You are on page 1of 9

Review

Renal glucose reabsorption inhibitors


to treat diabetes
Clifford J. Bailey
Life and Health Sciences, Aston University, Birmingham B4 7ET, UK

Current therapies to reduce hyperglycaemia in type 2 would be a valuable addition to the treatment choices for
diabetes mellitus (T2DM) mostly involve insulin-depen- hyperglycaemia in T2DM. Renewed interest in the role of
dent mechanisms and lose their effectiveness as pancre- the kidney in glucose homeostasis has prompted the de-
atic b-cell function declines. In the kidney, filtered velopment of sodium glucose cotransporter-2 (SGLT2)
glucose is reabsorbed mainly via the high-capacity, inhibitors that fulfil these criteria by reducing renal glu-
low-affinity sodium glucose cotransporter-2 (SGLT2) at cose reabsorption. This review examines the insulin-inde-
the luminal surface of cells lining the first segment of the pendent mechanism of action of these agents and recent
proximal tubules. Selective inhibitors of SGLT2 reduce clinical studies that indicate their potential as a novel
glucose reabsorption, causing excess glucose to be elim- approach in the treatment of T2DM.
inated in the urine; this decreases plasma glucose. In
T2DM, the glucosuria produced by SGLT2 inhibitors is Role of the kidney in glucose balance
associated with weight loss, and mild osmotic diuresis The renal cortex produces glucose by gluconeogenesis, main-
might assist a reduction in blood pressure. The mecha- ly for utilization in the renal medulla [9]. In T2DM, both the
nism is independent of insulin and carries a low risk of liver and kidney contribute to excess glucose production,
hypoglycaemia. This review examines the potential of and renal glucose production can contribute up to 20% of
SGLT2 inhibitors as a novel approach to the treatment of the total glucose released into the circulation in the post-
hyperglycaemia in T2DM. absorptive state [9,10]. However, the kidneys substantially
affect the circulating glucose pool through reabsorption of
Treating hyperglycaemia glucose filtered by the glomeruli. Renal glucose reabsorption
Treatment of hyperglycaemia in type 2 diabetes mellitus is a constitutive process that depends on the concentration of
(T2DM) is necessary to relieve acute symptoms and to glucose; it does not seem to be regulated by insulin and
reduce the risk of chronic vascular complications. Lifestyle might actually be increased in T2DM.
interventions, notably diet and exercise, are important but Almost all of the glucose entering the glomeruli in the
are generally insufficient to achieve or maintain glycaemic afferent glomerular arterioles is filtered into the nephron
control. Most current glucose-lowering therapies act to fluid and enters the proximal convoluted tubule. Normal
address the underlying endocrine pathogenesis of insulin kidneys (with a glomerular filtration rate of 125 mL/min)
resistance and b-cell dysfunction. However, these thera- filter approximately 180 L of plasma each day [11]. Thus, a
pies usually lose their effectiveness over time as b-cell healthy individual with an average plasma glucose con-
failure supervenes. centration of 5–5.5 mmol/L (90–100 mg/dL) will filter ap-
As a consequence, many patients receive multiple glu- proximately 160–180 g of glucose daily, all of which will be
cose-lowering therapies and eventually require exogenous reabsorbed [12]. In theory, the amount of filtered glucose
insulin. Indeed, half or more of patients with T2DM do not that is reabsorbed will increase in proportion to the plasma
achieve guideline targets for glycaemic control [glycated glucose concentration until the maximal reabsorptive
haemoglobin (HbA1c) of 6.5–7.0% (48–53 mmol/mol)] [1–4]. transport capacity of the tubules (Tm) is reached. Tm
A majority of patients with T2DM are overweight or usually occurs at an average filtered glucose load of ap-
obese [5], which increases insulin resistance and compli- proximately 375 mg/min, and all excess filtered glucose is
cates treatment [6]. However, several blood glucose-lower- excreted in urine. In practice, however, some glucose usu-
ing therapies (including insulin) are associated with ally escapes in the urine at plasma glucose concentrations
weight gain (Table 1), often of 2–5 kg [7]. This is predomi- above 11 mmol/L (200 mg/dL), the renal threshold. There
nantly due to reduced urinary glucose excretion, and ini- is a gradual increase in glucosuria above the renal thresh-
tially approximates to the correction of glycaemia [8]. old up to the Tm value for glucose, which is called the splay
Except for a-glucosidase inhibitors, which slow the rate effect. These features of renal glucose handling largely
of intestinal carbohydrate digestion, key antidiabetic reflect the properties of the sodium glucose cotransporters
agents act, at least in part, through insulin-dependent (SGLTs) in the proximal tubules.
mechanisms (Table 1 and Figure 1). Thus, a new thera-
peutic approach without reliance on insulin or weight gain,
Sodium glucose co-transporters
and suitable for use in combination with existing agents,
SGLT proteins are encoded by the solute carrier 5 (SLC5)
Corresponding author: Bailey, C.J. (c.j.bailey@aston.ac.uk). subfamily of sodium/substrate symporter genes [12]. The
0165-6147/$ – see front matter ß 2010 Elsevier Ltd. All rights reserved. doi:10.1016/j.tips.2010.11.011 Trends in Pharmacological Sciences, February 2011, Vol. 32, No. 2 63
Review Trends in Pharmacological Sciences February 2011, Vol. 32, No. 2

Table 1. Current antidiabetic agentsa [4,54–61]


