You are on page 1of 8

Euglycemic Ketoacidosis as a Complication of SGLT2

Inhibitor Therapy
Biff F. Palmer1 and Deborah J. Clegg2

Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors are drugs designed to lower plasma glucose concentration by
inhibiting Na1-glucose–coupled transport in the proximal tubule. Clinical trials demonstrate these drugs have 1
Division of
favorable effects on cardiovascular outcomes to include slowing the progression of CKD. Although most patients Nephrology,
tolerate these drugs, a potential complication is development of ketoacidosis, often with a normal or only a Department of
Medicine, University of
minimally elevated plasma glucose concentration. Inhibition of sodium-glucose cotransporter-2 in the proximal
Texas Southwestern
tubule alters kidney ATP turnover so that filtered ketoacids are preferentially excreted as Na1 or K1 salts, leading Medical Center, Dallas,
to indirect loss of bicarbonate from the body and systemic acidosis under conditions of increased ketogenesis. Texas
2
Risk factors include reductions in insulin dose, increased insulin demand, metabolic stress, low carbohydrate Associate Dean for
intake, women, and latent autoimmune diabetes of adulthood. The lack of hyperglycemia and nonspecific Research, College of
Nursing and Health
symptoms of ketoacidosis can lead to delays in diagnosis. Treatment strategies and various precautions are Professionals, Drexel
discussed that can decrease the likelihood of this complication. University,
CJASN 16: 1284–1291, 2021. doi: https://doi.org/10.2215/CJN.17621120 Philadelphia,
Pennsylvania

Introduction inhibitors were used in combination with insulin in Correspondence: Dr.


Underappreciated is the role the kidney plays in ketoa- Biff F. Palmer,
patients with type 1 diabetes mellitus (7–9). In the Kid- Department of Internal
cidosis defined as a primary decrease in plasma ney Disease: Improving Global Outcomes 2020 clinical Medicine, University of
bicarbonate concentration resulting from increased practice guideline for management of patients with dia- Texas Southwestern
ketoacids. Diabetic ketoacidosis (DKA) is a common betes and CKD, the risk of this disorder is described as Medical Center, 5323
cause of this acid-base disturbance and is accompanied Harry Hines Boulevard,
generally a rare event in patients with type 2 diabetes
Dallas, TX 75390.
by plasma glucose concentration .250 mg/dl, which is mellitus (10). Reports examining postmarketing use Email: biff.palmer@
a criterion for diagnosis. There are patients with diabe- suggest that the frequency of this disorder is much utsouthwestern.edu
tes mellitus who develop ketoacidosis with less severe greater in real-world practice. An analysis of the FDA
hyperglycemia, often ,200 mg/dl. This form of ketoa- Adverse Event Reporting System found a seven-fold
cidosis is euglycemic ketoacidosis and is a recognized higher risk of acidosis in treated patients with type 2
complication of sodium-glucose cotransporter-2 inhibi- diabetes mellitus when compared with dipeptidyl
tors (SGLT2is). There are new data about the benefits of peptidase-4 inhibitor therapy (11). This risk is approxi-
SGLT2i in individuals with kidney disorders; therefore, mately two- to four-fold greater than the numerical
here we remind the clinician of alterations in kidney imbalance reported in clinical trials of canagliflozin
function induced by these drugs that increase the risk and empagliflozin. Of those with ketoacidosis, euglyce-
of ketoacidosis. We highlight unique risk factors and mia was present in 71% of the patients. More recently, a
treatments of this disorder in individuals with kidney three-fold increase in the risk of DKA was found in a
disease. population cohort study of over 350,000 adults with
type 2 diabetes mellitus compared with those treated
with dipeptidyl peptidase-4 inhibitor (12). Importantly,
fatality rates of this complication with SGLT2i therapy
Risk of Ketoacidosis with Sodium-Glucose
are greater when compared with all patients with
Cotransporter-2 Inhibitor
SGLT2is are drugs developed to reduce blood glu- DKA (13).
cose levels in patients with diabetes mellitus. Random-
ized controlled trials demonstrate risk reductions for
myocardial infarction, heart failure, kidney disease pro- Increased Ketogenesis with Sodium-Glucose
gression, and cardiovascular mortality, which likely Cotransporter-2 Inhibitor
will increase their use (1–5). In 2015, the Food and SGLT2i lowers plasma glucose by pharmacologically
Drug Administration (FDA) issued a safety warning forcing excretion of glucose into the urine. The sodium-
on the risk of DKA (6). The frequency of ketoacidosis glucose cotransporter-2 (SGLT2) is located in the early
in clinical trials was small in patients with type 2 diabe- (S1) proximal tubule and is responsible for reabsorption
tes mellitus, but occurred in 4%–6% of patients when of 80%–90% of filtered glucose (14). SGLT1 is located in

1284 Copyright © 2021 by the American Society of Nephrology www.cjasn.org Vol 16 August, 2021
CJASN 16: 1284–1291, August, 2021 Euglycemic Ketoacidosis, Palmer and Clegg 1285

