Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Drugs

https://doi.org/10.1007/s40265-021-01573-3

REVIEW ARTICLE

A Role for SGLT‑2 Inhibitors in Treating Non‑diabetic Chronic Kidney


Disease
Lucia Del Vecchio1   · Angelo Beretta2 · Carlo Jovane1 · Silvia Peiti1,3 · Simonetta Genovesi3,4

Accepted: 12 July 2021


© The Author(s), under exclusive licence to Springer Nature Switzerland AG 2021

Abstract
In recent years, inhibitors of the sodium-glucose co-transporter 2 (SGLT2 inhibitors) have been shown to have significant
protective effects on the kidney and the cardiovascular system in patients with diabetes. This effect is also manifested in
chronic kidney disease (CKD) patients and is minimally due to improved glycaemic control. Starting from these positive
findings, SGLT2 inhibitors have also been tested in patients with non-diabetic CKD or heart failure with reduced ejection
fraction. Recently, the DAPA-CKD trial showed a significantly lower risk of CKD progression or death from renal or cardio-
vascular causes in a mixed population of patients with diabetic and non-diabetic CKD receiving dapagliflozin in comparison
with placebo. In patients with heart failure and reduced ejection fraction, two trials (EMPEROR-Reduced and DAPA-HF)
also found a significantly lower risk of reaching the secondary renal endpoint in those treated with an SGLT2 inhibitor in
comparison with placebo. This also applied to patients with CKD. Apart from their direct mechanism of action, SGLT2
inhibitors have additional effects that could be of particular interest for patients with non-diabetic CKD. Among these, SGLT2
inhibitors reduce blood pressure and serum acid uric levels and can increase hemoglobin levels. Some safety issues should
be further explored in the CKD population. SGLT2 inhibitors can minimally increase potassium levels, but this has not been
shown by the CREDENCE trial. They also increase magnesium and phosphate reabsorption. These effects could become
more significant in patients with advanced CKD and will need monitoring when these agents are used more extensively in
the CKD population. Conversely, they do not seem to increase the risk of acute kidney injury.

1 Introduction CKD is often a smouldering and insidious disease that


remains asymptomatic in the majority of cases until it
Chronic kidney disease (CKD) affects 697.5 million people is advanced; for this reason, it frequently remains unde-
globally [1]. According to data from the National Health and tected and overlooked. Consequently, late referral and
Nutrition Examination Survey (NHANES), the age-adjusted inadequate diagnosis and treatment are missed opportu-
prevalence of CKD ranges between 13 and 14% of the US nities for proper management of CKD and for delaying its
adult population, with minimal variation over the years [2]. progression towards kidney failure. Indeed, people with
As one would expect, CKD prevalence increases with age CKD have a much higher risk of death rather than kidney
and is slightly more common in women than men. African failure since they pay a severe toll in terms of increased
Americans and low-income and low-education people are cardiovascular (CV) disease [4]. Moreover, in comparison
the most affected groups. Among these patients, 2.5 million to the general population of similar age, CKD patients
people need renal replacement therapy (RRT) worldwide [3]. progressively experience a decrease in their wellbeing and
quality of life.
Despite different aetiologies, it is believed that CKD has
* Lucia Del Vecchio
common mechanisms that perpetuate and self-sustain the
lucia.delvecchio@asst-lariana.it
initial damage [5]. Indeed, microvasculature dysfunction
1
Department of Nephrology and Dialysis, Sant’Anna and glomerular hyperfiltration activate disruptive pathways
Hospital, ASST Lariana, Como, Italy inside the kidney, promoting mesangial activation, glomeru-
2
Internal Medicine Unit, Valduce Hospital, 22100 Como, Italy losclerosis, podocyte dysfunction, inflammation, hypoxia,
3
School of Medicine and Surgery, Nephrology Clinic, fibrosis, and tubular damage.
University of Milano, Bicocca, 20100 Milan, Italy Given these shared mechanisms of progression despite
4
Istituto Auxologico Italiano, IRCCS, 20100 Milan, Italy different underlying diseases, several nephroprotective

Vol.:(0123456789)
L. Del Vecchio et al.

Based on its findings, strict blood pressure control was sug-


Key Points  gested in CKD patients, especially in those with proteinuria.
However, the evidence supporting these recommendations
SGLT2 inhibitors act at the proximal tubule where they is weak and the enrolled population of the MDRD (Cau-
inhibit glucose and sodium reabsorption. The increased casian in the majority of cases with a mean age around 50
sodium output to the macula densa induces vasoconstric- years) possibly shows little representation of the average
tion of the afferent arteriole and reduces hyperfiltration. CKD patient at the current time. Subsequent trials did not
Several other nephroprotective mechanisms have also show any significant advantage on hard renal endpoints
been identified. associated with a more intensive blood-pressure control
Starting from the observed benefits on the kidney and [7–9]. More recently, the Systolic Blood Pressure Interven-
cardiovascular system in patients with diabetes, SGLT2 tion Trial (SPRINT) did not show a significant advantage on
inhibitors have also been proposed as a possible treat- renal outcomes (possibly because of insufficient statistical
ment for patients with non-diabetic chronic kidney dis- power in this respect) and possibly a higher incidence of
ease (CKD) or with heart failure with reduced ejection acute kidney insufficiency [10], despite a clear benefit on
fraction. the risk of reaching the primary composite cardiovascular
endpoint [11].
Recent large trials have shown the efficacy of SGLT2 Theoretically, any antihypertensive therapy is capa-
inhibitors on renal and cardiovascular endpoints in these ble of decreasing proteinuria in hypertensive patients.
two populations. However, RAS inhibitors also have hemodynamic and
Apart from direct benefits on the kidney and the heart, non-hemodynamic actions that confer additional nephro-
several additional benefits could be of help in non-dia- protective properties. Accordingly, they have become the
betic CKD, such as a decrease in blood pressure values first-choice therapy of hypertension in diabetic and non-
and of uric acid, improvement of anemia, and possibly diabetic nephropathies. Moreover, some metanalyses sup-
protection from acute kidney injury. port their use [12, 13]. However, single trials, especially
those performed in the 1990s, have somehow been critically
In people with advanced CKD, monitoring of potassium, reviewed, as the enrolled patient populations were not repre-
magnesium, and phosphate levels is warranted. sentative of today’s CKD patients, who are old, obese, with
low proteinuria levels, and with a high burden of comorbidi-
ties in many cases. This aspect is of importance, considering
that nephroprotection with RAS inhibitors is more evident in
the presence of significant proteinuria levels [14, 15].
agents have been tested and introduced into clinical prac-
Other critical points have also risen over the years. For
tice over the years.
instance, the two hallmark studies of angiotensin-II receptor
blockers (ARBs) in type 2 diabetes (T2D) gave consistent
1.1 State of the Art and Unmet Needs: RAS
findings, but the magnitude of nephroprotection was mod-
Blockade, Blood Pressure Reduction, and Low
est and the number of patients needed to be treated to avoid
Sodium Intake
one outcome was high. Moreover, as shown in non-diabetic
nephropathies, treatment with losartan did not significantly
Inhibitors of the renin-angiotensin system (RAS), blood
reduce the risk of a renal event when baseline albuminuria
pressure control, and reduced sodium intake are considered
was below 1.5 g/g [16].
the gold standard of nephroprotection.
Nearly 2 decades ago, dual blockade was proposed to
Starting from the observation of a clear relationship
increase RAS inhibition and counterbalance the effects of
between blood pressure values and CKD progression and
the single molecules. Several data showed a greater anti-
that higher blood pressure values increase proteinuria, in the
proteinuric effect of dual blockade in comparison to the sin-
last 30 years it has been widely accepted that lower blood
gle agents. However, large trials did not show a significant
pressure values could slow down CKD progression, espe-
beneficial effect on the risk of hard endpoints but increased
cially in proteinuric nephropathies. Indeed, proteinuria can
the risk of hyperkalaemia and acute kidney injury (AKI)
be worsened by the transmission of high systemic pressure to
[17–20]. Accordingly, the European Medicine Agency
the glomeruli. In addition to this, the treatment of hyperten-
(EMA) gave restrictions on dual blockade in patients with
sion can contribute to reducing the burden of cardiovascular
diabetic nephropathy, discouraging the use of an angiotensin
disease. Over the years, several studies compared different
converting enzyme (ACE) inhibitor with an ARB and strictly
blood pressure targets in CKD. The landmark trial is the
contraindicating the combination of the direct renin inhibitor
Modification of Diet in Renal Disease (MDRD) Study [6].
aliskiren with an ACE inhibitor or an ARB [21].
SGLT-2 Inhibitors in Non-diabetic CKD

The antiproteinuric effect of RAS inhibitors can be blood circulation through the action of GLUT2 and GLUT1
enhanced by reduced sodium intake [22]. Unfortunately, [35].
compliance with a low-salt diet is often suboptimal in eve- SGLT2 is mainly distributed in the upper part of the
ryday clinical practice. proximal tubule (S1 and S2 segments), whereas SGLT1
Finally, RAS inhibitors are often under-prescribed or dis- is distributed in the lower part, the S3 segment in humans
continued in everyday clinical practice [23] because of the [36–38]. In euglycemia, SGLT2 is mainly responsible for the
fear or occurrence of AKI and/or hyperkalaemia, especially reabsorption of filtered glucose: it accounts for more than
in old and frail patients. 80% of the glucose reabsorption; SGLT1 takes the remain-
ing glucose [39, 40]. When more glucose is delivered into
the distal proximal tubule, due to SGLT2 inhibition, SGLT1
2 The Physiology of Sodium and Glucose can compensate for the reabsorption of glucose. Euglycemic
Transport and Mechanism of Action humans treated with SGLT2 inhibitors (SGLT2is) maintain
of SGLT2 Inhibitors fractional glucose reabsorption of 40–50% [40, 41]. In the
presence of sustained hyperglycemia, the glucose flow to the
Glucose homeostasis is highly regulated, and the kidney distal proximal tubule is increased, and SLGT1 also medi-
plays a crucial role in these processes. Glucose reabsorp- ates glucose reabsorption.
tion from the proximal tubule is one of these tasks. The In this condition, SGLT2 activity leads to a decrease in
glomerulus filters ~ 162 g of glucose/day, and normally less NaCl delivery to the macula densa, sensed by the juxta-glo-
than 1% is finally excreted in the urine. merular apparatus as a reduction in the effective circulating
In 1960 at the Symposium on Membrane Transport and volume. This event induces a vasodilation of the afferent
Metabolism in Prague, Dr B. Crane first proposed “the glomerular arteriole with consequent hyperfiltration and
Na+/Glucose transport hypothesis”, according to which the intra-glomerular hypertension, a phenomenon called tubulo-
energy for active glucose transport is provided by the sodium glomerular feedback [42]. SGLT2is reverse this process and
gradient across the cell membrane. increase NaCl delivery to the macula densa, inducing vaso-
This reabsorptive capacity depends on the presence of constriction of the afferent arteriole via the paracrine release
two different classes of glucose transporters: the SLC5 of adenosine [43]. This phenomenon has been described in a
solute carriers, also called N ­ a+-glucose co-transporters series of experimental models of diabetes mellitus [44–46]
(SGLTs), and the SLC2A transporters (also called GLUTs). and also in young adults with type 1 diabetes (T1D) [47].
GLUTs are expressed on the basolateral membrane in the However, vasoconstriction of the afferent arteriole seems not
renal proximal tubule and they cause glucose to go back to be the only mechanism through which SGLT2is exert their
to the interstitial space by facilitated diffusion [24]. The nephroprotective effect. Recently, an experimental study
glucose gradient across the membrane is the driving force, showed by direct measurement that the tubule-glomerular
whereas for SGLTs the transmembrane sodium gradient is feedback response to SGLT2 blockade involves both preglo-
the driving force for glucose uptake [25, 26]. merular vasoconstriction and post-glomerular vasorelaxa-
In humans at least six different SGLT isoforms have been tion [48]. Moreover, in studies performed on animals with
identified: SGLT1 and SGLT2 are mainly expressed in intes- experimental diabetes, the suppression of SGLT2 activity
tinal and renal cells [25, 27]. reduces the production of glomerular markers of inflam-
SGLT1 is a high-affinity transporter for glucose (Michae- mation and fibrosis and suppresses oxidative stress in the
lis-Menten constant [Km] = 0.4 mmol/L) and galactose [28, diabetic mouse kidney [49, 50]. It has also been suggested
29]. Two sodium ions are transported through the SGLT1 that in T2D patients, the nephroprotective mechanism of
for each glucose molecule. SGLT2 Km values for glucose SGLT2is is not so much due to the rebalancing of tubulo-
and sodium are 2 and 25 mmol/L, respectively. SGLT2 is a glomerular feedback, but to the establishment of post-glo-
low-affinity and high-capacity glucose transporter. It is pre- merular vasodilation [51]. The evidence that SGLT2is exert
dominantly expressed in the kidneys of rodents and humans a nephroprotective effect even in non-diabetic CKD con-
[30, 31]. firms that the mechanisms by which these drugs protect renal
In the small intestine, SGLT1 is responsible for the function go beyond vasoconstriction of the afferent arteriole,
absorption of glucose [31–33], which enters into the epi- described in the presence of glomerular hyperfiltration, but
thelial cells. Thereafter it flows into the circulation through absent in advanced diabetic nephropathy or in non-diabetic
GLUT2 [34]. forms of CKD. Of note, improvement of cardiac function by
In the kidney, SGLT2 and SGLT1 are responsible for glu- SGLT2is may also have a favorable effect on the kidneys.
cose entry into the tubular cells across the apical membrane Moreover, the potential nephroprotective effect of SGLT2is
of the proximal convoluted tubule. Glucose then reaches the can be also related to their effects on multiple risk factors
L. Del Vecchio et al.

