114
Views
14
CrossRef citations to date
0
Altmetric
Review

Practical combination therapy based on pathophysiology of type 2 diabetes

Pages 355-369 | Published online: 31 Oct 2016

Abstract

Type 2 diabetes is a complex, chronic, and progressive condition that often necessitates the use of multiple medications to achieve glycemic goals. Clinical guidelines generally recommend intensifying pharmacotherapy if glycemic goals are not achieved after 3 months of treatment. However, for many patients with type 2 diabetes, treatment intensification is delayed or does not occur. Initiating combination therapy early in the disease course has the potential to delay disease progression and improve patient outcomes. Guidelines generally provide a list of agents that may be used in combination regimens and emphasize individualization of treatment. The purpose of this review is to discuss the rationale for combination therapy, considering treatment effects on pathophysiologic aspects of type 2 diabetes and individual drug profiles. The combination of newer antidiabetes therapies with complementary mechanisms of action provides the opportunity to target multiple sites of tissue, organ, and cellular dysfunction.

Introduction

More than 415 million adults worldwide are estimated to have diabetes, with an increase to 642 million expected by 2040.Citation1 Type 2 diabetes (T2D) is the most common type of diabetes in adults, and the prevalence is expected to increase largely because of a parallel increase in the prevalence of obesity.Citation1 There have been significant advances in the understanding of glucose homeostasis and the pathophysiology of T2D over the past few decades.Citation2 It is now understood that glucose concentrations are maintained by a complex network of finely tuned feedback mechanisms involving multiple organs and hormonal systems and that dysfunctions in this network contribute to the development and progression of insulin resistance and β-cell failure, hallmarks of T2D.Citation2 Although there is still much to be learned about the disease process,Citation2 current understanding suggests that early intervention with targeted pharmacotherapy to correct multiple sites of dysfunction might slow T2D progression and improve patient outcomes.

Treatment guidelines recommend metformin as initial oral pharmacotherapy for most adults,Citation3,Citation4 although study findings show that only 58% of patients are started on metformin in clinical practice.Citation5 Guidelines consistently advise follow-up at 3 months and a general stepwise approach to pharmacotherapy with the addition of another anti-diabetes agent if glycemic goals are not metCitation3,Citation4; however, many patients with T2D are subject to notable delays in treatment intensification, generally referred to as clinical inertia.Citation6 A retrospective cohort study in patients with T2D and glycated hemoglobin (A1C) ≥7% or ≥8% taking one oral antidiabetes drug (OAD) found that the median time to treatment intensification with an additional OAD was 2.9 and 1.6 years, respectively, and more than 7.2 and 6.9 years in patients already taking two OADs.Citation6 A recent retrospective database study found that in nearly 80% of patients with uncontrolled glycemia despite two OADs, a third OAD was added to the treatment regimen rather than insulin, which provided greater A1C reduction and lower health care costs, suggesting resistance to prescribing and taking insulin in current practice.Citation7 These prolonged periods of inadequate glycemic control can have a detrimental effect on long-term outcomes.Citation6,Citation8

Guidelines from the American Association of Clinical Endocrinologists (AACE) suggest early use of combination pharmacotherapy for patients with high A1C at treatment initiation ().Citation3 AACE recommends initial treatment with dual therapy for patients with an A1C ≥7.5% and advancement to triple therapy if A1C goals are not met after 3 months. For patients with an A1C >9.0% and no hyperglycemia symptoms (eg, polyuria, polydipsia, polyphagia), initial pharmacotherapy with three antidiabetes agents may be appropriate.Citation3 AACE guidelines further note that combination therapy should include medications with complementary mechanisms of action to maximize effectivenessCitation3,Citation9 to correct multiple sites of dysfunction. Targeting multiple sites of dysfunction using combination therapy early in the disease course may also produce more durable efficacy, particularly when using agents that preserve β-cell function.Citation10 The purpose of this review is to discuss the rationale for combination therapy based on the pathophysiology of T2D, providing an overview of the underlying defects in T2D and the primary mechanisms of action of commonly used antidiabetes medications for a greater insight into their use in combination.

Figure 1 Treatment algorithm based on A1C at entry.

Notes: Order of medications suggests hierarchy of usage by the AACE. Adapted with permission from American Association of Clinical Endocrinologists © 2016 AACE. Garber AJ, Abrahamson MJ, Barzilay JI, et al. AACE/ACE comprehensive type 2 diabetesmanagement algorithm 2016. Endocr Pract. 2016;22:84–113.Citation3 *Indicates few adverse events or possible benefits. #Use with caution.
Abbreviations: A1C, glycated hemoglobin; AACE, American Association of Clinical Endocrinologists; ACE, American College of Endocrinology; AGI, α-glucosidase inhibitor; DPP-4, dipeptidyl peptidase-4; GLN, glinide; GLP-1RA, glucagon-like peptide-1 receptor agonist; SGLT-2, sodium-glucose cotransporter-2; SU, sulfonylurea; TZD, thiazolidinedione.
Figure 1 Treatment algorithm based on A1C at entry.

Pathophysiology of T2D

Feedback regulation to maintain glucose homeostasis and the critical role of β-cells

In glucose-tolerant individuals, a feedback loop between β-cells and insulin-sensitive tissues maintains normal glucose homeostasis. In this cyclic process, insulin secreted in response to β-cell stimulation suppresses glucose production in the liver and facilitates uptake of glucose, amino acids, and fatty acids in adipose tissue and muscle. Plasma glucose concentrations are maintained within a narrow range (70–90 mg/dL) and are primarily regulated by the hormones insulin, glucagon, and incretins, with input from the central and peripheral nervous systems.Citation11 Pancreatic glucagon plays an important role in maintaining glucose homeostasis by stimulating glycogenolysis and gluconeogenesis, metabolic processes in the liver and kidney that convert glycogen and other substrates (lactate, amino acids, glycerol) to free glucose for circulation in the fasted state.Citation12,Citation13 It is estimated that renal glucose release accounts for 20% of overall endogenous glucose release and ~40% of all gluconeogenesis.Citation14 Glycogen is also stored in the skeletal muscle; however, muscle glycogen is not directly available to other tissues because it lacks the enzyme glucose-6-phosphatase needed to produce glucose.Citation14 When exogenous glucose is available, insulin suppresses glycogenolysis and gluconeogenesis. In patients with T2D, however, hepatic insulin resistance results in the inability of normal concentrations of insulin to inhibit glycogenolysis and gluconeogenesis,Citation15 resulting in increases in basal hepatic glucose production (HGP) and hyperglycemia in the fasted state.Citation16

Normally functioning adipose tissue is one of the key regulatory tissues involved in maintaining lipid and glucose homeostasis.Citation17 Thus, when adipose tissue is in a state of chronic low-level inflammation as occurs with obesity, a multitude of pathologies emerge, including peripheral insulin resistance.Citation18,Citation19 Preclinical studies have shown macrophage accumulation occurring at the onset of weight gain, and inflammation and macrophage-specific gene expression are upregulated in correlation with increased adiposity.Citation20 In both animals and humans, macrophage accumulation is in direct proportion to adipocyte size and body mass index.Citation21 This increased infiltration of macrophages into adipose tissue perpetuates the production of proinflammatory molecules, including free fatty acids that impair insulin sensitivity, which initiate additional activation and infiltration of peripheral monocytes and macrophages.Citation20,Citation21 Eventually, the continued state of inflammation and impaired insulin signaling in adipose tissue leads to lipolysis, necrosis, and the development of systemic insulin resistance.Citation20

The β-cell is highly attuned to changes in insulin sensitivity and can maintain glucose homeostasis by increasing insulin secretion during the early stages of insulin resistance. When the β-cell can no longer produce enough insulin to compensate for a diminished insulin response, glucose concentrations rise.Citation2 Exposure to chronically elevated glucose concentrations leads to production of reactive oxygen species that cause oxidative stress and increased production of free fatty acids, leading to glucolipotoxicity. This in turn interferes with β-cell compensatory mechanisms, collectively contributing to worsened β-cell function and apoptosis.Citation2,Citation22 The overall extent to which β-cell function is impaired determines the severity of hyperglycemia, and the progressive failure of β-cells accounts for the transition from impaired glucose tolerance to T2D.Citation2 The rate at which β-cell impairment occurs varies among patients, although certain factors, most notably poor metabolic control and duration of T2D, contribute to progression.Citation23

Multiple genetic and environmental factors also contribute to the development of insulin resistance and β-cell dysfunction.Citation2 Insulin resistance and reduced β-cell function are largely heritable and present in individuals with normal glucose tolerance but at risk for hyperglycemia (eg, first-degree relatives of patients with T2D).Citation24,Citation25 Obesity, nutrient composition (particularly intake of saturated fats), and other factors, such as age-related declines in β-cell response, also contribute to glucose intolerance.Citation2,Citation26 A genetic propensity for obesity has also been demonstratedCitation27; obesity and related insulin resistance are thought to arise when genes for abnormal body adiposity interact with certain environmental factors (eg, increased caloric intake, decreased energy expenditure).Citation2 Although it is not yet known how genes interact with the environment to cause progressive β-cell dysfunction, and there is inherent variability among patients, current data suggest that both a genetic propensity for β-cell dysfunction and increased body adiposity may be necessary for the interaction with environmental factors that results in T2D.Citation2

