DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
Contents available at ScienceDirect
Diabetes Research and Clinical Practice journ al h ome pa ge : www .elsevier.co m/lo cate/diabres
Invited review
SGLT2 inhibitors in the treatment of type 2 diabetes Farhad M. Hasan a,*, Mazen Alsahli b, John E. Gerich c a
University of Virginia School of Medicine, Charlottesville, VA, USA University of Toronto Faculty of Medicine, Toronto, Ontario, Canada c University of Rochester School of Medicine, Rochester, NY, USA b
article info
abstract
Article history:
The kidney plays an important role in glucose homeostasis via its production, utilization,
Received 27 January 2014
and, most importantly, reabsorption of glucose from glomerular filtrate which is largely
Received in revised form
mediated via the sodium glucose co-transporter 2 (SGLT2). Pharmacological inhibition of
6 February 2014
SGLT2 increases urinary glucose excretion and decreases plasma glucose levels in an
Accepted 19 February 2014
insulin-independent manner. Agents that inhibit SGLT2 represent a novel class of drugs,
Available online xxx
which has recently become available for treatment of type 2 diabetes. This article summarizes the rationale for use of these agents and reviews available clinical data on their
Keywords:
efficacy, safety, and risks/benefits. # 2014 Elsevier Ireland Ltd. All rights reserved.
Type 2 diabetes Glucose reabsorption SGLT2 inhibitors Dapagliflozin Canagliflozin Empagliflozin
Contents 1. 2.
3.
4. 5.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . The kidneys and normal glucose homeostasis . 2.1. Renal gluconeogenesis. . . . . . . . . . . . . . . 2.2. Renal glucose utilization . . . . . . . . . . . . . 2.3. Renal glucose reabsorption . . . . . . . . . . . Role of the kidney in diabetes . . . . . . . . . . . . . . 3.1. Renal gluconeogenesis. . . . . . . . . . . . . . . 3.2. Renal glucose utilization . . . . . . . . . . . . . 3.3. Renal glucose reabsorption . . . . . . . . . . . Therapeutic implications of SGLTs inhibition. . SGLT2 in clinical practice . . . . . . . . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
000 000 000 000 000 000 000 000 000 000 000
* Corresponding author at: 403 Woodcrest Road Wayne, PA 19087, USA. Tel.: +1 585 752 2860. E-mail address:
[email protected] (J.E. Gerich). Abbreviations: HbA1c, hemoglobin A1c; FRG, familial renal glucosuria; GLUT, glucose transporter; eGFR, estimated glomerular filtration rate; FPG, fasting plasma glucose; SGLT, sodium–glucose co-transporter; T2DM, type 2 diabetes mellitus; UGE, urinary glucose excretion. http://dx.doi.org/10.1016/j.diabres.2014.02.014 0168-8227/# 2014 Elsevier Ireland Ltd. All rights reserved.
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
2
diabetes research and clinical practice xxx (2014) xxx–xxx
5.1.
6.
7.
1.
Dapagliflozin . . . . . . . . . . . . . . . . . . . . . . . . . 5.1.1. Efficacy in monotherapy trials . . . . . 5.1.2. Efficacy in add-on trials . . . . . . . . . . 5.1.3. Effects on weight . . . . . . . . . . . . . . . 5.1.4. Effects on blood pressure . . . . . . . . . 5.1.5. Effects on lipids . . . . . . . . . . . . . . . . 5.1.6. Safety . . . . . . . . . . . . . . . . . . . . . . . . 5.1.7. Dosing recommendations. . . . . . . . . 5.1.8. Use in special populations . . . . . . . . 5.2. Canagliflozin . . . . . . . . . . . . . . . . . . . . . . . . . 5.2.1. Efficacy in monotherapy trials . . . . . 5.2.2. Efficacy in add-on trials . . . . . . . . . . 5.2.3. Effects on weight . . . . . . . . . . . . . . . 5.2.4. Effects on blood pressure . . . . . . . . . 5.2.5. Effects on lipids . . . . . . . . . . . . . . . . 5.2.6. Safety . . . . . . . . . . . . . . . . . . . . . . . . 5.2.7. Dosing recommendations. . . . . . . . . 5.2.8. Use in special populations . . . . . . . . 5.3. Other considerations . . . . . . . . . . . . . . . . . . . Selected SGLT2 inhibitors in clinical development . 6.1. Empagliflozin . . . . . . . . . . . . . . . . . . . . . . . . . 6.2. Ipragliflozin . . . . . . . . . . . . . . . . . . . . . . . . . . 6.3. LX4211 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
Introduction
Management of type 2 diabetes (T2DM) continues to be challenging despite the numerous therapeutic options available. Metformin is currently recommended as the first choice agent [1–3]. However side effects, primarily gastrointestinal, are common and approximately 10% of patients cannot tolerate it at any dose [4]. T2DM is a progressive disease so that as b cell function deteriorates, most patients will require additional therapy [5]. Alternatives and additions to metformin also have problems, which limit their usefulness. Sulfonylureas, meglitinides, and insulin are associated with weight gain and risk of hypoglycemia [5,6]. Thiazolidinediones are associated with risk of weight gain, edema, heart failure, and fractures [6,7]. Dipeptidyl peptidase 4 (DPP-4) inhibitors have only modest glucose lowering effect and their long-term safety remains to be established [6,8]. Similarly, the long-term safety of glucagon-like peptide-1 (GLP-1) analogs is unknown, and their use is often associated with significant gastrointestinal side effects [6,8]. The use of alpha glucosidase inhibitors outside the orient and Germany is generally low due to their common gastrointestinal side effects and frequent dosing schedule [6,9]. Additionally, renal insufficiency places major restrictions on many of the above agents (e.g. metformin, sulfonylureas, GLP-1 analogs, and alpha glucosidase inhibitors) [10]. Thus there is a need for additional treatment options. The ideal antidiabetes drug would be one associated with a robust and sustained HbA1c reduction, is well tolerated, can be administered easily, has low or no risk of hypoglycemia, has good long term safety, and has added benefit such as a favorable impact on b cell function, blood pressure, weight, albuminuria etc.
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . . .
000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000
The kidney plays an important role in glucose homeostasis and has recently become a target for treatment of diabetes. The majority of glucose reabsorption from glomerular filtrate is mediated via a transporter protein called sodium glucose cotransporter 2 (SGLT2) [11]. Pharmacological inhibition of SGLT2 increases urinary glucose excretion (UGE) and decreases plasma glucose levels in an insulin-independent manner [11]. SGLT2 inhibitors represent a novel class of drugs that has recently become available for treatment of T2DM. This article summarizes the rationale for use of these agents and reviews available clinical data on their efficacy, safety, and risks/benefits.
2. The kidneys and normal glucose homeostasis The kidney is involved in the regulation of glucose homeostasis via three different mechanisms: release of glucose into the circulation (gluconeogenesis), uptake of glucose from the circulation for its energy needs, and most importantly, glucose reabsorption from glomerular filtrate.
2.1.
Renal gluconeogenesis
After a 14–16 h overnight fast, approximately half of the glucose released into the circulation is from the breakdown of liver glycogen (glycogenolysis) stored in the liver and the other half is from the production of new glucose molecules (gluconeogenesis) by liver and kidneys [12–14]. In humans, only the liver and kidney contain significant amounts of the enzyme glucose-6-phosphatase and therefore are the only
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
3
Table 1 – The sodium glucose co-transporter family. Co-transporter SGLT1 SGLT2 SGLT4 SGLT5 SGLT6 SMIT1
Gene
Substrate
Tissue distribution
SLC5A1 SLC5A2 SLC5A9 SLC5A10 SMIT2/SLC5A11 SLC5A3
Glucose, galactose Glucose Glucose, mannose Not known Glucose, myo-inositol Glucose, myo-inositol
Intestine, trachea, kidney, heart, brain, testis, prostate Kidney, brain, liver, thyroid, muscle and heart Intestine, kidney, liver, brain, lung, trachea, uterus, pancreas Kidney Brain, kidney, intestine Brain, heart, kidney, lung
Adapted from Wright et al. [23].
organs that are able to perform gluconeogenesis. The human liver and kidneys provide about equal amounts of glucose via gluconeogenesis in the postabsorptive state (i.e. 12–16 h after the last meal). Consequently, after an overnight fast, 75–80% of glucose released into the circulation derives from the liver and the remaining 20–25% derives from the kidneys. As the duration of fasting increases, glycogen stores in the liver become further depleted until, after 48 h, virtually all the glucose released into the circulation is derived from gluconeogenesis. Consequently, as the length of fast increases, the proportion of overall glucose release accounted for by renal gluconeogenesis increases [13,15] After meal ingestion, overall endogenous glucose release decreases by 61%, with hepatic glycogenolysis virtually ceasing in the 4–6-h period [16]. Hepatic gluconeogenesis also decreases by 82% and glucose molecules generated through this pathway are not generally released in the circulation, but are largely directed into hepatic glycogen. Perhaps surprisingly, renal gluconeogenesis actually increases by approximately twofold and accounts for 60% of endogenous glucose release in the postprandial period [16]. This has been hypothesized to facilitate efficient repletion of glycogen stores in the liver [16]. These differences in regulation and reciprocal change in renal and hepatic glucose release have led to the concept of hepatorenal glucose reciprocity [17]. This concept refers to the situations in which a physiological or pathological decrease in glucose release by kidney or liver is associated with a compensatory increase in glucose release by liver or kidney so as to prevent hypoglycemia or to optimize homeostasis. Examples of this include the anhepatic phase after liver transplantation, prolonged fasting, acidosis, meal ingestion and insulin overdoses in diabetes mellitus [17,18]. With respect to hormonal influences, insulin suppresses glucose release by both organs with roughly comparable efficacy [19], whereas glucagon normally stimulates hepatic glucose release only, mainly via an early action on glycogenolysis [20]. Catecholamines normally exert a direct effect on renal glucose release only [21,22], although they may indirectly affect both hepatic and renal glucose release by increasing availability of gluconeogenic substrates and by suppressing insulin secretion. Cortisol, growth hormone and thyroid hormones have long-term stimulatory influences on hepatic glucose release (over a period of days) [12]. Their effects on renal glucose release in humans have yet to be determined.
2.2.
Renal glucose utilization
In the post-absorptive setting after an overnight fast, the kidneys utilize approximately 10% of all glucose utilized by the
body for its own energy needs [16]. Postprandially, renal glucose uptake increases approximately threefold; however, the proportion of overall systemic glucose disposal due to the kidney changes very little [16].
2.3.
Renal glucose reabsorption
Normally, the kidneys filter approximately 180 L of plasma each day. Since the average plasma glucose concentration throughout a 24-h period is 5.5 mmol/l (100 mg/dl), 180 g of glucose is filtered by the kidneys each day. In healthy individuals, virtually all of this is reabsorbed into the circulation and the urine is essentially free from glucose. To put this into perspective, in a typical day, the kidneys produce 15–55 g glucose via gluconeogenesis and metabolize 25–35 g glucose [11]. Therefore, in terms of glucose economy, it is clear that renal reabsorption (180 g) is the primary mechanism by which the kidney influences glucose homeostasis. Alterations in renal tubular glucose reabsorption may therefore be expected to have a considerable impact on glucose homeostasis. Reabsorption of glucose from glomerular filtrate occurs by means of sodium–glucose co-transporters (SGLT1 and SGLT2) in the proximal convoluted tubules. There are six members of this family (Table 1) [23]. In animal models, approximately 90% of glucose is reabsorbed by SGLT2, a high-capacity low-affinity glucose transporter thought to be located exclusively on the luminal surface of the epithelial cells lining the S1 and S2 segments of the proximal tubule [24,25]. Transport of sodium and glucose by SGLT2 occurs in a 1:1 ratio [24,26]. The remaining approximately 10% of glucose reabsorption is mediated by SGLT1, a high-affinity, low-capacity glucose/ galactose transporter located on the luminal surface of epithelial cells lining the S3 segment of the proximal tubule [26,27]. SGLT1 is also extensively expressed in the small intestine and in other tissues [26]. Glucose reabsorbed from the proximal tubules by SGLTs is then released into the circulation through the action of facilitative glucose transporters (GLUTs) at the basolateral membrane of the epithelial cells lining the proximal tubules (Fig. 1) [28]. Fig. 2 describes the renal handling of filtered glucose [29]. Glucose is freely filtered in the glomerulus, so that, as plasma glucose levels increase, the amount of glucose in the glomerular filtrate increases linearly. Reabsorption of filtered glucose also increases linearly until the maximal reabsorptive capacity (Tm) is exceeded. The plasma glucose concentration at which this occurs is often referred to as the renal threshold, which is approximately 11.0 mmol/l (200 mg/dl) in healthy adults and equates to a filtration rate of 260–350 mg/min/
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
4
diabetes research and clinical practice xxx (2014) xxx–xxx
Fig. 1 – Glucose reabsorption from the glomerular filtrate through a proximal tubular epithelial cell into the blood. GLUT, glucose transporter; SGLT, sodium–glucose co-transporter. Modified from Bakris et al. [28]. 1.73 m2 [30]. Once plasma glucose concentrations exceed this threshold, the percentage of filtered glucose that is reabsorbed decreases and the percentage of the filtered load of glucose that is excreted in the urine increases, resulting in glucosuria. The ‘rounding’ of the titration curve seen around the transition from complete reabsorption to urinary excretion of excess glucose (shown in Fig. 2 as ‘splay’) can be accounted for by heterogeneity in the glomerular filtration rate and glucose reabsorptive capacity of different individual nephrons [29]. The renal threshold for glucose is decreased in individuals with a rare condition known as familial renal glucosuria (FRG), caused by a range of mutations to the SLC5A2 gene, which encodes SGLT2 [31]. Depending on the nature of the mutations, these individuals have varying degrees of glucosuria, but in the most severe form (so-called ‘Type 0’ disease) they can lose > 100 g glucose per day to the urine [31]. Interestingly, the large majority of patients exhibit no symptoms and their condition is only identified incidentally. Typically, they do not become hypoglycemic or dehydrated and have no electrolyte imbalance or increased risk of urinary tract infections [31].
3.
Role of the kidney in diabetes
All of the ways in which the kidney normally affects glucose homeostasis are altered in patients with diabetes mellitus.
3.1.
Renal gluconeogenesis
Patients with T2DM have an increased release of glucose into the circulation by the kidney in the fasting state [32]. Although the liver is commonly viewed as being largely responsible for increased release of glucose into the circulation in T2DM, the absolute increase in renal glucose release is comparable in magnitude (2.60 and 2.21 mmol/(kg min) for liver and kidneys, respectively) [32]. Postprandially, renal glucose release also increases to a greater extent in people with T2DM than in people with normal glucose tolerance [33]. Much of this difference is as a result of an increase in endogenous glucose release of which 40% is due to increased renal glucose release [33].
3.2.
Renal glucose utilization
Meyer et al. showed that, in the post-absorptive state, renal glucose uptake is significantly greater in patients with T2DM than in normal individuals (353 mmol/min vs. 103 mmol/min), actually exceeding increased glucose production to result in a net glucose uptake of 92 mmol/min. This contrasts with a net output of 21 mmol/min in non-diabetic individuals [32]. In the postprandial state, uptake of glucose by tissues is increased in patients with T2DM and its distribution and fate are altered [33]. Glucose uptake by the kidneys is raised by more than twofold in diabetic vs. non-diabetic individuals [33], whereas glucose uptake in muscle is not significantly altered. Moreover, less glucose is oxidized [33].
