7 Non‐serious adverse events.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 8 Mild/moderate hypoglycaemia.
Figuras y tablas -
Analysis 3.8

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 8 Mild/moderate hypoglycaemia.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 9 Serious hypoglycaemia.
Figuras y tablas -
Analysis 3.9

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 9 Serious hypoglycaemia.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 10 Weight change (kg).
Figuras y tablas -
Analysis 3.10

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 10 Weight change (kg).

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 11 Change in HbA1c.
Figuras y tablas -
Analysis 3.11

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 11 Change in HbA1c.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 12 Fasting plasma glucose.
Figuras y tablas -
Analysis 3.12

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 12 Fasting plasma glucose.

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 13 BMI.
Figuras y tablas -
Analysis 3.13

Comparison 3 Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor, Outcome 13 BMI.

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 4.1

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 1 All‐cause mortality.

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 4.2

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 2 Serious adverse events.

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 3 Cardiovascular mortality.
Figuras y tablas -
Analysis 4.3

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 3 Cardiovascular mortality.

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 4 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 4.4

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 4 Non‐fatal myocardial infarction.

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 5 Non‐serious adverse events.
Figuras y tablas -
Analysis 4.5

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 5 Non‐serious adverse events.

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 6 Mild/moderate hypoglycaemia.
Figuras y tablas -
Analysis 4.6

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 6 Mild/moderate hypoglycaemia.

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 7 Serious hypoglycaemia.
Figuras y tablas -
Analysis 4.7

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 7 Serious hypoglycaemia.

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 8 Weight change (kg).
Figuras y tablas -
Analysis 4.8

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 8 Weight change (kg).

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 9 Change in HbA1c (%).
Figuras y tablas -
Analysis 4.9

Comparison 4 Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor, Outcome 9 Change in HbA1c (%).

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 5.1

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 1 All‐cause mortality.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 5.2

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 2 Serious adverse events.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 3 Cardiovascular mortality.
Figuras y tablas -
Analysis 5.3

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 3 Cardiovascular mortality.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 4 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 5.4

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 4 Non‐fatal myocardial infarction.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 5 Heart failure.
Figuras y tablas -
Analysis 5.5

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 5 Heart failure.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 6 Non‐fatal stroke.
Figuras y tablas -
Analysis 5.6

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 6 Non‐fatal stroke.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 7 Amputation of lower extremity.
Figuras y tablas -
Analysis 5.7

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 7 Amputation of lower extremity.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 8 Blindness or severe vision loss.
Figuras y tablas -
Analysis 5.8

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 8 Blindness or severe vision loss.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 9 End‐stage renal disease.
Figuras y tablas -
Analysis 5.9

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 9 End‐stage renal disease.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 10 Non‐serious adverse events.
Figuras y tablas -
Analysis 5.10

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 10 Non‐serious adverse events.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 11 Mild/moderate hypoglycaemia.
Figuras y tablas -
Analysis 5.11

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 11 Mild/moderate hypoglycaemia.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 12 Serious hypoglycaemia.
Figuras y tablas -
Analysis 5.12

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 12 Serious hypoglycaemia.

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 13 Weight (change).
Figuras y tablas -
Analysis 5.13

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 13 Weight (change).

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 14 Change in HbA1c.
Figuras y tablas -
Analysis 5.14

Comparison 5 Metformin plus sulphonylurea vs metformin plus thiazolidinedione, Outcome 14 Change in HbA1c.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 6.1

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 1 All‐cause mortality.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 6.2

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 2 Serious adverse events.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 3 Cardiovascular mortality.
Figuras y tablas -
Analysis 6.3

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 3 Cardiovascular mortality.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 4 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 6.4

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 4 Non‐fatal myocardial infarction.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 5 Heart failure.
Figuras y tablas -
Analysis 6.5

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 5 Heart failure.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 6 Non‐fatal stroke.
Figuras y tablas -
Analysis 6.6

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 6 Non‐fatal stroke.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 7 Non‐serious adverse events.
Figuras y tablas -
Analysis 6.7

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 7 Non‐serious adverse events.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 8 Mild/moderate hypoglycaemia.
Figuras y tablas -
Analysis 6.8

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 8 Mild/moderate hypoglycaemia.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 9 Serious hypoglycaemia.
Figuras y tablas -
Analysis 6.9

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 9 Serious hypoglycaemia.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 10 Weight change.
Figuras y tablas -
Analysis 6.10

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 10 Weight change.

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 11 Change in HbA1c.
Figuras y tablas -
Analysis 6.11

Comparison 6 Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention), Outcome 11 Change in HbA1c.

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 7.1

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 1 All‐cause mortality.

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 7.2

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 2 Serious adverse events.

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 3 Cardiovascular mortality.
Figuras y tablas -
Analysis 7.3

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 3 Cardiovascular mortality.

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 4 Mild/moderate hypoglycaemia.
Figuras y tablas -
Analysis 7.4

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 4 Mild/moderate hypoglycaemia.

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 5 Serious hypoglycaemia.
Figuras y tablas -
Analysis 7.5

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 5 Serious hypoglycaemia.

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 6 Weight change.
Figuras y tablas -
Analysis 7.6

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 6 Weight change.

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 7 Change in HbA1c.
Figuras y tablas -
Analysis 7.7

Comparison 7 Metformin plus sulphonylurea vs metformin plus glinide, Outcome 7 Change in HbA1c.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 8.1

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 1 All‐cause mortality.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 8.2

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 2 Serious adverse events.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 3 Cardiovascular mortality.
Figuras y tablas -
Analysis 8.3

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 3 Cardiovascular mortality.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 4 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 8.4

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 4 Non‐fatal myocardial infarction.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 5 Heart failure.
Figuras y tablas -
Analysis 8.5

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 5 Heart failure.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 6 Non‐fatal stroke.
Figuras y tablas -
Analysis 8.6

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 6 Non‐fatal stroke.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 7 Amputation of lower extremity.
Figuras y tablas -
Analysis 8.7

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 7 Amputation of lower extremity.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 8 Non‐serious adverse events.
Figuras y tablas -
Analysis 8.8

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 8 Non‐serious adverse events.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 9 Mild/moderate hypoglycaemia.
Figuras y tablas -
Analysis 8.9

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 9 Mild/moderate hypoglycaemia.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 10 Serious hypoglycaemia.
Figuras y tablas -
Analysis 8.10

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 10 Serious hypoglycaemia.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 11 Weight change.
Figuras y tablas -
Analysis 8.11

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 11 Weight change.

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 12 Change in HbA1c.
Figuras y tablas -
Analysis 8.12

Comparison 8 Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor, Outcome 12 Change in HbA1c.