of renal impairment such as high blood glucose (for T2D), infarction (MI). Empagliflozin [52] and canagliflozin [53]
high blood pressure, high serum uric acid, and body weight. did not significantly reduce MI risk when given to T2D
patients (HR: 0.87; 95% CI 0.70–1.09; P = 0.23 and HR:
0.85; 95% CI 0.69–1.05; P > 0.05, respectively). Data from
3 SGLT2 Inhibitors in Type 2 metanalyses are conflicting [59, 60], with only some show-
Diabetes: Mechanism of Actions ing a benefit in this respect [61, 62].
on the Cardiovascular System This heterogeneity across the class for selected outcomes,
and Available Evidence specifically for MACE and CV death, requires further explo-
ration; it remains to be explained whether this heterogene-
SGLT2is were developed to lower blood glucose levels in ity is due to differences in characteristics of patients and in
T2D patients. Several trials designed to evaluate SGLT2i their risk profiles or differences in enrolment, definition of
cardiovascular (CV) safety demonstrated that they had addi- outcomes, or among the drugs themselves.
tional benefits for the CV system [52–55]. This finding was Notably, the main CV benefit of SGLT2is is a reduction
confirmed by several analyses, albeit with a certain degree in hospitalization risk for heart failure, with only moder-
of heterogeneity between the different molecules in the class. ate heterogeneity between drugs in the class, with no dif-
The trials completed to date in T2D patients have predom- ferences in baseline ASCVD, previous history of heart
inantly focused on atherosclerotic CV disease (ASCVD)- failure, or baseline estimated (e) GFR of treated patients.
related outcomes. In this respect, a rather modest effect was In the EMPA-REG OUTCOME, CANVAS Program, and
found in risk reduction for MACE (major adverse cardiovas- DECLARE-TIMI 58 trials, the three SGLT2is significantly
cular events: myocardial infarction, stroke, or CV death). In reduced the risk of hospitalization for heart failure in T2D
particular, the Empagliflozin Cardiovascular Outcome Event patients [52, 53] with consistency of effectiveness across
Trial in Type 2 Diabetes Mellitus Patients-Removing Excess the trials, and similarly in those with or without prevalent
Glucose (EMPA–REG OUTCOME) demonstrated a 14% ASCVD. This was confirmed by the metanalysis of Zelniker
relative risk reduction in three-point MACE in association et al. [57], which demonstrated a 23% reduction in the risk of
with empagliflozin use (P = 0.04 for superiority) [51]. This CV death or hospitalization for heart failure (HR: 0.77; 95%
difference was largely driven by significantly lower rates CI 0.71–0.84, P < 0.0001) in patients with and without CV
of CV death in the empagliflozin group. The CANagliflo- disease and in patients with and without previous heart fail-
zin cardioVascular Assessment Study (CANVAS) Program ure. Other meta-analyses that excluded the CANVAS pro-
included data of 10,142 T2D patients at high risk for CV gram and EMPA–REG OUTCOME trials did not show any
events from two trials (CANVAS and CANVAS-R). In the decrease in hospitalization for heart failure [60, 61, 63, 64].
canagliflozin group the rate of triple MACE was signifi- Recently, data from the SCORED study were published
cantly lower than in the placebo group (hazard ratio (HR): [65]. This was a large, double-blind, placebo-controlled trial
0.86; 95% confidence interval (CI) 0.75–0.97; P = 0.02 for that tested the efficacy of sotagliflozin in more than 10,000
superiority) with regard to both primary and secondary T2D patients with CKD (eGFR of 25–60 mL/min/1.73 m ­ 2)
prevention [53]. Conversely, in the Dapagliflozin Effect on and risks for CV disease. Unfortunately, the trial ended
Cardiovascular Events—Thrombolysis in Myocardial Infarc- early because of lack of funding, thus reducing its statistical
tion 58 (DECLARE-TIMI 58) study, dapagliflozin failed power. In any case, the drug was found to be significantly
to demonstrate significant benefit in terms of a reduction effective in reducing the risk of reaching the primary com-
of MACE. As opposed to the trials reported above, in this posite endpoint of deaths from CV causes, hospitalizations
study, the majority of the patients (10,186 out of 17,160) had for heart failure, and urgent visits for heart failure in compar-
no ASCVD [56]. Accordingly, a meta-analysis including the ison to placebo (HR: 0.74; 95% CI 0.63–0.88; P < 0.001).
data from the EMPA-REG OUTCOME, CANVAS Program, It remains unclear exactly how SGLT2is exert these
and DECLARE-TIMI 58 trials, showed an 11% reduction effects in people with heart failure, but several mechanisms
in MACE associated with the use of SGLT2is (HR: 0.89; have been proposed for the CV effects of SGLT2is.
95% CI 0.83–0.96, P = 0.0014) only in patients with previ- These drugs reduce cardiac preload and lung and sys-
ous ACVD (HR: 0.86; 95% CI 0.80–0.93) [57]. A reduction temic congestion by increasing both natriuresis and gly-
in CV death risk was only found in empagliflozin-treated cosuria and causing osmotic diuresis; this reduces cardiac
patients, with moderate heterogeneity across the trials and preload and lung and systemic congestion. During the acute
no interaction of outcome for those with or without ASCVD. phase, increased natriuresis and polyuria possibly activate
Similarly, only empagliflozin was able to reduce the risk of the RAS. However, after nearly 3 months of treatment, com-
all-cause mortality [52, 58]. pensatory mechanisms take place that reduce the magnitude
Heterogeneity between the effect of different drugs of this effect. A steady state is reached in which the body
was also found in the reduction of the risk of myocardial has a slightly lower sodium content and blood volume [66,
SGLT-2 Inhibitors in Non-diabetic CKD

67]. This likely occurs as a consequence of increased food efficacy was confirmed across different eGFR and albumi-
and fluid intake as well as increased free-water reabsorp- nuria values [54].
tion, which is mediated by vasopressin [68]. As opposed
to traditional diuretics, which markedly stimulate the RAS
and sympathetic system as a compensatory mechanism of 4 Experimental Evidence of Possible Use
arterial underfilling, during the chronic phase SGLT2is do of SGLT2 Inhibitors in Non‑diabetic
not increase but rather suppress these systems [66]. This Chronic Kidney Disease (CKD)
is important, since the activation of these two systems
increases systemic vascular resistance and stimulates sodium Familiar renal glycosuria (FRG) is a well-known example of
and water retention, contributing to diuretic resistance. renal glycosuria in the absence of diabetes. This genetic con-
SGLT2is also decrease cardiac afterload by lowering arterial dition is characterized by SLC5A2 gene mutation, respon-
pressure by 3–5 mmHg, without increasing the heart rate. sible for encoding the SGLT2 carrier protein. As a result,
They may also decrease arterial stiffness [56, 69] and the affected subjects have “natural” inhibition of SGLT2 with
sympathetic nervous system hyperactivity related to heart glycosuria exceeding 100 g/day. Except for glycosuria, these
failure [70]. Norepinephrine upregulates the expression of patients are healthy without any other associated diseases
SGLT2, enhancing sodium and glucose reabsorption by the [76]. This suggests that SGLT2 inhibition is not harmful in
proximal tubule; conversely SGLT2is have the opposite euglycemic conditions but does not necessarily indicate any
effect [71]. benefit. Experimental models mimicking FRG have been lit-
tle used till now. Nespoux et al. [77] studied a mouse model
3.1 SGLT2 Inhibitors in Type 2 Diabetes: Renal of acute kidney injury (AKI) induced by bilateral ischemia-
Effects reperfusion (IR) with or without SGLT2 gene deletion. In
both genotypes, IR equally reduced renal mRNA expression
SGLT2is showed significant benefits on renal outcomes in of the ­Na+–K+–2Cl-co-transporter, suggesting comparable
T2D patients in terms of decrease in progression of albumi- thick ascending limb dysfunction, and similarly increased
nuria, doubling of serum creatinine, and need for RRT, with renal mRNA expression of markers of injury, inflammation,
no interaction of outcome through the presence of ASCVD, oxidative stress, and fibrosis.
baseline albuminuria, or history of heart failure [57, 72]. After promising results were obtained in a hyperglycae-
In the EMPAREG-OUTCOME trial, empagliflozin pre- mic setting in non-diabetic animal models, further inves-
vented the eGFR decline usually observed in T2D patients tigations have evaluated the potential role of pharmaco-
and also reduced by 38% the relative risk of progression logical SGLT2 inhibition in euglicemic non-diabetic CKD
to macroalbuminuria [73]. These benefits were observed in (Table 1).
patients with or without CKD [74]. Furthermore, the patients In several animal models and in vitro studies, SGLT2is
treated with empagliflozin had a significantly lower risk of showed significant protective effects on mechanisms leading
either doubling the serum creatinine from baseline or the to inflammation and renal fibrosis. This is possibly due to
need for RRT compared to placebo [74]. direct anti-inflammatory and anti-fibrotic effects of SGLT2is
Similarly, data from the CANVAS program showed that on tubular cells.
canagliflozin significantly reduced the risk of progression Priklbauer et al. [78] were the first to elucidate how empa-
to macroalbuminuria [53] and that of reaching the compos- gliflozin attenuated the expression of basal endothelin 1,
ite renal outcome, including the requirement for RRT (HR: which is involved in the early inflammatory pathogenesis of
0.60; 95% CI 0.47–0.77). CKD, in two independent human proximal tubular cell lines
Likely as a consequence of a hemodynamic effect, an under normoglycemic conditions. Moreover, empagliflozin
initial decline in eGFR occurred in the first 3 months of inhibited the expression of interleukin 1β and the TGF-
treatment with an SGLT2i, and stabilized thereafter [75]. β1-mediated expression of thrombospondin-1, tenascin-C,
Starting from the favorable effects on kidney function and platelet-derived growth factor-beta. Interestingly, the
and reduced progression to macroalbuminuria shown by the observed effects of empagliflozin were specific on tubular
secondary analysis of the large trials mentioned above, the cells, as endothelin-1 expression was not affected by cana-
CREDENCE trial was designed to primarily test the efficacy gliflozin in vascular endothelial cells.
of canagliflozin on renal outcomes in more than 4000 T2D In non-diabetic rats with Ang-II-dependent hyperten-
patients and albuminuric CKD [54]. The trial was stopped sion, empagliflozin exerted an antifibrotic effect with
earlier than planned due to excessive benefit since the rela- reduction of inflammatory infiltrates, regardless of blood
tive risk of the primary composite renal and CV endpoint glucose and blood pressure values [79]. Jaikumkao et al.
and the renal endpoint alone was significantly lower in the [80] examined the effects of SGLT2 inhibition in a predia-
canagliflozin group than in the placebo group. Canagliflozin betic obese rat model and found that dapagliflozin was able
L. Del Vecchio et al.

to reduce hyperfiltration, inflammation, microalbuminuria, of importance. Indeed, if the amelioration of hypoxia occurs
and tubulointerstitial fibrosis. Similar results were obtained at a late stage of CKD, it could be beneficial in reducing
by Yamato et al. [81], who tested the benefits of ipragli- renal damage and fibrosis through increased renal tubular
flozin in a mouse model of adenine-induced CKD. They expression of VEGF and its isoforms [84].
documented that ipragliflozin improved renal function dose- Podocytes can also be targeted by SGLT2 inhibition.
dependently and also decreased interleukin-6 and hematocrit Cassis et al. [85] demonstrated that the SGLT2 protein is
levels. Of note, the decrease in plasma creatinine levels was constitutively expressed in podocytes either in culture or in
independent of glucose plasma concentration and urinary a murine model of bovine serum albumin (BSA)-induced
glucose excretion. In another study, ipragliflozin was found overload proteinuria. In this experimental model of renal
to promote morphological changes in CKD mice by reducing disease, which is characterized by mild glomerulosclerosis,
renal tubular dilatation and fibrosis and by restoring renal interstitial inflammation, and interstitial fibrosis, the expres-
tubular cell mitochondrial abnormalities induced by a high- sion and concentration of SGLT2 on podocytes are enhanced
fat diet [82]. by albumin load in an NF-κ B-dependent manner; treatment
SGLT2is also seem to improve renal oxygenation, sug- with dapagliflozin was capable of reducing proteinuria and
gesting the involvement of the vascular endothelial growth improving podocyte dysfunction and loss. Moreover, in
factor (VEFG) pathway. Accordingly, luseogliflozin was cultured podocytes it limited the cytoskeletal remodelling
found to attenuate renal hypoxia, together with endothelial induced by the albumin load.
rarefaction and interstitial fibrosis, by increasing the expres- SGLT2 is also expressed in mesangial cells and has a
sion of VEGF [83]. Moreover, the improvement in renal oxy- major influence on their contractility according to glucose
genation could also activate/suppress the hypoxia-inducible concentrations [86]. This is of interest, considering that fol-
factor (HIF) system. The activation timing of HIF could be lowing different types of injury, mesangial cells upregulate

Table 1  Experimental studies of SGLT2 inhibitors in non-diabetic CKD


References Experimental model Conclusion

Wakisaka et al. [86] Rat mesangial cells Rat mesangial cells change their contractility according to
the extracellular glucose concentration via SGLT2
Zhang et al. [92] Non-diabetic subtotal nephrectomised rats No renoprotective effects
Ma et al. [93] Non-diabetic mouse model with progressive CKD due to No renoprotective effects
tubule-interstitial disease
Jaikumkao et al. [80] CKD obese rat model (high-fat diet) Dapagliflozin reduced hyperfiltration, inflammation, micro-
albuminuria and tubulointerstitial fibrosis
Cassis et al. [85] In vivo model of protein-overload proteinuria with BSA Dapagliflozin reduced the number of glomerular lesions,
with experimental CKD (nephrectomy) and in vitro improved proteinuria and podocyte dysfunction and loss
model with cultured podocyte.
Yamato et al. [81] Adenine-induced CKD in mice Ipragliflozin dose-dependently improved renal function and
decreased interleukin-6 and hematocrit levels
Zhang et at. [83] Nephrectomised mice with ischemic-reperfusion injury in Luseogliflozin attenuated renal hypoxia, endothelial rarefac-
the remaining kidney tion and interstitial fibrosis by increasing the expression of
VEGF
Mohamed et al. [88] Gentamicin-induced CKD rat model Dapagliflozin improved kidney function, oxidative stress
markers, decrease apoptosis of renal tubular cells by
modulating effects on RNA
Nespoux et al. [77] Kidney injury induced by bilateral ischemia-reperfusion in SGLT2 deletion does not have protective effect
SGLT2 gene deletion or wild-type mice
Castoldi et al. [79] Non-diabetic rats with angiotensin II-dependent hyperten- Empagliflozin had antifibrotic effect with reduction of
sion inflammatory infiltrates, regardless of blood glucose and
blood pressure values
Hasan et al. [89] Mouse model of sympathetic overactivity Canagliflozin reduced renal oxidative stress and inflamma-
tion
Onishi et al. [89] Nondiabetic mice with tubule-specific ­Na+-H+ exchanger 3 Low-dose empagliflozin causes acute small increase in urine
knockdown (NHE3-ko) and wild-type animals pH and bicarbonate excretion but reduced urine pH and
increased bicarbonate absorption in the long term in wild-
type mice but not in NHE3-ko mice

