Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2020 Oct 23;43(12):1211–1221. doi: 10.1007/s40264-020-01010-6

Potential Safety Issues with Use of Sodium-Glucose Cotransporter 2 Inhibitors, Particularly in People with Type 2 Diabetes and Chronic Kidney Disease

Tamara Y Milder 1,2,3,4, Sophie L Stocker 2,4, Richard O Day 2,4, Jerry R Greenfield 1,3,4,
PMCID: PMC7582030  PMID: 33095409

Abstract

Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a major advance in the fields of diabetology, nephrology, and cardiology. The cardiovascular and renal benefits of SGLT2 inhibitors are likely largely independent of their glycaemic effects, and this understanding is central to the use of these agents in the high-risk population of people with type 2 diabetes and chronic kidney disease. There are a number of potential safety issues associated with the use of SGLT2 inhibitors. These include the rare but serious risks of diabetic ketoacidosis and necrotising fasciitis of the perineum. The data regarding a possibly increased risk of lower limb amputation and fracture with SGLT2 inhibitor therapy are conflicting. This article aims to explore the potential safety issues associated with the use of SGLT2 inhibitors, with a particular focus on the safety of these drugs in people with type 2 diabetes and chronic kidney disease. We discuss strategies that clinicians can implement to minimise the risk of adverse effects including diabetic ketoacidosis and volume depletion. Risk mitigation strategies with respect to SGLT2 inhibitor-associated diabetic ketoacidosis are of particular importance during the current coronavirus disease 2019 (COVID-19) pandemic.

Key Points

Sodium-glucose cotransporter 2 (SGLT2) inhibitors have a number of adverse effects—the most serious of which are diabetic ketoacidosis and necrotising fasciitis of the perineum.
Clinicians should educate patients to temporarily stop taking their SGLT2 inhibitor when acutely unwell with reduced oral intake, to reduce their risk of diabetic ketoacidosis and acute kidney injury, and this education is especially important during the coronavirus disease 2019 (COVID-19) pandemic.
In very large randomised controlled trials, SGLT2 inhibitors have been associated with a lower risk of acute kidney injury. These drugs should not, however, be prescribed to a patient who is hypovolaemic or hypotensive, and a patient’s loop and/or thiazide diuretic dose may need to be reduced.

Introduction

Diabetes is the leading cause of chronic kidney disease (CKD) [1], and the coexistence of both conditions places an individual at high risk of cardiovascular disease and death [2]. Sodium-glucose cotransporter 2 (SGLT2) inhibitors inhibit proximal tubular glucose reabsorption, leading to glycosuria [3]. These drugs have important cardiovascular and renal benefits [49], leading one expert to refer to SGLT2 inhibitors as “cardiorenal risk reducing agents that have glucose lowering as a side effect” [10]. Furthermore, the cardiovascular and renal benefits of SGLT2 inhibitors appear to be largely independent of their glycaemic effects [9, 1113]. This point is pertinent to the use of these drugs in people with type 2 diabetes and CKD where glycosuria secondary to SGLT2 inhibition is reduced, resulting in potentially limited anti-hyperglycaemic efficacy [1417]. Indeed, in two heart failure trials (DAPA-HF and EMPEROR-Reduced) and a CKD trial (DAPA-CKD) where the primary endpoint was met, the effect of the SGLT2 inhibitor on the primary outcome was consistent in participants irrespective of the presence or absence of diabetes [9, 18, 19]. There are a multitude of proposed mechanisms for the cardioprotective properties of SGLT2 inhibitors, including natriuresis and osmotic diuresis, inhibition of the sodium-hydrogen exchanger in the myocardium, potential use of ketone bodies for cardiac metabolism, and reduced cardiac fibrosis and inflammation [20]. These results suggest that the benefits may be independent of effects on glycaemia. Given the use of these drugs by not only endocrinologists and primary care physicians, but also nephrologists and cardiologists, clinicians need to become familiar with the physiology, efficacy, and safety of SGLT2 inhibitors [10, 21, 22]. Indeed, SGLT2 inhibitors are associated with a number of adverse effects, including diabetic ketoacidosis (DKA), which is potentially life-threatening. Furthermore, adverse effects can occur very quickly. There have been many reviews exploring the efficacy of these agents in different population groups. Hence, this article examines the potential safety issues associated with the use of SGLT2 inhibitors, with a particular focus on the safety of these drugs in people with type 2 diabetes and CKD. We highlight measures that clinicians can implement to minimise the risk of adverse effects, including DKA, which is of particular relevance during the current coronavirus disease 2019 (COVID-19) pandemic.

Safety Issues in Patients With Versus Those Without CKD

The cardiovascular, renal and heart failure outcome trials to date have differed with respect to inclusion and exclusion criteria, including estimated glomerular filtration rate (eGFR) cut-offs [4, 69, 18, 23, 24]. Importantly, there is limited available data specifically  about SGLT2 inhibitor use in patients with severe CKD. Subgroup analyses from the EMPA-REG OUTCOME, CANVAS, and DECLARE-TIMI 58 trials found similar adverse event profiles with respect to specific SGLT2 inhibitors among participants with different baseline eGFR levels (Table 1) [2527]. In a subgroup analysis of the CREDENCE trial, severe adverse events were consistent among screening eGFR categories. There was a significant interaction test for volume depletion, with a higher risk with canagliflozin apparent in participants with screening eGFR 30 to < 45, but not eGFRs 45 to < 60 or 60 to < 90 mL/min/1.73 m2 (Table 1) [28]. Of note, empagliflozin, dapagliflozin, canagliflozin, and ertugliflozin exposure increases with advancing renal impairment; however, the area under the concentration–time curve does not exceed by twofold that reported in subjects with normal renal function [1417]. Canagliflozin is the only one of these four SGLT2 inhibitors for which use of the lower dose (100 mg once daily) is recommended for patients with renal impairment (specifically eGFR 30 to < 60 mL/min/1.73 m2) [29]. However, in Australia, empagliflozin, dapagliflozin, and ertugliflozin are contraindicated in patients with eGFR persistently < 45 mL/min/1.73 m2, largely due to limited anti-hyperglycaemic efficacy [3032].

Table 1.

Subgroup analyses from major SGLT2 inhibitor trials with respect to adverse event profiles of SGLT2 inhibitors in participants with different baseline eGFRs [2528]

Trial (SGLT2 inhibitor studied) Adverse event profile of SGLT2 inhibitors in participants with different baseline eGFRs
EMPA-REG OUTCOME (empagliflozin) Similar in participants with eGFR < 45, 45 to < 60, and ≥ 60 mL/min/1.73 m2
CANVAS Program (canagliflozin) Consistent across eGFR subgroups (< 45, 45 to < 60, 60 to < 90, and ≥ 90 mL/min/1.73 m2); however, trend (P heterogeneity = 0.06) for higher risk of hypoglycaemia in lower eGFR subgroup—noted that subgroups with lower eGFR had higher insulin use
DECLARE-TIMI 58 (dapagliflozin) Consistent across eGFR subgroups (< 60, 60 to < 90, and ≥ 90 mL/min/1.73 m2)
CREDENCE (canagliflozin) Consistent across eGFR subgroups (30 to < 45, 45 to < 60, and 60 to < 90 mL/min/1.73 m2 at screening) with respect to serious adverse events, amputation, and fracture. However, significant interaction test for volume depletion (P = 0.01), hazard ratio of volume depletion for participants with eGFR 30 to < 45 mL/min/1.73 m2 1.99 (95% CI 1.33–2.98) (canagliflozin vs placebo), compared with hazard ratio for participants with eGFR 60 to < 90 mL/min/1.73 m2 0.89 (0.58–1.38)