Agent Insulin-dependent mechanisms Comments
Insulin Insulin Body weight
action release change
a-Glucosidase inhibitors – – Neutral  Reduce rate of carbohydrate digestion in small
intestine, lowering postprandial glucose levels
 Only oral antidiabetic class currently available
that does not have an insulin-dependent
mechanism of action
Amylin agonistsb (pramlinitide) H – Reduce  Injected subcutaneously before each meal in
insulin-treated patients
 Slow gastric emptying, inhibit postprandial
glucagon production in a glucose-dependent
manner, thereby reducing the increase in
postprandial glucose levels
 Although not directly dependent on insulin,
suppression of glucagon by amylin agonists
might indirectly enhance insulin action
Biguanides (metformin) H – Neutral  Decrease hepatic glucose output and improve
peripheral glucose disposal
 Metformin is the only biguanide available in most
of the world
 Efficacy requires the presence of insulin
DPP-4 inhibitors – H Neutral  Prevent degradation of endogenous incretins,
c
especially GLP-1, thereby raising incretin levels
 Increased GLP-1 levels potentiate glucose-dependent
insulin secretion and suppress glucagon release
Glinides – H Gain  Stimulate insulin secretion by binding to the
benzamido site of the sulfonylurea receptor SUR1
on pancreatic b-cells
GLP-1 receptor agonists – H Reduce  Bind to GLP-1 receptors on pancreatic b-cells and
potentiate glucose-dependent insulin secretion
and suppress glucagon release, resulting in
reduced postprandial hepatic glucose production
and enhanced peripheral glucose uptake
Sulfonylureas – H Gain  Stimulate insulin secretion by binding to the
sulfonylurea receptor SUR1 on pancreatic b-cells
TZDs H – Gain  Modulate PPAR-g activity to increase sensitivity
of muscle, fat and liver to insulin; reduce hepatic
glucose production
Bromocriptine d H – Neutral  Dopamine D2 receptor agonist
Colesevelam d – H? Neutral  Bile acid sequestrant
 Mechanism uncertain, induces GLP-1 secretion in
rat models
a
Abbreviations: DPP-4, dipeptidyl peptidase-4; GLP-1, glucagon-like peptide-1; PPAR-g, peroxisome proliferator-activated receptor gamma; SUR1, sulfonylurea receptor;
TZDs, thiazolidinediones; –, no effect; H, increase or enhancement;?, not established.
b
Amylin is a neurohormone that is co-secreted with insulin in response to meals.
c
Incretins are hormones released from the gastrointestinal tract during meal digestion which enhance nutrient-induced insulin secretion.
d
The dopamine D2 receptor agonist bromocriptine and the bile sequestrant colesevelam have recently been licensed for treatment of T2DM in some territories.

SCL5 genes code for several proteins (75 kDa) with >59% SGLT2 and SGLT1 exhibit different transport charac-
amino acid identity, among which SGLT1 and SGLT2 are teristics [12]. SGLT2 transports glucose and sodium with a
the most well characterized [12]. SGLT1 is predominantly 1:1 stoichiometry, whereas SGLT1 transports one mole-
expressed in enterocytes, where it mediates glucose and cule of glucose with two sodium ions. SGLT2 is a low-
galactose uptake from the gut lumen [12,13]. SGLT1 is also affinity, high-capacity glucose transporter with K0.5 values
expressed in the more distal segments (S2–3) of the proxi- of 2 mM for glucose and 100 mM for sodium. By contrast,
mal convoluted tubule, where it mediates the reabsorption SGLT1 is a high-affinity, low-capacity glucose transporter
of glucose that has not been reabsorbed earlier in the with K0.5 values of 0.4 mM for glucose and 3 mM for sodium
tubule by SGLT2. SGLT2 is almost entirely confined to [14]. Thus, SGLT2 in the S1 segment is suited to reabsorp-
the first segment (S1) of the proximal tubules in the kidney tion of a high glucose concentration entering the proximal
cortex [13,14], where it mediates reabsorption of most of tubule [16], whereas SGLT1 is suited to reabsorption of the
the filtered glucose [15]. remaining lower glucose concentration in subsequent seg-
SGLT1 and SGLT2 transport glucose across the luminal ments (Figure 3). After reabsorption from the lumen,
membrane of epithelial cells lining the proximal tubules glucose within the proximal tubular cells passes into the
against a concentration gradient by coupling with the interstitium (and then to the plasma) by facilitative glu-
inward diffusion of sodium ions (Figure 2) [12]. This is a cose transporters in the basolateral membranes (GLUT2 in
secondary active process and is maintained by extrusion of S1 and GLUT1 in subsequent segments).
sodium across the basolateral membrane via the sodium- Although glucosuria is a feature of poorly controlled
potassium ATPase pump. diabetes, there is evidence that the renal threshold for

64
()TD$FIG][ Review Trends in Pharmacological Sciences February 2011, Vol. 32, No. 2

GLP-1 receptor agonist


SU/glinide
DPP-4i
Pancreas

Insulin

tin
Metformin TZD

Incre
Liver
Adipose
Muscle
tissue

Blood
glucose
Gut
Kidney

AGI
SGLT2i
TRENDS in Pharmacological Sciences

Figure 1. Sites of action of antidiabetic agents, including SGLT2 inhibitors. Metformin targets both the liver and skeletal muscle [62]. The thiazolidinediones (TZDs) improve
insulin sensitivity, reduce hepatic glucose production, and improve glucose disposal indirectly by altering lipid metabolism in adipose tissue [57,58]. Sulfonylureas,
meglitinides (glinides) and GLP-1 receptor agonists target pancreatic b-cells and increase insulin secretion. GLP-1 agonists also reduce excess glucagon secretion by
pancreatic a-cells and slow gastric emptying. DPP-4 inhibitors reduce the breakdown and increase the levels of endogenous incretin hormones GLP-1 and GIP [59]. Amylin
agonists (not illustrated) slow gastric emptying and reduce postprandial glucagon secretion [61]. a-Glucosidase inhibitors slow the rate of carbohydrate digestion by the
small intestine [4]. The dopamine receptor agonist bromocriptine, which improves diurnal hypothalamic control of glucose homeostasis, and the bile sequestrant
colesevelam, which might affect the secretion of incretin hormones (not illustrated), have recently been approved for use in the treatment of hyperglycaemia in the USA
[54–56]. Selective SGLT2 inhibitors reduce glucose reabsorption by the kidney. Abbreviations: AGI, a-glucosidase inhibitor; DPP-4i, dipeptidyl peptidase-4 inhibitor; GIP,
glucose-dependent insulinotropic polypeptide; GLP-1, glucagon-like peptide-1; SGLT2i, sodium glucose cotransporter-2 inhibitor; SU, sulfonylurea.
" Stimulatory or positive effect
¢ Inhibitory effect.