BSA bovine serum albumin, CKD chronic kidney disease, SGLT2 sodium-glucose co-transporter-2
SGLT-2 Inhibitors in Non-diabetic CKD

the expression of α-smooth muscle actin, change their con- 5 Clinical Trials with SGLT2 Inhibitors
tractile abilities, and increase the production of matrix pro- in Non‑diabetic CKD
teins, chemokines, and cytokines [87]. The contractility of
mesangial cells could also regulate glomerular blood flow The experience accumulated so far in patients with dia-
and filtration. It is unknown whether SGLT2 inhibition could betic CKD has shown that the nephroprotective properties
influence mesangial cells in euglycemic conditions. of SGLT2is are minimal due to improved glycemic con-
Amongst pleiotropic effects, SGLT2 inhibition also trol and that, among other effects, SGLT2is decrease the
exerts modulating effects on RNA. In a gentamicin-induced intraglomerular pressure and consequently hyperfiltration.
nephrotoxicity rat model, dapagliflozin showed early pro- Therefore, the idea has taken root that these agents can also
tection against drug-induced renal injury [88]. This was be nephroprotective in non-diabetic CKD. Similarly, many
mediated by a modulating effect on a specific class of non- of the mechanisms leading to reduced cardiovascular risk
coding RNA called microRNAs (miRNA 21 and 181a) and hospitalization for heart failure could also apply to the
known to play a major role in regulation of gene expres- non-diabetic CKD population.
sion and signalling pathways. Accordingly, dapagliflozin Bearing these considerations in mind, the Dapagliflozin
improved kidney function and oxidative stress markers, and Prevention of Adverse Outcomes in Chronic Kidney
decreased apoptosis of renal tubular cells, and increased Disease (DAPA-CKD) trial was designed [94].
miR-21, but decreased the expression of miR-181, with a This was a large, randomized, double-blind, parallel-
restoration of the renal architecture after 14 days of treat- group, placebo-controlled trial aimed at testing the efficacy
ment following injury. of the SGLT2i dapagliflozin on kidney and cardiovascular
Sympathetic nerve activation is among the factors impli- events in CKD patients with and without T2D [95]. Between
cated in CKD progression. Interestingly, canagliflozin February 2017 and July 2018 (for most countries), 4304
reduced renal oxidative stress and inflammation in a mouse subjects with a urinary albumin/creatinine ratio (UACR) ≥
model mimicking overactivity of the sympathetic nervous 200 mg/g and an eGFR between 25 and 75 mL/min/1.73
system [89]. ­m2 were randomized to dapagliflozin 10 mg once daily or
In non-diabetic mice, recent experimental data also placebo. Of these, 2906 (67.5%) had a diagnosis of diabetes
showed a possible contributory role of the N ­ a + –H + at baseline; the others were non-diabetics; the mean eGFR
exchanger 3 (NHE3) in the natriuretic effect of SGLT2is, was 43.1 mL/min/1.73 ­m2 and the median urinary albumin
leading to an acute small increase in urine pH and bicar- to creatinine ratio (UACR) was 949.3 mg/g. The study par-
bonate excretion but to reduced urine pH and increased ticipants were of mixed races, but predominantly Caucasians
bicarbonate absorption in the long term [90]. It is possible (53.2%), followed by Asians (34.1%), and only 4.4% were
that these changes may reflect a modestly increased acid African Americans; more than half of them had a history of
load following metabolic modifications following treatment cardiovascular disease, heart failure, myocardial infarction,
with empagliflozin. Whether increased H ­ + urinary excre- or stroke. All participants were given a maximum tolerated
tion could be an additional nephroprotective mechanism dose of an ACEi or ARB (except 2% who had documented
or possibly be negative needs further clarification [91]. intolerance).
Indeed, the mild worsening of metabolic acidosis could The trial was stopped early because of clear efficacy
somehow enhance the progression of CKD; however, the after a median follow-up of 2.4 years (interquartile range,
functional inhibition of NHE3 by chronic empagliflo- 2.0–2.7). The primary composite outcome of eGFR decrease
zin is probably counterbalanced by other mechanisms of of at least 50%, end-stage kidney disease, or death from
renal bicarbonate reabsorption. Conversely, the increase renal or cardiovascular causes occurred in a significantly
in bicarbonate reabsorption may reduce tubule-interstitial lower number of subjects in the dapagliflozin than the pla-
inflammation. cebo group (197 (9.2%) and 312 (14.5%), respectively; HR
Apart from these positive findings, other studies failed to 0.61; 95% CI 0.51–0.72; P < 0.001). To prevent one primary
provide evidence of reno-protective effects in non-diabetic endpoint, 19 subjects (95% CI 15–27) needed to be treated
CKD. In non-diabetic, subtotal nephrectomised rats, Zhang (NNT) during the trial. This NNT is not dissimilar to that
et al. [92] found that dapagliflozin did not significantly affect calculated in the IDNT and RENAAL trials for ARBs in
the development of hypertension and did not modify the diabetic nephropathy in comparison to placebo (16 and 29,
heavy proteinuria and the declining GFR. Similarly, Ma respectively) [96]; however, in DAPA-CKD it is obtained
et al. [93] documented that SGLT2 inhibition did not attenu- in addition to RAS blockade and not in comparison to sup-
ate the extent of glomerulosclerosis and tubulointerstitial portive therapy alone.
fibrosis in a mouse model of CKD with tubulointerstitial In DAPA-CKD, the effect of dapagliflozin on the primary
injury. endpoint was consistent for both diabetic and non-diabetic
L. Del Vecchio et al.

patients, as testified by the same NNT (19 in both cases, as well before the development of kidney failure. In this per-
in the whole population). It was also similar across severi- spective, DAPA-CKD has provided the first information
ties of CKD and proteinuria levels. Moreover, the patients on possible cardiovascular benefits of SGLT2is in non-
randomized to dapagliflozin had a significantly lower risk of diabetic CKD.
reaching the secondary renal endpoint (eGFR decrease of at As expected, the event rates for the composite outcome of
least 50%, ESKD, or death from renal causes). As expected, cardiovascular death or hospital admission for heart failure
the patients receiving the drug experienced a significant were higher in T2D subjects than in non-T2D subjects. How-
drop in GFR in the first 2 weeks of treatment; thereafter, ever, the benefit of dapagliflozin on this secondary outcome
the annual GFR decline was steeper in the placebo than in was similar, as testified by the non-significant interaction for
the dapagliflozin group (− 3.59 ± 0.11 mL/min/1.73 ­m2 diabetes status. Similarly, all-cause mortality was reduced
and –1.67 ± 0.11 mL/min/1.73 m ­ 2, respectively). These with dapagliflozin in participants with and without T2D.
trends are a further confirmation of the favorable actions Of note, in the overall trial population, the cardiovascular
of SGLT2is on intraglomerular pressure when given in benefits of dapagliflozin were confirmed in both primary and
conjunction with RAS blockers. Unfortunately, no informa- secondary prevention [99]. Moreover, the efficacy on kidney
tion has yet been published on proteinuria changes during endpoints was maintained independently from the presence/
follow-up. absence of cardiovascular disease at baseline.
Among pre-specified analyses, DAPA-CKD investigated Other than DAPA-CKD, only a small, short-term trial
possible differences in study outcomes between diabetic and tested the efficacy of SGLT2is in non-diabetic CKD. The
non-diabetic CKD [97]. The latter subpopulation included Effects of Dapagliflozin in Non-diabetic Patients With Pro-
1794 patients: 695 (38.7%) had chronic glomerulonephritis, teinuria (DIAMOND) was a randomized, double-blind,
687 (38.3%) had ischemic/hypertensive nephropathy, 412 placebo-controlled crossover trial conducted in Canada,
(23%) had other or unknown causes of nephropathy. Note Malaysia, and the Netherlands [100]. Fifty-three subjects
that in this study the recognised cause of nephropathy in dia- with GFR > 25 mL/min/1.73 m ­ 2 and proteinuria between
betic patients was not always the presence of diabetes mel- 500 and 3500 mg/day were randomized to either dapagliflo-
litus [98]. The separate analysis of the effect of dapagliflozin zin 10 mg or placebo over two 6-week periods with a 6-week
on the primary outcome showed consistency among patients washout period in between. All patients were on stable RAS
with diabetic and non-diabetic nephropathies. In particular, inhibitors. The mean change in proteinuria from baseline
the patients with glomerulonephritis who were randomized was similar in the two groups [difference of 0.9% (95% CI
to dapagliflozin had a 57% lower HR (95% CI 0.26–0.71) − 16.6 to 22.1; P = 0.93)]. As expected, patients experi-
of reaching the primary endpoint in comparison to placebo. enced a significant drop in GFR during their dapagliflozin
The effect on the primary outcome favoring dapagliflozin period (− 6.3 mL/min/1.73 ­m2 in in the dapagliflozin arm);
was also consistent for patients with ischemic/hypertensive this reduction in GFR was fully reversed after dapagliflozin
CKD or CKD of other or unknown cause (even if not statis- discontinuation.
tically significant in these two categories). This is in agree- A small pilot study tested the effect of dapagliflozin in ten
ment with the fact that the favorable effect of dapagliflozin patients with focal segmental glomerulosclerosis [101]. Over
was independent of baseline proteinuria levels, as stated the 8 weeks of treatment, no significant changes in renal
above, reinforcing the concept that the favorable effects of hemodynamics or proteinuria were observed. At present, it is
SGLT2is cannot be explained just by modifications of the difficult to reconcile the negative findings of these two small
tubule-glomerular feedback. studies with the significant and consistent nephroprotective
Among the cases of glomerulonephritis, 270 (38.8%) sub- effects shown by DAPA-CKD.
jects had IgA nephropathy [254 (94%) were biopsy-proven]. The use of SGLT2is in non-diabetic CKD is a rapidly
In this subgroup, dapagliflozin reduced the risk of the pri- evolving field; new and strong evidence is awaited in the
mary outcome compared with placebo by 71% (HR: 0.29; coming years. In particular, EMPA-KIDNEY (The Study
95% CI 0.12–0.73) [98]. Dapagliflozin also significantly of Heart and Kidney Protection with Empagliflozin) is a
decreased the urinary albumin-to-creatinine ratio by 26% large, phase III, double-blind, parallel-group clinical trial
in comparison with placebo (− 35.0% in the first 4 months) (NCT03594110) that will test the effect of empagliflozin on
[99]. Considering that this is an exploratory analysis, caution CKD progression or cardiovascular death versus placebo
is needed in interpreting the finding; however, it suggests on top of standard of care in more than 6,000 CKD patients
that in the future dapagliflozin has the potential to become with GFR ≥ 20 mL/min/1.73 ­m2. Subjects with polycystic
one of the most promising treatments of IgA nephropathy. kidney disease, T1D or T2D and prior atherosclerotic car-
As outlined in the introduction, cardiovascular disease diovascular disease with an eGFR > 60 mL/min/1.73 m ­ 2 are
is also a leading cause of morbidity and mortality in the excluded. Active recruitment is still ongoing; study comple-
CKD population in the absence of diabetes; often it occurs tion is expected in October 2022.
SGLT-2 Inhibitors in Non-diabetic CKD

6 Renal Outcomes in Trials of SGLT2 EMPEROR-Reduced and DAPA-HF trials: the difference
Inhibitors in Heart Failure in eGFR slope between the SGLT2i and placebo groups
was 1.73 mL/min/1.73 m ­ 2 for empagliflozin and 1.80 mL/
2
Two randomized controlled trials performed in patients with min/1.73 ­m for dapagliflozin (both P < 0.0001) [105].
heart failure with reduced ejection fraction (EF) also tested Finally, a recent trial, enrolling 1222 diabetic patients
renal outcomes: the EMPagliflozin outcomE tRial in Patients with a recent hospitalization for worsening heart failure ran-
With chrOnic heaRt Failure With Reduced Ejection Fraction domly assigned to take sotagliflozin (a combined SGLT1
(EMPEROR-Reduced) [102] and the Study to Evaluate the and SGLT2 inhibitor) or placebo, showed that the between-
Effect of Dapagliflozin on the Incidence of Worsening Heart group difference in the change in eGFR over time was − 0.16
Failure or Cardiovascular Death in Patients With Chronic mL/min/1.73 ­m2 in favor of the placebo group. The median
Heart Failure With Reduced Ejection Fraction (DAPA-HF) eGFR of the study population was about 50 mL/min/1.73
[103]. ­m2 [106].
The EMPEROR-Reduced study randomized 3,730
patients (49.8% diabetic) with heart failure and an EF of
40% or less to receive empagliflozin or placebo [102]. The 7 Additional Potential Benefits
mean baseline eGFR value was 62 mL/min/1.73 ­m2 and in Non‑diabetic CKD
48.3% of the patients had CKD (eGFR < 60 mL/min/1.73
­m2). Among the endpoints, the trial had a secondary, com- SGLT2is may have benefits including those that one could
posite renal outcome (chronic dialysis or renal transplanta- expect from increased natriuresis, activation of the tubular-
tion or a sustained reduction of eGFR); it occurred in 1.6% glomerular feedback, and vasoconstriction of the afferent
of patients in the empagliflozin group and 3.1% in the pla- glomerular arteriole (Table 2). Some of them could be useful
cebo group (HR 0.50; 95% CI 0.32–0.77). The decrease in in the overall management of CKD.
eGFR per year was − 0.55 versus − 2.28 mL/min/1.73 m ­ 2
in the empagliflozin group compared to the placebo group 7.1 Blood Pressure Decrease
(P < 0.001). Moreover, in 966 patients with paired measure-
ments before the start of the study and 23–45 days after the Hypertension is both a factor in worsening CKD and one
discontinuation of the trial, the eGFR decreased by − 0.93 of its main complications. Among the causes leading to
mL/min/1.73 ­m2 in patients taking empagliflozin and by an increase in blood pressure values, sodium-water reten-
− 4.21 mL/min/1.73 ­m2 in the placebo group. According tion has a strong contributory role. As a result of increased
to a prespecified analysis, the risk of the composite kidney natriuresis and diuresis, it is not then surprising that
outcome was reduced similarly in patients with and with- SGLT2is also have antihypertensive effects. Interestingly,
out CKD, including patients with eGFR as low as 20 mL/ osmotic diuresis occurring in the proximal tubule is rela-
min/1.73 ­m2 [104]. tively mild; conversely, the diuretic action of SGLT2 inhi-
The DAPA-HF study evaluated the effects of dapagliflo- bition is mostly due to the inhibition of reabsorption in the
zin versus placebo in 4744 patients with heart failure and loop of Henle rather than that in the proximal tubule. This
an EF ≤ 40% and elevated plasma N-terminal pro B-type is due to a reduced chloride concentration arriving at the
natriuretic peptide with and without diabetes [103]. Nearly loop of Henle [107]. Apart from this, SGLT2is can decrease
40% of the patients had an eGFR < 60 mL/min/1.73 m ­ 2. A arterial stiffness, reduce sympathetic nervous activity, and
composite renal outcome (the first occurrence of 50% or possibly have a direct effect on endothelial nitric oxide (NO)
greater sustained reduction in eGFR, kidney failure, or death [108].
from kidney-related causes) did not reach statistical signifi- Data coming from the trials performed in T2D patients
cance, neither in patients without nor in those with diabetes clearly showed a decrease in blood pressure values in those
mellitus (HR: 0.67; 95% CI 0.30–1.49 and HR: 0.73; 95% treated with SLGT2is. According to a recent metanalysis,
CI 0.39–1.34, respectively). The incidence of adverse kidney combination therapy with an SGLT2i and ACEI/ARB lead
events did not differ significantly between dapagliflozin and to an additional mean reduction in systolic blood pressure
placebo in either group. (SBP) of − 3.84 mmHg and of − 1.06 mmHg of diastolic
A meta-analysis, performed by pooling data from the blood pressure (DBP) compared with ACEI/ARB alone
two trials, showed that the risk of the composite renal end- [109]. Similar findings were obtained with 24-h ambulatory
point (50% or higher sustained decrease in eGFR, end-stage blood pressure measurements. Recently, a secondary analy-
renal disease, or renal death) was significantly reduced sis of the CREDENCE trial focused on the antihypertensive
in the SGLT2i group (HR: 0.62; 95% CI 0.43–0.90). The efficacy of canagliflozin in people with T2D and CKD [110].
eGFR modifications during follow-up were similar in the In this trial, canagliflozin reduced SBP by 3.50 mmHg.
L. Del Vecchio et al.