3.3. Fig. 2 – Renal glucose handling. Tm, transport maximum. Adapted from Silverman and Turner [29].
Renal glucose reabsorption
Glucosuria in diabetic patients does not occur at plasma glucose levels that would normally produce glucosuria in
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
non-diabetic individuals [34]. This is the result of increased glucose reabsorption from glomerular filtrate due to increased transport maximum (Tm) for glucose. This was shown in 1951 when Farber at al. demonstrated that Tm value for glucose is above mean normal in 10 out of 12 patients with diabetes (type 1 or 2). The administration of insulin decreased the Tm in each of the 12 patients [35]. In another study twenty years later in patients with type 1 diabetes mellitus (T1DM), Mogensen demonstrated that Tm for glucose increased from approximately 350 mg/min in normal individuals to approximately 420 mg/min in those with T1DM [34]. The responsible mechanism appears to involve increased expression of glucose transporter genes as studies of renal proximal tubular cells isolated from the urine of people with T2DM as well as cells from several experimental animal models have demonstrated enhanced expression of SGLT2 and GLUT2 transporters [36,37]. The up-regulation of SGLT2 in T2DM appears to be acquired and possibly secondary to hyperglycemia although the exact mechanism is unknown [36].
4. Therapeutic implications of SGLTs inhibition Phlorizin, a non-specific SGLT1 and SGLT2 inhibitor first isolated from the root bark of the apple tree in 1835 [38], was found to increase glucosuria, reduce hyperglycemia, and normalize insulin sensitivity in a partial pancreatectomized animal model of T2DM [39]. However, it was not developed as a treatment for diabetes because of a number of practical shortcomings. It is non-selective and inhibits SGLT1 at the intestinal brush border, which is responsible for absorption of dietary glucose [23]. Inhibition of SGLT1, therefore, has the potential to result in glucose–galactose malabsorption and thus diarrhea, as occurs in naturally occurring SGLT1 deficiency [40]. Furthermore, phlorizin is poorly absorbed in the intestine and is readily hydrolyzed to phloretin, a compound that blocks the facilitative glucose transporter, GLUT1. This might lead to interference with glucose uptake in a number of tissues [41]. To overcome phlorizin shortcomings, highly specific inhibitors of SGLT2 were developed (Fig. 3) [42]. Early SGLT2 inhibitor candidates, remogliflozin [43] and sergliflozin [44], were selective for SGLT2 versus SGLT1 (296- and 395-fold, respectively) but remained susceptible to glucosidase cleavage in the gut [45] and had relatively short half-lives [46–48]. The discovery of C-arylglucoside SGLT2 inhibitors, which are resistant to gastrointestinal tract b-glucosidases, followed. Initial C-arylglucoside compounds had lower affinities for SGLT2, but further modifications produced the more potent dapagliflozin [45]. In preclinical studies dapagliflozin had a longer half-life (13.8 h following a single oral 50-mg dose) than the earlier SGLT2 inhibitors [49]. The longer half-life allowed for once daily administration to achieve continuous controlled glucosuria. In addition, dapagliflozin is highly selective toward SGLT2, compared with SGLT1. Early in vitro studies with dapagliflozin determined that it was 3000-fold more selective for human SGLT2 versus SGLT1 [50]. In addition, dapagliflozin is 32-fold more potent than phlorizin in SGLT2 inhibition [51]. Dapagliflozin produces a dose-dependent increase in renal glucose excretion in
5
humans though SGLT2 inhibition [51–53]. For example, in a Phase IIa dose-ranging study, patients with T2DM (n = 47) were randomized to receive oral doses of dapagliflozin 5, 25 or 100 mg, or placebo, once daily for 14 days [52]. Dosedependent increases in renal glucose excretion occurred in patients with T2DM who received dapagliflozin. On day 1, renal glucose excretion of 45.2, 75.3 or 81.3 g/day occurred with dapagliflozin 5, 25 and 100 mg, respectively; after 14 days of dosing, renal glucose excretion was 36.6, 70.1 and 69.9 g/ day, corresponding to 20, 41 and 44% inhibition of glucose reabsorption. Fasting and postprandial glucose values decreased with treatment as well, and longer-term studies documented reduction in HbA1c as will be discussed later in this review. Dapagliflozin became recently available for clinical use. Canagliflozin, a similar stable, competitive, reversible, and highly selective SGLT2 inhibitor shortly followed. Currently, multiple agents in this class are being developed for the treatment of T2DM [42]. This approach to lowering hyperglycemia in T2DM is appealing for a number of reasons. Unlike the insulin secretagogues and insulin sensitizers, the action of SGLT2 inhibitors is independent of pancreatic b-cell function, which deteriorates over time. The insulin independence of their action plus the fact that they only lower the threshold without completely blocking renal glucose reabsorption means that hypoglycemic episodes are less likely. In fact, no major increase in hypoglycemic events was noted in clinical trials of SGLT2 inhibitors as will be discussed later. Furthermore, the glucosuric effects of these drugs translate into caloric loss and decrease in bodyweight. For example, the glucosuria induced by dapagliflozin monotherapy in patients with T2DM has been reported in one study to be associated with weight reduction of 2.5–3.4 kg in 12 weeks and a net loss of 200– 300 kcal/day [53]. Modest blood pressure reduction is consistently noted in clinical trials of SGLT2 inhibitors [54,55]. The mechanism of that is not entirely clear. The natriuretic effect of SGLT2 inhibitors, due to sodium-dependent co-transport mechanism of SGLT2, in addition to glucose-induced osmotic diuresis may be contributing. There is also a suggestion that some inhibition of the renin–angiotensin–aldosterone system, secondary to increased sodium delivery to the juxtaglomerular apparatus, may be present with SGLT2 inhibitors use [56,57]. As mentioned earlier, SGLT2 reabsorbs approximately 90% of the filtered glucose load in healthy individuals [24,25]. Unexpectedly, available SGLT inhibitors are incapable of completely blocking this in humans. The maximum effect noted is 80 g/day of urinary glucose excretion (UGE), which is less than 50% of the filtered glucose load. Increasing SGLT2 inhibitor dose after the 50% inhibition is achieved does not result in higher inhibition rate [58,59]. Dapagliflozin for example induced approximately 60 g/d of UGE when used at 20 mg/day or at the much higher dose of 500 mg/day in one study [59]. The reason for this incomplete inhibition is not known. Liu et al. has explored and proposed several potential explanations such as an inability of SGLT2 inhibitor to reach and interact with some SGLT2 transporters, or that SGLT2 could be in fact responsible for less than the previously reported 90% glucose reabsorption fraction [58].
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
6
diabetes research and clinical practice xxx (2014) xxx–xxx
Fig. 3 – The structure of selected SGLT2 and SGLT2/SGLT1 inhibitors. Modified from Tahrani et al. [42].
5.
SGLT2 in clinical practice
5.1.
Dapagliflozin
In January 2014 the US Food and Drug Administration (FDA) approved the use of dapagliflozin for adult patients with T2DM [60]. It is the second SGLT2 inhibitor approved by the FDA following the approval of canagliflozin. Dapagliflozin has been in use in Europe since 2012 when the European Medicines Agency approved its use for the treatment of T2DM as an adjunct to diet and exercise, in combination with other antidiabetes agents, including insulin, and as a monotherapy in metformin intolerant patients [61]. Currently, dapagliflozin is the SGLT2 inhibitor with the most published trials. Its efficacy and safety were evaluated in 14 clinical trials both as monotherapy [53,62–64] and in combination with other antidiabetes agents. Dapagliflozin trials involved more than 9000 patients with T2DM. Efficacy end points in major phase 3 trials are summarized in Tables 2 and 3.
5.1.1.
Efficacy in monotherapy trials
Four trials have evaluated the use of dapagliflozin as monotherapy for T2DM. In two separate 12-week trials including a total of 668 drug-naı¨ve patients with poorly controlled T2DM, dapagliflozin led to a significant reduction in HbA1c relative to placebo (0.7% by Kaku et al. [62], and 0.56% by List et al. [53]). List et al. also included a metformin arm that achieved a mean reduction in HbA1c of 0.55% relative to placebo, suggesting that dapagliflozin is as effective as metformin monotherpy in lowering HbA1c. Additionally, two
separate 24-week trials involving a total of 878 subjects with uncontrolled T2DM concluded there was a significant decrement in glycemic targets with dapagliflozin relative to placebo (0.52% by Ferrannini et al. [63], and 0.8% by Ji et al. [64]). Furthermore, fasting plasma glucose (FPG) was significantly reduced in all dapagliflozin monotherapy trials (mean decrement of 27 mg/dl, relative to placebo) [53,62–64]. In a double blind, active controlled trial, dapagliflozin 10 mg/day was as effective as extended release metformin (titrated to 2000 mg once daily) in reducing HbA1c (dapagliflozin, 1.45%; metformin, 1.4%) and superior to metformin in reducing FPG (dapagliflozin, 46.4 mg/dl; metformin 34 mg/dl) [65].
5.1.2.
Efficacy in add-on trials
In two phase-3 trials involving 728 subjects, dapagliflozin as an add-on to metformin significantly decreased HbA1c levels at 24 weeks compared to placebo (0.54% and 0.3%, placebo subtracted) without serious adverse effects [66,67]. In a 52week trial on 801 patients, dapagliflozin 20 mg/day was as affective as glipizide 20 mg/day in reducing HbA1c when added to metformin monotherapy (0.52% from baseline in both groups) [68], and both led to comparable reductions in fasting plasma glucose. Compared to placebo, dapagliflozin add-on to metformin led to a significant reduction in fasting plasma glucose (difference vs. placebo, 18 and 17 mg/dl) [66,67]. Dapagliflozin has also been studied as add-on therapy to glimepiride [69], pioglitazone [70], sitagliptin [71], and insulin [72–74]. At a dose of 10 mg/day, the reduction vs. placebo in HbA1c by dapagliflozin add-on to above agents ranged between 0.5% and 0.7%. Across the same studies FPG was reduced from baseline by 17 mg/dl to 33 mg/dl
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
Author (year)
n Patients
Monotherapy, placebo controlled Kaku (2013) 279
Duration (wk)
Initial HbA1c %
Initial FBG (md/dl)
Drug and dose
12
8.1
162
Background treatment Drug naı¨ve
DAP DAP DAP DAP PBO DAP DAP DAP DAP DAP PBO DAP DAP DAP PBO DAP DAP PBO
1 mg 2.5 mg 5 mg 10 mg
0.12 0.11 0.37 0.44 0.37 0.71 0.72 0.85 0.55 0.9 0.18 0.58 0.77 0.89 0.23 1.04 1.11 0.29
DHbA1c vs. comparator (%)
0.49 0.48 0.74 0.8
DFBG from baseline (mg/dl) 15.6 19.8 23.5 31.9 11.2 16 19 21 24 31 6 15.2 24.1 28.8 4.1 25.1 31.6 2.5
DFBG vs. comparator (mg/dl) 26.8 31 34.7 43.1
List (2009)
389
12
7.8
148.7
Drug naı¨ve
Ferrannini (2010)
485
24
8.3
172.4
Drug naı¨ve
Ji (2013)
393
24
8.3
161.2
Drug naı¨ve
Monotherapy, active-comparator controlled Henry (2012) 422
24
9.1
193.1
Drug naı¨ve
DAP 5 mg DAP 10 mg MET
1.19 1.45 1.4
0.21 0.1
41.5 46.4 34
7.5 12.4
Add-on to MET, placebo controlled Bailey (2010) 546
24
8
162.9
Metformin
148
Metformin
17.8 21.4 23.4 6.0 14.7 2.4
11.9 15.5 17.5
7.2
0.67 0.7 0.84 0.3 0.39 0.1
0.37 0.4 0.54
24
DAP DAP DAP PBO DAP PBO
Add-on to metformin trials, active-comparator controlled Nauck (2011) 801 52 7.7
NR
Metformin
22.3 18.7
3.6
Add-on to other AHAs, placebo controlled Strojek (2011) 597
172.6
Bolinder (2012)
182
24
8.1
2.5 mg 5 mg 10 mg 20 mg 50 mg
DHbA1c from baseline (%)
2.5 mg 5 mg 10 mg 5 mg 10 mg
2.5 mg 5 mg 10 mg 10 mg
0.53 0.54 0.67 0.37 0.72 0.35 0.54 0.66 0.75 0.82
0.28
27.6 34.1
17.1
DAP 10 mg GLIP 20 mg
0.52 0.52
Trial regimen Add-on to GLIM vs. PBO
DAP 2.5 mg
0.58
0.45
16.8
18.7
DAP DAP PBO DAP DAP PBO DAP
0.63 0.82 0.13 0.95 1.21 0.54 0.5
0.5 0.69
21.2 28.4 2.0 22.8 33.1 13.1 24.1
23.2 30.4
Rosenstock (2012)
420
48
8.4
164.8
Add-on to PIO vs. PBO
Jabbour (2013)
432
24
8
162.6
Add-on to SIT MET vs. PBO
5 mg 10 mg 5 mg 10 mg 10 mg
0 vs. GLIP
10 13 15 18 25 11.1 20.1 24.4
0.41 0.67 0.5
diabetes research and clinical practice xxx (2014) xxx–xxx
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
Table 2 – Dapagliflozin in major clinical trials.
9.7 20.0 27.9
7
DIAB-6018; No. of Pages 26
8
DAP, dapagliflozin; PBO, placebo; MET, metformin; GLIP, glipizide; GLIM, glimepiride; PIO, pioglitazone; INS, insulin; SIT, sitagliptin; n, number; wk, weeks; FPG, fasting plasma glucose; vs., versus; D, change in; LDL, low density lipoprotein cholesterol; NR, not reported.
16.1 5.4 34.1 23.4 18.0 0.82 0.78 0.43
179.0 808 Wilding (2013)
104
8.6
Add-on to INS vs. PBO
DAP 5 mg DAP 10 mg PBO
0.39 0.35
2.5 0.21
16.2 16.9 NR 20.5 0.96 1.01 0.47 0.64
177.5 800 Wilding (2012)
48
8.6
Add-on to INS vs. PBO
DAP 5 mg DAP 10 mg PBO DAP 2.5 mg
0.49 0.54
– 0.32
27.4
9.6 17.8 12.4 0.69 0.09 0.79 DAP 20 mg PBO DAP 2.5 mg
0.78
15.4 3.8 2.4 0.7 0 0.61 PBO DAP 10 mg 161.2 8.4 71 Wilding (2009)
12
Initial FBG (md/dl) Author (year)
Table 2 (Continued )
n Patients
Duration (wk)
Initial HbA1c %
Add-on to INS vs. PBO
DHbA1c from baseline (%) Drug and dose
DHbA1c vs. comparator (%)
DFBG from baseline (mg/dl)
DFBG vs. comparator (mg/dl)
diabetes research and clinical practice xxx (2014) xxx–xxx
compared with changes of +18 mg/dl to 13 mg/dl in placebo groups [69–73]. Additionally, dapagliflozin add-on to glimepiride [69], and add-on to pioglitazone [70] led to a significant reduction in postprandial glucose (PPG). Dapagliflozin has also been studied in combination with metformin in treatmentnaı¨ve patients with T2DM [65]. This combination was more effective than either drug alone in reducing HbA1c (dapagliflozin 10 mg/day, 1.5%; metformin, 1.4%; dapagliflozin 10 mg/dl plus metformin, 2%) and FPG (dapagliflozin 10 mg/day, 46 mg/dl; metformin, 35 mg/dl; dapagliflozin 10 mg/dl plus metformin, 60 mg/dl).
5.1.3.