[()TD$FIG]

Glucose

SGLT2

S1 segment of
proximal tubule
SGLT1
Up to 90%
Collecting duct
Glucose reabsorption

≥10%

Distal S2/3
segment of
proximal tubule

Negligible glucose
in urine
TRENDS in Pharmacological Sciences

Figure 2. Glucose reabsorption from the glomerular filtrate in the renal proximal tubule. Glucose is reabsorbed from the glomerular filtrate and into the blood through
proximal tubule epithelial cells. In segment S1 of the proximal tubules, SGLT2 transports glucose into cells against a concentration gradient by coupling glucose with the
inward diffusion of sodium ions down their concentration gradient. The inward passage of sodium ions is maintained by an Na+/K+ ATPase pump, which extrudes Na+ ions
across the basolateral membrane of the cell in exchange for K+ ions in 3:2 stoichiometry. In segment S1, glucose is transported from the cells into the interstitial
compartment by the facilitative glucose transporter GLUT2. A similar mechanism operates with SGLT1 and GLUT1 in segment S3. Based on Bakris et al. [63].

65
()TD$FIG][ Review Trends in Pharmacological Sciences February 2011, Vol. 32, No. 2

Segment S1
Basolateral membrane

SGLT2 GLUT2

Glucose
Glucose

Na+

Glucose

Na+

Na+
K+
Tight K+
junction Na+/K+ATPase
pump

Lateral intercellular space

Segment S3
Basolateral membrane

SGLT1 GLUT1

Glucose Glucose

Na+

Glucose

Na+

Na+
K+
K+
Na+/K+ATPase
pump

Lateral intercellular space


TRENDS in Pharmacological Sciences

Figure 3. Reabsorption of glucose from the renal proximal tubules by the sodium glucose cotransporters SGLT2 and SGLT1. Almost all of the glucose entering glomeruli in the
afferent glomerular arterioles is filtered into the nephron fluid of the proximal renal tubules. Most (up to 90%) of this filtered glucose is reabsorbed in the initial proximal
convoluted segment (S1) by SGLT2 located at the luminal surface of proximal tubular cells. Remaining glucose is reabsorbed from the filtrate in the more distal convoluted and
straight segments by SGLT1. Glucose within the proximal tubular cells is then transported back to the interstitial compartment and thence to the plasma by the facilitative
glucose transporters GLUT2 and GLUT1 in the S1 and S3 segments, respectively. In normal individuals with an average plasma glucose concentration of 5–5.5 mmol/L (90–
100 mg/dL), approximately 160–180 g of glucose is filtered daily, with less than 0.5 g/day of glucose appearing in the urine. Based on Bakris et al. [63] and Bays [64].

glucose might be higher in diabetic states, thereby reduc- glomerular filtration, approximately 324 g of glucose would
ing urinary glucose excretion [17–19]. Animal studies sug- be filtered daily. If there were 20–25% inhibition of renal
gest that SGLT2 mRNA expression is increased by chronic glucose reabsorption, the body would lose 65–80 g of glucose
hyperglycaemia, and this effect is reversed during insulin per day in urine. This equates to approximately 260–320
treatment, which is consistent with improved glycaemic kcal of energy per day. If diet and exercise were maintained
control [19]. There is also preliminary evidence that and metabolic efficiency was unchanged, this would assist
SGLT2 mRNA levels are elevated in renal proximal cells weight loss and improve glycaemic control, which would
isolated from the urine of subjects with T2DM [18], but this provide a rationale for selective SGLT2 inhibition.
has not been confirmed in other studies [20].
Inhibition of SGLT2 seems to exceed the capacity of Experiments of nature: familial renal glucosuria
SGLT1 to reabsorb all the filtered glucose in patients with Urinary glucose excretion can occur in the absence of either
T2DM, such that enough glucose is excreted in the urine to generalized proximal tubular dysfunction or hyperglycae-
improve glycaemic control. Assuming a mean blood glucose mia in inherited disorders of familial renal glucosuria (FRG)
concentration of 10 mmol/L (180 mg/dL) and a normal rate of [21–23]. Mutations in the SLC5A2 gene are responsible for