the more distal part of the proximal tubule (S2/S3) and is Sodium-Glucose Cotransporter-2 Inhibitor Effects on
responsible for the reabsorption of the remaining 10%–20% Kidney Function Predisposing to Ketoacidosis
of filtered glucose. Inhibition of these transporters reduces Although changes in insulin and glucagon levels are cen-
the transport maximum for glucose reabsorption, leading tral to increased production of ketoacids, SGLT2is further
to glycosuria and reductions in plasma glucose concentra- increase the risk of overt ketoacidosis by altering ATP turn-
tions. Decreased plasma glucose concentration signals a fall over in the proximal tubule (Figure 1). Na1-coupled glucose
in insulin levels and causes plasma glucagon levels to reabsorption is an example of secondary active transport
increase due to a paracrine effect induced by diminished where ATP hydrolysis fuels activity of the basolateral
inhibition of insulin on glucagon release in the pancreas. In Na1-K1-ATPase to maintain a low intracellular Na1 concen-
addition, inhibition of SGLT2 on pancreatic a-cells triggers tration and secondarily provide a favorable inward gradient
release of glucagon. Glucagon further suppresses insulin for Na1-coupled glucose transport across the apical mem-
by promoting hepatic release of kisspeptin-1 that, in turn, brane. This pathway normally reabsorbs approximately
suppresses glucose-stimulated insulin secretion (15). The 4%–6% of total filtered Na1 (22,23). In patients with diabetes
reduction in plasma insulin and glucose levels decreases mellitus, the threshold for glucose excretion is increased
carbohydrate oxidation and shifts metabolism toward from 150 to 250 mg/dl due to glucose-mediated hypertrophy
lipid oxidation fueled by increased circulating fatty acids of the proximal tubule and increased expression of SGLT2
and lipolysis. Changes in fuel preference are detected by (24,25). It is estimated these changes lead to a doubling in
measurable drops in the respiratory quotient following initi- percentage of filtered Na1 reabsorbed by this segment and
ation of SGLT2i therapy (16). By way of background, the a 50% increase in daily kidney ATP turnover (22,26).
respiratory quotient is a dimensionless number used in The energy required to fuel Na1-K1-ATPase pump
calculations of basal metabolic rate when estimated from activity to maintain a low intracellular Na1 concentration
carbon dioxide production. Greater amounts of fatty acid decreases following pharmacologic inhibition of SGLT2. In
metabolism lower the quotient toward 0.7, whereas a shift addition to decreasing Na1 entry into the cell coupled to glu-
cose, SGLT2is also decrease Na1 entry by inhibiting Na1-H1
to carbohydrate metabolism raises the quotient to values
antiporter (NHE3) activity. Studies in rodents show SGLT2
more than one.
and NHE3 colocalize in the proximal tubule brush border
SGLT2i causes daily glucose losses of 50–100 g corre-
membrane (27). Inhibition of SGLT2 decreases activity of
sponding to 200–400 kcal/d, which represent up to half of
NHE3, and the natriuretic effect of SGLT2i is diminished in
estimated carbohydrate intake in men and even higher
models where NHE3 is knocked down (28). The estimated
amounts in women (17). SGLT2i therapy leads to weight
daily kidney ATP turnover decreases by approximately
loss in both rodents and humans in association with reduc-
50% in patients with diabetes mellitus treated with SGLT2i
tions in visceral and subcutaneous fat mass as measured by
(26). Because ATP is not stored, reduced consumption
dual-energy x-ray absorptiometry (18). In rodent models,
will secondarily decrease pathways that normally generate
SGLT2is increase energy expenditure with activation and
ATP (29). Because metabolism of glutamine to NH41 and
recruitment of brown and beige adipocytes, respectively
HCO32 in the proximal tubule results in an obligatory
(19). Caloric deficits due to glucose loss in the urine com-
production of ATP, ammoniagenesis decreases following
bined with enhanced lipid oxidation and increases in energy SGLT2i therapy. A similar reduction in ammoniagenesis
expenditure account for reductions in fat mass (19,20). occurs when cellular ATP utilization is lowered by decreas-
The combination of increased lipolysis and a reduced ing the filtered Na1 or directly inhibiting Na1-K1-ATPase
insulin-glucagon ratio leads to increased hepatic production pump activity in the proximal tubule, supporting the notion
of ketoacids. Increased production of acetyl-CoA from fatty that the rate of ATP utilization places an upper limit on
acid b-oxidation is preferentially directed toward ketoacid ammonia production in the kidney (30).
production because glucagon stimulates and insulin inhibits Inhibition of ammonia synthesis indirectly predisposes to
the rate-limiting enzyme for this pathway (hydroxylmethyl- HCO32 loss from the body and development of metabolic
glutaryl-CoA). This pattern is similar to what occurs with acidosis. The production of b-hydroxybutyric acid consumes
fasting, except changes following SGLT2i are more rapid. HCO32 and generates Na-b-hydroxybutyrate and H2CO3,
In one study, fasting plasma ketone concentrations increased the latter of which quickly disassociates into CO2 (exhaled
four-fold over baseline in the first 2 weeks of therapy with by the lungs) and water. When b-hydroxybutyrate is
dapaglifozin (16). This shift to increased fat oxidation and excreted in the urine coupled to NH41, there is no net change
ketogenesis also occurs in SGLT2i-treated individuals with- in acid-base balance because production of NH41 from glu-
out diabetes mellitus, but to a lesser extent (21). Despite mea- tamine generates HCO32. By contrast, urinary excretion of
surable increases in plasma ketone levels in treated patients, b-hydroxybutyrate as either an Na1 or K1 salt is the equiv-
development of ketoacidosis is unusual. Those with the alent to losing HCO32 from the body and will generate
greatest increase in b-hydroxybutyrate levels have lower metabolic acidosis (31,32). The inhibitory effect on ammonia
fasting and postmeal insulin levels and impaired b-cell synthesis by SGLT2i favors excretion of Na1- and/or K1-
insulin sensitivity when compared with subjects in the lower coupled b-hydroxybutyrate in the urine.
quartiles, suggesting the amount of endogenous insulin SGLT2i also suppresses ammoniagenesis by causing a
secretion reserve may factor into the magnitude of ketogenic shift toward greater fatty acid oxidation by the proximal
response (22). As discussed below, factors leading to tubule. Under normal circumstances, this nephron segment
either a brisk reduction in exogenous insulin dose or inade- preferentially utilizes fatty acids as a fuel source to generate
quate endogenous insulin secretion predispose to overt ATP to meet the high metabolic demand of the tubule
ketoacidosis. because fatty acids provide more energy per gram as
1286 CJASN

Figure 1. | Alterations in ATP turnover predispose to systemic acidosis following sodium-glucose cotransporter-2 inhibitor (SGLT2i) admin-
istration. Decreased Na1 entry into the proximal tubular cell following inhibition of sodium-glucose cotransporter-2 decreases consumption
of ATP by the Na1-K1-ATPase pump. SGLT2i also restores fatty acid oxidation, which delivers more energy per gram than glucose. The com-
bined effect of reduced consumption along with increased generation of ATP decreases activity of other pathways that normally generate ATP to
include ammoniagenesis and uptake and oxidation of filtered ketone bodies. These changes predispose to urinary loss of filtered ketone bodies as
Na1 or K1 salts, causing indirect loss of bicarbonate from the body and development of metabolic acidosis under conditions of increased keto-
genesis. NaBOH, sodium b-hydroxybutyrate; NaHCO3, sodium bicarbonate; NH42, ammonium; NH4BOH, ammonium betahydroxybutyrate.