Effects on weight
Dapagliflozin monotherapy for 12 weeks resulted in a loss of 2.5 kg body weight compared with a loss of 1.2 kg and 1.7 kg in the placebo and metformin arms respectively [53]. In a 12week study of Japanese patients with T2DM, dapagliflozin monotherapy was associated with a significant weight reduction (1.9 kg) relative to placebo [62]. In a head-to-head comparison with metformin, dapagliflozin, both at 5 mg and 10 mg, led to a greater weight reduction (2.6 and 2.7 kg respectively) compared with metformin (1.4 kg) [65]. Ferrannini et al. [63], however, found no significant difference in body weight with dapagliflozin vs. placebo after 24-week treatment (3.2 kg vs. 2.2 kg). The authors attributed this to a high placebo effect likely due to improved adherence to diet and exercise. Yet, consistent with other studies, dapagliflozin produced a loss of 200–300 kcal/day. When used in combination or as add-on to other antidiabetes agents, dapagliflozin therapy resulted in a similar favorable effect on weight. The combination of dapagliflozin and metformin was more effective in reducing body weight than metformin alone (3.3 kg with dapagliflozin 10 mg/day plus metformin, vs. 1.4 kg with metformin alone) [65]. The weight reduction observed in dapagliflozin treated patients was predominantly attributable to reductions in body fat mass [67]. Dapagliflozin add-on to pioglitazone mitigated the weight gain associated with the latter, and the beneficial effects on body weight were sustained at 48 weeks of therapy [70]. When compared to glipizide in patients whose diabetes was inadequately controlled with metformin, dapagliflozin achieved significant reduction from baseline in body weight after 52 weeks (3.2 kg) compared with a weight gain of 1.4 kg with glipizide [68].
5.1.4.
Effects on blood pressure
Dapagliflozin monotherapy over 12–24 weeks was associated with a significant placebo-subtracted reduction in systolic blood pressure in the range of 2–9 mmHg. There was no significant change in heart rate or increase in syncopal episodes [53,62–64]. In a pooled analysis of 12 placebocontrolled studies, treatment with dapagliflozin 10 mg/day resulted in a systolic blood pressure change from baseline of 4.4 mmHg and diastolic blood pressure of 2.1 mmHg vs. 0.9 mmHg systolic and 0.5 mmHg diastolic blood pressure for placebo group at week 24 [54].
5.1.5.
Effects on lipids
An analysis addressing cardiovascular risk factors in the dapagliflozin trials concluded that dapagliflozin had an insignificant effect on lipid levels in individual studies [75].
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
9
diabetes research and clinical practice xxx (2014) xxx–xxx
Table 3 – Important seconday outcomes in dapagliflozin clinical trials. Author (year)
Drug and dose
DWeight from baseline (kg)
DWeight vs. comparator (kg)
DSBP from baseline (mmHg)
DSBP vs. comparator (mmHg)
DLDL from baseline (mg/dl)
DLDL vs. comparator (mg/dl)
Kaku (2013)
DAP 5 mg DAP 10 mg PBO
2.1 1.9 0.1
2.0 1.8
1.8 2.1 0.6
2.4 2.7
3.5 0.7 1.5
2 0.8
List (2009)
DAP DAP DAP DAP DAP PBO
2.5 mg 5 mg 10 mg 20 mg 50 mg
2.7 2.5 2.7 3.4 3.4 1.2
1.5 1.3 1.5 2.2 2.2
3.1 2.9 6.4 4.3 2.6 2.4
5.5 5.3 8.8 6.7 5
NR
–
Ferrannini (2010)
DAP 2.5 mg DAP 5 mg DAP 10 mg PBO
3.3 2.8 3.2 2.2
1.1 0.8 1.0
4.6 2.3 3.6 0.9
3.7 1.4 2.7
NR
–
Ji (2013)
DAP 5 mg DAP 10 mg PBO
1.6 2.3 0.3
1.4 2.0
1.2 2.3 0.8
2 3.1
3 7.4 1.7
1.3 9.1
Henry (2012)
DAP 5 mg DAP 10 mg MET
2.6 2.7 1.4
1.2 1.3
4.2 4 1.5
2.7 2.5
NR
–
Bailey (2010)
DAP 2.5 mg DAP 5 mg DAP 10 mg PBO
2.2 3 2.9 0.9
1.3 2.1 2
2.1 4.3 5.1 0.2
1.9 4.1 4.9
5 3.2 9.9 3.5
1.5 0.3 6.4
Bolinder (2012)
DAP 10 mg PBO
3 0.9
2.1
2.7 0.1
2.8
NR
–
Nauck (2011)
DAP 10 mg GLIP 20 mg
3.22 1.44
4.65
4.3 0.8
5.1
NR
–
Strojek (2011)
DAP 2.5 mg DAP 5 mg DAP 10 mg PBO
1.18 1.56 2.26 0.72
0.46 0.84 1.54
4.7 4 5 1.2
3.5 2.8 3.8
7.2 1 2.8 0.9
6.3 0.1 1.9
Rosenstock (2012)
DAP 5 mg DAP 10 mg PBO
0.09 0.14 1.64
1.55 1.78
0.8 3.4 1.3
2.1 4.7
NR
–
Jabbour (2013)
DAP 10 mg PBO
2.1 0.3
1.9
6 5.1
0.9
NR
–
Wilding (2009)
DAP 10 mg DAP 20 mg PBO
4.5 4.3 1.9
2.6 2.4
0.7 5.5 2.1
2.8 7.6
4.2 15.4 3
7.2 12.4
Wilding (2012)
DAP 2.5 mg DAP 5 mg DAP 10 mg PBO
1 1 1.6 0.8
1.8 1.8 2.4
5.3 4.3 4.1 1.5
3.8 2.8 2.6
NR
–
Wilding (2013)
DAP 2.5 mg DAP 5 mg DAP 10 mg PBO
1 1 1.5 1.8
2.8 2.9 3.3
NR 2.6 7.5 0.5
2.1 7
10.3 5.2 1.8 3.5
6.8 8.7 5.3
DAP, dapagliflozin; PBO, placebo; MET, metformin; GLIP, glipizide; vs., versus; D, change in; SBP, systolic blood pressure; LDL, low density lipoprotein cholesterol; NR, not reported.
Small changes in HDL-C (+2.1% to +9.3%), triglyceride (0.9% to 10.6%) and LDL-C (0.5% to +9.5%) were observed overall in patients receiving dapagliflozin therapy. Mean percent placebo-adjusted changes from baseline for dapagliflozin 10 mg were: total cholesterol +1%; HDL-C +1.7%; LDL-C +0.8%; and triglycerides 4.7% [75]. Table 3 summarizes relevant data on
the effect of dapagliflozin on body weight, systolic blood pressure, and LDL-C.
5.1.6.
Safety
Dapagliflozin was well tolerated both as monotherapy and as an add-on to other antidiabetes agents. The most frequent
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
10
diabetes research and clinical practice xxx (2014) xxx–xxx
adverse events were headache, diarrhea, back pain, and upper respiratory tract infections and there was no difference in their frequency across treatment arms [53,62,63,66]. Dropouts due to adverse events were rare. Hypoglycemic events were infrequent and not significantly different from placebo in patients treated with dapagliflozin monotherapy (0–3%) [62– 64] or as add-on metformin [66,67], pioglitazone [70], or sitagliptin (3% each) [71]. Hypoglycemic events, however, were significantly more frequent when dapagliflozin was used as add-on to insulin (27% vs. 13% in the placebo group at 48 weeks and 66% vs. 62% in the placebo group at 104 weeks) [72,73] or glimepiride therapy (8% vs. 5% in the placebo arm at 24 weeks) [69]. Consistent with several trials on SGLT2 inhibitors, dapagliflozin was associated with an increased incidence of genital infections, predominantly in females, and a modest increase in urinary tract infections when compared to placebo (Table 8). In an analysis, pooled data from 12 phase 2b and phase 3 clinical trials of dapagliflozin as monotherapy or as add-on to other antidiabetes agents, a higher incidence of genital infections, e.g. vulvovagnitis and balanitis, was observed in the dapagliflozin groups (4–6%) than placebo groups (1%) [76]. Another analysis of the same data observed an equivalent rate of urinary tract infections in the dapagliflozin groups (4–6%), but this was not significantly different from the placebo group (4%) [77]. Most of these events were reported to be mild to moderate in intensity and responded to a course of standard antimicrobial treatment. The analysis also demonstrated that higher doses of dapagliflozin were associated with a higher degree of glucosuria but there was no clear dose–response relationship between dapagliflozin and genital or urinary tract infections [76,77]. When added to standard care, dapagliflozin was shown to be effective and safe in older patients (mean age 63–64 years) with T2DM and history of cardiovascular disease [78]. Dapagliflozin therapy is associated with a mild but noticeable increase in hematocrit (Table 8). Mean placebo-adjusted changes from baseline in hematocrit were +1.7% for dapagliflozin 10 mg/day [79]. Despite no evidence of increased teratogenicity in animal studies, an increased number of breast and bladder cancers were reported among dapagliflozin users in clinical trials [80]. Breast cancer was reported in ten patients in the dapagliflozin group and three patients in the placebo group [81]. Also, there were 9 cases of bladder cancer in the treatment arm vs. one case in the control group. Most of the cases of bladder cancer, however, had hematuria at baseline suggesting a possible preexisting cancer [81]. Also, when considering all types of cancers, there is no increased risk with the use of dapagliflozin compared to placebo or comparator [82]. Currently, dapagliflozin is not recommended for patients with bladder cancer [79].
5.1.7.
Dosing recommendations
The manufacturers, Bristol–Myers Squibb and AstraZeneca, market dapagliflozin as Farxiga1. It is available as 10 mg tablets taken orally once daily at any time of day with or without food [79].
5.1.8. Use in special populations 5.1.8.1. Renal impairment. Dapagliflozin is not recommended for patients with T2DM and moderate to severe renal
impairment, end stage renal disease, or patients on dialysis [79]. In a 24-week study on type 2 diabetics who have moderate renal impairment (eGFR = 30–59 ml/min/1.73 m2] the mean reduction in HbA1c from baseline was not significantly different between dapagliflozin (0.43%) and placebo (0.32%) as expected since the efficacy of SGLT2 inhibitors requires adequate filtered load of glucose [83]. No dosage adjustment is indicated in patients with mild renal impairment [79]. Dapagliflozin is not recommended for use in patients receiving loop diuretics or who are volume depleted [79].
5.1.8.2. Hepatic impairment. There is limited experience in clinical trials in patients with hepatic impairment. No dosage adjustment is necessary for patients with mild or moderate hepatic impairment. In patients with severe hepatic impairment, a starting dose of 5 mg is recommended. If well tolerated, the dose may be increased to 10 mg [79]. 5.1.8.3. Older patients. Due to the limited experience in patients 75 years and older, initiation of dapagliflozin therapy is not recommended [79]. 5.1.8.4. Pregnancy and breastfeeding. Dapagliflozin therapy is not recommended in this population. No data are available [79]. 5.2.
Canagliflozin
In March 2013, canagliflozin became the first SGLT2 inhibitor to be approved in the United States [84]. It was similarly approved recently in the European Union [85]. Canagliflozin has been studied extensively in adult patients with T2DM as monotherapy [86,87], and as add-on to metformin therapy [88– 90], metformin plus sulfonylurea [91,92], metformin plus pioglitazone [93], and insulin [94]. The efficacy and safety of canagliflozin were also examined in vulnerable populations such as those with renal impairment [95] and the elderly [96]. Canagliflozin at 100 mg and 300 mg daily doses achieved significant HbA1c reductions compared to placebo and selected active comparators, with slightly greater reductions with canagliflozin 300 mg once daily (Table 4).
5.2.1.
Efficacy in monotherapy trials
Two published trials examined canagliflozin as monotherapy. The first trial studied 584 patients with T2DM who were drug naı¨ve [86]. At 26 weeks, canagliflozin resulted in a significant placebo-corrected reduction from baseline in HbA1c (canagliflozin 100 mg/day, 0.91%; canagliflozin 300 mg/day, 1.17%). In a separate study, Inagaki et al. [87] compared canagliflozin monotherapy with placebo in 383 Japanese patients with T2DM who were treatment naı¨ve over 12 weeks. Relative to placebo, significant reductions in HbA1c were observed in all canagliflozin groups relative to placebo (e.g. 0.91% and 0.99% with canagliflozin 100 mg and 300 mg/day, respectively). In both studies, canagliflozin achieved significant reductions in FPG and postprandial glycemic parameters. The beneficial effect on postprandial glucose is likely mediated through delaying intestinal glucose absorption via SGLT1 inhibition in addition to increasing UGE via SGLT2 inhibition [97].
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
Author (year)
n Patients
Monotherapy trials, placebo controlled Stenlof (2013) 584
Inagaki (2013)
383
Duration (wk)
Initial HbA1c %
26
8.0
173
12
8.1
165.6
Add-on to metformin trials, placebo and active comparator controlled Rosenstock (2012) 451 12 7.8
Initial FBG (md/dl)
Drug and dosea Background treatment Drug naı¨ve
Drug naı¨ve or washed out
162
PBO and SIT
7.9
169.2
PBO and SIT
Add-on to metformin trials, active-comparator controlled Cefalu (2013) 1450 52
7.8
NR
GLIM
Add-on to other AHAs trials, placebo controlled Wilding (2013) 469
8.1
NR
Trial regimen Add-on to MET + SU vs. PBO
Lavalle Gonzalez (2013)
Matthews (2013)
Yale (2013)
Bode (2013)
1020
1718
269
714
52
26
18
26
26
8.3
8
7.7
169.2
164.3
156.8
Add-on to INS vs. PBO
Add-on to different AHAs vs. PBO
Add-on to different AHAs vs. PBO
DHbA1c from baseline (%)
DHbA1c vs. comparator (%)
DFBG from baseline (mg/dl)
DFBG vs. comparator (mg/dl)
CAN 100 mg CAN 300 mg PBO CAN 50 mg
0.77 1.03 0.14 0.61
CAN 100 mg CAN 200 mg CAN 300 mg PBO
0.8 0.79 0.88 0.11
0.69 0.68 0.77
33.1 36.1 38.3 3
30.1 33.1 35.3
CAN 50 mg CAN 100 mg CAN 200 mg CAN 300 mg CAN 300 mg BID SIT PBO CAN 100 mg CAN 300 mg SIT 100 mg PBO
0.79 0.76 0.7 0.92 0.95 0.74 0.22 0.73 0.88 0.73 0.6
0.57 0.54 0.48 0.7 0.73 0.52 vs. PBO
16.2 25.2 27 25.2 23.4 12.6 3.6 26.2 35.2 17.7 17.8
19.8 28.8 30.6 28.8 27 16.2 vs. PBO
CAN 100 mg CAN 300 mg GLIM
0.82 0.93 0.81
0.01 0.12
25.2 27 18
7.2 9
CAN 100 mg
0.85
0.71
18.0
15.0
CAN 300 mg PBO CAN 100 mg
1.06 0.13 0.63
0.92
30.6 3.6 18.0
27.6
CAN 300 mg PBO CAN 100 mg
0.72 0.01 0.33
0.73
CAN 300 mg PBO CAN 100 mg
0.44 0.03 0.6
0.4
CAN 300 mg PBO
0.73 0.03
0.7
0.91 1.16 0.50
0.13 0.28 0.13 vs. PBO
0.65
0.3
0.57
27 34.2 9 24.7
27.4 39.1 21.7
8.4 17.4 0.1 vs. PBO
22.0
25.0 4.0 14.9
29.0
11.7 0.5 18.1
12.2
20.3 7.4
27.7
diabetes research and clinical practice xxx (2014) xxx–xxx
15.4
25.5
11
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
Table 4 – Canagliflozin in major clinical trials.
DIAB-6018; No. of Pages 26
12
5.4 0.66 SIT 100 mg
Once a day unless specified; CAN, canagliflozin; PBO, placebo; MET, metformin; GLIP, glipizide; GLIM, glimepiride; SU, sulfonylureas; PIO, pioglitazone; INS, insulin; SIT, sitagliptin; AHAs, antihyperglycemic agents; n, number; BID, twice daily; wk, weeks; FPG, fasting plasma glucose; vs., versus; D, change in; NR, not reported.
a
25.2 vs. SIT 1.03 CAN 300 mg Add-on to MET + SU vs. SIT 167.4 8.1 Add-on to other AHAs trials, active comparator controlled Schernthaner (2013) 755 52
0.77 1.03 0.26 CAN 300 mg PBO
0.37 vs. SIT
30.6
– NR 0.63 0.89 CAN 100 mg NR 7.9 26 342 Forst (2012)
Duration (wk) n Patients Author (year)
Table 4 (Continued )
Initial HbA1c %
Initial FBG (md/dl)
Add-on to MET + PIO vs. PBO
Drug and dosea
DHbA1c from baseline (%)
DHbA1c vs. comparator (%)
DFBG from baseline (mg/dl)
DFBG vs. comparator (mg/dl)
diabetes research and clinical practice xxx (2014) xxx–xxx
5.2.2.