66
Review Trends in Pharmacological Sciences February 2011, Vol. 32, No. 2

most FRG; heterozygous individuals usually develop mild vided that the compound is absorbed early in the upper
glucosuria (<10 g/1.73 m2/day) or sometimes no glucosuria, small intestine so that it does not prevent complete ab-
whereas those with homozygous or compound heterozygous sorption of glucose by the small intestine. In addition, it
SCL5A2 mutations show more severe glucosuria [21–23]. would require good bioavailability, safety and tolerability,
Twenty different mutations of the SCL5A2 gene have been and preferably a plasma half-life that would suit once-daily
reported, including missense, nonsense and splicing muta- oral administration.
tions [22,23]. Possible effects (other than glucosuria) have
been suggested for some patients with FRG, such as auto- Selective SGLT2 inhibitors
antibodies (but without clinical evidence of autoimmune The first orally active SGLT inhibitor was T-1095, devel-
disease) and bacteriuria, which is usually asymptomatic oped by Tanabe Seiyaku and Johnson & Johnson [32]. This
[24–26]. However, FRG does not seem to be associated with O-glucoside prodrug is converted in the intestine to an
significant clinical consequences and is considered a benign active form, T-1095A, which inhibits human SGLT2 (IC50
condition [27]. This supports the potential safety of life-long 50 nM) with four-fold greater selectivity than SGLT1 (IC50
glucosuria by loss of SGLT2 function. 200 nM) [33]. Administration of T-1095 to diabetic animal
models increased urinary glucose excretion and reduced
Development of SGLT inhibitors hyperglycaemia [33]. However, T-1095 and several other
Phlorizin (Figure 4) is a naturally occurring phenol glyco- early SGLT2 inhibitors (e.g. sergliflozin and remogliflozin)
side first isolated from the bark of apple trees in 1835 [28]. have not proceeded to clinical development, owing at least
In the late 19th and early 20th centuries, it was shown that partly to the lability of the O-glucosidic linkage and the
orally administered phlorizin increases urinary glucose resulting short half-life and poor bioavailability. Orally
excretion and loss of body weight. By the 1960s, it was active SGLT2 inhibitors now in clinical development in-
known that phlorizin inhibits glucose transport by erythro- clude BI 10733, canagliflozin (TA7284) and dapagliflozin
cytes and cells in the kidney and small intestine [28]. Proof (Figure 4). BI 10773 is a C-glucoside inhibitor with >2500-
of principle that promotion of renal glucose excretion could fold selectivity for SGLT2 (IC50 3.1 nM) over SGLT1 (IC50
be used in the treatment of diabetic states was provided by 7.7 mM) and it is not hydrolyzed by intestinal glucosidases
Rossetti and colleagues in 1987 [29]. Phlorizin restored [34]. Canagliflozin (TA7284) is also a C-glucoside and has
euglycaemia and insulin sensitivity in partially pancre- 200-fold selectivity for SGLT2 (IC50 2.2 nM) over SGLT1
atectomized rats, and the hyperglycaemia and insulin (IC50 0.44 mM) [35]. Dapagliflozin is a C-aryl glucoside with
resistance returned if phlorizin was discontinued [29]. In 1200-fold selectivity for SGLT2 (IC50 1.1 nM) over SGLT1
the same diabetic animal model, restoration of euglycae- (IC50 1.32 mM) [36,37]. It has been reported that each of
mia with phlorizin also improved insulin secretion, which these agents induces significant urinary glucose excretion,
indicates that hyperglycaemia itself (i.e. glucotoxicity) reduces blood glucose and reduces body weight in obese
makes an important contribution to insulin resistance diabetic rodent models.
and reduced b-cell function [30]. It was subsequently
shown that oral administration of phlorizin improves glu- Clinical studies: pharmacokinetics and
cose tolerance in normal mice without significantly affect- pharmacodynamics
ing the insulin response [31]. To date, papers describing clinical trial data have only
It was not until the genes for SGLT1 and SGLT2 were been published for dapagliflozin. This agent displayed
cloned in the 1990 s that phlorizin was identified as a dose-proportional plasma concentrations in single-dose
potent, competitive inhibitor of both transporters. Howev- (2.5–500 mg) and multiple-dose (2.5–100 mg daily for 14
er, the lack of selectivity towards SGLT2 is one reason why days) studies in healthy and T2DM subjects [38,39]. The
phlorizin was precluded as a drug candidate for the treat- amount of glucose excreted in urine increased with the
ment of T2DM [28]. In addition, phlorizin is poorly dose, and a near-maximal rate of urinary glucose excretion
absorbed in the small intestine and it has a short half-life, (3 g/h) occurred for 25 mg of dapagliflozin [38]. This
because the O-glucoside bond is highly susceptible to hy- represented approximately 20–25% inhibition of renal glu-
drolysis by intestinal glucosidases. From a therapeutic cose reabsorption. It is estimated that the maximum inhi-
perspective, an ideal renal glucose reabsorption inhibitor bition of glucose reabsorption by SGLT2 inhibition is self-
for T2DM treatment would selectively inhibit SGLT2, but limiting at approximately 30–40%. This probably reflects
could exert some non-selective inhibition of SGLT1, pro- the number of fully functional nephrons at any given time,
[()TD$FIG]
(a) (b) (c)

HO OH CH3 HO
O OH HO OH
O HO
HO O F
O
O HO CI
HO OH O S
HO
HO
HO OH

Phlorizin Dapagliflozin Canagliflozin


TRENDS in Pharmacological Sciences

Figure 4. Structure of (a) phlorizin, (b) dapagliflozin and (c) canagliflozin. The structure of BI 10773 is not available at this time.