compared with glucose. Studies in humans and rodent mod- oxidation. This additional amount of ATP adds to the inhib-
els of diabetes mellitus show metabolism in the kidney is itory effect on ammonia production. In starvation, the
shifted toward utilization of glucose for anaerobic glycolysis decrease in ammonia availability does not adversely affect
and away from oxidative phosphorylation of fatty acids acid-base balance because oxidation of the ketone body in
(33,34). Rather than being utilized as fuel, lipid accumulates the proximal tubule regenerates an amount of HCO32 equal
in the cell, where it has been linked to tubular injury through to what was consumed in their production. This suppressive
lipotoxicity. Upregulation of hypoxia-inducible factor is etio- effect on ammoniagenesis by way of ATP turnover is advan-
logically linked to this change in fuel preference (35). Admin- tageous during starvation because the requirement for gluta-
istration of dapagliflozin prevents stabilization of HIF and mine uptake by the kidney will be reduced, resulting in less
attenuates the metabolic shift, suggesting that reabsorption proteolysis and providing a protein-sparing effect. In this
of excess glucose by the diabetic kidney is responsible for regard, infusion of b-hydroxybutyrate has been shown to
the switch from fatty acid oxidation to glycolysis. The reduce kidney NH41 production in dogs and humans with
increase in ATP production following restoration of fatty chronic metabolic acidosis (39–41). Following SGLT2i, the
acid oxidation in combination with reduced utilization reduction in ATP utilization in combination with the large
accounts for the decrease in ammoniagenesis that occurs quantity of ATP produced from fat oxidation will not only
with SGLT2i (36). suppress ammoniagenesis but also decrease uptake and oxi-
The increase in fat oxidation following SGLT2i can further dation of ketone bodies. These changes cause increased uri-
exacerbate indirect loss of HCO32 from the body by impair- nary excretion of b-hydroxybutyrate coupled to Na1 or
ing uptake and oxidation of filtered b-hydroxybutyrate. The K1, predisposing to acidosis through the indirect loss
kidney normally has a large capacity to reabsorb filtered of HCO32 from the body (42,43). In a study of 66 patients
ketone bodies. With prolonged fasting, blood concentrations treated with empagliflozin, kidney b-hydroxybutyrate
and the filtered load of acetoacetate and b-hydroxybutyrate excretion rose three-fold, and fractional excretion increased
progressively increase. At the same time, the absolute reab- by approximately 70% after 28 days of therapy (42).
sorption rate of ketone bodies increases linearly with no evi-
dence of a tubular maximum (37,38). Reabsorption and sub-
sequent oxidation in the kidney regenerate consumed Risk Factors for Development of Ketoacidosis
HCO32, thereby lessening the degree of acidosis that would Changes in systemic metabolism and kidney ATP turn-
otherwise occur if the ketone was excreted as an Na1 or K1 over following SGLT2i do not cause overt metabolic acidosis
salt. The complete oxidation of ketones by the tubule also in most treated patients. Rather, these changes create a tenu-
generates ATP, adding to the pool derived from fatty acid ous state whereby metabolic acidosis can quickly develop
CJASN 16: 1284–1291, August, 2021 Euglycemic Ketoacidosis, Palmer and Clegg 1287