Table 2  Potential benefits and risks of SGLT2 inhibitors in non-diabetic chronic kidney disease
Action Positive Possibly negative

Lower HR for ESKD or death from renal causes YES


Lower HR for CV death YES
Improved renal outcomes in HF with reduced EF YES
Blood pressure decrease YES
Hb increase and improved renal ­O2 content YES
Decrease of serum uric acid YES
Acute kidney injury Possibly protective No increased risk
Hyperkalemia Minimal changes observed so far; more experience to be obtained
Hyperphosphatemia and bone effects No significant increase in fracture risk to date; more information
has to be collected on bone markers
Hypermagnesemia Increased magnesium More experience to be obtained, especially in advanced CKD
levels could be protec-
tive
Decreased urinary pH Experimental observation; no clinical data

HR hazard ratio, ESKD end-stage kidney disease, CV cardiovascular, HF heart failure, Hb hemoglobin, EF ejection fraction, CKD chronic kid-
ney disease

The experience accumulated so far in non-diabetic CKD Although it is still a matter of debate whether hyperurice-
is still limited. Indeed, the data published so far of DAPA- mia is a true risk factor or just a biomarker of renal and car-
CKD did not show data of blood pressure trends during fol- diovascular damage, in everyday clinical practice it is now
low-up [95]. Of note, no significant difference was observed treated more aggressively in comparison to the standard of
on the primary outcome in patients with SBP ≤ 130 mmHg care of previous decades.
or > 130 mmHg. Among the effects of SGLT2is, there is an increase in
The antihypertensive effect of SGLT2is is weaker in renal uric excretion and consequently a decrease in serum
patients with heart failure. According to a recent meta-anal- acid uric concentration. This occurs because in the proximal
ysis, the decrease in mean SBP is less than 1 mmHg [111]. tubule glucose competes with urates for the glucose trans-
However, this patient population is often characterized by porter (GLUT) 9 isoform 2 (GLUT9b): in the presence of
the presence of low rather than high blood pressure values. glycosuria more glucose is available for the transporter and
For this reason, blood pressure control is not necessarily less urate is thus reabsorbed [113]. At least in mice, the urate
needed, but rather avoided. transporter URAT1 is also required for the uricosuric effect
Considering that blood pressure control often requires the of SGLT2is [114].
use of several antihypertensive drugs in CKD patients, we Several clinical trials showed a decrease in uric acid lev-
can foresee this new class of drugs as an additional tool for els following therapy with SGLT2is. In particular, a meta-
helping to obtain adequate blood pressure control especially analysis of 62 trials of T2D patients showed a weighted
for patients with salt-sensitive hypertension (as is the case mean difference on uric acid levels of − 37.73 μmol/L (95%
with CKD patients in many instances). CI − 40.51 to − 34.95) between an SGLT2i and control, with
empagliflozin having the highest effect [115]. Apparently,
7.2 Decrease in Uric Acid the decrease in serum uric acid levels did not occur in CKD
patients (eGFR < 60 mL/min/1.73 ­m2).
Hyperuricemia is observed in nearly 25–30% of CKD Interestingly, in a subanalysis of the EMPA-REG OUT-
patients. Once considered as a secondary consequence of COME, empagliflozin improved all cardiorenal outcomes
reduced kidney function, causing gout and nephrolithiasis across tertiles of baseline serum acid uric levels, attenuating
in some cases, it has become more clear over the years that the trend towards a higher HR for those with high acid uric
hyperuricemia may also contribute to CKD progression and/ levels at baseline [116].
or to increased cardiovascular risk [112]. Possible patho- The experience accumulated so far in non-diabetic CKD
genic mechanisms include the worsening of oxidative stress patients is still limited. According to a phase II, cross-over
and inflammation. In addition to this, hyperuricemia is one study of subjects with asymptomatic hyperuricemia, dapa-
of the elements characterizing metabolic syndrome and gliflozin added to verinurad, a selective URAT1 inhibitor,
hyperinsulinemia. Both features are often observed in CKD and febuxostat further increased uric acid excretion and
patients, especially if obese. decreased serum levels [117].
SGLT-2 Inhibitors in Non-diabetic CKD

Increased uricosuria may enhance crystallization in renal [124]. As a result, HIF1-α is suppressed. Conversely, the
tubules and cause nephrolithiasis. However, available evi- HIF2-α subunit, which is the main one involved in stimu-
dence does not currently show any significant concern from lating EPO production, is not only regulated by hypoxia
this point of view [118]. Possibly, the increased urinary out- but also activated by the low carbohydrate content follow-
put would partly prevent nephrolithiasis. ing SGLT2 inhibition [125]. Indeed, SGLT2is are known
to activate sirtuin-1 (SIRT1). This is both a redox rheostat
7.3 Anemia Improvement and a nutrient and/or oxygen sensor; its activation causes
relative changes in HIF-1α/HIF-2α activities (a downreg-
Anemia is a common complication of CKD; it has been ulation of HIF-1α but an upregulation of HIF-2α) [126].
associated with reduced quality of life, increased need for Whether this also applies to non-diabetic CKD is still to
blood transfusions, higher risk of hospitalization, CV events, be determined.
and mortality. Despite the data coming from observational Another possible mechanism by which SGLT2is could
studies, the cause-effect relationship between anemia correc- stimulate erythropoiesis is the suppression of serum hep-
tion and improved outcomes has not been clearly established cidin, as shown by a small, randomized clinical trial of 52
so far. Moreover, after growing evidence showing possible T2D patients [127]. In this patient population, the increase
safety issues when aiming at near-to-normal hemoglobin in hemoglobin levels with therapy with dapagliflozin 10 mg/
levels with erythropoiesis-stimulating agents (ESAs), the day for 12 weeks was mediated by a 24% mean decrease of
hemoglobin target to aim for with these drugs is still a mat- serum hepcidin, together with a significant fall in ferritin
ter of discussion in the nephrology field. concentrations and an increase in erythroferrone levels.
Data from several clinical trials with SGLT2is have Similar data were shown by a substudy of the EMPA-
shown a mild increase in hematocrit/hemoglobin levels fol- HEART (Effects of Empagliflozin on Cardiac Structure
lowing therapy with these drugs [119]. Recently, a post hoc in Patients With Type 2 Diabetes) CardioLink-6 [128]. In
analysis of the CREDENCE trial evaluated the effects of this trial, individuals with T2D and stable coronary artery
canagliflozin in subjects with diabetic CKD [120]. Among disease were randomized to either empagliflozin 10 mg
the overall trial population, one-third of the participants had daily or placebo for 6 months. Hematocrit increased after
anemia at baseline (defined as hemoglobin < 13.0 g/dL in 6 months by 2.34% in those receiving empagliflozin. This
men or < 12.0 g/dL in women). During follow-up, the mean accompanied an increase in EPO levels and a decrease in
hemoglobin concentration was 0.7 g/dL higher in the cana- serum ferritin.
gliflozin group than the placebo group. Moreover, a lower Considering that hepcidin levels are often increased in
percentage of patients receiving canagliflozin had to start CKD, further confirmation is awaited in this setting.
iron or ESA therapy. No data about anemia and its treatment At the moment it is still difficult to foresee whether the
from the DAPA-CKD trial have been published so far. observed increase of hemoglobin levels during therapy with
Several hypotheses have arisen to explain this ancillary SGLT2is is meaningful from the clinical point of view in the
effect. The first and easiest one is hemoconcentration follow- CKD population (whether with or without diabetes).
ing the decrease in plasma volume. However, the increase
in hemoglobin levels is anticipated by reticulocytosis and 7.4 Metabolic Changes
increased erythropoietin (EPO) levels, suggesting increased
erythropoiesis [121]. Under conditions of stress, such as heart failure, glucose
Most of the available experimental data have been utilization is impaired and heart metabolism depends more
obtained in diabetes and are not necessarily applicable to on free fatty acid and ketone oxidation [129]; recently,
non-diabetic patients. Indeed, in diabetic patients, increased exogenous ketone administration has been proposed as a
glucose reabsorption generates a high-glucose environment possible therapeutic strategy for heart failure [130]. It has
in the renal tubule-interstitium, which may impair hypoxia- been suggested that SGLT2is can modify fuel consump-
inducible factor 1 (HIF-1), damage renal erythropoietin- tion by increasing fatty acid oxidation and ketogenesis and
producing cells (REPs), and decrease EPO secretion and concomitantly reducing carbohydrate disposal [71, 131].
erythropoiesis [122]. SGLT2is inhibit glucose reabsorption Indeed, SGLT2is significantly increase ketone body forma-
and may thus attenuate glucotoxicity in the renal tubule- tion in patients with type 2 diabetes [132]. This improved
interstitium, allowing REPs to resume their function and metabolic efficiency could result in less oxygen consumption
increase EPO secretion. In addition to this, SGLT2is have and improved myocardiocyte functioning [130].
been shown to cause decreased macrophage infiltration, oxi- Increased ketone body generation could also contribute
dative stress, and inflammation [123]. On the other hand, by to nephroprotection, as shown in two mouse models of non-
blocking proximal tubular sodium reabsorption, SGLT2is proteinuric and proteinuric diabetic kidney disease [133]. In
reduce oxygen consumption and improve renal hypoxia these animals, ketone bodies prevented decreases in renal
L. Del Vecchio et al.

ATP levels and lessened the podocyte damage mediated by of SGLT2is in patients with heart failure is preserved in the
the hyperactivation of rapamycin complex 1 (mTORC1). subgroup of those taking sacubitril/valsartan. A subanalysis
Moreover, SGLT2i can act directly on the adipose tis- of the DAPA-HF trial was aimed at assessing the efficacy
sue by increasing the percentage of brown adipocytes with and safety of dapagliflozin in the subgroup of patients who
respect to the white ones [134]. This effect is probably were taking sacubitril/valsartan at baseline (a total of 508
mediated by an increase in adiponectine and by polarization patients, 10.7%). Dapagliflozin had similar efficacy (HR:
towards M2 macrophages (with anti-inflammatory proper- 0.75 vs. 0.74 in patients taking and not taking sacubitril/
ties) [134]. valsartan) on the primary endpoint of cardiovascular death
Interestingly, cardiomyocytes express the glucagon recep- or worsening of heart failure. Even safety results were com-
tor, which contributes to cardiac inotropic and chronotropic parable in the two groups [144]. These encouraging findings
effects [135]. SGLT2is increase glucagon levels, possibly suggest that the use of both drugs together could further
due to glucose excretion and a direct effect on pancreatic reduce morbidity and mortality in patients with heart failure
alpha cells. The improvement of arrhythmia and myocardial and reduced EF.
function related to higher levels of glucagon may contribute
to a decrease in heart failure and cardiovascular mortality 8.2 Combination Therapy with Selective
risk [136, 137]. Mineralocorticoid Receptor Antagonists
Of note, modestly elevated circulating levels of the ketone
β-hydroxybutyrate (βOHB) may induce beneficial effects on Increased plasma aldosterone concentrations and mineralo-
the kidney, such as inhibiting inflammation, oxidative stress, corticoid receptor overactivation are known pathogenetic
and fibrosis [138]. factors for cardiovascular disease, heart failure, and kidney
Finally, a growing body of evidence is indicating that damage. Mineralocorticoid receptor antagonists (MRAs)
SGLT2is can also decrease oxidative stress through other have become the standard of care of symptomatic heart fail-
mechanisms, such as a decrease in activity of nicotinamide ure with low EF. In this setting, they also reduce the risk of
adenine dinucleotide phosphate (NADPH) oxidase [139] or cardiovascular events and sudden death. In addition to this,
by reducing the generation of advanced glycation end prod- several studies have underlined the properties of this class of
ucts (AGEs) [140]. drugs as antiproteinuric agents in diabetic and non-diabetic
nephropathies [145]. However, they are often poorly toler-
ated due to their side effects and their use is often limited by
8 SGLT2 Inhibitors in Combination the occurrence of hyperkalemia, especially in CKD patients.
with Other Drugs Selective, non-steroidal MRAs are a new class of drugs;
finerenone is the most used and will be licensed for clinical
8.1 Combination with a Neprilysin Inhibitor use next year in patients with T2D and CKD. As opposed to
non-selective antagonists, it is more selective for the miner-
Sacubitril/valsartan is a drug containing the neprilysin alocorticoid receptor with similar potency to spironolactone.
inhibitor sacubitril and the ANG-II receptor antagonist val- As a result, it has shown remarkable protective effects on
sartan. Neprilysin inhibition enhances the bioavailability of fibrosis and inflammation in animal models [146]. Recently,
natriuretic peptides. The Prospective Comparison of ARNI the Finerenone in Reducing Cardiovascular Mortality and
with ACEI to Determine Impact on Global Mortality and Morbidity in Diabetic Kidney Disease Trial (FIDELIO
Morbidity in Heart Failure (Paradigm-HF) trial compared DKD) showed that treatment with finerenone obtained a
sacubitril/valsartan with enalapril in patients with heart lower risk of reaching the primary renal endpoint (kidney
failure, and reduced EF and demonstrated that sacubitril/ failure, a decrease > 40% in the eGFR from baseline, or
valsartan was superior in reducing the risks of death and of death from renal causes) or cardiovascular events than pla-
hospitalization [141]. A subanalysis of Paradigm-HF showed cebo [147]. Hyperkalemia developed in a small percentage
that, compared with enalapril, sacubitril/valsartan led to a of patients. A second large trial, FIGARO, is still ongoing; it
slower rate of decrease in eGFR and improved cardiovascu- will primarily give information on cardiovascular endpoints
lar outcomes, even in patients with CKD [142]. At the renal in a trial population similar to that of the FIDELIO-DKD
level, sacubitril/valsartan induces preferential vasodilation study and test renal endpoints as secondary ones.
of the afferent arteriole and relative vasoconstriction of the Finerenone could be shown to be nephroprotective in the
efferent arteriole. Its action on intra-renal hemodynamics future in non-diabetic CKD as well.
is therefore different from that hypothesized for SGLT2is, Little information is available at the moment on the effi-
which would induce afferent arteriolar vasoconstriction cacy and safety of the combined use of selective MRAs and
through the tubule-glomerular feedback [143]. It is therefore SGLT2is. However, considering the entrance of finerenone
very interesting to know whether or not the positive effect
SGLT-2 Inhibitors in Non-diabetic CKD