67
Review Trends in Pharmacological Sciences February 2011, Vol. 32, No. 2

Table 2. Effects of dapagliflozin in clinical trials in subjects with T2DMa


Trial Treatment Treatment effect Reference
(duration) (change from baseline and
placebo subtracted)
Phase IIa Dapagliflozin (5–100 mg/day)  FSG #19–39 mg/dLb [39]
(14 days) vs placebo  OGTT AUC #10–23% a
n = 47
Phase IIb Dapagliflozin (2.5–50 mg/day)  HbA1c #0.37–0.72%b [40]
(12 weeks) vs placebo vs metformin c  FPG #10–25 mg/dLb
n = 348  PPG AUC #7050–10,150 mg min/dLb
 Body weight #1.3–2.0 kg b
Phase III (24 weeks) Dapagliflozin (2.5, 5, and  HbA1c #0.35–0.66%b [41]
n = 485 10 mg/day) vs placebo  FPG #11–25 mg/dLb
 Body weight #1.0–1.1kg
Phase III Dapagliflozin (2.5–10 mg/day)  HbA1c #0.37–0.54%b [42]
(24 weeks) + metformin vs placebo + metformin d  FPG #12–18 mg/dLb
n = 546  Body weight #1.3–2.1kg
Phase IIb Dapagliflozin (10 and 20 mg/day)  HbA1c #0.61% and 0.69% [45]
(12 weeks) n = 68 + insulin vs placebo + insulin e  FPG "2 and #10 mg/dL
 Body weight #4.5 kg and 4.3 kg
Phase III (24 weeks) Dapagliflozin (2.5–10 mg/day)  HbA1c #0.45–0.60%b [46]
n = 807 + insulin  OADs vs placebo  FPG #9–18 mg/dLb
+ insulin  OADs  Body weight #1.0–1.69 kg b
Phase III (24 weeks) Dapagliflozin (2.5, 5,10 mg/day)  HbA1c #0.45–0.69%b [44]
n = 597 + glimepiride vs placebo + glimepiride f  Body weight #0.46-1.54kg b
Phase III (52 weeks) Dapagliflozin (2.5–10 mg/day)  HbA1c #0.52%g [43]
n = 814 + metformin vs glipizide (5 mg/day  Body weight #3.2 kg h
up to  20 mg/day) + metformin
a
Abbreviations: AUC, area under the plasma concentration time curve; FSG, fasting serum glucose; FPG, fasting plasma glucose; OADs, oral antidiabetic drugs; OGTT oral
glucose tolerance test; # decrease; " increase.
b
Statistically significant versus placebo (P<0.05).
c
Metformin 1500 mg/day.
d
Pretreatment dose of metformin continued.
e
Pretreatment dose of insulin 50 U/day, halved during treatment phase of study.
f
Glimepiride 4 mg/day.
g
Statistically significant versus baseline and same reduction from baseline as glipizide group.
h
Statistically significant versus glipizide group (P < 0.0001).

anticipated competition for SGLT2 transporters by in- or an adjuvant to other antidiabetic agents have noted
creased glucose concentrations in proximal convoluted similar improvements in glycaemic control and reductions
tubule versus the SGLT2 inhibitor, and the compensating in body weight. In patients receiving insulin, the reduc-
capacity of SGLT1 in the S3 segment. tions in HbA1c and body weight during concomitant dapa-
gliflozin administration were achieved with a modest
Clinical studies in subjects with T2DM reduction in insulin dose [44,45].
Improved glycaemic control in T2DM has been observed for The mild osmotic diuresis observed with dapagliflozin,
dapagliflozin as monotherapy [39–41] and for combination as evidenced by initial increases in urinary volume up to
therapy with metformin [42,43] or glimepiride [44] and as 450 mL/day, was accompanied by small increases in
combination therapy for subjects treated with insulin plus blood urea nitrogen and haematocrit and a modest de-
oral antidiabetic agents (Table 2) [45,46]. In a randomized, crease in systolic blood pressure (–3 to –6 mm Hg) [40–
double-blind, placebo-controlled (RDBPC) 12-week study, 42]. A long-term increase in urinary glucose can predispose
348 previously untreated subjects with T2DM received patients to genitourinary infections, and although dapagli-
2.5–50 mg/day dapagliflozin, placebo, or 1500 mg/day met- flozin increased the incidence of these infections, they were
formin (as a therapeutic benchmark) [40]. Dapagliflozin transient and none was severe. More patients also reported
increased urinary glucose excretion from 6–11 g/day at symptoms of hypoglycaemia with dapagliflozin than with
baseline to 52–85 g/day at study end, whereas placebo- placebo, but these were mild and similar to those in
and metformin-treated patients continued to excrete metformin-treated subjects [40].
6 g/day. HbA1c levels were reduced by 0.55% to 0.90% Further clinical trials to examine the effects of dapagli-
(6–10 mmol/mol) with dapagliflozin compared with 0.18% flozin in conjunction with other anti-diabetes medications
(2 mmol/mol) for placebo and 0.73% (8 mmol/mol) for are in progress; there are additional clinical trial pro-
metformin. The reduction in body weight was 1.3–2.0 kg grammes with canagliflozin, BI 10773 and other candidate
more with dapagliflozin than with placebo, a difference SGLT2 inhibitors. Details are available on the clinical
that was particularly evident during the first week. This trials website at http://clinicaltrials.gov/.
might reflect an initial diuretic effect of SGLT2 inhibition,
whereas continued weight loss is consistent with the calo- Concluding remarks
rific value of ongoing glucose elimination in urine. Further Because T2DM is a progressive disease, glucotoxicity poses
RDBPC or parallel-group studies in T2DM for up to 52 an insidious cumulative risk that requires early and effec-
weeks with dapagliflozin (2.5–10 mg/day) as monotherapy tive intervention to prevent, defer and ameliorate chronic