following some precipitating factor that leads to accelerated events higher in patients with latent autoimmune diabetes
ketogenesis (Table 1). This situation occurs when insulin of adulthood or those testing positive for glutamic acid
deficiency becomes more pronounced, explaining reports decarboxylase 65-kD isoform antibodies (57). In one series
of ketoacidosis when SGLT2is are used off label in patients of SGLT2i-associated ketoacidosis, approximately one third
with type 1 diabetes mellitus or when the insulin dose is of patients had this form of the disease (49). These patients
reduced by either patients or health care workers due to have circulating islet cell antibodies and are diagnosed
reductions in plasma glucose levels. with diabetes mellitus later in life. Upon presentation, insulin
Adjunctive therapy with SGLT2i to intensive insulin regi- may not be required due to the presence of residual b-cell
mens improves glycemic control in type 1 diabetes mellitus function, leading to misclassification as type 2 diabetes mel-
but is associated with a dose-dependent increase in the abso- litus. Soon after diagnosis, there is a relatively rapid decline
lute risk of DKA (44–47). The higher risk is attributable to the in insulin release due to autoantibody-mediated injury.
failure to recognize early manifestations of metabolic decom- SGLT2i can acutely accelerate this decline due to increased
pensation secondary to a blunted rise in plasma glucose lev- release of glucagon, rendering the patient at risk for ketoaci-
els due to increased urinary glucose excretion (48). Use of dosis following a minimal metabolic stress.
SGLT2i in type 1 diabetes mellitus should be restricted to Because urinary excretion of ketoacid salts coupled to NH41
specialists with the resources necessary to train, educate, is neutral with regard to acid-base balance, it will be important
and support carefully selected patients. to determine if people with diabetes and reduced GFR or those
A sudden restriction in carbohydrate availability will on renin angiotensin system blockers are at increased risk for
increase lipolysis and lipid oxidation, explaining reports of ketoacidosis because these conditions can limit ammoniagen-
ketoacidosis under conditions of surgical stress, intercurrent esis in the kidney. There may also be additional concerns in
illness, or prolonged fasting (49). A weight loss strategy cen- individuals with type 4 renal tubular acidosis because hyper-
tered on carbohydrate restriction is a risk factor and should kalemia further suppresses ammoniagenesis.
be discouraged or implemented with close monitoring. The
glycosuric response of SGLT2i relative to the filtered glucose
load is independent of body size. This finding may explain
Clinical Features and Treatment of Sodium-Glucose
the higher risk of acidosis in lean individuals on a low-
Cotransporter-2 Inhibitor–Induced Ketoacidosis
carbohydrate diet because they are exposed to a higher def-
A normal anion gap hyperchloremic metabolic acidosis is
icit of carbohydrate when compared with obese subjects (50).
present in the initial stages of ketoacidosis (31). Loss of Na1-
Use of SGLT2i in the setting of alcohol use or salicylate toxic-
b-hydroxybutyrate in the urine secondarily causes kidney
ity is a risk factor because these conditions are characterized
retention of NaCl in an attempt to maintain extracellular
by increased ketogenesis (32,51).
fluid volume near normal. Volume depletion or a reduction
Sex-based differences in substrate metabolism may
in GFR will limit filtration of ketoacid salts, causing Na1-
explain the higher risk of ketoacidosis reported in women
b-hydroxybutyrate to accumulate in the plasma resulting
(11,45,52). During fasting, women demonstrate a greater
in an increased anion gap metabolic acidosis. At presenta-
increase in circulating nonesterified fatty acids and ketone
tion, most patients exhibit some combination of increased
bodies and a greater fall in the rate of endogenous glucose
production and utilization when compared with men (53). anion gap and a normal anion gap acidosis (58). Because
Women also demonstrate increased lipid and reduced carbo- insulin deficiency induced by SGLT2i is less severe com-
hydrate utilization as a fuel during exercise, whereas the pared with DKA, glucose overproduction and underutiliza-
opposite is true in men (54). This proclivity to utilize lipids tion are quantitatively less pronounced (Table 2). This effect,
as a fuel under conditions of metabolic stress is due to the combined with pharmacologic inhibition of glucose reab-
permissive effect of estrogen on lipolysis through enhance- sorption in the kidney, likely accounts for why the plasma
ment of hormone-sensitive lipase in adipocytes. In addition, glucose is only minimally elevated in patients with
a sexually dimorphic response of autonomic nerve activity SGLT2i-induced ketoacidosis. The lack of significant hyper-
may play a role (55). Circulating catecholamines and skeletal glycemia despite development of ketoacidosis can delay
muscle sympathetic nerve activity with exercise are lower in the diagnosis because plasma glucose is what is typically
women as compared with men. Despite the blunted monitored by the patients on a day-to-day basis. Most
response, women demonstrate a greater degree of lipolysis patients present with abdominal pain, nausea, anorexia,
but lower rates of glucose production and utilization. Lipol- vomiting, or other nonspecific symptoms (49) (Table 3).
ysis in women is solely mediated by b-adrenergic receptors, Given the benefits demonstrated in nondiabetic kidney
whereas in men, there is also activation of a-adrenergic disease in the Dapagliflozin and Prevention of Adverse Out-
receptors, which exert an inhibitory effect on lipolysis (56). comes in Chronic Kidney Disease study, use of SGLT2i by
Given the divergent effects of a- and b-receptor activation, nephrologists is likely to increase. Little is known about
stimulation of both receptors in men will blunt the degree development of ketoacidosis in such individuals, but certain
of lipolysis when compared with women, in whom only lipo- risk factors may increase the risk of this complication. Insulin
lytic b-receptors are stimulated. The shift to greater fat oxida- resistance is a feature of uremia and increases in severity
tion in women during metabolic stress increases the risk of with advancing stages of CKD (59). Concomitant use of
ketoacidosis with SGLT2i. immunosuppressive drugs, particularly corticosteroids, can
Patients with a longer duration of disease or the require- worsen insulin resistance and predispose to ketoacidosis.
ment for insulin have more impaired insulin secretory In nondiabetic rodents, euglycemic ketoacidosis developed
reserve and are at higher risk. In addition to type 1 diabetes with SGLT2i when animals were stressed with volume
mellitus, an analysis of canagliflozin trial data found DKA depletion (60). This observation suggests clinical conditions
1288 CJASN

Table 1. Risk factors for sodium-glucose cotransporter-2 inhibitor–associated ketoacidosis

Risk Factor Mechanism Precautions to Reduce Risk


Reduction in insulin dose Lowering of plasma glucose concentration Avoid .20% reductions in insulin dose;
following SGLT2i may prompt lowering of implement frequent pre- and post-
insulin dose by patient or prescriber prandial glucose monitoring following
Decreased insulin effect stimulates lipolysis and changes in insulin dose
increases glucagon-insulin ratio, predisposing Frequent monitoring of urine ketones
to ketogenesis in liver following insulin dose adjustments

Off-label use in type 1 diabetes Increased risk with pump therapy due to Reduce insulin up to 20% following first
mellitus operational failures causing sudden decrease dose of SGLT2i, then up titrate to
in insulin dose; lower risk with subcutaneous original baseline or maintain stable
regimens utilizing longer-acting formulations insulin regimen for first 7 treatment d,
compared with rapid-acting analogs in pump and adjust thereafter according to
regimens glucose levels.a Assess ketones to
detect early DKA in event of
nonspecific symptoms regardless of
glucose levels; adhere to optimized
sick-day protocols

Restriction in dietary carbohydrate Glycosuric effect of SGLT2i combined with Avoid very low–carbohydrate diets
availability as seen with the use decreased dietary carbohydrate intake leads
of ketogenic diets to decreased insulin levels
Increased lipolysis due to reductions in
carbohydrate intake leads to an increased
glucagon-insulin ratio, which predisposes to
ketogenesis

Surgical stress, trauma, Decreased food intake leads to reduced insulin Discontinue SGLT2i 3 d prior to elective
intercurrent illness such as levels, thereby increasing likelihood of surgery
gastroenteritis where there is an ketogenesis Discontinue SGLT2i with acute illness
inability to eat or drink Nonspecific symptoms, such as malaise and Educate patient to monitor for ketonuria,
nausea, along with mild or absent glycemia in addition to glucose, and contact
may cause patient to decrease insulin dose, provider with any symptoms or
thus increasing risk of ketosis detectable ketonuria
Increased levels of counter-regulatory hormones Supplemental doses of rapid-acting
(catecholamines and cortisol) accelerate insulin, along with fluids and
lipolysis and increase insulin demand ingestion of carbohydrates
Lean body habitus Carbohydrate deficit is greater in lean versus Educate lean/muscle-bound patients as
obese individuals because the degree of to their increased risk and symptoms
glycosuria relative to filtered load is
independent of body size following SGLT2i

Alcohol use disorder and salicylate Alcohol withdrawal and salicylate toxicity Discontinue SGLT2i
toxicity are both associated with increased lipolysis Be aware of patients with chronic
and decreased insulin-glucagon ratio, thereby salicylate toxicity
increasing risk of ketogenesis, conditions
made worse following SGLT2i

Women Women preferentially oxidize fatty acids and Patient education that increased risk is
exhibit greater increase in ketogenesis with confined to premenopausal women or
metabolic stress postmenopausal women taking
Estrogen enhances hormone-sensitive lipase in hormonal replacement therapy to
adipocytes include estrogens