Dysfunction in other body systems

Gastrointestinal tract

In healthy subjects, more insulin is secreted after nutrient ingestion compared with a similar nutrient load given intravenously.Citation28 The gastrointestinal-derived incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic hormone (GIP), are largely responsible for this effect and primarily stimulate insulin secretion in a glucose-dependent manner by acting at their receptors on β-cells.Citation28 GLP-1 has also been shown to directly inhibit glucagon secretion via GLP-1 receptors on pancreatic α cells or indirectly by stimulating insulin and somatostatin secretion.Citation29 In vivo, GLP-1 and GIP facilitate β-cell proliferation and inhibit apoptosis, in addition to stimulating insulin secretion. GLP-1 also slows gastric emptying and decreases food intake, likely through multiple mechanisms.Citation29,Citation30 Under basal conditions, circulating GLP-1 and GIP concentrations are low but rise rapidly in the postprandial state and then decline as GIP and GLP-1 are rapidly degraded by enzymes, primarily dipeptidyl peptidase-4 (DPP-4).Citation31 In patients with T2D, the insulinotropic response to GIP is notably reduced,Citation32 which likely accounts for much of the decrease in incretin activity.Citation33 In contrast, the insulinotropic response to GLP-1 remains largely intact in T2D,Citation28 although certain patient factors, such as high body mass index or high glucagon concentrations, may reduce GLP-1 secretion.Citation33

Kidney

In glucose-tolerant subjects, nearly all glucose filtered by the renal glomeruli is reabsorbed by the proximal tubule for recirculation.Citation34 Glucose is excreted into the urine to help maintain glucose homeostasis only if the maximum glucose transport capacity is exceeded.Citation34 Glucose reabsorption from the glomerular filtrate is primarily mediated by sodium-glucose cotransporter-2 (SGLT-2), a low-affinity, high-capacity transporter located in the proximal convoluted tubule, in a process independent of insulin secretion or action.Citation34 Compared with subjects with normal glucose tolerance, patients with T2D have increased renal glucose uptake and release in the postprandial state.Citation35 As the threshold for glucose reabsorption increases, uptake continues despite elevated plasma glucose concentrations, minimizing glucosuria and exacerbating the hyperglycemic environment.Citation36 In addition, the kidneys also contribute to glucose release through increased gluconeogenic activity.Citation14

Brain

The nervous system also plays an important role in metabolic regulation. Through direct and indirect pathways, the nervous system controls glucose metabolism, affecting insulin and glucagon secretion and HGP.Citation2 The hypothalamus, in particular, is an important mediator of β-cell function and plasma insulin concentrations.Citation2,Citation37 Insulin action at the hypothalamus also plays a critical role in regulating appetite and body weight,Citation2,Citation35 and high-fat diets have been shown to lead to inflammatory signaling in the hypothalamus and subsequent insulin and leptin resistance.Citation38 Decreased activity at this site is associated with the development of obesity, which is consistent with imaging studies showing structural changes (increased gliosis) in the hypothalamus of obese subjects.Citation2,Citation39 In preclinical studies, disruptions in circadian rhythms were associated with the development of insulin resistance and obesity,Citation40 as well as an increase in β-cell dysfunction and apoptosis.Citation41

It is apparent that continuous interactions between β-cells and insulin-sensitive tissues and input from other essential organs, such as the brain, kidney, and gastrointestinal tract, collectively ensure an adequate supply of glucose to tissues and organs while keeping plasma glucose concentrations within a specific range to prevent hypoglycemia or hyperglycemia. However, disruptions in these interactions by genetic or environmental triggers, such as inherited insulin resistance, β-cell dysfunction, or obesity, lead to a series of interconnected disturbances that contribute to the development and progression of T2D.

Actions of commonly used antidiabetes medications

New approaches to targeted pharmacotherapy have been developed in response to a better understanding of the underlying pathophysiology of T2D. Knowledge about the mechanisms of action of the more traditional agents has also increased in recent years. summarizes the primary actions and key clinical effects of each class of commonly used antidiabetes medications.

Table 1 Overview of antidiabetes medications

As recognized by the AACE, many patients will not reach glycemic goals with monotherapy and will require a combination of antidiabetes medications.Citation3 However, association recommendations are generally not prescriptive with regard to which treatments to use in combination; rather, individualized treatment is emphasized. Current knowledge provides a sensible rationale for choosing medications that act in a synergistic manner (eg, improved insulin sensitivity and increased insulin secretion) to control glycemia, minimize adverse effects, including hypoglycemia, and correct underlying pathologies.

Suppression of HGP

The primary action of metformin is as an agonist of 5-adenosine monophosphate-activated protein kinase, which reduces HGP by inhibiting gluconeogenesis in the liver. Additionally, metformin yields improved hepatic insulin sensitivity and decreased fasting plasma insulin concentrations.Citation10,Citation42 Data also indicate that metformin has a beneficial effect on lipid metabolism secondary to improved insulin sensitivity and may help reverse hepatic steatosis,Citation43,Citation44 a condition closely associated with insulin resistance.Citation42 Because 5-adenosine monophosphate-activated protein kinase actions also lead to reduced growth of human cancer cells, an antitumorigenic role has also been suggested for metformin,Citation45 which is supported by a meta-analysis demonstrating a significant 50% reduction in hepatocellular carcinoma incidence in patients with diabetes receiving metformin versus not receiving metformin.Citation46

Insulin sensitizers

Thiazolidinediones (TZDs), synthetic peroxisome proliferator-activated receptor-γ agonists,Citation47 are potent insulin sensitizers in the liver, muscle, and adipose tissueCitation48Citation50; evidence has shown TZDs to additionally alleviate lipotoxicity by transporting lipid deposits out of muscle, liver, and β-cells.Citation10 Similar to that of metformin, the insulin-sensitizing effects of TZDs may counteract hepatic steatosis.Citation44

Insulin secretagogues

Sulfonylureas (SUs) primarily increase insulin secretion by binding to the adenosine triphosphate-sensitive potassium (KATP) channel on pancreatic β-cell plasma membranes.Citation51Citation53 Specifically, SUs bind to the SU receptor 1, a subunit on the KATP channel, and induce closure.Citation53 As a result, concentrations of intracellular potassium rise, the β-cell membrane is depolarized, and insulin-containing secretory granules move to the cell surface for release into circulation.Citation53,Citation54

In contrast to the actions of SUs, the actions of incretin-based therapies are glucose-dependent.Citation55 Incretin mimetics, or GLP-1 receptor agonists (GLP-1RAs), primarily increase insulin secretion and decrease glucagon secretion by activating the GLP-1 receptor.Citation30,Citation31 Delayed gastric emptying has also been associated with GLP-1RAs and shown to occur within minutes of administration.Citation29 It is likely that this effect, as well as enhanced satiety and reduced food intake, involves interactions between the central and peripheral nervous systems.Citation29

It is estimated that the half-life of biologically active GLP-1 in circulation is less than 2 minutes because of rapid degradation by DPP-4 activity.Citation56,Citation57 By inhibiting the DPP-4 enzyme and thereby increasing the half-life of endogenous incretin hormones in circulation, DPP-4 inhibitors i ndirectly increase stimulation of GLP-1 receptors.Citation58 DPP-4 inhibitors are rapidly absorbed following oral administration and competitively and reversibly bind to the active site of DPP-4, reducing DPP-4 activity by 70% to 90% and, consequently, increasing GLP-1 concentrations approximately 1.5- to 4-fold at therapeutic doses.Citation59 Preclinical and clinical evidence has demonstrated the benefits of DPP-4 pharmacologic inhibition, including increased concentrations of GIP and GLP-1, stimulation of insulin in circulation, improved glucose tolerance and insulin sensitivity, suppressed plasma glucagon, and prevention of weight gain.Citation29 The clinical efficacy of DPP-4 inhibitors has been demonstrated in randomized controlled trials showing significant reductions from baseline in fasting plasma glucose, postprandial glucose, and A1C.Citation59 DPP-4 inhibitors typically provide mean A1C reductions ranging from approximately 0.4% to 1.0% as monotherapy and from 0.7% to 1.2% in combination therapy regimens.Citation59

Insulin independent agents

SGLT-2 inhibitors reduce elevated glucose concentrations through increased glucosuria by decreasing renal glucose reabsorption.Citation34 The mechanism of action of SGLT-2 inhibitors is independent of insulin and may therefore complement a variety of other antidiabetes medications that are insulin dependent.Citation34 It is also theorized that because SGLT-2 inhibitors target renal glucose handling, efficacy should not be compromised as further declines in β-cell function and increased insulin resistance occur.Citation34

The α-glucosidase inhibitors (AGIs) primarily reduce postprandial plasma glucose concentrations through the competitive and reversible inhibition of α-glucosidase located in the brush border of the small intestine.Citation60 Following nutrient ingestion, α-glucosidase hydrolyzes complex carbohydrates to monosaccharides for absorption. Thus, AGIs obstruct the degradation of starches and sucrose and delay carbohydrate reabsorption in a nonsystemic manner.Citation60 However, a patient’s diet may alter efficacy. Studies indicate that AGIs may have a more potent effect in patients who consume a typical Eastern diet (whole grains, vegetables, fruits) compared with patients whose diet is characterized as Western (processed meat, high-fat dairy products, refined grains).Citation60,Citation61

Central nervous system

The development of therapies that target the central nervous system to reduce glucose concentrations is challenging.Citation2 Bromocriptine, a quick-release sympatholytic D2-dopamine agonist, is currently the only approved antidiabetes medication that has a centrally mediated mechanism of action.Citation2 Although the mechanism by which bromocriptine lowers plasma glucose concentrations is unknown, it is believed that early-morning treatment with bromocriptine resets circadian rhythms of hypothalamic dopamine and serotonin and thereby improves insulin sensitivity.Citation62

Bile acid sequestrants

Bile acid sequestrants were initially developed to treat hypercholesterolemia; therefore, these agents have significant lipid-lowering properties.Citation63 The mechanism by which colesevelam reduces plasma glucose concentrations is not fully understood, although study findings suggest that colesevelam reduces hyperglycemia by improving tissue glucose metabolism in both the fasting (increased plasma glucose clearance) and postprandial (increased glycolytic disposal of oral glucose) states.Citation64 Colesevelam has also been associated with increased GLP-1 and GIP plasma concentrations, which likely contribute to observed improvements in β-cell function and increased glycolytic disposal.Citation64

The American Diabetes Association currently does not favor the use of AGIs, colesevelam, or bromocriptine because of modest effects on glucose lowering, frequent dosing schedules, and occurrence of side effects.Citation51 Therefore, discussion of the use of these agents in combination is limited in this review.