68
Review Trends in Pharmacological Sciences February 2011, Vol. 32, No. 2

complications [47]. Glucotoxicity represents a pathogenic doubled during administration of dapagliflozin. However,
spiral that is caused by insulin resistance and b-cell dys- the incidence of urinary tract infections was not consistently
function and also aggravates these conditions. Indeed, increased [42–44]. In most cases, genital infections were
chronic hyperglycaemia can accelerate the loss of b-cell recognized and dealt with by patients such that they re-
function and mass by generating reactive oxygen species solved before the patient attended the next clinic visit. Few
and through wide daily fluctuations in blood glucose [48–50]. patients required medical intervention or discontinued the
Selective inhibition of SGLT2 decreases plasma glucose by SGLT2 inhibitor.
increasing urinary glucose excretion, which offers a novel In summary, selective SGLT2 inhibition is a novel
mechanism that is independent of the amount of circulating approach for reducing hyperglycaemia in patients with
insulin and the degree of insulin resistance. This approach is T2DM through increased renal glucose elimination. Al-
additive and complementary with other blood-glucose-low- though natural mutations that inactivate SGLT2 are gen-
ering therapies and carries low risk of hypoglycaemia be- erally well tolerated in patients with FRG, long-term
cause SGLT1 can reabsorb an increasing proportion of clinical trials with pharmacological inhibitors of SGLT2
filtered glucose at low plasma glucose concentrations. More- are awaited to confirm safety in patients with T2DM.
over, inhibition of SGLT2 does not impede counter-regula- Selective SGLT2 inhibitors have potential advantages
tory mechanisms of glucose homoeostasis. conferred by their independence from insulin secretion
Urinary elimination of excess glucose by inhibiting or action; notably, their glucose-lowering efficacy should
SGLT2 facilitates body weight reduction. Obesity is a not decrease significantly with disease progression. Weight
strong risk factor for insulin resistance and T2DM, and loss is a further benefit of SGLT2 inhibition, and there is
each of these conditions is an independent risk factor for low potential to cause hypoglycaemia due to glucose reab-
cardiovascular disease [51]. Weight loss in obese patients sorption by SGLT1 and lack of interference with counter-
with T2DM can reduce the risk of all-cause mortality [52], regulation. SGLT2 inhibition is compatible with other
and weight management is therefore a valuable component antidiabetic therapies, including insulin, and might there-
of diabetes management [5,6,53]. Patients with T2DM fore benefit any stage in the natural history of T2DM.
typically require escalating insulin doses, often in combi-
nation with oral agents to counter the progressive increase Disclosure statement
in hyperglycaemia [4]. This frequently promotes weight Professor Bailey has received research grants from and
gain and potentially compromises the value of glycaemic provided remunerated ad hoc consultancy to pharmaceu-
control to reduce long-term vascular complications [4]. tical companies that produce antidiabetic and antiobesity
Thus, a reduced insulin dose requirement with SGLT2 agents. He has undertaken research on dapagliflozin (Bris-
inhibition constitutes an additional mechanism to assist tol-Myers Squibb and AstraZeneca) but records no conflict
weight reduction alongside the caloric dissipation of extra of interest for this review.
glucose in urine [44]. Weight loss plus a modest decrease in
plasma volume during SGLT2 inhibition could at least Acknowledgements
partly account for a reduction in blood pressure. The author gratefully acknowledges the assistance of Dr Ann P. Tighe of
Diabetes is typically associated with a deterioration in Parexel, which receives funding from Bristol-Myers Squibb and
AstraZeneca.
renal function, especially in the elderly. Inhibition of SGLT2
does not interfere with the glomerular filtration of glucose,
but does require a filtration rate that is sufficient to deliver References
1 Turner, R.C. et al. (1999) Glycemic control with diet, sulfonylurea,
substantial amounts of glucose into the proximal tubules. metformin, or insulin in patients with type 2 diabetes mellitus:
Thus, SGLT2 inhibition should not adversely affect the progressive requirement for multiple therapies (UKPDS 49). UK
progression of diabetic glomerular pathology, but this ther- Prospective Diabetes Study (UKPDS) Group. J. Am. Med. Assoc.
apy is not suitable for patients with a severely reduced 281, 2005–2012
2 Holman, R.R. et al. (2007) Addition of biphasic, prandial, or basal
glomerular filtration rate. SGLT2 inhibition might reduce
insulin to oral therapy in type 2 diabetes. N. Engl. J. Med. 357,
the availability of glucose as an energy source within cells 1716–1730
lining the S1 segment of the proximal tubules, which could 3 Roden, M. (2009) Optimal insulin treatment in type 2 diabetes. N. Engl.
potentially influence active transport functions in this part J. Med. 361, 1801–1803
of the tubule. The nephron retains considerable spare ca- 4 Nathan, D.M. et al. (2009) Medical management of hyperglycemia in
type 2 diabetes: a consensus algorithm for the initiation and
pacity to compensate for small changes in sodium reuptake
adjustment of therapy: a consensus statement of the American
in the S1 segment, but initial transient changes in renal Diabetes Association and the European Association for the Study of
sodium handling might not be unexpected. Evidence from Diabetes. Diabetes Care 32, 193–203
patients with FRG suggests that protracted exposure to 5 Pi-Sunyer, F.X. (2009) The impact of weight gain on motivation,
glucose along subsequent regions of the nephron and to compliance, and metabolic control in patients with type 2 diabetes
mellitus. Postgrad. Med. 121, 94–107
the urothelium of the urinary tracts does not have detri- 6 Kahn, S.E. et al. (2006) Mechanisms linking obesity to insulin
mental effects. However, renal glucose elimination does resistance and type 2 diabetes. Nature 444, 840–846
generate modest osmotic diuresis (although there has been 7 UK Prospective Diabetes Study (UKPDS) Group (1998) Effect of
no evidence of dehydration in trials to date), and diuresis intensive blood-glucose control with sulfonylureas or insulin
might explain the small but consistent reduction in blood compared with conventional treatment and risk of complications in
patients with type 2 diabetes (UKPDS 33). Lancet 352, 837–853
pressure. High levels of urinary glucose excretion increase 8 Makimattila, S. et al. (1999) Causes of weight gain during insulin
the risk of genitourinary infections in uncontrolled diabetes, therapy with and without metformin in patients with type II diabetes
and the incidence of genital infections was approximately mellitus. Diabetologia 42, 406–412