LADA LADA is phenotypically similar to type 2 Increased awareness of disorder


diabetes mellitus, but insulin dependency Prevalence is about 10% among patients
develops rapidly (5–6 yr) due to immune- diagnosed with type 2 diabetes
mediated b-cell injury mellitus with islet antibodies
Unrecognized rapid loss of b-cell secretory Presence of impaired b-cell function at
reserve combined with metabolic stress diagnosis warrants early insulin
increases risk in the setting of SGLT2i therapy

Long-standing type 2 diabetes Longer duration of disease increases likelihood Patient education and more frequent
mellitus of marked b-cell insufficiency monitoring when SGLT2i is given to
Imposition of metabolic stress readily unmasks patients with long duration of disease
limited insulin secretory reserve

SGLT2i, sodium-glucose cotransporter-2 inhibitor; DKA, diabetic ketoacidosis; LADA, latent autoimmune diabetes in adults.
a
Strategies utilized to decrease risk of ketoacidosis in clinical trials of type 1 diabetes mellitus (44,45).
CJASN 16: 1284–1291, August, 2021 Euglycemic Ketoacidosis, Palmer and Clegg 1289

Table 2. Selected clinical and laboratory findings distinguishing ketoacidosis due to sodium-glucose cotransporter-2 inhibitor from typ-
ical diabetic ketoacidosis

Sodium-Glucose Cotransporter-2
Factor Diabetic Ketoacidosis
Inhibitor–Induced Diabetic Ketoacidosis
Endogenous glucose production " ""
Insulin release # ##
Insulin resistance " ""
Tissue glucose disposal # ##
Kidney glucose clearance "" "
Plasma glucose levels Normal or increased, often ,250 mg/dla Typically 350–800 mg/dl
Extracellular fluid volume # ##
Presenting symptoms More nonspecific to include malaise, Polyuria and polydipsia due to osmotic
nausea, anorexia, abdominal pain diuresis, nausea, vomiting, shortness
of breath

a
Because the plasma glucose is either normal or only modestly increased in most patients, transcellular shift in water and K1 is
minimal, making disturbances in plasma Na1 and K1 concentration less severe when compared with typical diabetic ketoacidosis
(mechanisms are reviewed in ref. 31).

predisposing to insulin resistance, such as activation of up to 14 days after drug discontinuation (63). Patients should
sympathetic nerve activity or increased circulating glucocor- be educated about possible symptoms of ketoacidosis and, if
ticoids, may be sufficient to cause this complication in present, instructed to contact their physician even if glucose
susceptible individuals without known diabetes mellitus. monitoring does not indicate hyperglycemia (Table 3). When
The risk of SGLT2i-induced ketoacidosis can be minimized SGLT2is are prescribed to patients treated with insulin, the
by close monitoring and making appropriate adjustments in dose of insulin may need to be reduced to avoid hypoglyce-
medications upon changing clinical conditions. For example, mia. Such a change requires close monitoring as the reduc-
major surgery or trauma is a common precipitating risk fac- tion in inulin dose increases the risk for ketoacidosis.
tor. These conditions lead to stress-induced increases in cir- There are no specific guidelines for treatment of SGLT2i-
culating catecholamines and cortisol, which, in combination, induced ketoacidosis; however, the treatment approach is
can reduce insulin sensitivity. The glycosuric effect of like that of typical DKA (31). In addition to drug discontinu-
SGLT2i may prevent significant increases in plasma glucose ation, restoration of extracellular fluid volume with normal
concentration, masking the true requirement for insulin and saline should be the initial approach. The degree of volume
increasing the risk of ketoacidosis. Current recommenda- depletion may be less severe because hyperglycemia is less
tions suggest SGLT2i should be discontinued at least 3 pronounced, minimizing the severity of osmotic diuresis.
days prior to an elective surgical or invasive procedure Early intravenous insulin is required to facilitate utilization
(61). Insulin can be used as a bridge for glycemic control dur- of glucose and diminish ketogenesis. In typical DKA, a 5%
ing this period. Although this time period provides approx- dextrose solution is recommended after the plasma glucose
imately five t1/2 of elimination, it should be noted that phar- falls to 250 mg/dl. This solution along with insulin may be
macologic effects of the drugs, such as glycosuria and appropriate earlier in the treatment of SGLT2i-induced ketoa-
ketonemia, can be detected up to 10 days after discontinua- cidosis because the plasma glucose concentration may only
tion (62). Patient reports have described the onset of DKA be minimally increased. In the absence of life-threatening

Table 3. Recommendations to patients when prescribed sodium-glucose cotransporter-2 inhibitor

Recommendations
Maintain appropriate fluid intake
Ensure adequate carbohydrate intake and avoid low-carbohydrate diets
Avoid skipping insulin doses and skipping meals
In situations of acute illness, vomiting, diarrhea, or inability to eat or drink
Discontinue SGLT2i
Contact provider even if blood glucose levels are not elevated
Continue to monitor glucose levels and monitor for presence of urinary ketones
If on insulin therapy, contact provider for dose adjustments; do not stop insulin
Ketoacidosis symptoms are nonspecific (malaise, nausea, anorexia, vomiting) and can occur despite normal or minimally
elevated blood glucose level
If ketonuria detected, patient may be advised to administer dose of rapid-acting insulin and consume 30 g carbohydrate
Restart SGLT2i when eating and drinking normally, usually after 24–48 h as directed by medical provider

