Abstract
For decades, sulfonylureas (SUs) have been important drugs in the antidiabetic therapeutic armamentarium. They have been used as monotherapy as well as combination therapy. Focus on newer drugs and concerns about the risk of severe hypoglycemia and weight gain with some SUs have led to discussion on their safety and utility. It has to be borne in mind that the adverse events associated with SUs should not be ascribed to the whole class, as many modern SUs, such as glimepiride and gliclazide modified release, are associated with better safety profiles. Furthermore, individualization of treatment, using SUs in combination with other drugs, backed with careful monitoring and patient education, ensures maximum benefits with minimal side effects. The current guidelines, developed by experts from Africa, Asia, and the Middle East, promote the safe and smart use of SUs in combination with other glucose-lowering drugs.
Keywords: Gliclazide, glimepiride, sulfonylureas, type 2 diabetes
EXECUTIVE SUMMARY
Sulfonylureas (SUs) in oral combination therapy:
A1. Modern SUs (glimepiride and gliclazide modified release [MR]) are effective and safe second-line agents in patients who have not achieved predecided glycemic targets with metformin monotherapy (Grade A; evidence level [EL] 1)
A2. Modern SUs are effective and safe as initial therapy if used in combination with lifestyle modification and metformin, in patients with a baseline glycated hemoglobin ≥7.5% (Grade A; EL 1)
A3. SUs may be considered for use in combination with all classes of oral antidiabetic drugs except glinides (Grade A, EL1)
A4. If not used earlier, modern SUs may be preferred as third-line agents for the management of uncontrolled diabetes with dual combination therapy, owing to better safety profile than older SUs (Grade A, EL 1)
A5. Fixed-dose combinations (FDCs) containing SUs reduce cost, offer convenience, and improve patient adherence (Grade B; EL 1); hence, FDCs with varying strengths of SU + metformin should be made available, while SU + other drugs may be considered (Grade A; EL 4).
Comparative assessment as dual therapy with metformin:
B1. Compared to metformin uptitration beyond half-maximal dose, the addition of SU to metformin demonstrates better glucose-lowering efficacy, safety, and tolerability (Grade A, EL 1)
B2. Compared to pioglitazone, SUs demonstrate good glucose-lowering efficacy with significantly lower risk of weight gain (Grade A, EL 1)
B3. Compared to dipeptidyl peptidase 4 inhibitors, SUs demonstrate better and more durable glucose-lowering efficacy; however, the likelihood of increase in body weight and risk of hypoglycemia should be taken into consideration (Grade A, EL 1)
B4. Compared to sodium glucose co-transporter 2 inhibitors, SUs show noninferior glycemic control; however, safety criteria need to be considered while preferring either class (Grade A, EL 1)
B5. Compared to glucagon-like peptide 1 receptor agonists, SUs show similar glycemic efficacy, with acceptable safety at lower cost (Grade A, EL 1).
SU and insulin combination:
C1. Modern SUs may be continued, with appropriate precaution, when basal insulin is initiated (Grade A; EL1)
C2. Modern SUs may be continued, in the antipodal meal, if premixed insulin is initiated once daily (Grade A; EL1)
C3. Short-acting SUs, or glinides, may be continued or added to the third meal, with appropriate glucose monitoring if premixed insulin is initiated twice daily (Grade B; EL 1).
Use in special populations:
D1. Combinations containing modern SUs can be used in elderly patients as they are associated with low risk of hypoglycemia (Grade A; EL 1)
D2. SUs (glibenclamide) may be used in the glycemic control of neonatal diabetes (KCNJ11, ABCC8 gene mutations) and Maturity-Onset Diabetes of the Young 3 (MODY 3) (Grade A; EL 3)
D3. The evidence base for the use of SUs in adolescents with type 2 diabetes is limited (Grade A; EL 4)
D4. There is insufficient evidence to recommend the use of SUs, as monotherapy or in combination, to be used during pregnancy and lactation (Grade A; EL 2).
Use in comorbid conditions:
E1. There is insufficient evidence to suggest that modern SUs increase cardiovascular (CV) risk. Modern SUs are preferred over conventional SUs in patients with diabetes and cardiovascular disease (CVD) (Grade A; EL 1)
E2. Among SUs, short-acting drugs, especially those metabolized in the liver (glipizide), should be preferred in patients with moderate/severe renal impairment. In mild/moderate renal impairment, modern SUs may also be used, preferably at lower doses (Grade A; EL 3)
E3. Reductions of SU dose and/or longer intervals between dosing are recommended in patients with mild/moderate hepatic impairment (Grade B; EL 4).
SUs in combination and Ramadan:
F1. Modern SUs may be used in combination with other drugs during Ramadan, with appropriate counseling and dose modification (Grade A; EL 3)
F2. Individuals on once-daily SU should take their medication at Iftar (evening meal) (Grade A; EL 3)
F3. Individuals on twice-daily SU may shift the morning dose to Iftar and half of the evening dose to Suhur (morning meal) (Grade A; EL 4)
F4. Patients on SU and premixed insulin should consider reducing the dose of either drug or shifting from premix to low-peak basal insulin during Ramadan (Grade A; EL 4)
F5. Dose titration during Ramadan should be based on twice-weekly or weekly glucose monitoring (Grade A; EL 3).
INTRODUCTION
Epidemiology and burden of diabetes
Type 2 diabetes mellitus (T2DM), a progressive metabolic disorder, is continuously gaining the status of a potential epidemic in the world. According to the 2015 global estimates of International Diabetes Federation (IDF), about 415 million people (1 in 11 adults) have been shown to present with diabetes and is expected to reach 642 million (1 in 10 adults) by 2040.[1] Moreover, around 318 million adults are associated with impaired glucose tolerance, who are at a high risk of developing diabetes in the future. The 2015 regional fact sheet of IDF estimates the prevalence to be 10.7%, 8.5%, and 3.2% in the Middle East and North Africa (MENA), South East Asia (SEA), and African regions, respectively.[1] This increasing burden of the disease may contribute to increased rate of complications, reduction of quality of life, and premature mortality. As per the 2016 World Health Organization (WHO) global report on diabetes, the high blood glucose age-standardized mortality rates are 139.6, 115.3, and 111.3/100,000 in WHO Eastern Mediterranean, South-East Asia, and African regions, respectively.[2]
Prescription pattern of oral antidiabetic drugs
Tight glycemic control reduces the associated complications and improves the quality of life in patients with T2DM. The United Kingdom Prospective Diabetes Study trial reported that each 1% reduction of glycated hemoglobin (A1C) decreases approximately 12%–43% risk of diabetes-related mortality and morbidity.[3] Numerous antidiabetic agents are currently available as monotherapy or in combination therapy for the treatment of T2DM. However, oral antidiabetic drugs (OADs) still dominate the prescribing pattern (56.4%) followed by insulin alone (43.6%).[4,5] Furthermore, sulfonylureas (SUs) alone or in combination with metformin have been the most commonly prescribed OADs in some Afro-Asian countries.[6,7,8,9]
SUs can be classified either according to their hierarchy of development (conventional and modern SUs) or based on the duration of action (short-, intermediate-, and long-acting). The classification has been described in Table 1. This helps to avoid confusion during their use and can be effectively utilized in patients with variable clinical scenario.
Table 1.
Situational analysis of sulfonylureas
The conventional or modern SUs are widely used as second-line agents in the management of T2DM in most countries of Africa, MENA, and SEA region due to low cost and high efficacy. Among all, the combination of glimepiride (GLIM) and metformin is available in most of these countries. The National List of Essential Medicines (NLEM) in different countries containing SUs along with metformin is shown in Table 2. In 2015, the Indian NLEM was updated to align with the current treatment guidelines. Where more drugs were available within a therapeutic class, the core committee considered the best-suited one after due deliberation and careful evaluation of their relative safety, efficacy, and cost. Accordingly, glibenclamide (GLIB) has been replaced with GLIM in the diabetes section.[10] Tolbutamide (TOLB) and chlorpropamide (CHOL) are still used in Sri Lanka and Tanzania, respectively; gliquidone is available in Indonesia and Egypt. Fixed-dose combinations (FDCs) containing either conventional or modern SUs are available in most of the countries in Africa, MENA, and SEA regions except Oman and Sri Lanka. Furthermore, basal or premix insulin in combination with SUs is also prescribed for the management of T2DM in different parts of the world. A complete list of situational analysis of SUs in participating countries is summarized in Table 3.
Table 2.
Table 3.
RATIONALE AND METHODOLOGY
Pathophysiological basis
During the course of treatment, progressive nature of T2DM with a gradual decline in the functional β-cells leads to continuing the decrease in the glucose-lowering efficacy of OADs over time.[11] Evidence suggests that early combination therapy with intensive glycemic control can be an effective approach for better preservation of β-cell function, which may quickly attain the target glycemic level and reduce diabetic-related complications.[12,13,14,15,16,17] Early introduction of combination therapy also reduces complications associated with uptitration of monotherapies. The delay in stepping up from monotherapy to combination therapy in the step-wise approach contributes to long periods of hyperglycemia and an increased risk of macro- and microvascular complications.[18]
Pharmaceutical advances
Metformin produces its antihyperglycemic action without affecting the insulin secretion; hence, it is beneficial in combining metformin with insulin secretagog, like an SU. Among OADs available as add-on therapies to metformin, modern SUs can be considered as an ideal option owing to their high efficacy, relative cardiovascular (CV) safety, and low cost. The risk of hypoglycemia and weight gain can be minimized using modern SUs such as GLIM and gliclazide (GLIC) modified release (MR) with fewer side effects and better efficacy, which also has contributed to their wider use. Furthermore, combination therapies show a greater blood glucose-lowering effect than that of a single agent, which has been demonstrated in a number of studies and has resulted in the marketing of FDC preparations.[11,19]
Methodology
The current consensus reviews the recent evidence on SUs and presents evidence-based recommendations on the use of SUs and their combination for the management of T2DM. In order to impart the highest possible evidence base for the use of SUs in combination in the management of T2DM, a systematic review of the literature was initiated. Existing guidelines, meta-analyses, systematic reviews, randomized controlled trials (RCTs), non-RCTs, and key cited articles relating to T2DM management were reviewed and recommendations were framed. Recommendations for each section of the consensus statement were discussed by the expert panels and where there was a little or no evidence, the panel relied on logical empiricism and consensus to make their recommendations. The current consensus is developed in accordance with the American Association of Clinical Endocrinologists protocol for standardized production of clinical practice guidelines.[20] Recommendations are based on clinical importance (graded as A: strong, B: intermediate, C: weak, and D: no evidence based), which were coupled with four intuitive levels of evidence (1 = “at least one RCT or meta-analysis of RCTs,” 2 = “at least one nonrandomized or noncontrolled, prospective epidemiological study,” 3 = “cross-sectional or observational or surveillance or pilot study,” and 4 = “existing guideline or consensus expert opinion on extensive patient experience or review”) [Table 4.]
Table 4.
SULFONYLUREAS AND TYPE 2 DIABETES MELLITUS
History
SUs have been a cornerstone in the T2DM management for the past 60 years. TOLB was the first SU marketed in the 1950s. This was followed by the introduction of the other first-generation agents such as CHOL, acetohexamide, and tolazamide. The next advancement in SU therapy was the development of potent second-generation agents such as GLIB and glipizide (GLIP) in the year 1984 in the United States. Furthermore, GLIM, a third-generation agent with eminent characteristics, was released into the market in the year 1995.[21,22,23] SUs with respect to their generation, history of development, duration of action, and other pharmacokinetic/pharmacodynamics profiles are described in Table 5.[5]
Table 5.
Mechanism of action and differential effects of sulfonylureas
SUs have been categorized as insulin secretagogs. They act by stimulating the pancreatic β-cells to secrete insulin. SUs mainly bind to the SU receptors (SURs), a subunit of potassium ATP-dependent (KATP) channels located in the β-cell membrane, which eventually blocks the potassium channels and facilitates the influx Ca2+ into the cell. This leads to cell depolarization and subsequently accelerates insulin exocytosis [Figure 1a].[5,23] Furthermore, owing to their cell-mediated and nonglucose-mediated action, all SUs are more effective in the early stages of T2DM when the β-cell function is to its greatest ability.[11,24] All the SUs are eliminated by liver and kidney and well tolerated by adult patients; however, hypoglycemia and weight gain are the concerns with conventional SUs.[25,26]
The affinity of SUs varies with different SUR subunits present in KATP channels.[5] It is reported that GLIB blocks both SUR1 (pancreatic β-cells) and SUR2 (cardiac and skeletal muscles) subunits with similar affinity. The modern SU, GLIM, blocks SUR1 and preferentially the sarcolemmal SUR2 while sparing the mitochondrial SUR. A number of studies, however, have demonstrated that GLIM does not have a negative impact on cardiac function and has less effect on the electrical properties of the heart.[27,28] GLIC and TOLB selectively block only SUR1 compared to SUR2.[29,30] The affinity of SUR1 protein toward sulfonyl moiety is 100–1000 folds more compared to SUR2 protein.[31] Furthermore, GLIC is the only SU which does not bind to the Epac2 receptor [Figure 1b][5], a stimulating factor for insulin exocytosis, which may confer a lower risk of hypoglycemia.[5,32] Similarly, GLIM also confers a low rate of hypoglycemia and weight gain than conventional SUs due to its lower binding affinity (2–3 folds) and quick association and dissociation with SUR proteins.[33,34,35] Furthermore, SUs inhibit the mitochondrial KATP channels in cardiac myocytes, which contributes to impairment of ischemic preconditioning; however, GLIM does not exert this effect and preserves myocardial ischemic preconditioning.[5] Moreover, a lesser pancreatic overstimulation and resultant low hypoglycemia by GLIC could be due to the restoration of the early insulin peak in response to glucose stimulation and higher reversibility of binding of GLIC to the SUR1 of β-cell.[36] In addition, modern SUs also exhibit certain pleiotropic effects such as insulin clearance, glucagon secretion, insulin sensitization, anti-oxidative effect, angiogenesis, vascular health, and ischemic preconditioning.[5]
Summary of guidelines recommending SUs and combinations for the management of type 2 diabetes mellitus
The addition of SUs has been the gold standard combination therapy for decades in patients who fail to achieve target glycemic control with metformin monotherapy.[37] In addition, pertaining to their low cost and high efficacy, SUs are widely used as a second-line agent in different regions of the world for the management of diabetes. Moreover, modern SUs such as GLIM and GLIC MR are preferred over other SUs in diabetic patients due to low risk of hypoglycemia and CV neutrality.[36,38,39,40] A summary of guidelines recommending SUs in combination therapy for the management of T2DM in various countries is depicted in Table 6.
Table 6.
SULFONYLUREAS AND COMBINATIONS IN THE MANAGEMENT OF TYPE 2 DIABETES MELLITUS
Sulfonylurea + metformin
Mechanism
A combination of two drugs with the complementary mechanism of action may help in addressing multiple etiologies of hyperglycemia in patients with T2DM.[5] Metformin has insulin-sensitizing properties. It facilitates insulin uptake by the peripheral tissues and enhances the glucose utilization in adipose and intestinal tissues. SUs increase the sensitivity of β-cells to glucose and facilitate endogenous secretion of insulin. Furthermore, both metformin and SUs may reduce hepatic glucose overproduction by decreasing hepatic gluconeogenesis and glycogenolysis; however, the relative contribution of gluconeogenesis and glycogenolysis by metformin remains controversial.[56,57] Moreover, SUs may inhibit secretion of glucagon from islet cells[58] and also stimulates glycogen synthesis in the liver.[57] Diagrammatic representation of the complementary mechanism is shown in Figure 2.