Efficacy in add-on trials
In add-on to metformin studies, canagliflozin was superior to placebo and sitagliptin, and non-inferior to glimepiride in achieving glycemic targets [88–90]. In a phase 3 trial, 1020 patients with T2DM who were being treated with metformin were randomized to receive canagliflozin 100 mg, 300 mg, sitagliptin 100 mg, or placebo [89]. At week 26, daily canagliflozin at 100 mg and 300 mg significantly reduced HbA1c relative to placebo (0.62% and 0.77%, respectively). At week 52, canagliflozin 100 mg and 300 mg demonstrated noninferiority, and canagliflozin 300 mg demonstrated superiority, to sitagliptin in lowering HbA1c (0.73%, 0.88%, and 0.73%, respectively). Both canagliflozin doses significantly reduced FPG vs. placebo (week 26) and sitagliptin (week 52). In a separate study, canagliflozin add-on to metformin was noninferior to glimepiride and metformin combination [90]; and at 300 mg/day canagliflozin provided greater HbA1c reductions compared to glimepiride (0.93% vs. 0.81%). Additionally, a more favorable FPG profile was observed after 52 weeks with canagliflozin 100 mg/day and 300 mg/day compared to glimepiride (24.3 mg/dl to 27.5 mg/dl vs. 18.4 mg/dl). Canagliflozin add-on was shown to be beneficial and safe in patients inadequately controlled with insulin with or without additional oral agents [94]. Canagliflozin 100 mg and 300 mg daily for 18 weeks achieved significant placebo-adjusted reductions in HbA1c (0.64% and 0.73%) and FPG (22 and 29 mg/dl). The efficacy of canagliflozin was sustained when used as a third agent in patients with T2DM. In a study by Wilding et al. [91] canagliflozin add-on to a background treatment with metformin plus sulfonylurea significantly reduced HbA1c and FPG. Placebo-adjusted changes from baseline in HbA1c were 0.72% for canagliflozin 100 mg/day and 0.93% for 300 mg/ day. These reductions were sustained at week 52 (0.75% and 0.97%). Schernthaner et al. [92] compared canagliflozin 300 mg/day with sitagliptin 100 mg/day in patients inadequately controlled with metformin plus sulfonyurea. At 52 weeks, canagliflozin 300 mg was superior to sitagliptin 100 mg in reducing HbA1C (1.03% and 0.66%, respectively) [92]. In patients inadequately controlled with a combination therapy of metformin and pioglitazone, the addition of canagliflozin resulted in significant placebo corrected reductions in HbA1c (0.63% and 0.77% with canagliflozin 100 mg and 300 mg, respectively) [93]. In a study of 269 patients with T2DM and stage-3 chronic kidney disease (an eGFR 30 and <50 ml/min/ 1.73 m2), canagliflozin at 100 mg and 300 mg/day was generally well tolerated and led to a lower, but still significant, reduction in HbA1c relative to placebo (0.30% and 0.41%) [95]. Compared to placebo group, more patients treated with canagliflozin had increases in serum creatinine (9% and 10% vs. 4%) and blood urea nitrogen (9% and 6% vs. 2%). The changes in renal function occurred more rapidly in the first three weeks of treatment, but trended back towards baseline over time.
5.2.3.
Effects on weight
Canagliflozin monotherapy, both at 100 and 300 mg doses, was associated with a significant reduction in body weight from baseline compared to placebo (2.2% and 3.3%, respectively) [86]. When added to metformin, canagliflozin was superior to glimepiride in reducing body weight at 52 weeks (3.7 kg with 100 mg and 4.0 kg with 300 mg daily, vs. +0.7 kg with
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
glimepiride) [90]. Compared with sitagliptin 100 mg, canagliflozin 300 mg add-on to metformin plus sulfonylurea was superior in achieving weight reduction (2.3 kg vs. +0.1 kg) [92]. Furthermore, weight reduction was an additional benefit when canagliflozin was added to insulin with or without additional oral agents (1.9 kg with 100 mg and 2.4 kg with 300 mg of canagliflozin, both placebo-corrected) [94].
5.2.4.
Effects on blood pressure
Across the placebo-controlled phase 3 studies, the placebosubtracted mean changes from baseline in systolic blood pressure ranged from 2.2 to 5.7 mmHg with canagliflozin 100 mg/day and from 1.6 to 7.9 mmHg with canagliflozin 300 mg/day [55]. At 300 mg/day, canagliflozin achieved a significant reduction (5.1 mmHg) in systolic blood pressure compared to sitagliptin 100 mg/day (+0.9 mmHg) over 52 weeks [92]. Mean changes from baseline in heart rate were 0.1 and 0.7 beats/min with canagliflozin and sitagliptin, respectively. Additionally, treatment with canagliflozin was associated with larger mean decreases from baseline in systolic blood pressure compared to glimepiride (mean differences relative to glimepiride = 3.48 and 4.76 mmHg with canagliflozin 100 mg and 300 mg doses, respectively) [90]. The reductions in SBP were sustained over the 26-week treatment periods in the placebo-controlled studies and the 52-week treatment periods in the active controlled studies [55].
5.2.5.
Effects on lipids
Mean percent increases in HDL-C were observed in the canagliflozin 100 mg and 300 mg groups relative to placebo (ranging from 0.8 to 6.8% with the 100 mg dose and 0.9 to 8.4% with the 300 mg dose) in phase 3 studies, and were significantly greater than those observed with placebo in most studies [55]. While larger mean percent reductions in fasting triglycerides with the canagliflozin groups compared with placebo group were seen in most studies, the treatment difference was generally small and often not statistically significant for individual studies. In a pooled analysis of the placebo-controlled studies, the placebo-subtracted mean percent changes from baseline for LDL-C were 4.5% and 8.0%, for the canagliflozin 100 mg and 300 mg groups, respectively. In 52-week studies, no consistent further increases in LDL-C were observed from weeks 26 to 52 [55]. With regard to the clinical implications of the increase in LDL-C observed with canagliflozin, data from the Cholesterol Treatment Trialists Collaboration [98] suggest that a population mean increase of 8.15 mg/dl in LDL-C for 5 years (the extent of increase observed with the canagliflozin 300 mg group) could translate into a 4% to 5% increase in the incidence of major adverse cardiovascular events over that same time period [55]. This estimated change in cardiovascular risk assumes no change in other cardiovascular risk factors. However, canagliflozin treatment is proven to improve other cardiovascular risk surrogates such as blood pressure, weight, HDL-C, and glycemic control, which may counterbalance the slight rise in LDL-C. Table 5 summarizes relevant data on the effect of canagliflozin on body weight, systolic blood pressure, and LDL-C.
5.2.6.
13
Safety
Canagliflozin was generally well tolerated in the published trials, and the incidence of serious adverse events was comparable to control. Consistent with dapagliflozin trials, hypoglycemia was rare with canagliflozin monotherapy and with combination therapy that did not include insulin or an insulin secretagogue [55,99]. When added to metformin, canagliflozin therapy over 2 years was associated with a significantly lower risk of hypoglycemia compared with glimepiride (6.8% and 8.2% with canagliflozin 100 mg and 300 mg per day respectively, vs. 40.9% with glimepiride) [90]. Furthermore, rates of severe hypoglycemia were lower with canagliflozin 100 and 300 mg relative to glimepiride (0.6%, 0.2%, and 3.3%, respectively). When added to a background therapy of metformin plus sulfonylurea, both canagliflozin 300 mg/day and sitagliptin 100 mg/day had a similar rate of documented hypoglycemia (43.2% vs. 40.7%) and severe hypoglycemia (4% and 3.4%, respectively) [92]. Severe hypoglycemia was defined as events requiring the assistance of another person, or with loss of consciousness or a seizure regardless of whether biochemically documented. Like dapagliflozin, two of the most common adverse events in patients treated with canagliflozin were female genital mycotic infections and urinary tract infections (Table 8) [100,101]. Male genital mycotic infections were less common (2%) and occurred predominantly in uncircumcised males and males with a prior history of balanitis or balanoposthitis [99]. In analysis of canagliflozin trials, the incidence of urinary tract infections in the broad population (n = 9439) was 5.5% with canagliflozin 100 mg, 5.7% with canagliflozin 300 mg, and 4.3% with placebo or active comparator [55]. Upper urinary tract infections were rare (0.2% across all groups). A recent safety meta-analysis of SGLT2 inhibitors and DPP-4 inhibitors, concluded there were no statistically significant differences in the incidence of urinary tract infections in the comparison of both, DPP-4 inhibitors (RR = 1.15, 95% CI: 0.80–1.65) and SGLT2 inhibitors (RR = 1.02, 95% CI: 0.54–1.91) with the placebo [102]. However, the analysis concluded that treatment with SGLT2 inhibitors resulted in a significantly increased risk of genital infection compared with placebo (RR = 2.36, 95% CI: 1.17–4.74). Patients developing genital mycotic infections responded well to a standard course of antimicrobial therapy, usually without discontinuation of canagliflozin during clinical trials [55]. Table 8 summarizes the rates of adverse events of special interest. Owing to its mild diuretic effect, several adverse events related to reductions in intravascular volume were observed in canagliflozin trials, such as orthostatic hypotension, postural dizziness, thirst, increased urination, and dehydration [99]. Factors associated with volume-related adverse effects were older age (75 years), concomitant use of loop diuretics, and moderate renal impairment (eGFR between 30 and 59 ml/min/ 1.73 m2). The incidence rates of adverse effects related to reduced intravascular volume in the broad population (n = 9439) were 2.3%, 3.4%, and 1.5% with canagliflozin 100 mg, 300 mg, and placebo or comparator, respectively [55]. Small, transient, and reversible decreases in eGFR were observed with canagliflozin therapy consistent with its hemodynamic effect [55]
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
14
diabetes research and clinical practice xxx (2014) xxx–xxx
Table 5 – Important seconday outcomes in canagliflozin clinical trials. Author (year)
Drug and dosea
Stenlof (2013)
CAN 100 mg CAN 300 mg PBO
2.5 3.6 0.3
2.2 3.3
3.3 5.0 0.4
3.7 5.4
8.5 4.6 6.2
2.3 1.6
Inagaki (2013)
CAN 100 mg CAN 300 mg PBO
2.5 3.2 0.8
1.7 2.4
7.1 8.7 1.2
5.9 7.5
4.9 5.5 0.9
5.8 6.4
Rosenstock (2012)
CAN CAN CAN CAN SIT PBO
2.3 2.4 3.0 2.9 0.5 0.9
1.4 1.5 2.1 2.0 0.4 vs. PBO
1.0 2.1 4.9 3.6 0.8 1.3
0.3 0.8 3.6 2.3 0.5 vs. PBO
4.9 3.2 1.1 7.8 8 NR
–
Lavalle Gonzalez (2013)
CAN 100 mg CAN 300 mg SIT 100 mg PBO
3.3 3.7 1.2 NR
Cefalu (2013)
CAN 100 mg CAN 300 mg GLIM
3.7 4.0 0.7
4.4 4.7
3.3 4.6 0.2
3.5 4.8
NR
–
Wilding (2013)
CAN 100 mg CAN 300 mg PBO
2.0 2.6 0.6
1.4 2.0
4.9 4.3 2.7
2.2 1.6
0.8 4.3 0
0.8 4.3
Matthews (2013)
CAN 100 mg CAN 300 mg PBO
1.8 2.3 0.1
1.9 2.4
2.6 4.4
6.3 6.6
Yale (2013)
CAN 100 mg CAN 300 mg PBO
1.2 1.4 0.2
1.4 1.6
6.1 6.4 0.3
5.7 6.1
3.5 3.1 3.4
0.1 7
Bode (2013)
CAN 100 mg CAN 300 mg PBO
2.4 3.1 0.1
2.3 3.0
3.5 6.8 1.1
4.6 7.9
13.2 13 6.2
7 6.8
Forst (2012)
CAN 100 mg CAN 300 mg PBO
NR
2.7 3.7
NR
4.1 3.5
NR
–
Schernthaner (2013)
CAN 300 mg SIT 100 mg
2.3 0.1
2.4 vs. SIT
5.1 0.9
5.9 vs. SIT
6.3 0.4
5.9 vs. SIT
100 mg 200 mg 300 mg 300 mg BID
DWeight from baseline (kg)
DWeight vs. comparator (kg)
DSBP from baseline (mmHg)
DSBP vs. comparator (mmHg)
DLDL from baseline (mg/dl)
DLDL vs. comparator (mg/dl)
4.3 4.3 3.1 NR
3.5 4.7 0.7 NR
a Once a day unless specified; CAN, canagliflozin; PBO, placebo; MET, metformin; GLIM, glimepiride; SIT, sitagliptin; SBP, systolic blood pressure; LDL, low density lipoprotein cholesterol; BID, twice daily; vs., versus; D, change in; NR, not reported.
Effects of canagliflozin on the macrovascular complications of T2DM remain to be determined. The CANagliflozin Cardiovascular Assessment Study (CANVAS) is an ongoing prospective placebo-controlled trial to evaluate the effects of canagliflozin on the risk of cardiovascular disease and to assess safety and tolerability in patients with inadequately controlled T2DM and increased cardiovascular risk [103]. Intermittent data (CANVAS sub-study) have been released and, so far, have been reassuring [104,105]. However, final results are not expected until 2015. Small reductions in bone density and numerical increases in the number of low-trauma upper extremity fractures were observed in older patients receiving canagliflozin therapy relative to placebo [55,106]. The reduction in bone density is likely explained by the associated weight loss. Yet, there were
no meaningful changes in serum or urine calcium excretion, and there were variable, but overall no meaningful changes in 1,25-dihydroxy vitamin D levels. There was a transient increase in PTH at week 3 with no substantive changes at week 12 (phase 2), or at weeks 26 or 52 (phase 3). The FDA has required the manufacturer to conduct a study to assess the effects of canagliflozin on bone health [106].
5.2.7.
Dosing recommendations
Janssen Pharmaceuticals markets canagliflozin as Invokana1. It is administered once daily before the first meal of the day [99]. The recommended starting dose is 100 mg once daily, but can be increased to the maximum dose of 300 mg in patients with an eGFR 60 ml/min/1.73 m2 who require additional glucose control.
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
5.2.8. Use in special populations 5.2.8.1. Renal impairment. Canagliflozin has not been studied in advanced chronic kidney disease (eGFR < 30 ml/min/ 1.73 m2), end stage renal disease or dialysis patients [99]. Based on its pharmacological properties, canagliflozin is likely not effective in these patients. Patients with stage 3 chronic kidney disease who received canagliflozin achieved less glycemic efficacy compared to patients with normal renal function and had a higher incidence of renal-related adverse effects such as hypovolemia and hyperkalemia [55,99]. Canagliflozin therapy was also associated with transient decreases in eGFR and increases in blood urea nitrogen in such patients [95]. However, similar, but smaller changes were also observed in patients receiving placebo, suggesting that these changes might be attributable to the underlying diabetic nephropathy rather than to the intervention. Current prescription guidelines for canagliflozin recommend a maximum daily dose of 100 mg for patients with an eGFR between 46 and 59 ml/min/m2, and do not recommend it for eGFR < 45 ml/ min/m2 [99].