CI confidence interval, eGFR estimated glomerular filtration rate, SGLT2 sodium-glucose cotransporter 2

Infections of the Genitalia and Perineum

Diabetes, particularly with poor control and glycosuria, is a known risk factor for genital infection [33]. Diabetes may suppress the immune response to infection [34]. Glucose present on the genitalia due to SGLT2 inhibition is believed to aid growth and adherence of yeast and impair the local immune response [35]. A meta-analysis of randomised controlled trials (RCTs) and two large population-based studies have demonstrated an approximate threefold increase in risk of genital infection with SGLT2 inhibitor use compared with placebo or other diabetes drug classes [3638]. In the CREDENCE trial, the event rate for genital mycotic infection in the canagliflozin versus placebo groups for females was 12.6 versus 6.1 per 1000 person-years, and for men, 8.4 versus 0.9 per 1000 person-years [8]. A study of two large cohorts of commercially insured patients in the United States found that the elevated risk of genital infections was apparent within the first month of SGLT2 inhibitor treatment and remained elevated during the course of treatment [37]. Furthermore, the risk of genital infections with SGLT2 inhibitor use was greater in the subgroup of patients aged 60 years and over. History of prior genital infection (especially recent history) is a clear risk factor for the development of genital infection during SGLT2 inhibitor treatment [39]. Toyama et al. conducted a meta-analysis of RCTs of SGLT2 inhibitors in patients with type 2 diabetes and CKD (defined as eGFR < 60 mL/min/1.73 m2) and found an approximate threefold increase in the risk of genital infections [40]. This suggests that the increase in the risk of genital infections in patients with CKD is similar to that in the non-CKD population. Most genital infections are mild to moderate in severity and are responsive to topical antifungals or a single dose of fluconazole [41, 42]. These infections do not necessitate cessation of the agent [42]. Clinicians should recommend good perineal hygiene to patients [43].

In 2018, the Food and Drug Administration (FDA) issued a warning about the risk of necrotising fasciitis of the perineum (Fournier’s gangrene) with SGLT2 inhibitor treatment [44]. There were 55 cases of Fournier’s gangrene requiring surgical debridement associated with SGLT2 inhibitor use reported to the FDA Adverse Event Reporting System (FAERS) between March 2013 and January 2019 [45]. All patients were severely ill, at least 25 patients needed more than one surgery, and three patients died. A number of patients had complicating DKA, sepsis, and/or acute kidney injury (AKI). The time between initiation of SGLT2 inhibitor therapy and infection was variable—5 days to 49 months. Large RCTs have not demonstrated an increased risk of Fournier’s gangrene with SGLT2 inhibitor treatment. In the DECLARE-TIMI 58 trial, one patient in the dapagliflozin group compared with five patients in the placebo group experienced Fournier’s gangrene [7]. However, RCTs are not designed or powered to demonstrate or refute an increased risk of extremely rare events such as Fournier’s gangrene. The documented case reports submitted to regulatory and surveillance agencies need to be carefully considered.

Urinary Tract Infections

In 2015, the FDA issued a warning about the risk of serious urinary tract infections (UTIs) with SGLT2 inhibitor use due to 19 cases of urosepsis and pyelonephritis reported over an 18-month period [46]. In contrast, SGLT2 inhibitors have generally not been associated with an elevated risk of UTIs in large meta-analyses and population-based studies [36, 38, 47, 48]. One of the four cardiovascular outcome trials to date, however, has demonstrated a significant increase in risk of UTIs with SGLT2 inhibitor therapy; in the VERTIS CV trial approximately 12% versus 10% of participants randomised to ertugliflozin and placebo, respectively, experienced a UTI [49]. In the CREDENCE trial, there was no significant difference in the rate of UTIs between the canagliflozin and placebo groups [8]. The exact rate of pyelonephritis and urosepsis was not reported. Whether there are differences in the risk of UTI based on the type of SGLT2 inhibitor is yet to be established. With respect to why SGLT2 inhibition may not increase the risk of UTIs despite causing glycosuria, Fralick and MacFadden have hypothesised that diuresis and polyuria secondary to SGLT2 inhibition counters potential bacterial growth due to glycosuria and/or prevents bacterial ascension of the urinary tract [35]. Severe CKD may lead to reduced urine output, and glycosuria secondary to SGLT2 inhibition is reduced [1417]. The influence of these factors on the risk of UTIs is uncertain as data regarding SGLT2 inhibitor treatment in patients with stage 4 and 5 CKD (eGFR 15–29 mL/min/1.73 m2 and < 15 mL/min/1.73 m2 or requiring dialysis, respectively) are limited. Also unclear is the risk of UTIs in higher-risk populations such as people with urinary tract structural or functional abnormalities or people who are immunosuppressed. With regard to post-transplant diabetes mellitus in renal transplant recipients, in one RCT (n = 49 patients), which compared empagliflozin or placebo treatment for 24 weeks, three patients in both the empagliflozin and placebo groups experienced a UTI [50]. However, two patients in the empagliflozin group had to discontinue treatment—one because of urosepsis and one because of repeated UTIs. The patient who experienced urosepsis had a history of recurrent UTIs, and clinicians should be cautious when considering prescribing SGLT2 inhibitors to patients with a history of recurrent UTIs.

Hypoglycaemia

Given the insulin-independent mechanism of action of SGLT2 inhibitors, these agents are not associated with an increased risk of hypoglycaemia [4, 6, 7, 51]. These drugs lower plasma glucose by inducing glycosuria, resulting in a reduction in plasma insulin concentration and an increase in plasma glucagon concentration, leading to an increase in endogenous glucose production [51, 52]. The absence of hypoglycaemia risk was clearly seen in the DAPA-HF trial, where 55% of patients did not have diabetes [9]. However, if a patient is prescribed insulin and/or a sulfonylurea, the doses of these medications may need to be reduced, as SGLT2 inhibitors reduce glycated haemoglobin (HbA1c) by 0.6–0.9% (Table 2) [53]. With regard to adjusting concomitant diabetes medications in patients with moderate to severe CKD, clinicians should note that SGLT2 inhibitors have limited anti-hyperglycaemic efficacy, due to reduced glycosuria [54].

Table 2.

Suggested adjustment of concomitant diabetic and non-diabetic medication when initiating an SGLT2 inhibitor [43, 93]

Medication Suggested adjustment
Diabetes
Insulin Consider reducing dose if HbA1c < 8.0%. However, do not excessively reduce insulin dose (for example, > 20%) as this increases the risk of DKA
Sulfonylurea Consider reducing dose or stopping if HbA1c < 8.0%
In patients with advanced CKD, SGLT2 inhibitors have limited anti-hyperglycaemic efficacy due to reduced glycosuria, which should factor into decisions about potentially changing other diabetes medications. Insulin doses need to be reduced in advanced CKD due to an increased half-life of the drug irrespective of concomitant SGLT2 inhibitor use
Non-diabetes
Loop and/or thiazide diuretics Consider reducing dose of diuretic if systolic blood pressure < 120 mmHg. If evidence of dehydration based on fluid balance assessment, recommend reducing dose or stopping diuretic and only starting SGLT2 inhibitor when dehydration resolved

DKA diabetic ketoacidosis, CKD chronic kidney disease, HbA1c glycated haemoglobin, SGLT2 sodium-glucose cotransporter 2