in clinical practice and expected wider use of SGLT2is, it is meta-analysis of 18 trials enrolling more than 150,000 T2D
something that we need to consider. subjects, SGLT2is were associated with a lower risk of AKI
Theoretically, SGLT2is could enhance aldosterone syn- compared to placebo (odds ratio, 0.76; 95% CI 0.66–0.88)
thesis as a consequence of natriuresis and osmotic diuresis. [152]. Similar findings were reported from real-life observa-
However, according to available data, they do not seem to tions [153].
alter plasma aldosterone levels significantly [148]. Apart Patients with CKD could be more susceptible to AKI
from this aspect, the mechanism of action of SGLT2is and development. In this respect, a secondary analysis of the
MRAs displays potential synergistic renoprotective and car- CREDENCE trial did not show an increased risk of AKI at
diovascular properties if given in combination. different levels of baseline GFR [154]. However, for baseline
A secondary analysis of DAPA-HF investigated the effi- GFR between 30 and ≤ 45 mL/min/1.78 ­m2, a higher num-
cacy and safety of dapagliflozin in patients with heart fail- ber of patients receiving canagliflozin were reported as hav-
ure and reduced EF [149]. Of the total 4744 patients, 71% ing volume depletion compared to placebo. Of note, nearly
received an MRA. Dapagliflozin showed similar efficacy on half of the participants had an acute drop in eGFR of > 10%
the primary endpoint compared to placebo in patients either without impairing the clinical benefit of canagliflozin [155].
taking or not taking an MRA; similar findings were observed A more acute drop exceeding 30% occurred infrequently.
for secondary endpoints. In both MRA subgroups, safety Similarly, more patients randomized to dapagliflozin
outcomes were similar in patients randomized to dapagli- developed volume depletion compared to placebo in DAPA-
flozin or placebo. CKD; however, renal-related adverse events (including AKI)
Following a protocol amendment, SGLT2is were allowed occurred with a lower frequency.
as concomitant medications during FIDELIO-DKD and Of note, a case report showed an association between
FIGARO trials. A secondary analysis giving information empagliflozin use and the development of acute interstitial
about the combined use of these two classes of drugs is very nephritis [156].
likely to be performed. Conversely, treatment with MRA
was not admitted in the CREDENCE and DAPA-CKD trials. 9.2 Hyperkalaemia
Recently, a consensus statement by the European Renal
and Cardiovascular Medicine (EURECA-m) working group The kidney has an important role in potassium homeostasis;
of the European Renal Association—European Dialysis and accordingly, hyperkalaemia is one of the most common com-
Transplant Association (ERA-EDTA) and the Hyperten- plications of CKD. Among the contributory factors, reduced
sion and Kidney working group of the European Society of ­K+ urinary excretion, metabolic acidosis, and hyperinsulin-
Hypertension (ESH) recommended the use of finerenone in ism are the most important causes.
addition to an ACEi or an ARB in maximum tolerated doses For a similar degree of kidney function, it is more likely
and independent of the use of an SGLT2i in patients with to develop in diabetic than in non-diabetic CKD patients.
T2D and CKD (eGFR 25–75 mL/min/1.73 m ­ 2), albuminuria, Hyperkalemia is not only associated with an increased
and normal serum potassium [150]. risk of cardiovascular events and mortality, but it often pre-
Of note, a clinical trial has just started aimed at com- vents full use of RAS blockers as kidney- and cardioprotec-
paring the efficacy and safety of a new, selective MRA tive therapies.
(AZD9977, AstraZeneca) alone and in combination with Data coming from clinical trials with SGLT2is and
dapagliflozin in participants with heart failure with low EF everyday use are quite reassuring in this respect, showing
and severe CKD (at least 30% of participants with eGFR minimal increases in potassium levels (in addition to RAS
< 30 mL/min) (NCT04595370). blockers). The observed increase in potassium levels is pos-
sibly explained by small transient changes in GFR or vol-
ume status resulting in less sodium exchanged for potassium
9 Safety Issues of Particular Interest [157]. Reduced glucose levels and improvement of insulin
for Non‑Diabetic CKD resistance could also result in a redistribution of potassium
out of the cells [157]. Conversely, SGLT2 inhibition may
9.1 Acute Kidney Injury determine enhanced aldosterone activity with increased
kaliuresis, possibly counterbalancing the mechanisms men-
Given that SGLT2is cause osmotic diuresis and an acute tioned above.
drop in GFR in the first weeks of treatment, AKI was con- Focusing on CKD, data from the CREDENCE trial
sidered as a possible safety concern at the very beginning. showed that a lower number of patients randomized to
However, expanding experience with SGLT2is not only canagliflozin developed hyperkalemia as an adverse event
did prove this concern [151], but also suggested a possi- in comparison to those on placebo [6.8% vs. 8.2%, respec-
ble protective effect in this respect. According to a recent tively; HR 0.80 (95% CI 0.65–1.00)] [54].
L. Del Vecchio et al.

Unfortunately, available publications of DAPA-CKD do that canagliflozin was associated with normalization of
not give detailed information on serum potassium changes serum magnesium in hypo-magnesemic patients with T2D
during follow-up. [165], who frequently have low magnesium levels due to
renal magnesium wasting.
9.3 Mineral Bone Metabolism A number of mechanisms have been hypothesized to
explain this effect of SGLT2is on plasma magnesium values.
The inhibition of sodium-glucose co-transport increases Among others, the reduced insulin resistance observed with
­Na+ availability in the proximal tubule and, in turn, influ- SGLT2i administration may be associated with a decrease
ences renal phosphate handling by enhancing its reabsorp- in renal magnesium excretion and may induce a redistribu-
tion [158]. Moreover, osmotic diuresis produces a decline tion of magnesium out of the cells to the extracellular space
in medullary osmolarity causing a moderate impairment [166]. It has been hypothesized that this effect may play
of calcium reabsorption in the ascending limbs of Henle’s a role in the beneficial cardiovascular outcomes observed
loop [159]. Accordingly, data on healthy volunteers showed in the SGLT2 trials. However, a recent post hoc analysis
increased levels of serum phosphate, fibroblast growth fac- pooling data from CANVAS and CANVAS-R trials did not
tor 23 (FGF-23), and parathyroid hormone (PTH), as well confirm this hypothesis; the authors concluded that change
as decreased 1,25(OH) vitamin D levels following treatment in serum magnesium was not a mediator of the effects of
with canagliflozin [160]. However, some of these changes canagliflozin on cardiovascular outcomes [167].
could only be transient. Similar changes were reported by a The kidney is crucial in the maintenance of normal mag-
post hoc analysis of the IMPROVE trial in T2D patients and nesium levels; when renal function declines, the ability of
albuminuria [161]. Compared with placebo, dapagliflozin magnesium excretion is then reduced [168]. However, a
significantly increased serum phosphate, PTH, and FGF23, number of compensatory mechanisms are put in place. As
with a non-statistically significant trend towards decreased a consequence, CKD patients often in reality have normal
serum 1,25(OH) vitamin D. Calcium and 25(OH) vitamin serum levels or even hypomagnesemia. How much increased
D were unaffected. Some of the patients considered in the magnesium levels can have a positive effect in CKD patients
study had a certain degree of kidney insufficiency (eGFR is still a controversial topic. Experimental data suggest a
> 45 mL/min/1.73 m ­ 2). Despite this, the overall changes protective effect of magnesium on vascular calcifications
were of a mild degree. Changes in bone biomarkers could [169]. Moreover, two recent metanalyses showed that plasma
be more intense and become of clinical relevance in patients magnesium concentration was inversely associated with all-
with more advanced CKD. No detailed information is avail- cause mortality, cardiovascular mortality, and cardiovascular
able to date for the non-diabetic CKD population at different events in CKD patients [170, 171], indirectly suggesting a
stages of CKD. potential positive effect rather than a safety concern.
Apart from the CANVAS study, data from clinical stud-
ies are quite reassuring with respect to an increased risk of 9.5 Risk of Infectious Complications
bone fractures following SGLT2 inhibition [162]. Moreover,
according to the DAPA-CKD study, there is no difference in A significantly higher risk of genital infections has been
fracture risk among the patients receiving dapagliflozin or described in diabetic patients taking SGLT2is compared
placebo in both the diabetic- and non-diabetic groups [95]. with placebo and other antidiabetic treatments [172].
However, the follow-up period of all these trials could be too Comprehensive data on urinary infections in patients with
short with respect to the time needed to develop clinically non-diabetic glomerular disease treated with SGLT2is are
significant bone fragility leading to fractures. currently lacking. However, considering that patients with
glomerulonephritis are often treated with immunosuppres-
9.4 Hypermagnesemia sive drugs, it is possible that the use of immunosuppressive
therapy in combination with SGLT2is may increase the risk
A meta-analysis including 18 studies demonstrated a signifi- of infectious complications.
cant increase in plasma magnesium levels in T2D patients
taking SGLT2is compared with placebo. This association
has been described for four different molecules (canagliflo- 10 Future Perspectives
zin, dapagliflozin, empagliflozin, and ipragliflozin) in the
absence of CKD [163]. Furthermore, it was found that cana- SGLT2is can provide nephroprotection to non-diabetic CKD
gliflozin induced a higher increase in serum magnesium in thanks to the combined contribution of several mechanisms
patients with eGFR < 60 mL/min/1.73 ­m2 [164]. A post hoc (Fig. 1). In the coming years, SGLT2is are likely to enter on
analysis based on pooled data from four randomized con- a large scale in everyday clinical practice for the treatment
trolled trials comparing canagliflozin with placebo showed of patients with diabetic and non-diabetic CKD. Indeed,
SGLT-2 Inhibitors in Non-diabetic CKD

clinical trials have given remarkable and consistent find- in both diabetic and non-diabetic CKD [95]. The ongoing
ings in terms of reno- and cardiovascular protection. This EMPA-KIDNEY will include patients with eGFR ≥ 20 mL/
efficacy was obtained on top of RAS inhibition and with a min. It is difficult to imagine significant nephroprotection for
good safety profile. Data from the ongoing EMPA-KIDNEY stage V CKD patients. However, it is possible that SGLT2is
are likely to further reinforce the evidence supporting the use also maintain their beneficial effects on the CV system at the
of SGLT2is in non-diabetic CKD. final stages of CKD, providing diuresis conservation.
In addition to this, several treatment indications may open In addition to this, SGLT2i-induced osmotic diuresis can
in the future with SGLT2is. be helpful in conditions of fluid retention (as is often the
It is well known that the antidiabetic efficacy of SGLT2is case in advanced CKD) alone or in combination with other
progressively decreases as CKD worsens. Accordingly, the diuretics. However, the combined use with loop diuretics
initial treatment indication in T2D patients was for eGFR should be done with caution and avoided in subjects with
> 45/mL/min/1.73 ­m2. However, accumulating evidence unstable volume status or hypovolemia. Of note, the effects
has shown that the nephroprotective and CV efficacy is of SGLT2is on kidney and cardiovascular outcomes are quite
also maintained at lower levels of GFR. However, to which consistent in patients receiving or not receiving diuretics
degree of kidney function SGLT2is remain effective is still [174]. In the future, the diuretic properties of SGLT2is could
an open question. Data from the CREDENCE trial showed be used in cases of acute decompensated heart failure, as
consistent efficacy across all levels of kidney function down suggested by preliminary experiences [175] or in patients
to an eGFR of 30 mL/min/1.73 ­m2. A recent subgroup analy- with nephrotic syndrome. Interestingly, the intraperitoneal
sis of this trial of 174 participants who had an eGFR < 30 administration of SGLT2is has been proposed to reduce peri-
mL/min/1.73 ­m2 indicated that the effects of canagliflozin toneal fibrosis and ultrafiltration failure in patients receiving
on kidney, cardiovascular, and mortality outcomes were peritoneal dialysis [176].
consistent with those of patients with higher GFR [173]. The use of SGLT2is could also offer benefits for reduc-
According to its inclusion criteria, DAPA-CKD gave evi- ing/preventing the nephrotoxic effects of contrast media,
dence of efficacy up to GFR values of 25 mL/min/1.73 ­m2

Fig. 1  Possible mechanisms of nephroprotection of SGLT2 inhibi- reports possible pathophysiological mechanisms. IL1β interleukin 1β;
tors in non-diabetic chronic kidney disease. The left panel of the fig- TGF- β1 tumor growth factor β1
ure describes the clinical effects of SGLT2 inhibitors, the right one
L. Del Vecchio et al.