Recommendations to patients when prescribed sodium-glucose cotransporter-2 inhibitor (SGLT2i) are especially important for lean
women who are premenopausal or postmenopausal on hormone replacement therapy.
1290 CJASN

acidosis or a pH,7.1, alkali therapy is generally not required 7. Bonora BM, Avogaro A, Fadini GP: Sodium-glucose co-trans-
because insulin administration slows the rate of ketoacid pro- porter-2 inhibitors and diabetic ketoacidosis: An updated review
of the literature. Diabetes Obes Metab 20: 25–33, 2018
duction, and oxidation of ketoanions regenerates HCO32. 8. Peters AL, Henry RR, Thakkar P, Tong C, Alba M: Diabetic
The presence of a predominately normal gap acidosis indi- ketoacidosis with canagliflozin, a sodium-glucose cotransporter
cates that only a small amount of organic anions is available 2 inhibitor, in patients with type 1 diabetes. Diabetes Care 39:
for regeneration of HCO32, and the indirect loss of HCO32 532–538, 2016
9. Dandona P, Mathieu C, Phillip M, Hansen L, Tscho €pe D, Thoren
from the body has been considerable. Regeneration of
F, Xu J, Langkilde AM; DEPICT-1 Investigators: Efficacy and
HCO32 and establishment of normal acid-base balance can safety of dapagliflozin in patients with inadequately controlled
take up to 72 hours or even longer in the presence of CKD type 1 diabetes: The DEPICT-1 52-week study. Diabetes Care 41:
in this setting. Alkali therapy may be useful to shorten the 2552–2559, 2018
total duration of acidemia in such patients. 10. Kidney Disease: Improving Global Outcomes (KDIGO) Diabetes
In summary, use of SGLT2i is likely to increase dramatically Work Group: KDIGO 2020 clinical practice guideline for dia-
betes management in chronic kidney disease (CKD). Available
given the evidence that these drugs confer cardiovascular at: https://kdigo.org/guidelines/diabetes-ckd/
benefit. These drugs alter ATP turnover in the kidney, predis- 11. Blau JE, Tella SH, Taylor SI, Rother KI: Ketoacidosis associated
posing to euglycemic ketoacidosis under conditions of accel- with SGLT2 inhibitor treatment: Analysis of FAERS data. Diabe-
erated ketogenesis. Clinicians should be familiar with risk fac- tes Metab Res Rev 33: e2924, 2017
tors so the likelihood of this disorder can be minimized. 12. Douros A, Lix LM, Fralick M, Dell’Aniello S, Shah BR, Ronksley
PE, Tremblay E, Hu N, Alessi-Severini S, Fisher A, Bugden SC,
Ernst P, Filion KB: Sodium-glucose cotransporter-2 inhibitors and
Disclosures the risk for diabetic ketoacidosis: A multicenter cohort study.
All authors have nothing to disclose. Ann Intern Med 173: 417–425, 2020
13. Diaz-Ramos A, Eilbert W, Marquez D: Euglycemic diabetic
ketoacidosis associated with sodium-glucose cotransporter-2
Funding inhibitor use: A case report and review of the literature. Int J
None. Emerg Med 12: 27, 2019
14. Ghezzi C, Loo DDF, Wright EM: Physiology of renal glucose
References handling via SGLT1, SGLT2 and GLUT2. Diabetologia 61: 2087–
1. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, 2097, 2018
Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, 15. Song WJ, Mondal P, Wolfe A, Alonso LC, Stamateris R, Ong BW,
Lim OC, Yang KS, Radovick S, Novaira HJ, Farber EA, Farber CR,
Inzucchi SE; EMPA-REG OUTCOME Investigators: Empagliflozin,
Turner SD, Hussain MA: Glucagon regulates hepatic kisspeptin
cardiovascular outcomes, and mortality in type 2 diabetes.
to impair insulin secretion. Cell Metab 19: 667–681, 2014
N Engl J Med 373: 2117–2128, 2015
16. Daniele G, Xiong J, Solis-Herrera C, Merovci A, Eldor R, Tripathy
2. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod
D, DeFronzo RA, Norton L, Abdul-Ghani M: Dapagliflozin
MN, Martinez FA, Ponikowski P, Sabatine MS, Anand IS,
enhances fat oxidation and ketone production in patients with
Belohl €hm M, Chiang CE, Chopra VK, de Boer RA,
avek J, Bo
type 2 diabetes. Diabetes Care 39: 2036–2041, 2016
Desai AS, Diez M, Drozdz J, Dukat A, Ge J, Howlett JG, Katova
17. Rosenstock J, Ferrannini E: Euglycemic diabetic ketoacidosis: A
T, Kitakaze M, Ljungman CEA, Merkely B, Nicolau JC, O’Meara
predictable, detectable, and preventable safety concern with
E, Petrie MC, Vinh PN, Schou M, Tereshchenko S, Verma S, Held
SGLT2 inhibitors. Diabetes Care 38: 1638–1642, 2015
C, DeMets DL, Docherty KF, Jhund PS, Bengtsson O, Sjo €strand € Kullberg J, Johansson L, Wilding J,
18. Bolinder J, Ljunggren O,
M, Langkilde AM; DAPA-HF Trial Committees and Investigators: Langkilde AM, Sugg J, Parikh S: Effects of dapagliflozin on body
Dapagliflozin in patients with heart failure and reduced ejection weight, total fat mass, and regional adipose tissue distribution in
fraction. N Engl J Med 381: 1995–2008, 2019 patients with type 2 diabetes mellitus with inadequate glycemic
3. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, control on metformin. J Clin Endocrinol Metab 97: 1020–1031,
Januzzi J, Verma S, Tsutsui H, Brueckmann M, Jamal W, Kimura 2012
K, Schnee J, Zeller C, Cotton D, Bocchi E, Bo€hm M, Choi DJ,
19. Xu L, Nagata N, Nagashimada M, Zhuge F, Ni Y, Chen G,
Chopra V, Chuquiure E, Giannetti N, Janssens S, Zhang J, Gon- Mayoux E, Kaneko S, Ota T: SGLT2 inhibition by empagliflozin
zalez Juanatey JR, Kaul S, Brunner-La Rocca HP, Merkely B, promotes fat utilization and browning and attenuates inflam-
Nicholls SJ, Perrone S, Pina I, Ponikowski P, Sattar N, Senni M, mation and insulin resistance by polarizing M2 macrophages in
Seronde MF, Spinar J, Squire I, Taddei S, Wanner C, Zannad F; diet-induced obese mice. EBioMedicine 20: 137–149, 2017
EMPEROR-Reduced Trial Investigators: Cardiovascular and renal 20. Ferrannini E, Muscelli E, Frascerra S, Baldi S, Mari A, Heise T,
outcomes with empagliflozin in heart failure. N Engl J Med 383: Broedl UC, Woerle HJ: Metabolic response to sodium-glucose
1413–1424, 2020 cotransporter 2 inhibition in type 2 diabetic patients. J Clin Invest
4. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, 124: 499–508, 2014
Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon 21. Ferrannini E, Baldi S, Frascerra S, Astiarraga B, Heise T, Bizzotto
CP, Capuano G, Chu PL, de Zeeuw D, Greene T, Levin A, Pol- R, Mari A, Pieber TR, Muscelli E: Shift to fatty substrate utilization
lock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, in response to sodium-glucose cotransporter 2 inhibition in
Brenner BM, Mahaffey KW; CREDENCE Trial Investigators: subjects without diabetes and patients with type 2 diabetes.
Canagliflozin and renal outcomes in type 2 diabetes and Diabetes 65: 1190–1195, 2016
nephropathy. N Engl J Med 380: 2295–2306, 2019 22. Brady JA, Hallow KM: Model-based evaluation of proximal
5. Heerspink HJL, Stefansson BV, Correa-Rotter R, Chertow GM, sodium reabsorption through SGLT2 in health and diabetes and
Greene T, Hou FF, Mann JFE, McMurray JJV, Lindberg M, Ross- the effect of inhibition with canagliflozin. J Clin Pharmacol 58:
ing P, Sjo€stro
€m CD, Toto RD, Langkilde AM, Wheeler DC; 377–385, 2018
DAPA-CKD Trial Committees and Investigators: Dapagliflozin in 23. Thomas MC, Cherney DZI: The actions of SGLT2 inhibitors on
patients with chronic kidney disease. N Engl J Med 383: 1436– metabolism, renal function and blood pressure. Diabetologia 61:
1446, 2020 2098–2107, 2018
6. US Food and Drug Administration: Drug Safety Communication: 24. Wang XX, Levi J, Luo Y, Myakala K, Herman-Edelstein M, Qiu L,
FDA warns that SGLT2 inhibitors for diabetes may result in a Wang D, Peng Y, Grenz A, Lucia S, Dobrinskikh E, D’Agati VD,
serious condition of too much acid in the blood, 2015. Available Koepsell H, Kopp JB, Rosenberg AZ, Levi M: SGLT2 protein
at: www.fda.gov/downloads/Drugs/DrugSafety/UCM446954. expression is increased in human diabetic nephropathy: SGLT2
pdf. Accessed June 22, 2015 protein inhibition decreases renal lipid accumulation,
CJASN 16: 1284–1291, August, 2021 Euglycemic Ketoacidosis, Palmer and Clegg 1291