Volume Depletion

In 2015/2016, the FDA issued a warning about the risk of AKI with canagliflozin and dapagliflozin based on 101 cases over an approximate 2.5-year period, some requiring hospitalisation and dialysis [55]. In approximately half of the cases, AKI occurred within 1 month of SGLT2 inhibitor initiation. CREDENCE and the cardiovascular outcome trials have clearly demonstrated the important renoprotective effects of SGLT2 inhibitors [8, 56, 57]. In CREDENCE and the cardiovascular outcome trials (EMPA-REG OUTCOME, CANVAS Program, and DECLARE-TIMI 58), there was a 25% lower risk of AKI with SGLT2 inhibitor treatment compared with placebo [57]. There is often a mild acute decrease in eGFR with SGLT2 inhibitor initiation that is reversible on treatment cessation—the reduction in the CREDENCE trial at 3 weeks was −3.7 mL/min/1.73 m2 [8, 58]. In the trial, the mean change in eGFR slope was lower in the canagliflozin group compared with the placebo group (−3.19 vs −4.71 mL/min/1.73 m2 per year) [8]. The mild reduction in eGFR with SGLT2 inhibitor initiation does not represent AKI. This effect is thought to be due to increased tubular sodium delivery to the macula densa activating tubuloglomerular feedback, resulting in afferent arteriolar vasoconstriction, which is protective in the long-term because of the reduction in intraglomerular pressure [58]. This theory is partly based on data in young adults with type 1 diabetes and hyperfiltration [59]. However, the recent Renoprotective Effects of Dapagliflozin in Type 2 Diabetes trial questioned this theory [60]. In patients with type 2 diabetes without overt nephropathy, 12 weeks of dapagliflozin reduced GFR, filtration fraction, and intraglomerular pressure without increasing renal vascular resistance—suggesting that the acute eGFR decline is due to efferent arteriolar vasodilation rather than afferent arteriolar vasoconstriction [60, 61]. In the EMPA-REG OUTCOME trial, the acute dip in eGFR with empagliflozin treatment was greater in users of an angiotensin converting enzyme (ACE) inhibitor, an angiotensin receptor blocker (ARB), or any diuretic compared with non-users [62]. However, in users of these medications, adding empagliflozin did not increase the risk of AKI compared with adding placebo [62].

SGLT2 inhibitors should not be initiated in patients who are hypovolaemic and/or hypotensive, because this could contribute to AKI. Further, patients prescribed loop and/or thiazide diuretics may need dose reduction of these medications to prevent volume depletion (Table 2) [63, 64]. Patients should be instructed when acutely unwell (for example, vomiting, diarrhoea, and reduced oral intake) to withhold their SGLT2 inhibitor (part of a sick day management plan) [43].

There has been a recent reported case of AKI secondary to osmotic nephrosis attributed to recent prescription of canagliflozin, postulated to be due to increased tubular osmotic pressure secondary to glucose reabsorption inhibition [65]. The authors of this report recommend consideration of a kidney biopsy in cases of prolonged AKI despite SGLT2 inhibitor discontinuation. Furthermore, the issue of possible hypoxic medullary injury secondary to SGLT2 inhibition, due to increased distal natriuresis augmenting transport workload in the medulla and oxygen consumption, has been raised by Heyman et al. [66]. These authors caution against concomitant administration of agents that could worsen medullary hypoxia, and recommend cessation of SGLT2 inhibitors prior to radiocontrast studies.

Diabetic Ketoacidosis

DKA is a rare but potentially life-threatening adverse effect of SGLT2 inhibitor therapy [67, 68], estimated to occur in approximately one in 1000 SGLT2 users who have type 2 diabetes (although the precise incidence is unknown) [69]. The event rate of SGLT2 inhibitor-associated DKA in the CREDENCE trial was higher compared with the cardiovascular outcome trials (2.2 vs < 1 event per 1000 patient-years) [8, 56]. This may be, at least in part, related to the higher use of insulin at baseline in CREDENCE; all except one of the 12 patients in CREDENCE who developed DKA had concomitant insulin treatment [4, 68]. In contrast, there were no reported DKA events in patients randomised to dapagliflozin in the DAPA-CKD trial; however, this trial included patients with and without diabetes [19]. The higher risk of DKA in insulin-treated patients is pertinent to nephrologists as patients with advanced CKD have relatively limited therapeutic options for the management of type 2 diabetes. SGLT2 inhibitor-associated DKA is commonly referred to as “euglycaemic” DKA as the degree of hyperglycaemia is often lower than expected due to glycosuria [70]. However, in a review of 105 cases of SGLT2 inhibitor-associated DKA, 35% of cases had an admission plasma glucose concentration < 200 mg/dL (11.1 mmol/L) [71]. A more precise term for this adverse effect is “DKA with lower-than-anticipated glucose levels”, as recommended by the American Association of Clinical Endocrinologists and American College of Endocrinology [70]. The duration of SGLT2 inhibitor treatment prior to the onset of DKA is highly variable (0.3–420 days) [69, 71].

With regard to the pathophysiology of DKA, SGLT2 inhibitor use leads to a reduction in plasma insulin concentration and an increase in plasma glucagon concentration [52]. Additionally, free fatty acid suppression post-meal is impaired [52]. This decrease in the insulin-to-glucagon ratio and increase in free fatty acids promotes ketogenesis [52, 72]. SGLT2 inhibitor treatment increases plasma ketone levels, and an elevated ketone level does not necessarily indicate DKA [7275]. SGLT2 inhibitor-associated DKA most frequently occurs in patients with one or more additional risk factor(s) for insulin deficiency and/or ketogenesis (Table 3) [6771]. In a recent Australian retrospective cohort study of SGLT2 inhibitor-associated DKA cases, 22% of patients with presumed type 2 diabetes were subsequently diagnosed as having type 1 diabetes [69]. Fourteen of 37 cases of DKA related to SGLT2 inhibition occurred during hospital admission. Eleven of the 14 patients were fasting due to surgery, and SGLT2 inhibitor therapy was continued during admission in six of these cases. Eleven of the 14 inpatients were on insulin treatment prior to hospitalisation, and insulin was generally ceased prior to the onset of DKA. These findings highlight the need to employ specific strategies to reduce the risk of DKA, including educating patients to temporarily withhold their SGLT2 inhibitor when acutely unwell with reduced oral intake (Table 4) [67, 70, 71].

Table 3.

Risk factors for SGLT2 inhibitor-associated DKA

Type 1 diabetes including latent autoimmune diabetes in adults (patients with presumed type 2 diabetes where there is clinical suspicion of type 1 diabetes should have autoantibodies tested)
Type 2 diabetes with insulin deficiency
Excessive reduction in exogenous insulin dose or insulin cessation
Diabetes due to pancreatic disease
Fasting, including during the perioperative state
Very low carbohydrate diet
Hypovolaemia
Excessive alcohol consumption (daily consumption and/or binge drinking)
Metabolic stress including acute infection, surgery, myocardial infarction, pancreatitis, and intensive exercise

DKA diabetic ketoacidosis, SGLT2 sodium-glucose cotransporter 2

Table 4.

Strategies to reduce the risk of SGLT2 inhibitor-associated DKA

Careful prescription of an SGLT2 inhibitor in light of a patient’s risk factors for DKA (see Table 1)
Reducing a patient’s insulin dose cautiously when commencing an SGLT2 inhibitor, as excessive insulin dose reduction or cessation of insulin therapy can contribute to the risk of DKA
Informing patients about the risk of SGLT2 inhibitor-associated DKA, including when to withhold an SGLT2 inhibitor, including acute illness with reduced oral intake (part of a sick day management plan), symptoms of DKA (nausea, vomiting, abdominal pain, tiredness, rapid breathing), and the need to seek medical attention if symptoms occur (provision of written information is recommended)
Cessation of an SGLT2 inhibitor ≥ 3 days prior to an operation and only recommencing therapy when a patient is eating and drinking normally

DKA diabetic ketoacidosis, SGLT2 sodium-glucose cotransporter 2

Treatment of SGLT2 inhibitor-associated DKA involves rehydration and an insulin-dextrose infusion [70]. A higher rate of intravenous dextrose (10–20%) is often needed to enable sufficient dosage of insulin for resolution of ketoacidosis [67]. An endocrinologist should be involved in the management of DKA and decisions regarding subsequent diabetes pharmacotherapy.