Summary of findings for the main comparison. Metformin‐sulphonylurea (second‐ or third‐generation) combination therapy compared with metformin plus another antidiabetic drug for adults with type 2 diabetes mellitus

Metformin‐sulphonylurea (second‐ or third‐generation) combination therapy compared with metformin plus another antidiabetic drug for adults with type 2 diabetes mellitus

Patient: people with type 2 diabetes mellitus

Settings: outpatients

Intervention: metformin + sulphonylurea

Comparison: metformin plus another antidiabetic drug

Outcomes

Metformin + antidiabetic drug

Metformin + sulphonylurea

Relative effect
(95% CI)

No. of participants
(trials)

Certainty of the evidence
(GRADE)

Comments

All‐cause mortality (N)

M + GLP1‐A
Follow‐up: 2‐3 years

7 per 1000

8 per 1000 (4 to 19)

RR 1.15 (0.49 to 2.67)

2594 (3)

⊕⊕⊝⊝a1

Low

M + DPP4‐I
Follow‐up: 1‐3 years

4 per 1000

5 per 1000 (3 to 9)

RR 1.32 (0.76 to 2.28)

11,694 (9)

⊕⊕⊝⊝
Lowb1

M + thiazolidinedione
Follow‐up: 1‐5.5 years

34 per 1000

37 per 1000 (29 to 48)

RR 1.09 (0.85 to 1.40)

6654 (6)

⊕⊕⊝⊝
Lowc1

M + nateglinide

Follow‐up: 1‐2 years

See comment

874 (3)

⊕⊕⊝⊝
Lowd1

1 participant died in each intervention group

M + SGLT2‐I
Follow‐up: 2‐4 years

6 per 1000

6 per 1000 (3 to 13)

RR 0.96 (0.44 to 2.09)

5134 (4)

⊕⊝⊝⊝
Very lowe1

Cardiovascular mortality (N)

M + GLP1‐A
Follow‐up: 2‐3 years

See comment

609 (1)

⊕⊕⊝⊝
Lowa2

1/307 (0.3%) participants died due to cardiovascular disease in the M+S group compared with 1/302 (0.3%) participants in the M + GLP1‐A group

M + DPP4‐I
Follow‐up: 1‐3 years

2 per 1000

4 per 1000 (1 to 9)

RR 1.54 (0.63 to 3.79)

6874 (6)

⊕⊕⊝⊝
Lowb2

M + thiazolidinedione
Follow‐up: 1‐5.5 years

14 per 1000

11 per 1000 (5 to 23)

RR 0.78 (0.36 to 1.67)

5940 (4)

⊕⊕⊝⊝
Lowc2

M + nateglinide

Follow‐up: 1 year

See comment

446 (2)

⊕⊕⊝⊝
Lowd2

No cardiovascular death was reported

M + SGLT2‐I
Follow‐up: 2‐4 years

3 per 1000

3 per 1000 (1 to 12)

RR 1.22 (0.33 to 4.41)

3589 (3)

⊕⊝⊝⊝
very lowe2

Serious adverse events (N)

M + GLP1‐A
Follow‐up: 2‐3 years

126 per 1000

114 per 1000 (92 to 140)

RR 0.90 (0.73 to 1.11)

2594 (3)

⊕⊝⊝⊝
Very lowa3

M + DPP4‐I
Follow‐up: 1‐3 years

124 per 1000

132 per 1000 (120 to 146)

RR 1.07 (0.97 to 1.18)

11,694 (9)

⊕⊝⊝⊝
Very lowb3

M + thiazolidinedione
Follow‐up: 1‐5.5 years

200 per 1000

202 per 1000 (186 to 222)

RR 1.01 (0.93 to 1.11)

6654 (6)

⊕⊝⊝⊝
Very lowc3

M + nateglinide

Follow‐up:

60 per 1000

101 per 1000 (32 to 313)

RR 1.68 (0.54 to 5.21)

874 (3)

⊕⊕⊝⊝
Lowd3

M + SGLT2‐I
Follow‐up: 2‐4 years

124 per 1000

126 per 1000 (94 to 170)

RR 1.02 (0.76 to 1.37)

5134 (4)

⊕⊝⊝⊝
Very lowe3

Non‐fatal stroke (N)

M + GLP1‐A

Not reporteda4

M + DPP4‐I
Follow‐up: 1‐2 years

3 per 1000

6 per 1000 (2 to 18)

RR 2.21 (0.74 to 6.58)

5093 (4)

⊕⊝⊝⊝
Very lowb4

M + thiazolidinedione
Follow‐up: 1‐4.8 years

10 per 1000

13 per 1000 (7 to 25)

RR 1.29 (0.67 to 2.47)

3123 (2)

⊕⊝⊝⊝
Very lowc4

M + nateglinide

Follow‐up: 52 weeks

See comment

233 (1)

⊕⊝⊝⊝
Very lowd4

No non‐fatal stroke was reported

M + SGLT2‐I
Follow‐up: 2 years

4 per 1000

3 per 1000 (1 to 13)

RR 0.87 (0.22 to 3.34)

2775 (2)

⊕⊝⊝⊝
Very lowe4

Non‐fatal myocardial infarction (N)

M + GLP1‐A
Follow‐up: 2‐3 years

6 per 1000

3 per 1000 (1 to 16)

RR 0.57 (0.12 to 2.82)

1575 (2)

⊕⊝⊝⊝
Very lowa5

M + DPP4‐I
Follow‐up: 1‐3 years

3 per 1000

5 per 1000 (2 to 10)

RR 1.45 (0.69 to 3.07)

6874 (6)

⊕⊝⊝⊝
very lowb5

M + thiazolidinedione
Follow‐up: 1‐4.8 years

11 per 1000

14 per 1000 (8 to 24)

RR 1.21 (0.68 to 2.14)

3718 (3)

⊕⊝⊝⊝
Very lowc5

M + nateglinide

Follow‐up: 1 year

See comment

446 (2)

⊕⊕⊝⊝
Lowd5

In 1 trial 2/101 (2%) participants had a non‐fatal myocardial infarction in the M+S group compared with 0/112 participant in the metformin plus nateglinide group

M + SGLT2‐I
Follow‐up: 2‐4 years

6 per 1000

8 per 1000 (3 to 24)

RR 1.43 (0.49 to 4.18)

2264 (2)

⊕⊝⊝⊝
Very lowe5

Microvascular complications (N), definition: end‐stage renal disease, blindness or severe vision loss, amputation of lower extremity

M + GLP1‐A

Not reporteda6

M + DPP4‐I
Follow‐up: 1 year

See comment

64 (1)

⊕⊝⊝⊝
Very lowb6

In 1 trial no participants had a lower‐extremity amputation, developed blindness or severe vision loss, or end‐stage renal disease

M + thiazolidinedione
Follow‐up: 1‐4.8 years

See comment

3123 (2)

⊕⊝⊝⊝
Very lowc6

2 trials (3123 participants) reported that no participants had a lower‐extremity amputation
1 trial (95 participants) reported that no participants developed blindness or severe vision loss, or end‐stage renal disease

M + nateglinide

Follow‐up: 52 weeks

See comment

233 (1)

⊕⊕⊝⊝
Lowd6

No microvascular complications were reported

M + SGLT2‐I
Follow‐up: 2 years

See comment

1325 (1)

⊕⊝⊝⊝
Very lowe6

In 1 trial 1/437 (0.2%) participants had an amputation of the lower extremity in the M+S group compared with 1/888 (0.1%) in the M + SGLT2‐I group

Health‐related quality of life

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; DPP4‐I: dipeptidyl peptidase‐4 inhibitor; GLP1‐A: glucagon‐like peptide 1 analogue; HbA1c: glycosylated haemoglobin A1c; M: metformin; M+S: metformin + sulphonylurea; N: number; N/R: not reported; RR: risk ratio; SGLT2‐I: sodium‐glucose co‐transporter 2 inhibitor; T: thiazolidinedione

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

All‐cause mortality
a1Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 17.
b1Downgraded by one level because of inconsistency (non‐consistent direction of effect) and by one level because of imprecision (CI consistent with both benefit and harm) ‐ see Appendix 18.
c1Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 20.
d1Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 21.
e1Downgraded by one level because of inconsistency (point estimates varied widely) and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Cardiovascular mortality
a2Downgraded by two levels because of serious imprecision (small number of trials, CI consistent with both benefit and harm) ‐ see Appendix 17.
b2Downgraded by one level because of because of inconsistency (non‐consistent direction of effect) and by one level because of imprecision (CI consistent with both benefit and harm) ‐ see Appendix 18.
c2Downgraded by two levels because of serious imprecision (small number of trials, CI consistent with both benefit and harm) ‐ see Appendix 20.
d2Downgraded by two levels because of serious imprecision (small number of trials, unknown event rate) ‐ see Appendix 20.
e2Downgraded by one level because of inconsistency (point estimates varied widely) and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Serious adverse events
a3Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 17.
b3Downgraded by one level because of attrition bias, by one level because of inconsistency (non‐consistent direction of effect) and by one level because of imprecision (CI consistent with both benefit and harm) ‐ see Appendix 18.
c3Downgraded by one level because of attrition bias, by one level because of inconsistency (non‐consistent direction of effect) and by one level because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 20.
d3Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 21.
e3Downgraded by one level because of attrition bias and inconsistency (point estimates varied widely) and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 22.