Glycemic efficacy
The addition of SUs to ongoing metformin monotherapy has shown good glycemic control with acceptable safety and tolerability in numerous meta-analyses and RCTs.
Sulfonylureas add-on to metformin versus metformin monotherapy
A meta-analysis of 15 RCTs each lasting <1 year compared metformin monotherapy with the combination of metformin and SUs. All included studies favored the combination arm over monotherapy for glycemic efficacy with a pooled between-group difference of 0.9% (95% confidence interval [CI]: 0.7%–1.2%).[59] In another meta-analysis, when SU was added to oral medication, A1C was reduced by 1.62% (95% CI: 1.00, 2.24; I2 = 94.1%) in the SU group than in the comparator group.[60] A placebo-controlled study by Ahren et al. with a duration lasting up to 104 weeks compared metformin (>1500 mg daily) with the combination of metformin (≥1500 mg daily) plus GLIM (up to 4 mg daily). The study showed a between-group difference in A1C of 0.63% at the end, favoring the combination arm.[61] In a randomized, open-label, parallel group, multicenter trial, GLIM/metformin FDC therapy provided significantly greater adjusted mean decreases in A1C (−1.2 vs. −0.8%, P < 0.0001) and fasting plasma glucose (FPG) (−35.7 vs. −18.6 mg/dL, P < 0.0001) compared with metformin uptitration. Furthermore, a significantly greater proportion of patients with GLIM/metformin FDC therapy achieved A1C <7% (74.7 vs. 46.6%, P < 0.0001) at the end of the study.[62] In a multicentric epidemiologic surveillance protocol of 60 days, patients (n = 759) with T2DM were prospectively prescribed 1–2 tablets of GLIC 60 mg + metformin 500 mg during the course of daily practice. The study reported that 62.5% of patients had achieved the primary outcome (FPG of 90–130 mg/dL) at the end of the study. Mean (95% CI) FPG (mg/dL) decreased from baseline by 48.7 (45.0–51.4) with 1 tablet, by 71.3 (66.0–76.6) with 1½ tablets, and by 86.3 (75.7–96.9) with 2 tablets. Furthermore, the frequency of hypoglycemia was reported as 0.7%.[63]
A summary of published RCTs comparing the combinations of different SUs and metformin with metformin monotherapy in the management of T2DM is summarized in Table 7.
Table 7.
Sulfonylureas versus thiazolidinedione as add-on to metformin
Compared to add-on therapies with other OADs, SUs as an add-on to metformin have shown a favorable glycemic control in terms of significant A1C reduction. A meta-analysis comparing the outcomes of thiazolidinedione (TZD) and SUs, both add-ons to metformin, reported a pooled between-group difference in A1C of −0.06% (95% CI: −0.19%–0.06%).[59] Furthermore, a multicentric randomized, parallel group, open-label, forced titration study reported that GLIM therapy resulted in a more rapid decline in A1C levels at weeks 6, 12, and 20 in comparison to pioglitazone (P < 0.05) in patients with uncontrolled glycemic level with metformin. A target A1C ≤7% was reached faster in the GLIM group (median, 80–90 days vs. 140–150 days [P = 0.024]).[72] Evidence reported no significant difference in glycemic parameters between GLIC and pioglitazone when added in patients with uncontrolled glycemic level with metformin monotherapy.[73,74]
Sulfonylureas versus dipeptidyl peptidase 4inhibitor as add-on to metformin
SUs compared to dipeptidyl peptidase 4 (DPP-4) inhibitors (DPP-4I) when added to metformin are associated with a significantly greater reduction in A1C from baseline to 12 weeks (mean difference [MD]: 0.21; 95% CI: 0.06, 0.35) but no significant difference at 52 and 104 weeks (MD: 0.06 and 0.02, respectively, 95% CI: −0.03, 0.15 and −0.13, 0.18 respectively).[75] Furthermore, accumulated evidence from 14 RCTs (n > 10,000) reported that SUs were associated with a larger decline in A1C compared to DPP-4I (weighted mean difference [WMD]: 0.08, 95% CI: 0.03, 0.14, P = 0.001).[76] Moreover, a systematic review and meta-analysis reported that GLIM was associated with a 12% greater reduction in A1C compared to DPP-4I (WMD: −0.12; 95% CI: −0.16, −0.07), with a clinically irrelevant weight difference between the treatment (2.1 kg).[77] A 52-week RCT comparing GLIC and vildagliptin as a second-line agent reported a noninferiority of vildagliptin, with a mean change from A1C (−0.85% ± 0.06 vs. −0.81% ± 0.06).[78] Another 52-week RCT also demonstrated noninferiority of saxagliptin versus GLIP; adjusted mean changes from baseline A1C were 0.74% versus 0.80%; the between-group difference was 0.06% (95% CI: −0.05, 0.16).[79] However, in a population-based cohort study in Denmark (4734 patients), A1C reduction with SUs as a second-line agent was better compared to that of DPP-4I (1.2% vs. 0.8%).[80]
Sulfonylureas versus sodium glucose co-transporter 2 inhibitor as add-on to metformin
In a meta-analysis, the combination of metformin plus an SU with the combination of metformin plus a sodium-glucose co-transporter 2 inhibitor (SGLT-2I) was compared. The results supported the combination of metformin plus an SGLT-2I for A1C reduction (pooled between-group difference in A1C of 0.17%; 95% CI: 0.14%–0.20%).[59] In EMPA-REG-H2HSU-trial, a noninferiority trend in terms of change of A1C has been reported with empagliflozin when compared to GLIM at 104 weeks.[81] Furthermore, in CANTATA-SU trial, no difference in glycemic control was observed with less dose of canagliflozin and GLIM (canagliflozin 100 mg vs. GLIM 6–8 mg [MD − 0.01%]).[82] Similarly a 52-week, double-blind, active-controlled, noninferiority RCT reported a statistically noninferiority in adjusted mean A1C reduction with dapagliflozin (−0.52%) and GLIP (−0.52%) in T2DM patients with inadequate glycemic control with metformin.[83]
Sulfonylureas versus glucagon-like peptide 1 analogs as add-on to metformin
The dose-dependent variability has been observed in terms of glycemic control between glucagon-like peptide-1 (GLP-1) analogs and SUs when added to metformin. A meta-analysis of several RCTs has found favorable outcomes with the combination of metformin plus SU despite submaximal doses of SUs being compared with maximal doses of daily exenatide (pooled between-group difference in A1C, −0.26%; 95% CI: −0.48%–0.03%).[59] In the LEAD-2 trial, liraglutide reported noninferiority in reduction of A1C compared to GLIM, both added on to metformin.[84] Similarly a 16-week RCT including patients from China, India, and South Korea reported that liraglutide 1.2 and 1.8 mg was noninferior to GLIM (mean A1C reduction: 1.36%, 1.45%, and 1.39%, respectively).[85] However, combination of metformin plus maximum dose of albiglutide (titrated to 50 mg weekly) compared to metformin plus submaximal dose of GLIM (titrated to 4 mg daily) favored the metformin plus albiglutide arm (A1C reduction, −0.9% vs. −0.3%) in HARMONY 3 trial.[61]
Sulfonylureas versus insulin as add-on to metformin
In DiaRegis registry (n = 3810), the addition of insulin reduced the A1C level more as compared to SU after the failure of metformin monotherapy in a duration of 2 years (−0.9 ± 2.0% vs. −0.6 ± 1.4%).[86]
Body weight
SU use is associated with weight gain, a secondary effect that also occurs with insulin, TZD, and glinides. Modern SUs such as GLIM and GLIC MR are associated with weight neutralizing/reducing effect compared to conventional SUs.[5] Furthermore, evidence suggests that GLIM may be the least in the class to endorse weight gain.[87,88] Nonetheless, weight gain associated with SUs may be due to improved utilization of consumed glucose and a subsequent reduction in glycosuria, thereby indicating reduction in glucotoxicity.[88]
An extra increment in body weight has been observed when SUs were added to ongoing metformin therapy.[59] When compared to TZD, a pooled mean between-group difference of 0.9 kg (95% CI: 0.4–1.3 kg) favored the combination of metformin plus SU.[59] Similarly, the addition of pioglitazone was associated with more weight gain (2.5 kg) than GLIC (1.2 kg) in patients with metformin.[89] Several meta-analyses and RCTs reported that metformin-SU combination was associated with more weight gain compared to metformin-DPP-4I combinations and metformin-SGLT-2I combinations; however, the difference was nonsignificant.[59,77] Zhang et al. reported that DPP-4I was associated with a reduction in body weight (WMD, −1.652 kg; 95% CI values here as –1.658, –1.646) compared to SUs.[90] Furthermore, in a 16-week prospective study, a variation of weight change has been observed between vildagliptin and GLIC (−0.3 kg vs. +1.4 kg, P = 0.048) after adding to metformin.[91] Similarly, in another trial, canagliflozin 100 and 300 mg and GLIM 6–8 mg/day were associated with −4.1%, −4.2%, and 0.9% reductions in body weight, respectively.[92] Furthermore, several RCTs reported a weight loss in patients with the combination of metformin and GLP-1 receptor agonists and weight gain in patients with the combination of metformin and SUs.[61,85,93,94] Compared to insulin, in BETA trial, addition of GLIM to metformin produced a less weight gain (mean between-group difference in weight of −1.7 kg, P = 0.02) in patients with T2DM.[95] In DiaRegis registry (n = 3810), the addition of insulin also reported increment of body weight from baseline as compared to SU after the failure of metformin monotherapy in a duration of 2 years (+0.8 ± 9.0 vs. −0.4 ± 4.8 kg).[86]
Safety and tolerability
Hypoglycemia is a primary clinical concern during the intensification of the antidiabetic regimen in patients with T2DM. The hypoglycemic potentials of SUs are different pertaining to their variable mode of action and pharmacokinetic and pharmacodynamic properties. Evidence suggests that modern SUs are associated with less risk of hypoglycemia compared to conventional SUs.[39,40,96,97] Furthermore, the European GUIDE (GlUcose control in type 2 diabetes: Diamicron MR vs. GLIM) study (n = 845) was the first double-blind, 27-week, parallel-group, large-scale, head-to-head study compared once-daily GLIC MR (maximum dose up to 120 mg) to once-daily GLIM (maximum dose up to 6 mg) either as monotherapy or in combination. Hypoglycemia occurred significantly lesser with GLIC MR compared to GLIM (3.7% vs. 8.9%, respectively, P = 0.003), though a higher number of patients reached A1C <6.5% with GLIM (17% vs. 2%) which may influence the hypoglycemic episodes. Moreover, no episodes of severe hypoglycemia were reported during the study in patients, reiterating the safety of both these modern SUs.[98] Kim et al. in their randomized, open-label, parallel group, multicentric study reported that patients with GLIM/metformin combination therapy experienced more hypoglycemia compared with metformin uptitration therapy (41% vs. 5.6%, P < 0.0001), but there was no serious hypoglycemia reported in any group.[62] In a 52-week RCT, patients taking GLIC or vildagliptin experienced similar incidence of any adverse events (~61%); however, GLIC patients had more serious adverse events (8.7% vs. 6.7%) and more vildagliptin patients discontinued as a result of an unsatisfactory effect (n = 22 vs. 13).[78]
Evidence shows an increase in hypoglycemia risk with SUs compared to other OADs when added to metformin monotherapy. The hypoglycemic risk, when compared with SUs, for TZD was as follows: pooled odds ratio (OR): 7.5; 95% CI: 4.0–13.8;[59] DPP-4I, risk ratio: 0.24, 95% CI: 0.21–0.27, P < 0.001;[76] and SGLT-2I, OR: 0.08 (95% CI: 0.03–0.17).[59] Furthermore, DPP-4I was associated with lower risk of total adverse events (Mantel–Haenszel odds ratio [MHOR]: 0.79; 95% CI: 0.72–0.87) and CV events (MHOR: 0.53; 95% CI: 0.32–0.87) compared with SUs.[90]
Several RCTs reported that the rate of genital and urinary tract infections was more common with SGLT-2I compared to SUs.[92,99] Furthermore, there were increased odds of genital infections for metformin plus SGLT-2I and differences in relative odds by gender were as follows: pooled OR: 5.2 (95% CI: 3.4–7.8) for women and pooled OR: 7.6 (95% CI: 4.0–14.4) for men.[59] Moreover, volume depletion was observed frequently with canagliflozin than GLIM.[92] Similarly, gastrointestinal events with liraglutide and exenatide[84,100] and diarrhea with albiglutide[61] were more commonly reported.
In a retrospective cohort study, 20,070 patients were newly treated with an SU, DPP-4I, or a TZD following metformin therapy failure. It was reported that the therapy failure at 1 year was 15% with SU, 23% with DPP-4I, and 8% with TZD.[101] Furthermore, multivariate analysis of the data showed that addition of a DPP-4I was associated with an increased risk of treatment failure (adjusted hazard ratio (aHR): 1.58; 95% CI: 1.48–1.68) compared to the SU group, while adding a TZD was associated with a adjusted hazard ratio (aHR: 0.45; 95% CI: 0.41–0.50).
Moreover, choice of drugs should be based on the risk of hypoglycemia in an individual patient; further details regarding this are provided in patient and dose selection sections in this article.