Lower Limb Amputation

The CANVAS Program is the only cardiovascular outcome trial that has shown an increased risk of lower limb amputation with SGLT2 inhibitor therapy compared with placebo [6, 7, 76]. Approximately six versus three participants per 1000 person-years in the canagliflozin versus placebo groups experienced lower limb amputation; 71% of amputations occurred at the level of the toe or metatarsal [6]. Multivariate modelling revealed a number of baseline characteristics that were significantly associated with amputation during follow-up, including male sex, prior amputation, peripheral vascular disease, neuropathy, albuminuria, and higher HbA1c [77]. However, the effect of canagliflozin on amputation risk did not vary according to any baseline characteristic or dose of canagliflozin (100 or 300 mg daily) [77]. Interestingly, there was no difference in the risk of amputation between the canagliflozin and placebo groups in the CREDENCE trial [8]. During the CREDENCE trial, there was a protocol amendment asking investigators to examine patients’ feet and temporarily withhold the study drug if there was any active condition present that might lead to amputation [8].

A cohort study using nationwide health and administrative registers in Sweden and Denmark found that compared with new users of glucagon-like peptide 1 (GLP-1) receptor agonists, new users of SGLT2 inhibitors had an increased risk of lower limb amputation (incidence rate 2.7 vs 1.1 events per 1000 person-years, hazard ratio 2.3) [48]. Ninety-nine per cent of SGLT2 inhibitor users were taking dapagliflozin (61%) or empagliflozin (38%). These results contrast with safety results of the EMPA-REG OUTCOME, DECLARE-TIMI 58, and DAPA-HF trials [7, 9, 76].

In summary, whether there is a definite increase in risk of lower limb amputation with canagliflozin treatment is unclear. Furthermore, the mechanisms underlying such a potential adverse effect are unknown. Postulated mechanisms include volume depletion secondary to diuresis, and an effect on calcium, magnesium, and vitamin D metabolism that may impair foot ulcer healing [78, 79]. Based on available evidence to date, we recommend that clinicians provide education to patients about preventive foot care and perform regular foot screening, as well as avoiding canagliflozin in patients with an acute heightened risk of amputation (as per the CREDENCE protocol—history of amputation within past 12 months, active ulcer, osteomyelitis, gangrene, or critical leg ischaemia within 6 months) [8].

Mineral Metabolism and Fracture

Blau et al. examined the acute effect of canagliflozin on mineral metabolism in healthy adults [80]. Subjects received canagliflozin 300 mg daily or placebo for 5 days, and later crossed over to the other treatment. Canagliflozin administration rapidly increased serum phosphorus, corresponding to an increase in urinary phosphorus reabsorption. Additionally, canagliflozin treatment increased plasma fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH) and reduced 1,25-dihydroxyvitamin D. The differences in mean serum phosphorus and plasma FGF23 between the canagliflozin and placebo groups were no longer significant by day 5. In contrast, differences in 1,25-dihydroxyvitamin D and PTH were still significant at this time point. There was no significant difference in serum calcium, but there was a significant decrease in urinary calcium excretion on day 4. de Jong et al. performed a post hoc analysis of the IMPROVE trial, a randomised, placebo-controlled, crossover trial involving dapagliflozin in patients with type 2 diabetes and albuminuric CKD (eGFR ≥ 45 mL/min/1.73 m2) [81]. Compared with the start of treatment, 6 weeks of dapagliflozin increased serum phosphorus (+  11%), PTH (+ 15%), and FGF23 (+ 20%) and decreased 1,25-dihydroxyvitamin D (− 19%). Importantly, these changes did not correlate with change in eGFR. The increase in serum phosphorus with SGLT2 inhibition is believed to be due to increased sodium in the proximal tubule driving sodium-dependent phosphate reabsorption [82]. This is postulated to increase FGF23, which decreases 1,25-dihydroxyvitamin D, leading to an increase in PTH [80, 83]. In the study by Blau et al., the increase in serum phosphorus correlated with urinary sodium excretion, but not urinary glucose excretion [80]. In severe CKD, the effects of SGLT2 inhibition on urinary glucose and presumably also urinary sodium excretion are attenuated, perhaps resulting in a less marked effect on serum phosphorus. However, more data are needed with regard to patients with stage 4 CKD, where control of hyperphosphataemia can be difficult.

Changes to mineral metabolism secondary to SGLT2 inhibition may be relevant to the heightened risk of fracture evident in the CANVAS Program (15.4 vs 11.9 participants with fracture randomised to canagliflozin vs placebo per 1000 patient-years) [6], although this is the only very large RCT to date with a fracture safety signal. Meta-analyses of RCTs of SGLT2 inhibitors have not demonstrated an increased risk of fractures compared with placebo [84, 85]. The increased risk of fracture with canagliflozin was only seen in one of the two trials that compose the CANVAS Program (CANVAS, not CANVAS-R). Furthermore, there was no difference in risk of fracture between the canagliflozin and placebo groups in CREDENCE [8]. The mean follow-up was longer in CANVAS compared with CANVAS-R (5.7 vs 2.1 years). The median follow-up of CREDENCE was 2.6 years. Interestingly, in a fracture analysis of CANVAS, there was no difference between canagliflozin-treated patients with or without fractures with respect to post-randomisation per cent changes from baseline in serum phosphate [86]. There was a similar fracture incidence in the canagliflozin 100 mg and 300 mg groups. A possible relationship between falls (potentially caused by volume depletion) and fractures cannot be excluded. An RCT of dapagliflozin in patients with stage 3 CKD demonstrated a higher risk of fracture with the SGLT2 inhibitor compared with placebo; seven of the 13 participants randomised to dapagliflozin who sustained a fracture exhibited orthostatic hypotension or had a history of diabetic neuropathy [87].

In summary, meta-analyses and population-based studies of SGLT2 inhibitor therapy have largely not demonstrated an increased risk of fracture [40, 47, 84, 85, 88]. However, given the changes in mineral metabolism and the results of the CANVAS Program described, longer-term data are needed with respect to risk of fracture. This issue is of relevance to the population of patients with CKD, who have or are at risk of CKD–mineral and bone disorder (CKD-MBD).

Safety Considerations of SGLT2 Inhibitor Use During the COVID-19 Pandemic

Patients with comorbidities including diabetes, older age and hypertension are at risk for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [89]. Therefore, it is important that clinicians inform patients taking SGLT2 inhibitors that they should temporarily stop their SGLT2 inhibitor when acutely unwell with reduced oral intake (as part of a sick day management plan), and that patients understand this advice, to try to reduce the risk of DKA and dehydration potentially leading to AKI [90, 91]. Patients admitted to hospital with SARS-CoV-2 infection should have their SGLT2 inhibitor discontinued [90, 91]. In a systematic literature review of cases involving patients with COVID-19 and DKA or combined DKA and hyperglycaemic hyperosmolar syndrome, at least seven of the 110 patients were taking SGLT2 inhibitors. Self-monitoring of plasma ketone levels when DKA is suspected has been recommended [92], although there are cost and possibly test kit availability issues. Additionally, patients must understand how to correctly use a meter that measures plasma ketone levels and be able to seek advice, preferably from a diabetes educator or endocrinologist, as to what actions to take based on their symptoms, plasma glucose, and ketone level.