Basal insulin

Many patients will eventually require insulin to maintain glycemic control.Citation51 The primary mechanism of action of insulin and its analogs is the activation of insulin receptors.Citation51 Thus, insulin analogs lower plasma glucose concentrations by stimulating peripheral glucose uptake and inhibiting HGP.Citation65,Citation66 Although various formulations of insulin are available, guidance from the AACE recommends initial insulin therapy with long-acting basal insulin analogs because basal formulations are associated with less glycemic variability and a lower risk for hypoglycemia compared with neutral protamine Hagedorn, an intermediate-acting insulin.Citation3 Several types of long-acting basal insulin analogs are available in the US, including insulin glargine (Lantus® [Sanofi-Aventis, Bridgewater, NJ, USA] and Toujeo® [Sanofi-Aventis]), insulin detemir (Levemir® [Novo Nordisk, Bagsværd, Denmark]), and insulin degludec (Tresiba® [Novo Nordisk]).Citation51,Citation66,Citation67 Although both formulations of insulin glargine provide prolonged activity and thus allow once-daily dosing, Toujeo is a concentrated glargine, and its duration of action exceeds that of Lantus (approximately 36 vs 24 hours).Citation68 In addition, Toujeo has been shown to significantly reduce nighttime low blood sugar events compared with Lantus (P=0.0002).Citation69 Insulin detemir or Levemir is also associated with a constant duration of action, but similar to that of Lantus (approximately 24 hours), and may be administered once or twice daily in divided doses.Citation66 Of the currently available long-acting basal insulin analogs, Tresiba provides the longest duration of effect, lasting at least 42 hours, primarily because of delayed absorption of insulin degludec from the subcutaneous tissue into systemic circulation.Citation67

Potential combinations to improve pathophysiologic dysfunction

Enhancing endogenous insulin action

There are many combination regimens that have the potential to address multiple sites of pathophysiologic dysfunction of T2D, and those that may exert complementary effects are outlined in . The combination of metformin and a TZD has been suggestedCitation10 because metformin reduces HGP by inhibiting gluconeogenesis in the liver but is only a weak sensitizer in the muscle,Citation42,Citation70,Citation71 and TZDs are potent insulin sensitizers in liver, muscle, and adipose tissue.Citation48 Metformin and TZDs individually have a robust effect on A1C, and each exerts its actions through different pathways, providing an additive effect on glucose lowering.Citation10 Metformin and TZDs are also associated with a low risk for hypoglycemia,Citation3 making these agents appealing for use in combination. However, because metformin primarily relies on renal function for elimination,Citation72 it is contraindicated in patients with renal disease or dysfunction.Citation73 Metformin is also associated with gastrointestinal adverse effects that may lead to discontinuation for some patients but may dissipate with continued use or with dosing adjustments.Citation74 TZDs have been shown to have beneficial effects on cognitive disorders related to insulin resistance in the central nervous system in studies in miceCitation75 and to exert favorable effects on cardiovascular (CV) risk markers in patients with T2D.Citation76 However, TZDs are associated with an increased risk for weight gain and fluid retention, which may worsen or lead to congestive heart failure (HF).Citation77,Citation78

Table 2 Potential regimens for targeted combination therapy to address major pathophysiologic defectsTable Footnotea

Insulin secretagogues, such as SUs or incretin-based therapies, may also act in a complementary manner to metformin by facilitating insulin secretion. SUs are associated with moderate glycemic efficacy, although these agents are recommended for patients with a disease duration of less than 5 yearsCitation55 because the mechanism of action is dependent on functioning β-cells.Citation53 In addition, because SUs act independently of plasma glucose concentrations, the main risk is hypoglycemia, which may be exacerbated by coadministration with other drugs, especially those that inhibit SU metabolism via the cytochrome P450 2C9 isozyme.Citation79 To avoid hypoglycemia, the addition of a DPP-4 inhibitor or a GLP-1RA to metformin may be considered instead. Although GLP-1RAs are associated with a more robust effect on A1C lowering compared with DPP-4 inhibitors,Citation80 each has been shown to improve glycemic control without increasing the risk for hypoglycemia.Citation51 GLP-1RAs are also associated with gastrointestinal side effects, particularly transient nausea, which may be attributed to delayed gastric emptying.Citation80 Preclinical and clinical studies also demonstrate additional complementary actions between metformin and a DPP-4 inhibitor or a GLP-1RA. In vivo, metformin increased circulating concentrations of active GLP-1 but did not affect the activity of the DPP-4 enzyme.Citation81 Thus, as demonstrated in healthy subjects, metformin and a DPP-4 inhibitor increased GLP-1 concentrations through two separate pathways, providing an additive increase in GLP-1 concentrations.Citation82 Similar benefits were observed with metformin in combination with exogenous GLP-1 administration in patients with T2D, although findings in this study suggested that metformin enhanced injected active GLP-1 concentrations by inhibiting DPP-4 activity.Citation83

Preserving β-cell function

Maintaining β-cell function is a key aspect in delaying disease progression,Citation84 and as noted above, the efficacy of drugs that depend on β-cell function for their actions, such as the SUs, declines as β-cell function declines.Citation10 Furthermore, results of in vitro studies have suggested that prolonged exposure to SUs may in fact cause disturbances in islet cell functionCitation85 and may induce β-cell apoptosis as a secondary effect to the closure of the KATP channel.Citation86

In contrast, substantial evidence has demonstrated that TZDs positively affect β-cells by improving insulin secretory capacity and function, preserving structure and mass, and exerting protective effects from oxidative stress.Citation87 Preclinical studies have also shown that GLP-1RAs and DPP-4 inhibitors improve or preserve β-cell mass by promoting β-cell proliferation and inhibiting apoptosis.Citation30,Citation88 Clinical trials in patients with T2D further demonstrate that GLP-1RAs preserve β-cell function and sensitivity to glucose with a durable effect,Citation89,Citation90 and DPP-4 inhibitors sustain β-cell function during 2 years of treatment.Citation91 More recently, SGLT-2 inhibitors have been shown to improve β-cell function in patients with T2D, despite having no direct effect on pancreatic β-cells, through the reversal of glucotoxicity and improved insulin sensitivity.Citation92 Therefore, the combination of a TZD with a DPP-4 inhibitor, GLP-1RA, or SGLT-2 inhibitor may provide both insulin-sensitizing and insulin secretory effects, in addition to helping preserve β-cell function.

Promoting weight loss

Treatment-related effects on weight are also important to consider because weight gain can worsen insulin resistance and thereby diminish efficacy.Citation89 TZDs are associated with weight gain, in part due to fluid retention and fat cell proliferation,Citation93 as are SUs, as a secondary effect on adipocyte KATP channels.Citation94 Metformin is associated with weight loss or is weight neutral and has been shown to maintain efficacy across all body weight categories, including patients who are overweight or obese,Citation55 providing a rationale for use in combination. Because AGIs slow intestinal carbohydrate digestion and absorption,Citation51 weight gain is not expected, and treatment has been shown to minimize weight gain when used in combination with SUs.Citation95 However, because AGIs have limited potency in lowering A1C and are also commonly associated with gastrointestinal side effects, their use in combination regimens may not always be suitable.Citation3

For patients in whom weight loss is difficult, the use of a GLP-1RA with an SGLT-2 inhibitor may be particularly beneficial because GLP-1RAs slow gastric emptying, increase satiety, and reduce food intake,Citation29 and SGLT-2 inhibitors decrease weight through increased caloric loss via glucosuria.Citation3 Additionally, GLP-1RA s and SGLT-2 inhibitors reduce blood pressure in patients with T2D, possibly as a secondary effect of weight loss and other factors, such as the mild osmotic diuretic effect of the SGLT-2 inhibitors.Citation34,Citation96 In a recent retrospective analysis, patients had a two-fold greater weight loss with GLP-1RA and SGLT-2 inhibitor combination therapy (−7.2 kg) compared with patients taking an SGLT-2 inhibitor alone (−3.0 kg).Citation97 Because the actions of GLP-1RAs are distinct from those of SGLT-2 inhibitors, the reduction in weight is likely additive when used in combination. DPP-4 inhibitors may also complement SGLT-2 inhibitors in this regard. Inhibition of the DPP-4 enzyme is associated with prevention of weight gain,Citation29 and clinical study findings have shown an initial combination therapy with a DPP-4 inhibitor and an SGLT-2 inhibitor in treatment-naïve patients to significantly reduce weight compared with a DPP-4 inhibitor administered alone.Citation98