All-cause mortality and macrovascular complications
A meta-analysis including several RCTs demonstrated that SUs (GLIB, GLIP, GLIC, and GLIM) as an add-on to metformin were not associated with all-cause mortality (OR: 1.26; 95% CI: 0.94–1.68; I2 = 0%) and CV mortality (OR: 1.40; 95% CI: 0.61–3.22; I2 = 6%). Furthermore, among all SUs, GLIP was associated with increased all-cause and CV mortality, while the risk was least with GLIM (numerically, though did not achieve statistical significance).[38] Modern SUs show less CV mortality than conventional SUs due to more pancreatic selectivity action. A randomized, double-blind, crossover study of GLIC 80 mg twice daily and GLIM 2 mg once daily, each for 4 weeks as add-on therapy to metformin, found no evidence between SUR1-specific (GLIC) and nonspecific SU (GLIM) in differential effects on arterial distensibility, endothelial function, or vasodilator mechanisms.[102]
Metformin remains an ideal first-line agent and among the SUs, the relative safety of a GLIC makes it a preferred SU. In a population-based study on 107,806 patients from the Danish registry with a median follow-up of 3.3 years, Schramm et al. evaluated several mortality end points with different insulin secretagogs compared with metformin in patients with or without a previous history of myocardial infarction (MI).[103] The all-cause mortality was significantly higher with all SUs (GLIM: HR: 1.32, 95% CI: 1.24–1.40; GLIP: HR: 1.27, 95% CI: 1.17–1.38; and GLIB: HR: 1.19, 95% CI: 1.11–1.28) except GLIC (HR: 1.05, 95% CI: 0.94–1.16) in patients with a previous history of MI. Similar significant increase in all-cause mortality was observed with all SUs except GLIC in those with a previous history of MI. Furthermore, this was a retrospective study and therefore there is a likelihood of selection bias (patients on GLIM had a higher baseline risk and likely contributed to more morbidity and mortality). In another nationwide study on 202,272 Danish patients, GLIP, GLIB, GLIM, and TOLB appeared to be associated with an increased risk compared with GLIC when used in combination with metformin.[104]
When added to metformin, pioglitazone found to produce potential benefits in terms of improvements in specific lipid abnormalities compared to GLIC or GLIM.[74,89,105] Nonetheless, a single retrospective cohort study from a Veterans Affairs population with Medicare (n = 80,936) found a nonsignificant increase in the risk of stroke or MI (composite outcome) for SU-based versus TZD-based therapy: aHR: 1.15 (95% CI: 0.8–1.66; P = 0.46).[106]
The pooled OR from five RCTs on mortality between DPP-4I and SUs, when added on to metformin, was 0.64 (95% CI: 0.27–1.51).[59] In the Danish National Registry, combination therapies with incretin-based drugs and metformin were compared with a combination of metformin and SU in T2DM for all-cause mortality, CV mortality, and combined end point of MI, stroke, and CV mortality. By keeping metformin + SU as a reference, the study demonstrated a significantly decreased risk of death among metformin plus DPP-4I users (n = 11,138) with a relative risk (RR) of 0.65 (0.54–0.80) for mortality, 0.57 (0.40–0.80) for CV mortality, and 0.70 (0.57–0.85) for the combined end point. For metformin + GLP-1 receptor agonist, the RR for mortality was 0.77 (0.51–1.17), for CV mortality was 0.89 (0.47–1.68), and for the combined end point was 0.82 (0.55–1.21).[104] However, a meta-analysis suggested that long-term all-cause mortality (which was low [< 1%] across studies) was similar for metformin plus SGLT-2I and metformin plus SU (pooled OR: 0.86; 95% CI: 0.29–2.55).[59] Moreover, a significant reduction in urine albumin was observed in the metformin plus exenatide arm (37.97%) compared to the metformin plus GLIM arm (5.76%).[107]
Nonetheless, in a meta-analysis of 301 clinical trials which utilized the glucose-lowering drugs including metformin, SUs, TZD, DPP-4I, alpha-glucosidase inhibitor (AGI), SGLT-2I, GLP-1 receptor agonists, meglitinides, and insulin either alone or in combination suggested no significant difference in the risk of CV mortality between the antidiabetic drugs.[108]
A summary of landmark trials comparing outcomes of SUs with other antidiabetic drugs as add-on therapy is summarized in Table 8.
Table 8.
Sulfonylurea + other oral antidiabetic drugs
Mechanism
In patients with inadequate glycemic control on SUs, there are a number of OADs available which can be used in combination with an SU. However, the choice of therapy should be individualized based on patient characteristics, preferences, and cost. TZDs decrease insulin resistance in the periphery and in the liver resulting in increased insulin-dependent glucose disposal and decreased hepatic glucose output. Thus, the combination of SUs and TZD may be beneficial for supporting each other in T2DM.
DPP-4 inhibitors elevate cellular cyclic adenosine monophosphate (cAMP) levels in pancreatic β-cells, leading to potentiated insulin secretion. SUs mimic the glucose-induced KATP channel-dependent pathway. Therefore, an increase in cAMP induced by incretin therapy potentiates KATP channel-independent insulinotropic action by glucose.[111,112,113] Therefore, SUs and DPP-4I may be an effective combination for supporting inappropriate insulin secretion in T2DM; however, careful consideration is required when initiated in the elderly and/or patients with renal insufficiency.[113]
Glycemic efficacy
A combination therapy with SU and TZD resulted in remarkable glycemic control when compared to each monotherapy. A randomized, double-blind, placebo-controlled, multicentric study comparing the efficacy of add-on of GLIM to on-going rosiglitazone reported that combination therapy with GLIM produced greater reductions in A1C (mean [standard error (SE)]: −1.2% [0.1%] vs. −0.3% [02%]; P < 0.001) and FPG (mean [SE]: −24.4 [6.0] mg/dL vs. 5.9 [8.0] mg/dL; P < 0.006).[114] Similarly a 28-week, double-blind, parallel-group RCT revealed that GLIM/rosiglitazone FDC significantly reduced A1C (2.5 ± 1.4%) than rosiglitazone (1.8 ± 1.5%) or GLIM (1.7 ± 1.4%) monotherapy (model-adjusted mean treatment difference, P < 0.0001 vs. both rosiglitazone and GLIM).[115] Evidence suggests that treatment with a combination of pioglitazone and SUs provided extensive glycemic control as compared to baseline glycemic level; however, no significant difference was found when compared to metformin and SU-based combinations.[89,109,116]
In a multicentric, prospective, randomized, open-label study, patients who had an uncontrolled glycemic level with sitagliptin and low-dose GLIM were randomized to receive uptitration with either sitagliptin or GLIM. There was no significant difference in the A1C-lowering effects between the two groups. However, a significant A1C-lowering effect from baseline of GLIM uptitration was found (P < 0.01 vs. baseline).[117] Furthermore in a 52-week, prospective, single-arm study, sitagliptin and low dose GLIC or GLIM also reduced A1C by −0.80% (95% CI: −0.90 to −0.68) (P < 0.001) from baseline.[118]
In conditions where dual therapy failed to obtain optimal glycemic control, the addition of a third agent (SUs) could be helpful in achieving glycemic targets. GLIM as an add-on to metformin and TZD resulted in significant improvement in A1C level (mean [SE]: −1.31% [0.08] vs. −0.33% [0.08], respectively; P < 0.001) from baseline with more patients achieving target A1C ≤ 7% compared with metformin and TZD combination (62.2% vs. 26.0%, P < 0.001).[119] Evidence suggests that GLIM strongly enhances the glucose-lowering effect in triple oral antidiabetic therapy with sitagliptin and metformin for patients with T2DM.[120]
A systematic review and network meta-analysis evaluated the efficacy of triple therapy regimen for T2DM. The study included SUs in all combinations except one (MET + TZD + DPP-4I). For A1C reduction, all triple therapies were statistically superior to MET + SU dual therapy. However, none of the triple therapy combinations demonstrated differences in A1C compared with other triple therapies.[121]
Body weight
In a randomized, double-blind, placebo-controlled, multicentric study at the end of the 1st year, GLIM with pioglitazone (4.9%) and GLIM with rosiglitazone (6.2%) treatment groups had significant increases from baseline in body mass index (BMI) (P < 0.05).[122] Furthermore, when added to GLIC, pioglitazone resulted in an increment of body weight in comparison to metformin (3.7 vs.−1.7 kg).[89] However, in a 52-week, prospective, single-arm study, sitagliptin and low-dose GLIC or GLIM reduced BMI by −0.38 kg/m2 (95% CI:−0.72, –0.04) (P < 0.05) from baseline.[118]
In a prospective observational study, no change in body weight (69.6 ± 3.0–69.1 ± 2.9 kg in the low-dose group and 62.1 ± 2.6–61.9 ± 3.0 kg in the high-dose group; P > 0.05 for both groups) was observed in patients taking combination of 50 mg/day sitagliptin and low-dose GLIM (2 or 3 mg decreased to 1 mg: n = 15) compared to high-dose GLIM (4 or 6 mg decreased to 1 mg).[111]
Safety and tolerability
Evidence suggests that the combination regimens of SU and TZD were well tolerated by the patients with no significant difference in adverse effects with the comparators.[114,115,122,123]
Furthermore, no hypoglycemic episodes were reported by various studies assessing the efficacy of sitagliptin and SU combinations.[111,124] Moreover, several studies reported that the combination treatments with sitagliptin and SU were safe and well tolerated in patients with T2DM.[111,117,118]
In a 30-week, randomized, double-blind, placebo-controlled, parallel-group study, the risk of hypoglycemia (51.2% vs. 8.3%, P < 0.001) was greater with GLIM add-on to metformin-TZD than placebo.[119] Similarly, in EUREXA trial, the ratio of documented symptomatic (blood glucose ≤70 mg/dl [3.9 mmol/L]) hypoglycemic rates for add-on GLIM to add-on TZD was 8.48 (P < 0.0001).[125]
All-cause mortality and macrovascular complications
The increase of lipid risk factors for CV diseases (CVDs) from baseline was more significant in GLIM-rosiglitazone patients compared to GLIM-pioglitazone patients.[122,126] In an Indian RCT, FDC containing GLIM (2 mg), pioglitazone (15 mg), and metformin sustained release (500 mg) significantly reduced the levels of triglyceride, low-density lipoprotein cholesterol, and total cholesterol.[127]
A summary of published RCTs comparing combinations of SUs and other OADs in the management of T2DM is shown in Table 9.
Table 9.
Sulfonylurea + insulin
Mechanism
SUs when added to insulin increase endogenous insulin secretion and possibly exert some extra pancreatic actions on muscle and liver, thereby improving glycemic control and decreasing daily insulin requirements.[128] A subset of T2DM patients who are mild to moderately obese, have adequate endogenous insulin secretory reserve, and are in poor glycemic regulation (A1C >10%), despite twice-daily insulin administration, may show significant improvement in glycemic regulation and/or decrease in insulin daily dose of insulin-SU therapy.[129] Evidence suggests that addition of SUs to insulin in patients with uncontrolled glycemic levels reduces the insulin dose by 20%–30% and avoids hypoglycemia.[130] Therefore, studies have compared the treatments with a smaller insulin dose with SU and a larger insulin dose without SU. A summary of published RCTs comparing combinations of SUs and insulin in the management of T2DM is shown in Table 10.
Table 10.
Glycemic efficacy
In a Cochrane systematic review including nine trials (316 patients), insulin-SU combination therapy compared with insulin monotherapy was associated with a reduction of A1C; MD of −1% (95% CI: −1.6 to −0.5; P < 0.01). However, insulin-metformin (−0.9%), insulin-AGIs (−0.4%), and insulin-DPP4I (−0.4%) combinations had revealed a less significant change in A1C compared to insulin-SU combination therapy.[138] Furthermore, a meta-analysis comprising 17 RCTs reported a significantly lower A1C in SU groups (in combination with insulin) compared with placebo (0.46% lower; 95% CI: 0.24, 0.69, I2 = 43.6%).[60] Moreover, another meta-analysis also found favorable outcomes with insulin-SU combination therapy in terms of glycemic control compared to insulin monotherapy (P < 0.0001).[139]
Past studies show that approximately half of the patients attain A1C <7% with dual insulin therapy whereas patients taking insulin alone met goals only one-third of the time.[133,134,140,141,142] Yki-Järvinen et al. determined that a basal insulin regimen containing GLIM decreased total daily insulin requirements by approximately one-third.[143] A combination of once-daily basal insulin with an SU versus monotherapy with premixed insulin has proven to decrease total insulin dose by the same factor.[133,143]
A Korean RCT after 24 weeks of observation period reported a pronounced reduction of A1C with the addition of GLIM to insulin glargine and metformin than insulin glargine plus metformin (0.49% [95% CI: 0.16%–0.82%], P = 0.005).[136]
In “4-T” trial, patients with uncontrolled glycemic level with SUs and metformin were randomized to receive twice-daily biphasic insulin aspart 30 or thrice-daily prandial insulin aspart, or once-daily (twice if required) basal insulin detemir. After 1-year follow-up, the mean reduction in A1C was significantly greater in the biphasic (1.3%) and prandial groups (1.4%) than in the basal group (0.8%).[144] However, after 3 years, basal or prandial insulin-based regimen had better A1C control than biphasic insulin-based regimen.[145]
Safety tolerability and body weight
The combination of insulin and SU results in a reduction of necessary insulin dose per day.[60,138] However, most studies adding SUs to insulin reported milder hypoglycemic episodes versus insulin monotherapy (range: 2.2–6.1 vs. 2.0–2.6 episodes per participant).[138] The addition of SUs into insulin monotherapy resulted in an additional weight gain of 0.4 to 1.9 kg versus −0.8 to 2.1 kg in the insulin monotherapy group.[138] However, no change in body weight was observed by addition of SUs to insulin in several older RCTs.[146,147]
ROLE OF SULFONYLUREAS IN COMBINATION
Cost
Medication cost always plays a significant role in the management of any disease in underdeveloped and developing countries, as it directly affects the drug utilization and patient compliance. Patients without any medical insurance and below the poverty line encounter numerous challenges for the use of expensive medicines in the management of T2DM. A study revealed that the medication cost for diabetes was USD 138 per patient per year which is equivalent to 8.1% of the total budgeted health expenditure for that financial year in sub-Saharan Africa.[148] However, SUs are associated with a significantly lower cost per quality-adjusted-life-years (QALYs) and result in the longest time to insulin dependence.[149] Sensitivity analysis on the medication cost reported the difference in the expected medication cost per QALY from the base-case cost; metformin costs 81.75 USD/month, SU costs 54.85 USD/month, DPP-4I costs 232.84 USD/month, GLP-1 receptor agonist costs 325.97 USD/month, and insulin therapy costs 245.70 USD/month.[149] This cost comparison indicates that SUs should be preferred as an add-on to metformin if the choice of drug is based on cost.[19] Furthermore, FDC users on an average had higher out-of-pocket costs for their prescriptions and had a less restricted heath plan.[150] In summary, lower cost without compromising the glycemic efficacy and tolerability could make SUs the prime choice of treatment of T2DM.
Adherence
Medication compliance is directly related to the cost, availability, dosage regimen, and complications associated with the treatment.[151,152] In the current scenario, FDCs increase the patient compliance by reducing the frequency of drug administration and cost of the medication. Pan F et al. reported that the FDC of metformin-glyburide resulted in 13% increase in patient adherence (P < 0.001) compared to the 2-pill regimen.[150] Similarly, a meta-analysis also reported that FDC decreases the risk of medication noncompliance, improves clinical outcomes, and should be considered in patients with chronic conditions.[153]
Fixed-dose combinations
Combining two or three antihyperglycemic agents with complementary mechanisms of action is a cornerstone of T2DM management.[154] Apart from that, low-dose combination therapy compared to high-dose monotherapy might exhibit lesser side effects and can achieve similar or better glycemic control.[155] Furthermore, FDCs reduce number, frequency, and flexibility of dosage administration and minimize the treatment complexity, thereby improving patient adherence.[156] In a meta-analysis, use of FDCs with antihyperglycemic agents was associated with lower A1C and higher mean possession ratio values compared to dual therapies in patients with T2DM.[157] Furthermore, a systematic review suggested that T2DM patients treated with FDC therapy may have better adherence, improved satisfaction, and lower direct medical costs, compared to those treated with loose pill combination therapy.[158] A list of available SUs either individually or in FDCs with their strength, dose, and required dose titration is summarized in Table 11.
Table 11.