Conclusion

SGLT2 inhibitors have clinically important cardio-renal benefits, especially for people with type 2 diabetes and CKD, who are at high risk of cardiovascular disease and end-stage kidney disease. Clinicians need to be aware of the potential safety issues with SGLT2 inhibitor therapy in order to try to minimise the occurrence of adverse events, as well as to detect and intervene early if these events occur. Our understanding regarding the safety of these agents is evolving, and longer-term data will provide greater knowledge. Additionally, there is a need for greater  specific data with respect to people with severe CKD.

Declarations

Funding

No funding was received for the preparation of this study.

Conflict of interest

Authors have no conflicts of interest that are directly relevant to the content of this article.

Ethics approval

Not applicable.

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Availability of data and material

Not applicable.

Code availability

Not applicable.

Author contributions

TYM drafted the manuscript. TYM, SLS, ROD, and JRG edited and approved the final manuscript.

References

  • 1.Saran R, Robinson B, Abbott KC, Bragg-Gresham J, Chen X, Gipson D et al. US Renal Data System 2019 Annual data report: epidemiology of kidney disease in the United States. Am J Kidney Dis. 2020;75(1S1):A6–A7. 10.1053/j.ajkd.2019.09.003. [DOI] [PubMed]
  • 2.Branch M, German C, Bertoni A, Yeboah J. Incremental risk of cardiovascular disease and/or chronic kidney disease for future ASCVD and mortality in patients with type 2 diabetes mellitus: ACCORD trial. J Diabetes Compl. 2019;33(7):468–472. doi: 10.1016/j.jdiacomp.2019.04.004. [DOI] [PubMed] [Google Scholar]
  • 3.Rieg T, Vallon V. Development of SGLT1 and SGLT2 inhibitors. Diabetologia. 2018;61(10):2079–2086. doi: 10.1007/s00125-018-4654-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117–2128. doi: 10.1056/NEJMoa1504720. [DOI] [PubMed] [Google Scholar]
  • 5.Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, et al. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med. 2016;375(4):323–334. doi: 10.1056/NEJMoa1515920. [DOI] [PubMed] [Google Scholar]
  • 6.Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377(7):644–657. doi: 10.1056/NEJMoa1611925. [DOI] [PubMed] [Google Scholar]
  • 7.Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380(4):347–357. doi: 10.1056/NEJMoa1812389. [DOI] [PubMed] [Google Scholar]
  • 8.Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380(24):2295–2306. doi: 10.1056/NEJMoa1811744. [DOI] [PubMed] [Google Scholar]
  • 9.McMurray JJV, Solomon SD, Inzucchi SE, Kober L, Kosiborod MN, Martinez FA, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381(21):1995–2008. doi: 10.1056/NEJMoa1911303. [DOI] [PubMed] [Google Scholar]
  • 10.Bakris GL. Major advancements in slowing diabetic kidney disease progression: focus on SGLT2 Inhibitors. Am J Kidney Dis. 2019;74(5):573–575. doi: 10.1053/j.ajkd.2019.05.009. [DOI] [PubMed] [Google Scholar]
  • 11.Inzucchi SE, Kosiborod M, Fitchett D, Wanner C, Hehnke U, Kaspers S, et al. Improvement in cardiovascular outcomes with empagliflozin is independent of glycemic control. Circulation. 2018;138(17):1904–1907. doi: 10.1161/CIRCULATIONAHA.118.035759. [DOI] [PubMed] [Google Scholar]
  • 12.Cooper ME, Inzucchi SE, Zinman B, Hantel S, von Eynatten M, Wanner C, et al. Glucose control and the effect of empagliflozin on kidney outcomes in type 2 diabetes: an analysis from the EMPA-REG OUTCOME Trial. Am J Kidney Dis. 2019;74(5):713–715. doi: 10.1053/j.ajkd.2019.03.432. [DOI] [PubMed] [Google Scholar]
  • 13.Cannon CP, Perkovic V, Agarwal R, Baldassarre J, Bakris G, Charytan DM, et al. Evaluating the effects of canagliflozin on cardiovascular and renal events in patients With type 2 diabetes mellitus and chronic kidney disease according to baseline HbA1c, including those with HbA1c < 7%: results from the CREDENCE trial. Circulation. 2020;141(5):407–410. doi: 10.1161/CIRCULATIONAHA.119.044359. [DOI] [PubMed] [Google Scholar]
  • 14.Macha S, Mattheus M, Halabi A, Pinnetti S, Woerle HJ, Broedl UC. Pharmacokinetics, pharmacodynamics and safety of empagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, in subjects with renal impairment. Diabetes Obes Metab. 2014;16(3):215–222. doi: 10.1111/dom.12182. [DOI] [PubMed] [Google Scholar]
  • 15.Kasichayanula S, Liu X, Pe Benito M, Yao M, Pfister M, LaCreta FP, et al. The influence of kidney function on dapagliflozin exposure, metabolism and pharmacodynamics in healthy subjects and in patients with type 2 diabetes mellitus. Br J Clin Pharmacol. 2013;76(3):432–444. doi: 10.1111/bcp.12056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Devineni D, Curtin CR, Marbury TC, Smith W, Vaccaro N, Wexler D et al. Effect of hepatic or renal impairment on the pharmacokinetics of canagliflozin, a sodium glucose co-transporter 2 inhibitor. Clin Ther. 2015;37(3):610–28 e4. 10.1016/j.clinthera.2014.12.013. [DOI] [PubMed]
  • 17.Sahasrabudhe V, Terra SG, Hickman A, Saur D, Shi H, O’Gorman M, et al. The effect of renal impairment on the pharmacokinetics and pharmacodynamics of ertugliflozin in subjects with type 2 diabetes mellitus. J Clin Pharmacol. 2017;57(11):1432–1443. doi: 10.1002/jcph.955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med. 2020 doi: 10.1056/NEJMoa2022190. [DOI] [PubMed] [Google Scholar]
  • 19.European Society of Cardiology Press Office: DAPA-CKD trial meets primary endpoint in patients with chronic kidney disease. 2020. https://www.escardio.org/The-ESC/Press-Office/Press-releases/DAPA. Accessed 8 Sep 2020.
  • 20.Cowie MR, Fisher M. SGLT2 inhibitors: mechanisms of cardiovascular benefit beyond glycaemic control. Nat Rev Cardiol. 2020 doi: 10.1038/s41569-020-0406-8. [DOI] [PubMed] [Google Scholar]
  • 21.Tuttle KR, Cherney DZ, Diabetic Kidney Disease Task Force of the American Society of N. Sodium glucose cotransporter 2 inhibition heralds a call-to-action for diabetic kidney disease. Clin J Am Soc Nephrol. 2020;15(2):285–8. 10.2215/cjn.07730719. [DOI] [PMC free article] [PubMed]
  • 22.Neumiller JJ, Kalyani RR. How does CREDENCE inform best use of SGLT2 inhibitors in CKD? Clin J Am Soc Nephrol. 2019;14(11):1667–1669. doi: 10.2215/CJN.05340419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cannon CP, McGuire DK, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, et al. Design and baseline characteristics of the eValuation of ERTugliflozin effIcacy and Safety CardioVascular outcomes trial (VERTIS-CV) Am Heart J. 2018;206:11–23. doi: 10.1016/j.ahj.2018.08.016. [DOI] [PubMed] [Google Scholar]