Triple oral combination therapy

Much of the previous discussion has focused on potential dual therapy combinations; however, some patients with high A1C may require initial triple oral pharmacotherapy or intensified treatment with a third agent if dual therapy regimens do not provide adequate glycemic control. In patients with more recent disease, triple oral therapy with metformin, an SU, and a DPP-4 inhibitor may help address key disease aspects, including decreased insulin secretion and increased HGP. SUs in this population may also provide greater efficacy because β-cell function may still be intact. In patients with inadequate glycemic control despite metformin and SU dual therapy, the addition of a DPP-4 inhibitor, an SGLT-2 inhibitor, or a TZD has been shown to improve glycemic control.Citation99,Citation100 However, as previously mentioned, SUs increase the risk for hypoglycemia,Citation3 making a TZD in combination with metformin and a DPP-4 inhibitor or an SGLT-2 inhibitor an alternative choice if avoiding hypoglycemia is a primary treatment goal.Citation101,Citation102 TZDs in these combinations should also provide additive benefits on β-cell function and mass, although adverse effects on body weight may limit use. Triple therapy with metformin, a DPP-4 inhibitor, and an SGLT-2 inhibitor has also been shown to be efficacious in reducing A1C without increasing the risk for hypoglycemia or weight gain.Citation103Citation107 Based on the respective mechanisms of action, this particular combination allows improved insulin sensitivity and enhanced insulin production, with additional clinical benefits on weight and blood pressure. In addition, SGLT-2 inhibitors are associated with a rise in fasting plasma glucagon concentrations and endogenous glucose production, which could limit efficacy.Citation108,Citation109 However, preclinical studies have shown the suppressive effects of metformin on HGP to counteract the rise in endogenous glucose production and thereby enhance the glucose-lowering properties of SGLT-2 inhibitors.Citation110 DPP-4 inhibitors, which suppress glucagon secretion and therefore inhibit endogenous glucose production,Citation111 have been shown to reduce SGLT-2 inhibitor-induced increases in glucagon when used concomitantly.Citation112

Combination with basal insulin

In general, the initiation of insulin should be considered for patients with initial A1C >9% and symptomatic hyperglycemia or for patients with A1C >8.0% despite dual therapy and/or long-standing T2D, because adding a third antidiabetes drug in this scenario will be less likely to achieve glycemic targets ().Citation3 Although previous guidance has suggested delaying the initiation of insulin therapy until after several rounds of treatment failure, more recent recommendations suggest initiating insulin earlier, often in combination with DPP-4 inhibitors, SGLT-2 inhibitors, or GLP-1RAs.Citation3 Research also supports the early use of insulin to protect β-cell function through the rapid reversal of glucolipotoxicity.Citation113

Weight gain is common with insulin therapy and has been attributed to the correction of severe hyperglycemia leading to rebuilding of muscle and fat mass, as well as increased caloric retention resulting from decreased glucosuria.Citation93 In addition, patients experiencing hypoglycemia may increase their food intake.Citation93 For patients requiring increasing insulin doses, TZDs have been suggested as adjunct therapy to help reduce insulin daily doses and establish glycemic control.Citation51 Although TZDs may additionally help to preserve β-cell function,Citation87 weight gain and fluid retention associated with TZD therapy may be exacerbated with concomitant insulin use,Citation114 limiting use in some patients. In patients taking SU therapy, it is generally recommended that the dose be reduced or treatment stopped after starting basal insulin.Citation3

In patients with T2D receiving basal insulin, controlled clinical trials support the use of incretin-based therapies or SGLT-2 inhibitors. For example, in patients on background insulin glargine with or without OADs, add-on exenatide for 30 weeks was associated with significantly greater reductions from baseline in A1C (−1.74% vs −1.04%, P<0.001) and weight (−1.8 vs +1.0 kg) versus add-on placebo, with no increased risk for hypoglycemia.Citation115 Additional clinical studies of other GLP-1RAs added to existing basal insulin regimens have demonstrated similar results.Citation116,Citation117 Clinical trials of add-on DPP-4 inhibitor therapy to basal insulin (or other insulin formulations) have also demonstrated significant improvements in A1C without an increased risk for weight gain or hypoglycemia versus controls.Citation118Citation121 Importantly, findings observed from clinical trials of basal insulin in combination with incretin-based therapies are consistent with those from real-world observational studies.Citation122,Citation123 Available clinical trial data have also shown that the addition of an SGLT-2 inhibitor to basal insulin or other formulations (with or without other OADs) effectively reduces A1C and weight and maintains a low risk for hypoglycemia.Citation124Citation126 Of interest, insulin detemir may have a more favorable effect on weight compared with other basal insulin formulations, and patients treated with detemir plus other OADs have been shown to consistently experience weight loss.Citation127,Citation128

Based on clinical trial findings, the AACE suggests a DPP-4 inhibitor, GLP-1RA, or SGLT-2 inhibitor in combination with basal insulin to decrease basal and postprandial glucose and weight gain, without increasing the risk for hypoglycemia and weight gain.Citation3 With more complex insulin regimens, SGLT-2 inhibitors are also suggested to reduce the amount of insulin needed.Citation51 Consistently, clinical trials have shown that patients treated with GLP-1RAs, DPP-4 inhibitors, and SGLT-2 inhibitors in combination with insulin (basal and other formulations) require fewer increases in daily insulin doses compared with controls.Citation115,Citation116,Citation118,Citation119,Citation124Citation126 DPP-4 inhibitors and SGLT-2 inhibitors have the advantage of oral administration; however, because patients are already receiving insulin injections, the route of administration may not be a primary factor in the treatment selection.Citation129 In addition, injection pens with single administration of combination basal insulin and GLP-1RA (insulin degludec and liraglutide and insulin glargine and lixisenatide fixed ratio) are currently in development.Citation130,Citation131

More recently, there has been concern about whether SGLT-2 inhibitors are associated with an increased risk of diabetic ketoacidosis (DKA), particularly in patients with a recent reduction in concomitant insulin dose.Citation132 Based on postmarketing reports of 73 cases in patients treated with SGLT-2 inhibitors (March 2013–June 2015), the US Food and Drug Administration required labeling of SGLT-2 inhibitors to include warnings of an increased risk of DKA.Citation133 More expansive reviews of data have shown that most patients had only slightly elevated plasma glucose concentrations (euglycemic DKA), and some cases occurred in patients with type 1 diabetes.Citation132 In addition, data from the clinical development programs for the three US-approved SGLT-2 inhibitors have shown no clear association between DKA and SGLT-2 inhibitor treatment.Citation132 For example, in the EMPA-REG OUTCOME trial in 7,020 patients, the incidence of DKA was ≤0.1% and did not differ between empagliflozin and placebo groups.Citation134 A similar incidence rate of <0.1% for DKA was also reported from the clinical development programs for canagliflozin (N=17,596) and dapagliflozin (N>18,000).Citation132,Citation135 The AACE has also undertaken a rigorous examination of this potential risk and concluded that the occurrence of DKA in treated patients is infrequent, and the risk–benefit ratio remains in favor of continued SGLT-2 inhibitor use, with no changes in current recommendations required.Citation136 Regulatory agencies and pharmaceutical companies will continue to monitor this potential risk, and attention to risk factors for euglycemic DKA, such as reductions in concomitant insulin doses, has been advised when treating patients with SGLT-2 inhibitors.Citation132

CV safety

The effect of antidiabetes therapies on CV outcomes remains a major concern. Research has reported mixed findings as to whether SUs or insulin are associated with an increased risk for CV events.Citation137Citation140 Prescribing information for glipizide, glyburide, and glimepiride includes a warning of an increased risk of CV mortality with the first-generation SU, tolbutamide, versus insulin as add-on to diet or diet alone.Citation141Citation143 Although the use of SUs in patients with CV risk appears to be declining in clinical practice,Citation144 recent findings of an increased risk for mortality and CV events with treatment warrant continued caution.Citation145,Citation146 For example, initial SU monotherapy in a German cohort of patients with T2D significantly increased the risk of major CV events, overall mortality, and T2D-related hospitalization compared with initial metformin monotherapy (all P<0.001).Citation146 Findings from a systematic review additionally found an increased risk of all-cause mortality and CV-related mortality with SUs compared with other antidiabetes drugs and a significantly higher risk of myocardial infarction (vs DPP-4 inhibitors) in randomized clinical trials, which was also confirmed in observational studies.Citation145 Further, the risk of stroke was significantly higher in patients treated with SUs in clinical trials versus those treated with DPP-4 inhibitors, GLP-1RAs, TZDs, or insulin.Citation145

TZDs have been associated with HF, attributed to treatment-induced fluid retention.Citation3,Citation51 These agents are therefore contraindicated in patients with preexisting HF, and the prescribing information for pioglitazone and rosiglitazone contains warnings for congestive HF.Citation77,Citation78 Although there is not yet conclusive evidence to establish CV benefit with this treatment,Citation147 fewer patients with T2D had a CV disease event with bromocriptine quick-release as add-on to standard of care compared with placebo in a 1-year trial.Citation148

