You are on page 1of 13

JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 75, NO.

4, 2020

ª 2020 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION

PUBLISHED BY ELSEVIER

THE PRESENT AND FUTURE

JACC STATE-OF-THE-ART REVIEW

Mechanisms of Cardiorenal Effects


of Sodium-Glucose
Cotransporter 2 Inhibitors
JACC State-of-the-Art Review

Thomas A. Zelniker, MD, MSC, Eugene Braunwald, MD

ABSTRACT

Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a new drug class approved for treatment of diabetes, have been
shown to possess a favorable metabolic profile and to significantly reduce atherosclerotic events, hospitalization for
heart failure, cardiovascular and total mortality, and progression of chronic kidney disease. Although initially considered
to be only glucose-lowering agents, the effects of SGLT2i have expanded far beyond that, and their use is now being
studied in the treatment of heart failure and chronic kidney disease, even in patients without diabetes. It is therefore
critical for cardiologists, diabetologists, nephrologists, and primary care physicians to be familiar with this drug class.
This first part of this 2-part review provides an overview of the current understanding of the mechanisms of the
cardio-metabolic-renal benefits of SGLT2i. The second part summarizes the recent clinical trials of SGLT2i.
(J Am Coll Cardiol 2020;75:422–34) © 2020 by the American College of Cardiology Foundation.

T ype 2 diabetes mellitus (T2DM) and its most


serious consequences—heart
kidney disease—represent 1 of the greatest
pandemics of chronic disease in the world today
disease and
cular risk of new glucose-lowering agents in pa-
tients with T2DM (8,9). A number of these trials
confirmed the safety of approved
drugs. However, quite unexpectedly, they also
antidiabetic

(1–4). Modern treatment of T2DM began with paren- identified 2 drug classes, sodium-glucose cotrans-
teral insulin, discovered by Banting and Best, porter 2 inhibitors (SGLT2i) and glucagon like pep-
almost a century ago (5). Since then, several classes tide 1 receptor agonists, that reduce major adverse
of orally active drugs that improve glycemic control cardiovascular events, including cardiovascular
have become available and have been widely used. death (10,11). These findings are revolutionizing
Concerns emerged early in this century about the the care of patients with T2DM, and in the case of
cardiovascular safety of some of these antidiabetic the SGLT2i, have been found to be beneficial in
compounds (6,7). As a consequence, regulatory nondiabetic patients with heart failure (HF) as
bodies now require pharmaceutical manufacturers well (12). Important new information on SGLT2i is
to conduct trials to exclude an excess of cardiovas- growing rapidly, and although we reviewed this

Listen to this manuscript’s


audio summary by
Editor-in-Chief From the TIMI Study Group, Cardiovascular Division, Brigham and Women’s Hospital, and Department of Medicine, Harvard
Dr. Valentin Fuster on Medical School, Boston, Massachusetts. Dr. Zelniker is currently affiliated with the Division of Cardiology, Vienna General Hos-
JACC.org. pital, Medical University of Vienna, Vienna, Austria. Dr. Zelniker is supported by the German Research Foundation (Deutsche
Forschungsgemeinschaft ZE 1109/1-1); and has received lecture fees from AstraZeneca. Dr. Braunwald has received research grants
through his institution from AstraZeneca, Daiichi-Sankyo, GlaxoSmithKline, Merck, and Novartis; has consultancies with Amgen,
Cardurion, MyoKardia, NovoNordisk, and Verve; has received personal fees for lectures from Medscape; and has performed
uncompensated consultancies and lectures for Novartis and The Medicines Company.

Manuscript received October 2, 2019; revised manuscript received November 15, 2019, accepted November 17, 2019.

ISSN 0735-1097/$36.00 https://doi.org/10.1016/j.jacc.2019.11.031


JACC VOL. 75, NO. 4, 2020 Zelniker and Braunwald 423
FEBRUARY 4, 2020:422–34 Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors

then reabsorbed by SGLT1 located further ABBREVIATIONS


HIGHLIGHTS AND ACRONYMS
distal in the S2 /S3 segments of the proximal
 T2DM, HF, and CKD are closely convoluted tubules.
AMPK = 50 adenosine
intertwined. SGLT2 are high-capacity/low-affinity monophosphate-activated
transporters and are expressed in higher protein kinase
 SGLT2i exert pleiotropic metabolic and
concentrations as compared with SGLT1, CaMKII = CaDD/calmodulin
direct cardioprotective and neph-
which are low-capacity/high-affinity trans- dependent kinase
roprotective effects.
porters (13). Moreover, the 2 transporters are CKD = chronic kidney disease

 SGLT2i reduce inflammation, oxidative characterized by different stoichiometries: eGFR = estimated glomerular

stress, fibrosis, intraglomerular hyper- SGLT1 transports 2 Na þ per molecule of filtration rate

glucose and SGLT2 transports 1 (15). SGLT2, NAFLD = nonalcoholic fatty


tension, and sympathetic nervous system
þ liver disease
activation, and may improve mitochon- but not SGLT1, colocalize with the renal Na /
RAAS = renin-angiotensin-
drial function and myocardial efficiency. hydrogen exchanger NHE3, which is largely
aldosterone system
responsible for Naþ reabsorption in the
 Cardiologists, diabetologists, nephrolo- SGLT2i = sodium-glucose
proximal tubule, and SGLT2i appear to cross- cotransporter 2 inhibitor
gists, and primary care physicians should
react with NHE3 and thereby inhibit reab-
T2DM = type 2 diabetes
be familiar with this drug class.
sorption and augment natriuresis (16,17). mellitus
Both homozygous as well as heterozygous
mutations in the SGLT2 coding gene SLC5A2 can
subject in the Journal in 2018 (13), we now bring it
result in familial renal glucosuria (18,19). Patients
up to date in this 2-part series. Here we focus on
with these rare conditions are normoglycemic, have
the mechanisms of the cardiac and renal effects of
normal kidney function, and may exhibit incomplete
these agents.
penetrance.
GLUCOSE-LOWERING AND METABOLIC The glucose-lowering ability of the inhibition of
EFFECTS MEDIATED BY SGLT2i SGLT2 through urinary glucose excretion has been
reported to reduce glycated hemoglobin A1c in pa-
Under physiological conditions in normal adults, tients with T2DM, generally by between 0.5% and
approximately 180 g of glucose are filtered by the 1.0%. The glucosuria depends on the circulating
renal glomeruli and reabsorbed completely by the glucose concentration and kidney function (20).
renal tubules each day. In patients with T2DM, SGLT2i requires glomerular filtration rate for
glucose can be detected in the urine when blood glucose-lowering, which declines in patients
glucose concentration exceeds a threshold of with estimated glomerular filtration rate (eGFR)
approximately 200 mg/dl (11.1 mmol/l); marked glu- <45 ml/min/1.73 m 2. Similarly, glucosuria with
cosuria is a hallmark of untreated T2DM. SGLT2i is attenuated in patients without hypergly-
Cell membranes are impermeable to glucose and cemia. Therefore, the risk of hypoglycemia with this
require facilitated diffusion carriers or specific mem- drug class is low.
brane transport proteins for its uptake. In the kid- SGLT2i also promote weight loss, resulting from
neys, tubular glucose reabsorption is coupled with loss of adipose tissue. Although the caloric loss
sodium (Naþ) that follows the electrochemical through urinary glucose excretion equals approxi-
gradient of a higher Naþ concentration in the mately 200 kcal/day, data from randomized clinical
glomerular filtrate to a lower intracellular concen- trials indicate that weight loss of approximately 2 to
tration in epithelial tubular cells. This gradient is 3 kg develops in about 6 months and then plateaus. It
maintained through the basolateral Na þ/K þ ATPase of has been suggested that compensatory hyperphagia
the epithelial tubular cell. Glucose enters the cell with hunger for sugar-rich foods may explain the
coupled with Na þ through sodium-glucose cotrans- diminishing effect of continued caloric loss (21).
porters 1 and 2 (SGLT1/2) and then exits the cell Experimental data suggest that SGLT2i triggers a
passively via the basolateral glucose transporter 2. fasting-like state with increased fatty acid oxidation
SGLT2 are expressed nearly exclusively in the kidney; and ketogenesis, and reduces adipose tissue and liver
SGLT1 is present in the kidney as well, but has also steatosis (22). Molecular mechanisms mediating these
been detected in the intestines and heart (14). SGLT2 metabolic effects include: 1) activation of the 5 0
is located in the S 1 (first) segment of the proximal adenosine monophosphate-activated protein kinase
convoluted tubule and accounts for approximately (AMPK); 2) inhibition of mechanistic target of rapa-
90% of the reabsorbed glucose. Residual glucose is mycin; and 3) increased hepatic and plasma levels of
424 Zelniker and Braunwald JACC VOL. 75, NO. 4, 2020

Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors FEBRUARY 4, 2020:422–34

fibroblast growth factor 21, independent of insulin or activation of the RAAS, leading to a vicious circle (25).
glucagon activity, as has been shown in obese, SGLT2i have the potential to mitigate these patho-
nondiabetic mice (22). Moreover, SGLT2i have been physiological changes.
linked to a reduction in epicardial fat (23), a biologi- Patients with T2DM are also at high risk of
cally highly-active tissue involved in leptin and the developing CKD, which serves as an additional
renin-angiotensin-aldosterone system (RAAS) driver for multiple cardiovascular complications
signaling, and may thereby be cardioprotective. In a including HF. Diabetic CKD is the leading cause of
mouse model, ipragliflozin, an SGLT2i approved for end-stage kidney disease and, therefore, the need
clinical use in Japan, has also been shown to reduce for renal replacement therapy. SGLT2i have emerged
inflammation of adipose tissue by a reduction in the as a promising drug class that may interrupt this
ratio of pro-inflammatory M-1-like adipose tissue other vicious circle in patients with T2DM.
macrophages to anti-inflammatory M-2 tissue mac-
EFFECTS OF SGLT2i ON THE
rophages (24).
CARDIOVASCULAR SYSTEM

CARDIORENAL INTERACTIONS IN SGLT2i-mediated natriuresis and glucosuria lower


PATIENTS WITH TYPE 2 DIABETES cardiac pre-load and reduce pulmonary congestion
and systemic edema. These effects appear to play a
T2DM, particularly long-standing and/or requiring role in the reduction of hospitalizations for HF
insulin, is frequently complicated by HF and chronic observed in the cardiovascular and kidney outcomes
kidney disease (CKD). These 3 entities (T2DM, HF, trials in patients with T2DM discussed in Part 2 of this
and CKD) have interconnected pathways and share review (31). Urinary output returns to normal within
several metabolic and signaling cascades; the pres- 12 weeks after treatment is begun (32,33). As is the
ence of any of the 3 conditions worsens the other 2 case with other diuretic agents, the natriuresis
(25). Dyslipidemia, hypertension (Figure 1), and a induced by SGLT2i is attenuated over time through
prothrombotic state, which occur commonly in pa- compensatory mechanisms and achievement of a
tients with T2DM, contribute to the development of stable state. It is possible that SGLT2i-mediated glu-
accelerated atherosclerosis. The latter increases the coresis (compared with other diuretic agents whose
risk of myocardial infarction, which may be respon- actions are primarily natriuretic) results in greater
sible for, or contribute to, the development of HF proportional reductions of interstitial compared with
with reduced ejection fraction (26–28). intravascular volume. This may occur as a conse-
A variety of pathophysiological mechanisms that quence of greater electrolyte-free water clearance by
contribute to the development of HF in T2DM are peripheral sequestration of osmotically inactive so-
shown in Table 1. A specific diabetic cardiomyopathy, dium (34).
considered to be a separate entity independent of SGLT2i have been shown to significantly reduce
coronary artery disease and arterial hypertension, has the Na þ content of the skin, as measured by 23
Na-
been described (27,29,30). The hyperglycemia of magnetic resonance imaging (35). An increased
T2DM, especially when it is prolonged, results in (cutaneous) tissue Naþ content has been correlated
advanced glycation end-products, oxidative stress, with left ventricular hypertrophy (36). The SGLT2i-
inflammation, and apoptosis, which together with induced Naþ depletion may improve left ventricular
microvascular coronary artery disease appear to be remodeling and ejection fraction. SGLT2i also reduce
responsible for the development of diabetic cardio- cardiac afterload by lowering arterial pressure by 3 to
myopathy. This condition, like myocardial infarction, 5 mm Hg without increasing heart rate, and have been
may also be responsible for and/or contribute shown to reduce arterial stiffness (37,38). The
importantly to the development of HF. In long- reduction of blood pressure is preserved, even in
standing T2DM, the combination of multiple patients with reduced glomerular filtration rate,
myocardial infarcts and diabetic cardiomyopathy can suggesting that SGLT2i may reduce the sympathetic
cause HF, which, until the development of SGLT2i, nervous system overdrive of HF (Figure 1) (39).
had been difficult to treat and associated with poor Studies carried out both in vitro and in vivo have
prognosis. The low cardiac output of HF causes a shown that norepinephrine up-regulates expression
reduction of renal blood flow and elevated renal of SGLT2, thereby enhancing Na þ and glucose reab-
venous pressure, which are accompanied by sorption by the proximal tubule, while, conversely,
JACC VOL. 75, NO. 4, 2020 Zelniker and Braunwald 425
FEBRUARY 4, 2020:422–34 Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors

F I G U R E 1 Inter-Relationships Between SGLT2i and the Sympathetic Nervous System and Illustration of the Positive Role of SGLT2i in
Hypertension and Heart Failure

Adapted from Scheen (39). SGLT2i ¼ sodium-glucose cotransporter 2 inhibitors; SNS ¼ sympathetic nervous system.

SGLT2i reduces tyrosine hydroxylase and noradren- improve the state of an “energy starved” heart, a
aline in the kidney and heart thereby contributing to condition observed in T2DM and HF. In support of the
the natriuresis and glucoresis (40). suggestion that SGLT2i improves cardiac metabolism
in these patients is the observation that treatment of
EFFECTS OF SGLT2i ON nondiabetic pigs with ischemic HF with empagliflozin
CARDIAC METABOLISM
reduces adverse cardiac remodeling by switching
myocardial substrate utilization from glucose toward
The heart requires energy continuously and is capable
oxidation of free fatty acids, ketone bodies, and
of consuming a variety of substrates, including both
branched-chain amino acids (46,47). Treatment of
glucose and free fatty acids. Under conditions of
diabetic mice with empagliflozin resulted in an
stress, such as HF and/or T2DM, glucose utilization is
increased cardiac production of ATP by about 30%
impaired, and the heart relies more heavily on the
metabolism of free fatty acids and ketones (41,42).
Beta-hydroxybutyrate has been suggested to act as a T A B L E 1 Pathophysiological Mechanisms That Contribute to
“superfuel” by increasing the metabolic efficiency of the Development of Heart Failure in Patients With Type 2

the heart (43). The shift toward more consumption of Diabetes Mellitus

ketone bodies has been hypothesized to improve the Altered myocardial substrate metabolism

ATP supply to the failing heart (44). Chronic infusion Mitochondrial bioenergetics
Oxidative stress
of beta-hydroxybutyrate has been shown to improve
Lipotoxicity
cardiac function, efficiency, and remodeling in a
Inflammation
canine tachypacing-induced HF model (44). Further-
Endoplasmic reticulum stress
more, ketone bodies also exert anti-inflammatory ef- Insulin signaling
fects by suppressing activation of the nucleotide- Beta-2 adrenergic receptor signaling
binding oligomerization domain-like receptor P3 G protein-coupled receptor kinase 2 signaling
(NLRP3) inflammasome activity (45) (Table 2). Renin angiotensin aldosterone signaling