  • 24.Heerspink HJL, Stefansson BV, Chertow GM, Correa-Rotter R, Greene T, Hou FF, et al. Rationale and protocol of the Dapagliflozin And Prevention of Adverse outcomes in Chronic Kidney Disease (DAPA-CKD) randomized controlled trial. Nephrol Dial Transpl. 2020;35(2):274–282. doi: 10.1093/ndt/gfz290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wanner C, Lachin JM, Inzucchi SE, Fitchett D, Mattheus M, George J, et al. Empagliflozin and clinical outcomes in patients with type 2 diabetes mellitus, established cardiovascular disease, and chronic kidney disease. Circulation. 2018;137(2):119–129. doi: 10.1161/CIRCULATIONAHA.117.028268. [DOI] [PubMed] [Google Scholar]
  • 26.Neuen BL, Ohkuma T, Neal B, Matthews DR, de Zeeuw D, Mahaffey KW, et al. Cardiovascular and renal outcomes with canagliflozin according to baseline kidney function. Circulation. 2018;138(15):1537–1550. doi: 10.1161/CIRCULATIONAHA.118.035901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cahn A, Raz I, Bonaca M, Mosenzon O, Murphy SA, Yanuv I, et al. Safety of dapagliflozin in a broad population of patients with type 2 diabetes: analyses from the DECLARE-TIMI 58 study. Diabetes Obes Metab. 2020;22(8):1357–1368. doi: 10.1111/dom.14041. [DOI] [PubMed] [Google Scholar]
  • 28.Jardine MJ, Zhou Z, Mahaffey KW, Oshima M, Agarwal R, Bakris G, et al. Renal, cardiovascular, and safety outcomes of canagliflozin by baseline kidney function: a secondary analysis of the CREDENCE randomized trial. J Am Soc Nephrol. 2020;31(5):1128–1139. doi: 10.1681/ASN.2019111168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Janssen Pharmaceuticals, Inc. Invokana (canagliflozin) tablets—full prescribing information. 2020. http://www.janssenlabels.com/package-insert/product-monograph/prescribing-information/INVOKANA-pi.pdf. Accessed 11 Sep 2020.
  • 30.Boehringer Ingelheim Pty Limited. Australian Product Information—Jardiance empagliflozin film-coated tablets. 2020. https://www.ebs.tga.gov.au/ebs/picmi/picmirepository.nsf/pdf?OpenAgent&id=CP-2014-PI-01783-1&d=202009211016933&d=202009221016933. Accessed 11 Sep 2020.
  • 31.AstraZeneca Pty Ltd. Australian Product Information Forxiga. 2020. https://www.ebs.tga.gov.au/ebs/picmi/picmirepository.nsf/pdf?OpenAgent&id=CP-2012-PI-02861-1&d=202009221016933. Accessed 11 Sep 2020.
  • 32.Merck Sharp & Dohme (Australia) Pty Limited. Australian Product Information—Steglatro (Ertugliflozin). 2019. https://www.ebs.tga.gov.au/ebs/picmi/picmirepository.nsf/pdf?OpenAgent&id=CP-2018-PI-01724-1&d=202009221016933. Accessed 11 Sep 2020.
  • 33.Rodrigues CF, Rodrigues ME, Henriques M. Candida sp. infections in patients with diabetes mellitus. J Clin Med. 2019. 10.3390/jcm8010076. [DOI] [PMC free article] [PubMed]
  • 34.Geerlings S, Fonseca V, Castro-Diaz D, List J, Parikh S. Genital and urinary tract infections in diabetes: impact of pharmacologically-induced glucosuria. Diabetes Res Clin Pract. 2014;103(3):373–381. doi: 10.1016/j.diabres.2013.12.052. [DOI] [PubMed] [Google Scholar]
  • 35.Fralick M, MacFadden DR. A hypothesis for why sodium glucose co-transporter 2 inhibitors have been found to cause genital infection, but not urinary tract infection. Diabetes Obes Metab. 2020;22(5):755–758. doi: 10.1111/dom.13959. [DOI] [PubMed] [Google Scholar]
  • 36.Liu J, Li L, Li S, Jia P, Deng K, Chen W, et al. Effects of SGLT2 inhibitors on UTIs and genital infections in type 2 diabetes mellitus: a systematic review and meta-analysis. Sci Rep. 2017;7(1):2824. doi: 10.1038/s41598-017-02733-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Dave CV, Schneeweiss S, Patorno E. Comparative risk of genital infections associated with sodium-glucose co-transporter-2 inhibitors. Diabetes Obes Metab. 2019;21(2):434–438. doi: 10.1111/dom.13531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lega IC, Bronskill SE, Campitelli MA, Guan J, Stall NM, Lam K, et al. Sodium glucose cotransporter 2 inhibitors and risk of genital mycotic and urinary tract infection: a population-based study of older women and men with diabetes. Diabetes Obes Metab. 2019;21(11):2394–2404. doi: 10.1111/dom.13820. [DOI] [PubMed] [Google Scholar]
  • 39.McGovern AP, Hogg M, Shields BM, Sattar NA, Holman RR, Pearson ER et al. Risk factors for genital infections in people initiating SGLT2 inhibitors and their impact on discontinuation. BMJ Open Diabetes Res Care. 2020;8(1). 10.1136/bmjdrc-2020-001238. [DOI] [PMC free article] [PubMed]
  • 40.Toyama T, Neuen BL, Jun M, Ohkuma T, Neal B, Jardine MJ, et al. Effect of SGLT2 inhibitors on cardiovascular, renal and safety outcomes in patients with type 2 diabetes mellitus and chronic kidney disease: a systematic review and meta-analysis. Diabetes Obes Metab. 2019;21(5):1237–1250. doi: 10.1111/dom.13648. [DOI] [PubMed] [Google Scholar]
  • 41.Vardeny O, Vaduganathan M. Practical guide to prescribing sodium-glucose cotransporter 2 inhibitors for cardiologists. JACC Heart Fail. 2019;7(2):169–172. doi: 10.1016/j.jchf.2018.11.013. [DOI] [PubMed] [Google Scholar]
  • 42.Lupsa BC, Inzucchi SE. Use of SGLT2 inhibitors in type 2 diabetes: weighing the risks and benefits. Diabetologia. 2018;61(10):2118–2125. doi: 10.1007/s00125-018-4663-6. [DOI] [PubMed] [Google Scholar]
  • 43.Fitchett D. A safety update on sodium glucose co-transporter 2 inhibitors. Diabetes Obes Metab. 2019;21(suppl 2):34–42. doi: 10.1111/dom.13611. [DOI] [PubMed] [Google Scholar]
  • 44.U.S. Food and Drug Administration: FDA warns about the rare occurrences of a serious infection of the genital area with SGLT2 inhibitors for diabetes. 2018. https://www.fda.gov/drugs/drug-safety-and-availability/fda-warns-about-rare-occurrences-serious-infection-genital-area-sglt2-inhibitors-diabetes. Accessed 18 Oct 2018.
  • 45.Bersoff-Matcha SJ, Chamberlain C, Cao C, Kortepeter C, Chong WH. Fournier gangrene associated with sodium-glucose cotransporter-2 inhibitors: a review of spontaneous postmarketing cases. Ann Intern Med. 2019;170(11):764–769. doi: 10.7326/M19-0085. [DOI] [PubMed] [Google Scholar]
  • 46.U.S. Food and Drug Administration: FDA Drug Safety Communication: FDA revises labels of SGLT2 inhibitors for diabetes to include warnings about too much acid in the blood and serious urinary tract infections. 2015. https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-fda-revises-labels-sglt2-inhibitors-diabetes-include-warnings-about. Accessed 13 July 2018.