CV outcome trials

Findings have been published from three CV outcome trials for DPP-4 inhibitors showing no increased risk for major CV events with alogliptin, saxagliptin, or sitagliptin, respectively, versus placebo as add-on to standard of care.Citation149Citation151 However, a potential for an increased risk of hospitalization for HF versus placebo was observed in patients treated with saxagliptinCitation149 and in a subgroup of patients with no history of HF at baseline treated with alogliptin.Citation152 A recent observational cohort study using data from a US insurance claims database evaluated the risk of hospitalization for HF between patients with T2D treated with DPP-4 inhibitors or an SU and between patients treated with saxagliptin or sitagliptin.Citation153 In patients with no baseline CV disease, DPP-4 inhibitors were associated with a significantly lower risk of hospitalization for HF compared with those treated with an SU (P=0.013). The risk of hospitalization for HF between saxagliptin and sitagliptin in patients with or without baseline CV disease and between DPP-4 inhibitors and SUs in patients with baseline CV disease was not significantly different.Citation153 Additional CV outcome studies for linagliptin are ongoing.Citation154,Citation155 Until additional findings are known, it is recommended that DPP-4 inhibitors be used with caution, or not at all, in patients with preexisting HF.Citation4

In the EMPA-REG OUTCOME CV outcomes trial in patients with T2D and established CV disease, empagliflozin as add-on to standard of care was associated with significantly lower rates of major CV events (ie, death from CV causes, nonfatal myocardial infarction, nonfatal stroke), primarily driven by a significant reduction in death from CV causes, as well as death from any cause and hospitalization for HF compared with add-on placebo.Citation134 CV outcome trials for dapagliflozin and canagliflozin are underway and are expected to be completed in 2017 (canagliflozin) and 2019 (dapagliflozin). Each trial includes patients with T2D at high risk for CV events.Citation156,Citation157 Most of the GLP-1RA CV trials are ongoing, although one study for lixisenatide recently reported preliminary findings.Citation158 In this study, no increased risk for major CV events was observed in patients with a recent acute coronary syndrome event treated with lixisenatide versus placebo.Citation158

Conclusion

The multitude of therapeutic options for T2D adds to the complexity of managing patients with this disease but also allows clinicians to readjust and rebalance treatment over time as physiologic changes occur throughout the course of the disease. As understanding of the pathophysiology of T2D grows, it is becoming apparent that early use of targeted combination therapy has the potential to slow disease progression, potentially through the correction of known pathologies and the preservation of β-cell function. Obesity plays a central role in the development and worsening of T2D, and thus it can be argued that treatments that facilitate weight loss may be most beneficial. Whereas the underlying pathophysiology of T2D should be a key factor in choosing which medications to combine, the treatment choice must also be individualized to patients’ needs and tolerability. Based on individual therapeutic profiles, the newer incretin-based therapies and SGLT-2 inhibitors are appropriate for use in combination with one another or in combination with more traditional agents (eg, metformin, TZDs, insulin) to reduce A1C without increasing the risk of hypoglycemia. In addition, because these newer agents act on different pathways, the beneficial actions on glycemia and weight are likely additive when used in combination. Although clinical inertia remains an issue, emerging research helps to support and advocate for the use of early combination therapy as a practical approach to preempt the development of long-term complications and disease progression.

Acknowledgments

Medical writing support for the preparation of this manuscript was provided by Meg Shurak, MS, and Janet E Matsuura, PhD, from Complete Healthcare Communications, LLC (Chadds Ford, PA, USA), with funding from AstraZeneca.

Disclosure

PL has received a research grant and is a speaker and consultant for AstraZeneca, Sanofi-Aventis, LLC, and Novo Nordisk, Inc. He has also received a research grant and is a speaker for Eli Lilly and Company and Boehringer Ingelheim, GmbH; is a speaker and consultant for Janssen Pharmaceuticals, Inc.; and is a speaker for GlaxoSmithKline, plc. The author reports no other conflicts of interest in this work.