5.2.8.2. Hepatic impairment. No dosage adjustments are necessary in patients with mild to moderate hepatic impairment [99]. Canagliflozin has not been studied in patients with severe hepatic impairment and is not recommended in this patient population. 5.2.8.3. Older patients. In clinical trials, 2034 patients aged 65 years were treated with canagliflozin [99]. Older patients had a higher incidence of volume related adverse events such as hypotension, syncope and postural dizziness. These effects were more common with the 300 mg daily dose relative to 100 mg doses [96]. Thus, 100 mg maximum doses may be considered in older patients unless the benefits outweigh the risks of using the higher dose [99]. 5.2.8.4. Pregnancy and breastfeeding. Canagliflozin has not been studied extensively in this population and is therefore considered a category C agent [99]. The extent of canagliflozin excretion in human milk is unknown. 5.3.
Other considerations
The reduction in body weight associated with SGLT2 inhibitors is noteworthy given that over 80% of patients with T2DM are overweight or obese [6,107], weight loss is difficult for patients with T2DM to achieve [6], and many antidiabetes treatments (including pioglitazone) are associated with weight gain. Owing to the mechanism of action of SGLT2 inhibitors, involving glucosuria and the associated osmotic duiresis, there is a concern of developing side effects such as dehydration, electrolytes disturbance, and renal dysfunction. Fortunately, this is generally not supported by the results of the studies discussed above. Further reassurance comes from the evidence that subjects with familial renal glucosuria (FRG), a rare condition caused by a mutation in SGLT2 gene and a disease model of SGLT2 inhibition, do not develop dehydration, electrolytes disturbance, or hypoglycemia [108]. Given the novelty of these agents it is premature to describe their long-term effect on important end points such as cancer
15
risk and cardiovascular outcomes, including myocardial infarction and stroke. Time and further evidence from postmarketing studies are required to quantify any potential associations.
6. Selected SGLT2 inhibitors in clinical development 6.1.
Empagliflozin
Empagliflozin (BI 10773) is a potent and selective SGLT2 inhibitor being developed by Boehringer Ingelheim and Eli Lilly pharmaceuticals as a treatment for T2DM [109]. In a preclinical study, empagliflozin had the highest selectivity for SGLT2 over SGLT1 (>2500-fold), followed by tofogliflozin (>1875-fold), dapagliflozin (>1200 fold), ipragliflozin (>550-fold) and canagliflozin (>250-fold) [110]. Empagliflozin has been shown to induce UGE both in healthy volunteers [111] and in patients with T2DM [112]. A New Drug Application (NDA) has been submitted to the US FDA for empagliflozin [113]. To date, 8 major clinical trials (phases 2b and 3) addressing the efficacy and safety of empagliflozin in patients with T2DM have been published (Tables 6 and 7). Empagliflozin has been studied as monotherapy [114,115], and as add-on to metformin [116,117], two oral agents [118,119] and insulin [120]. Empagliflozin at 25 mg for 12 weeks significantly decreased HbA1c (0.72%), FPG (31.9 mg/dl), and body weight (1.2 kg vs.) relative to placebo in patients with T2DM [114]. In a placebo and active comparator controlled trial, 899 patients were randomized to empagliflozin 10 mg, empagliflozin 25 mg, sitagliptin, and placebo [115]. Compared with placebo, mean differences in change from baseline HbA1c at week 24 were 0.74% for empagliflozin 10 mg, 0.85% for empagliflozin 25 mg, and 0.73% for sitagliptin. Empagliflozin was beneficial when used as an add-on therapy. In a 12-week study in patients with T2DM inadequately controlled with metformin, empagliflozin add-on at 25 mg resulted in HbA1c reduction by up to 0.69% relative to placebo [117]. Additionally it led to significant reductions in FPG (up to 32.7 mg/dl vs. placebo) and weight (up to 1.7 kg vs. placebo). Sustained glycemic control and weight reduction were observed with empagliflozin therapy in a 78-week, openlabel extension of this trial [121]. In a separate trial, 666 patients inadequately controlled on metformin and sulfonylureas were randomized to once-daily empagliflozin 10 mg, empagliflozin 25 mg, or placebo [118]. At week 24, placebosubtracted changes from baseline in HbA1c were 0.65% and 0.60% for empagliflozin 10 mg and 25 mg doses, respectively. Additionally, empagliflozin significantly reduced mean daily glucose levels, weight, and systolic (but not diastolic) blood pressure vs. placebo. In a 78-week phase 3 clinical trial of empagliflozin as add-on to basal insulin in adults with T2DM, placebo-adjusted changes in HbA1c for empagliflozin 10 mg and 25 mg were 0.6% and 0.7% ( p < 0.001), respectively, at week 18, and 0.5% and 0.6%, respectively, at week 78 ( p < 0.001) [120]. The study included an 18-week fixed insulin dose period, after which the dose was adjusted at investigator’s discretion. At week 78, empagliflozin therapy resulted in significant reduction in the required daily insulin dose.
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
Author (year)
n, Patients
Duration (wk)
Initial HbA1c %
Initial FBG (md/dl)
Empagliflozin Monotherapy trials, placebo and active comparator controlled Ferrannini (2013) 408 12 7.9
899
24
Monotherapy trials, active comparator controlled Ferrannini (2013) 659 78
Add-on to metformin trials, placebo controlled Ha¨ring (2013) 637 24
D HbA1c vs. comparator (%)
DFBG from baseline (mg/dl)
DFBG vs. comparator (mg/dl)
Background treatment Drug naı¨ve or a single OADs
EMP 10 mg
0.48
0.58
28.9
29.7
EMP 25 mg MET PBO EMP 10 mg EMP 25 mg SIT 100 mg PBO
0.63 0.75 0.1 0.66 0.78 0.66 0.08
0.73 0.85 vs. PBO
31.1 29.7 0.8 19.4 24.5 6.8 11.7
31.9 30.5 vs. PBO
EMP 10 mg
0.34
0.22
30.4
4.4
EMP 25 mg MET
0.47 0.56
0.09
27.7 26
1.7
7.9
151.6
Drug naı¨ve
7.9
178
12-wk prior therapy with EMP
NR
D HbA1c from baseline (%)
0.74 0.85 0.73 vs. PBO
31.1 36.2 18.7 vs. PBO
NR
MET
EMP 10 mg EMP 25 mg PBO
0.7 0.77 0.13
0.57 0.64
20.0 22.3 6.4
26.0 28.6
Add-on to metformin trials, placebo and active comparator controlled Rosenstock (2013) 495 12 7.9
NR
MET
EMP 10 mg EMP 25 mg EMP 50 mg SIT PBO
0.56 0.55 0.49 0.45 0.14
0.7 0.69 0.63 0.59 vs. PBO
22.1 26.8 27.9 12.2 5.0
27.2 31.9 32.9 17.2 vs. PBO
Add-on to metformin trials, active-comparator controlled Ferrannini (2013) 166 78 7.9
178
SIT
EMP 10 mg EMP 25 mg SIT
0.34 0.63 0.4
0.06 0.23
21.3 31.8 16.0
5.3 15.8
Add-on to other AHAs trials, placebo controlled Ha¨ring (2013) 666 24
8.1
152.8
Trial regimen Add-on to MET + SU vs. PBO
EMP EMP PBO EMP EMP PBO EMP EMP PBO
0.82 0.77 0.17 0.59 0.72 0.11 0.48 0.64 0.02
0.64 0.59
23.2 23.2 5.6 16.9 22.0 6.5 10.0 15.0 3.0
28.8 20.9
0.76 0.47
1.23
39.9 5.9
45.8
Kovacs (2013)
498
24
8.1
151.7
Add-on to PIO MET vs. PBO
Rosenstock (2013)
494
78
8.2
142.1
Add-on to basal INS vs. PBO
16
8.3
175
Background treatment Drug naı¨ve or washout
Ipragliflozin Monotherapy trials, placebo controlled Kashiwagi (2011) 129
10 mg 25 mg 10 mg 25 mg 10 mg 25 mg
IPR 50 mg PBO
0.48 0.61 0.46 0.62
23.4 28.4 13.0 18.0
diabetes research and clinical practice xxx (2014) xxx–xxx
Roden (2013)
NR
Drug and dosea
DIAB-6018; No. of Pages 26
16
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
Table 6 – Selected SGLT2 inhibitors in clinical trials.
Wilding (2013)
343
12
Add-on to other AHAs trials, placebo controlled Kashiwagi (2012) 242 24 Kashiwagi (2012)
Schwartz (2011)
151
61
LX4211 Monotherapy trials, placebo controlled Zambrowicz (2012) 36
24
Drug naı¨ve or washout
8.32
168.1
MET
7.8
154.8
MET
8.36
178
SU
8.32
171.5
PIO
4
NR
NR
Trial regimen Mono and add-on to OADs vs. PBO
4
8.3
185
Background treatment NR
8.1
172
MET
Add-on to metformin trials, placebo controlled Rosenstock (2012) 299 12
IPR 50 mg IPR 150 mg IPR 300 mg PBO MET
0.39 0.47 0.55 0.26
IPR 50 mg PBO IPR 50 mg IPR 150 mg IPR 300 mg PBO
0.87 0.38 0.65 0.72 0.79 0.31
1.29
IPR 50 mg PBO IPR 50 mg PBO
NR
1.14
NR
38
NR
0.88
NR
41
IPR 50 mg IPR 100 mg IPR 200 mg IPR 300 mg PBO
0.73 0.61 0.84 0.73 0.1
0.63 0.51 0.74 0.63
60.3 49.0 70.6 65.0 10.4
50.0 38.5 60.1 54.5
LX 150 mg LX 300 mg PBO
1.15 1.25 0.49
0.66 0.76
52 68 12
40 56
LX 75 mg LX 200 mg LX 200 mg BID LX 400 mg PBO
0.41 0.53 0.8 0.92 0.08
0.49 0.61 0.88 1
NR
0.65 0.73 0.81
19.8 23.4 30.2
0.72 vs. PBO
21.2 vs. PBO
0.34 -0.4 0.48
22 11 14.2 24.3 27.7 1.1
DIAB-6018; No. of Pages 26
Add-on to metformin trials, placebo controlled Goto (2012) 168 24
165
39.4 13.1 23.2 26.6
a Once a day unless specified; EMP, empagliflozin; IPR, ipragliflozin; LX, LX4211; PBO, placebo; mono, monotherapy; MET, metformin; SU, sulfonylureas; PIO, pioglitazone; INS, insulin; SIT, sitagliptin; OADs, oral antihyperglycemic drugs; AHAs, antihyperglycemic agents; n, number; BID, twice daily; wk, weeks; FPG, fasting plasma glucose; vs., versus; D, change in; NR, not reported.
diabetes research and clinical practice xxx (2014) xxx–xxx
17
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
Monotherapy trials, placebo and active comparator controlled Fonseca (2013) 412 12 7.9
DIAB-6018; No. of Pages 26
18
diabetes research and clinical practice xxx (2014) xxx–xxx
Table 7 – Important secondary outcomes in empagliflozin, ipragliflozin, and LX4211 trials. Author (year)
Empagliflozin Ferrannini (2013)
Roden (2013)
Ferranini (2013)
Ha¨ring (2013)
Rosenstock (2013)
Ferrannini (2013)
Ha¨ring (2013)
Kovacs (2013)
Rosenstock (2013)
Ipragliflozin Kashiwagi (2011) Fonseca (2013)
Goto (2012) Wilding (2013)
Kashiwagi (2012) Kashiwagi (2012) LX4211 Zambrowicz (2012)
Rosenstock (2012)
Drug and dosea
DWeight from baseline (kg)
DWeight vs. comparator (kg)
DSBP from baseline (mmHg)
DSBP vs. comparator (mmHg)
DLDL from baseline (mg/dl)
DLDL vs. comparator (mg/dl)
EMP 10 mg EMP 25 mg MET PBO EMP 10 mg EMP 25 mg PBO EMP 10 mg EMP 25 mg MET EMP 10 mg EMP 25 mg PBO EMP 10 mg EMP 25 mg EMP 50 mg SIT PBO EMP 10 mg EMP 25 mg SIT EMP 10 mg EMP 25 mg PBO EMP 10 mg EMP 25 mg PBO EMP 10 mg EMP 25 mg PBO
2.3 2.0 1.3 0.8 2.3 2.5 0.3 2.2 2.6 1.3 2.08 2.46 NR 2.7 2.6 2.9 0.8 1.2 3.1 4.0 0.4 2.2 2.4 0.4 1.6 1.5 0.3 2.2 2.0 0.7
1.6 1.3 0.5 vs. PBO
NR
–
0 0 9.6 vs. PBO
1.9 2.2
2.9 3.7 0.3 0.1 1.7 2 4.5 5.2 NR 4.4 8.5 3.2 2.2 1.8 3.3 3 1.8 4.1 3.5 1.4 3.1 4 0.7 4.1 2.4 0.1
2.6 3.4
1.2 1.2 10.8 1.2 NR
1.9 3.7
NR
–
–
NR
–
2.6 6.7 1.4 0.4 vs. PBO
10 5 8.9 7 1.2 NR
8.8 3.8 7.7 5.8 vs. PBO
1.5 3.9 0.8 3.5 1.6 0 NR
0.4 3.1
IPR 50 mg PBO IPR 50 mg IPR 150 mg IPR 300 mg MET IPR 50 mg PBO IPR 50 mg IPR 150 mg IPR 300 mg IPR PBO IPR 50 mg PBO IPR 50 mg PBO
NR
1.5
NR
3.2
NR
–
1.6 2.0 2.6 0.78 2.3 0.6 2.1 2.0 2.2 0.5 NR
0.7 1.1 1.7 0.12 vs. PBO 2.7
2.6 3 2.6 3.1 NR
NR
–
3.6
NR
1.6 1.5 1.7
3.3 2.2 4.3
NR
4.2
NR
–
NR
2.8
3.8 2.7 4.8 0.5 5.5 1.3 5.9 2.5
3.4
NR
–
LX 150 mg LX 300 mg PBO LX 75 mg LX 200 mg LX 200 mg BID LX 400 mg PBO
3 4 2.0 1.0 1.9 1.8 2.5 0.4
1 2
10 13 4 0.2 3.9 4.5 5.7 0.5
6 9
6 15 22 NR
16 7
0.9 1.3 1.6 2.0 1.5 1.4 1.7 0.4 vs. PBO 2.7 3.6 1.8 2.0 2.0 1.8 2.9 2.7
1.3
0.6 1.5 1.4 2.1
5.1 4.8 2.7 2.1 3.9 4.7 4.2 2.5
0.3 3.4 4 5.2
–
–
3.5 1.6 –
–
–
a Once a day unless specified; EMP, empagliflozin; IPR, ipragliflozin; LX, LX4211; PBO, placebo; MET, metformin; SIT, sitagliptin; SBP, systolic blood pressure; LDL, low density lipoprotein cholesterol; BID, twice daily; vs., versus; D, change in; NR, not reported.