Non‐fatal stroke
a4No adequate data for analysis.
b4Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 18.
c4Downgraded by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 20.
d4Downgraded by two levels of evidence because of serious imprecision (low number of trials, unknown event rate) ‐ see Appendix 21.
e4Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Non‐fatal myocardial infarction
a5Downgraded by one level because of attrition bias and by two levels because of serious imprecision (small number of trials, CI consistent with both benefit and harm) ‐ see Appendix 17.
b5Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 18.
c5Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials) ‐ see Appendix 20.
d5Downgraded by two levels because of serious imprecision (low number of trials, low event rate) ‐ see Appendix 21.
e15Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Microvascular complications
a6No adequate data for analysis.
b6Downgraded by three levels because of very serious imprecision (small number of participants, one trial only, unknown event rate) ‐ see Appendix 18.
c6Downgraded by one level because of attrition bias and reporting bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, unknown event rate) ‐ see Appendix 20.
d6Downgraded by two levels because of serious imprecision (low number of trials, unknown event rate) ‐ see Appendix 21.
e6Downgraded by one level because of attrition bias and by two levels because of serious imprecision (CI consistent with both benefit and harm, small number of trials, low event rate) ‐ see Appendix 22.

Figuras y tablas -
Summary of findings for the main comparison. Metformin‐sulphonylurea (second‐ or third‐generation) combination therapy compared with metformin plus another antidiabetic drug for adults with type 2 diabetes mellitus
Table 1. Overview of trial populations

Trial ID

(design)

Intervention(s) and comparator(s)

Description of power and sample size calculation

Screened/eligible
(N)

Randomised
(N)

Analysed
(N)

Finishing trial
(N)

Randomised finishing trial
(%)

Follow‐up
(extended follow‐up)a

Handelsman 2017

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg/day + glimepiride 1‐6 mg/day + placebo

Quote from publication: "A sample size of ˜340 patients randomized to each treatment group was calculated to have 91% power to declare non‐inferiority for a margin of δ=0.35% at an overall two sided 5% alpha‐level, assuming that the true mean difference in HbA1c between omarigliptin and glimepiride is 0.0%"

1197

375

375

284

75.7

54 weeks

C: metformin ≥ 1500 mg/day + omarigliptin 25 mg/week + placebo

376

375

290

77.1

Total:

751

750

574

76.4

Hollander 2017

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg/day + glimepiride 1‐8 mg/day + placebo

Quote from publication: "With a non‐inferiority margin of 3.3 mmol/mol (0.3%), and assuming a true mean difference in HbA1c of 0 mmol/mol, randomisation of approximately 1230 patients (410 patients per group, to yield a sample size of 337 per group at week 52) was estimated to provide 97% power to demonstrate non‐inferiority of a given ertugliflozin dose to glimepiride in HbA1c reduction at week 52"

2985

437

352

348

79.6

52 weeks (104 weeks)b

C1: metformin ≥ 1500 mg/day + ertugliflozin 5 mg/day + placebo

448

335

340

75.9

C2: metformin ≥ 1500 mg/day + ertugliflozin 15 mg/day + placebo

441

350

357

81.0

Total:

1326

1037

1045

78.8

Vaccaro 2017

(parallel RCT)c

I: metformin 2000 mg/day + sulphonylurea (glibenclamide 5‐15 mg/day, gliclazide 30‐120 mg/day or glimepiride 2‐6 mg/day)

Quote from publication: "The study was designed to be event driven. The initial sample size calculation was based on an estimated primary endpoint rate of 3.5% per year, with the study intended to have 80% power to detect a reduction of 20% in the primary outcome in either group versus the other, based on the results of the PROACTIVE trial. On the basis of these assumptions, 652 events were needed for the primary efficacy analysis. Therefore, 4396 patients had to be enrolled and followed up for at least 4 years; assuming a trial discontinuation rate of 15%, 5172 patients needed to be recruited and randomly assigned (2586 in each treatment group). However, because of the lower than expected rate of recruitment and because the number of participants discontinuing the study was lower than initially foreseen, an approved protocol amendment (January, 2012) subsequently reduced the sample size requirement. Accordingly, 3371 patients should have been enrolled to expect the 498 endpoint events needed to detect a 20% reduction in the incidence of events with a statistical power of 80% (hazard ratio [HR] 0.80, p=0.05 [one‐sided log‐rank test]), assuming an estimated occurrence rate of the primary endpoint of 3.5% per year and a 5% loss to follow‐up. Nonetheless, nearly 9 years after the beginning of the study, the number of events needed was still not reached, and a futility analysis was done as recommended by the data and safety monitoring board"

4956

1500

1493

1255

83.6

Median follow‐up 57.3 months

C: metformin 2000 mg/day + pioglitazone 15‐45 mg/day

1541

1535

1103

71.6

Total:

3041

3028

2358

77.5

Dei Cas 2017

(parallel RCT)

I: metformin ≥ 1500 mg/day + glibenclamide 10 mg/day

Quote from publication: "Sample size was calculated to achieve 80% power to reject the null hypothesis of equal mean changes in the primary endpoint when the population mean difference is 0.15 with a standard deviation of 0.2 in both groups and with a significance level (∝) of 0.05 using a two‐sided two sample equal‐variance T test (difference between the two treatments at 12 months) (software PASS‐ NCSS, USA). In addition, 40 subjects were sufficient to guarantee a delta value between 0 and 12 months in the treatment group of ˜10% (SD of pair differences 20%) with a alpha value of 5% and β = 80%"

73

24

24

19

79.2

12 months

C: metformin ≥ 1500 mg/day + vildagliptin 100 mg/day

40

40

40

100

Total:

64

64

59

92.2

Leiter 2015

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg/day + glimepiride 1‐8 mg/day

Quote from publication: "Sample size was calculated on the basis of the per‐protocol analysis; an estimated 277 patients per group would be needed to provide approximately 90% power to show non‐inferiority of canagliflozin to glimepiride for HbA1c lowering, with an assumed difference of 0.0% between canagliflozin and glimepiride and an assumed common SD of 1.0%. We assumed that 35% of patients would discontinue the study before week 52; therefore, about 427 patients were planned for inclusion in each group. For the body composition substudy, 46 or more patients per group would provide 90% power for the comparisons between groups in percentage of total fat and visceral adipose tissue; to assure collection of imaging at both baseline and week 52, approximately 70 patients per group were planned for inclusion"

3316

484

482

314

64.9

52 weeks (+ 52 weeks)

C1: metformin ≥ 1500 mg/day + canagliflozin 100 mg/day

483

483

343

71.0

C2: metformin ≥ 1500 mg/day + canagliflozin 300 mg/day

485

485

323

66.6

Total:

1452

Del Prato 2015

(non‐inferiority parallel RCT)

I: metformin 1500‐2500 mg/day + glipizide 5‐20 mg/day

Quote from publication: "To demonstrate non‐inferiority of dapagliflozin in comparison with glipizide as add‐on therapy to metformin for changes from baseline to week 52 in HbA1c with a non‐inferiority margin of 0.35%, assuming a standard deviation (SD) of 1.25%, and at a one‐sided significance level of 0.025, 280 evaluable patients were needed in each treatment group to provide approximately 90% power (given a true difference of zero between the 2 treatment groups). Assuming a 5% exclusion rate from the full analysis set, 295 patients per treatment group are needed for the full analysis set. Additionally, to have 90% power for the per‐protocol population and assuming a 25% exclusion rate from the per‐protocol population, 373 patients per treatment group (746 patients in total) were planned for randomization"