References

  • International Diabetes Federation, Global burden Sixth edition 2014 Available from: http://www.idf.org/diabetesatlas/5e/the-global-burden Accessed February 9, 2015
  • Kahn SE Cooper ME Del Prato S Pathophysiology and treatment of type 2 diabetes: perspectives on the past, present, and future Lancet 2014 383 9922 1068 1083 24315620
  • Garber AJ Abrahamson MJ Barzilay JI Consensus statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the comprehensive type 2 diabetes management algorithm - 2016 executive summary Endocr Pract 2016 22 1 84 113 26731084
  • American Diabetes Association 7. Approaches to glycemic treatment Diabetes Care 2016 39 Suppl 1 S52 S59 26696682
  • Berkowitz SA Krumme AA Avorn J Initial choice of oral glucose-lowering medication for diabetes mellitus: a patient-centered comparative effectiveness study JAMA Intern Med 2014 174 12 1955 1962 25347323
  • Khunti K Wolden ML Thorsted BL Andersen M Davies MJ Clinical inertia in people with type 2 diabetes: a retrospective cohort study of more than 80,000 people Diabetes Care 2013 36 11 3411 3417 23877982
  • Levin PA Wei W Zhou S Xie L Baser O Outcomes and treatment patterns of adding a third agent to 2 OADs in patients with type 2 diabetes J Manag Care Spec Pharm 2014 20 5 501 512 24761822
  • Brown JB Nichols GA Perry A The burden of treatment failure in type 2 diabetes Diabetes Care 2004 27 7 1535 1540 15220224
  • Garber AJ Abrahamson MJ Barzilay JI American Association of Clinical Endocrinologists’ comprehensive diabetes management algorithm 2013 consensus statement–executive summary Endocr Pract 2013 19 3 536 557 23816937
  • DeFronzo RA Banting Lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus Diabetes 2009 58 4 773 795 19336687
  • Triplitt CL Examining the mechanisms of glucose regulation Am J Manag Care 2012 18 1 suppl S4 S10 22559855
  • Ravnskjaer K Hogan MF Lackey D Glucagon regulates gluconeogenesis through KAT2B- and WDR5-mediated epigenetic effects J Clin Invest 2013 123 10 4318 4328 24051374
  • Triplitt C Understanding the kidney’s role in blood glucose regulation Am J Manag Care 2012 18 S11 S16 22559853
  • Gerich JE Meyer C Woerle HJ Stumvoll M Renal gluconeogenesis: its importance in human glucose homeostasis Diabetes Care 2001 24 2 382 391 11213896
  • Basu R Chandramouli V Dicke B Landau B Rizza R Obesity and type 2 diabetes impair insulin-induced suppression of glycogenolysis as well as gluconeogenesis Diabetes 2005 54 7 1942 1948 15983193
  • Barthel A Schmoll D Novel concepts in insulin regulation of hepatic gluconeogenesis Am J Physiol Endocrinol Metab 2003 285 4 E685 E692 12959935
  • Guilherme A Virbasius JV Puri V Czech MP Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes Nat Rev Mol Cell Biol 2008 9 5 367 377 18401346
  • Wellen KE Hotamisligil GS Inflammation, stress, and diabetes J Clin Invest 2005 115 5 1111 1119 15864338
  • Wensveen FM Valentic S Sestan M Turk Wensveen T Polic B The “Big Bang” in obese fat: Events initiating obesity-induced adipose tissue inflammation Eur J Immunol 2015 45 9 2446 2456 26220361
  • Xu H Barnes GT Yang Q Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance J Clin Invest 2003 112 12 1821 1830 14679177
  • Weisberg SP McCann D Desai M Rosenbaum M Leibel RL Ferrante AWJr Obesity is associated with macrophage accumulation in adipose tissue J Clin Invest 2003 112 12 1796 1808 14679176
  • Poitout V Robertson RP Glucolipotoxicity: fuel excess and beta-cell dysfunction Endocr Rev 2008 29 3 351 366 18048763
  • Russo GT Giorda CB Cercone S Nicolucci A Cucinotta D BetaDecline Study Group Factors associated with beta-cell dysfunction in type 2 diabetes: the BETADECLINE study PLoS One 2014 9 10 e109702 25347846
  • DeFronzo RA Tripathy D Skeletal muscle insulin resistance is the primary defect in type 2 diabetes Diabetes Care 2009 32 suppl 2 S157 S163 19875544
  • Cnop M Vidal J Hull RL Progressive loss of beta-cell function leads to worsening glucose tolerance in first-degree relatives of subjects with type 2 diabetes Diabetes Care 2007 30 3 677 682 17327340
  • Hu FB van Dam RM Liu S Diet and risk of Type II diabetes: the role of types of fat and carbohydrate Diabetologia 2001 44 7 805 817 11508264
  • Guenard F Deshaies Y Cianflone K Kral JG Marceau P Vohl MC Differential methylation in glucoregulatory genes of offspring born before vs. after maternal gastrointestinal bypass surgery Proc Natl Acad Sci U S A 2013 110 28 11439 11444 23716672
  • Nauck MA Baller B Meier JJ Gastric inhibitory polypeptide and glucagon-like peptide-1 in the pathogenesis of type 2 diabetes Diabetes 2004 53 suppl 3 S190 S196
  • Drucker DJ The biology of incretin hormones Cell Metab 2006 3 3 153 165 16517403
  • Drucker DJ Nauck MA The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes Lancet 2006 368 9548 1696 1705 17098089
  • Drucker DJ Sherman SI Gorelick FS Bergenstal RM Sherwin RS Buse JB Incretin-based therapies for the treatment of type 2 diabetes: evaluation of the risks and benefits Diabetes Care 2010 33 2 428 433 20103558
  • Nauck MA Heimesaat MM Orskov C Holst JJ Ebert R Creutzfeldt W Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus J Clin Invest 1993 91 1 301 307 8423228
  • Nauck MA Vardarli I Deacon CF Holst JJ Meier JJ Secretion of glucagon-like peptide-1 (GLP-1) in type 2 diabetes: what is up, what is down? Diabetologia 2011 54 1 10 18 20871975
  • Nauck MA Update on developments with SGLT2 inhibitors in the management of type 2 diabetes Drug Des Devel Ther 2014 8 1335 1380
  • Meyer C Woerle HJ Dostou JM Welle SL Gerich JE Abnormal renal, hepatic, and muscle glucose metabolism following glucose ingestion in type 2 diabetes Am J Physiol Endocrinol Metab 2004 287 6 E1049 E1056 15304374
  • Abdul-Ghani MA Norton L Defronzo RA Role of sodium-glucose cotransporter 2 (SGLT 2) inhibitors in the treatment of type 2 diabetes Endocr Rev 2011 32 4 515 531 21606218
  • Murphy KG Bloom SR Gut hormones in the control of appetite Exp Physiol 2004 89 5 507 516 15184357
  • Thaler JP Schwartz MW Minireview: Inflammation and obesity pathogenesis: the hypothalamus heats up Endocrinology 2010 151 9 4109 4115 20573720
  • Thaler JP Yi CX Schur EA Obesity is associated with hypothalamic injury in rodents and humans J Clin Invest 2012 122 1 153 162 22201683
  • Shi SQ Ansari TS McGuinness OP Wasserman DH Johnson CH Circadian disruption leads to insulin resistance and obesity Curr Biol 2013 23 5 372 381 23434278
  • Gale JE Cox HI Qian J Block GD Colwell CS Matveyenko AV Disruption of circadian rhythms accelerates development of diabetes through pancreatic beta-cell loss and dysfunction J Biol Rhythms 2011 26 5 423 433 21921296
  • Viollet B Guigas B Sanz Garcia N Leclerc J Foretz M Andreelli F Cellular and molecular mechanisms of metformin: an overview Clin Sci (Lond) 2012 122 6 253 270 22117616
  • Wang CC Lin SK Tseng YF Elevation of serum aminotransferase activity increases risk of carotid atherosclerosis in patients with non-alcoholic fatty liver disease J Gastroenterol Hepatol 2009 24 8 1411 1416 19702910
  • Idilman R Mizrak D Corapcioglu D Clinical trial: insulin-sensitizing agents may reduce consequences of insulin resistance in individuals with non-alcoholic steatohepatitis Aliment Pharmacol Ther 2008 28 2 200 208 18445142
  • Facciorusso A The influence of diabetes in the pathogenesis and the clinical course of hepatocellular carcinoma: recent findings and new perspectives Curr Diabetes Rev 2013 9 5 382 386 23845075
  • Singh S Singh PP Singh AG Murad MH Sanchez W Anti-diabetic medications and the risk of hepatocellular cancer: a systematic review and meta-analysis Am J Gastroenterol 2013 108 6 881 891 quiz 892 23381014
  • Zhao D Shi Z Warriner AH Molecular mechanism of thiazolidinedione-mediated inhibitory effects on osteoclastogenesis PLoS One 2014 9 7 e102706 25032991
  • Miyazaki Y Mahankali A Matsuda M Improved glycemic control and enhanced insulin sensitivity in type 2 diabetic subjects treated with pioglitazone Diabetes Care 2001 24 4 710 719 11315836
  • Miyazaki Y Matsuda M DeFronzo RA Dose-response effect of pioglitazone on insulin sensitivity and insulin secretion in type 2 diabetes Diabetes Care 2002 25 3 517 523 11874940
  • Miyazaki Y Glass L Triplitt C Effect of rosiglitazone on glucose and non-esterified fatty acid metabolism in Type II diabetic patients Diabetologia 2001 44 12 2210 2219 11793023
  • American Diabetes Association Standards of Medical Care in Diabetes-2016 Diabetes Care 2016 39 Suppl 1 S1 S119 26696671
  • Proks P Reimann F Green N Gribble F Ashcroft F Sulfonylurea stimulation of insulin secretion Diabetes 2002 51 suppl 3 S368 S376 12475777
  • Aquilante CL Sulfonylurea pharmacogenomics in type 2 diabetes: the influence of drug target and diabetes risk polymorphisms Expert Rev Cardiovasc Ther 2010 8 3 359 372 20222815
  • Reis AF Velho G Sulfonylurea receptor -1 (SUR1): genetic and metabolic evidences for a role in the susceptibility to type 2 diabetes mellitus Diabetes Metab 2002 28 1 14 19 11938023
  • Handelsman Y Bloomgarden ZT Grunberger G 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
  • Baggio L Drucker DJ Biology of incretins: GLP-1 and GIP Gastroenterology 2007 132 2131 2157 17498508
  • Deacon CF Johnsen AH Holst JJ Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo J Clin Endocrinol Metab 1995 80 3 952 957 7883856
  • Nauck MA El-Ouaghlidi A The therapeutic actions of DPP-IV inhibition are not mediated by glucagon-like peptide-1 Diabetologia 2005 48 4 608 611 15761719
  • Baetta R Corsini A Pharmacology of dipeptidyl peptidase-4 inhibitors: similarities and differences Drugs 2011 71 11 1441 1467 21812507
  • He K Shi JC Mao XM Safety and efficacy of acarbose in the treatment of diabetes in Chinese patients Ther Clin Risk Manag 2014 10 505 511 25061309
  • Zhu Q Tong Y Wu T Li J Tong N Comparison of the hypoglycemic effect of acarbose monotherapy in patients with type 2 diabetes mellitus consuming an Eastern or Western diet: a systematic meta-analysis Clin Ther 2013 35 6 880 899 23602502