  • 47.Donnan JR, Grandy CA, Chibrikov E, Marra CA, Aubrey-Bassler K, Johnston K, et al. Comparative safety of the sodium glucose co-transporter 2 (SGLT2) inhibitors: a systematic review and meta-analysis. BMJ Open. 2019;9(1):e022577. doi: 10.1136/bmjopen-2018-022577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ueda P, Svanstrom H, Melbye M, Eliasson B, Svensson AM, Franzen S, et al. Sodium glucose cotransporter 2 inhibitors and risk of serious adverse events: nationwide register based cohort study. BMJ. 2018;363:k4365. doi: 10.1136/bmj.k4365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kumbhani DJ. Evaluation of Ertugliflozin Effcacy and Safety Cardiovascular Outcome Trial—VERTIS CV. American College of Cardiology. 2020. https://www.acc.org/latest-in-cardiology/clinical-trials/2020/06/16/11/24/vertis. Accessed 29 June 2020.
  • 50.Halden TAS, Kvitne KE, Midtvedt K, Rajakumar L, Robertsen I, Brox J, et al. Efficacy and safety of empagliflozin in renal transplant recipients with posttransplant diabetes mellitus. Diabetes Care. 2019;42(6):1067–1074. doi: 10.2337/dc19-0093. [DOI] [PubMed] [Google Scholar]
  • 51.Ferrannini E. Sodium-glucose co-transporters and their inhibition: clinical physiology. Cell Metab. 2017;26(1):27–38. doi: 10.1016/j.cmet.2017.04.011. [DOI] [PubMed] [Google Scholar]
  • 52.Ferrannini E, Muscelli E, Frascerra S, Baldi S, Mari A, Heise T, et al. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J Clin Invest. 2014;124(2):499–508. doi: 10.1172/JCI72227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zaccardi F, Webb DR, Htike ZZ, Youssef D, Khunti K, Davies MJ. Efficacy and safety of sodium-glucose co-transporter-2 inhibitors in type 2 diabetes mellitus: systematic review and network meta-analysis. Diabetes Obes Metab. 2016;18(8):783–794. doi: 10.1111/dom.12670. [DOI] [PubMed] [Google Scholar]
  • 54.Milder TY, Stocker SL, Samocha-Bonet D, Day RO, Greenfield JR. Sodium-glucose cotransporter 2 inhibitors for type 2 diabetes-cardiovascular and renal benefits in patients with chronic kidney disease. Eur J Clin Pharmacol. 2019;75(11):1481–1490. doi: 10.1007/s00228-019-02732-y. [DOI] [PubMed] [Google Scholar]
  • 55.U.S. Food and Drug Administration: FDA Drug Safety Communication: FDA strengthens kidney warnings for diabetes medicines canagliflozin (Invokana, Invokamet) and dapagliflozin (Farxiga, Xigduo XR). 2016. https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-fda-strengthens-kidney-warnings-diabetes-medicines-canagliflozin. Accessed 18 June 2018.
  • 56.Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet. 2019;393(10166):31–39. doi: 10.1016/S0140-6736(18)32590-X. [DOI] [PubMed] [Google Scholar]
  • 57.Neuen BL, Young T, Heerspink HJL, Neal B, Perkovic V, Billot L, et al. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol. 2019;7(11):845–854. doi: 10.1016/S2213-8587(19)30256-6. [DOI] [PubMed] [Google Scholar]
  • 58.Cherney DZ, Kanbay M, Lovshin JA. Renal physiology of glucose handling and therapeutic implications. Nephrol Dial Transplant. 2020;35(suppl 1):i3–i12. doi: 10.1093/ndt/gfz230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cherney DZ, Perkins BA, Soleymanlou N, Maione M, Lai V, Lee A, et al. Renal hemodynamic effect of sodium-glucose cotransporter 2 inhibition in patients with type 1 diabetes mellitus. Circulation. 2014;129(5):587–597. doi: 10.1161/CIRCULATIONAHA.113.005081. [DOI] [PubMed] [Google Scholar]
  • 60.van Bommel EJM, Muskiet MHA, van Baar MJB, Tonneijck L, Smits MM, Emanuel AL, et al. The renal hemodynamic effects of the SGLT2 inhibitor dapagliflozin are caused by post-glomerular vasodilatation rather than pre-glomerular vasoconstriction in metformin-treated patients with type 2 diabetes in the randomized, double-blind RED trial. Kidney Int. 2020;97(1):202–212. doi: 10.1016/j.kint.2019.09.013. [DOI] [PubMed] [Google Scholar]
  • 61.Bjornstad P, Nelson RG, Pavkov ME. Do sodium-glucose cotransporter-2 inhibitors affect renal hemodynamics by different mechanisms in type 1 and type 2 diabetes? Kidney Int. 2020;97(1):31–33. doi: 10.1016/j.kint.2019.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Mayer GJ, Wanner C, Weir MR, Inzucchi SE, Koitka-Weber A, Hantel S, et al. Analysis from the EMPA-REG OUTCOME((R)) trial indicates empagliflozin may assist in preventing the progression of chronic kidney disease in patients with type 2 diabetes irrespective of medications that alter intrarenal hemodynamics. Kidney Int. 2019;96(2):489–504. doi: 10.1016/j.kint.2019.02.033. [DOI] [PubMed] [Google Scholar]
  • 63.Neuen BL, Jardine MJ, Perkovic V. Sodium-glucose cotransporter 2 inhibition: which patient with chronic kidney disease should be treated in the future? Nephrol Dial Transplant. 2020;35(suppl 1):i48–i55. doi: 10.1093/ndt/gfz252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Triantafylidis LK, Hawley CE, Fagbote C, Li J, Genovese N, Paik JM. A pilot study embedding clinical pharmacists within an interprofessional nephrology clinic for the initiation and monitoring of empagliflozin in diabetic kidney disease. J Pharm Pract. 2019. 10.1177/0897190019876499. [DOI] [PMC free article] [PubMed]
  • 65.Phadke G, Kaushal A, Tolan DR, Hahn K, Jensen T, Bjornstad P, et al. Osmotic nephrosis and acute kidney injury associated with SGLT2 inhibitor use: a case report. Am J Kidney Dis. 2020;76(1):144–147. doi: 10.1053/j.ajkd.2020.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Heyman SN, Khamaisi M, Rosen S, Rosenberger C, Abassi Z. Potential hypoxic renal injury in patients with diabetes on SGLT2 inhibitors: caution regarding concomitant use of NSAIDs and iodinated contrast media. Diabetes Care. 2017;40(4):e40–e41. doi: 10.2337/dc16-2200. [DOI] [PubMed] [Google Scholar]
  • 67.Isaacs M, Tonks KT, Greenfield JR. Euglycaemic diabetic ketoacidosis in patients using sodium-glucose co-transporter 2 inhibitors. Intern Med J. 2017;47(6):701–704. doi: 10.1111/imj.13442. [DOI] [PubMed] [Google Scholar]
  • 68.Meyer EJ, Gabb G, Jesudason D. SGLT2 inhibitor-associated euglycemic diabetic ketoacidosis: a South Australian clinical case series and Australian Spontaneous Adverse Event Notifications. Diabetes Care. 2018;41(4):e47–e49. doi: 10.2337/dc17-1721. [DOI] [PubMed] [Google Scholar]
  • 69.Hamblin PS, Wong R, Ekinci EI, Fourlanos S, Shah S, Jones AR, et al. SGLT2 inhibitors increase the risk of diabetic ketoacidosis developing in the community and during hospital admission. J Clin Endocrinol Metab. 2019;104(8):3077–3087. doi: 10.1210/jc.2019-00139. [DOI] [PubMed] [Google Scholar]
  • 70.Handelsman Y, Henry RR, Bloomgarden ZT, Dagogo-Jack S, DeFronzo RA, Einhorn D, et al. American Association of Clinical Endocrinologists and American College of Endocrinology position statement on the association of SGLT-2 inhibitors and diabetic ketoacidosis. Endocr Pract. 2016;22(6):753–762. doi: 10.4158/EP161292.PS. [DOI] [PubMed] [Google Scholar]