69
Review Trends in Pharmacological Sciences February 2011, Vol. 32, No. 2

9 Gerich, J.E. et al. (2001) Renal gluconeogenesis: its importance in 35 Liang, Y. et al. (2009) JNJ-28431754/TA-7284, an SGLT inhibitor,
human glucose homeostasis. Diabetes Care 24, 382–391 lowers blood glucose and reduces body weight in obese and type 2
10 Meyer, C. et al. (1998) Abnormal renal and hepatic glucose metabolism diabetic animal models. Diabetes 58 (Suppl 1), Abstract 534-P
in type 2 diabetes mellitus. J. Clin. Invest. 102, 619–624 36 Han, S. et al. (2008) Dapagliflozin, a selective SGLT2 inhibitor,
11 Abdul-Ghani, M.A. and DeFronzo, R.A. (2008) Inhibition of renal improves glucose homeostasis in normal and diabetic rats. Diabetes
glucose absorption: a novel strategy for achieving glucose control in 57, 1723–1729
type 2 diabetes mellitus. Endocr. Pract. 14, 782–790 37 Meng, W. et al. (2008) Discovery of dapagliflozin: a potent,
12 Wright, E.M. et al. (2007) Active sugar transport in health and disease. selective renal sodium-dependent glucose cotransporter 2 (SGLT2)
J. Intern. Med. 261, 32–43 inhibitor for the treatment of type 2 diabetes. J. Med. Chem. 51, 1145–
13 Chen, J. et al. (2008) Tissue expression profiling of the sodium-glucose 1149
co-transporter (SGLT) family: implication for targeting SGLT2 in type 38 Komoroski, B. et al. (2009) Dapagliflozin, a novel SGLT2 inhibitor,
2 diabetes patients. Diabetes 57 (Suppl. 1), A682 induces dose-dependent glucosuria in healthy subjects. Clin.
14 Wright, E.M. (2001) Renal Na+-glucose cotransporters. Am. J. Physiol. Pharmacol. Ther. 85, 520–526
Renal Physiol. 280, F10–F18 39 Komoroski, B. et al. (2009) Dapagliflozin, a novel, selective SGLT2
15 Kanai, Y. et al. (1994) The human kidney low affinity Na+/glucose inhibitor, improved glycemic control over 2 weeks in patients with type
cotransporter SGLT2. Delineation of the major renal reabsorptive 2 diabetes mellitus. Clin. Pharmacol. Ther. 85, 513–519
mechanism for D-glucose. J. Clin. Invest. 93, 397–404 40 List, J.F. et al. (2009) Sodium-glucose cotransport inhibition with
16 Hediger, M.A. and Rhoads, D.B. (1994) Molecular physiology of dapagliflozin in type 2 diabetes. Diabetes Care 32, 650–657
sodium-glucose transporters. Physiol. Rev. 74, 993–1026 41 Ferrannini, E. et al. (2010) Dapagliflozin monotherapy in type 2
17 Mogensen, C.E. (1971) Maximum tubular reabsorption capacity for diabetic patients with inadequate glycemic control by diet and
glucose and renal hemodynamcis during rapid hypertonic glucose exercise: a randomized, double-blind, placebo-controlled, phase III
infusion in normal and diabetic subjects. Scand. J. Clin. Lab. Invest. trial. Diabetes Care 33, 2217–2224
28, 101–109 42 Bailey, C.J. et al. (2010) Effect of dapagliflozin in patients with type 2
18 Rahmoune, H. et al. (2005) Glucose transporters in human renal diabetes who have inadequate glycaemic control with metformin: a
proximal tubular cells isolated from the urine of patients with non- randomised, double-blind, placebo-controlled trial. Lancet 375, 2223–
insulin-dependent diabetes. Diabetes 54, 3427–3434 2233
19 Freitas, H.S. et al. (2008) Na+-glucose transporter-2 messenger 43 Nauck, M. et al. (2010) Dapagliflozin vs glipizide in patients with type 2
ribonucleic acid expression in kidney of diabetic rats correlates with diabetes mellitus inadequately controlled on metformin: 52-week
glycemic levels: involvement of hepatocyte nuclear factor-1alpha results of a double-blind, randomised, controlled trial, In
expression and activity. Endocrinology 149, 717–724 Proceedings of: the 46th Annual Meeting of the European Association
20 Lee, Y.J. et al. (2007) Regulatory mechanisms of Na+/glucose for the Study of Diabetes (EASD), September 20–24, 2010, Stockholm,
cotransporters in renal proximal tubule cells. Kidney Int. Suppl. 72, Sweden Abstract 241
S27–S35 44 Strojek, K. et al. (2010) Efficacy and safety of dapagliflozin in patients
21 Calado, J. et al. (2006) Familial renal glucosuria: SLC5A2 mutation with type 2 diabetes mellitus and inadequate glycaemic control on
analysis and evidence of salt-wasting. Kidney Int. 69, 852–855 glimepiride monotherapy, In Proceedings of the 46th Annual Meeting of
22 Calado, J. et al. (2008) Twenty-one additional cases of familial renal the European Association for the Study of Diabetes (EASD), September
glucosuria: absence of genetic heterogeneity, high prevalence of private 20–24, 2010, Stockholm, Sweden Abstract 870
mutations and further evidence of volume depletion. Nephrol. Dial. 45 Wilding, J.P.H. et al. (2009) A study of dapagliflozin in patients with
Transplant. 23, 3874–3879 type 2 diabetes receiving high doses of insulin plus insulin sensitizers.
23 Santer, R. et al. (2003) Molecular analysis of the SGLT2 gene Applicability of a novel insulin-independent treatment. Diabetes Care
in patients with renal glucosuria. J. Am. Soc. Nephrol. 14, 2873– 32, 1656–1662
2882 46 Wilding, J.P.H. et al. (2010) Dapagliflozin in patients with type 2
24 Scholl-Burgi, S. et al. (2004) Long-term outcome of renal glucosuria diabetes poorly controlled on insulin therapy—efficacy of a novel
type 0: the original patient and his natural history. Nephrol. Dial. insulin-independent treatment. Diabetes 59 (Suppl. 1), A21–A22
Transplant. 19, 2394–2396 47 Hsu, W.C. (2009) Consequences of delaying progression to optimal
25 De Marchi, S. et al. (1983) Is renal glycosuria a benign condition? Proc. therapy in patients with type 2 diabetes not achieving glycemic goals.
Eur. Dial. Transplant Assoc. 20, 681–685 South. Med. J. 102, 67–76
26 De Paoli, P. et al. (1984) Immunological characterization of renal 48 Newsholme, P. et al. (2007) Diabetes associated cell stress and
glycosuria patients. Clin. Exp. Immunol. 56, 289–294 dysfunction: role of mitochondrial and non-mitochondrial ROS
27 Santer, R. and Calado, J. (2010) Familial renal glucosuria and SGLT2; production and activity. J. Physiol. 583, 9–24
from a Mendelian trait to a therapeutic target. Clin. J. Am. Soc. 49 Del Guerra, S. et al. (2007) Gliclazide protects human islet beta-cells
Nephrol. 5, 133–141 from apoptosis induced by intermittent high glucose. Diabetes Metab.
28 Ehrenkranz, J.R. et al. (2005) Phlorizin: a review. Diabetes Metab. Res. Res. Rev. 23, 234–238
Rev. 21, 31–38 50 Lupi, R. et al. (2007) Insulin secretion defects of human type 2 diabetic
29 Rossetti, L. et al. (1987) Correction of hyperglycemia with phlorizin islets are corrected in vitro by a new reactive oxygen species scavenger.
normalizes tissue sensitivity to insulin in diabetic rats. J. Clin. Invest. Diabetes Metab. 33, 340–345
79, 1510–1515 51 Expert Panel on Detection, Evaluation, and Treatment of High Blood
30 Rossetti, L. et al. (1987) Effect of chronic hyperglycemia on in vivo Cholesterol in Adults (2001) Third Report of the National Cholesterol
insulin secretion in partially pancreatectomized rats. J. Clin. Invest. Education Program (NCEP) Expert Panel on Detection, Evaluation,
80, 1037–1044 and Treatment of High Blood Cholesterol in Adults (Adult Treatment
31 Takii, H. et al. (1997) Lowering effect of phenolic glycosides on the rise Panel III). J. Am. Med. Assoc. 285, 2486–2497
in postprandial glucose in mice. Biosci. Biotechnol. Biochem. 61, 1531– 52 Poobalan, A.S. et al. (2007) Long-term weight loss effects on all
1535 cause mortality in overweight/obese populations. Obes. Rev. 8, 503–
32 Tsujihara, K. et al. (1999) Na+-glucose cotransporter (SGLT) inhibitors 513
as antidiabetic agents. 4. Synthesis and pharmacological properties of 53 Ford, E.S. et al. (2008) Metabolic syndrome and incident diabetes:
40 -dehydroxyphlorizin derivatives substituted on the B ring. J. Med. current state of the evidence. Diabetes Care 31, 1898–1904
Chem. 42, 5311–5324 54 Scranton, R.E. et al. (2007) A randomized, double-blind, placebo-
33 Oku, A. et al. (1999) T-1095, an inhibitor of renal Na+-glucose controlled trial to assess safety and tolerability during treatment of
cotransporters, may provide a novel approach to treating diabetes. type 2 diabetes with usual diabetes therapy and either Cycloset or
Diabetes 48, 1794–1800 placebo. BMC Endocr. Disord. 7, 3
34 Grempler, R. et al. (2009) In vitro properties and in vivo effect on 55 Bays, H.E. et al. (2008) Colesevelam hydrochloride therapy in patients
urinary glucose excretion of BI 10773, a novel selective SGLT2 with type 2 diabetes mellitus treated with metformin: glucose and lipid
inhibitor. Diabetes 58 (Suppl. 1). Abstract 521-P effects. Arch. Intern. Med. 168, 1975–1983