  • Holt RI Barnett AH Bailey CJ Bromocriptine: old drug, new formulation and new indication Diabetes Obes Metab 2010 12 12 1048 1057 20977575
  • Handelsman Y Role of bile acid sequestrants in the treatment of type 2 diabetes Diabetes Care 2011 34 suppl 2 S244 S250 21525463
  • Beysen C Murphy EJ Deines K Effect of bile acid sequestrants on glucose metabolism, hepatic de novo lipogenesis, and cholesterol and bile acid kinetics in type 2 diabetes: a randomised controlled study Diabetologia 2012 55 2 432 442 22134839
  • Lantus® (insulin glargine [rDNA origin] injection). [prescribing information] Bridgewater, NJ sanofiaventis 2009
  • Levemir® (insulin detemir) [prescribing information] Bagsvaerd, Denmark Novo Nordisk A/S 2015
  • Tresiba® (insulin degludec injection) [prescribing information] Bagsvaerd, Denmark Novo Nordisk A/S 2015
  • Toujeo® (insulin glargine) [prescribing Information] Summary of Product Characteristics Frankfurt, Germany Sanofi-Aventis Deutschland, GmbH 2015
  • Sanofi Press Release: Sanofi reports positive phase 3 results for Toujeo® (insulin glargine [rDNA origin] injection, 300 U/mL) 2015 Available from: http://www.news.sanofi.us/2014-06-14-Sanofi-Reports-Positive-Phase-3-Results-for-Toujeo-insulin-glargine-rDNA-origin-injection-300-U-mL Accessed December 7, 2015
  • Hallsten K Virtanen KA Lonnqvist F Rosiglitazone but not metformin enhances insulin- and exercise-stimulated skeletal muscle glucose uptake in patients with newly diagnosed type 2 diabetes Diabetes 2002 51 12 3479 3485 12453903
  • Malin SK Gerber R Chipkin SR Braun B Independent and combined effects of exercise training and metformin on insulin sensitivity in individuals with prediabetes Diabetes Care 2012 35 1 131 136 22040838
  • Graham GG Punt J Arora M Clinical pharmacokinetics of metformin Clin Pharmacokinet 2011 50 2 81 98 21241070
  • Glucophage® (metformin hydrochloride) and Glucophage XR® (metformin hydrochloride extended-release) [prescribing information] Princeton, NJ Bristol-Myers Squibb Company 2009
  • Kim CH Han KA Oh HJ Safety, tolerability, and efficacy of metformin extended-release oral antidiabetic therapy in patients with type 2 diabetes: an observational trial in Asia J Diabetes 2012 4 4 395 406 22742083
  • Yu Y Li X Blanchard J Insulin sensitizers improve learning and attenuate tau hyperphosphorylation and neuroinflammation in 3xTg-AD mice J Neural Transm (Vienna) 2015 122 4 593 606 25113171
  • Genovese S De Berardis G Nicolucci A Effect of pioglitazone versus metformin on cardiovascular risk markers in type 2 diabetes Adv Ther 2013 30 2 190 202 23359066
  • Actos® (pioglitazone hydrochloride) [prescribing information] Deerfield, IL Takeda Pharmaceuticals America, Inc 2012
  • Avandia® (rosiglitazone maleate) [prescribing information] Research Triangle Park, NC GlaxoSmithKline 2011
  • Amin M Suksomboon N Pharmacotherapy of type 2 diabetes mellitus: an update on drug-drug interactions Drug Saf 2014 37 11 903 919 25249046
  • Reid T Choosing GLP-1 receptor agonists or DPP-4 inhibitors: weighing the clinical trial evidence Clin Diabetes 2012 30 1 3 12
  • Mulherin AJ Oh AH Kim H Grieco A Lauffer LM Brubaker PL Mechanisms underlying metformin-induced secretion of glucagon-like peptide-1 from the intestinal L cell Endocrinology 2011 152 12 4610 4619 21971158
  • Migoya EM Bergeron R Miller JL Dipeptidyl peptidase-4 inhibitors administered in combination with metformin result in an additive increase in the plasma concentration of active GLP-1 Clin Pharmacol Ther 2010 88 6 801 808 21048706
  • Cuthbertson J Patterson S O’Harte FP Bell PM Addition of metformin to exogenous glucagon-like peptide-1 results in increased serum glucagon-like peptide-1 concentrations and greater glucose lowering in type 2 diabetes mellitus Metabolism 2011 60 1 52 56 20152998
  • Kahn SE Lachin JM Zinman B Effects of rosiglitazone, glyburide, and metformin on beta-cell function and insulin sensitivity in ADOPT Diabetes 2011 60 5 1552 1560 21415383
  • Del Guerra S Marselli L Lupi R Effects of prolonged in vitro exposure to sulphonylureas on the function and survival of human islets J Diabetes Complications 2005 19 1 60 64 15642492
  • Maedler K Carr RD Bosco D Zuellig RA Berney T Donath MY Sulfonylurea induced beta-cell apoptosis in cultured human islets J Clin Endocrinol Metab 2005 90 1 501 506 15483097
  • Campbell IW Mariz S Beta-cell preservation with thiazolidinediones Diabetes Res Clin Pract 2007 76 2 163 176 17052795
  • Vilsboll T The effects of glucagon-like peptide-1 on the beta cell Diabetes Obes Metab 2009 11 suppl 3 11 18 19878257
  • Garber AJ Incretin effects on beta-cell function, replication, and mass: the human perspective Diabetes Care 2011 34 Suppl 2 S258 S263 21525465
  • Garber A Henry R Ratner R Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial Lancet 2009 373 9662 473 481 18819705
  • Leibowitz G Cahn A Bhatt DL Impact of treatment with saxagliptin on glycaemic stability and beta-cell function in the SAVOR-TIMI 53 study Diabetes Obes Metab 2015 17 5 487 494 25656169
  • Merovci A Mari A Solis C Dapagliflozin lowers plasma glucose concentration and improves beta cell function J Clin Endocrinol Metab 2015 100 5 1927 1932 25710563
  • Fonseca V Effect of thiazolidinediones on body weight in patients with diabetes mellitus Am J Med 2003 115 suppl 8A 42S 48S 14678865
  • Shi H Moustaid-Moussa N Wilkison WO Zemel MB Role of the sulfonylurea receptor in regulating human adipocyte metabolism FASEB J 1999 13 13 1833 1838 10506587
  • Acarbose [prescribing information] Morgantown, WV Mylan Pharmaceuticals, Inc 2015
  • Wang B Zhong J Lin H Blood pressure-lowering effects of GLP-1 receptor agonists exenatide and liraglutide: a meta-analysis of clinical trials Diabetes Obes Metab 2013 15 8 737 749 23433305
  • McGovern A Dutta N Watters K Munro N Feher M Additive weight loss effect with a combination of an oral sodium-glucose cotransporter 2 (SGLT2) inhibitor and a glucagon-like peptide 1 (GLP-1) agonist in type 2 diabetes. A6 (P143) Paper presented at: Diabetes UK Professional Conference March 11–13, 2015 London, UK
  • Lewin A DeFronzo R Patel S Initial combination of empagliflozin and linagliptin in subjects with type 2 diabetes Diabetes Care 2015 38 3 394 402 25633662
  • Matthaei S Bowering K Rohwedder K Grohl A Parikh S Study 05 Group Dapagliflozin improves glycemic control and reduces body weight as add-on therapy to metformin plus sulfonylurea: a 24-week randomized, double-blind clinical trial Diabetes Care 2015 38 3 365 372 25592197
  • Liu SC Chien KL Wang CH Chen WC Leung CH Efficacy and safety of adding pioglitazone or sitagliptin to patients with type 2 diabetes insufficiently controlled with metformin and a sulfonylurea Endocr Pract 2013 1 30
  • DeFronzo RA Burant CF Fleck P Wilson C Mekki Q Pratley RE Efficacy and tolerability of the DPP-4 inhibitor alogliptin combined with pioglitazone, in metformin-treated patients with type 2 diabetes J Clin Endocrinol Metab 2012 97 5 1615 1622 22419732
  • Kovacs CS Seshiah V Swallow R Empagliflozin improves glycaemic and weight control as add-on therapy to pioglitazone or pioglitazone plus metformin in patients with type 2 diabetes: a 24-week, randomized, placebo-controlled trial Diabetes Obes Metab 2014 16 2 147 158 23906415
  • Rosenstock J Hansen L Zee P Dual add-on therapy in type 2 diabetes poorly controlled with metformin monotherapy: a randomized double-blind trial of saxagliptin plus dapagliflozin addition versus single addition of saxagliptin or dapagliflozin to metformin Diabetes Care 2014 38 3 376 383 25352655
  • DeFronzo RA Lewin A Patel S Combination of empagliflozin and linagliptin as second-line therapy in subjects with type 2 diabetes inadequately controlled on metformin Diabetes Care 2015 38 3 384 393 25583754
  • Jabbour SA Hardy E Sugg J Parikh S Study 10 Group Dapagliflozin is effective as add-on therapy to sitagliptin with or without metformin: a 24-week, multicenter, randomized, double-blind, placebo-controlled study Diabetes Care 2014 37 3 740 750 24144654
  • Matthaei S Catrinoiu D Celinski A Randomized, double-blind trial of triple therapy with saxagliptin add-on to dapagliflozin plus metformin in patients with type 2 diabetes Diabetes Care 2015 38 11 2018 2024 26324329
  • Mathieu C Ranetti AE Li D Randomized, double-blind, phase 3 trial of triple therapy with dapagliflozin add-on to saxagliptin plus metformin in type 2 diabetes Diabetes Care 2015 38 11 2009 2017 26246458
  • Merovci A Solis-Herrera C Daniele G Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production J Clin Invest 2014 124 2 509 514 24463448
  • Ferrannini E Muscelli E Frascerra S Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients J Clin Invest 2014 124 2 499 508 24463454
  • Neschen S Scheerer M Seelig A Metformin supports the antidiabetic effect of a sodium glucose cotransporter 2 inhibitor by suppressing endogenous glucose production in diabetic mice Diabetes 2015 64 1 284 290 25071027
  • Balas B Baig MR Watson C The dipeptidyl peptidase IV inhibitor vildagliptin suppresses endogenous glucose production and enhances islet function after single-dose administration in type 2 diabetic patients J Clin Endocrinol Metab 2007 92 4 1249 1255 17244786
  • Hansen L Iqbal N Ekholm E Cook W Hirshberg B Postprandial dynamics of plasma glucose, insulin, and glucagon in patients with type 2 diabetes treated with saxagliptin plus dapagliflozin add-on to metformin therapy Endocr Pract 2014 20 11 1187 1197 25370334
  • Raz I Mosenzon O Early insulinization to prevent diabetes progression Diabetes Care 2013 36 suppl 2 S190 S197 23882045
  • Scheen AJ Combined thiazolidinedione-insulin therapy: should we be concerned about safety? Drug Saf 2004 27 12 841 856 15366973
  • Buse JB Bergenstal RM Glass LC Use of twice-daily exenatide in basal insulin-treated patients with type 2 diabetes: a randomized, controlled trial Ann Intern Med 2011 154 2 103 112 21138825