inflammation, and the development of nephropathy in diabetic 45. Rosenstock J, Marquard J, Laffel LM, Neubacher D, Kaspers S,
mice. J Biol Chem 292: 5335–5348, 2017 Cherney DZ, Zinman B, Skyler JS, George J, Soleymanlou N,
25. Mather A, Pollock C: Glucose handling by the kidney. Kidney Int Perkins BA: Empagliflozin as adjunctive to insulin therapy in type
Suppl 120: S1–S6, 2011 1 diabetes: The EASE trials. Diabetes Care 41: 2560–2569, 2018
26. Ferrannini E: Sodium-glucose co-transporters and their inhibi- 46. Garg SK, Henry RR, Banks P, Buse JB, Davies MJ, Fulcher GR,
tion: Clinical physiology. Cell Metab 26: 27–38, 2017 Pozzilli P, Gesty-Palmer D, Lapuerta P, Simo  R, Danne T,
27. Pessoa TD, Campos LC, Carraro-Lacroix L, Girardi AC, Malnic McGuire DK, Kushner JA, Peters A, Strumph P: Effects of sota-
G: Functional role of glucose metabolism, osmotic stress, and gliflozin added to insulin in patients with type 1 diabetes. N Engl
sodium-glucose cotransporter isoform-mediated transport on J Med 377: 2337–2348, 2017
Na1/H1 exchanger isoform 3 activity in the renal proximal 47. Wolfsdorf JI, Ratner RE: SGLT inhibitors for type 1 diabetes:
tubule. J Am Soc Nephrol 25: 2028–2039, 2014 Proceed with extreme caution. Diabetes Care 42: 991–993,
28. Onishi A, Fu Y, Patel R, Darshi M, Crespo-Masip M, Huang W, 2019
Song P, Freeman B, Kim YC, Soleimani M, Sharma K, Thomson 48. Patel NS, Van Name MA, Cengiz E, Carria LR, Weinzimer SA,
SC, Vallon V: A role for tubular Na1/H1 exchanger NHE3 in the Tamborlane WV, Sherr JL: Altered patterns of early metabolic
natriuretic effect of the SGLT2 inhibitor empagliflozin. Am J decompensation in type 1 diabetes during treatment with a
Physiol Renal Physiol 319: F712–F728, 2020 SGLT2 inhibitor: An insulin pump suspension study. Diabetes
29. Halperin ML, Cheema-Dhadli S: Renal and hepatic aspects of Technol Ther 19: 618–622, 2017
ketoacidosis: A quantitative analysis based on energy turnover. 49. Burke KR, Schumacher CA, Harpe SE: SGLT2 inhibitors: A sys-
Diabetes Metab Rev 5: 321–336, 1989 tematic review of diabetic ketoacidosis and related risk factors in
30. Halperin ML, Vinay P, Gougoux A, Pichette C, Jungas RL: Reg- the primary literature. Pharmacotherapy 37: 187–194, 2017
ulation of the maximum rate of renal ammoniagenesis in the 50. Herring RA, Shojaee-Moradie F, Garesse R, Stevenage M, Jack-
acidotic dog. Am J Physiol 248: F607–F615, 1985 son N, Fielding BA, Mendis A, Johnsen S, Umpleby AM, Davies
31. Palmer BF, Clegg DJ: Electrolyte and acid-base disturbances in M, Russell-Jones DL: Metabolic effects of an SGLT2 inhibitor
patients with diabetes mellitus [published correction appears in (Dapagliflozin) during a period of acute insulin withdrawal and
N Engl J Med 381: 1598, 2019 10.1056/NEJMx190026]. N Engl J development of ketoacidosis in people with type 1 diabetes.
Med 373: 548–559, 2015 Diabetes Care 43: 2128–2136, 2020
32. Palmer BF, Clegg DJ: Electrolyte disturbances in patients with 51. Palmer BF, Clegg DJ: Salicylate toxicity. N Engl J Med 382: 2544–
chronic alcohol-use disorder. N Engl J Med 377: 1368–1377, 2555, 2020
2017 52. Fadini GP, Bonora BM, Avogaro A: SGLT2 inhibitors and dia-
33. Jang HS, Noh MR, Kim J, Padanilam BJ: Defective mitochondrial betic ketoacidosis: Data from the FDA adverse event reporting
fatty acid oxidation and lipotoxicity in kidney diseases. Front system. Diabetologia 60: 1385–1389, 2017
Med (Lausanne) 7: 65, 2020 53. Marinou K, Adiels M, Hodson L, Frayn KN, Karpe F, Fielding BA:
34. Kang HM, Ahn SH, Choi P, Ko YA, Han SH, Chinga F, Park AS, Young women partition fatty acids towards ketone body pro-
Tao J, Sharma K, Pullman J, Bottinger EP, Goldberg IJ, Susztak K: duction rather than VLDL-TAG synthesis, compared with young
Defective fatty acid oxidation in renal tubular epithelial cells has men. Br J Nutr 105: 857–865, 2011
a key role in kidney fibrosis development. Nat Med 21: 37–46, 54. Hedrington MS, Davis SN: Sexual dimorphism in glucose and
2015 lipid metabolism during fasting, hypoglycemia, and exercise.