considering their direct anti-inflammatory and antifibrotic intensive versus standard blood-pressure control. N Engl J Med.
nephroprotective effects. 2015;373:2103–16.
12. Xie X, Liu Y, Perkovic V, Li X, Ninomiya T, Hou W, et al. Renin-
Finally, the combined use of SGLT1 and SGLT2 inhibi- angiotensin system inhibitors and kidney and cardiovascular out-
tion could have additional advantages in non-diabetic CKD, comes in patients with CKD: a Bayesian network meta-analysis
given that in this subset there is no significant risk of devel- of randomized clinical trials. Am J Kidney Dis. 2016;67:728–41.
oping ketoacidosis [177]. 13. Zhang Y, He D, Zhang W, Xing Y, Guo Y, Wang F, et al. ACE
inhibitor benefit to kidney and cardiovascular outcomes for
patients with non-dialysis chronic kidney disease stages 3–5:
Declarations  a network meta-analysis of randomised clinical trials. Drugs.
2020;80(8):797–811.
Funding  This article received no funding. 14. Jafar TH, Stark PC, Schmid CH, Landa M, Maschio G, de Jong
PE, AIPRI Study Group, et al. Progression of chronic kidney
disease: the role of blood pressure control, proteinuria, and
Conflicts of interest/Competing interests  Lucia Del Vecchio received angiotensin-convert- ing enzyme inhibition: a patient-level meta-
speaker fees from Mundipharma. Simonetta Genovesi received speak- analysis. Ann Intern Med. 2003;139:244–52.
er fees from Astra-Zeneca. Angelo Beretta, Carlo Jovane and Silvia 15. Casas JP, Chua W, Loukogeorgakis S, Vallance P, Smeeth L,
Peiti have no conflicts of interest to declare. All the Authors contrib- Hingorani AD, et al. effect of inhibitors of the renin-angiotensin
uted to the writing and revision of the manuscript. system and other antihypertensve drugs on renal outcomes: sys-
tematic review and meta-analysis. Lancet. 2005;366:2026–33.
Ethics approval, Consent to participate, Consent for publication, Avail‑ 16. de Zeeuw D, Remuzzi G, Parving HH, Keane WF, Zhang Z, Sha-
ability of data and material, Code availability  Not applicable. hinfar S, et al. Proteinuria, a target for renoprotection in patients
with type 2 diabetic nephropathy: lessons from RENAAL. Kid-
ney Int. 2004;65:2309–20.
17. Yusuf S, Teo KK, Pogue J, Dyal L, Copland I, Schumacher H,
References et al. Telmisartan, ramipril, or both in patients at high risk for
vascular events. N Engl J Med. 2008;358(15):1547–59.
1. GBD Chronic Kidney Disease Collaboration. Global, regional, 18. Parving HH, Brenner BM, McMurray JJ, de Zeeuw D, Haffner
and national burden of chronic kidney disease, 1990–2017: a SM, Solomon SD, ALTITUDE Investigators, et al. Cardiorenal
systematic analysis for the Global Burden of Disease Study 2017. end points in a trial of aliskiren for type 2 diabetes. N Engl J
Lancet. 2020;395(10225):709–33. Med. 2012;367(23):2204–13.
2. Al-Kibria GM, Crispen R. Prevalence and trends of chronic kid- 19. Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA,
ney disease and its risk factors among US adults: an analysis of Duckworth W, VA NEPHRON-D Investigators, et al. Combined
NHANES 2003–18. Rev Med Rep. 2020;20: 101193. angiotensin inhibition for the treatment of diabetic nephropathy.
3. Liyanage T, Ninomiya T, Jha V. Worldwide access to treat- N Engl J Med. 2013;369(20):1892–903.
ment for end-stage kidney disease: a systematic review. Lancet. 20. Feng Y, Huang R, Kavanagh J, Li L, Zeng X, Li Y, Fu P. Effi-
2015;385:1975–82. cacy and safety of dual blockade of analysis. J Cardiovasc Drugs.
4. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic 2019;19(3):259–86.
kidney disease and the risks of death, cardiovascular events, and 21. https://​www.​ema.​europa.​eu/​en/​docum​ents/​refer ​ral/​restr​iction-​
hospitalization. N Engl J Med. 2004;351:1296–305. combi​ned-​use-​medic​ines-​affec​ting-​renin-​angio​tensin-​system-​
5. Webster AC, Nagler EV, Morton RL, Masson P. Chronic kidney ras_​en.​pdf. Accessed 12 June 2021
disease. Lancet. 2017;389(10075):1238–52. 22. D’Elia L, Rossi G, di Cola MS, Savino I, Galletti F, Strazzullo
6. Klahr S, Levey AS, Beck GJ, Caggiula AW, Hunsicker L, Kusek P. Meta-analysis of the effect of dietary sodium restriction with
JW, et al. The effects of dietary protein restriction and blood- or without concomitant renin-angiotensin-aldosterone system-
pressure control on the progression of chronic renal disease. inhibiting treatment on albuminuria. Clin J Am Soc Nephrol.
Modification of diet in renal disease study group. N Engl J Med. 2015;10(9):1542–52.
1994;330:877–84. 23. Qiao Y, Shin JI, Sang Y, Inker LA, Secora A, Luo S, et al. Dis-
7. ACCORD Study Group, Cushman WC, Evans GW, Byington RP, continuation of angiotensin converting enzyme inhibitors and
Goff DC Jr, Grimm RH Jr, Cutler JA, et al. Effects of intensive angiotensin receptor blockers in chronic kidney disease. Mayo
blood-pressure control in type 2 diabetes mellitus. N Engl J Med. Clin Proc. 2019;94(11):2220–9.
2010;362:1575–85. 24. Joost HG, Thorens B. The extended GLUT-family of sugar/polyol
8. Schrier RW, Abebe KZ, Perrone RD, Torres VE, Braun WE, transport facilitators: nomenclature, sequence characteristics, and
Steinman TI, HALT-PKD Trial Investigators, et al. Blood pres- potential function of its novel members (review). Mol Membr
sure in early autosomal dominant polycystic kidney disease. N Biol. 2001;18:247–56.
Engl J Med. 2014;371:2255–66. 25. Wright EM. Renal Na(+)-glucose co-transporters. Am J Physiol
9. Wright JT Jr, Bakris G, Greene T, Agodoa LY, Appel LJ, Renal Physiol. 2001;280:F10–8.
Xharleston J, African American Study of Kidney Disease and 26. Scheepers A, Joost HG, Schurmann A. The glucose transporter
Hypertension Study Group, et al. Effect of blood pressure low- families SGLT and GLUT: molecular basis of normal and aber-
ering and antihypertensive drug class on progression of hyper- rant function. J Parenter Enteral Nutr. 2004;28:364–71.
tensive kidney disease: results from the AASK trial. JAMA. 27. Hirayama BA, Wong HC, Smith CD, Hagenbuch BA, Hediger
2002;288:2421–31. MA, Wright EM. Intestinal and renal Na+/glucose co-transport-
10. Cheung AK, Rahman M, Reboussin DM, Craven TE, Greene T, ers share common structures. Am J Physiol. 1991;261:C296-304.
Kimmel PL, et al. Effects of intensive BP control in CKD. J Am 28. Sabino-Silva R, Mori RC, David-Silva A, Okamoto MM, Frei-
Soc Nephrol. 2017;28:2812–23. tas HS, Machado UF. The Na(+)/glucose co-transporters: from
11. SPRINT Research Group, Wright JT Jr, Williamson JD, Whel- genes to therapy. Braz J Med Biol Res. 2010;43:1019–26.
ton PK, Snyder JK, Sink Km, et  al. A randomized trial of
SGLT-2 Inhibitors in Non-diabetic CKD

29. Santer R, Calado J. Familial renal glucosuria and SGLT2: from 47. Cherney DZI, Perkins BA, Soleymanlou N, Maione M, Lai V,
a mendelian trait to a therapeutic target. Clin J Am Soc Nephrol. Lee A, et al. Renal hemodynamic effect of sodium-glucose co-
2010;5:133–41. transporter 2 inhibition in patients with type 1 diabetes mellitus.
30. Shepard BD, Cheval L, Peterlin Z, Firestein S, Koepsell H, Circulation. 2014;129:587–97.
Doucet A, et al. A renal olfactory receptor aids in kidney glucose 48. Thomson SC, Vallon V. Effects of SGLT2 inhibitor and dietary
handling. Sci Rep. 2016;6:35215. NaCl on glomerular hemodynamics assessed by micropuncture
31. Sopjani M, Bhavsar SK, Fraser S, Kemp BE, Föller M, Lang F. in diabetic rats. Am J Physiol Renal Physiol. 2021;320:F761–71.
Regulation of Na+-coupled glucose carrier SGLT1 by AMP- 49. Osorio H, Coronel I, Arellano A, Pacheco U, Bautista R, Franco
activated protein kinase. Mol Membr Biol. 2010;27:137–44. M, et al. Sodium-glucose co-transporter inhibition prevents oxi-
32. Birnir B, Lee HS, Hediger MA, Wright EM. Expression and dative stress in the kidney of diabetic rats. Oxid Med Cell Lon-
characterization of the intestinal Na+/glucose co-transporter in gev. 2012;2012: 542042.
COS-7 cells. Biochim Biophys Acta. 1990;1048:100–4. 50. Tanaka S, Sugiura Y, Saito H, Sugahara M, Higashijima Y,
33. Tabatabai NM, Sharma M, Blumenthal SS, Petering DH. Yamaguchi J, et al. Sodium–glucose co-transporter 2 inhibition
Enhanced expressions of sodium-glucose co-transporters in normalizes glucose metabolism and suppresses oxidative stress
the kidneys of diabetic Zucker rats. Diabetes Res Clin Pract. in the kidneys of diabetic mice. Kidney Int. 2018;94:912–25.
2009;83:e27-30. 51. van Bommel EJM, Muskiet MHA, van Baar MJB, Tonneijck L,
34. Augustin R. The protein family of glucose transport facili- Smits MM, Emanuel AL, et al. The renal hemodynamic effects
tators: it’s not only about glucose after all. IUBMB Life. of the SGLT2 inhibitor dapagliflozin are caused by post-glomer-
2010;62:315–33. ular vasodilatation rather than pre-glomerular vasoconstriction
35. Chintalapati C, Keller T, Mueller TD, Gorboulev V, Schäfer N, in metformin-treated patients with type 2 diabetes in the rand-
Zilkowski I, et al. Protein RS1 (RSC1A1) downregulates the omized, double-blind RED trial. Kidney Int. 2020;97:202–12.
exocytotic pathway of glucose transporter SGLT1 at low intra- 52. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel
cellular glucose via inhibition of ornithine decarboxylase. Mol S, et al. Empagliflozin, cardiovascular outcomes, and mortality
Pharmacol. 2016;90:508–21. in type 2 diabetes. N Engl J Med. 2015;373:2117–28.
36. Veyhl M, Keller T, Gorboulev V, Vernaleken A, Koep- 53. Neal B, Perkovic V, Matthews DR. Canagliflozin and cardio-
sell H. RS1 (RSC1A1) regulates the exocytotic pathway of vascular and renal events in type 2 diabetes. N Engl J Med.
­Na +- d -glucose co-transporter SGLT1. Am J Physiol Renal 2017;377:2099.
Physiol. 2006;291:F1213–23. 54. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL,
37. Balen D, Ljubojevic M, Breljak D, Brzica H, Zlender V, Koepsell Charytan DM, et al. Canagliflozin and renal outcomes in type 2
H, et al. Revised immunolocalization of the Na+-d-glucose co- diabetes and nephropathy. N Engl J Med. 2019;380:2295–306.
transporter SGLT1 in rat organs with an improved antibody. Am 55. Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S,
J Physiol Cell Physiol. 2008;295:C475–89. Masiukiewicz U, VERTIS CV Investigators, et al. Ertugliflozin
38. Vallon V, Platt KA, Cunard R, Schroth J, Whaley J, Thomson SC, and cardiovascular outcomes in type 2 diabetes. N Engl J Med.
et al. SGLT2 mediates glucose reabsorption in the early proximal 2020;383:1425–35.
tubule. J Am Soc Nephrol. 2011;22:104–12. 56. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn
39. Gorboulev V, Schurmann A, Vallon V, Kipp H, Jaschke A, A, DECLARE–TIMI 58 Investigators, et al. Dapagliflozin and
Klessen D, et al. Na(+)-d-glucose co-transporter SGLT1 is piv- cardiovascular outcomes in type 2 diabetes. N Engl J Med.
otal for intestinal glucose absorption and glucose-dependent 2019;380(4):347–57.
incretin secretion. Diabetes. 2012;61:187–96. 57. Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Furtado
40. Rieg T, Masuda T, Gerasimova M, Mayoux E, Platt K, Pow- RHM, et al. SGLT2 inhibitors for primary and secondary preven-
ell DR, et al. Increase in SGLT1-mediated transport explains tion of cardiovascular and renal outcomes in type 2 diabetes: a
renal glucose reabsorption during genetic and pharmacological systematic review and meta-analysis of cardiovascular outcome
SGLT2 inhibition in euglycemia. Am J Physiol Renal Physiol. trials. Lancet. 2019;393(10166):31–9.
2014;306:F188–93. 58. Tang H, Fang Z, Wang T, Cui W, Zhai S, Song Y. Meta-analysis
41. Abdul-Ghani MA, DeFronzo RA, Norton L. Novel hypothesis of effects of sodium-glucose co-transporter 2 inhibitors on cardi-
to explain why SGLT2 inhibitors inhibit only 30–50% of filtered ovascular outcomes and all-cause mortality among patients with
glucose load in humans. Diabetes. 2013;62:3324–8. type 2 diabetes mellitus. Am J Cardiol. 2016;118(11):1774–80.
42. Ruggenenti P, Porrini EL, Gaspari F, Motterlini N, Cannata A, 59. Saad M, Mahmoud AN, Elgendy IY, Abuzaid A, Barakat AF,
Carrara F, et al. Glomerular hyperfiltration and renal disease pro- Elgendy AY, et al. Cardiovascular outcomes with sodium- glu-
gression in type 2 diabetes. Diabetes Care. 2012;35:2061–8. cose co-transporter-2 inhibitors in patients with type II diabetes
43. Heerspink HJL, Perkins BA, Fitchett DH, Husain M, Cherney mellitus: a meta-analysis of placebo-controlled randomized tri-
DZI. Sodium glucose co-transporter 2 inhibitors in the treat- als. Int J Cardiol. 2017;228:352–8.
ment of diabetes mellitus: cardiovascular and kidney effects, 60. Wu JH, Foote C, Blomster J, Toyama T, Perkovic V, Sundström
potential mechanisms, and clinical applications. Circulation. J, et al. Effects of sodium- glucose co-transporter- 2 inhibitors on
2016;134:752–72. cardiovascular events, death, and major safety outcomes in adults
44. Pollock CA, Lawrence JR, Field MJ. Tubular sodium handling with type 2 diabetes: a systematic review and meta- analysis.
and tubuloglomerular feedback in experimental diabetes mellitus. Lancet Diabetes Endocrinol. 2016;4(5):411–9.
Am J Physiol Renal Physiol. 1991;260:946-F952. 61. Savarese G, D’Amore C, Federici M, De Martino F, Delle Srot-
45. Vallon V, Richter K, Blantz RC, Thomson S, Osswald H. taglie S, Marciano C, et al. Effects of dipeptidyl peptidase 4
Glomerular hyperfiltration in experimental diabetes melli- inhibitors and sodium-glucose linked co-transporter-2 Inhibitors
tus: potential role of tubular reabsorption. J Am Soc Nephrol. on cardiovascular events in patients with type 2 diabetes mellitus:
1999;10:2569–76. a meta- analysis. Int J Cardiol. 2016;220:595–601.
46. Kidokoro K, Cherney DZI, Bozovic A, Nagasu H, Satoh M, 62. Monami M, Dicembrini I, Mannucci E. Effects of SGLT-2 inhibi-
Kanda E, et al. Evaluation of glomerular hemodynamic func- tors on mortality and cardiovascular events: a comprehensive
tion by empagliflozin in diabetic mice using in vivo imaging. meta-analysis of randomized controlled trials. Acta Diabetol.
Circulation. 2019;140:303–15. 2017;54(1):19–36.
L. Del Vecchio et al.