1217

408

401

141

34.6

52 weeks (+156 weeks)

C: metformin 1500‐2500 mg/day + dapagliflozin 2.5‐10 mg/day

406

400

161

39.7

Total:

814d

801

302

37.0

Schernthaner 2015

(parallel RCT)

I: metformin at any dose + glimepiride 1‐6 mg/day + placebo

Quote from publication: "A sample size of 698 patients (349/treatment arm) was calculated for detecting superiority of saxagliptin in the primary endpoint, with a two‐sided significance level of 0.05 and 80% power. This assumed a 10% dropout rate and an odds ratio (OR) of 1.55 for achieving target HbA1c without hypoglycaemia with saxagliptin compared with glimepiride"

957

360

359

285

79.2

52 weeks

C: metformin at any dose + saxagliptin 5 mg/day + placebo

360

359

289

80.3

Total:

720

718

574

79.7

Del Prato 2014e

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg once daily or maximum tolerated dose + glipizide 5‐20 mg once daily

Quote from publication: "The planned randomization sample size for the study was between 815 and 897 patients per treatment arm. This ensured at least 95% power to declare non‐inferiority between either alogliptin dose (12.5 or 25 mg) and glipizide at week 104, assuming a non‐inferiority margin of 0.3%, no difference between either alogliptin dose and glipizide, a standard deviation of change from baseline of 1.2%, an evaluability rate of 60%, and a one‐sided 0.0125 significance level. The 0.0125 significance level was chosen so that, combined with similar analyses conducted at week 52, the overall one‐sided type 1 error rate for the trial was maintained at the 0.025 level"

5789

874

336

427

48.9

104 weeks (+ 2 weeks)

C1: metformin ≥ 1500 mg once daily or maximum tolerated dose + alogliptin 12.5 mg once daily

880

371

472

53.6

C2: metformin ≥ 1500 mg once daily or maximum tolerated dose + alogliptin 25 mg once daily

885

382

493

55.7

Total:

2639

1089

1392

52.7

Ahrén 2014

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg daily + glimepiride 2‐4 mg once daily + placebo

Quote from publication: "The planned sample size provided >90% power to demonstrate superiority versus placebo and noninferiority versus sitagliptin and glimepiride (noninferiority margin = 0.3%). Superiority of albiglutide versus sitagliptin and glimepiride was tested if noninferiority was established"

1525

317

102

191

60.3

104 weeks (+ 52 weeks)

C1: metformin ≥ 1500 mg daily + albiglutide 30‐50 mg once weekly + placebo

315

115

192

61.0

C2: metformin ≥ 1500 mg daily + sitagliptin 100 mg once daily + placebo

313

88

190

60.7

C3: metformin ≥ 1500 mg daily + placebo

104

16

55

52.9

Total:

1049

321

628

59.9

Ridderstråle 2014

(non‐inferiority parallel RCT)

I: metformin immediate release ≥ 1500 mg/day plus glimepiride 1‐4 mg/day

Quote from publication: "698 patients per group were needed to provide a power of at least 95% to show non‐inferiority, based on a margin of 0.3%, for the primary endpoint at weeks 52 and 104 if the true treatment effect is 0.05% (in favour of glimepiride) and SD is 1.2%"

2637

780

780

648 ( 2 years)

589 (4 years)

83.1 (2 years)

75.5 (4 years)

208 weeks

C: metformin immediate release ≥ 1500 mg/day plus empagliflozin 25 mg/day

769

765

652 (2 years)

610 (4 years)

84.8 (2 years)

79.3 (4 years)

Total:

1549

1545

1300 (2 years)

1199 (4 years)

83.9 (2 years)

77.4 (4 years)

Göke 2013

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg daily + glipizide 5‐20 mg/day

Quote from publication: "With 419 patients per treatment group, there was a 95% power to establish the non‐inferiority comparison on change from baseline to week 52 HbA1c at the 5% level, assuming that the standard deviation of change from baseline HbA1c was 1.1%, with a non‐inferiority limit set at 0.35% and a zero true difference between the two randomised treatments. The sample size assumed that 35% of randomised patients would be excluded from the PP analysis set"

1377

430

426

147

34.2

52 weeks (+ 52 weeks)

C: metformin ≥ 1500 mg daily + saxagliptin 5 mg/day

428

426

165

38.6

Total:

858

852

312

36.4

Maffioli 2013

(parallel RCT)

I: metformin 2550 mg/day plus glibenclamide 10 mg/day

Quote from publication: "Considering a difference of at least 10% as clinically significant compared with the baseline and an α error of 0.05, the actual sample size was adequate to obtain a power higher than 0.80 to detect a significant between‐group difference in variables related to ultrasonography parameters"

84

80

80

95.2

12 months

C: metformin 2550 mg/day plus pioglitazone 30 mg/day

86

80

80

93.0

Total:

170

160

160

94.1

Nauck 2013

(parallel RCT)

I: metformin 1500‐2000 mg/day + glimepiride 1‐4 mg/day + placebo

Quote from publication: "Sample size calculations were based on showing A1C and body weight differences of 0.5 and 3%, respectively, after 6 months of treatment. The assumed standard deviation for A1C and the coefficient of variance for weight were 1.2 and 3%, respectively. The combined power (calculated as the product of the marginal powers for A1C and weight) was at least 85%"

1662

244

234

113

46.3

26 weeks (+ 18 months)

C1: metformin 1500‐2000 mg/day + liraglutide 0.6 mg/day + placebo

242

239

130

53.7

C2: metformin 1500‐2000 mg/day + liraglutide 1.2 mg/day + placebo

241

231

137

56.8

C3: metformin 1500‐2000 mg/day + liraglutide 1.8 mg/day + placebo

242

235

118

48.8

C4: metformin 1500‐2000 mg/day + placebo

122

120

31

25.4

Total:

1091

1059

529

48.5

Gallwitz 2012a

(non‐inferiority parallel RCT)

I: metformin median dose 2000 mg/day + glimepiride mean dose 2.01 mg/day + placebo

Quote from publication: "We calculated sample size on the basis of the non‐inferiority test of exenatide versus glimepiride, an expected mean baseline HbA1c concentration of 8.2%, a 1 year patient accrual, maximum follow‐up of 3 years, dropout rate of 15% per year (for reasons other than treatment failure), and a 58% event rate in each group after 1 year. With these assumptions, 527 patients per study group would provide about a 90% power to conclude non‐inferiority of exenatide"

1404

514

485

386

75.1

Max. follow‐up 3 years

C: metformin median dose 2000 mg/day + exenatide mean dose 17.35 μg/day + placebo

515

488

341

66.2

Total:

1029

973

727

71.1

Gallwitz 2012b

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg/day + glimepiride 1‐4 mg/day

Quote from publication: "On the assumption of an SD of change in HbA1c from baseline of 1.3%, a sample size of 707 participants per treatment group was needed for 90% power to show non‐inferiority through a 97.5% CI for treatment difference in the adjusted mean change from baseline to endpoint of < 0.35% HbA1c at the level of α=0.0125 (one‐sided)"

2283

775

755

604

78

104 weeks (+ 1 week)

C: metformin ≥ 1500 mg/day + linagliptin 5 mg/day

777

764

587

76

Total:

1552

1519

1191

77

Derosa 2011a

(parallel RCT)

I: metformin 1000‐2000 mg/day + glimepiride 6 mg/day

Quote from publication: "Considering as clinically significant a difference of at least the 10% compared to the baseline and an alpha error of 0.05, the actual sample size was adequate to obtain a power higher than 0.80 for all measured variable"