SPECIAL SITUATIONS
Elderly
SUs are usually well tolerated; however, mild hypoglycemia has been reported with long-acting SUs (e.g., CHOL, glyburide,).[128] For older patients who have persistent hyperglycemia with lifestyle intervention and metformin, the addition of a short-acting SU such as GLIP is a preferred option. A double-blind randomized and open-label comparative study after 2 years of treatment concluded that GLIC MR in combination with other OADs significantly improved glycemic control in the elderly and renal-impaired T2DM patients with a very good safety profile.[159] A recent review evaluated the comparative safety and efficacy of commonly available SUs (GLIB, GLIC, GLIM, and GLIP) for the management of T2DM in older patients.[160] The study reported that GLIC can be used in older patients due to its low risk of hypoglycemia; however, it is suggested to restrict the use of GLIB in such populations. Furthermore, several guidelines including IDF and Canadian guidelines also recommend modern SUs (GLIC MR and GLIM) as a drug of choice for elder T2DM patients.[161,162] In GENERATION trial, a similar proportion of patients achieved the primary end point of A1C <7.0% at week 52 without confirmed/severe hypoglycemia with saxagliptin compared to GLIM when added to metformin (37.9% vs. 38.2%; OR: 0.99, 95% CI: 0.73, 1.34; P = 0.9415). However, it is noteworthy that saxagliptin was numerically (but not significantly) inferior for patients aged ≥75 years (35.9% vs. 45.5%).[110]
Children and adolescents
The evidence on the use of SUs in children is limited.[163] A 26-week, single-blind, active-controlled study, conducted in 285 pediatric patients with T2DM, found that GLIM was as effective as metformin in A1C reduction (−0.54%, P = 0.001 and −0.71%, P = 0.0002, respectively) with similar rates of hypoglycemia (4.9% vs. 4.2%, respectively).[164] Furthermore, a prospective trial evaluated the effect of SUs (mainly CHOL) in patients with Maturity-Onset Diabetes of the Young (MODY) patients with 9–29 years of age and found that long-term administration of SUs significantly enhanced glucose-induced insulin concentrations by approximately 68%.[165] Evidence and guidelines suggested that SUs are the drug of choice for the treatment of MODY.[166,167,168] Neonatal diabetes mellitus (NDM) is generally caused by several genetic abnormalities and might be either permanent or transient. In patients with NDM, SUs facilitate insulin secretion through the hormone GLP-1 pathway in response to food present in the gut. Evidence suggests that patients with permanent or transient NDM due to mutations in KCNJ11 or ABCC8 gene were successfully treated with SU (GLIB) therapy rather than insulin.[169,170,171,172] An observational case study reported that young children with neonatal diabetes even with blood glucose level of 350 mg/dL also responded to a usual dose of SU.[173]
Pregnancy and lactation
Second-generation SUs such as GLIB despite low molecular weight do not cross the placenta in significant amounts.[174] This phenomenon may be attributed either to its high protein-binding capacity (99.8%) or reverting the total GLIB content into the maternal system by an unidentified placental transport system. Furthermore, the absence of fetal adverse effects such as malformations and hypoglycemia makes GLIB an acceptable treatment option for patients with T2DM during pregnancy.[174,175] In a meta-analysis including one RCT from India, the outcomes of GLIB, metformin, and insulin were assessed. The study reported similar rate of cesarean delivery (range: 23%–52%) and infant birth weights (MD: −93 g, 95% CI: −191 to 5 g), with no significant difference in maternal glycemic control between the treatments.[176] Furthermore, a systemic review and meta-analysis reported no significant difference in terms of glycemic control or pregnancy outcomes between GLIB and insulin when used during pregnancy.[177] Moreover, in a meta-analysis, no significant difference was observed in the rate of neonatal major abnormality or neonatal death among women treated with GLIB-metformin compared with nonexposed women during first trimester; however, the study was limited by heterogeneity.[178] Nonetheless, long-term efficacy studies of GLIB during pregnancy and lactation are limited.[175] GLIB compared to metformin was more effective in lowering blood glucose in women with gestational diabetes with a lower treatment failure rate.[175] Moreover, there are some contradicting results from other studies and the paucity of data regarding the usage of SU combination therapy available during pregnancy and lactation.
Patients with comorbidity
Patients with diabetes are always at an increased risk of developing CVD. The risk of stroke, heart disease, and death due to heart disease is 2 folds high in patients with diabetes than in those without diabetes. Newer-generation SUs such as GLIM and GLIC MR reduce CV risk and may prevent protective ischemic cardiac preconditioning after MI.[179] In the ADVANCE trial, intensive glucose control with a GLIC-containing regimen has been shown to reduce the incidence of combined major macro- and microvascular events (18.1% vs. 20.0% with standard control; HR: 0.90; 95% CI: 0.82–0.98; P = 0.01), as well as that of major microvascular events (9.4% vs. 10.9%; HR: 0.86; 95% CI: 0.77–0.97; P = 0.01).[17] Similarly, the Veterans Affairs Diabetes Trial including 1791 military veterans (median follow-up: 5.6 years) reported that GLIM-based intensive regimen was associated with a 17% relative risk reduction in the rate of CV events compared to standard therapy.[13] Moreover, a recent meta-analysis demonstrates that SUs are not associated with increased risk for all-cause mortality, CV mortality, MI, or stroke.[38]
SUs such as GLIP and GLIC are mainly excreted as unchanged drug or inactive metabolite. Therefore, they may produce less hypoglycemia in patients with renal impairment. GLIM has been reported to be safe and effective in diabetic patients with renal impairment.[180,181] However, GLIB may aggravate the risk of hypoglycemia in such populations.[182] Short-acting SUs such as GLIC and GLIP can be used with proper dose adjustment and monitoring in patients with renal insufficiency.[5] An RCT reported that in patients with chronic renal insufficiency, sitagliptin and GLIP provided similar A1C-lowering efficacy, and GLIP produced more hypoglycemia and weight gain than sitagliptin.[183] Furthermore, SUs should be cautiously used in patients with liver diseases, as most of the SUs are inactivated in the liver, they might accumulate in the body during liver dysfunction, and ultimately cause hypoglycemia. Moreover, in hypoalbuminemia, the concentration of free drug will increase and may produce hypoglycemia.[5,184] SUs with a short half-life such as GLIP or GLIB are preferred in liver disease patients. Patients with decompensated cirrhosis, i.e., encephalopathy, ascites, or coagulopathy, may have a reduced ability to counteract hypoglycemia, and thus, the response to therapy should be monitored closely.[185]
Diabetes and Ramadan
Patients who are continuing antidiabetic medications and undergoing fasting during Ramadan may encounter several difficulties such as hypoglycemia and other health-related complications.[5] Due to the increased risk of hypoglycemia, individuals with SUs should be careful during fasting period; however, some modern SUs (GLIC, GLIM, and GLIP) are associated with lower risk of hypoglycemia compared to GLIB.[96] The recent IDF-DAR practical guideline advocates that patients continuing second-generation SUs can fast safely during Ramadan.[186] The Glimepiride in Ramadan study reported that the efficacy and safety of GLIM have remained unchanged during the period of Ramadan in patients with T2DM.[187] In the STEADFAST study, there was no significant difference reported in hypoglycemic episodes between vildagliptin and GLIC (both add-ons to metformin) (P = 0.039).[188] Similarly, in Treat 4 Ramadan Trial, liraglutide compared to SUs (both add-ons to metformin) did not reveal any episodes of severe hypoglycemia in either group.[189] Table 12 summarizes the efficacy and safety of SU combination therapy during Ramadan. Incretin-based therapies such as DPP-4I and GLP-1 receptor agonist work by increasing insulin secretion in a glucose-dependent manner; therefore, they may not produce any risk of hypoglycemia. However, they may amplify the hypoglycemic effect of SUs when used in combination.[193] Studies conducted during Ramadan found higher hypoglycemic episodes with SUs compared to DPP-4I; however, subgroup analysis found GLIC having least hypoglycemic episodes among all SUs, almost similar to DPP-4I.[36,192]
Table 12.
Diabetes and other fasting
Fasting is a universal religiocultural tradition observed in varying forms, in different religions across the world. Hindus observe fasting during Navaratri, Karva Chauth, and Guru Purnima; Buddhists observe during Lent; Jains observe in the occasion of Paryushana.[194,195,196] In the diabeto-centric viewpoint, patients taking antidiabetic agents should be monitored regularly for the event of hypoglycemia and other associated complications. Furthermore, the antidiabetic dose should be modified or omitted during the fasting period in order to prevent future complications. The dose modification or omission should be tailored according to the type of medication, food plan, and patient characteristics. Long-acting SUs such as GLIB should not be used; however, modern SUs with low tendency of hypoglycemia such as GLIM and GLIC MR can be considered with dose reduction.[194,195,196] In addition, patients with diabetes and who are undergoing fasting should be educated properly for the daily blood glucose monitoring and about hypoglycemic symptoms in order to maintain a safe fasting during rituals.
TRANSFORMING EVIDENCE INTO CLINICAL PRACTICE
Patient selection
Selection of patient plays a vital role in optimizing the SU combination therapy. Combination therapies should be introduced early in all patients with T2DM for preserving β-cell functions. The initiation of combination therapy depends on the individual patients’ A1C at entry level. Modern SUs can be used in both obese and lean patients owing to the low risk of weight gain.[5] SUs should be cautiously used in the elderly patients and in patients at high risk of hypoglycemia.[32,34] In addition, patients with T2DM using SU combination therapy who wish to go for fasting should be closely monitored during this period to avoid further complications.[5]
Drug selection
All SUs are not similar in terms of their efficacies, adverse effects, and tolerability. Clinical factors such as levels of fasting, postprandial hyperglycemia, comorbid hypertension or other CVDs, and hepatic or renal dysfunction determine the selection of SU combination therapy.[5] Shortacting SUs should be preferred in the case of postprandial hyperglycemia; however, twice-daily SU in combination with metformin should be preferred in fasting hyperglycemia.[197,198,199] Combining two different SUs is not logical as they have a similar mechanism of action but SUs may be combined with other OADs such as metformin and TZD (with the complementary mechanism of actions), and even with insulin.[5] Patients taking a combination of SUs with incretin-based therapy should be under vigilant monitoring due to the risk of hypoglycemia.[193] FDCs containing SUs should be preferred over combination regimens.
Dose selection
Dose adjustment or modification is inevitable in T2DM patients with the comorbid conditions. Dose selection for FDCs containing SUs should be done as per individual patient characteristics and properties of SUs.[5] The South Asian Federation of Endocrine Societies consensus recommends initiation of SUs at low doses and uptitrating them gradually based on the glycemic responses to prevent hypoglycemia, but when hypoglycemic episodes are confirmed, reduction of dose should be considered.[113] The consensus also recommends educating the patients and family members about the signs and symptoms of hypoglycemia.[5] When using combination therapy containing SUs and incretin-based therapies, the dose of SUs should be reduced in the elderly and/or patients with renal insufficiency.
Diabetes education and patient and physician empowerment
Due to chronic nature of the disease, diabetes necessitates self-management plan in day-to-day basis.[200] Diabetic education enables the patients to make informed decisions and effectively manage the disease without any complications. Diabetic education, which gives information on physical activity, glucose monitoring, diet, hypoglycemia, dosage and timing of medications, and identification of the symptoms of complications, should be provided to patients and their families. Patients should be encouraged and supported to become active partners in the decision-making process, to set realistic goals, select appropriate management strategies, enhance adherence, and improve treatment outcomes.[5] Self-monitoring of blood glucose (SMBG) at home and self-down titration of doses in case of hypoglycemia by patients are recommended. The patient should be trained for the safe use of FDC containing SUs and should be able to detect the hypoglycemic complications. Furthermore, patients along with their family members should be educated about the usage of SMBG system. When FDCs containing SUs are prescribed, physicians should consider the clinical profile of the patient and implement strategies that will not only help to minimize patient's concerns over SUs treatment but also empower them for self-management. At each visit, physicians should look into symptoms suggestive of hypoglycemia and adjust doses accordingly when risks outweigh glycemic benefits.
CONCLUSION
Intensification of diabetic therapy with a proactive approach is crucial to achieve target glycemic levels. SUs are an important component of pharmacological armamentarium in the treatment of T2DM. Owing to their well-established efficacy and safety profile, they are the most commonly recommended class of agents when glycemic targets are not achieved with metformin alone. Owing to their disparity in pharmacokinetic and pharmacodynamic profile, which results in variability in safety and tolerability, the selection of SUs should be highly individualized with careful monitoring in high-risk patients. Due to their unique characteristics such as increased efficacy, CV safety, fewer episodes of hypoglycemia, and weight-neutralizing effects, modern SUs should be used over conventional SUs in the management of T2DM. Lower treatment cost without compromising the glycemic efficacy and tolerability together with good compliance rate positions SUs as the front-line agent in the management of T2DM. In addition, the complementary mechanisms of action of GLIM and metformin contribute to their enhanced glycemic effects. Appropriate patient selection, suitable drug and dose, along with proper patient and physician education may help in the effective and vigilant use of SUs in the management of T2DM.
Financial support and sponsorship
This study was funded by Sanofi Diabetes India.
Conflicts of interest
There are no conflicts of interest.
Acknowledgment
The authors acknowledge Sanofi INDIA for supporting this initiative with an academic grant and Rabi Narayan Panigrahy, Syam Kumar Yelamanchi, and Leena Patel from Jeevan Scientific Technology Limited (Hyderabad, India) for writing/editorial assistance in the development of this manuscript.
REFERENCES
- 1.International Diabetes Federation. The International Federation (IDF) Diabetes Atlas. 7th ed. Belgium: International Diabetes Federation; 2015. [Last accessed on 2017 Jan 25]. ISBN: 978-2-930229-81-2. Available from: http://www.diabetesatlas.org/ [Google Scholar]
- 2.Global Report on Diabetes – World Health Organization. 2016. [Last accessed on 2017 Jan 25]. Available from: http://www.apps.who.int/iris/bitstream/10665/204871/1/9789241565257_eng.pdf .