  • Ahmann A Rodbard HW Rosenstock J Efficacy and safety of liraglutide versus placebo added to basal insulin analogues (with or without metformin) in patients with type 2 diabetes: a randomized, placebo-controlled trial Diabetes Obes Metab 2015 17 11 1056 1064 26179619
  • Rosenstock J Fonseca VA Gross JL Advancing basal insulin replacement in type 2 diabetes inadequately controlled with insulin glargine plus oral agents: A comparison of adding albiglutide, a weekly GLP-1 receptor agonist, versus thrice-daily prandial insulin lispro Diabetes Care 2014 37 8 2317 2325 24898300
  • Barnett AH Charbonnel B Donovan M Fleming D Chen R Effect of saxagliptin as add-on therapy in patients with poorly controlled type 2 diabetes on insulin alone or insulin combined with metformin Curr Med Res Opin 2012 28 4 513 523 22313154
  • Yki-Jarvinen H Rosenstock J Duran-Garcia S Effects of adding linagliptin to basal insulin regimen for inadequately controlled type 2 diabetes: a ≥52-week randomized, double-blind study Diabetes Care 2013 36 12 3875 3881 24062327
  • Rosenstock J Rendell MS Gross JL Fleck PR Wilson CA Mekki Q Alogliptin added to insulin therapy in patients with type 2 diabetes reduces HbA(1C) without causing weight gain or increased hypoglycaemia Diabetes Obes Metab 2009 11 12 1145 1152 19758359
  • Vilsboll T Rosenstock J Yki-Jarvinen H Efficacy and safety of sitagliptin when added to insulin therapy in patients with type 2 diabetes Diabetes Obes Metab 2010 12 2 167 177 20092585
  • Yoon NM Cavaghan MK Brunelle RL Roach P Exenatide added to insulin therapy: a retrospective review of clinical practice over two years in an academic endocrinology outpatient setting Clin Ther 2009 31 7 1511 1523 19695400
  • Seufert J Pegelow K Bramlage P Efficacy and safety of insulin glargine added to a fixed-dose combination of metformin and a dipeptidyl peptidase-4 inhibitor: results of the GOLD observational study Vasc Health Risk Manag 2013 9 711 717 24259985
  • Rosenstock J Jelaska A Zeller C Kim G Broedl UC Woerle HJ Impact of empagliflozin added on to basal insulin in type 2 diabetes inadequately controlled on basal insulin: a 78-week randomized, double-blind, placebo-controlled trial Diabetes Obes Metab 2015 17 10 936 948 26040302
  • Wilding JP Woo V Soler NG Long-term efficacy of dapagliflozin in patients with type 2 diabetes mellitus receiving high doses of insulin: a randomized trial Ann Intern Med 2012 156 6 405 415 22431673
  • Neal B Perkovic V de Zeeuw D Efficacy and safety of canagliflozin, an inhibitor of sodium-glucose cotransporter 2, when used in conjunction with insulin therapy in patients with type 2 diabetes Diabetes Care 2015 38 3 403 411 25468945
  • Damci T Emral R Svendsen AL Balkir T Vora J Lower risk of hypoglycaemia and greater odds for weight loss with initiation of insulin detemir compared with insulin glargine in Turkish patients with type 2 diabetes mellitus: local results of a multinational observational study BMC Endocr Disord 2014 14 61 25048824
  • Yale JF Damci T Kaiser M Initiation of once daily insulin detemir is not associated with weight gain in patients with type 2 diabetes mellitus: results from an observational study Diabetol Metab Syndr 2013 5 1 56 24499517
  • Vora J Combining incretin-based therapies with insulin: realizing the potential in type 2 diabetes Diabetes Care 2013 36 suppl 2 S226 S232 23882050
  • American Diabetes Association Fast Facts Data and Statistics about Diabetes 2015 Available from: http://professional.diabetes.org/ResourcesForProfessionals.aspx?cid=91777&loc=dorg-statistics Accessed March 23, 2015
  • Gough SC Bode B Woo V Efficacy and safety of a fixed-ratio combination of insulin degludec and liraglutide (IDegLira) compared with its components given alone: results of a phase 3, open-label, randomised, 26-week, treat-to-target trial in insulin-naive patients with type 2 diabetes Lancet Diabetes Endocrinol 2014 2 11 885 893 25190523
  • Rosenstock J Ferrannini E Euglycemic diabetic ketoacidosis: a predictable, detectable, and preventable safety concern with SGLT2 inhibitors Diabetes Care 2015 38 9 1638 1642 26294774
  • FDA Drug Safety Communication: FDA revises labels of SGLT2 inhibitors for diabetes to include warnings about too much acid in the blood and serious urinary tract infections 2015 Available from: http://www.fda.gov/Drugs/DrugSafety/ucm475463.htm Accessed January 29, 2016
  • Zinman B Wanner C Lachin JM Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes N Engl J Med 2015 373 22 2117 2128 26378978
  • Erondu N Desai M Ways K Meininger G Diabetic ketoacidosis and related events in the canagliflozin type 2 diabetes clinical program Diabetes Care 2015 38 9 1680 1686 26203064
  • AACE/ACE scientific and clinical review: association of SGLT2 inhibitors and DKA 2015 Available from: http://resources.aace.com/in-the-news/aaceace-scientific-and-clinical-review-association-sglt2-inhibitors-and-dka Accessed January 29, 2016
  • Hsu PF Sung SH Cheng HM Association of clinical symptomatic hypoglycemia with cardiovascular events and total mortality in type 2 diabetes: a nationwide population-based study Diabetes Care 2013 36 4 894 900 23223349
  • ORIGIN Trial Investigators Gerstein HC Bosch J Basal insulin and cardiovascular and other outcomes in dysglycemia N Engl J Med 2012 367 4 319 328 22686416
  • UK Prospective Diabetes Study (UKPDS) Group Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes Lancet 1998 352 9131 837 853 9742976
  • Roumie CL Hung AM Greevy RA Comparative effectiveness of sulfonylurea and metformin monotherapy on cardiovascular events in type 2 diabetes mellitus: a cohort study Ann Intern Med 2012 157 9 601 610 23128859
  • Glucotrol® (glipizide tablets) [prescribing information] New York, NY Pfizer Inc 2010
  • Micronase® (glyburide tablets) [full prescribing information] New York, NY Pfizer Inc 2010
  • Amaryl® (glimepiride) [prescribing information] Bridgewater, NJ sanofiaventis 2013
  • Kachroo S Kawabata H Colilla S Use of sulfonylureas in patients with multiple risk factors for CV disease or those with existing CV disease - CREST Study: are we following current ADA/EASD guidelines? [A1112] Paper presented at: 51st Annual Meeting, European Association for the Study of Diabetes September 14–18, 2015 Stockholm, Sweden
  • Baxter C Das R Langerman H Increased risk of cardiovascular-related events associated with sulfonylureas compared to other antihyperglycaemic drugs: a Bayesian meta-analysis of survival data. [A128] Paper presented at: 51st Annual Meeting, European Association for the Study of Diabetes September 14–18, 2015 Stockholm, Sweden
  • Berg B Wilke T Groth A Treatment characteristics and outcomes associated with sulphonylurea versus metformin therapy in incident type 2 diabetes mellitus patients: results of the German CREST study. [A130] Paper presented at: 51st Annual Meeting, European Association for the Study of Diabetes September 14–18, 2015 Stockholm, Sweden
  • Cycloset® (bromocriptine mesylate) [prescribing information] Tiverton, RI VeroScience, LLC 2010
  • Gaziano JM Cincotta AH O’Connor CM Randomized clinical trial of quick-release bromocriptine among patients with type 2 diabetes on overall safety and cardiovascular outcomes Diabetes Care 2010 33 7 1503 1508 20332352
  • Scirica BM Bhatt DL Braunwald E Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus N Engl J Med 2013 369 14 1317 1326 23992601
  • White WB Cannon CP Heller SR Alogliptin after acute coronary syndrome in patients with type 2 diabetes N Engl J Med 2013 369 14 1327 1335 23992602
  • Green JB Bethel MA Armstrong PW Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes N Engl J Med 2015 373 3 232 242 26052984
  • Zannad F Cannon C Cushman WC Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial Lancet 2015 385 9982 2067 2076 25765696
  • Fu AZ Johnston SS Ghannam A Tsai K Cappell K Fowler R Riehle E Cole AL Kalsekar I Sheehan J Association between hospitalization for heart failure and dipeptidyl peptidase 4 inhibitors in patients with type 2 diabetes: an observational study Diabetes Care 2016 39 5 726 734 26740636
  • Rosenstock J Marx N Kahn SE Cardiovascular outcome trials in type 2 diabetes and the sulphonylurea controversy: rationale for the active-comparator CAROLINA trial Diab Vasc Dis Res 2013 10 4 289 301 23449634
  • Cardiovascular and renal microvascular outcome study with linagliptin in patients with type 2 diabetes mellitus at high vascular risk (CARME-LINA) 2015 Available from: http://clinicaltrials.gov/ct2/show/NCT01897532?term=NCT01897532&rank=1 Accessed March 16, 2015
  • Multicenter trial to evaluate the effect of dapagliflozin on the incidence of cardiovascular events (DECLARE-TIMI58) 2015 Available from: http://clinicaltrials.gov/ct2/show/NCT01730534?term=declare&rank=2 Accessed September 29, 2015
  • Neal B Perkovic V de Zeeuw D Rationale, design, and baseline characteristics of the Canagliflozin Cardiovascular Assessment Study (CANVAS)–a randomized placebo-controlled trial Am Heart J 2013 166 2 217 223.e11 23895803
  • American Diabetes Association First CVD outcome trial of a GLP-1 agonist finds no cardiac risk or benefit 2015 Available from: http://www.diabetes.org/newsroom/press-releases/2015/elixa.html?referrer=http://google.diabetes.org/search?site=Diabetes&client=diabetes&entqr=3&oe=ISO-8859-1&ie=ISO-8859-1&ud=1&proxystylesheet=diabetes&output=xml_no_dtd&proxyreload=1&q=ELIXA Accessed June 18, 2015
  • Jiang Y Huang W Wang J Metformin plays a dual role in MIN6 pancreatic beta cell function through AMPK-dependent autophagy Int J Biol Sci 2014 10 3 268 277 24644425
  • Chao EC SGLT-2 inhibitors: a new mechanism for glycemic control Clin Diabetes 2014 32 1 4 11 26246672
  • Barnett AH Redefining the role of thiazolidinediones in the management of type 2 diabetes Vasc Health Risk Manag 2009 5 1 141 151 19436665
  • Green JB Feinglos MN Are sulfonylureas passe? Curr Diab Rep 2006 6 5 373 377 17076998
  • Harrison LB Adams-Huet B Raskin P Lingvay I Beta-cell function preservation after 3.5 years of intensive diabetes therapy Diabetes Care 2012 35 7 1406 1412 22723578