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
Hypoglycemic events were not increased with empagliflozin therapy compared to placebo. A pooled analysis of data obtained from two phase 2b trials examined the effect of empagliflozin on blood pressure [122]. At week 12, significant reductions in mean SBP of 2.6 mmHg and 3.3 mmHg were observed with empagliflozin 10 mg and 25 mg, respectively, relative to placebo, and more pronounced reductions were seen in patients with higher SBP (>140 mmHg at baseline). There was no associated increase in heart rate, and the authors observed that changes in SBP from baseline to week 12 were not correlated with change in body weight or change in HbA1c, suggesting that the antihypertensive effects of empagliflozin are independent of its ability to reduce plasma glucose and body weight [122]. Empagliflozin was superior to metformin and sitagliptin in achieving weight reduction [123]. Adjusted mean reductions in body weight were 2.1 kg and 1.9 kg for empagliflozin 10 mg and 25 mg, respectively, vs. a reduction of 0.9 kg with metformin monotherapy. As add-on to metformin, empagliflozin 10 mg and 25 mg produced mean weight reductions of 2.9 and 3.8 kg, respectively, compared with a negligible change in weight (0.6 kg) with sitagliptin (Table 7). In a separate study, empagliflozin was well tolerated when used as add-on to pioglitazone alone or pioglitazone plus metformin for 24 weeks [119], and it led to significant improvements in glycemic control, body weight, and blood pressure (Tables 6 and 7). Significantly more patients receiving empagliflozin achieved >5% reduction from baseline in body weight at week 24 (14– 19%), compared with placebo (6%). Hypoglycemia was rare with this add-on and comparable to placebo, and there was no increase in the risk of side effects associated with pioglitazone. Hypoglycemia was infrequent and comparable to placebo with empagliflozin therapy [114–120,123]. Yet, the incidence of genital mycotic infections and female urinary tract infections was higher with empagliflozin therapy compared to placebo or active comparator (Table 8). But there was no increased incidence of male urinary tract infections with empagliflozin therapy compared to other groups. A single 50 mg dose of empagliflozin was well tolerated in patients with different stages of renal or hepatic impairment [124,125], and the authors concluded that no dose adjustment of empagliflozin is required for such patients. However, consistent with other SGLT2 inhibitors, UGE produced by empagliflozin decreased with increasing renal impairment and correlated with decreased eGFR [124]. In severe renal impairment or renal failure, empagliflozin therapy showed little to no effect on UGE. A large, 4-year, ongoing head-to-head trial is comparing empagliflozin with glimepiride as add-on to metformin in T2DM with insufficient glycemic control [126]. In addition to determining the effects of these treatments on glycemic control over the long term, this study will investigate their effects on beta-cell function, cardiovascular risk and renal function.
6.2.
Ipragliflozin
Ipragliflozin (ASP1941) is another selective SGLT2 inhibitor that is co-developed by Astellas and Kotobuki Pharmaceuticals [127]. It was recently approved in Japan for the treatment
19
of T2DM [128]. In healthy human subjects [129] and T2DM patients [130], ipragliflozin once daily was associated with a dose dependent UGE. In patients with T2DM, ipragliflozin therapy was associated with UGE of up to 90 g/day, and the amount of UGE positively correlated with plasma glucose levels and the eGFR [130]. In healthy subjects, ipragliflozin did not affect the pharmacokinetics of sitagliptin, pioglitazone, or glimepiride, and vice versa, suggesting that ipragliflozin can be safely co-administered with other oral antidiabetes agents [131]. Published clinical trials have examined ipragliflozin as monotherapy [132,133] and as add-on to metformin [134,135], SU or pioglitazone [136], and other antihyperglycemic agents [130] in patients with T2DM (Tables 6 and 7). In a 12-week dosefinding study, ipragliflozin doses 50 mg/day were comparable to metformin (1500 mg/day) in lowering HbA1c and FPG [133]. Similarly, in a 16-week, placebo-controlled, monotherapy trial in Japanese patients with T2DM, ipragliflozin (50 mg/ day) resulted in significant reductions from baseline in HbA1c (1.23%), FPG (46 mg/dl) and body weight (1.47 kg), compared with placebo [132]. Ipragliflozin was beneficial as an add-on to monotherapy in T2DM patients with inadequate glycemic control. In a 24-week metformin add-on trial, ipragliflozin (50 mg/day) significantly reduced HbA1c (1.29%), FPG (39.4 mg/dl), and body weight (1.69 kg) compared with placebo [134]. Ipragliflozin was generally well tolerated and was not associated with increased risk of hypoglycemia relative to placebo. However, it was associated with increased incidence of genital infections but not urinary tract infections [132,134]. A separate add-on study concluded that ipragliflozin (12.5–300 mg/day) and metformin (1500 mg/day) combination treatment for 12 weeks was well tolerated in patients with T2DM and that dose-dependent reduction in the HbA1c (up to 0.50%, vs. placebo) was observed in more patients treated with ipragliflozin than with placebo [135]. Treatment with ipragliflozin (50 mg/day) added to either an SU or pioglitazone for 24 weeks was well tolerated and significantly reduced HbA1c, body weight, and blood pressure, relative to placebo [136]. Similar to other SGLT2 inhibitors, more patients on ipragliflozin therapy reported diuresis-related adverse effects, such as polyuria and pollakauria, than placebo [136] (see Table 8).
6.3.
LX4211
LX4211 is a novel dual inhibitor of SGLT1 and SGLT2 glucose transporters that is being developed by Lexicon Pharmaceuticals for the treatment of T2DM [137]. In addition to inducing glucosuria through blocking glucose reabsorption by the kidney, LX4211 may also block glucose reabsorption in the small intestine via inhibiting SGLT1 [138]. Hence it may have enhanced benefits in patients with T2DM through its dual mechanism of action. The combination of LX4211 and DPP-4 inhibitors is hypothetically promising and has been tested in preclinical and clinical studies [139]. LX4211 plus sitagliptin was well tolerated and produced significant reductions in glucose levels throughout the day and decreases in insulin levels relative to sitagliptin monotherapy. Additionally, the combination led to synergistic increases in active GLP-1, with no reported GI adverse effects [139].
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
Reference (year)
a
Hypoglycemic events, %
Genital tract infections, %
Urinary tract infections, %
SGL2-i agent
PBO or comparator
SGL2-i agent (femalejmale)
PBO or comparator (femaljmale)
SGL2-i agent (femalejmale)
PBO or comparator (femalejmale)
Mono Mono vs. PBO vs. MET Mono Mono vs. MET Add-on to MET Add-on to MET Add-on to MET vs. GLIP Add-on to GLIM Add-on to PIO Add-on to INS Add-on to INS Add-on to INS Add-on to MET vs. Add-on to SIT MET vs. PBO
3 6–10 1 0–2 1 2–4 2 3 8 2 27 60 61–70 3 3
3 4 2 2 3 3 3 40 5 0 13 52 62 3 2
13 2–7 3–5 0–2 13 8–13 5j2 15 7 8–9 21 11 10–21j3–13 9 10
1 0 1 0 2 5 0 3 1 3 4 3 6j0 2 1
13 5–12 4–5 0–4 11 8 12j2 7 7 5–9 4 11 10–20j7 8 7
4 6 3 2 4 8 2 4 6 8 0 5 7j4 4 6
26 12 12 52 52 26 18 26 26 52
Mono Mono Add-on to Add-on to Add-on to Add-on to Add-on to Add-on to Add-on to vs. SITA
4 1–4 2–6 NR 6 35 49 52 6 43
3 0 2 – 34 18 37 36 3 41
7–9j3–6 2 3–8 10–11j2–5 11–14j7–8 19j7 10–12j4–8 3j2 13j5 12
4j0 0 2 1 1j2 5j1 2j1 0 2j0 2
5–7 0 3–9 5–8 6 8 3 6–8 7 4
4 0 6 7 5 8 2 6 5 6
12 24 24 12 24 24 78 78 78
Mono vs. MET vs. PBO Mono vs. PBO vs. SIT Add-on to MET Add-on to MET vs. SIT vs. PBO Add-on to MET + SU Add-on to PIO MET Add-on to INS Mono vs. MET Add-on to MET vs. SIT
NR <1 1–2 0–4 14 2 36 1–2 2
– <1 1 0 8 2 35 4 4
0–4 4–9j1–3 4–5 1–10 1–5j1 6–10j1–7 5–8 5–6j4–5 4j2–3
0 0 0 0 1 3j1 2 4j0 0
1–3 13–15j1–2 5–6 3–6 13–18j0–3 22–31j2–4 NR 6–7j0–7 16–23j2–3
1 9 5 3 3 23j1 7j0 15j10
16 12 24 12 24 24
Mono Mono Add-on Add-on Add-on Add-on
MET MET SU PIO
<1 1 0 0–6 1 1
0 0 0 3 1 0
3 0–12 5 0–4 7j2 9j0
0 1 0 2 2j0 0
0 2–13 2 1–7 10j0 14j0
<1 9 4 6 0j2 6j0
4 12
Mono Add-on to MET
0 NR
0 –
NR 3–5
– 0
8 2
0 2
24 12 24 12 24 24 24 104 24 24 12 48 104 24 24
to to to to
MET vs. SIT vs. PBO MET MET vs. GLIM MET + SU INS different AHAs different AHAs
vs. placebo unless specified; PBO, placebo; mono, monotherapy; MET, metformin; GLIP, glipizide; GLIM, glimepiride; SU, sulfonylureas; PIO, pioglitazone; INS, insulin; SIT, sitagliptin; AHAs, antihyperglycemic agents; n, number; BID, twice daily; wk, weeks; FPG, fasting plasma glucose; vs., versus; D, change in; NR, not reported.
diabetes research and clinical practice xxx (2014) xxx–xxx
Dapagliflozin Ferrannini (2010) List (2009) Ji (2013) Kaku (2013) Henry (2012) Bailey (2010) Bolinder (2012) Nauck (2011) Strojek (2011) Rosenstock (2012) Wilding (2009) Wilding (2012) Wilding (2013) Henry (2012) Jabbour (2013) Canagliflozin Stenlof (2013) Inagaki (2013) Rosenstock (2012) Lavalle Gonzalez (2013) Cefalu (2013) Wilding (2012) Matthews (2013) Yale (2013) Bode (2013) Schernthaner (2013) Empagliflozin Ferrannini (2013) Roden (2013) Ha¨ring (2013) Rosenstock (2013) Ha¨ring (2013) Kovacs (2013) Rosenstock (2013) Ferrannini (2013) Ferrannini (2013) Ipragliflozin Kashiwagi (2011) Fonseca (2013) Goto (2012) Wilding (2013) Kashiwagi (2012) Kashiwagi (2012) LX4211 Zambrowicz (2012) Rosenstock (2012)
Regimena
Duration (wk)
DIAB-6018; No. of Pages 26
20
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
Table 8 – Adverse events of special interest in SGLT2 inhibitors trials.
DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
In a 4-week, phase 2 trial, LX4211 (300 mg/day) produced significant reductions in HbA1c (0.76%) and FPG (56 mg/dl) relative to placebo [138]. In a 12-week metformin add-on trial in 299 patients with T2DM, LX4211 maximized UGE at 200 mg daily dose [140]; and it produced a dose-dependent decrease in HbA1c levels, up to 1% at 400 mg daily dose. Additionally, LX4211 add-on produced significant reductions in body weight (up to 2.1 kg) and systolic blood pressure (up to 5.2 mmHg), relative to placebo. LX4211 treatment significantly reduced triglyceride concentrations in all but the group receiving 200 mg LX4211 per day [140]. A phase 1 study is assessing the drug properties, safety, and tolerability of LX4211 in patients with T2DM and moderate to severe renal impairment [141], and a phase-2 study is examining the safety and efficacy of LX4211 in patients with T1DM [142].
7.
Conclusion
Current therapeutic agents for T2DM have not achieved the desired glycemic goals or have been associated with adverse effects that limit their efficacy. Newer agents with different or complementary mechanisms of action, and better safety profile, are needed. SGLT2 inhibitors are novel agents that improve glycemic targets in addition to ameliorating comorbidities commonly associated with T2DM, such as obesity and hypertension. Their unique, insulin-independent, mechanism of action makes them advantageous where other agents may fail as a result of the disease progression. This article reviews the currently approved SGLT2 inhibitors for the treatment of T2DM and other agents within the class that are in advanced stages of clinical development. The major disadvantage of SGLT2 therapy, perhaps, is the increased risk of genital mycotic infections, and to a lesser extent, urinary tract infections, in small percentage of patients, particularly in women. Additionally, since the efficacy of SGLT2 inhibitors requires adequate filtered load of glucose in the kidney, their efficacy diminishes in renal impairment. Yet, in the absence of moderate or severe renal impairment, SGLT2 inhibitors are associated with significant and sustained HbA1c reduction when used as monotherapy or in combination with other antidiabetes agents, with low incidence of hypoglycemia. Furthermore, they are simple to administer with once daily oral dosing, and they improve beta cell function, promote weight loss, and reduce blood pressure. Data so far are reassuring regarding the effect of SGLT2 inhibitors on important outcomes such as cardiovascular disease and cancer risk; however, longer duration of therapy is needed to study the effect of SGLT2 inhibitors on these outcomes. Dapagliflozin and canagliflozin are two competitive, reversible, and highly selective SGLT2 inhibitors recently became available for clinical use in management of T2DM. Clinical trials to date on these two agents have been well conducted and have shown significant and sustained HbA1c reduction when used as monotherapy or in combination with other antidiabetes agents. Multiple other SGLT2 inhibitors are currently undergoing preclinical testing.
21
Conflict of interest JEG is a consultant/member of the speaker bureau for AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Janssen Pharmaceuticals, MannKind, Merck, and Sanofi. FMH and MA have no conflicts.
references
[1] Executive summary: standards of medical care in diabetes—2014. Diabetes Care 2014:S5–13. [2] National Institute for Health and Clinical Excellence. The management of type 2 diabetes: 2010 NICE guidelines [Internet]. London, U.K.: National Institute for Health and Clinical Excellence; 2010, Available from http:// www.nice.org.uk/nicemedia/live/12165/44320/44320.pdf [accessed 30.10.13].. [3] Bennett WL, Odelola OA, Wilson LM, Bolen S, Selvaraj S, Robinson KA, et al. Evaluation of guideline recommendations on oral medications for type 2 diabetes mellitus: a systematic review. Ann Intern Med 2012;1(Pt 1):27–36. [4] Bailey C, Krentz A. Oral antidiabetic agents. In: Pickup J, Williams G, editors. Textbook of diabetes. 4th ed. Oxford: Blackwell Publishing Ltd.; 2010. [5] Kahn SE, Haffner SM, Heise MA, Herman WH, Holman RR, Jones NP, et al. Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl J Med 2006;23:2427–43. [6] Inzucchi SE, Bergenstal RM, Buse JB, Diamant M, Ferrannini E, Nauck M, et al. Management of hyperglycemia in type 2 diabetes: a patient-centered approach: position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2012;6:1364–79. [7] Philippe J, Raccah D. Treating type 2 diabetes: how safe are current therapeutic agents? Int J Clin Pract 2009;2: 321–32. [8] Tahrani AA, Bailey CJ, Del Prato S, Barnett AH. Management of type 2 diabetes: new and future developments in treatment. Lancet 2011;9786:182–97. [9] Krentz AJ, Bailey CJ. Oral antidiabetic agents: current role in type 2 diabetes mellitus. Drugs 2005;3:385–411. [10] Alsahli M, Gerich J. Hypoglycemia, chronic kidney disease and diabetes. Diabetic Hypoglycemia 2013;6(September (2)):12–6. [11] Gerich JE. Role of the kidney in normal glucose homeostasis and in the hyperglycaemia of diabetes mellitus: therapeutic implications. Diabet Med 2010;2:136– 42. [12] Gerich JE. Physiology of glucose homeostasis. Diabetes Obes Metab 2000;6:345–50. [13] Landau BR, Wahren J, Chandramouli V, Schumann WC, Ekberg K, Kalhan SC. Contributions of gluconeogenesis to glucose production in the fasted state. J Clin Invest 1996;2:378–85. [14] Stumvoll M, Meyer C, Mitrakou A, Nadkarni V, Gerich JE. Renal glucose production and utilization: new aspects in humans. Diabetologia 1997;7:749–57. [15] Consoli A, Kennedy F, Miles J, Gerich J. Determination of Krebs cycle metabolic carbon exchange in vivo and its use to estimate the individual contributions of gluconeogenesis and glycogenolysis to overall glucose output in man. J Clin Invest 1987;5:1303–10.