- 3.Stratton IM, Adler AI, Neil HA, Matthews DR, Manley SE, Cull CA, et al. Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (UKPDS 35): Prospective observational study. BMJ. 2000;321:405–12. doi: 10.1136/bmj.321.7258.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Agarwal AA, Jadhav PR, Deshmukh YA. Prescribing pattern and efficacy of anti-diabetic drugs in maintaining optimal glycemic levels in diabetic patients. J Basic Clin Pharm. 2014;5:79–83. doi: 10.4103/0976-0105.139731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kalra S, Aamir AH, Raza A, Das AK, Azad Khan AK, Shrestha D, et al. Place of sulfonylureas in the management of type 2 diabetes mellitus in south Asia: A consensus statement. Indian J Endocrinol Metab. 2015;19:577–96. doi: 10.4103/2230-8210.163171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Truter I. An investigation into antidiabetic medication prescribing in South Africa. J Clin Pharm Ther. 1998;23:417–22. doi: 10.1046/j.1365-2710.1998.00182.x. [DOI] [PubMed] [Google Scholar]
- 7.Chiang CW, Chiu HF, Chen CY, Wu HL, Yang CY. Trends in the use of oral antidiabetic drugs by outpatients in Taiwan: 1997-2003. J Clin Pharm Ther. 2006;31:73–82. doi: 10.1111/j.1365-2710.2006.00705.x. [DOI] [PubMed] [Google Scholar]
- 8.Principal JK. A study on drug utilization of oral hypoglycemic agents in type-2 diabetic patients. Asian J Pharm Clin Res. 2011;4:60–4. [Google Scholar]
- 9.Mandal S, Maiti T, Das AK, Das A, Mandal A, Sarkar BS, et al. Drug utilization study in patients with type 2 diabetes mellitus attending diabetes clinic of a tertiary care hospital in rural Bengal. Int J Basic Clin Pharmacol. 2016;5:1647–54. [Google Scholar]
- 10.National List of Essential Medicines. India: 2015. [Last accessed on 2017 Mar 21]. Available from: http://www.apps.who.int/medicinedocs/en/d/Js23088en/ [Google Scholar]
- 11.Mkele G. What's the latest on sulfonylureas in the management of type 2 diabetes? South Afr Fam Pract. 2013;55:501–3. [Google Scholar]
- 12.Cahn A, Cefalu WT. Clinical considerations for use of initial combination therapy in type 2 diabetes. Diabetes Care. 2016;39(Suppl 2):S137–45. doi: 10.2337/dcS15-3007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hayward RA, Reaven PD, Wiitala WL, Bahn GD, Reda DJ, Ge L, et al. Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2015;372:2197–206. doi: 10.1056/NEJMoa1414266. [DOI] [PubMed] [Google Scholar]
- 14.Phung OJ, Sobieraj DM, Engel SS, Rajpathak SN. Early combination therapy for the treatment of type 2 diabetes mellitus: Systematic review and meta-analysis. Diabetes Obes Metab. 2014;16:410–7. doi: 10.1111/dom.12233. [DOI] [PubMed] [Google Scholar]
- 15.Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359:1577–89. doi: 10.1056/NEJMoa0806470. [DOI] [PubMed] [Google Scholar]
- 16.Derosa G, Sibilla S. Optimizing combination treatment in the management of type 2 diabetes. Vasc Health Risk Manag. 2007;3:665–71. [PMC free article] [PubMed] [Google Scholar]
- 17.ADVANCE Collaborative Group. Patel A, MacMahon S, Chalmers J, Neal B, Billot L, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med. 2008;358:2560–72. doi: 10.1056/NEJMoa0802987. [DOI] [PubMed] [Google Scholar]
- 18.Del Prato S, Felton AM, Munro N, Nesto R, Zimmet P, Zinman B, et al. Improving glucose management: Ten steps to get more patients with type 2 diabetes to glycaemic goal. Int J Clin Pract. 2005;59:1345–55. doi: 10.1111/j.1742-1241.2005.00674.x. [DOI] [PubMed] [Google Scholar]
- 19.Abrahamson MJ. Should sulfonylureas remain an acceptable first-line add-on to metformin therapy in patients with type 2 diabetes? Yes, they continue to serve us well!iabetes Care. 2015;38:166–9. doi: 10.2337/dc14-1945. [DOI] [PubMed] [Google Scholar]
- 20.Handelsman Y, Bloomgarden ZT, Grunberger G, Umpierrez G, Zimmerman RS, Bailey TS, et al. American Association of clinical endocrinologists and American College of endocrinology – Clinical practice guidelines for developing a diabetes mellitus comprehensive care plan-2015. Endocr Pract. 2015;21(Suppl 1):1–87. doi: 10.4158/EP15672.GL. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Aquilante CL. Sulfonylurea pharmacogenomics in type 2 diabetes: The influence of drug target and diabetes risk polymorphisms. Expert Rev Cardiovasc Ther. 2010;8:359–72. doi: 10.1586/erc.09.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.White JR., Jr A brief history of the development of diabetes medications. Diabetes Spectr. 2014;27:82–6. doi: 10.2337/diaspect.27.2.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Quianzon CC, Cheikh IE. History of current non-insulin medications for diabetes mellitus. J Community Hosp Intern Med Perspect. 2012;2:1–4. doi: 10.3402/jchimp.v2i3.19081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sarkar A, Tiwari A, Bhasin PS, Mitra M. Pharmacological and pharmaceutical profile of gliclazide: A review. J Appl Pharm Sci. 2011;1:11–9. [Google Scholar]
- 25.Oderda G, Richards K, Turpin S. Sulfonylurea Agents & Combination Products Drug Class Review. University of Utah College of Pharmacy. 2013 Jul; Final Report. [Google Scholar]
- 26.Lexi-Comp Inc. Drug Information Handbook. 21st ed. Hudson, OH: Lexi-Comp Inc; 2013. [Google Scholar]
- 27.Ballagi-Pordány G, Németh M, Aranyi Z, Kékesi E, Koltai MZ, Papp G, et al. Effect of glimepiride on the electrical activity of isolated rabbit heart muscle. Arzneimittelforschung. 1992;42:111–3. [PubMed] [Google Scholar]
- 28.Nakamura I, Oyama J, Komoda H, Shiraki A, Sakamoto Y, Taguchi I, et al. Possible effects of glimepiride beyond glycemic control in patients with type 2 diabetes: A preliminary report. Cardiovasc Diabetol. 2014;13:15. doi: 10.1186/1475-2840-13-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ashcroft FM, Gribble FM. Tissue-specific effects of sulfonylureas: Lessons from studies of cloned K(ATP) channels. J Diabetes Complications. 2000;14:192–6. doi: 10.1016/s1056-8727(00)00081-7. [DOI] [PubMed] [Google Scholar]
- 30.Gribble FM, Ashcroft FM. Sulfonylurea sensitivity of adenosine triphosphate-sensitive potassium channels from beta cells and extrapancreatic tissues. Metabolism. 2000;49:3–6. [PubMed] [Google Scholar]
- 31.Vila-Carriles WH, Zhao G, Bryan J. Defining a binding pocket for sulfonylureas in ATP-sensitive potassium channels. FASEB J. 2007;21:18–25. doi: 10.1096/fj.06-6730hyp. [DOI] [PubMed] [Google Scholar]
- 32.Thulé PM, Umpierrez G. Sulfonylureas: A new look at old therapy. Curr Diab Rep. 2014;14:473. doi: 10.1007/s11892-014-0473-5. [DOI] [PubMed] [Google Scholar]
- 33.Kramer W, Müller G, Girbig F, Gutjahr U, Kowalewski S, Hartz D, et al. Differential interaction of glimepiride and glibenclamide with the beta-cell sulfonylurea receptor. II. Photoaffinity labeling of a kDa protein by [3H] glimepiride glimepiride. Biochim Biophys Acta. 1994;1191:278–90. doi: 10.1016/0005-2736(94)90178-3. [DOI] [PubMed] [Google Scholar]
- 34.Briscoe VJ, Griffith ML, Davis SN. The role of glimepiride in the treatment of type 2 diabetes mellitus. Expert Opin Drug Metab Toxicol. 2010;6:225–35. doi: 10.1517/17425250903512955. [DOI] [PubMed] [Google Scholar]
- 35.Holstein A, Plaschke A, Egberts EH. Lower incidence of severe hypoglycaemia in patients with type 2 diabetes treated with glimepiride versus glibenclamide. Diabetes Metab Res Rev. 2001;17:467–73. doi: 10.1002/dmrr.235. [DOI] [PubMed] [Google Scholar]
- 36.Singh AK, Singh R. Is gliclazide a sulfonylurea with difference? A review in 2016. Expert Rev Clin Pharmacol. 2016;9:839–51. doi: 10.1586/17512433.2016.1159512. [DOI] [PubMed] [Google Scholar]
- 37.Halimi S, Schweizer A, Minic B, Foley J, Dejager S. Combination treatment in the management of type 2 diabetes: Focus on vildagliptin and metformin as a single tablet. Vasc Health Risk Manag. 2008;4:481–92. doi: 10.2147/vhrm.s2503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rados DV, Pinto LC, Remonti LR, Leitão CB, Gross JL. Correction: The association between sulfonylurea use and all-cause and cardiovascular mortality: A Meta-analysis with trial sequential analysis of randomized clinical trials. PLoS Med. 2016;13:e1002091. doi: 10.1371/journal.pmed.1002091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Basit A, Riaz M, Fawwad A. Glimepiride: Evidence-based facts, trends, and observations (GIFTS). [corrected] Vasc Health Risk Manag. 2012;8:463–72. doi: 10.2147/HIV.S33194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Draeger KE, Wernicke-Panten K, Lomp HJ, Schüler E, Rosskamp R. Long-term treatment of type 2 diabetic patients with the new oral antidiabetic agent glimepiride (Amaryl): A double-blind comparison with glibenclamide. Horm Metab Res. 1996;28:419–25. doi: 10.1055/s-2007-979830. [DOI] [PubMed] [Google Scholar]
- 41.Garber AJ, Abrahamson MJ, Barzilay JI, Blonde L, Bloomgarden ZT, Bush MA, et al. Consensus statement by the American association of clinical endocrinologists and American college of endocrinology on the comprehensive type 2 diabetes management algorithm-2017 executive summary. Endocr Pract. 2017;23:207–38. doi: 10.4158/EP161682.CS. [DOI] [PubMed] [Google Scholar]
- 42.American Diabetes Association. Standards of Medical Care in Diabetes. 2017. [Last accessed on 2017 Feb 24]. Available from: http://www.professional.diabetes.org/sites/professional.diabetes.org/files/media/dc_40_s1_final.pdf .
- 43.Ministry of Health, Kingdom of Bahrain. Guidelines for Management of Type 2 Diabetes Mellitus in Primary Care Settings and Outpatient Clinics in the Kingdom of Bahrain. [Last accessed on 2017 Mar 08]. Available from: http://www.nhra.bh/files/files/CEO/Hypertension%20Guideline%2031052015.pdf .
- 44.Chinenye S, Ofoegbu EN, Onyemelukwe GC, Uloko AE, Ogbera AO. Clinical Practice Guidelines for Diabetes Management in Nigeria. Diabetes Association of Nigeria (DAN) 2013 [Google Scholar]
- 45.Rutten GE, De Grauw WJ, Nijpels G, Houweling ST, Van de Laar FA, Bilo HJ, et al. Diabetes guideline of the Dutch College of General Practitioners (NHG-Standard Diabetes mellitus type 2 (derde herziening)) Huisarts Wet. 2013;56:512–25. [Google Scholar]
- 46.Diabetes Mellitus Guidelines, WHO EMRO. 2006. [Last accessed on 2017 Apr 06]. Available from: http://www.applications.emro.who.int/dsaf/dsa664.pdf .
- 47.National Clinical Guidelines for Management of Diabetes Mellitus; Republic of Kenya Ministry. 2010 [Google Scholar]
- 48.International Diabetes Federation Guideline Development Group. Global guideline for type 2 diabetes. Diabetes Res Clin Pract. 2014;104:1–52. doi: 10.1016/j.diabres.2012.10.001. [DOI] [PubMed] [Google Scholar]
- 49.Type 2 Diabetes in Adults: Management, NICE Guideline [NG28] 2015. [Last accessed on 2017 Mar 14]. Available from: https://www.nice.org.uk/guidance/ng28?unlid=739233160201610316820 .
- 50.Madhu SV, Saboo B, Makkar BM, Reddy GC, Jana J, Panda JK, et al. RSSDI clinical practice recommendations for management of type 2 diabetes mellitus, 2015. Int J Diabetes Dev Ctries. 2015;35:1–71. [Google Scholar]
- 51.Mishra S, Ray S, Dalal JJ, Sawhney JP, Ramakrishnan S, Nair T, et al. Management protocols of stable coronary artery disease in India: Executive summary. Indian Heart J. 2016;68:868–73. doi: 10.1016/j.ihj.2016.11.318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.The Society for Endocrinology, Metabolism and Diabetes of South Africa Type 2 Diabetes Guidelines Expert Committee. The 2017 SEMDSA Guideline for the Management of Type 2 Diabetes Guideline Committee. JEMDSA. 2017;21(Supplement 1):S1–S196. [Google Scholar]
- 53.The United Republic of Tanzania, Standard Treatment Guidelines and Essential Medicines List; Ministry of Health and Social Welfare. (4th ed) 2013 May; [Google Scholar]
- 54.National Diabetes Guidelines, United Arab Emirates. 2009. [Last accessed on 2017 Mar 08]. Available from: cms.wounds-uk.com/media/NationalDiabetesGuidelinesUAE.pdf .