54

49

49

90.7

12 months

C: metformin 1000‐2000 mg/day + exenatide 20 μg/day

57

52

52

91.2

Total:

111

101

101

91.0

Derosa 2011b

(parallel RCT)

I: metformin 1700 ± 850 mg/day + glibenclamide 5‐15 mg/day

Quote from publication: "Considering as clinically significant a difference of at least the 10% compared to the baseline and an alpha error of 0.05, the actual sample size was adequate to obtain a power higher than 0.80 for all measured variables"

99

95

95

96.0

12 months

C: metformin 1700 ± 850 mg/day + pioglitazone 15‐45 mg/day

102

99

99

97.1

Total:

201

194

194

96.5

Petrica 2011

(parallel RCT)

I: metformin 1700 mg/day plus glimepiride 4 mg/day

124

39

34

34

87.2

1 year

C: metformin 1700 mg/day plus pioglitazone 30 mg/day

39

34

34

87.2

Total:

78

68

68

87.2

Derosa 2010

(parallel RCT)

I: metformin 1500 ± 500 mg/day + glibenclamide 15 mg/day

65

57

57

87.7

12 months

C: metformin 1500 ± 500 mg/day + exenatide 20 μg/day

63

59

59

93.7

Total:

128

116

116

90.6

Matthews 2010

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg twice a day + glimepiride 2‐6 mg/day

Quote from publication: "With 3120 patients randomized (i.e. 1560 patients per treatment arm) and an overall 20% discontinuation rate, the study had 96% power to show non‐inferiority of vildagliptin compared with glimepiride (one‐sided α level of 0.0125, assuming a non‐inferiority margin of 0.3% HbA1c and a standard deviation of 1.25%)"

approx. 6000

1556

1518

953

61.2

2 years

C: metformin ≥ 1500 mg twice a day + vildagliptin 50 mg twice a day

1562

1476

994

63.6

Total:

3118

2994

1947

62.4

Filozof 2010

(non‐inferiority parallel RCT)

I: metformin 1500 mg/day plus gliclazide 80‐320 mg/day

Quote from publication: "Eight hundred patients (400 per group) were required to demonstrate non‐inferiority of vildagliptin to gliclazide in HbA1c reduction, with a one‐sided α level of 0.025 at the end of the study with 92% power (assuming a true difference of 0.1% in favour of gliclazide, standard deviation of HbA1c reduction at week 52 of 1.25 units and discontinuation rate of 20% over the 52‐week period)"

494

393

412

83.4

52 weeks

C: metformin 1500 mg/day plus vildagliptin 100 mg/day

513

386

407

79.3

Total:

1007

779

819

81.3

Seck 2010

(non‐inferiority parallel RCT)

I: metformin ≥ 1500 mg/day plus glipizide 5‐20 mg/day

2141

584

559

264

45.2

2 years

C: metformin ≥ 1500 mg/day plus sitagliptin 100 mg/day

588

576

255

43.4

Total:

1172

1135

519

44.3

Home 2009

(non‐inferiority parallel RCT)

I: metformin up to 2550 mg/day + glibenclamide (or equivalent for different preparations) up to 15 mg/day or gliclazide up to 240 mg/day or glimepiride up to 4 mg/day

Quote from publication: "For the non‐inferiority hypothesis, 4000 participants followed for a median time of 6 years were needed to give 99% power, provided that the active control group had an 11% event rate per year, allowing 2% annual loss to follow‐up. Blinded overall event tracking showed the event rate during the study was well below this rate. Therefore, endpoint sweeps were implemented to identify any missed events. An in‐depth review of a sample of individual records showed very few missed events"

7428

1108

1105

906

82.0

Mean follow‐up: 5.5 years

C: metformin up to 2550 mg/day + rosiglitazone up to 8 mg/day

1120

1117

939

84.1

Total:

2228

2222

1845

83.0

Derosa 2009a

(parallel RCT)

I: metformin 850 mg/day + glimepiride 2‐6 mg/day

Quote from publication: "Considering as clinically significant a difference of at least 10% compared with the baseline and an α error of 0.05, the actual sample size is adequate to obtain a power higher than 0.80 for all variables related to glucose metabolism..."

66

60

90.9

15 months

C1: metformin 850‐2550 mg/day + pioglitazone 15‐45 mg/day

69

60

87.0

C2: metformin 1000‐3000 mg/day

67

60

90.0

Total:

202

180

89.1

Derosa 2009b

(parallel RCT)

I: metformin 1500‐3000 mg/day + glibenclamide 7.5‐15 mg/day

124

114

114

91.9

1 year

C: metformin 1500‐3000 mg/day + nateglinide 180‐360 mg/day

124

119

119

96.0

Total:

248

233

233

94.0

Petrica 2009

(parallel RCT)

I: metformin 1700 mg/day + glimepiride 4 mg/day

65

22

17

17

77.3

1 year

C: metformin 1700 mg/day plus rosiglitazone 4 mg/day

22

17

17

77.3

Total:

44

34

34

77.3

NCT00367055

(parallel RCT)

I: metformin 2000 mg/day + gliclazide 80‐320 mg/day

44

41

30

68.2

36 months

C: metformin 2000 mg/day + rosiglitazone 4‐8 mg/day

45

43

32

71.1

Total:

89

62

62

69.7

Hamann 2008

(non‐inferiority parallel RCT)

I: metformin 2000 mg/day + glibenclamide 5‐15 mg/day or gliclazide 80‐320 mg/day

Quote from publication: "The non‐inferiority margin was set at 0.4%. A sample size of 190 per treatment group was required to give a 90% probability that the upper limit of a two‐sided 95% CI for the difference in treatment means would be below 0.4% (significance level of 0.025 in a one‐sided test), assuming an SD of 1.2%. Assuming an attrition rate of 30%, 544 subjects were to be recruited"

818

302

288

230

76.2

52 weeks

C: metformin 2000 mg/day + rosiglitazone 4‐8 mg/day

294

285

233

79.3

Total:

596

573

463

77.7

Ristic 2007

(parallel RCT)

I: metformin > 1000 mg/day + gliclazide 80‐240 mg/day

Quote from publication: "A planned sample size of 120 patients per treatment was considered sufficient to detect an HbA1c difference of 0.5% with 90% power, assuming a dropout rate of 15% and an SD of 1.1 (calculated for 24 weeks treatment)"

129

101

98

76.0

24 weeks (+6 months)

C: metformin > 1000 mg/day + nateglinide 180‐540 mg/day

133

112

108

81.2

Total:

262

213

206

78.6

Charbonnel 2005

(parallel RCT)

I: metformin at pre‐study dose + gliclazide 80‐320 mg/day

Quote from publication: "Sample size was based on demonstrating a between‐group difference of 0.35% in the change in HbA1c from baseline to week 52 (the primary efficacy variable) using a two‐sided t‐test. A total of 225 patients/group completing at least 24 weeks of the study was required, on the basis of a level of 95% power at 5% significance"

1071

313

238

76.0

104 weeks

C: metformin at pre‐study dose + pioglitazone 15‐45 mg/day

317

233

73.5

Total:

630

471

74.8

Derosa 2005

(parallel RCT)

I: metformin 1500 mg/day plus glimepiride 2 mg/day

Quote from publication: "The study power was a priori calculated by using the World Wide Web‐available power calculator of the university of California, Los Angeles, Department of statistics (Los Angeles, CA)"

49

47

47

95.9

12 months

C: metformin 1500 mg/day plus rosiglitazone 4 mg/day

50

48

48

96.0

Total:

99

95

95

96.0

Gerich 2005

(parallel RCT)

I: metformin 500‐2000 mg/day + glyburide 1.25‐15 mg/day + placebo

908

209

198

122

58.4

104 weeks

C: metformin 500‐2000 mg/day + nateglinide 180‐540 mg/day + placebo

219

208

141

64.4

Total:

428

406

263

61.4

Grand total

All interventions

12,863

All comparators

15,883

All interventions and comparators

28,746

‐ denotes not reported

aFollow‐up under randomised conditions until end of trial (= duration of intervention + follow‐up post‐intervention or identical to duration of intervention); extended follow‐up refers to follow‐up of participants once the original trial was terminated as specified in the power calculation.
bThe trial was conducted over 104 weeks in two 52‐week phases; the primary and secondary hypotheses were pre‐specified for testing at week 52 (phase A); treatment was continued for another 52 weeks (phase B) to evaluate longer‐term safety and efficacy.
cThe median duration of therapy was 12 (IQR 11–13) months, 12 (11–12) months for vildagliptin and 12 (7–13) months for glibenclamide.
dTwo participants did not take the drug.
eThe primary efficacy endpoint was the change in HbA1c from baseline to week 52 and to week 104.