It has been suggested that SGLT2i alter fuel con- Autophagy


Advanced glycation end products
sumption with an increase in oxidation of fatty acids
and ketogenesis and with a concomitant reduction in Modified with permission from Kenny and Abel (26).
utilization of carbohydrates. These alterations
426 Zelniker and Braunwald JACC VOL. 75, NO. 4, 2020

Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors FEBRUARY 4, 2020:422–34

Studies in patients with T2DM have linked mito-


T A B L E 2 Lessons Learned on Presumed Mechanisms of SGLT2i Activity
chondrial dysfunction with ventricular hypertrophy
SGLT2i and heart failure
1. Osmotic diuresis and natriuresis reduce cardiac overload.
and fibrosis (54). It has been suggested that SGLT2i
a. Greater reductions of interstitial fluid compared with other diuretic agents (34) may improve mitochondrial function and reduce
2. Direct cardiac effects:
a. SGLT2i inhibit NHE-1 (60–62)
oxygen-reactive stress (55).
b. SGLT2i reduce CaMKII (reduces sarcoplasmic Caþþ leak / improves contractility) (59) Hyperglycemia contributes to activation of the
c. Increased phosphorylation levels of myofilament regulatory proteins? (66)
d. Epigenetic modification? (65)
RAAS, which leads to overproduction of angiotensin
3. Reduction of sympathetic nervous system overdrive (39,40) II and aldosterone, both of which induce cardiac hy-
4. Possible improvement in myocardial efficiency (43,44,46–48,69)
5. Improved oxygen delivery through stimulation of renal EPO secretion (52,90)
pertrophy and fibrosis and impair cardiac relaxation.
6. Reduction in inflammation (/ activation of adenosine monophosphate-activated Hyperglycemia also results in the formation of
protein kinase) (57,64)
7. Reduction of oxidative stress (improving mitochondrial function) (58,96,101)
advanced glycation end products, which cause cross-
8. Metabolic: lowering of HbA1c, blood pressure, body weight, vascular stiffness (71) linking of collagen (56), leading to increased fibrosis
SGLT2i and atherosclerosis with increased myocardial stiffness and impaired
1. Reduction in inflammation (72)
2. Reduction in epicardial fat and noxious signals including leptin and RAAS (23) cardiac relaxation. Studies in mice suggest that
3. Reduction in oxidative stress (58,95,96,100,101) dapagliflozin reduces inflammation in myofibro-
4. Improved endothelial function (58,73)
5. Cardioprotective effects by shifting circulating vascular progenitor cell toward M2 blasts, an effect mediated through increased activa-
polarization (74) tion of AMPK independently of SGLT2i (57).
6. Reduction in uric acid (91,92)
7. Improvement in NAFLD? (78,79) Moreover, empagliflozin has been shown to prevent
8. Metabolic: lowering of HbA1c (/AGE, fibrosis), blood pressure, body weight (link to HF) hyperglycemic impairment of proteinase-activated
a. Reduction in vascular stiffness (71)
9. BUT! Small increases LDL-C (clinical implications unknown) (80) receptor 2–mediated vasodilation by inhibition of
SGLT2i and progression of kidney disease aortic endothelial SGLT2 and minimizing oxidative
1. Metabolic: lowering of HbA1c (/AGE, fibrosis), and body weight (link to HF) (20)
stress (58).
2. Reduction in blood pressure and vascular stiffness (71)
3. Restoration of the tubuloglomerular feedback (84,85,87) Empagliflozin has been shown to reduce Ca þþ/
4. Reduction in workload regarding ATP production (90)
calmodulin dependent kinase (CaMKII) activity in
5. Anti-inflammatory and antifibrotic effects, reduction in oxidative stress (95–102)
6. Reduction in uric acid (91,92) murine ventricular myocytes, which is increased in
Interaction between heart and kidney HF (Table 2). Empagliflozin also reduces the CaMKII
1. Prevention of progress of disease in one organ may prevent deterioration of the
phosphorylation of the cardiac ryanodine receptor in
other (25)
both murine and human ventricular myocytes,
CaMKII ¼ Caþþ/calmodulin dependent kinase; EPO ¼ erythropoietin; HbA1c ¼ glycated hemoglobin A1c; resulting in significantly reduced sarcoplasmic Ca þþ
HF ¼ heart failure; LDL-C ¼ low-density lipoprotein cholesterol; NAFLD ¼ nonalcoholic fatty liver disease;
RAAS ¼ renin-angiotensin-aldosterone system; SGLT2i ¼ sodium glucose cotransporter inhibitor. leak and thereby improved contractility (59). Another
important biological explanation for the benefit of
SGLT2i has been proposed by Uthman et al. (60,61),
who found that SGLT2i directly inhibits the Naþ/
through an increase in the rate of glucose and fatty Hydrogen exchanger 1 (NHE1) in the myocardium.
acid oxidation (48–50). Inhibition of cardiac NHE1 by SGLT2i reduces cyto-
Furthermore, treatment with the SGLT2i cana- plasmic Na þ and Caþþ levels and increases mito-
gliflozin for 4 weeks has been shown to attenuate chondrial Ca þþ levels (Figure 2) (60,62). Empagliflozin
myocardial ischemia and reperfusion injury ex vivo also exerts direct cardioprotective effects during
in both diabetic and nondiabetic rats (51). More- ischemia by inhibiting NHE1 (61).
over, SGLT2i-mediated stimulation of renal eryth- Experimental data also suggest that treatment with
ropoietin secretion leading to new red blood cell empagliflozin protects cardiac function in a nondia-
formation and increased hematocrit has been hy- betic mouse model of HF with preserved ejection
pothesized to contribute to improved oxygen de- fraction induced by transverse aortic constriction
livery to the heart with resultant improvement in (63). In a nondiabetic porcine model of HF with pre-
cardiac function (52). The improved cardiac meta- served ejection fraction, treatment with dapagliflozin
bolism and the increased reabsorption of magne- reversed left ventricular concentric remodeling by
sium and potassium from the tubular fluid might reducing sympathetic tone, mitigating inflammatory
also exert antiarrhythmic effects that could result response in the aorta and reactivation of the nitric
in lower rates of sudden cardiac death (53). Dysre- oxide–cyclic guanosine 30 ,5 0 -monophosphate (cGMP)–
gulation of metabolic substrate, inflammation, cGMP-dependent protein kinase (NO-cGMP-PKG)
increased apoptosis, and impaired calcium handling pathway (64).
have all been suggested to explain the cardiac In addition, a transcriptomics study suggested
impairment in diabetic cardiomyopathy (29). that SGLT2i may exhibit favorable effects on HF by
JACC VOL. 75, NO. 4, 2020 Zelniker and Braunwald 427
FEBRUARY 4, 2020:422–34 Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors

F I G U R E 2 Direct Effects of SGLT2i on the Cardiomyocyte

HF and/or T2DM Na+/Ca++ Na+/K+


NHE1 Anporter ATPase
Na+ Ca++ 2K+

H+ 3Na+ 3Na+
ATP
Nac+ Cac++
H+

3Na++
Na++ CaM++
Ca++
ATP
Mitochondrion
A Cardiomyocyte
SGLT2i Na+/Ca++ Na+/K+
SGLT2i NHE1 Anporter ATPase
Na+ Ca++ 2K+