63. Sonesson C, Johansson PA, Johnsson E, Gause-Nilsson I. Car- 79. Castoldi G, Carletti R, Ippolito S, Colzani M, Barzaghi F, Stella
diovascular effects of dapagliflozin in patients with type 2 dia- A, et al. Renal anti-fibrotic effect of sodium glucose co-trans-
betes and different risk categories: a meta-analysis. Cardiovasc porter 2 inhibition in angiotensin II-dependent hypertension. Am
Diabetol. 2016;15:37. J Nephrol. 2020;51:119–29.
64. Salsali A, Kim G, Woerle HJ, Broedl UC, Hantel S. Cardiovas- 80. Jaikumkao K, Pongchaidecha A, Chueakula N, Thongnak LO,
cular safety of empagliflozin in patients with type 2 diabetes: a Wanchai K, Chatsudthipong V, et al. Dapagliflozin, a sodium-
meta-analsis of data from randomized placebo-controlled trials. glucose co-transporter-2 inhibitor, slows the progression of renal
Diabetes Obes Metab. 2016;18(10):1034–40. complications through the suppression of renal inflammation,
65. Bhatt DL, Szarek M, Pitt B, Cannon CP, Leiter LA, McGuire endoplasmic reticulum stress and apoptosis in prediabetic rats.
DK, SCORED Investigators, et  al. Sotagliflozin in patients Diabetes Obes Metab. 2018;20:2617–26.
with diabetes and chronic kidney disease. N Engl J Med. 81. Yamato M, Kato N, Kakino A, Yamada KI, Inoguchi T. Low dose
2021;384(2):129–39. of sodium-glucose transporter 2 inhibitor ipragliflozin attenu-
66. Schork A, Saynisch J, Vosseler A, Jaghutriz BA, Heyne N, Peter ated renal dysfunction and interstitial fibrosis in adenine-induced
A, et al. Effect of SGLT2 inhibitors on body composition, fluid chronic kidney disease in mice without diabetes. Metabol Open.
status and renin-angiotensin-aldosterone system in type 2 dia- 2020;7: 100049.
betes: a prospective study using bioimpedance spectroscopy. 82. Takagi S, Li J, Takagaki Y, Kitada M, Nitta K, Takasu T,
Cardiovasc Diabetol. 2019;18(1):46. et  al. Ipragliflozin improves mitochondrial abnormalities in
67. Sarzani R, Giulietti F, Di Pentima C, Spannella F. Sodium-glu- renal tubules induced by a high-fat diet. J Diabetes Investig.
cose co-transporter-2 inhibitors: peculiar “hybrid” diuretics that 2018;9:1025–32.
protect from target organ damage and cardiovascular events. Nutr 83. Zhang Y, Nakano D, Guan Y, Hitomi H, Uemura A, Masaki
Metab Cardiovasc Dis. 2020;30(10):1622–32. T, et al. A sodium-glucose co-transporter 2 inhibitor attenu-
68. Masuda T, Muto S, Fukuda K, Watanabe M, Ohara K, Koep- ates renal capillary injury and fibrosis by a vascular endothelial
sell H, et al. Osmotic diuresis by SGLT2 inhibition stimulates growth factor-dependent pathway after renal injury in mice. Kid-
vasopressin-induced water reabsorption to maintain body fluid ney Int. 2018;94:524–35.
volume. Physiol Rep. 2020;8(2): e14360. 84. Kong KH, Oh HJ, Lim BJ, Kim M, Han KH, Choi YH, et al.
69. Kario K, Okada K, Kato M, Nishizawa M, Yoshida T, Asano Selective tubular activation of hypoxia-inducible factor-2alpha
T, et al. 24-hour blood pressure-lowering effect of an SGLT-2 has dual effects on renal fibrosis. Sci Rep. 2017;7(1):11351.
inhibitor in patients with diabetes and uncontrolled nocturnal 85. Cassis P, Locatelli M, Cerullo D, Corna D, Buelli S, Zanchi C,
hypertension: results from the randomized, placebo-controlled et al. SGLT2 inhibitor dapagliflozin limits podocyte damage in
SACRA study. Circulation. 2018;139(18):2089–97. proteinuric nondiabetic nephropathy. JCI Insight. 2018;3(15):
70. Scheen AJ. Effect of SGLT2 inhibitors on the sympathetic nerv- e98720.
ous system and blood pressure. Curr Cardiol Rep. 2019;21:70. 86. Wakisaka M, Nagao T, Yoshinari M. Sodium glucose cotrans-
71. Matthews VB, Elliot RH, Rudnicka C, Hricova J, Herat L, porter 2 (SGLT2) plays as a physiological glucose sensor and
Schlaich MP. Role of the sympathetic nervous system in reg- regulates cellular contractility in rat mesangial cells. PLoS ONE.
ulation of the sodium glucose co-transporter 2. J Hypertens. 2016;11(3): e0151585.
2017;35:2059–68. 87. Paccosi S, Giachi M, Di Gennaro P, Guglielmotti A, Parenti A.
72. McGuire DK, Shih WJ, Cosentino F, Charbonnel B, Cherney The chemokine (C-C motif) ligand protein synthesis inhibitor
DZI, Dagogo-Jack S, et al. Association of SGLT2 inhibitors with bindarit prevents cytoskeletal rearrangement and contraction of
cardiovascular and kidney outcomes in patients with type 2 dia- human mesangial cells. Cytokine. 2016;85:92–100.
betes. A meta-analysis. JAMA Cardiol. 2021;6(2):148–58. 88. Mohamed DI, Khairy E, Saad SST, Habib EK, Hamouda MA.
73. Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Potential protective effects of dapagliflozin in gentamicin
Mattheus M, EMPA-REG OUTCOME Investigators, et al. Empa- induced nephrotoxicity rat model via modulation of apoptosis
gliflozin and progression of kidney disease in type 2 diabetes. N associated miRNAs. Gene. 2019;707:198–204.
Engl J Med. 2016;375(4):323–34. 89. Hasan R, Lasker S, Hasan A, Zerin F, Zamila M, Parvez F, et al.
74. Seidu S, Kunutsor SK, Cos X, Gillani S, Khunti K, For and on Canagliflozin ameliorates renal oxidative stress and inflammation
behalf of Primary Care Diabetes Europe, et al. SGLT2 inhibi- by stimulating AMPK-Akt-eNOS pathway in the isoprenaline-
tors and renal outcomes in type 2 diabetes with or without renal induced oxidative stress model. Sci Rep. 2020;10(1):14659.
impairment: a systematic review and meta-analysis. Prim Care 90. Onishi A, Fu Y, Patel R, Darshi M, Crespo-Masip M, Huang W,
Diabetes. 2018;12(3):265–83. et al. A role for tubular Na+/H+ exchanger NHE3 in the natriu-
75. Zhang XL, Zhu QQ, Chen YH, Li XL, Chen F, Huang JA, et al. retic effect of the SGLT2 inhibitor empagliflozin. Am J Physiol
Cardiovascular safety, long- term non cardiovascular safety, and Renal Physiol. 2020;319(4):F712–28.
efficacy of sodium-glucose co-transporter 2 inhibitors in patients 91. Mannon EC, O’Connor PM. Alkali supplementation as a thera-
with type 2 diabetes mellitus: a systemic review and meta-anal- peutic in chronic kidney disease: what mediates protection? Am
ysis with trial sequential analysis. J Am Heart Assoc. 2018;7(2): J Physiol Renal Physiol. 2020;319(6):F1090–104.
e007165. 92. Zhang Y, Thai K, Kepecs DM, Gilbert RE. Sodium-glucose
76. Santer R, Kinner M, Lassen CL, Schneppenheim R, Eggert P, linked co-transporter-2 inhibition does not attenuate disease
Bald M, et al. Molecular analysis of the SGLT2 gene in patients progression in the rat remnant kidney model of chronic kidney
with renal glucosuria. J Am Soc Nephrol. 2003;14:2873–82. disease. PLoS ONE. 2016;11(1): e0144640.
77. Nespoux J, Patel R, Zhang H, Huang W, Freeman B, Sanders PW, 93. Ma Q, Steiger S, Anders HJ. Sodium glucose transporter-2 inhi-
et al. Gene knockout of the Na+-glucose co-transporter SGLT2i bition has no renoprotective effects on non-diabetic chronic kid-
in a murine model of acute kidney injury induced by ischemia- ney disease. Physiol Rep. 2017;5: e13228.
reperfusion. Am J Physiol Renal Physiol. 2020;318(5):F1100–12. 94. Wheeler DC, Stefansson BV, Batiushin M, Bilchenko O, Cher-
78. Pirklbauer M, Bernd M. Empagliflozin inhibits basal and IL-1β- ney DZI, Chertow GM, et al. The dapagliflozin and preven-
mediated MCP-1/CCL2 and endothelin-1 expression in human tion of adverse outcomes in chronic kidney disease (DAPA-
proximal tubular cells. Int J Mol Sci. 2020;21:8189. CKD) trial: baseline characteristics. Nephrol Dial Transplant.
2020;35(10):1700–11.
SGLT-2 Inhibitors in Non-diabetic CKD

95. Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, 2 diabetes: a systematic review and meta-analysis. Nephrol Dial
Greene T, Hou FF, DAPA-CKD Trial Committees and Investiga- Transplant. 2021 (Online ahead of print)
tors, et al. Dapagliflozin in patients with chronic kidney disease. 110. Ye N, Jardine MJ, Oshima M, Hockham C, Heerspink HJL, Agar-
N Engl J Med. 2020;383(15):1436–46. wal R, et al. Blood pressure effects of canagliflozin and clinical
96. Locatelli F, Del Vecchio L, Pozzoni P, D’Amico M, Andrulli S. outcomes in type 2 diabetes and chronic kidney disease: insights
Is it the agent or the blood pressure level that matters for renal from the CREDENCE Trial. Circulation. 2021;143(18):1735–49.
and vascular protection in chronic nephropathies? Kidney Int. 111. Chambergo-Michilot D, Tauma-Arrué A, Loli-Guevara S. Effects
2005;67(suppl 93):S15-19. and safety of SGLT2 inhibitors compared to placebo in patients
97. Wheeler DC, Stefánsson BV, Jongs N, Chertow GM, Greene T, with heart failure: a systematic review and meta-analysis. Int J
Hou FF, DAPA-CKD Trial Committees and Investigators, et al. Cardiol Heart Vasc. 2020;32: 100690.
Effects of dapagliflozin on major adverse kidney and cardiovas- 112. Feig DI, Kang D-H, Johnson RJ. Uric acid and cardiovascular
cular events in patients with diabetic and non-diabetic chronic risk. N Engl J Med. 2008;359(17):1811–21.
kidney disease: a prespecified analysis from the DAPA-CKD 113. Lytvyn Y, Škrtić M, Yang GK, Yip PM, Perkins BM, Cherney
trial. Lancet Diabetes Endocrinol. 2021;9(1):22–31. DZI. Glycosuria-mediated urinary uric acid excretion in patients
98. Wheeler DC, Toto RD, Stefansson BV, Jongs N, Chertow GM, with uncomplicated type 1 diabetes mellitus. Am J Physiol Renal
Greene T, for the DAPA-CKD Trial Committees and Investi- Physiol. 2015;308(2):F77-83.
gators, et al. A pre-specified analysis of the DAPA-CKD trial 114. Novikov A, Fu Y, Huang W, Freeman B, Patel R, van Ginkel C,
demonstrates the effects of dapagliflozin on major adverse et al. SGLT2 inhibition and renal urate excretion: role of lumi-
kidney events in patients with IgA nephropathy. Kidney Int. nal glucose, GLUT9, and URAT1. Am J Physiol Renal Physiol.
2021. https:// ​ d oi. ​ o rg/ ​ 1 0. ​ 1 016/j. ​ k int. ​ 2 021. ​ 0 3. ​ 0 33 (S0085- 2019;316(1):F173–85.
2538(21)00396-3; Online ahead of print). 115. Zhao Y, Xu L, Tian D, Xia P, Zheng H, Wang L, et al. Effects of
99. McMurray JJV, Wheeler DC, Stefánsson BV, Jongs N, Postmus sodium-glucose co-transporter 2 (SGLT2) inhibitors on serum
D, Correa-Rotter R, DAPA-CKD Trial Committees and Inves- uric acid level: a meta-analysis of randomized controlled trials.
tigators, et al. Effect of dapagliflozin on clinical outcomes in Diabetes Obes Metab. 2018;20(2):458–62.
patients with chronic kidney disease, with and without cardio- 116. Verma S, Ji Q, Bhatt DL, Mazer CD, Al-Omran M, Inzucchi
vascular disease. Circulation. 2021;143(5):438–48. SE, et  al. Association between uric acid levels and cardio-
100. Cherney DZI, Dekkers CCJ, Barbour SJ, Cattran D, Gafor renal outcomes and death in patients with type 2 diabetes: a
AHA, Greasley PJ, DIAMOND investigators, et  al. Effects subanalysis of EMPA-REG OUTCOME. Diabetes Obes Metab.
of the SGLT2 inhibitor dapagliflozin on proteinuria in non- 2020;22(7):1207–14.
diabetic patients with chronic kidney disease (DIAMOND): a 117. Stack AG, Han D, Goldwater R, Johansson S, Dronam-
randomised, double-blind, crossover trial. Lancet Diabetes Endo- raju N, Oscarsson J, et al. Dapagliflozin added to verinurad
crinol. 2020;8(7):582–93. plus febuxostat further reduces serum uric acid in hyper-
101. Rajasekeran H, Reich HN, Hladunewich MA, Cattran D, Lovshin uricemia: the QUARTZ Study. J Clin Endocrinol Metab.
JA, Lytvyn Y, et al. Dapagliflozin in focal segmental glomeru- 2021;106(5):e2347–56.
losclerosis: a combined human-rodent pilot study. Am J Physiol 118. Cosentino C, Dicembrini I, Nreu B, Mannucci E, Monami M.
Renal Physiol. 2018;314(3):F412–22. Nephrolithiasis and sodium-glucose co-transporter-2 (SGLT-2)
102. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson inhibitors: a meta-analysis of randomized controlled trials. Dia-
P, et al. Cardiovascular and renal outcomes with empagliflozin betes Res Clin Pract. 2019;155: 107808.
in heart failure. N Engl J Med. 2020;383:1413–24. 119. Stefánsson BV, Heerspink HJL, Wheeler DC, Sjöström CD,
103. Petrie MC, Verma S, Docherty KF, Inzucchi SE, Anand I, Greasley PJ, Sartipy P, et al. Correction of anemia by dapagli-
Bělohlávek J, et al. Effect of dapagliflozin on worsening heart flozin in patients with type 2 diabetes. J Diabetes Complicat.
failure and cardiovascular death in patients with heart failure 2020;34(12): 107729.
with and without diabetes. JAMA. 2020;323:1353–68. 120. Oshima M, Neuen BL, Jardine MJ, Bakris G, Edwards R, Levin
104. Zannad F, Ferreira JP, Pocock SJ, Zeller C, Anker SD, Butler A, et al. Effects of canagliflozin on anaemia in patients with
J, et al. Cardiac and kidney benefits of empagliflozin in heart type 2 diabetes and chronic kidney disease: a post-hoc analy-
failure across the spectrum of kidney function: insights from sis from the CREDENCE trial. Lancet Diabetes Endocrinol.
EMPEROR-Reduced. Circulation. 2021;143(4):310–21. 2020;8(11):903–14.
105. Zannad F, Ferreira JP, Pocock SJ, Anker SD, Butler J, Filippatos 121. Maruyama T, Takashima H, Oguma H, Nakamura Y, Ohno M,
G, et al. SGLT2 inhibitors in patients with heart failure with Utsunomiya K, et al. Canagliflozin improves erythropoiesis in
reduced ejection fraction: a meta-analysis of the EMPEROR- diabetes patients with anemia of chronic kidney disease. Diabetes
Reduced and DAPA-HF trials. Lancet. 2020;396:819–29. Technol Ther. 2019;21(12):713–20.
106. Bhatt DL, Szarek M, Steg PG, Cannon CP, Leiter LA, McGuire 122. Marathias KP, Lambadiari VA, Markakis KP, Vlahakos VD,
DK, et al. Sotagliflozin in patients with diabetes and recent wors- Bacharaki D, Raptis AE, et al. Competing effects of renin angio-
ening heart failure. N Engl J Med. 2020;384:117–28. tensin system blockade and sodium-glucose co-transporter-2
107. Kimura G. Importance of inhibiting sodium-glucose co- inhibitors on erythropoietin secretion in diabetes. Am J Nephrol.
transporter and its compelling indication in type 2 diabe- 2020;51(5):349–56.
tes: pathophysiological hypothesis. J Am Soc Hypertens. 123. Panchapakesan U, Pegg K, Gross S, Komala MG, Mudaliar H,
2016;10(3):271–8. Forbes J, et al. Effects of SGLT2 inhibition in human kidney
108. Sanidas EA, Papadopoulos DP, Hatziagelaki E, Grassos C, Vel- proximal tubular cells–renoprotection in diabetic nephropathy?
liou M, Barbetseas J. Sodium glucose co-transporter 2 (SGLT2) PLoS ONE. 2013;8(2): e54442.
inhibitors across the spectrum of hypertension. Am J Hypertens. 124. Packer M. Mutual antagonism of hypoxia-inducible factor iso-
2020;33(3):207–13. forms in cardiac, vascular, and renal disorders. JACC Basic
109. Tian B, Deng Y, Cai Y, Han M, Xu G. Efficacy and safety of Transl Sci. 2020;5(9):961–8.
combination therapy with sodium-glucose transporter 2 inhibi- 125. Krishan P, Singh G, Bedi O. Carbohydrate restriction amelio-
tors and renin-angiotensin system blockers in patients with type rates nephropathy by reducing oxidative stress and upregulating
L. Del Vecchio et al.