C: comparator; CI: confidence interval; FPG: fasting plasma glucose; HbA1c: glycosylated haemoglobin A1c; I: intervention; IQR: interquartile range; PP: per protocol; RCT: randomised controlled trial; SD: standard deviation

Figuras y tablas -
Table 1. Overview of trial populations
Table 2. Overview of trials (trial arms), comparators, intervention and number of randomised participants

Drug class

Trials (trial arms) (N)a

Metformin + comparator: randomised participants (N)

Metformin + sulphonylurea: randomised participants (N)b

Thiazolidinediones

1

1

1

1

1

1

1

1

1

1

1

Total:11 (11)

Pioglitazone 15‐45 mg: 1541

Pioglitazone 15‐45 mg: 102

Pioglitazone 15‐45 mg: 69

Pioglitazone: 15‐45 mg: 317

Pioglitazone 30 mg: 86

Pioglitazone 30 mg: 39

Rosiglitazone 4 mg: 22

Rosiglitazone 4 mg: 50

Rosiglitazone 4‐8 mg: 45

Rosiglitazone 4‐8 mg: 294

Rosiglitazone up to 8 mg: 1120

Total:3685

Glimepiride/glibenclamide/gliclazide: 1500

Glibenclamide: 99

Glimepiride: 66

Gliclazide: 313

Glibenclamide: 84

Glimepiride: 39

Glimepiride: 22

Glimepiride: 49

Gliclazide: 44

Glibenclamide/gliclazide: 302

Glibenclamide/gliclazide/glimepiride: 1108

Total:3626

DPP‐4 inhibitors

1

1

1

1

1

1

1

1

1

1

1

Total: 10 (11)

Alogliptin 12.5 mg: 880

Alogliptin 25 mg: 885

Linagliptin 5 mg: 777

Omarigliptin 25 mg: 376

Saxagliptin 5 mg: 360

Saxagliptin 5 mg: 428

Sitagliptin 100 mg: 313

Sitagliptin 100 mg: 588

Vildagliptin 50 mg: 1562

Vildagliptin 100 mg: 40

Vildagliptin 100 mg: 513

Total: 6722

Glipizide: 874
(Glipizide: 874)

Glimepiride: 775

Glimepiride: 375

Glimepiride: 360

Glipizide: 430

Glimepiride: 317

Glipizide: 584

Glimepiride: 1556

Glibenclamide: 24

Gliclazide: 494

Total: 5789

GLP‐1 agonists

1

1

1

1

1

1

1

Total: 5 (7)

Albiglutide 30‐50 mg: 315

Exenatide 17.35 µg: 515

Exenatide 20 µg: 57

Exenatide 20 µg: 63

Liraglutide 0.6 mg: 242

Liraglutide 1.2 mg: 241

Liraglutide 1.8 mg: 242

Total: 1675

Glimepiride: 317

Glimepiride: 514

Glimepiride: 54

Glibenclamide: 65

Glimepiride: 244

(Glimepiride: 244)

(Glimepiride: 244)

Total: 1194

SGLT‐2 inhibitors

1

1

1

1

1

1

Total: 4 (6)

Canagliflozin 100 mg: 483

Canagliflozin 300 mg: 485

Dapagliflozin 2.5‐10 mg: 406

Empagliflozin 25 mg: 769

Ertugliflozin 5 mg: 448

Ertugliflozin 15 mg: 441

Total: 3032

Glimepiride: 484

(Glimepiride: 484)

Glipizide: 408

Glimepiride: 780

Glimepiride: 437

(Glimepiride: 437)

Total: 2109

Glinides

1

1

1

Total: 3 (3)

Nateglinide 180‐360 mg: 124

Nateglinide 180‐540 mg: 133

Nateglinide 180‐540 mg: 219

Total: 476

Glibenclamide: 124

Gliclazide: 129

Glyburide: 209

Total: 462

Metformin monotherapy

1

1

1

Total:3 (3)

Metformin ≥1500 mg: 104

Metformin 1500‐2000 mg: 122

Metformin 1000‐3000 mg: 67

Total: 293

(Glimepiride: 317)

(Glimepiride: 244)

(Glimepiride: 66)

(Total: 627)

aTotal number of unique included trials was 32 with 41 trial arms.
bNumbers of randomised participants for metformin combination therapy do not add up correctly because several trials had one intervention with several comparator groups, the intervention group may therefore appear in several drug classes and is characterised by parentheses; combination therapy data for metformin monotherapy are shown for illustrative purposes.

DPP4‐I: dipeptidyl‐peptidase 4; GLP‐1: glucagon‐like peptide‐1 agonist; SGLT‐2: sodium‐glucose transport 2

Figuras y tablas -
Table 2. Overview of trials (trial arms), comparators, intervention and number of randomised participants
Table 3. Overview of HbA1c and hypoglycaemia across comparisons

Comparison

Better HbA1c for M+S

Better HbA1c for comparator

Less hypoglycaemia for M+S

Less hypoglycaemia for comparator

M+S vs metformin + placebo

Yes

No

No

Yes: mild or moderate episodes

M+S vs metformin + GLP‐1 agonist

No

No

No

Yes: mild or moderate episodes

M+S vs metformin + DPP‐4 inhibitor

Yes

No

No

Yes: mild or moderate and serious episodes

M+S vs metformin + thiazolidinedione

No

Yes

No

Yes: mild or moderate and serious episodes

M+S vs metformin + glinide

No

Yes

No

Yes: mild or moderate episodes

M+S vs metformin + SGLT‐2 inhibitor

No

Yes

No

Yes: mild or moderate and serious episodes

DPP4‐I: dipeptidyl‐peptidase 4; GLP‐1: glucagon‐like peptide‐1 agonist; HbA1c: glycosylated haemoglobin A1c; M+S: metformin + sulphonylurea; SGLT‐2: sodium‐glucose transport 2

Figuras y tablas -
Table 3. Overview of HbA1c and hypoglycaemia across comparisons
Comparison 1. Metformin plus sulphonylurea vs metformin plus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2 Serious adverse events Show forest plot

2

771

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.59, 1.61]

3 Cardiovascular mortality Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4 Non‐fatal myocardial infarction Show forest plot

2

771

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.08, 5.10]

5 Heart failure Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

6 Non‐serious adverse events Show forest plot

2

771

Risk Ratio (M‐H, Random, 95% CI)

1.25 [0.96, 1.64]

7 Mild/moderate hypoglycaemia Show forest plot

2

771

Risk Ratio (M‐H, Random, 95% CI)

3.93 [0.71, 21.88]

8 Serious hypoglycaemia Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

9 Weight change Show forest plot

2

476

Mean Difference (IV, Random, 95% CI)

3.37 [1.35, 5.39]

10 Change in HbA1c Show forest plot

2

472

Mean Difference (IV, Random, 95% CI)

‐0.47 [‐1.07, 0.14]