  • 71.Bonora BM, Avogaro A, Fadini GP. Sodium-glucose co-transporter-2 inhibitors and diabetic ketoacidosis: an updated review of the literature. Diabetes Obes Metab. 2018;20(1):25–33. doi: 10.1111/dom.13012. [DOI] [PubMed] [Google Scholar]
  • 72.Ferrannini E, Baldi S, Frascerra S, Astiarraga B, Heise T, Bizzotto R, et al. Shift to fatty substrate utilization in response to sodium-glucose cotransporter 2 inhibition in subjects without diabetes and patients with type 2 diabetes. Diabetes. 2016;65(5):1190–1195. doi: 10.2337/db15-1356. [DOI] [PubMed] [Google Scholar]
  • 73.Polidori D, Iijima H, Goda M, Maruyama N, Inagaki N, Crawford PA. Intra- and inter-subject variability for increases in serum ketone bodies in patients with type 2 diabetes treated with the sodium glucose co-transporter 2 inhibitor canagliflozin. Diabetes Obes Metab. 2018;20(5):1321–1326. doi: 10.1111/dom.13224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ferrannini E, Baldi S, Frias JP, Guja C, Hardy E, Repetto E, et al. Hormone-substrate changes with exenatide plus dapagliflozin versus each drug alone: the randomized, active-controlled DURATION-8 study. Diabetes Obes Metab. 2020;22(1):99–106. doi: 10.1111/dom.13870. [DOI] [PubMed] [Google Scholar]
  • 75.Sato Y, Nunoi K, Kaku K, Yoshida A, Suganami H. Basal insulin secretion capacity predicts the initial response and maximum levels of beta-hydroxybutyrate during therapy with the sodium-glucose co-transporter-2 inhibitor tofogliflozin, in relation to weight loss. Diabetes Obes Metab. 2020;22(2):222–230. doi: 10.1111/dom.13890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Inzucchi SE, Iliev H, Pfarr E, Zinman B. Empagliflozin and assessment of lower-limb amputations in the EMPA-REG OUTCOME Trial. Diabetes Care. 2018;41(1):e4–e5. doi: 10.2337/dc17-1551. [DOI] [PubMed] [Google Scholar]
  • 77.Matthews DR, Li Q, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, et al. Effects of canagliflozin on amputation risk in type 2 diabetes: the CANVAS Program. Diabetologia. 2019;62(6):926–938. doi: 10.1007/s00125-019-4839-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Potier L, Roussel R, Velho G, Saulnier PJ, Bumbu A, Matar O, et al. Lower limb events in individuals with type 2 diabetes: evidence for an increased risk associated with diuretic use. Diabetologia. 2019;62(6):939–947. doi: 10.1007/s00125-019-4835-z. [DOI] [PubMed] [Google Scholar]
  • 79.Monteiro-Soares M, Ribeiro-Vaz I, Boyko EJ. Canagliflozin should be prescribed with caution to individuals with type 2 diabetes and high risk of amputation. Diabetologia. 2019;62(6):900–904. doi: 10.1007/s00125-019-4861-x. [DOI] [PubMed] [Google Scholar]
  • 80.Blau JE, Bauman V, Conway EM, Piaggi P, Walter MF, Wright EC et al. Canagliflozin triggers the FGF23/1,25-dihydroxyvitamin D/PTH axis in healthy volunteers in a randomized crossover study. JCI Insight. 2018. 10.1172/jci.insight.99123. [DOI] [PMC free article] [PubMed]
  • 81.de Jong MA, Petrykiv SI, Laverman GD, van Herwaarden AE, de Zeeuw D, Bakker SJL, et al. Effects of dapagliflozin on circulating markers of phosphate homeostasis. Clin J Am Soc Nephrol. 2019;14(1):66–73. doi: 10.2215/CJN.04530418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Vinke JSJ, Heerspink HJL, de Borst MH. Effects of sodium glucose cotransporter 2 inhibitors on mineral metabolism in type 2 diabetes mellitus. Curr Opin Nephrol Hypertens. 2019;28(4):321–327. doi: 10.1097/MNH.0000000000000505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Edmonston D, Wolf M. Good guys, bad guys, guesses, and near misses in nephrology. Clin J Am Soc Nephrol. 2019;14(1):7–9. doi: 10.2215/CJN.13801118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Li X, Li T, Cheng Y, Lu Y, Xue M, Xu L, et al. Effects of SGLT2 inhibitors on fractures and bone mineral density in type 2 diabetes: an updated meta-analysis. Diabetes Metab Res Rev. 2019;35(7):e3170. doi: 10.1002/dmrr.3170. [DOI] [PubMed] [Google Scholar]
  • 85.Cheng L, Li YY, Hu W, Bai F, Hao HR, Yu WN, et al. Risk of bone fracture associated with sodium-glucose cotransporter-2 inhibitor treatment: a meta-analysis of randomized controlled trials. Diabetes Metab. 2019;45(5):436–445. doi: 10.1016/j.diabet.2019.01.010. [DOI] [PubMed] [Google Scholar]
  • 86.Watts NB, Bilezikian JP, Usiskin K, Edwards R, Desai M, Law G, et al. Effects of canagliflozin on fracture risk in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab. 2016;101(1):157–166. doi: 10.1210/jc.2015-3167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kohan DE, Fioretto P, Tang W, List JF. Long-term study of patients with type 2 diabetes and moderate renal impairment shows that dapagliflozin reduces weight and blood pressure but does not improve glycemic control. Kidney Int. 2014;85(4):962–971. doi: 10.1038/ki.2013.356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hidayat K, Du X, Shi BM. Risk of fracture with dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, or sodium-glucose cotransporter-2 inhibitors in real-world use: systematic review and meta-analysis of observational studies. Osteoporos Int. 2019;30(10):1923–1940. doi: 10.1007/s00198-019-04968-x. [DOI] [PubMed] [Google Scholar]
  • 89.Guan WJ, Liang WH, Zhao Y, Liang HR, Chen ZS, Li YM et al. Comorbidity and its impact on 1590 patients with COVID-19 in China: a nationwide analysis. Eur Respir J. 2020. 10.1183/13993003.00547-2020. [DOI] [PMC free article] [PubMed]
  • 90.Ceriello A, Standl E, Catrinoiu D, Itzhak B, Lalic NM, Rahelic D, et al. Issues of cardiovascular risk management in people with diabetes in the COVID-19 era. Diabetes Care. 2020 doi: 10.2337/dc20-0941. [DOI] [PubMed] [Google Scholar]
  • 91.Bornstein SR, Rubino F, Khunti K, Mingrone G, Hopkins D, Birkenfeld AL, et al. Practical recommendations for the management of diabetes in patients with COVID-19. Lancet Diabetes Endocrinol. 2020;8(6):546–550. doi: 10.1016/S2213-8587(20)30152-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gorriz JL, Navarro-Gonzalez JF, Ortiz A, Vergara A, Nunez J, Jacobs-Cacha C, et al. Sodium-glucose cotransporter 2 inhibition: towards an indication to treat diabetic kidney disease. Nephrol Dial Transplant. 2020;35(suppl 1):i13–i23. doi: 10.1093/ndt/gfz237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Biesenbach G, Raml A, Schmekal B, Eichbauer-Sturm G. Decreased insulin requirement in relation to GFR in nephropathic type 1 and insulin-treated type 2 diabetic patients. Diabet Med. 2003;20(8):642–645. doi: 10.1046/j.1464-5491.2003.01025.x. [DOI] [PubMed] [Google Scholar]

Articles from Drug Safety are provided here courtesy of Nature Publishing Group

RESOURCES