H+ 3Na+ 3Na+
ATP

Nac + ++
Cac
Na+
CaM++
Ca++ ATP
Mitochondrion
B Cardiomyocyte

(A) Conditions, such as heart failure and/or type 2 diabetes mellitus (T2DM), increase the expression of Naþ/Hydrogen exchanger 1 (NHE-1)
and result in increased cytosolic Naþ and Caþþ concentrations, but reduced mitochondrial Caþ and mitochondrial ATP generation.
(B) Inhibition of cardiac NHE-1 reduces cytoplasmic Naþ and Caþþ levels and increases mitochondrial Caþþ levels, resulting in improved
mitochondrial respiration (and increase in ATP production) and viability of cardiomyocytes. Modified from Uthman et al. (55).
SGLT2i ¼ sodium-glucose cotransporter 2 inhibitors.

epigenetic modification (65). Furthermore, empagli- absence of T2DM, and this is now being studied in
flozin has also been shown to reduce passive dia- patients, as summarized in Part 2 of this review (31).
stolic tension in isolated ventricular trabeculae In summary, SGLT2i improve ventricular loading
obtained from patients with end-stage HF with conditions, cardiac metabolism, bioenergetics, ven-
preserved ejection fraction (66) (Figure 3). This tricular remodeling, and exert direct cardioprotective
improvement was observed immediately after and possibly antiarrhythmic effects (Tables 2 and 3,
intravenous administration of empagliflozin, sug- Central Illustration) (70).
gesting that SGLT2i increased phosphorylation
levels of myofilament regulatory proteins (66). VASCULAR EFFECTS
SGLT2i have also been shown to significantly reduce
E/eʹ from 13.7 to 12.1 cm/s and from 9.3 to 8.5 cm/s SGLT2i lowers systolic and diastolic pressures
in patients with heart failure with preserved ejec- without increasing heart rate (71) (Tables 2 and 3).
tion fraction (67,68). Empagliflozin has also SGLT2i exert anti-inflammatory effects through
been reported to improve left ventricular pump multiple mechanisms, including weight loss,
function in mice with doxorubicin-induced cardio- reduction in inflammation of adipose tissue, in-
myopathy (69). Taken together, these findings crease in ketone bodies, and reduction in uric acid
suggest that SGLT2i may be effective in HF with (72). Canagliflozin may improve endothelial function
preserved ejection fraction (Figure 3), even in the by inhibiting inflammatory pathways through
428 Zelniker and Braunwald JACC VOL. 75, NO. 4, 2020

Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors FEBRUARY 4, 2020:422–34

F I G U R E 3 Skinned Cardiomyocytes Isolated From Patients With HFpEF and From Healthy Donors

4.0

3.5

3.0
Fpassive norm.
2.5
P < 0.0001
2.0

1.5
P < 0.0001
1.0

0.5

0
1.8 1.9 2 2.1 2.2 2.3 2.4
Sarcomere Length (µm)
HFpEF HFpEF After Empagliflozin
NF NF After Empagliflozin

Normalized passive stiffness (F) measured at various sarcomere lengths from 1.8 to 2.4 mm of cardiomyocytes from HFpEF and nonfailing (NF)
myocardium from patients incubated 60 min with empagliflozin. Reproduced with permission from Pabel et al. (66)

blocking interleukin-1 b–stimulated cytokine and NONALCOHOLIC FATTY LIVER DISEASE


chemokine secretion in vascular endothelial cells
(73). Empagliflozin may exert cardioprotective anti- Nonalcoholic fatty liver disease (NAFLD) is
inflammatory and vessel-regenerative effects by frequently observed in patients with T2DM as well as
shifting circulating vascular progenitor cell toward in patients with metabolic syndrome, and is associ-
M2 polarization (74). Furthermore, empagliflozin ated with an increased risk of adverse cardiovascular
has been shown to improve coronary microvascular events (76). Multiple mechanisms, including sys-
function and increase cardiac output in mice, as temic inflammation, oxidative stress, endothelial
evidenced by increases in coronary blood flow and dysfunction, and changes in lipid profiles, by which
fractional ventricular area change as shown by ul- NAFLD increases cardiovascular risk have been
trasound imaging (75). identified (76). To date, only limited treatment stra-
tegies exist to address this condition with an esti-
mated worldwide prevalence of 25% (77). SGLT2i
T A B L E 3 Overview of Mechanisms of Favorable Cardio-Metabolic-Renal Effects
have been shown to reduce adipose tissue,
Heart Atherosclerotic Diabetic Kidney and therefore, may favorably influence NAFLD. In
Failure Effect Disease
diabetic mice, dapagliflozin compared with insulin
Glucose lowering U
prevented NAFLD (78). In a double blind, controlled,
Reduction in body weight U U U
randomized trial of patients with T2DM, dapagli-
Lowering of blood pressure U U U
Natriuresis U U
flozin lowered serum aminotransferases, as corre-
Anti-inflammation U U U lates of liver fat, and reduced liver fat content as
Antifibrotic U U quantified using CT/MR imaging (79). No data for
Reduction in extracellular matrix turnover U U the effect of SGLT2i on NAFLD in the absence of
Amelioration of intrarenal hypoxia U T2DM have been reported to date. However, SGLT2i
Restoration of the tubuloglomerular feedback U
have been shown to increase levels of low-density
Reduction in natriuretic peptides U U
lipoprotein cholesterol by 3 to 5 mg/dl (80), an
Reduction in energy demand U U
effect that could oppose slightly the aforementioned
Reduction in liver fat U
benefits of this drug class.
JACC VOL. 75, NO. 4, 2020 Zelniker and Braunwald 429
FEBRUARY 4, 2020:422–34 Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors

C ENTR AL I LL U STRA T I O N Sodium-Glucose Cotransporter 2 Inhibitor Cardiorenal Protection


Mechanistic Overview

Zelniker, T.A. et al. J Am Coll Cardiol. 2020;75(4):422–34.

Adapted from Vergara et al. (106).

EFFECTS OF SGLT2i ON KIDNEY FUNCTION pathways promoting inflammation, and glomerular


fibrosis causing progressive reduction of glomerular
T2DM results in multiple metabolic and hemody- filtration rate, progressive albuminuria, and ulti-
namic changes that promote structural changes in the mately, end-stage kidney disease.
kidneys, affecting primarily the microcirculation The protective effects of SGLT2i on the kidney are
(Table 2). In the early stage of diabetic kidney disease, believed to be mediated by a number of both hemo-
glomerular hyperfiltration is observed, which is dynamic and nonhemodynamic mechanisms (Central
associated with an increase of single-glomerular Illustration). The improvements of cardiac function
filtration rate to adapt to a reduced number of by SGLT2i may contribute to their favorable effects on
nephrons, systemic arterial hypertension, or the kidneys halting the “vicious cardiorenal circle”
increased metabolic demand (81). Hemodynamic (Tables 2 and 3, Figure 4) (25). Activation of the
changes through vasodilation of afferent and/or tubuloglomerular feedback has been hypothesized to
contraction of efferent glomerular arterioles exert be mainly responsible. The action of SGLT2i in the
mechanical (shear and tensile) stress on the glomer- proximal convoluted tubule results in increased
ular capillaries, basement membrane, podocytes, and concentrations of Na þ at the macula densa. Primarily
the proximal tubular epithelium, ultimately causing driven through adenosine-mediated signal cascades,
renal hypertrophy and expansion of the mesangial this causes vasoconstriction of the afferent arterioles
matrix (82). These changes activate further harmful and thereby lowers the intraglomerular pressure and
430 Zelniker and Braunwald JACC VOL. 75, NO. 4, 2020

Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors FEBRUARY 4, 2020:422–34

F I G U R E 4 Suggested Mechanisms for Cardiorenal Protection With SGLT2i

Adapted from Evans et al. (105).