Figuras y tablas -
Comparison 1. Metformin plus sulphonylurea vs metformin plus placebo
Comparison 2. Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

3

2594

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.49, 2.67]

2 Serious adverse events Show forest plot

3

2594

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.73, 1.11]

3 Cardiovascular mortality Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4 Non‐fatal myocardial infarction Show forest plot

2

1575

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.12, 2.82]

5 Heart failure Show forest plot

3

2594

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.10, 2.77]

6 End‐stage renal disease Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

7 Non‐serious adverse events Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

8 Mild/moderate hypoglycaemia Show forest plot

3

2594

Risk Ratio (M‐H, Random, 95% CI)

3.24 [2.05, 5.13]

9 Serious hypoglycaemia Show forest plot

3

2594

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.16, 6.30]

10 Weight (change) Show forest plot

5

1777

Mean Difference (IV, Random, 95% CI)

5.54 [3.62, 7.46]

11 Change in HbA1c Show forest plot

5

2346

Mean Difference (IV, Random, 95% CI)

0.01 [‐0.15, 0.17]

Figuras y tablas -
Comparison 2. Metformin plus sulphonylurea vs metformin plus GLP‐1 analogue
Comparison 3. Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

9

11694

Risk Ratio (M‐H, Random, 95% CI)

1.32 [0.76, 2.28]

1.1 Trials with long duration (≥ 2 years)

6

9909

Risk Ratio (M‐H, Random, 95% CI)

1.38 [0.72, 2.64]

1.2 Trials with short duration (< 2 years)

3

1785

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.14, 7.20]

2 Serious adverse events Show forest plot

9

11694

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.97, 1.18]

2.1 Trials with long duration (≥ 2 years)

6

9909

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.97, 1.19]

2.2 Trials with short duration (< 2 years)

3

1785

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.61, 1.68]

3 Cardiovascular mortality Show forest plot

6

6874

Risk Ratio (M‐H, Random, 95% CI)

1.54 [0.63, 3.79]

3.1 Trials with long duration (≥ 2 years)

5

6810

Risk Ratio (M‐H, Random, 95% CI)

1.54 [0.63, 3.79]

3.2 Trials with short duration (< 2 years)

1

64

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Non‐fatal myocardial infarction Show forest plot

6

6874

Risk Ratio (M‐H, Random, 95% CI)

1.45 [0.69, 3.07]

4.1 Trials with long duration (≥ 2 years)

5

6810

Risk Ratio (M‐H, Random, 95% CI)

1.45 [0.69, 3.07]

4.2 Trials with short duration (< 2 years)

1

64

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

5 Heart failure Show forest plot

8

10691

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.47, 2.34]

5.1 Trials with long duration (≥ 2 years)

6

9909

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.33, 1.86]

5.2 Trials with short duration (< 2 years)

2

782

Risk Ratio (M‐H, Random, 95% CI)

6.00 [0.73, 49.59]

6 Non‐fatal stroke Show forest plot

4

5093

Risk Ratio (M‐H, Random, 95% CI)

2.21 [0.74, 6.58]

6.1 Trials with long duration (≥ 2 years)

3

5029

Risk Ratio (M‐H, Random, 95% CI)

2.21 [0.74, 6.58]

6.2 Trials with short duration (< 2 years)

1

64

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

7 Non‐serious adverse events Show forest plot

7

7592

Risk Ratio (M‐H, Random, 95% CI)

1.18 [1.03, 1.35]

7.1 Trials with long duration (≥ 2 years)

5

6810

Risk Ratio (M‐H, Random, 95% CI)

1.21 [1.04, 1.42]

7.2 Trials with short duration (< 2 years)

2

782

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.82, 1.21]

8 Mild/moderate hypoglycaemia Show forest plot

7

9973

Risk Ratio (M‐H, Random, 95% CI)

7.42 [4.77, 11.53]

8.1 Trials with long duration (≥ 2 years)

5

9051

Risk Ratio (M‐H, Random, 95% CI)

6.67 [4.32, 10.28]

8.2 Trials with short duration (< 2 years)

2

922

Risk Ratio (M‐H, Random, 95% CI)

39.09 [7.69, 198.82]

9 Serious hypoglycaemia Show forest plot

8

10691

Risk Ratio (M‐H, Random, 95% CI)

8.04 [3.31, 19.53]

9.1 Trials with long duration (≥ 2 years)

5

9051

Risk Ratio (M‐H, Random, 95% CI)

8.66 [3.10, 24.16]

9.2 Trials with short duration (< 2 years)

3

1640

Risk Ratio (M‐H, Random, 95% CI)

6.46 [1.10, 37.85]

10 Weight change (kg) Show forest plot

9

10228

Mean Difference (IV, Random, 95% CI)

2.15 [1.71, 2.58]

10.1 Trials with long duration (≥ 2 years)

6

8667

Mean Difference (IV, Random, 95% CI)

2.25 [1.71, 2.78]

10.2 Trials with short duration (< 2 years)

3

1561

Mean Difference (IV, Random, 95% CI)

1.78 [1.27, 2.30]

11 Change in HbA1c Show forest plot

9

9320

Mean Difference (IV, Random, 95% CI)

‐0.05 [‐0.13, 0.03]

11.1 Trials with long duration (≥ 2 years)

6

7779

Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.14, 0.07]

11.2 Trials with short duration (< 2 years)

3

1541

Mean Difference (IV, Random, 95% CI)

‐0.09 [‐0.25, 0.07]

12 Fasting plasma glucose Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

13 BMI Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 3. Metformin plus sulphonylurea vs metformin plus DPP‐4 inhibitor
Comparison 4. Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2 Serious adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3 Cardiovascular mortality Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4 Non‐fatal myocardial infarction Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5 Non‐serious adverse events Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

6 Mild/moderate hypoglycaemia Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

7 Serious hypoglycaemia Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

8 Weight change (kg) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

9 Change in HbA1c (%) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 4. Metformin plus sulphonylurea vs metformin plus long‐acting DPP‐4 inhibitor
Comparison 5. Metformin plus sulphonylurea vs metformin plus thiazolidinedione

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

6

6654

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.85, 1.40]

1.1 Rosiglitazone

4

2996

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.86, 1.68]

1.2 Pioglitazone

2

3658

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.42, 2.80]

2 Serious adverse events Show forest plot

6

6654

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.93, 1.11]

2.1 Rosiglitazone

4

2996

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.81, 1.29]

2.2 Pioglitazone

2

3658

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.83, 1.18]

3 Cardiovascular mortality Show forest plot

4

5940

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.36, 1.67]

3.1 Rosiglitazone

3

2912

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.30, 2.72]

3.2 Pioglitazone

1

3028

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.14, 1.48]

4 Non‐fatal myocardial infarction Show forest plot

3

3718

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.68, 2.14]

4.1 Rosiglitazone

2

690

Risk Ratio (M‐H, Random, 95% CI)

2.93 [0.12, 71.65]

4.2 Pioglitazone

1

3028

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.66, 2.10]

5 Heart failure Show forest plot

5

6570

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.43, 1.04]

5.1 Rosiglitazone

3

2912

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.41, 1.33]

5.2 Pioglitazone

2

3658

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.31, 1.16]

6 Non‐fatal stroke Show forest plot

2

3123

Risk Ratio (M‐H, Random, 95% CI)

1.29 [0.67, 2.47]

6.1 Rosiglitazone

1

95

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6.2 Pioglitazone

1

3028

Risk Ratio (M‐H, Random, 95% CI)

1.29 [0.67, 2.47]

7 Amputation of lower extremity Show forest plot

2

3123

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

7.1 Rosiglitazone

1

95

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

7.2 Pioglitazone

1

3028

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

8 Blindness or severe vision loss Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

9 End‐stage renal disease Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10 Non‐serious adverse events Show forest plot

5

6024

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.44, 2.01]