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
22
diabetes research and clinical practice xxx (2014) xxx–xxx
[16] Meyer C, Dostou JM, Welle SL, Gerich JE. Role of human liver, kidney, and skeletal muscle in postprandial glucose homeostasis. Am J Physiol Endocrinol Metab 2002;2:E419– 27. [17] Meyer C, Dostou JM, Gerich JE. Role of the human kidney in glucose counterregulation. Diabetes 1999;5:943–8. [18] Gerich JE. Hepatorenal glucose reciprocity in physiologic and pathologic conditions. Diabetes Nutr Metab 2002;5:298–302 [discussion 302–3]. [19] Cersosimo E, Garlick P, Ferretti J. Renal glucose production during insulin-induced hypoglycemia in humans. Diabetes 1999;2:261–6. [20] Stumvoll M, Meyer C, Kreider M, Perriello G, Gerich J. Effects of glucagon on renal and hepatic glutamine gluconeogenesis in normal postabsorptive humans. Metabolism 1998;10:1227–32. [21] Meyer C, Stumvoll M, Welle S, Woerle HJ, Haymond M, Gerich J. Relative importance of liver, kidney, and substrates in epinephrine-induced increased gluconeogenesis in humans. Am J Physiol Endocrinol Metab 2003;4:E819–26. [22] Stumvoll M, Chintalapudi U, Perriello G, Welle S, Gutierrez O, Gerich J. Uptake and release of glucose by the human kidney. Postabsorptive rates and responses to epinephrine. J Clin Invest 1995;5:2528–33. [23] Wright EM, Hirayama BA, Loo DF. Active sugar transport in health and disease. J Intern Med 2007;1:32–43. [24] Lee YJ, Lee YJ, Han HJ. Regulatory mechanisms of Na(+)/ glucose cotransporters in renal proximal tubule cells. Kidney Int Suppl 2007;106:S27–35. [25] Chen J, Feder J, Neuhaus I, Whaley J. Tissue expression profiling of the sodium–glucose co-transporter (SGLT) family: implication for targeting SGLT2 in type 2 diabetes patients (2493-PO). Diabetes 2008;57:A682. [26] Wright EM. Renal Na(+)-glucose cotransporters. Am J Physiol Renal Physiol 2001;1:F10–8. [27] Brown GK. Glucose transporters: structure, function and consequences of deficiency. J Inherit Metab Dis 2000;3:237–46. [28] Bakris GL, Fonseca VA, Sharma K, Wright EM. Renal sodium-glucose transport: role in diabetes mellitus and potential clinical implications. Kidney Int 2009;12:1272–7. [29] Silverman M, Turner J. Glucose transport in the renal proximal tubule. In: Windhager EE, editor. Handbook of physiology. New York: Oxford University Press; 1992. p. 2017–38. Copyright # 1992 by the American Physiological Society. By permission of Oxford University Press Inc.. [30] Zelikovic I. Aminoaciduria and glycosuria. In: Avner ED, Harmon WE, Niaudet P, editors. Pediatric nephrology. 5th ed. Philadelphia: Lippincott Williams & Wilkins; 2004. p. 701–28. [31] Santer R, Kinner M, Lassen CL, Schneppenheim R, Eggert P, Bald M, et al. Molecular analysis of the SGLT2 gene in patients with renal glucosuria. J Am Soc Nephrol 2003;11:2873–82. [32] Meyer C, Stumvoll M, Nadkarni V, Dostou J, Mitrakou A, Gerich J. Abnormal renal and hepatic glucose metabolism in type 2 diabetes mellitus. J Clin Invest 1998;3:619–24. [33] 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;6:E1049–56. [34] Mogensen CE. Maximum tubular reabsorption capacity for glucose and renal hemodynamcis during rapid hypertonic glucose infusion in normal and diabetic subjects. Scand J Clin Lab Invest 1971;1:101–9. [35] Farber SJ, Berger EY, Earle DP. Effect of diabetes and insulin of the maximum capacity of the renal tubules to reabsorb glucose. J Clin Invest 1951;2:125–9.
[36] Rahmoune H, Thompson PW, Ward JM, Smith CD, Hong G, Brown J. Glucose transporters in human renal proximal tubular cells isolated from the urine of patients with noninsulin-dependent diabetes. Diabetes 2005;12:3427–34. [37] Vestri S, Okamoto MM, de Freitas HS, Aparecida Dos Santos R, Nunes MT, Morimatsu M, et al. Changes in sodium or glucose filtration rate modulate expression of glucose transporters in renal proximal tubular cells of rat. J Membr Biol 2001;2:105–12. [38] Ehrenkranz JR, Lewis NG, Kahn CR, Roth J. Phlorizin: a review. Diabetes Metab Res Rev 2005;1:31–8. [39] Rossetti L, Smith D, Shulman GI, Papachristou D, DeFronzo RA. Correction of hyperglycemia with phlorizin normalizes tissue sensitivity to insulin in diabetic rats. J Clin Invest 1987;5:1510–5. [40] Wright EMI. Glucose galactose malabsorption. Am. J. Physiol. 1998;5(Pt 1):G879–82. [41] Isaji M. Sodium-glucose cotransporter inhibitors for diabetes. Curr Opin Investig Drugs 2007;4:285–92. [42] Tahrani AA, Barnett AH, Bailey CJ. SGLT inhibitors in management of diabetes. Lancet Diabetes Endocrinol 2013;2:140–51. [43] Fujimori Y, Katsuno K, Nakashima I, Ishikawa-Takemura Y, Fujikura H, Isaji M. Remogliflozin etabonate, in a novel category of selective low-affinity sodium glucose cotransporter (SGLT2) inhibitors, exhibits antidiabetic efficacy in rodent models. J Pharmacol Exp Ther 2008;1:268–76. [44] Fujimori Y, Katsuno K, Ojima K, Nakashima I, Nakano S, Ishikawa-Takemura Y, et al. Sergliflozin etabonate, a selective SGLT2 inhibitor, improves glycemic control in streptozotocin-induced diabetic rats and Zucker fatty rats. Eur J Pharmacol 2009;1–3:148–54. [45] Washburn WN. Development of the renal glucose reabsorption inhibitors: a new mechanism for the pharmacotherapy of diabetes mellitus type 2. J Med Chem 2009;7:1785–94. [46] Dobbins RL, O‘Connor-Semmes R, Kapur A, Kapitza C, Golor G, Mikoshiba I, et al. Remogliflozin etabonate, a selective inhibitor of the sodium-dependent transporter 2 reduces serum glucose in type 2 diabetes mellitus patients. Diabetes Obes Metab 2012;1:15–22. [47] Hussey EK, Kapur A, O‘Connor-Semmes RL, Tao W, Poo JL, Dobbins RL. Safety, pharmacokinetics and pharmacodynamics of Remogliflozin etabonate (SGLT2 Inhibitor) and metformin when co-administered in type 2 diabetes mellitus (T2DM) patients. Diabetes 2010;59(Suppl. 1) [abstract 564-P, 2009:A157]. [48] Hussey EK, Clark RV, Amin DM, Kipnes MS, O‘ConnorSemmes RL, O‘Driscoll EC, et al. Single-dose pharmacokinetics and pharmacodynamics of sergliflozin etabonate, a novel inhibitor of glucose reabsorption, in healthy volunteers and patients with type 2 diabetes mellitus. J Clin Pharmacol 2010;6:623–35. [49] Obermeier M, Yao M, Khanna A, Koplowitz B, Zhu M, Li W, et al. In vitro characterization and pharmacokinetics of dapagliflozin (BMS-512148), a potent sodium-glucose cotransporter type II inhibitor, in animals and humans. Drug Metab Dispos 2010;3:405–14. [50] Bellamine A, Smarsh M, Onorato J, Dapagliflozin. (BMS512148) is a competitive, selective and reversible inhibitor of human SGLT2. In: Presented at: biomedical transporters conference; 2009. [51] Han S, Hagan DL, Taylor JR, Xin L, Meng W, Biller SA, et al. Dapagliflozin, a selective SGLT2 inhibitor, improves glucose homeostasis in normal and diabetic rats. Diabetes 2008;6:1723–9. [52] Komoroski B, Vachharajani N, Feng Y, Li L, Kornhauser D, Pfister M. Dapagliflozin, a novel, selective SGLT2 inhibitor,
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
[53]
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
improved glycemic control over 2 weeks in patients with type 2 diabetes mellitus. Clin Pharmacol Ther 2009;5:513– 9. List JF, Woo V, Morales E, Tang W, Fiedorek FT. Sodiumglucose cotransport inhibition with dapagliflozin in type 2 diabetes. Diabetes Care 2009;4:650–7. Sjo¨stro¨m D, Sugg J, Ptaszynska A, Parikh S. Presentation at the European Society of Cardiology; 2014, Available at: http://spo.escardio.org/eslides/ view.aspx?eevtid=54&fp=3972 [accessed January 10]. Janssen Research and Development, LLC. Canagliflozin slides presented at the Food and Drug Administration (FDA) Endocrinologic and Metabolic Drugs Advisory Committee (EMDAC) Meeting, January 10, 2013. Available at: http://www.fda.gov/AdvisoryCommittees/ CommitteesMeetingMaterials/Drugs/ EndocrinologicandMetabolicDrugsAdvisoryCommittee/ ucm336233.htm [accessed 11.01.14]. Ferrannini E, Solini A. SGLT2 inhibition in diabetes mellitus: rationale and clinical prospects. Nat Rev Endocrinol 2012;8:495–502. Thomson SC, Vallon V, Blantz RC. Kidney function in early diabetes: the tubular hypothesis of glomerular filtration. Am J Physiol Renal Physiol 2004;1:F8–15. Liu JJ, Lee T, DeFronzo RA. Why Do SGLT2 inhibitors inhibit only 30–50% of renal glucose reabsorption in humans? Diabetes 2012;9:2199–204. Komoroski B, Vachharajani N, Boulton D, Kornhauser D, Geraldes M, Li L, et al. Dapagliflozin, a novel SGLT2 inhibitor, induces dose-dependent glucosuria in healthy subjects. Clin Pharmacol Ther 2009;5:520–6. FDA approves dapagliflozin. Available at: http:// www.fda.gov/NewsEvents/Newsroom/ PressAnnouncements/ucm380829.htm [accessed 10.01.14]. European Medicines Agency authorizing the use of dapagliflozin in November 2012. Available at: http:// www.ema.europa.eu/ema/index.jsp?curl=pages/ medicines/human/medicines/002322/ human_med_001546.jsp&mid=WC0b01ac058001d124 [accessed 10.01.13]. Kaku K, Inoue S, Matsuoka O, Kiyosue A, Azuma H, Hayashi N, et al. Efficacy and safety of dapagliflozin as a monotherapy for type 2 diabetes mellitus in Japanese patients with inadequate glycaemic control: a phase II multicentre, randomized, double-blind, placebocontrolled trial. Diabetes Obes Metab 2013;5:432–40. Ferrannini E, Ramos SJ, Salsali A, Tang W, List JF. Dapagliflozin monotherapy in type 2 diabetic patients with inadequate glycemic control by diet and exercise: a randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care 2010;10:2217–24. Ji L, Ma J, Li H, Mansfield TA, T‘joen CL, Iqbal N, et al. Dapagliflozin as monotherapy in drug-naive asian patients with type 2 diabetes mellitus: a randomized, blinded, prospective phase III study. Clin Ther 2013;36(1):84–100. Henry RR, Murray AV, Marmolejo MH, Hennicken D, Ptaszynska A, List JF. Dapagliflozin, metformin XR, or both: initial pharmacotherapy for type 2 diabetes, a randomised controlled trial. Int J Clin Pract 2012;5:446–56. Bailey CJ, Gross JL, Pieters A, Bastien A, List JF. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with metformin: a randomised, double-blind, placebo-controlled trial. Lancet 2010;9733:2223–33. Bolinder J, Ljunggren O, Kullberg J, Johansson L, Wilding J, Langkilde AM, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
[77]
[78]
[79] [80] [81]
[82]
[83]
23
distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J Clin Endocrinol Metab 2012;3:1020–31. Nauck MA, Del Prato S, Meier JJ, Duran-Garcia 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;9: 2015–22. Strojek K, Yoon KH, Hruba V, Elze M, Langkilde AM, Parikh S. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with glimepiride: a randomized, 24-week, double-blind, placebo-controlled trial. Diabetes Obes. Metab 2011;10:928–38. Rosenstock J, Vico M, Wei L, Salsali A, List JF. Effects of dapagliflozin, an SGLT2 inhibitor, on HbA(1c), body weight, and hypoglycemia risk in patients with type 2 diabetes inadequately controlled on pioglitazone monotherapy. Diabetes Care 2012;7:1473–8. Jabbour SA, Hardy E, Sugg J, Parikh S, for the Study 10 Group. Dapagliflozin Is effective as add-on therapy to sitagliptin with or without metformin: a 24-week, multicenter, randomized, double-blind, placebocontrolled studys. Diabetes Care 2013. DC_130467. Wilding JP, Norwood P, T‘joen C, Bastien A, List JF, Fiedorek FT. A study of dapagliflozin in patients with type 2 diabetes receiving high doses of insulin plus insulin sensitizers: applicability of a novel insulin-independent treatment. Diabetes Care 2009;9:1656–62. Wilding JP, Woo V, Soler NG, Pahor A, Sugg J, Rohwedder K, et al. Long-term efficacy of dapagliflozin in patients with type 2 diabetes mellitus receiving high doses of insulin: a randomized trial. Ann Intern Med 2012;6:405–15. Wilding JP, Woo V, Rohwedder K, Sugg J, Parikh S, for the Dapagliflozin 006 Study Group. Dapagliflozin in patients with type 2 diabetes receiving high doses of insulin: efficacy and safety over 2 years. Diabetes Obes Metab 2013;16(2):124–36. Ptaszynska A, Hardy E, Johnsson E, Parikh S, List J. Effects of dapagliflozin on cardiovascular risk factors. Postgrad Med 2013;3:181–9. Johnsson KM, Ptaszynska A, Schmitz B, Sugg J, Parikh SJ, List JF. Vulvovaginitis and balanitis in patients with diabetes treated with dapagliflozin. J Diabetes Complications 2013;5:479–84. Johnsson KM, Ptaszynska A, Schmitz B, Sugg J, Parikh SJ, List JF. Urinary tract infections in patients with diabetes treated with dapagliflozin. J Diabetes Complications 2013;5:473–8. Leiter L, Cefalu W, de Bruin T, Gause-Nilsson I, Sugg J, Parikh S. Efficacy and safety of dapagliflozin for type 2 diabetes mellitus patients with a history of cardiovascular disease. In: 72nd American Diabetes Association Scientific Sessions; 2012. FORXIGA1. Summary of product characteristics. BristolMyers Squibb/AstraZeneca EEIG; 2013. Burki TK. FDA rejects novel diabetes drug over safety fears. Lancet 2012;9815:507. Ptaszynska A, Johnsson K, Apanovitch AM, Sugg J, Parikh S, List J. Safety of dapagliflozin in clinical trials for type 2 diabetes mellitus. Int Med J 2013;43:p50. Jones D. Diabetes field cautiously upbeat despite possible setback for leading SGLT2 inhibitor. Nat Rev Drug Discov 2011;9:645–6. Kohan D, Fioretto P, List J. Efficacy and safety of dapagliflozin in patients with type 2 diabetes and moderate renal impairment [abstract no. TH-PO524]. J Am Soc Nephrol 2011;232A.