consequently reduces hyperfiltration and related SGLT2i-induced reduction in glucose reabsorption


damage (83). An elegant study recently confirmed reduces the ATP demand of the basolateral Naþ/Kþ
the SGLT2i-mediated restoration of the tubuloglo- ATPase of the tubular cells, which maintain the
merular feedback, demonstrating afferent arteriolar electrochemical Na þ-gradient between the tubular
vasoconstriction after administration of empagli- fluid and the tubular cells. Consequently, myofibro-
flozin in a diabetes mouse model (84,85). A recent blasts, transformed through cell injury, revert into
secondary analysis from the EMPA-REG (Empagli- erythropoietin-producing fibroblasts. It has been
flozin, Cardiovascular Outcomes, and Mortality in proposed that by this mechanism, SGLT2i attenuates
Type 2 Diabetes) Outcome trial reported that empa- metabolic stress in kidney cells and subsequently also
gliflozin reduced the incidence of a composite renal reduces the sympathetic nervous system overdrive
outcome irrespective of baseline medication but the (90).
magnitude of the observed reductions tended to be SGLT2i-mediated glucosuria and consequent
larger in patients treated with angiotensin-converting increased glucose concentrations in the distal seg-
enzyme inhibitors/angiotensin receptor blocker (86). ments of the renal tubular cells trigger the secretion
After causing reduction of eGFR in the range of 3 to of uric acid through urate transporter 1 and glucose
5 ml/min/1.73 m 2 over the first few weeks of SGLT2i, transporter 9 into the tubular fluid (91,92). SGLT2i
eGFR then stabilizes and SGTL2i preserves and delays might thereby prevent harmful downstream effects of
the progression of CKD (Figure 5) (87,88). uric acid including inflammation, oxidative stress,
A mediation analysis from the EMPA-REG Outcome and activation of RAAS (93).
trial suggested that changes in hematocrit and he- Hyperglycemia generates reactive oxygen species
moglobin were important mediators of the risk and activates inflammatory responses, which
reduction of cardiovascular death by empagliflozin contribute to the pathogenesis of diabetic kidney
(89). Given that the urinary output returns to baseline disease (94). In a diet-induced mouse model of T2DM,
values shortly after start of treatment, Sano and canagliflozin reduced intrarenal angiotensinogen
Goto (90) have suggested that the hypoxic microen- production, monocyte/macrophage infiltration and
vironment of renal tubular cells was improved by the oxidative stress and thereby reduced renal inflam-
persistent increase in hematocrit resulting from an mation and renal tubular fibrosis (95,96). In a diabetic
elevation in erythropoietin levels by lowering the mouse model with nephropathy, SGLT2i have been
energy requirement of renal tubular cells. The found to reduce the activity of NLRP3 inflammasome
JACC VOL. 75, NO. 4, 2020 Zelniker and Braunwald 431
FEBRUARY 4, 2020:422–34 Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors

F I G U R E 5 eGFR Over Time Was Different Between Empagliflozin and Placebo in the EMPA-REG OUTCOME Trial in Type 2 Diabetes Mellitus Patients

78

76
Adjusted Mean (SE) eGFR
(mL/min/1.73m2)

74

72

70

68

66
4 12 28 52 66 80 94 108 122 136 150 164 178 192
Baseline
Week
Patients analyzed
Placebo 2,323 2,295 2,267 2,205 2,121 2,064 1,927 1,981 1,763 1,479 1,262 1,123 977 731 448

Empagliflozin 4,644 4,578 4,533 4,451 4,318 4,225 4,018 4,131 3,710 3,103 2,654 2,387 2,087 1,623 1,037

Estimated glomerular filtration rate (eGFR) is according to Modification of Diet in Renal Disease formula. Reproduced with permission from Wanner et al. (87).

(97), a multimeric protein complex that is involved in Although the reductions in arterial pressure and
inflammatory processes including the production of circulating glucose concentrations may contribute to
interleukin-1 b and interleukin-18 (98). Further evi- the salutary effects of SGLT2i on the kidney (Table 2)
dence of anti-inflammatory and antifibrotic effects of (71), it is unlikely that the rather modest reductions
SGLT2i stems from an intriguing study that selected in these functions would translate into an effect size
biomarkers through a systems biology approach and of the observed magnitude of approximately 45% of
linked data from diabetic kidney disease models with the composite kidney outcome (worsening in eGFR,
information on SGLT2i molecular effects (99). When end-stage kidney disease, or death from renal cause)
compared with the widely used sulfonylurea agent as observed in a pooled analysis of the 3 SGLT2i
glimepiride, canagliflozin reduced plasma levels of cardiovascular outcomes trials (10). Exploratory an-
tumor necrosis factor receptor 1, IL-6, matrix metal- alyses from the EMPA-REG Outcome trial and
loproteinase 7, and fibronectin (99). These findings CANTATA-SU (Canagliflozin Versus Glimepiride in
lend further support to antifibrotic and anti- Patients With Type 2 Diabetes Inadequately
inflammatory properties of SGLT2i. Controlled With Metformin), a phase 3 trial that
Empagliflozin has also been shown to improve compared canagliflozin with glimepiride, indicate
mitochondrial function and autophagy, improving that the nephroprotective effects of SGLT2i are in-
renal morphology in diabetic mice by reducing frag- dependent of their hypoglycemic actions (103,104).
mentation of renal tubular mitochondria through
activation of AMPK (100). In a study of patients with
T2DM and urinary albumin to creatinine ratio CONCLUSIONS
>100 mg/g dapagliflozin significantly reduced urinary
metabolites linked to mitochondrial metabolism The cardio-metabolic-renal effects of SGLT2i are most
(101). These findings suggest that improvements in likely multifactorial, and although initially conceived
mitochondrial function may mediate, or at least only as glucose-lowering agents, they have expanded
contribute to, the nephroprotective properties of this far beyond this action (Table 2). The unexpected and
drug class. Another putative mechanism is that impressive reductions in important clinical endpoints
SGLT2i regulate endoplasmic reticulum stress- observed in large-scale clinical trials, described in
mediated apoptosis in proximal tubular cells (102). Part 2 of this review (31), have led investigators from
432 Zelniker and Braunwald JACC VOL. 75, NO. 4, 2020

Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors FEBRUARY 4, 2020:422–34

the bedside back to the bench to improve the under-


standing of the drugs in this class and elucidate their ADDRESS FOR CORRESPONDENCE: Dr. Eugene
mechanisms of action. Braunwald, TIMI Study Group, Brigham and Women’s
For several important papers on this subject that Hospital, 60 Fenwood Road, 7th Floor, Boston,
were published after submission of this paper on Massachusetts 02115. E-mail: ebraunwald@partners.
October 3, 2019, please see the Online Appendix. org. Twitter: @TIMIStudyGroup, @ZelnikerThomas.