70
Review Trends in Pharmacological Sciences February 2011, Vol. 32, No. 2

56 Shang, Q. et al. (2010) Colesevelam improves insulin resistance in a 60 Flatt, P.R. et al. (2009) Recent advances in antidiabetic drug therapies
diet-induced obesity (F-DIO) rat model by increasing the release of targeting the enteroinsular axis. Curr. Drug. Metab. 10, 125–137
GLP-1. Am. J. Physiol. Gastrointest Liver Physiol. 298, G419–424 61 Ryan, G.J. et al. (2005) Pramlintide in the treatment of type 1 and type
57 Drucker, D.J. (2005) Biologic actions and therapeutic potential of the 2 diabetes mellitus. Clin. Ther. 27, 1500–1512
proglucagon-derived peptides. Nat. Clin. Pract. Endocrinol. Metab. 1, 62 Bailey, C.J. and Turner, R.C. (1996) Metformin. N. Engl. J. Med. 334,
22–31 574–579
58 Natali, A. and Ferrannini, E. (2006) Effects of metformin and 63 Bakris, G.L. et al. (2009) Renal sodium-glucose transport: role in
thiazolidinediones on suppression of hepatic glucose production and diabetes mellitus and potential clinical implications. Kidney Int. 75,
stimulation of glucose uptake in type 2 diabetes: a systematic review. 1272–1277
Diabetologia 49, 434–441 64 Bays, H. (2009) From victim to ally: the kidney as an emerging target
59 Yki-Jarvinen, H. (2004) Thiazolidinediones. N. Engl. J. Med. 351, for the treatment of diabetes mellitus. Curr. Med. Res. Opin. 25, 671–
1106–1118 681

71

You might also like