- 55.The Republic of Uganda, Ministry of Health, National Guidelines for Management of Common Conditions; Uganda Clinical Guidelines. 2016 [Google Scholar]
- 56.Hundal RS, Krssak M, Dufour S, Laurent D, Lebon V, Chandramouli V, et al. Mechanism by which metformin reduces glucose production in type 2 diabetes. Diabetes. 2000;49:2063–9. doi: 10.2337/diabetes.49.12.2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Del Prato S, Vigili de Kreutzenberg S, Riccio A, Tiengo A. Hepatic sensitivity to insulin: Effects of sulfonylurea drugs. Am J Med. 1991:29S–36S. doi: 10.1016/0002-9343(91)90415-t. [DOI] [PubMed] [Google Scholar]
- 58.Ramracheya R, Ward C, Shigeto M, Walker JN, Amisten S, Zhang Q, et al. Membrane potential-dependent inactivation of voltage-gated ion channels in alpha-cells inhibits glucagon secretion from human islets. Diabetes. 2010;59:2198–208. doi: 10.2337/db09-1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Bolen S, Tseng E, Hutfless S, Segal JB, Suarez-Cuervo C, Berger Z, et al. Diabetes Medications for Adults with Type 2 Diabetes: An Update. Rockville (MD): Agency for Healthcare Research and Quality (US); 2016. [Last accessed on 2017 Jan 12]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK362863/ [PubMed] [Google Scholar]
- 60.Hirst JA, Farmer AJ, Dyar A, Lung TW, Stevens RJ. Estimating the effect of sulfonylurea on hbA1c in diabetes: A systematic review and meta-analysis. Diabetologia. 2013;56:973–84. doi: 10.1007/s00125-013-2856-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ahrén B, Johnson SL, Stewart M, Cirkel DT, Yang F, Perry C, et al. HARMONY 3: 104-week randomized, double-blind, placebo- and active-controlled trial assessing the efficacy and safety of albiglutide compared with placebo, sitagliptin, and glimepiride in patients with type 2 diabetes taking metformin. Diabetes Care. 2014;37:2141–8. doi: 10.2337/dc14-0024. [DOI] [PubMed] [Google Scholar]
- 62.Kim HS, Kim DM, Cha BS, Park TS, Kim KA, Kim DL, et al. Efficacy of glimepiride/metformin fixed-dose combination vs. metformin uptitration in type 2 diabetic patients inadequately controlled on low-dose metformin monotherapy: A randomized, open label, parallel group, multicenter study in Korea. J Diabetes Investig. 2014;5:701–8. doi: 10.1111/jdi.12201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kalra S, Das AK. Epidemiologic surveillance of glycemic response to a scored, breakable, extended release, fixed dose combination of gliclazide and metformin in persons with type 2 diabetes. J Assoc Physicians India. 2017;65:38–41. [PubMed] [Google Scholar]
- 64.Charpentier G, Fleury F, Kabir M, Vaur L, Halimi S. Improved glycaemic control by addition of glimepiride to metformin monotherapy in type 2 diabetic patients. Diabet Med. 2001;18:828–34. doi: 10.1046/j.1464-5491.2001.00582.x. [DOI] [PubMed] [Google Scholar]
- 65.González-Ortiz M, Martínez-Abundis E. Grupo para el Tratamiento de la Diabetes Mellitus con Combinaciones. Efficacy and safety of glimepiride plus metformin in a single presentation, as combined therapy, in patients with type 2 diabetes mellitus and secondary failure to glibenclamide, as monotherapy. Rev Invest Clin. 2004;56:327–33. [PubMed] [Google Scholar]
- 66.Ristic S, Collober-Maugeais C, Cressier F, Tang P, Pecher E. Nateglinide or gliclazide in combination with metformin for treatment of patients with type 2 diabetes mellitus inadequately controlled on maximum doses of metformin alone: 1-year trial results. Diabetes Obes Metab. 2007;9:506–11. doi: 10.1111/j.1463-1326.2006.00632.x. [DOI] [PubMed] [Google Scholar]
- 67.Goldstein BJ, Pans M, Rubin CJ. Multicenter, randomized, double-masked, parallel-group assessment of simultaneous glipizide/metformin as second-line pharmacologic treatment for patients with type 2 diabetes mellitus that is inadequately controlled by a sulfonylurea. Clin Ther. 2003;25:890–903. doi: 10.1016/s0149-2918(03)80112-1. [DOI] [PubMed] [Google Scholar]
- 68.Feinglos M, Dailey G, Cefalu W, Osei K, Tayek J, Canovatchel W, et al. Effect on glycemic control of the addition of 2.5 mg glipizide GITS to metformin in patients with T2DM. Diabetes Res Clin Pract. 2005;68:167–75. doi: 10.1016/j.diabres.2004.09.002. [DOI] [PubMed] [Google Scholar]
- 69.Marre M, Howlett H, Lehert P, Allavoine T. Improved glycaemic control with metformin-glibenclamide combined tablet therapy (Glucovance) in type 2 diabetic patients inadequately controlled on metformin. Diabet Med. 2002;19:673–80. doi: 10.1046/j.1464-5491.2002.00774.x. [DOI] [PubMed] [Google Scholar]
- 70.Chien HH, Chang CT, Chu NF, Hsieh SH, Huang YY, Lee IT, et al. Effect of glyburide-metformin combination tablet in patients with type 2 diabetes. J Chin Med Assoc. 2007;70:473–80. doi: 10.1016/S1726-4901(08)70044-3. [DOI] [PubMed] [Google Scholar]
- 71.Ray JA, Huet D, Valentine WJ, Palmer AJ, Cugnardey N, Renaudin C, et al. Long-term costs and clinical outcomes associated with metformin-glibenclamide combination tablets (Glucovance®) in patients with type 2 diabetes sub-optimally controlled by metformin: A modelling study in the French setting. Br J Diabetes Vasc Dis. 2008;8:39–44. [Google Scholar]
- 72.Umpierrez G, Issa M, Vlajnic A. Glimepiride versus pioglitazone combination therapy in subjects with type 2 diabetes inadequately controlled on metformin monotherapy: Results of a randomized clinical trial. Curr Med Res Opin. 2006;22:751–9. doi: 10.1185/030079906X104786. [DOI] [PubMed] [Google Scholar]
- 73.Lee YK, Song SO, Kim KJ, Cho Y, Choi Y, Yun Y, et al. Glycemic effectiveness of metformin-based dual-combination therapies with sulphonylurea, pioglitazone, or DPP4-inhibitor in drug-naïve Korean type 2 diabetic patients. Diabetes Metab J. 2013;37:465–74. doi: 10.4093/dmj.2013.37.6.465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Matthews DR, Charbonnel BH, Hanefeld M, Brunetti P, Schernthaner G. Long-term therapy with addition of pioglitazone to metformin compared with the addition of gliclazide to metformin in patients with type 2 diabetes: A randomized, comparative study. Diabetes Metab Res Rev. 2005;21:167–74. doi: 10.1002/dmrr.478. [DOI] [PubMed] [Google Scholar]
- 75.Mishriky BM, Cummings DM, Tanenberg RJ. The efficacy and safety of DPP4 inhibitors compared to sulfonylureas as add-on therapy to metformin in patients with type 2 diabetes: A systematic review and meta-analysis. Diabetes Res Clin Pract. 2015;109:378–88. doi: 10.1016/j.diabres.2015.05.025. [DOI] [PubMed] [Google Scholar]
- 76.Zhou JB, Bai L, Wang Y, Yang JK. The benefits and risks of DPP4-inhibitors vs. sulfonylureas for patients with type 2 diabetes: Accumulated evidence from randomised controlled trial. Int J Clin Pract. 2016;70:132–41. doi: 10.1111/ijcp.12761. [DOI] [PubMed] [Google Scholar]
- 77.Amate JM, Lopez-Cuadrado T, Almendro N, Bouza C, Saz-Parkinson Z, Rivas-Ruiz R, et al. Effectiveness and safety of glimepiride and iDPP4, associated with metformin in second line pharmacotherapy of type 2 diabetes mellitus: Systematic review and meta-analysis. Int J Clin Pract. 2015;69:292–304. doi: 10.1111/ijcp.12605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Filozof C, Gautier JF. A comparison of efficacy and safety of vildagliptin and gliclazide in combination with metformin in patients with type 2 diabetes inadequately controlled with metformin alone: A 52-week, randomized study. Diabet Med. 2010;27:318–26. doi: 10.1111/j.1464-5491.2010.02938.x. [DOI] [PubMed] [Google Scholar]
- 79.Göke B, Gallwitz B, Eriksson J, Hellqvist A, Gause-Nilsson I D1680C00001 Investigators. Saxagliptin is non-inferior to glipizide in patients with type 2 diabetes mellitus inadequately controlled on metformin alone: A 52-week randomised controlled trial. Int J Clin Pract. 2010;64:1619–31. doi: 10.1111/j.1742-1241.2010.02510.x. [DOI] [PubMed] [Google Scholar]
- 80.Thomsen RW, Baggesen LM, Søgaard M, Pedersen L, Nørrelund H, Buhl ES, et al. Early glycaemic control in metformin users receiving their first add-on therapy: A population-based study of 4,734 people with type 2 diabetes. Diabetologia. 2015;58:2247–53. doi: 10.1007/s00125-015-3698-1. [DOI] [PubMed] [Google Scholar]
- 81.Ridderstråle M, Andersen KR, Zeller C, Kim G, Woerle HJ, Broedl UC, et al. Comparison of empagliflozin and glimepiride as add-on to metformin in patients with type 2 diabetes: A 104-week randomised, active-controlled, double-blind, phase 3 trial. Lancet Diabetes Endocrinol. 2014;2:691–700. doi: 10.1016/S2213-8587(14)70120-2. [DOI] [PubMed] [Google Scholar]
- 82.Cefalu WT, Leiter LA, Yoon KH, Arias P, Niskanen L, Xie J, et al. Efficacy and safety of canagliflozin versus glimepiride in patients with type 2 diabetes inadequately controlled with metformin (CANTATA-SU): 52 week results from a randomised, double-blind, phase 3 non-inferiority trial. Lancet. 2013;382:941–50. doi: 10.1016/S0140-6736(13)60683-2. [DOI] [PubMed] [Google Scholar]
- 83.Nauck MA, Del Prato S, Meier JJ, Durán-García S, Rohwedder K, Elze M, et al. Dapagliflozin versus glipizide as add-on therapy in patients with type 2 diabetes who have inadequate glycemic control with metformin: A randomized, 52-week, double-blind, active-controlled noninferiority trial. Diabetes Care. 2011;34:2015–22. doi: 10.2337/dc11-0606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Nauck M, Frid A, Hermansen K, Thomsen AB, During M, Shah N, et al. Long-term efficacy and safety comparison of liraglutide, glimepiride and placebo, all in combination with metformin in type 2 diabetes: 2-year results from the LEAD-2 study. Diabetes Obes Metab. 2013;15:204–12. doi: 10.1111/dom.12012. [DOI] [PubMed] [Google Scholar]
- 85.Yang W, Chen L, Ji Q, Liu X, Ma J, Tandon N, et al. Liraglutide provides similar glycaemic control as glimepiride (both in combination with metformin) and reduces body weight and systolic blood pressure in Asian population with type 2 diabetes from China, South Korea and India: A 16-week, randomized, double-blind, active control trial(*) Diabetes Obes Metab. 2011;13:81–8. doi: 10.1111/j.1463-1326.2010.01323.x. [DOI] [PubMed] [Google Scholar]
- 86.Gitt AK, Bramlage P, Schneider S, Tschöpe D. A real world comparison of sulfonylurea and insulin vs. incretin-based treatments in patients not controlled on prior metformin monotherapy. Cardiovasc Diabetol. 2015;14:13. doi: 10.1186/s12933-015-0172-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Massi-Benedetti M. Glimepiride in type 2 diabetes mellitus: A review of the worldwide therapeutic experience. Clin Ther. 2003;25:799–816. doi: 10.1016/s0149-2918(03)80109-1. [DOI] [PubMed] [Google Scholar]
- 88.Green JB, Feinglos MN. Are sulfonylureas passé? Curr Diab Rep. 2006;6:373–7. doi: 10.1007/s11892-006-0008-9. [DOI] [PubMed] [Google Scholar]
- 89.Charbonnel B, Schernthaner G, Brunetti P, Matthews DR, Urquhart R, Tan MH, et al. Long-term efficacy and tolerability of add-on pioglitazone therapy to failing monotherapy compared with addition of gliclazide or metformin in patients with type 2 diabetes. Diabetologia. 2005;48:1093–104. doi: 10.1007/s00125-005-1751-1. [DOI] [PubMed] [Google Scholar]
- 90.Zhang Y, Hong J, Chi J, Gu W, Ning G, Wang W, et al. Head-to-head comparison of dipeptidyl peptidase-IV inhibitors and sulfonylureas – A meta-analysis from randomized clinical trials. Diabetes Metab Res Rev. 2014;30:241–56. doi: 10.1002/dmrr.2482. [DOI] [PubMed] [Google Scholar]
- 91.Hissa MR, Cavalcante LL, Guimarães SB, Hissa MN. A 16-week study to compare the effect of vildagliptin versus gliclazide on postprandial lipoprotein concentrations and oxidative stress in patients with type 2 diabetes inadequately controlled with metformin monotherapy. Diabetol Metab Syndr. 2015;7:62. doi: 10.1186/s13098-015-0058-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Leiter LA, Yoon KH, Arias P, Langslet G, Xie J, Balis DA, et al. Canagliflozin provides durable glycemic improvements and body weight reduction over 104 weeks versus glimepiride in patients with type 2 diabetes on metformin: A randomized, double-blind, phase 3 study. Diabetes Care. 2015;38:355–64. doi: 10.2337/dc13-2762. [DOI] [PubMed] [Google Scholar]
- 93.Derosa G, Putignano P, Bossi AC, Bonaventura A, Querci F, Franzetti IG, et al. Exenatide or glimepiride added to metformin on metabolic control and on insulin resistance in type 2 diabetic patients. Eur J Pharmacol. 2011;666:251–6. doi: 10.1016/j.ejphar.2011.05.051. [DOI] [PubMed] [Google Scholar]
- 94.Derosa G, Maffioli P, Salvadeo SA, Ferrari I, Ragonesi PD, Querci F, et al. Exenatide versus glibenclamide in patients with diabetes. Diabetes Technol Ther. 2010;12:233–40. doi: 10.1089/dia.2009.0141. [DOI] [PubMed] [Google Scholar]
- 95.Moon JS, Ha KS, Yoon JS, Lee HW, Lee HC, Won KC, et al. The effect of glargine versus glimepiride on pancreatic β-cell function in patients with type 2 diabetes uncontrolled on metformin monotherapy: Open-label, randomized, controlled study. Acta Diabetol. 2014;51:277–85. doi: 10.1007/s00592-013-0553-z. [DOI] [PubMed] [Google Scholar]
- 96.Chan SP, Colagiuri S. Systematic review and meta-analysis of the efficacy and hypoglycemic safety of gliclazide versus other insulinotropic agents. Diabetes Res Clin Pract. 2015;110:75–81. doi: 10.1016/j.diabres.2015.07.002. [DOI] [PubMed] [Google Scholar]
- 97.Andersen SE, Christensen M. Hypoglycaemia when adding sulphonylurea to metformin: A systematic review and network meta-analysis. Br J Clin Pharmacol. 2016:1291–302. doi: 10.1111/bcp.13059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schernthaner G, Grimaldi A, Di Mario U, Drzewoski J, Kempler P, Kvapil M, et al. GUIDE study: Double-blind comparison of once-daily gliclazide MR and glimepiride in type 2 diabetic patients. Eur J Clin Invest. 2004;34:535–42. doi: 10.1111/j.1365-2362.2004.01381.x. [DOI] [PubMed] [Google Scholar]
- 99.Del Prato S, Nauck M, Durán-Garcia S, Maffei L, Rohwedder K, Theuerkauf A, et al. Long-term glycaemic response and tolerability of dapagliflozin versus a sulphonylurea as add-on therapy to metformin in patients with type 2 diabetes: 4-year data. Diabetes Obes Metab. 2015;17:581–90. doi: 10.1111/dom.12459. [DOI] [PubMed] [Google Scholar]
- 100.Gallwitz B, Guzman J, Dotta F, Guerci B, Simó R, Basson BR, et al. Exenatide twice daily versus glimepiride for prevention of glycaemic deterioration in patients with type 2 diabetes with metformin failure (EUREXA): An open-label, randomised controlled trial. Lancet. 2012;379:2270–8. doi: 10.1016/S0140-6736(12)60479-6. [DOI] [PubMed] [Google Scholar]
- 101.Mamza J, Mehta R, Donnelly R, Idris I. Important differences in the durability of glycaemic response among second-line treatment options when added to metformin in type 2 diabetes: A retrospective cohort study. Ann Med. 2016;48:224–34. doi: 10.3109/07853890.2016.1157263. [DOI] [PubMed] [Google Scholar]
- 102.Dhindsa P, Davis KR, Donnelly R. Comparison of the micro- and macro-vascular effects of glimepiride and gliclazide in metformin-treated patients with type 2 diabetes: A double-blind, crossover study. Br J Clin Pharmacol. 2003;55:616–9. doi: 10.1046/j.1365-2125.2003.01781.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Schramm TK, Gislason GH, Vaag A, Rasmussen JN, Folke F, Hansen ML, et al. Mortality and cardiovascular risk associated with different insulin secretagogues compared with metformin in type 2 diabetes, with or without a previous myocardial infarction: A nationwide study. Eur Heart J. 2011;32:1900–8. doi: 10.1093/eurheartj/ehr077. [DOI] [PubMed] [Google Scholar]
- 104.Mogensen UM, Andersson C, Fosbøl EL, Schramm TK, Vaag A, Scheller NM, et al. Cardiovascular safety of combination therapies with incretin-based drugs and metformin compared with a combination of metformin and sulphonylurea in type 2 diabetes mellitus – A retrospective nationwide study. Diabetes Obes Metab. 2014;16:1001–8. doi: 10.1111/dom.12314. [DOI] [PubMed] [Google Scholar]
- 105.Pfützner A, Schöndorf T, Tschöpe D, Lobmann R, Merke J, Müller J, et al. PIOfix-study: Effects of pioglitazone/metformin fixed combination in comparison with a combination of metformin with glimepiride on diabetic dyslipidemia. Diabetes Technol Ther. 2011;13:637–43. doi: 10.1089/dia.2010.0233. [DOI] [PubMed] [Google Scholar]