REFERENCES

1. Dabelea D, Mayer-Davis EJ, Saydah S. Preva- 12. McMurray JJV, Solomon SD, Inzucchi SE, et al. 25. Braunwald E. Diabetes, heart failure, and renal
lence of type 1 and type 2 diabetes among children Dapagliflozin in patients with heart failure and dysfunction: The vicious circles. Prog Cardiovasc
and adolescents from 2001 to 2009. JAMA 2014; reduced ejection fraction. N Engl J Med 2019;381: Dis 2019;62:298–302.
311:1778–86. 1995–2008.
26. Kenny HC, Abel ED. Heart failure in type 2
2. Menke A, Casagrande S, Cowie CC. Prevalence 13. Zelniker TA, Braunwald E. Cardiac and renal diabetes mellitus. Circ Res 2019;124:121–41.
of diabetes in adolescents aged 12 to 19 Years in effects of sodium-glucose co-transporter 2 in-
27. Jia G, Hill MA, Sowers JR. Diabetic cardio-
the United States, 2005-2014. JAMA 2016;316: hibitors in diabetes: JACC State-of-the-Art Re-
myopathy: An update of mechanisms contrib-
344–5. view. J Am Coll Cardiol 2018;72:1845–55.
uting to this clinical entity. Circ Res 2018;122:
3. Cho NH, Shaw JE, Karuranga S, et al. IDF Dia- 14. Wright EM, Turk E. The sodium/glucose 624–38.
betes atlas: global estimates of diabetes preva- cotransport family SLC5. Pflugers Arch 2004;447:
lence for 2017 and projections for 2045. Diabetes 28. Dunlay SM, Givertz MM, Aguilar D, et al. Type
510–8.
Res Clin Pract 2018;138:271–81. 2 diabetes mellitus and heart failure: A Scientific
15. Kanai Y, Lee WS, You G, Brown D, Hediger MA. Statement From the American Heart Association
4. Benjamin EJ, Muntner P, Alonso A, et al. Heart The human kidney low affinity Naþ/glucose and the Heart Failure Society of America. Circula-
disease and stroke statistics-2019 update: a report cotransporter SGLT2. Delineation of the major tion 2019;140:e294–324.
from the American Heart Association. Circulation renal reabsorptive mechanism for D-glucose.
2019;139:e56–528. J Clin Invest 1994;93:397–404. 29. Marwick TH, Ritchie R, Shaw JE, Kaye D. Im-
plications of underlying mechanisms for the
5. Vecchio I, Tornali C, Bragazzi NL, Martini M. The 16. Alicic RZ, Neumiller JJ, Johnson EJ, Dieter B, recognition and management of diabetic cardio-
discovery of insulin: an important milestone in the Tuttle KR. Sodium-glucose cotransporter 2 inhi- myopathy. J Am Coll Cardiol 2018;71:339–51.
history of medicine. Front Endocrinol (Lausanne) bition and diabetic kidney disease. Diabetes 2019;
2018;9:613. 68:248–57. 30. Dillmann WH. Diabetic cardiomyopathy. Circ
Res 2019;124:1160–2.
6. Nissen SE, Wolski K. Effect of rosiglitazone on the 17. Packer M. Activation and inhibition of sodium-
risk of myocardial infarction and death from car- hydrogen exchanger is a mechanism that links the 31. Zelniker TA, Braunwald E. Clinical benefit of
diovascular causes. N Engl J Med 2007;356:2457–71. pathophysiology and treatment of diabetes mel- cardiorenal effects of sodium-glucose cotrans-

7. U.S. Food and Drug Administration. FDA back- litus with that of heart failure. Circulation 2017; porter 2 inhibitors: JACC state-of-the-art review. J
ground document: Endocrinologic and Metabolic 136:1548–59. Am Coll Cardiol 2020;75:436–48.

Drugs Advisory Committee Meeting. 2018. Avail- 18. Santer R, Kinner M, Lassen CL, et al. Mo- 32. Tanaka H, Takano K, Iijima H, et al. Factors
able at: https://www.fda.gov/media/121272/ lecular analysis of the SGLT2 gene in patients affecting canagliflozin-induced transient urine
download. Accessed September 27, 2019. with renal glucosuria. J Am Soc Nephrol 2003; volume increase in patients with type 2 diabetes
8. U.S. Food and Drug Administration. Guidance 14:2873–82. mellitus. Adv Ther 2017;34:436–51.
for industry: diabetes mellitus—evaluating cardio- 19. Santer R, Calado J. Familial renal glucosuria 33. Sha S, Polidori D, Heise T, et al. Effect of the
vascular risk in new antidiabetic therapies to treat and SGLT2: from a mendelian trait to a therapeutic sodium glucose co-transporter 2 inhibitor cana-
type 2 diabetes. 2008. Available at: https://www. target. Clin J Am Soc Nephrol 2010;5:133–41. gliflozin on plasma volume in patients with type 2
fda.gov/downloads/Drugs/GuidanceCompliance diabetes mellitus. Diabetes Obes Metab 2014;16:
20. Cherney DZI, Cooper ME, Tikkanen I, et al.
RegulatoryInformation/Guidances/ucm071627. 1087–95.
Pooled analysis of Phase III trials indicate con-
pdf. Accessed September 27, 2019.
trasting influences of renal function on blood 34. Hallow KM, Helmlinger G, Greasley PJ,
9. European Medical Agency. Guideline on clinical pressure, body weight, and HbA1c reductions with McMurray JJV, Boulton DW. Why do SGLT2 in-
investigation of medicinal products in the treat- empagliflozin. Kidney Int 2018;93:231–44. hibitors reduce heart failure hospitalization? A
ment or prevention of diabetes mellitus. 2018.
21. Horie I, Abiru N, Hongo R, et al. Increased differential volume regulation hypothesis. Dia-
Available at: https://www.ema.europa.eu/
sugar intake as a form of compensatory hyper- betes Obes Metab 2018;20:479–87.
documents/scientific-guideline/draft-guideline-
phagia in patients with type 2 diabetes under 35. Karg MV, Bosch A, Kannenkeril D, et al. SGLT-
clinical-investigation-medicinal-products-treatment-
dapagliflozin treatment. Diabetes Res Clin Pract 2-inhibition with dapagliflozin reduces tissue so-
prevention-diabetes-mellitus_en.pdf. Accessed
2018;135:178–84. dium content: a randomised controlled trial. Car-
September 27, 2019.
22. Osataphan S, Macchi C, Singhal G, et al. SGLT2 diovasc Diabetol 2018;17:5.
10. Zelniker TA, Wiviott SD, Raz I, et al. SGLT2 inhibitors
inhibition reprograms systemic metabolism via 36. Schneider MP, Raff U, Kopp C, et al. Skin So-
for primary and secondary prevention of cardiovascular
FGF21-dependent and -independent mechanisms. dium concentration correlates with left ventricular
and renal outcomes in type 2 diabetes: a systematic
JCI Insight 2019;4:123130. hypertrophy in CKD. J Am Soc Nephrol 2017;28:
review and meta-analysis of cardiovascular outcome
trials. Lancet 2019;393:31–9. 23. Sato T, Aizawa Y, Yuasa S, et al. The effect of 1867–76.
dapagliflozin treatment on epicardial adipose tis-
11. Kristensen SL, Rorth R, Jhund PS, et al. Car- 37. Kario K, Okada K, Kato M, et al. 24-hour blood
sue volume. Cardiovasc Diabetol 2018;17:6.
diovascular, mortality, and kidney outcomes with pressure-lowering effect of an SGLT-2 inhibitor in
GLP-1 receptor agonists in patients with type 2 24. Miyachi Y, Tsuchiya K, Shiba K, et al. A reduced patients with diabetes and uncontrolled nocturnal
diabetes: a systematic review and meta-analysis of M1-like/M2-like ratio of macrophages in healthy hypertension: Results from the randomized,
cardiovascular outcome trials. Lancet Diabetes adipose tissue expansion during SGLT2 inhibition. placebo-controlled SACRA study. Circulation 2018
Endocrinol 2019;7:776–85. Sci Rep 2018;8:16113. Nov 29 [E-pub ahead of print].
JACC VOL. 75, NO. 4, 2020 Zelniker and Braunwald 433
FEBRUARY 4, 2020:422–34 Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors

38. Wiviott SD, Raz I, Bonaca MP, et al. Dapagli- with impaired mitochondrial function and dy- function in patients with type 2 diabetes. Car-
flozin and cardiovascular outcomes in type 2 dia- namics in type 2 diabetic but not in obese patients. diovasc Diabetol 2018;17:73.
betes. N Engl J Med 2019;380:347–57. Circulation 2014;130:554–64.
68. Soga F, Tanaka H, Tatsumi K, et al. Impact of
39. Scheen AJ. Effect of SGLT2 inhibitors on the 55. Uthman L, Baartscheer A, Schumacher CA, dapagliflozin on left ventricular diastolic function
sympathetic nervous system and blood pressure. et al. Direct cardiac actions of sodium glucose of patients with type 2 diabetic mellitus with
Curr Cardiol Rep 2019;21:70. cotransporter 2 inhibitors target pathogenic chronic heart failure. Cardiovasc Diabetol 2018;17:
mechanisms underlying heart failure in diabetic 132.
40. Matthews VB, Elliot RH, Rudnicka C, et al.
Role of the sympathetic nervous system in regu- patients. Front Physiol 2018;9:1575. 69. Oh CM, Cho S, Jang JY, et al. Cardioprotective
lation of the sodium glucose cotransporter 2. 56. Zelniker TA, Jarolim P, Scirica BM, et al. potential of an SGLT2 inhibitor against
J Hypertens 2017;35:2059–68. Biomarker of collagen turnover (C-terminal telo- doxorubicin-induced heart failure. Korean Circ J
peptide) and prognosis in patients with non- ST 2019;49:1183–95.
41. Tran DH, Wang ZV. Glucose metabolism in
cardiac hypertrophy and heart failure. J Am Heart -elevation acute coronary syndromes. J Am Heart 70. Verma S, McMurray JJV. SGLT2 inhibitors and
Assoc 2019;8:e012673. Assoc 2019;8:e011444. mechanisms of cardiovascular benefit: a state-of-
57. Ye Y, Bajaj M, Yang HC, Perez-Polo JR, the-art review. Diabetologia 2018;61:2108–17.
42. Sowton AP, Griffin JL, Murray AJ. Metabolic
profiling of the diabetic heart: Toward a richer Birnbaum Y. SGLT-2 inhibition with dapagliflozin 71. Cherney DZ, Perkins BA, Soleymanlou N, et al.
picture. Front Physiol 2019;10:639. reduces the activation of the Nlrp3/ASC inflam- The effect of empagliflozin on arterial stiffness
masome and attenuates the development of dia- and heart rate variability in subjects with uncom-
43. Ferrannini E, Baldi S, Frascerra S, et al. Shift to
betic cardiomyopathy in mice with type 2 diabetes. plicated type 1 diabetes mellitus. Cardiovasc Dia-
fatty substrate utilization in response to sodium-
Further augmentation of the effects with sax- betol 2014;13:28.
glucose cotransporter 2 inhibition in subjects
agliptin, a DPP4 inhibitor. Cardiovasc Drugs Ther
without diabetes and patients with type 2 dia- 72. Bonnet F, Scheen AJ. Effects of SGLT2 in-
2017;31:119–32.
betes. Diabetes 2016;65:1190–5. hibitors on systemic and tissue low-grade inflam-
58. El-Daly M, Pulakazhi Venu VK, et al. Hyper- mation: The potential contribution to diabetes
44. Horton JL, Davidson MT, Kurishima C, et al.
glycaemic impairment of PAR2-mediated vasodi- complications and cardiovascular disease. Dia-
The failing heart utilizes 3-hydroxybutyrate as a
lation: prevention by inhibition of aortic betes Metab 2018;44:457–64.
metabolic stress defense. JCI Insight 2019;4:
endothelial sodium-glucose-co-transporter-2 and 73. Mancini SJ, Boyd D, Katwan OJ, et al. Cana-
12407.
minimizing oxidative stress. Vascul Pharmacol gliflozin inhibits interleukin-1beta-stimulated
45. Youm YH, Nguyen KY, Grant RW, et al. The 2018;109:56–71. cytokine and chemokine secretion in vascular
ketone metabolite beta-hydroxybutyrate blocks
59. Mustroph J, Wagemann O, Lucht CM, et al. endothelial cells by AMP-activated protein kinase-
NLRP3 inflammasome-mediated inflammatory
Empagliflozin reduces Ca/calmodulin-dependent dependent and -independent mechanisms. Sci Rep
disease. Nat Med 2015;21:263–9.
kinase II activity in isolated ventricular car- 2018;8:5276.
46. Santos-Gallego CG, Requena-Ibanez JA, San diomyocytes. ESC Heart Fail 2018;5:642–8. 74. Hess DA, Terenzi DC, Trac JZ, et al. SGLT2
Antonio R, et al. Empagliflozin ameliorates
60. Uthman L, Baartscheer A, Bleijlevens B, et al. Inhibition with empagliflozin increases circulating
adverse left ventricular remodeling in non-
Class effects of SGLT2 inhibitors in mouse car- provascular progenitor cells in people with type 2
ndiabetic heart failure by enhancing myocardial
diomyocytes and hearts: inhibition of Na(þ)/H(þ) diabetes mellitus. Cell Metab 2019;30:609–13.
energetics. J Am Coll Cardiol 2019;73:1931–44.
exchanger, lowering of cytosolic Na(þ) and vaso- 75. Adingupu DD, Gopel SO, Gronros J, et al.
47. Lehrke M. SGLT2 inhibition: Changing what
dilation. Diabetologia 2018;61:722–6. SGLT2 inhibition with empagliflozin improves
fuels the heart. J Am Coll Cardiol 2019;73:1945–7.
coronary microvascular function and cardiac
61. Uthman L, Nederlof R, Eerbeek O, et al.
48. Verma S, Rawat S, Ho KL, et al. Empagliflozin contractility in prediabetic ob/ob(-/-) mice. Car-
Delayed ischemic contracture onset by Empagli-
increases cardiac energy production in diabetes: diovasc Diabetol 2019;18:16.
flozin associates with NHE-1 inhibition and is
Novel translational insights into the heart failure
dependent on insulin in isolated mouse hearts. 76. Stahl EP, Dhindsa DS, Lee SK, et al. Nonalco-
benefits of SGLT2 inhibitors. J Am Coll Cardiol
Cardiovasc Res 2019;115:1533–45. holic fatty liver disease and the heart: JACC State-
Basic Transl Sci 2018;3:575–87.
of-the-Art Review. J Am Coll Cardiol 2019;73:
62. Packer M. Reconceptualization of the molec-
49. Abdurrachim D, Teo XQ, Woo CC, et al. 948–63.
ular mechanism by which sodium-glucose
Empagliflozin reduces myocardial ketone utiliza-
cotransporter 2 inhibitors reduce the risk of heart 77. Younossi ZM, Koenig AB, Abdelatif D, et al.
tion while preserving glucose utilization in diabetic
failure events. Circulation 2019;140:443–5. Global epidemiology of nonalcoholic fatty liver
hypertensive heart disease: A hyperpolarized (13)
disease-Meta-analytic assessment of prevalence,
C magnetic resonance spectroscopy study. Dia- 63. Connelly KA, Zhang Y, Visram A, et al. Empa-
incidence, and outcomes. Hepatology 2016;64:
betes Obes Metab 2019;21:357–65. gliflozin improves diastolic function in a nondia-
73–84.
betic rodent model of heart failure with preserved
50. Nakamura M, Sadoshima J. Ketone body can
ejection fraction. J Am Coll Cardiol Basic Transl Sci 78. Omori K, Nakamura A, Miyoshi H, et al. Effects
be a fuel substrate for failing heart. Cardiovasc Res
2019;4:27–37. of dapagliflozin and/or insulin glargine on beta
2019;115:1567–9.
cell mass and hepatic steatosis in db/db mice.
51. Lim VG, Bell RM, Arjun S, et al. SGLT2 Inhibitor, 64. Zhang N, Feng B, Ma X, Sun K, Xu G, Zhou Y. Metabolism 2019;98:27–36.
canagliflozin, attenuates myocardial infarction in Dapagliflozin improves left ventricular remodeling
and aorta sympathetic tone in a pig model of heart 79. Latva-Rasku A, Honka MJ, Kullberg J, et al.
the diabetic and nondiabetic heart. J Am Coll
failure with preserved ejection fraction. Cardiovasc The SGLT2 inhibitor dapagliflozin reduces liver fat
Cardiol Basic Transl Sci 2019;4:15–26.
Diabetol 2019;18:107. but does not affect tissue insulin sensitivity: a
52. Maruyama T, Takashima H, Oguma H, et al. randomized, double-blind, placebo controlled
Canagliflozin improves erythropoiesis in diabetes 65. Solini A, Seghieri M, Giannini L, et al. The ef- study with 8-week treatment in type 2 diabetes
patients with anemia of chronic kidney disease. fects of dapagliflozin on systemic and renal patients. Diabetes Care 2019;42:931–7.
Diabetes Technol Ther 2019;21:713–20. vascular function display an epigenetic signature.
80. Basu D, Huggins LA, Scerbo D, et al. Mecha-
J Clin Endocrinol Metab 2019;104:4253–63.
53. Toto RD, Goldenberg R, Chertow GM, et al. nism of increased LDL (low-Density lipoprotein)
Correction of hypomagnesemia by dapagliflozin in 66. Pabel S, Wagner S, Bollenberg H, et al. and decreased triglycerides with SGLT2 (sodium-
patients with type 2 diabetes: a post hoc analysis Empagliflozin directly improves diastolic function glucose cotransporter 2) inhibition. Arterioscler
of 10 randomized, placebo-controlled trials. in human heart failure. Eur J Heart Fail 2018;20: Thromb Vasc Biol 2018;38:2207–16.
J Diabetes Complications 2019:107402. 1690–700.
81. Chagnac A, Zingerman B, Rozen-Zvi B, Her-
54. Montaigne D, Marechal X, Coisne A, et al. 67. Matsutani D, Sakamoto M, Kayama Y, et al. man-Edelstein M. Consequences of glomerular
Myocardial contractile dysfunction is associated Effect of canagliflozin on left ventricular diastolic hyperfiltration: the role of physical forces in the
434 Zelniker and Braunwald JACC VOL. 75, NO. 4, 2020

Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors FEBRUARY 4, 2020:422–34

pathogenesis of chronic kidney disease in diabetes 90. Sano M, Goto S. Possible mechanism of he- 99. Heerspink HJL, Perco P, Mulder S, et al.
and obesity. Nephron 2019;143:38–42. matocrit elevation by sodium glucose cotrans- Canagliflozin reduces inflammation and
porter 2 inhibitors and associated beneficial renal fibrosis biomarkers: a potential mechanism of
82. Lowen J, Grone E, Grone HJ, Kriz W. Hernia-
and cardiovascular effects. Circulation 2019;139: action for beneficial effects of SGLT2 in-
tion of the tuft with outgrowth of vessels through
1985–7. hibitors in diabetic kidney disease. Dia-
the glomerular entrance in diabetic nephropathy
betologia 2019;62:1154–66.
damages the juxta-glomerular- apparatus. Am J 91. Novikov A, Fu Y, Huang W, et al. SGLT2 inhi-
100. Lee YH, Kim SH, Kang JM, et al. Empagli-
Physiol Renal Physiol 2019;317:F399–410. bition and renal urate excretion: role of luminal
glucose, GLUT9, and URAT1. Am J Physiol Renal flozin attenuates diabetic tubulopathy by
83. Heerspink HJ, Perkins BA, Fitchett DH, improving mitochondrial fragmentation and
Physiol 2019;316:F173–85.
Husain M, Cherney DZ. Sodium glucose cotrans- autophagy. Am J Physiol Renal Physiol 2019;317:
porter 2 inhibitors in the treatment of diabetes 92. Wilcox CS, Shen W, Boulton DW, Leslie BR, F767–80.
mellitus: cardiovascular and kidney effects, po- Griffen SC. Interaction between the sodium-
101. Mulder S, Heerspink HJL, Darshi M, et al. Ef-
tential mechanisms, and clinical applications. Cir- glucose-linked transporter 2 inhibitor dapagli-
fects of dapagliflozin on urinary metabolites in
culation 2016;134:752–72. flozin and the loop diuretic bumetanide in normal
patients with type 2 diabetes. Diabetes Obes
human subjects. J Am Heart Assoc 2018;7: Metab 2019;21:2422–8.
84. Kidokoro K, Cherney DZI, Bozovic A, et al.
e007046.
Evaluation of glomerular hemodynamic function 102. Shibusawa R, Yamada E, Okada S, et al.
by empagliflozin in diabetic mice using in vivo 93. Lytvyn Y, Perkins BA, Cherney DZ. Uric acid as Dapagliflozin rescues endoplasmic reticulum
imaging. Circulation 2019;140:303–15. a biomarker and a therapeutic target in diabetes. stress-mediated cell death. Sci Rep 2019;9:9887.
Can J Diabetes 2015;39:239–46.
85. Wanner C. Sodium glucose cotransporter 2 103. Cooper ME, Inzucchi SE, Zinman B, et al.
inhibition and the visualization of kidney hemo- 94. Alicic RZ, Rooney MT, Tuttle KR. Diabetic Glucose control and the effect of empagliflozin on
dynamics. Circulation 2019;140:316–8. kidney disease: Challenges, progress, and possi- kidney outcomes in type 2 diabetes: An analysis
bilities. Clin J Am Soc Nephrol 2017;12:2032–45. from the EMPA-REG OUTCOME Trial. Am J Kidney
86. Mayer GJ, Wanner C, Weir MR, et al. Analysis from
Dis 2019;74:713–5.
the EMPA-REG OUTCOME((R)) trial indicates empa- 95. Woods TC, Satou R, Miyata K, et al. Canagli-
104. Heerspink HJ, Desai M, Jardine M, et al.
gliflozin may assist in preventing the progression of flozin prevents intrarenal angiotensinogen
Canagliflozin slows progression of renal func-
chronic kidney disease in patients with type 2 diabetes augmentation and mitigates kidney injury and
tion decline independently of glycemic effects.
irrespective of medications that alter intrarenal he- hypertension in mouse model of type 2 diabetes
J Am Soc Nephrol 2017;28:368–75.
modynamics. Kidney Int 2019;96:489–504. mellitus. Am J Nephrol 2019;49:331–42.
105. Evans M, Morgan AR, Yousef Z. What next
87. Wanner C, Heerspink HJL, Zinman B, et al. 96. Yaribeygi H, Atkin SL, Butler AE, Sahebkar A. after metformin? Thinking beyond glycaemia: are
Empagliflozin and kidney function decline in pa- Sodium-glucose cotransporter inhibitors and SGLT2 inhibitors the answer? Diabetes Ther 2019;
tients with type 2 diabetes: a slope analysis from oxidative stress: an update. J Cell Physiol 2019; 10:1719–31.
the EMPA-REG OUTCOME trial. J Am Soc Nephrol 234:3231–7.
106. Vergara A, Jacobs-Cacha C, Soler MJ. So-
2018;29:2755–69.
97. Birnbaum Y, Bajaj M, Yang HC, Ye Y. Combined dium-glucose cotransporter inhibitors: beyond
88. Neuen BL, Ohkuma T, Neal B, et al. Effect of SGLT2 and DPP4 inhibition reduces the activation glycaemic control. Clin Kidney J 2019;12:322–5.
canagliflozin on renal and cardiovascular out- of the Nlrp3/ASC inflammasome and attenuates
comes across different levels of albuminuria: data the development of diabetic nephropathy in mice
from the CANVAS program. J Am Soc Nephrol with type 2 diabetes. Cardiovasc Drugs Ther 2018;
KEY WORDS diabetes, heart failure, renal
2019;30:2229–42. 32:135–45.
function, SGLT2 inhibitor
89. Inzucchi SE, Zinman B, Fitchett D, et al. How 98. Masters SL, Dunne A, Subramanian SL, et al.
does empagliflozin reduce cardiovascular mortal- Activation of the NLRP3 inflammasome by islet
ity? Insights from a mediation analysis of the amyloid polypeptide provides a mechanism for
EMPA-REG OUTCOME trial. Diabetes Care 2018; enhanced IL-1beta in type 2 diabetes. Nat Immu- A PPE NDI X For an addendum, please see the
41:356–63. nol 2010;11:897–904. online version of this paper.

You might also like