HIF-1a levels in type-1 diabetic rats. J Diabetes Metab Disord. treated with sacubitril/valsartan: the DAPA-HF trial. JACC Heart
2017;16:47. Fail. 2020;8:811–8.
126. Bessho R, Takiyama Y, Takiyama T, et al. Hypoxia-inducible 145. Sarafidis PA, Memmos E, Alexandrou ME, Papagianni A.
factor-1α is the therapeutic target of the SGLT2 inhibitor for Mineralocorticoid receptor antagonists for nephroprotection:
diabetic nephropathy. Sci Rep. 2019;9:14754. current evidence and future perspectives. Curr Pharm Des.
127. Ghanim H, Abuaysheh S, Hejna J, Green K, Batra M, Makdissi 2018;24(46):5528–36.
A, et al. Dapagliflozin suppresses hepcidin and increases eryth- 146. Barrera-Chimal J, Girerd S, Jaisser F. Mineralocorticoid recep-
ropoiesis. J Clin Endocrinol Metab. 2020;105(4): dgaa057. tor antagonists and kidney diseases: pathophysiological basis.
128. Mazer CD, Hare GMT, Connelly PW, Gilbert RE, Shehata N, Kidney Int. 2019;96(2):302–19.
Quan A, et al. Effect of empagliflozin on erythropoietin levels, 147. Bakris GL, Agarwal R, Anker SD, Pitt B, Ruilope LM, Rossing
iron stores, and red blood cell morphology in patients with type P, FIDELIO-DKD Investigators, et al. Effect of finerenone on
2 diabetes mellitus and coronary artery disease. Circulation. chronic kidney disease outcomes in type 2 diabetes. N Engl J
2020;141:704–7. Med. 2020;383(23):2219–29.
129. Sowton AP, Griffin JL, Murray AJ. Metabolic profiling of the dia- 148. Sawamura T, Karashima S, Nagase S, Nambo H, Shimizu E,
betic heart: toward a richer picture. Front Physiol. 2019;10:639. Higashitani T, et al. Effect of sodium-glucose co-transporter-2
130. Selvaraj S, Kelly DP, Margulies KB. Implications of altered inhibitors on aldosterone-to-renin ratio in diabetic patients with
ketone metabolism and therapeutic ketosis in heart failure. Cir- hypertension: a retrospective observational study. BMC Endocr
culation. 2020;141(22):1800–12. Disord. 2020;20(1):177.
131. Cavaiola TS, Pettus J. Cardiovascular effects of sodium glu- 149. Shen L, Kristensen SL, Bengtsson O, Böhm M, de Boer RA,
cose co-transporter 2 inhibitors. Diabetes Metab Syndr Obes. Docherty KF, et al. Dapagliflozin in HFrEF patients treated with
2018;11:133–48. mineralocorticoid receptor antagonists: an analysis of DAPA-HF.
132. Inagaki N, Kazuoki Kondo K, Yoshinari T, Kuki H. Efficacy and JACC Heart Fail. 2021;S2213–1779(20):30704–6.
safety of canagliflozin alone or as add-on to other oral antihy- 150. Ortiz A, Ferro CJ, Balafa O, Burnier M, Ekart R, Halimi JM,
perglycemic drugs in Japanese patients with type 2 diabetes: A et al. Mineralocorticoid receptors antagonists for nephroprotec-
52-week open-label study. J Diabetes Investig. 2015;6(2):210–8. tion and cardioprotection in patients with diabetes mellitus and
133. Tomita I, Kume S, Sugahara S, Osawa N, Yamahara Y, Yas- chronic kidney disease. A consensus statement by the European
uda-Yamahara M, et al. SGLT2 inhibition mediates protection Renal and Cardiovascular Medicine (EURECA-m) working
from diabetic kidney disease by promoting ketone body-induced group of the European Renal Association—European Dialysis
mTORC1 inhibition. Cell Metab. 2020;32(3):404–19. and Transplant Association (ERA-EDTA) and the Hypertension
134. Xu L, Nagata N, Nagashimada M, Zhuge F, Ni Y, Chen G, May- and Kidney working group of the European Society of Hyperten-
oux E, et al. SGLT2 Inhibition by empagliflozin promotes fat sion (ESH). Nephrol Dial Transplant. 2021. https://​doi.​org/​10.​
utilization and browning and attenuates inflammation and insulin 1093/​ndt/​gfab1​67 (Online ahead of print).
resistance by polarizing M2 macrophages in diet-induced obese 151. Pelletier R, Ng K, Alkabbani W, Labib Y, Mourad N, Gam-
mice. EBioMedicine. 2017;20:137–49. ble JM. Adverse events associated with sodium glucose co-
135. Jones BJ, Tan T, Bloom SR. Minireview: glucagon in stress and transporter 2 inhibitors: an overview of quantitative systematic
energy homeostasis. Endocrinology. 2012;153(3):1049–54. reviews. Ther Adv Drug Saf. 2021;12:2042098621989134.
136. Ferrannini E, Muscelli E, Frascerra S, et al. Metabolic response 152. Zhao M, Sun S, Huang Z, Wang T, Tang H. Network meta-anal-
to sodium- glucose co-transporter 2 inhibition in ty pe 2 diabetic ysis of novel glucose-lowering drugs on risk of acute kidney
patients. J Clin Invest. 2014;124(2):499–508. injury. Clin J Am Soc Nephrol. 2020;16(1):70–8.
137. Bonner C, Kerr-Conte J, Gmyr V, et al. Inhibition of the glucose 153. Rampersad C, Kraut E, Whitlock RH, Komenda P, Woo V,
transporter SGLT2 with dapagliflozin in pancreatic alpha cells Rigatto C, et al. Acute kidney injury events in patients with
triggers glucagon secretion. Nat Med. 2015;21(5):512–7. type 2 diabetes using SGLT2 inhibitors versus other glucose-
138. Hattori Y. Beneficial effects on kidney during treatment with lowering drugs: a retrospective cohort study. Am J Kidney Dis.
sodium-glucose co-transporter 2 inhibitors: proposed role of 2020;76(4):471–9.
ketone utilization. Heart Fail Rev. 2021;26(4):947–52. 154. Jardine MJ, Zhou Z, Mahaffey KW, Oshima M, Agarwal R,
139. Oelze M, Kröller-Schön S, Welschof P, Ansen T, Hausding M, Bakris G, CREDENCE Study Investigators, et al. Renal, cardio-
Mikhed Y, et al. The sodium-glucose co-transporter 2 inhibitor vascular, and safety outcomes of canagliflozin by baseline kidney
empagliflozin improves diabetes-induced vascular dysfunction in function: a secondary analysis of the CREDENCE randomized
the streptozotocin diabetes rat model by interfering with oxida- trial. J Am Soc Nephrol. 2020;31(5):1128–39.
tive stress and glucotoxicity. PLoS ONE. 2014;9(11): e112394. 155. Oshima M, Jardine MJ, Agarwal R, Bakris G, Cannon CP, Char-
140. Yaribeygi H, Atkin SL, Butler AE, Sahebkar A. Sodium–glucose ytan DM, et al. Insights from CREDENCE trial indicate an acute
co-transporter inhibitors and oxidative stress: an update. J Cell drop in estimated glomerular filtration rate during treatment with
Physiol. 2019;234(4):3231–7. canagliflozin with implications for clinical practice. Kidney Int.
141. McMurray JJV, Packer M, Desai AS, Gong J, Lefkowitz MP, Riz- 2021;99(4):999–1009.
kala AR, et al. Angiotensin–neprilysin inhibition versus enalapril 156. Ryan R, Choo S, Willows J, Walker J, Prasad K, Tez D. Acute
in heart failure. N Engl J Med. 2014;371:993–1004. interstitial nephritis due to sodium-glucose co-transporter 2
142. Damman K, Gori M, Claggett B, Jhund PS, Senni M, Lefkowitz inhibitor empagliflozin. Clin Kidney J. 2020;14(3):1020–2.
MP, et al. Renal effects and associated outcomes during angi- 157. Cianciolo G, De Pascalis A, Gasperoni L, Tondolo F, Zappulo
otensin-neprilysin inhibition in heart failure. JACC Hear Fail. F, Capelli I, et al. The off-target effects, electrolyte and mineral
2018;6:489–98. disorders of SGLT2i. Molecules. 2020;25(12):2757.
143. Ruggenenti P, Remuzzi G. Combined neprilysin and RAS inhibi- 158. Barrett PQ, Aronson PS. Glucose and alanine inhibition of phos-
tion for the failing heart: Straining the kidney to help the heart? phate transport in renal microvillus membrane vesicles. Am J
Eur J Heart Fail. 2015;17:468–71. Physiol. 1982;242:126–31.
144. Solomon SD, Jhund PS, Claggett BL, Dewan P, Køber L, Kosi- 159. Lang F. Osmotic diuresis. Renal Physiol. 1987;10:160–73.
borod MN, et al. Effect of dapagliflozin in patients with hfref 160. Blau JE, Bauman V, Conway EM, Piaggi P, Wal-

ter MF, Wright EC, et  al. Canaglif lozin triggers the
SGLT-2 Inhibitors in Non-diabetic CKD

FGF23/1,25-dihydroxyvitamin D/PTH axis in healthy volunteers 170. Leenders NHJ, Vermeulen EA, van Ballegooijen AJ, Hoek-
in a randomized crossover study. JCI Insight. 2018;3: e99123. stra T, de Vries R, Beulens JW, Vervloet MG. The association
161. de Jong MA, Petrykiv SI, Laverman GD, van Herwaarden AE, de between circulating magnesium and clinically relevant outcomes
Zeeuw D, Bakker SJL, et al. Effects of dapagliflozin on circulat- in patients with chronic kidney disease: a systematic review and
ing markers of phosphate homeostasis. Clin J Am Soc Nephrol. meta-analysis. Clin Nutr. 2021;40(5):3133–47.
2019;14(1):66–73. 171. Xiong J, He T, Wang M, Nie L, Zhang Y, Wang Y, et al. Serum
162. Li X, Li T, Cheng Y, Lu Y, Xue M, Xu L, et  al. Effects of magnesium, mortality, and cardiovascular disease in chronic kid-
SGLT2 inhibitors on fractures and bone mineral density in type ney disease and end-stage renal disease patients: a systematic
2 diabetes: an updated meta-analysis. Diabetes Metab Res Rev. review and meta-analysis. J Nephrol. 2019;32(5):791–802.
2019;35(7): e3170. 172. Li D, Wang T, Shen S, Fang Z, Dong Y, Tang H. Urinary tract
163. Tang H, Zhang X, Zhang J, Li Y, Del Gobbo LC, Zhai S, et al. and genital infections in patients with type 2 diabetes treated
Elevated serum magnesium associated with SGLT2 inhibitor use with sodium-glucose co-transporter 2 inhibitors: a meta-anal-
in type 2 diabetes patients: a meta-analysis of randomised con- ysis of randomized controlled trials. Diabetes Obes Metab.
trolled trials. Diabetologia. 2016;59(12):2546–51. 2017;19(3):348–55.
164. Weir MR, Kline I, Xie J, Edwards R, Usiskin K. Effect of cana- 173. Bakris G, Oshima M, Mahaffey KW, Agarwal R, Cannon CP,
gliflozin on serum electrolytes in patients with type 2 diabetes Capuano G, et al. Effects of canagliflozin in patients with base-
in relation to estimated glomerular filtration rate (eGFR). Curr line eGFR < 30 mL/min per 1.73 m(2): subgroup analysis of
Med Res Opin. 2014;30(9):1759–68. the randomized CREDENCE Trial. Clin J Am Soc Nephrol.
165. Gilbert RE, Mende C, Vijapurkar U, Sha S, Davies MJ, Desai 2020;15(12):1705–14.
M. Effects of canagliflozin on serum magnesium in patients with 174. Yu J, Arnott C, Neuen BL, Heersprink HL, Mahaffey KW, Can-
type 2 diabetes mellitus: a post hoc analysis of randomized con- non CP, et al. Cardiovascular and renal outcomes with canagli-
trolled trials. Diabetes Ther. 2017;8(2):451–8. flozin according to baseline diuretic use: a post hoc analysis from
166. Filippatos TD, Tsimihodimos V, Liamis G, Elisaf MS. the CANVAS Program. ESC Heart Fail. 2021;8(2):1482–93.
SGLT2 inhibitors-induced electrolyte abnormalities: an analy- 175. Griffin M, Riello R, Rao VS, Ivey-Miranda J, Fleming J, Maulion
sis of the associated mechanisms. Diabetes Metab Syndr. C, et al. Sodium glucose co-transporter 2 inhibitors as diuretic
2018;12(1):59–63. adjuvants in acute decompensated heart failure: a case series.
167. Wang KM, Li JW, Bhalla V, Jardine MJ, Neal B, de Zeeuw D. ESC Heart Fail. 2020;7(4):1966–71.
Canagliflozin, serum magnesium and cardiovascular outcomes— 176. Shentu Y, Li Y, Xie S, Jiang H, Sun S, Lin R, et al. Empagli-
analysis from the CANVAS Program. Endocrinol Diab Metab. flozin, a sodium glucose co-transporter-2 inhibitor, ameliorates
2021. https://​doi.​org/​10.​1002/​edm2.​247. peritoneal fibrosis via suppressing TGF-beta/Smad signaling. Int
168. van de Wal-Visscher ER, Kooman JP, van der Sande FM. Mag- Immunopharmacol. 2021;93: 107374.
nesium in chronic kidney disease: should we care? Blood Purif. 177. He Y, Pachori A, Chen P, Ma S, Mendonza AE, Amer A, et al.
2018;45(1–3):173–8. Glucosuric, renal and haemodynamic effects of licogliflozin, a
169. Ter Braake AD, Vervloet MG, de Baaij JHF, Hoenderop JGJ. dual inhibitor of sodium-glucose co-transporter-1 and sodium-
Magnesium to prevent kidney disease-associated vascular cal- glucose co-transporter-2, in patients with chronic kidney disease:
cification: crystal clear? Nephrol Dial Transplant. 2020 (Online a randomized trial. Diabetes Obes Metab. 2021;23(5):1182–90.
ahead of print).

You might also like