35. Cai T, Ke Q, Fang Y, Wen P, Chen H, Yuan Q, Luo J, Zhang Y, Front Endocrinol (Lausanne) 6: 61, 2015
Sun Q, Lv Y, Zen K, Jiang L, Zhou Y, Yang J: Sodium-glucose 55. Davis SN, Galassetti P, Wasserman DH, Tate D: Effects of gender
cotransporter 2 inhibition suppresses HIF-1a-mediated meta-
on neuroendocrine and metabolic counterregulatory responses
bolic switch from lipid oxidation to glycolysis in kidney tubule
to exercise in normal man. J Clin Endocrinol Metab 85: 224–230,
cells of diabetic mice. Cell Death Dis 11: 390, 2020
2000
36. Vinay P, Lemieux G, Cartier P, Ahmad M: Effect of fatty acids on
56. Palmer BF, Clegg DJ: The sexual dimorphism of obesity. Mol Cell
renal ammoniagenesis in in vivo and in vitro studies. Am J Physiol
Endocrinol 402: 113–119, 2015
231: 880–887, 1976
57. Erondu N, Desai M, Ways K, Meininger G: Diabetic ketoacidosis
37. Sapir DG, Owen OE: Renal conservation of ketone bodies during
and related events in the canagliflozin type 2 diabetes clinical
starvation. Metabolism 24: 23–33, 1975
program. Diabetes Care 38: 1680–1686, 2015
38. Owen OE, Caprio S, Reichard GA Jr, Mozzoli MA, Boden G,
58. Sampani E, Sarafidis P, Papagianni A: Euglycaemic diabetic
Owen RS: Ketosis of starvation: A revisit and new perspectives.
ketoacidosis as a complication of SGLT-2 inhibitors: Epidemi-
Clin Endocrinol Metab 12: 359–379, 1983
39. Desir G, Bratusch-Marrain P, DeFronzo RA: Effect of hyperke- ology, pathophysiology, and treatment. Expert Opin Drug Saf 19:
tonemia on renal ammonia excretion in man. Metabolism 35: 673–682, 2020
736–743, 1986 59. Kobayashi S, Maesato K, Moriya H, Ohtake T, Ikeda T: Insulin
40. Sherwin RS, Hendler RG, Felig P: Effect of ketone infusions on resistance in patients with chronic kidney disease. Am J Kidney
amino acid and nitrogen metabolism in man. J Clin Invest 55: Dis 45: 275–280, 2005
1382–1390, 1975 60. Perry RJ, Rabin-Court A, Song JD, Cardone RL, Wang Y, Kibbey
41. Lemieux G, Pichette C, Vinay P, Gougoux A: Cellular mecha- RG, Shulman GI: Dehydration and insulinopenia are necessary
nisms of the antiammoniagenic effect of ketone bodies in the and sufficient for euglycemic ketoacidosis in SGLT2 inhibitor-
dog. Am J Physiol 239: F420–F426, 1980 treated rats. Nat Commun 10: 548, 2019
42. Ferrannini E, Baldi S, Frascerra S, Astiarraga B, Barsotti E, Clerico 61. US Food and Drug Administration: FDA approves label changes
A, Muscelli E: Renal handling of ketones in response to sodium- to SGLT2 inhibitors regarding temporary discontinuation of
glucose cotransporter 2 inhibition in patients with type 2 dia- medication before scheduled surgery. Available at: https://www.
betes. Diabetes Care 40: 771–776, 2017 fda.gov/drugs/drug-safety-and-availability/fda-revises-labels-
43. Yokono M, Takasu T, Hayashizaki Y, Mitsuoka K, Kihara R, sglt2-inhibitors-diabetes-include-warnings-about-too-much-
Muramatsu Y, Miyoshi S, Tahara A, Kurosaki E, Li Q, Tomiyama acid-blood-and-serious. Accessed March 23, 2020
H, Sasamata M, Shibasaki M, Uchiyama Y: SGLT2 selective 62. Yehya A, Sadhu A: Sodium-glucose cotransporter 2 inhibitor-
inhibitor ipragliflozin reduces body fat mass by increasing fatty associated prolonged euglycemic diabetic ketoacidosis in type 2
acid oxidation in high-fat diet-induced obese rats. Eur J Phar- diabetes: A case report and literature review. Clin Diabetes 38:
macol 727: 66–74, 2014 112–116, 2020
44. Mathieu C, Dandona P, Gillard P, Senior P, Hasslacher C, Araki 63. Iqbal I, Hamid M, Khan MAA, Kainat A, Tariq S: Dapagliflozin-
E, Lind M, Bain SC, Jabbour S, Arya N, Hansen L, Thoren F, induced late-onset euglycemic diabetic ketoacidosis. Cureus 11:
Langkilde AM; DEPICT-2 Investigators: Efficacy and safety of e6089, 2019
dapagliflozin in patients with inadequately controlled type 1
diabetes (the DEPICT-2 study): 24-week results from a random- Published online ahead of print. Publication date available at
ized controlled trial. Diabetes Care 41: 1938–1946, 2018 www.cjasn.org.

You might also like