- 106.Prentice JC, Conlin PR, Gellad WF, Edelman D, Lee TA, Pizer SD, et al. Capitalizing on prescribing pattern variation to compare medications for type 2 diabetes. Value Health. 2014;17:854–62. doi: 10.1016/j.jval.2014.08.2674. [DOI] [PubMed] [Google Scholar]
- 107.Zhang H, Zhang X, Hu C, Lu W. Exenatide reduces urinary transforming growth factor-β1 and type IV collagen excretion in patients with type 2 diabetes and microalbuminuria. Kidney Blood Press Res. 2012;35:483–8. doi: 10.1159/000337929. [DOI] [PubMed] [Google Scholar]
- 108.Palmer SC, Mavridis D, Nicolucci A, Johnson DW, Tonelli M, Craig JC, et al. Comparison of clinical outcomes and adverse events associated with glucose-lowering drugs in patients with type 2 diabetes: A Meta-analysis. JAMA. 2016;316:313–24. doi: 10.1001/jama.2016.9400. [DOI] [PubMed] [Google Scholar]
- 109.Comaschi M, Demicheli A, Di Pietro C, Bellatreccia A, Mariz S COM06 Study Investigators. Effects of pioglitazone in combination with metformin or a sulfonylurea compared to a fixed-dose combination of metformin and glibenclamide in patients with type 2 diabetes. Diabetes Technol Ther. 2007;9:387–98. doi: 10.1089/dia.2006.0023. [DOI] [PubMed] [Google Scholar]
- 110.Schernthaner G, Durán-Garcia S, Hanefeld M, Langslet G, Niskanen L, Östgren CJ, et al. Efficacy and tolerability of saxagliptin compared with glimepiride in elderly patients with type 2 diabetes: A randomized, controlled study (GENERATION) Diabetes Obes Metab. 2015;17:630–8. doi: 10.1111/dom.12461. [DOI] [PubMed] [Google Scholar]
- 111.Ishii H, Ohkubo Y, Takei M, Nishio S, Yamazaki M, Kumagai M, et al. Efficacy of combination therapy with sitagliptin and low-dose glimepiride in Japanese patients with type 2 diabetes. J Clin Med Res. 2014;6:127–32. doi: 10.14740/jocmr1701w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Komatsu M, Takei M, Ishii H, Sato Y. Glucose-stimulated insulin secretion: A newer perspective. J Diabetes Investig. 2013;4:511–6. doi: 10.1111/jdi.12094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yabe D, Seino Y. Dipeptidyl peptidase-4 inhibitors and sulfonylureas for type 2 diabetes: Friend or foe? J Diabetes Investig. 2014;5:475–7. doi: 10.1111/jdi.12229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.McCluskey D, Touger MS, Melis R, Schleusener DS, McCluskey D. Results of a randomized, double-blind, placebo-controlled study administering glimepiride to patients with type 2 diabetes mellitus inadequately controlled with rosiglitazone monotherapy. Clin Ther. 2004;26:1783–90. doi: 10.1016/j.clinthera.2004.11018. [DOI] [PubMed] [Google Scholar]
- 115.Chou HS, Palmer JP, Jones AR, Waterhouse B, Ferreira-Cornwell C, Krebs J, et al. Initial treatment with fixed-dose combination rosiglitazone/glimepiride in patients with previously untreated type 2 diabetes. Diabetes Obes Metab. 2008;10:626–37. doi: 10.1111/j.1463-1326.2007.00753.x. [DOI] [PubMed] [Google Scholar]
- 116.Hanefeld M, Brunetti P, Schernthaner GH, Matthews DR, Charbonnel BH QUARTET Study Group. One-year glycemic control with a sulfonylurea plus pioglitazone versus a sulfonylurea plus metformin in patients with type 2 diabetes. Diabetes Care. 2004;27:141–7. doi: 10.2337/diacare.27.1.141. [DOI] [PubMed] [Google Scholar]
- 117.Shimoda S, Iwashita S, Sekigami T, Furukawa N, Matsuo Y, Ichimori S, et al. Comparison of the efficacy of sitagliptin and glimepiride dose-up in Japanese patients with type 2 diabetes poorly controlled by sitagliptin and glimepiride in combination. J Diabetes Investig. 2014;5:320–6. doi: 10.1111/jdi.12151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Harashima SI, Ogura M, Tanaka D, Fukushima T, Wang Y, Koizumi T, et al. Sitagliptin add-on to low dosage sulphonylureas: Efficacy and safety of combination therapy on glycaemic control and insulin secretion capacity in type 2 diabetes. Int J Clin Pract. 2012;66:465–76. doi: 10.1111/j.1742-1241.2012.02903.x. [DOI] [PubMed] [Google Scholar]
- 119.Roberts VL, Stewart J, Issa M, Lake B, Melis R. Triple therapy with glimepiride in patients with type 2 diabetes mellitus inadequately controlled by metformin and a thiazolidinedione: Results of a 30-week, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther. 2005;27:1535–47. doi: 10.1016/j.clinthera.2005.10.017. [DOI] [PubMed] [Google Scholar]
- 120.Arai K, Maeda H, Sirabe S, Yamamoto R, Yamauchi M, Hirao T, et al. Glimepiride strongly enhances the glucose-lowering effect in triple oral antidiabetes therapy with sitagliptin and metformin for Japanese patients with type 2 diabetes mellitus. Diabetes Technol Ther. 2013;15:335–41. doi: 10.1089/dia.2012.0287. [DOI] [PubMed] [Google Scholar]
- 121.Downes MJ, Bettington EK, Gunton JE, Turkstra E. Triple therapy in type 2 diabetes; a systematic review and network meta-analysis. PeerJ. 2015;3:e1461. doi: 10.7717/peerj.1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Derosa G, Cicero AF, D’Angelo A, Gaddi A, Ciccarelli L, Piccinni MN, et al. Effects of 1 year of treatment with pioglitazone or rosiglitazone added to glimepiride on lipoprotein (a) and homocysteine concentrations in patients with type 2 diabetes mellitus and metabolic syndrome: A multicenter, randomized, double-blind, controlled clinical trial. Clin Ther. 2006;28:679–88. doi: 10.1016/j.clinthera.2006.05.012. [DOI] [PubMed] [Google Scholar]
- 123.Davidson JA, McMorn SO, Waterhouse BR, Cobitz AR. A 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study of the efficacy and tolerability of combination therapy with rosiglitazone and sulfonylurea in African American and Hispanic American patients with type 2 diabetes inadequately controlled with sulfonylurea monotherapy. Clin Ther. 2007;29:1900–14. doi: 10.1016/j.clinthera.2007.09.011. [DOI] [PubMed] [Google Scholar]
- 124.Umayahara R, Yonemoto T, Kyou C, Morishita K, Ogawa T, Taguchi Y, et al. Low-dose glimepiride with sitagliptin improves glycemic control without dose-dependency in patients with type 2 diabetes inadequately controlled on high-dose glimepiride. Endocr J. 2014;61:1163–70. doi: 10.1507/endocrj.EJ14-0233. [DOI] [PubMed] [Google Scholar]
- 125.Schernthaner G, Rosas-Guzmán J, Dotta F, Guerci B, Simó R, Festa A, et al. Treatment escalation options for patients with type 2 diabetes after failure of exenatide twice daily or glimepiride added to metformin: Results from the prospective European Exenatide (EUREXA) study. Diabetes Obes Metab. 2015;17:689–98. doi: 10.1111/dom.12471. [DOI] [PubMed] [Google Scholar]
- 126.Derosa G, Cicero AF, Gaddi A, Ragonesi PD, Fogari E, Bertone G, et al. Metabolic effects of pioglitazone and rosiglitazone in patients with diabetes and metabolic syndrome treated with glimepiride: A twelve-month, multicenter, double-blind, randomized, controlled, parallel-group trial. Clin Ther. 2004;26:744–54. doi: 10.1016/s0149-2918(04)90074-4. [DOI] [PubMed] [Google Scholar]
- 127.Meshram DM, Langade DG, Kinagi SB, Naikwadi AA, Morye V, Chopra D, et al. Evaluation of efficacy and safety of fixed dose combination of glimepiride 2 mg plus pioglitazone 15 mg plus metformin SR 500 mg in the management of patients with type-2 diabetes mellitus. J Indian Med Assoc. 2005;103:447–50. [PubMed] [Google Scholar]
- 128.Sola D, Rossi L, Schianca GP, Maffioli P, Bigliocca M, Mella R, et al. Sulfonylureas and their use in clinical practice. Arch Med Sci. 2015;11:840–8. doi: 10.5114/aoms.2015.53304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Lebovitz HE, Pasmantier R. Combination insulin-sulfonylurea therapy. Diabetes Care. 1990;13:667–75. doi: 10.2337/diacare.13.6.667. [DOI] [PubMed] [Google Scholar]
- 130.Groop LC, Groop PH, Stenman S. Combined insulin-sulfonylurea therapy in treatment of NIDDM. Diabetes Care. 1990;13(Suppl 3):47–52. doi: 10.2337/diacare.13.3.47. [DOI] [PubMed] [Google Scholar]
- 131.Zhou J, Zheng F, Guo X, Yang H, Zhang M, Tian H, et al. Glargine insulin/gliclazide MR combination therapy is more effective than premixed insulin monotherapy in Chinese patients with type 2 diabetes inadequately controlled on oral antidiabetic drugs. Diabetes Metab Res Rev. 2015;31:725–33. doi: 10.1002/dmrr.2661. [DOI] [PubMed] [Google Scholar]
- 132.Schiel R, Müller UA. Efficacy and treatment satisfaction of once-daily insulin glargine plus one or two oral antidiabetic agents versus continuing premixed human insulin in patients with type 2 diabetes previously on long-term conventional insulin therapy: The SWITCH pilot study. Exp Clin Endocrinol Diabetes. 2008;116:58–64. doi: 10.1055/s-2007-985367. [DOI] [PubMed] [Google Scholar]
- 133.Janka HU, Plewe G, Busch K. Combination of oral antidiabetic agents with basal insulin versus premixed insulin alone in randomized elderly patients with type 2 diabetes mellitus. J Am Geriatr Soc. 2007;55:182–8. doi: 10.1111/j.1532-5415.2007.01043.x. [DOI] [PubMed] [Google Scholar]
- 134.Standl E, Maxeiner S, Raptis S, Karimi-Anderesi Z, Schweitzer MA HOE901/4009 Study Group. Good glycemic control with flexibility in timing of basal insulin supply: A 24-week comparison of insulin glargine given once daily in the morning or at bedtime in combination with morning glimepiride. Diabetes Care. 2005;28:419–20. doi: 10.2337/diacare.28.2.419. [DOI] [PubMed] [Google Scholar]
- 135.Olsson PO, Lindström T. Combination-therapy with bedtime nph insulin and sulphonylureas gives similar glycaemic control but lower weight gain than insulin twice daily in patients with type 2 diabetes. Diabetes Metab. 2002;28:272–7. [PubMed] [Google Scholar]
- 136.Park CY, Kang JG, Chon S, Noh J, Oh SJ, Lee CB, et al. Comparison between the therapeutic effect of metformin, glimepiride and their combination as an add-on treatment to insulin glargine in uncontrolled patients with type 2 diabetes. PLoS One. 2014;9:e87799. doi: 10.1371/journal.pone.0087799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Li CJ, Zhang JY, Yu DM, Zhang QM. Adding glimepiride to current insulin therapy increases high-molecular weight adiponectin levels to improve glycemic control in poorly controlled type 2 diabetes. Diabetol Metab Syndr. 2014;6:41. doi: 10.1186/1758-5996-6-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Vos RC, van Avendonk MJ, Jansen H, Goudswaard AN, van den Donk M, Gorter K, et al. Insulin monotherapy compared with the addition of oral glucose-lowering agents to insulin for people with type 2 diabetes already on insulin therapy and inadequate glycaemic control. Cochrane Database Syst Rev. 2016;9:CD006992. doi: 10.1002/14651858.CD006992.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Pugh JA, Wagner ML, Sawyer J, Ramirez G, Tuley M, Friedberg SJ, et al. Is combination sulfonylurea and insulin therapy useful in NIDDM patients? A metaanalysis. Diabetes Care. 1992;15:953–9. doi: 10.2337/diacare.15.8.953. [DOI] [PubMed] [Google Scholar]
- 140.Esposito K, Ciotola M, Maiorino MI, Gualdiero R, Schisano B, Ceriello A, et al. Addition of neutral protamine lispro insulin or insulin glargine to oral type 2 diabetes regimens for patients with suboptimal glycemic control: A randomized trial. Ann Intern Med. 2008;149:531–9. doi: 10.7326/0003-4819-149-8-200810210-00005. [DOI] [PubMed] [Google Scholar]
- 141.Janka HU, Plewe G, Riddle MC, Kliebe-Frisch C, Schweitzer MA, Yki-Järvinen H, et al. Comparison of basal insulin added to oral agents versus twice-daily premixed insulin as initial insulin therapy for type 2 diabetes. Diabetes Care. 2005;28:254–9. doi: 10.2337/diacare.28.2.254. [DOI] [PubMed] [Google Scholar]
- 142.Wright A, Burden AC, Paisey RB, Cull CA, Holman RR U.K. Prospective Diabetes Study Group. Sulfonylurea inadequacy: Efficacy of addition of insulin over 6 years in patients with type 2 diabetes in the U.K. Prospective diabetes study (UKPDS 57) Diabetes Care. 2002;25:330–6. doi: 10.2337/diacare.25.2.330. [DOI] [PubMed] [Google Scholar]
- 143.Yki-Järvinen H, Ryysy L, Nikkilä K, Tulokas T, Vanamo R, Heikkilä M, et al. Comparison of bedtime insulin regimens in patients with type 2 diabetes mellitus. A randomized, controlled trial. Ann Intern Med. 1999;130:389–96. doi: 10.7326/0003-4819-130-5-199903020-00002. [DOI] [PubMed] [Google Scholar]
- 144.Holman RR, Thorne KI, Farmer AJ, Davies MJ, Keenan JF, Paul S, et al. Addition of biphasic, prandial, or basal insulin to oral therapy in type 2 diabetes. N Engl J Med. 2007;357:1716–30. doi: 10.1056/NEJMoa075392. [DOI] [PubMed] [Google Scholar]
- 145.Holman RR, Farmer AJ, Davies MJ, Levy JC, Darbyshire JL, Keenan JF, et al. Three-year efficacy of complex insulin regimens in type 2 diabetes. N Engl J Med. 2009;361:1736–47. doi: 10.1056/NEJMoa0905479. [DOI] [PubMed] [Google Scholar]
- 146.Stuart CA, Gilkison CR, Carlson RF, Stuart CA, Gilkison CR, Carlson RF, et al. Effect of adding a sulfonylurea in patients with non-insulin-dependent diabetes mellitus previously well controlled with insulin. Endocr Pract. 1997;3:344–8. doi: 10.4158/EP.3.6.344. [DOI] [PubMed] [Google Scholar]
- 147.Riddle M, Hart J, Bingham P, Garrison C, McDaniel P. Combined therapy for obese type 2 diabetes: Suppertime mixed insulin with daytime sulfonylurea. Am J Med Sci. 1992;303:151–6. doi: 10.1097/00000441-199203000-00003. [DOI] [PubMed] [Google Scholar]
- 148.Chale SS, Swai AB, Mujinja PG, McLarty DG. Must diabetes be a fatal disease in Africa? Study of costs of treatment. BMJ. 1992;304:1215–8. doi: 10.1136/bmj.304.6836.1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Zhang Y, McCoy RG, Mason JE, Smith SA, Shah ND, Denton BT, et al. Second-line agents for glycemic control for type 2 diabetes: Are newer agents better? Diabetes Care. 2014;37:1338–45. doi: 10.2337/dc13-1901. [DOI] [PubMed] [Google Scholar]
- 150.Pan F, Chernew ME, Fendrick AM. Impact of fixed-dose combination drugs on adherence to prescription medications. J Gen Intern Med. 2008;23:611–4. doi: 10.1007/s11606-008-0544-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Saini SD, Schoenfeld P, Kaulback K, Dubinsky MC. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15:e22–33. [PubMed] [Google Scholar]
- 152.Meece J. Improving medication adherence among patients with type 2 diabetes. J Pharm Pract. 2014;27:187–94. doi: 10.1177/0897190013513803. [DOI] [PubMed] [Google Scholar]
- 153.Bangalore S, Kamalakkannan G, Parkar S, Messerli FH. Fixed-dose combinations improve medication compliance: A meta-analysis. Am J Med. 2007;120:713–9. doi: 10.1016/j.amjmed.2006.08.033. [DOI] [PubMed] [Google Scholar]
- 154.Blonde L, San Juan ZT. Fixed-dose combinations for treatment of type 2 diabetes mellitus. Adv Ther. 2012;29:1–3. doi: 10.1007/s12325-011-0094-1. [DOI] [PubMed] [Google Scholar]
- 155.Riddle M. Combining sulfonylureas and other oral agents. Am J Med. 2000;108(Suppl 6a):15S–22S. doi: 10.1016/s0002-9343(00)00338-7. [DOI] [PubMed] [Google Scholar]
- 156.Kalra S. Aggressive treatment in newly diagnosed diabetes with fixed dose combinations. Medicine. 2012;22:249–53. [Google Scholar]
- 157.Han S, Iglay K, Davies MJ, Zhang Q, Radican L. Glycemic effectiveness and medication adherence with fixed-dose combination or coadministered dual therapy of antihyperglycemic regimens: A meta-analysis. Curr Med Res Opin. 2012;28:969–77. doi: 10.1185/03007995.2012.684045. [DOI] [PubMed] [Google Scholar]
- 158.Hutchins V, Zhang B, Fleurence RL, Krishnarajah G, Graham J. A systematic review of adherence, treatment satisfaction and costs, in fixed-dose combination regimens in type 2 diabetes. Curr Med Res Opin. 2011;27:1157–68. doi: 10.1185/03007995.2011.570745. [DOI] [PubMed] [Google Scholar]
- 159.Drouin P, Standl E Diamicron MR Study Group. Gliclazide modified release: Results of a 2-year study in patients with type 2 diabetes. Diabetes Obes Metab. 2004;6:414–21. doi: 10.1111/j.1462-8902.2004.00404.x. [DOI] [PubMed] [Google Scholar]
- 160.Chanal H. Should elderly patients with type 2 diabetes be treated with glibenclamide (glyburide) or different sulphonylurea? Geneva: World Health Organization; 2013. [Google Scholar]
- 161.International Diabetes Federation, Managing Older People with Type 2 Diabetes. Global Guideline. 2013. [Last accessed on 2017 Apr 27]. Available from: https://www.idf.org/e-library/guidelines/78-global-guideline-for-managing-older-peoplewith-type-2-diabetes.html .