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
24
diabetes research and clinical practice xxx (2014) xxx–xxx
[84] FDA news release approving Invokana in March 29, 2013. Available at: http://www.fda.gov/NewsEvents/Newsroom/ PressAnnouncements/ucm345848.htm [accessed 11.01.14]. [85] European Commission approving Invokana on November 22, 2013. Available at: http://ec.europa.eu/health/ documents/community-register/html/newproc.htm#h [accessed 11.01.14]. [86] Stenlof K, Cefalu WT, Kim KA, Alba M, Usiskin K, Tong C, et al. Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise. Diabetes Obes Metab 2013;4:372–82. [87] Inagaki N, Kondo K, Yoshinari T, Maruyama N, Susuta Y, Kuki H. Efficacy and safety of canagliflozin in Japanese patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, 12-week study. Diabetes Obes Metab 2013;12:1136–45. [88] Rosenstock J, Aggarwal N, Polidori D, Zhao Y, Arbit D, Usiskin K, et al. Dose-ranging effects of canagliflozin, a sodium-glucose cotransporter 2 inhibitor, as add-on to metformin in subjects with type 2 diabetes. Diabetes Care 2012;6:1232–8. [89] Lavalle-Gonzalez FJ, Januszewicz A, Davidson J, Tong C, Qiu R, Canovatchel W, et al. Efficacy and safety of canagliflozin compared with placebo and sitagliptin in patients with type 2 diabetes on background metformin monotherapy: a randomised trial. Diabetologia 2013;12:2582–92. [90] 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 noninferiority trial. Lancet 2013;9896:941–50. [91] Wilding JP, Charpentier G, Hollander P, Gonzalez-Galvez G, Mathieu C, Vercruysse F, et al. Efficacy and safety of canagliflozin in patients with type 2 diabetes mellitus inadequately controlled with metformin and sulphonylurea: a randomised trial. Int J Clin Pract 2013;12:1267–82. [92] Schernthaner G, Gross JL, Rosenstock J, Guarisco M, Fu M, Yee J, et al. Canagliflozin compared with sitagliptin for patients with type 2 diabetes who do not have adequate glycemic control with metformin plus sulfonylurea: a 52week randomized trial. Diabetes Care 2013;9:2508–15. [93] Forst T, Guthrie R, Goldenberg R, et al. Efficacy and safety of canagliflozin in subjects with type 2 diabetes on metformin and pioglitazone [abstract]. In: 4th world congress on controversies to consensus in diabetes, obesity, and hypertension; 2012. [94] Matthews D, Fulcher G, Perkovic V, de Zeeuw D, Mahaffey K, Rosenstock J, et al. Efficacy and safety of Canagliflozin (CANA), an inhibitor of Sodium Glucose Co-Transporter 2 (SGLT2), added on to insulin therapy with or without oral agents in type 2 diabetes (T2D). Diabetologie und Stoffwechsel S01:P251, 2012. [95] Yale JF, Bakris G, Cariou B, Yue D, David-Neto E, Xi L, et al. Efficacy and safety of canagliflozin in subjects with type 2 diabetes and chronic kidney disease. Diabetes Obes Metab 2013;5:463–73. [96] Bode B, Stenlof K, Sullivan D, Fung A, Usiskin K. Efficacy and safety of canagliflozin treatment in older subjects with type 2 diabetes mellitus: a randomized trial. Hosp Pract (1995) 2013;2:72–84. [97] Polidori D, Sha S, Mudaliar S, Ciaraldi TP, Ghosh A, Vaccaro N, et al. Canagliflozin lowers postprandial glucose and insulin by delaying intestinal glucose absorption in addition to increasing urinary glucose excretion results of
[98]
[99]
[100]
[101]
[102]
[103]
[104]
[105]
[106]
[107]
[108]
[109]
[110]
[111]
a randomized, placebo-controlled study. Diabetes Care 2013;36(8):2154–61. Baigent C, Keech A, Kearney PM, et al. Cholesterol Treatment Trialists’ (CTT) Collaborators. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet 2005;366: 1267–78. INVOKANA1 [package insert]. Titusville, NJ: Janssen Pharmaceuticals Inc; 2013. Available at: http:// www.invokanahcp.com/prescribing-information.pdf [accessed 11.01.14]. Nyirjesy P, Zhao Y, Ways K, Usiskin K. Evaluation of vulvovaginal symptoms and Candida colonization in women with type 2 diabetes mellitus treated with canagliflozin, a sodium glucose co-transporter 2 inhibitor. Curr Med Res Opin 2012;7:1173–8. Nicolle L, Capuano G, Ways K, Usiskin K. Effect of canagliflozin, a sodium glucose co-transporter 2 (SGLT2) inhibitor, on bacteriuria and urinary tract infection in subjects with type 2 diabetes enrolled in a 12-week, phase 2 study. Curr Med Res Opin 2012;7:1167–71. Kawalec P, Mikrut A, ˇˇLopuch S. The safety of dipeptidyl peptidase-4 (DPP-4) inhibitors or sodium-glucose cotransporter 2 (SGLT-2) inhibitors added to metformin background therapy in patients with type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Metab Res 2013. Janssen Research & Development, LLC. CANVAS – CANagliflozin cardioVascular Assessment Study. NLM identifier: NCT01032629. Available at: http:// www.clinicaltrials.gov/ct2/show/NCT01032629 [accessed 12.01.14]. Tucker ME. Experts welcome canagliflozin approval, but questions remain. Heart Org 2014. Available at: http:// www.theheart.org/article/1530799.do [accessed January 11]. Matthews DR, Fulcher G, Perkovic V, et al. Efficacy and safety of canagliflozin (CANA), an inhibitor of sodium glucose co-transporter 2 (SGLT2), added-on to insulin therapy +/ oral agents in type 2 diabetes [abstract]. In: 48th European Association for the Study of Diabetes. Annual meeting; 2012. Lamos EM, Younk LM, Davis SN. Canagliflozin, an inhibitor of sodium-glucose cotransporter 2, for the treatment of type 2 diabetes mellitus. Expert Opin Drug Metab Toxicol 2013;6:763–75. Daousi C, Casson IF, Gill GV, MacFarlane IA, Wilding JP, Pinkney JH. Prevalence of obesity in type 2 diabetes in secondary care: association with cardiovascular risk factors. Postgrad Med J 2006;966:280–4. Nair S, Joseph F, Ewins D, Wilding J, Goenka N. From history to reality: sodium glucose co-transporter 2 inhibitors–a novel therapy for type 2 diabetes mellitus. Pract Diabetes Int 2010;7:311–6. Clinical development pipline on Lilly’s website. Available at: http://www.lilly.com/SiteCollectionDocuments/ Pipeline/Clinical Development Pipeline/13.html [accessed 14.01.14]. Grempler R, Thomas L, Eckhardt M, Himmelsbach F, Sauer A, Sharp D, et al. Empagliflozin, a novel selective sodium glucose cotransporter-2 (SGLT-2) inhibitor: characterisation and comparison with other SGLT-2 inhibitors. Diabetes Obes Metab 2012;1:83–90. Seman L, Macha S, Nehmiz G, Simons G, Ren B, Pinnetti S, et al. Empagliflozin (BI 10773), a potent and selective SGLT2 inhibitor, induces dose-dependent glucosuria in healthy subjects. Clin Pharmacol Drug Dev 2013;2(2): 152–61.
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26 diabetes research and clinical practice xxx (2014) xxx–xxx
[112] Heise T, Seewaldt-Becker E, Macha S, Hantel S, Pinnetti S, Seman L, et al. Safety, tolerability, pharmacokinetics and pharmacodynamics following 4 weeks’ treatment with empagliflozin once daily in patients with type 2 diabetes. Diabetes Obes Metab 2013;7:613–21. [113] Boehringer Ingelheim and Eli Lilly and Company submit new drug application to FDA for empagliflozin. Available at: https://investor.lilly.com/ releasedetail.cfm?ReleaseID=750602 [accessed 14.01.14]. [114] Ferrannini E, Seman L, Seewaldt-Becker E, Hantel S, Pinnetti S, Woerle HJ. A Phase IIb, randomized, placebocontrolled study of the SGLT2 inhibitor empagliflozin in patients with type 2 diabetes. Diabetes Obes Metab 2013;8:721–8. [115] Roden M, Weng J, Eilbracht J, Delafont B, Kim G, Woerle HJ, et al. Empagliflozin monotherapy with sitagliptin as an active comparator in patients with type 2 diabetes: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol 2013;3:208–19. [116] Haring HU, Merker L, Seewaldt-Becker E. Empagliflozin as add-on to metformin for 24 weeks improves glycemic control in patients with type 2 diabetes(T2DM). Diabetes 2013;62(suppl 1):A282. [117] Rosenstock J, Seman LJ, Jelaska A, Hantel S, Pinnetti S, Hach T, et al. Efficacy and safety of empagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, as addon to metformin in type 2 diabetes with mild hyperglycaemia. Diabetes Obes Metab 2013;12:1154–60. [118] Haring HU, Merker L, Seewaldt-Becker E, Weimer M, Meinicke T, Woerle HJ, et al. Empagliflozin as add-on to metformin plus sulfonylurea in patients with type 2 diabetes: a 24-week, randomized, double-blind, placebocontrolled trial. Diabetes Care 2013;11:3396–404. [119] Kovacs CS, Seshiah V, Swallow R, Jones R, Rattunde H, Woerle HJ, et al. 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):147–58. [120] Rosenstock J, Jelaska A, Kim G, Broedl U, Woerle H. Empagliflozin as add-on to basal insulin for 78 weeks improves glycemic control with weight loss in insulintreated type 2 diabetes (T2DM). Diabetes 2013;62(Suppl. 1):A285 [Asbtract 1102-P]. [121] Woerle H, Ferrannini E, Berk A, Manun’Ebo M, Pinnetti S, Broedl U. Safety and efficacy of empagliflozin as monotherapy or add-on to metformin in a 78-week openlabel extension study in patients with type 2 diabetes. In: 72nd scientific sessions of the American Diabetes Association; 2012. [122] Hach T, Heerspink HJL, Pfarr E, Lund S, Ley L, Broedl UC, et al. The sodium glucose cotransporter-2 (SGLT-2) inhibitor empagliflozin lowers blood pressure independent of weight or HbA1c change. In: Presented at the 48th European Association for the Study of Diabetes (EASD) annual meeting. 2012. p. 1–5. [123] Ferrannini E, Berk A, Hantel S, Pinnetti S, Hach T, Woerle HJ, et al. Long-term safety and efficacy of empagliflozin, sitagliptin, and metformin: an active-controlled, parallelgroup, randomized, 78-week open-label extension study in patients with type 2 diabetes. Diabetes Care 2013;12:4015–21. [124] Macha S, Mattheus M, Halabi A, Pinnetti S, Woerle HJ, Broedl UC. Pharmacokinetics, pharmacodynamics and safety of empagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, in subjects with renal impairment. Diabetes Obes Metab 2014;16:215–22. [125] Macha S, Rose P, Mattheus M, Cinca R, Pinnetti S, Broedl UC, et al. Pharmacokinetics, safety and tolerability of
[126]
[127]
[128]
[129]
[130]
[131]
[132]
[133]
[134]
[135]
[136]
[137]
[138]
[139]
25
empagliflozin, a sodium glucose cotransporter 2 inhibitor, in patients with hepatic impairment. Diabetes Obes Metab 2014;16:118–23. Ridderstrale M, Svaerd R, Zeller C, Kim G, Woerle HJ, Broedl UC, et al. Rationale, design and baseline characteristics of a 4-year (208-week) phase III trial of empagliflozin, an SGLT2 inhibitor, versus glimepiride as add-on to metformin in patients with type 2 diabetes mellitus with insufficient glycemic control. Cardiovasc Diabetol 2013. 129-2840-12-129. Astellas submits application for approval of ipragliflozin in Japan [March, 2013]. Available at: http:// www.astellas.com/en/corporate/news/pdf/ 130313_1_Eg.pdf [accessed 16.01.14]. Ipragliflozin approved in Japan in January 2014. Available at: http://www.astellas.com/en/corporate/news/detail/ approval-of-suglat-tablets-a-s.html [accessed 25.01.14]. Veltkamp SA, Kadokura T, Krauwinkel WJ, Smulders RA. Effect of Ipragliflozin (ASP1941), a novel selective sodiumdependent glucose co-transporter 2 inhibitor, on urinary glucose excretion in healthy subjects. Clin Drug Investig 2011;12:839–51. Schwartz SL, Akinlade B, Klasen S, Kowalski D, Zhang W, Wilpshaar W. Safety, pharmacokinetic, and pharmacodynamic profiles of ipragliflozin (ASP1941), a novel and selective inhibitor of sodium-dependent glucose co-transporter 2, in patients with type 2 diabetes mellitus. Diabetes Technol Ther 2011;12:1219–27. Smulders RA, Zhang W, Veltkamp SA, van Dijk J, Krauwinkel WJ, Keirns J, et al. No pharmacokinetic interaction between ipragliflozin and sitagliptin, pioglitazone, or glimepiride in healthy subjects. Diabetes Obes Metab 2012;10:937–43. Kashiwagi A, Takinami Y, Kazuta K, Yoshida S, Utsuno A, Nagase I. Ipragliflozin improved glycaemic control with additional benefits of reductions of body weight and blood pressure in Japanese patients with type 2 diabetes mellitus: BRIGHTEN Study; 2011;S68–9. Fonseca VA, Ferrannini E, Wilding JP, Wilpshaar W, Dhanjal P, Ball G, et al. Active- and placebo-controlled dose-finding study to assess the efficacy, safety, and tolerability of multiple doses of ipragliflozin in patients with type 2 diabetes mellitus. J Diabetes Complications 2013;3:268–73. Goto K, Kashiwagi A, Kazuta K. Ipragliflozin reduces A1C and body weight in type 2 diabetes patients who have inadequate glycemic control on metformin alone: ILLUMINATE study. Diabetes 2012;Suppl. 1:A269. Wilding JP, Ferrannini E, Fonseca VA, Wilpshaar W, Dhanjal P, Houzer A. Efficacy and safety of ipragliflozin in patients with type 2 diabetes inadequately controlled on metformin: a dose-finding study. Diabetes Obes Metab 2013;5:403–9. Kashiwagi A, Shiga T, Akiyama N. Ipragliflozin reduced HbA1c and body weight in Japanese type 2 diabetes patients who have inadequate glycaemic control on sulfonylurea or pioglitazone alone. In: Presented at the 48th EASD annual meeting; 2012 [Poster 739]. LX4211 in the pipeline by Lexicon Pharmaceuticals. Available at: http://www.lexgen.com/pipeline/lx4211.html [accessed 16.01.14]. Zambrowicz B, Freiman J, Brown P, Frazier K, Turnage A, Bronner J, et al. LX4211, a dual SGLT1/SGLT2 inhibitor, improved glycemic control in patients with type 2 diabetes in a randomized, placebo-controlled trial. Clin Pharmacol Ther 2012;2:158–69. Zambrowicz B, Ding ZM, Ogbaa I, Frazier K, Banks P, Turnage A, et al. Effects of LX4211, a dual SGLT1/SGLT2 inhibitor, plus sitagliptin on postprandial active GLP-1 and
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014
DIAB-6018; No. of Pages 26
26
diabetes research and clinical practice xxx (2014) xxx–xxx
glycemic control in type 2 diabetes. Clin Ther 2013;3. 273– 285e7. [140] Rosenstock J, Zambrowicz B, Lapuerta P, Ogbaa I, Banks P, Freiman J, et al. Impact of LX4211, a dual inhibitor of sodium glucose transporter 1 and 2 on cardiovascular risk factors in type 2 diabetes. Abstract presented at: 2012 American Heart Association annual meeting.
[141] LX4211 in subjects with T2DM and renal impairment. Available at: http://www.clinicaltrials.gov/ct2/show/ NCT01555008 [accessed 17.01.14]. [142] Efficacy and safety of LX4211 in patients with inadequately controlled type 1 diabetes mellitus. Available at: http://www.clinicaltrials.gov/ct2/show/ NCT01742208 [accessed 17.01.14].
Please cite this article in press as: Hasan FM, et al. SGLT2 inhibitors in the treatment of type 2 diabetes. Diabetes Res Clin Pract (2014), http:// dx.doi.org/10.1016/j.diabres.2014.02.014