10.1 Rosiglitazone

4

2996

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.92, 1.08]

10.2 Pioglitazone

1

3028

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.82, 1.05]

11 Mild/moderate hypoglycaemia Show forest plot

5

6059

Risk Ratio (M‐H, Random, 95% CI)

3.63 [2.98, 4.44]

11.1 Rosiglitazone

3

2401

Risk Ratio (M‐H, Random, 95% CI)

3.76 [2.81, 5.02]

11.2 Pioglitazone

2

3658

Risk Ratio (M‐H, Random, 95% CI)

4.78 [1.93, 11.87]

12 Serious hypoglycaemia Show forest plot

5

6570

Risk Ratio (M‐H, Random, 95% CI)

3.98 [0.34, 46.01]

12.1 Rosiglitazone

3

2912

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.37, 3.68]

12.2 Pioglitazone

2

3658

Risk Ratio (M‐H, Random, 95% CI)

24.68 [3.34, 182.16]

13 Weight (change) Show forest plot

7

6877

Mean Difference (IV, Random, 95% CI)

‐0.55 [‐2.75, 1.64]

13.1 Rosiglitazone

3

2865

Mean Difference (IV, Random, 95% CI)

‐0.96 [‐4.77, 2.86]

13.2 Pioglitazone

4

4012

Mean Difference (IV, Random, 95% CI)

‐0.44 [‐1.36, 0.47]

14 Change in HbA1c Show forest plot

10

7020

Mean Difference (IV, Random, 95% CI)

0.17 [0.04, 0.30]

14.1 Rosiglitazone

5

2940

Mean Difference (IV, Random, 95% CI)

0.20 [‐0.02, 0.42]

14.2 Pioglitazone

5

4080

Mean Difference (IV, Random, 95% CI)

0.15 [‐0.04, 0.34]

Figuras y tablas -
Comparison 5. Metformin plus sulphonylurea vs metformin plus thiazolidinedione
Comparison 6. Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

6

6654

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.85, 1.40]

1.1 Trials with long duration (≥ 2 years)

4

5964

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.85, 1.40]

1.2 Trials with short duration (< 2 years)

2

690

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.14, 6.89]

2 Serious adverse events Show forest plot

6

6654

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.93, 1.11]

2.1 Trials with long duration (≥ 2 years)

4

5964

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.93, 1.11]

2.2 Trials with short duration (< 2 years)

2

690

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.32, 1.42]

3 Cardiovascular mortality Show forest plot

4

5940

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.36, 1.67]

3.1 Trials with long duration (≥ 2 years)

2

5250

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.38, 1.89]

3.2 Trials with short duration (< 2 years)

2

690

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.01, 4.05]

4 Non‐fatal myocardial infarction Show forest plot

3

3718

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.68, 2.14]

4.1 Trials with long duration (≥ 2 years)

1

3028

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.66, 2.10]

4.2 Trials with short duration (< 2 years)

2

690

Risk Ratio (M‐H, Random, 95% CI)

2.93 [0.12, 71.65]

5 Heart failure Show forest plot

5

6570

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.43, 1.04]

5.1 Trials with long duration (≥ 2 years)

3

5880

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.43, 1.04]

5.2 Trials with short duration (< 2 years)

2

690

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.06, 15.54]

6 Non‐fatal stroke Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

6.1 Trials with long duration (≥ 2 years)

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6.2 Trials with short duration (< 2 years)

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

7 Non‐serious adverse events Show forest plot

5

6024

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.44, 2.01]

7.1 Trials with long duration (≥ 2 years)

3

5334

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.39, 2.42]

7.2 Trials with short duration (< 2 years)

2

690

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.48, 1.81]

8 Mild/moderate hypoglycaemia Show forest plot

5

6059

Risk Ratio (M‐H, Random, 95% CI)

3.63 [2.98, 4.44]

8.1 Trials with long duration (≥ 2 years)

4

5964

Risk Ratio (M‐H, Random, 95% CI)

3.73 [2.95, 4.72]

8.2 Trials with short duration (< 2 years)

1

95

Risk Ratio (M‐H, Random, 95% CI)

2.04 [0.39, 10.63]

9 Serious hypoglycaemia Show forest plot

5

6570

Risk Ratio (M‐H, Random, 95% CI)

3.98 [0.34, 46.01]

9.1 Trials with long duration (≥ 2 years)

3

5880

Risk Ratio (M‐H, Random, 95% CI)

4.61 [0.14, 149.68]

9.2 Trials with short duration (< 2 years)

2

690

Risk Ratio (M‐H, Random, 95% CI)

2.93 [0.12, 71.65]

10 Weight change Show forest plot

7

6877

Mean Difference (IV, Random, 95% CI)

‐0.55 [‐2.75, 1.64]

10.1 Trials with long duration (≥ 2 years)

3

5833

Mean Difference (IV, Random, 95% CI)

‐1.49 [‐4.79, 1.81]

10.2 Trials with short duration (< 2 years)

4

1044

Mean Difference (IV, Random, 95% CI)

0.20 [‐2.15, 2.56]

11 Change in HbA1c Show forest plot

10

7020

Mean Difference (IV, Random, 95% CI)

0.17 [0.04, 0.30]

11.1 Trials with long duration (≥ 2 years)

4

5896

Mean Difference (IV, Random, 95% CI)

0.17 [‐0.04, 0.39]

11.2 Trials with short duration (< 2 years)

6

1124

Mean Difference (IV, Random, 95% CI)

0.17 [‐0.02, 0.37]

Figuras y tablas -
Comparison 6. Metformin plus sulphonylurea vs metformin plus thiazolidinedione (subgroups duration of intervention)
Comparison 7. Metformin plus sulphonylurea vs metformin plus glinide

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2 Serious adverse events Show forest plot

3

874

Risk Ratio (M‐H, Random, 95% CI)

1.68 [0.54, 5.21]

3 Cardiovascular mortality Show forest plot

2

446

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Mild/moderate hypoglycaemia Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5 Serious hypoglycaemia Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

6 Weight change Show forest plot

2

619

Mean Difference (IV, Fixed, 95% CI)

1.11 [‐0.06, 2.29]

7 Change in HbA1c Show forest plot

3

852

Mean Difference (IV, Random, 95% CI)

0.16 [‐0.64, 0.96]

Figuras y tablas -
Comparison 7. Metformin plus sulphonylurea vs metformin plus glinide
Comparison 8. Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

4

5134

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.44, 2.09]

2 Serious adverse events Show forest plot

4

5134

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.76, 1.37]

3 Cardiovascular mortality Show forest plot

3

3589

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.33, 4.41]

4 Non‐fatal myocardial infarction Show forest plot

2

2264

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.49, 4.18]

5 Heart failure Show forest plot

3

3809

Peto Odds Ratio (Peto, Fixed, 95% CI)

9.21 [1.26, 67.24]

6 Non‐fatal stroke Show forest plot

2

2775

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.22, 3.34]

7 Amputation of lower extremity Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

8 Non‐serious adverse events Show forest plot

3

3809

Risk Ratio (M‐H, Random, 95% CI)

1.27 [1.01, 1.59]

9 Mild/moderate hypoglycaemia Show forest plot

3

3309

Risk Ratio (M‐H, Random, 95% CI)

5.60 [2.38, 13.14]

10 Serious hypoglycaemia Show forest plot

4

5134

Risk Ratio (M‐H, Random, 95% CI)

6.16 [2.92, 12.97]

11 Weight change Show forest plot

3

3294

Mean Difference (IV, Random, 95% CI)

4.41 [4.05, 4.77]

12 Change in HbA1c Show forest plot

4

4182

Mean Difference (IV, Random, 95% CI)

0.09 [‐0.07, 0.24]

Figuras y tablas -
Comparison 8. Metformin plus sulphonylurea vs metformin plus SGLT‐2 inhibitor