- 162.Canadian Diabetes Association Clinical Practice Guidelines Expert Committee. Meneilly GS, Knip A, Tessier D. Diabetes in the elderly. [Last accessed on 2015 Aug 11];Can J Diabetes. 2013 37(Suppl 1):S184–90. doi: 10.1016/j.jcjd.2013.01.045. Available from: http://www.canadianjournalofdiabetes.com/article/S1499-2671(13)00046-4/pdf . [DOI] [PubMed] [Google Scholar]
- 163.Onge ES, Miller SA, Motycka C, DeBerry A. A review of the treatment of type 2 diabetes in children. J Pediatr Pharmacol Ther. 2015;20:4–16. doi: 10.5863/1551-6776-20.1.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Gottschalk M, Danne T, Vlajnic A, Cara JF. Glimepiride versus metformin as monotherapy in pediatric patients with type 2 diabetes: A randomized, single-blind comparative study. Diabetes Care. 2007;30:790–4. doi: 10.2337/dc06-1554. [DOI] [PubMed] [Google Scholar]
- 165.Fajans SS, Brown MB. Administration of sulfonylureas can increase glucose-induced insulin secretion for decades in patients with maturity-onset diabetes of the young. Diabetes Care. 1993;16:1254–61. doi: 10.2337/diacare.16.9.1254. [DOI] [PubMed] [Google Scholar]
- 166.Raile K, Schober E, Konrad K, Thon A, Grulich-Henn J, Meissner T, et al. Treatment of young patients with HNF1A mutations (HNF1A-MODY) Diabet Med. 2015;32:526–30. doi: 10.1111/dme.12662. [DOI] [PubMed] [Google Scholar]
- 167.Thanabalasingham G, Owen KR. Diagnosis and management of maturity onset diabetes of the young (MODY) BMJ. 2011;343:d6044. doi: 10.1136/bmj.d6044. [DOI] [PubMed] [Google Scholar]
- 168.Hanas R, Donaghue K, Klingensmith G, Swift P, Colagiuri S. Global IDF/ISPAD Guideline for Diabetes in Childhood and Adolescence. Brussels: International Diabetes Federation; 2011. [Google Scholar]
- 169.Oztekin O, Durmaz E, Kalay S, Flanagan SE, Ellard S, Bircan I, et al. Successful sulfonylurea treatment of a neonate with neonatal diabetes mellitus due to a novel missense mutation, p.P1199L, in the ABCC8 gene. J Perinatol. 2012;32:645–7. doi: 10.1038/jp.2012.46. [DOI] [PubMed] [Google Scholar]
- 170.Pearson ER, Flechtner I, Njølstad PR, Malecki MT, Flanagan SE, Larkin B, et al. Switching from insulin to oral sulfonylureas in patients with diabetes due to kir6.2 mutations. N Engl J Med. 2006;355:467–77. doi: 10.1056/NEJMoa061759. [DOI] [PubMed] [Google Scholar]
- 171.Babenko AP, Polak M, Cavé H, Busiah K, Czernichow P, Scharfmann R, et al. Activating mutations in the ABCC8 gene in neonatal diabetes mellitus. N Engl J Med. 2006;355:456–66. doi: 10.1056/NEJMoa055068. [DOI] [PubMed] [Google Scholar]
- 172.Sagen JV, Raeder H, Hathout E, Shehadeh N, Gudmundsson K, Baevre H, et al. Permanent neonatal diabetes due to mutations in KCNJ11 encoding kir6.2: Patient characteristics and initial response to sulfonylurea therapy. Diabetes. 2004;53:2713–8. doi: 10.2337/diabetes.53.10.2713. [DOI] [PubMed] [Google Scholar]
- 173.Codner E, Flanagan SE, Ugarte F, García H, Vidal T, Ellard S, et al. Sulfonylurea treatment in young children with neonatal diabetes: Dealing with hyperglycemia, hypoglycemia, and sick days. Diabetes Care. 2007;30:e28–9. doi: 10.2337/dc06-2134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Kavitha N, De S, Kanagasabai S. Oral hypoglycemic agents in pregnancy: An update. J Obstet Gynaecol India. 2013;63:82–7. doi: 10.1007/s13224-012-0312-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Kalra B, Gupta Y, Singla R, Kalra S. Use of oral anti-diabetic agents in pregnancy: A pragmatic approach. N Am J Med Sci. 2015;7:6–12. doi: 10.4103/1947-2714.150081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Nicholson W, Bolen S, Witkop CT, Neale D, Wilson L, Bass E, et al. Benefits and risks of oral diabetes agents compared with insulin in women with gestational diabetes: A systematic review. Obstet Gynecol. 2009;113:193–205. doi: 10.1097/AOG.0b013e318190a459. [DOI] [PubMed] [Google Scholar]
- 177.Dhulkotia JS, Ola B, Fraser R, Farrell T. Oral hypoglycemic agents vs. insulin in management of gestational diabetes: A systematic review and metaanalysis. Am J Obstet Gynecol. 2010;203:457. doi: 10.1016/j.ajog.2010.06.044. e1-9. [DOI] [PubMed] [Google Scholar]
- 178.Gutzin SJ, Kozer E, Magee LA, Feig DS, Koren G. The safety of oral hypoglycemic agents in the first trimester of pregnancy: A meta-analysis. Can J Clin Pharmacol. 2003;10:179–83. [PubMed] [Google Scholar]
- 179.Azimova K, San Juan Z, Mukherjee D. Cardiovascular safety profile of currently available diabetic drugs. Ochsner J. 2014;14:616–32. [PMC free article] [PubMed] [Google Scholar]
- 180.Ioannidis I. Diabetes treatment in patients with renal disease: Is the landscape clear enough? World J Diabetes. 2014;5:651–8. doi: 10.4239/wjd.v5.i5.651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Rosenkranz B, Profozic V, Metelko Z, Mrzljak V, Lange C, Malerczyk V, et al. Pharmacokinetics and safety of glimepiride at clinically effective doses in diabetic patients with renal impairment. Diabetologia. 1996;39:1617–24. doi: 10.1007/s001250050624. [DOI] [PubMed] [Google Scholar]
- 182.van Dalem J, Brouwers MC, Stehouwer CD, Krings A, Leufkens HG, Driessen JH, et al. Risk of hypoglycaemia in users of sulphonylureas compared with metformin in relation to renal function and sulphonylurea metabolite group: Population based cohort study. BMJ. 2016;354:i3625. doi: 10.1136/bmj.i3625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Arjona Ferreira JC, Marre M, Barzilai N, Guo H, Golm GT, Sisk CM, et al. Efficacy and safety of sitagliptin versus glipizide in patients with type 2 diabetes and moderate-to-severe chronic renal insufficiency. Diabetes Care. 2013;36:1067–73. doi: 10.2337/dc12-1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Saini JS. Management guidelines for use of oral hypoglycemic agents (OHA) in complex clinical situations and important drug interactions with OHA's. Acta Endocrinol. 1991;108:85–90. [Google Scholar]
- 185.Tolman KG, Fonseca V, Dalpiaz A, Tan MH. Spectrum of liver disease in type 2 diabetes and management of patients with diabetes and liver disease. Diabetes Care. 2007;30:734–43. doi: 10.2337/dc06-1539. [DOI] [PubMed] [Google Scholar]
- 186.Hassanein M, Al-Arouj M, Hamdy O, Bebakar WM, Jabbar A, Al-Madani A, et al. Diabetes and Ramadan: Practical guidelines. Diabetes Res Clin Pract. 2017;126:303–16. doi: 10.1016/j.diabres.2017.03.003. [DOI] [PubMed] [Google Scholar]
- 187.Glimepiride in Ramadan (GLIRA) Study Group. The efficacy and safety of glimepiride in the management of type 2 diabetes in Muslim patients during Ramadan. Diabetes Care. 2005;28:421–2. doi: 10.2337/diacare.28.2.421. [DOI] [PubMed] [Google Scholar]
- 188.Hassanein M, Abdallah K, Schweizer A. A double-blind, randomized trial, including frequent patient-physician contacts and Ramadan-focused advice, assessing vildagliptin and gliclazide in patients with type 2 diabetes fasting during Ramadan: The STEADFAST study. Vasc Health Risk Manag. 2014;10:319–26. doi: 10.2147/VHRM.S64038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Brady EM, Davies MJ, Gray LJ, Saeed MA, Smith D, Hanif W, et al. A randomized controlled trial comparing the GLP-1 receptor agonist liraglutide to a sulphonylurea as add on to metformin in patients with established type 2 diabetes during Ramadan: The treat 4 Ramadan trial. Diabetes Obes Metab. 2014;16:527–36. doi: 10.1111/dom.12249. [DOI] [PubMed] [Google Scholar]
- 190.Azar ST, Echtay A, Wan Bebakar WM, Al Araj S, Berrah A, Omar M, et al. Efficacy and safety of liraglutide compared to sulphonylurea during Ramadan in patients with type 2 diabetes (LIRA-Ramadan): A randomized trial. Diabetes Obes Metab. 2016;18:1025–33. doi: 10.1111/dom.12733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Malha LP, Taan G, Zantout MS, Azar ST. Glycemic effects of vildagliptin in patients with type 2 diabetes before, during and after the period of fasting in Ramadan. Ther Adv Endocrinol Metab. 2014;5:3–9. doi: 10.1177/2042018814529062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Al Sifri S, Basiounny A, Echtay A, Al Omari M, Harman-Boehm I, Kaddaha G, et al. The incidence of hypoglycaemia in Muslim patients with type 2 diabetes treated with sitagliptin or a sulphonylurea during Ramadan: A randomised trial. Int J Clin Pract. 2011;65:1132–40. doi: 10.1111/j.1742-1241.2011.02797.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Almalki MH, Alshahrani F. Options for controlling type 2 diabetes during Ramadan. Front Endocrinol (Lausanne) 2016;7:32. doi: 10.3389/fendo.2016.00032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Kalra S, Bajaj S, Gupta Y, Agarwal P, Singh SK, Julka S, et al. Fasts, feasts and festivals in diabetes-1: Glycemic management during Hindu fasts. Indian J Endocrinol Metab. 2015;19:198–203. doi: 10.4103/2230-8210.149314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Latt TS, Kalra S. Managing diabetes during fasting – A focus on Buddhist lent. Diabetes Voice. 2012;57:42–5. [Google Scholar]
- 196.Julka S, Sachan A, Bajaj S, Sahay R, Chawla R, Agrawal N, et al. Glycemic management during Jain fasts. Indian J Endocrinol Metab. 2017;21:238–41. doi: 10.4103/2230-8210.192489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Glyade: Gliclazide Tablets: Label Information of TGA. Diabeta. [Last accessed on 2015 Mar 25]. Available from: http://www.medicines.org.au/files/afpglyad.pdf .
- 198.Glyburide Tablets. Label Information of FDA. [Last accessed on 2015 Mar 25]. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/017532Orig1s034lbl.pdf .
- 199.Amaryl: Glimepiride Tablets. Label Information of FDA. [Last accessed on 2015 Mar 25]. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/020496s027lbl.pdf .
- 200.Powers MA, Bardsley J, Cypress M, Duker P, Funnell MM, Fischl AH, et al. Diabetes self-management education and support in type 2 diabetes: A joint position statement of the American Diabetes Association, the American association of diabetes educators, and the academy of nutrition and dietetics. Diabetes Educ. 2015;41:417–30. doi: 10.1177/0145721715588904. [DOI] [PubMed] [Google Scholar]