Skip to main content

OPINION article

Front. Endocrinol., 07 October 2024
Sec. Renal Endocrinology
This article is part of the Research Topic Endocrine Regulation of Homeostasis of Water, Electrolytes and Organic Solutes View all articles

Empagliflozin: a wonder drug for the treatment of SIAD?

  • Department of Metabolism & Experimental Therapeutics, Division of Medicine, University College London, London, United Kingdom

Introduction

The syndrome of inappropriate antidiuresis (SIAD), the commonest cause of euvolemic hyponatremia, is associated with significant morbidity and mortality (1, 2). Despite the poor efficacy of fluid restriction (35), second line agents, such as urea (6, 7) and tolvaptan (810), are underutilized in clinical practice. This unmet clinical need may be satisfied by empagliflozin, with emerging evidence supporting its use as a promising new treatment option for SIAD (11, 12). Empagliflozin, a potent, selective inhibitor of sodium glucose cotransporter 2 (SGLT2), is an oral glucose-lowering agent widely used for the treatment of type 2 diabetes mellitus (DM) (1315) (16), especially in patients with established cardiovascular disease (17) or heart failure or chronic kidney disease (CKD) (18) because of its favorable cardiorenal outcomes. In recent years, empagliflozin has gained approval for two new indications, regardless of the presence or absence of DM; firstly, for the treatment of heart failure across the spectrum of ejection fraction (19, 20), and second, as a nephroprotective agent in patients with CKD at risk of progression (21).

Mechanisms of empagliflozin action in SIAD

SGLT2, a high capacity and low affinity glucose transporter located in the first segment of the proximal convoluted tubule, accounts for the reabsorption of approximately 90% of filtered glucose (22, 23). Therefore, empagliflozin, through SGLT2 inhibition, induces pronounced glycosuria, leading to osmotic diuresis and increased renal electrolyte-free water excretion (24). Of note is that SGLT2 inhibitors may induce estivation-like metabolic patterns, evolutionarily conserved survival strategies enabling physiological adaptation to energy and water shortage, which potentially contribute to observed improvements in cardiac and renal function (25). After the first dose of empagliflozin, natriuresis occurs due to SGLT2 inhibition, where sodium is coupled with glucose reabsorption (2628), and blockage of sodium-hydrogen exchanger-3 (NHE3) in the proximal tubule (29). However, natriuresis is short-lived and transient, recovering to the baseline a few days later (30), following compensatory increase of sodium reabsorption in the distal tubule and collecting duct (31), increase in plasma renin activity and reduction in natriuretic peptides (2628). Animal studies showing that empagliflozin reduces expression of Na+-K+-2Cl- cotransporter and epithelial Na+ channels (ENaC) (32) and decreases mRNA and protein levels of aquaporin 2 (32), the main channel for water reabsorption, suggest that empagliflozin may partially contribute to polyuria via its direct effect on sodium and water channels (26, 32).

Given the well-known effect of SGLT2 inhibition on enhancing urinary water excretion in patients with type 2 DM, an artificial SIAD model was created in healthy volunteers via intravenous desmopressin administration and overhydration (33). This double-blind placebo-controlled randomized crossover study, including 14 healthy volunteers, examined the effect of empagliflozin on urine volume and glycosuria for a 6-hour period (33). In this artificial SIAD model, a single dose of empagliflozin 25 mg led to significantly increased urine volume in association with glycosuria, but without a significant difference in total natriuresis, demonstrating a strong aquaretic action of empagliflozin (33).

An interesting observation is that administration of SGLT2 inhibitors in patients with type 2 DM promotes release of arginine vasopressin (AVP) in order to compensate for persistent osmotic diuresis (34). Empagliflozin-induced losses of electrolyte-free water produce water flow from intracellular to extracellular fluid which are detected by osmoreceptors stimulating AVP release (34). Following these findings, a recent study investigated whether empagliflozin improves hyponatremia through affecting apelin and AVP, two neuropeptides which regulate water homeostasis in opposing ways; AVP by promoting antidiuresis, whereas apelin by augmenting diuresis (3537). In patients with SIAD, empagliflozin had no effect on plasma apelin concentration, but led to a significant increase by 25% of median serum levels of copeptin, an equimolar surrogate marker of AVP (38). These findings suggest that the efficacy of empagliflozin in SIAD is neither mediated through apelin nor blunted by the adaptive stimulation of AVP release (38). The finding that the increase in serum AVP levels does not compromise the efficacy of empagliflozin in SIAD is also supported by the fact that the overall effect of empagliflozin on ENaC is reduced activity (32) contrary to the marked upregulation of ENaC expression in states of chronic exposure to AVP (39).

Studies of empagliflozin as treatment for SIAD

Two randomized controlled trials (RCTs) have evaluated the efficacy and safety of empagliflozin for treatment of SIAD (11, 12). The first trial, recruiting 88 hospitalized patients with SIAD and serum sodium < 130 mmol/l (median sodium 126 mmol/l) and assigning them to empagliflozin 25 mg or placebo once daily for 4 days in addition to standard fluid restriction less than 1000 ml/day, showed that empagliflozin led to a significantly higher median sodium increase of 10 mmol/l versus 7 mmol/l in the placebo group (P = 0.04) (11). The difference in serum sodium concentration between empagliflozin and placebo arm was already noted after 24 hours (4 versus 3 mmol/l), increased after 48 hours (8 versus 5 mmol/l) and persisted for the whole 4-day period. It is worth mentioning that the control arm of mild fluid restriction showed a median sodium increment of 7 mmol/l, much higher than previously reported (3), suggesting a potentially significant contribution of hyponatremia autocorrection after reversing the cause of SIAD (40). Empagliflozin was well tolerated with no events of hypoglycemia or hypotension. Out of 43 empagliflozin-treated patients, there were two cases (4.6%) of overly rapid hyponatremia correction and 4 cases (9.3%) of transient decrease in renal function (11). Patients in the empagliflozin arm exhibited marked glycosuria, as evidenced by median urinary glucose of 111.2 mmol/l versus 0.3 mmol/l in the placebo arm, and empagliflozin treatment produced a statistically significant increase in urine osmolality by 164 mOsm/kg compared with the placebo group (P < 0.001) (11).The magnitude of serum sodium increase was negatively correlated with baseline serum sodium and osmolality, with more pronounced changes in serum sodium being observed in those with profound hyponatremia (serum sodium < 125 mmol/l) at the outset (11). In total, this study demonstrated that empagliflozin in combination with fluid restriction is an effective treatment option for hospitalized patients with SIAD over a 4-day period.

Following these encouraging findings, a randomized, double-blind, placebo-controlled, crossover trial evaluated the efficacy and safety of empagliflozin compared to placebo combined with limitation of fluid intake to around 1500 ml/day in 14 outpatients with median serum sodium of 131 mmol/l due to chronic SIAD (12). Four-week treatment with empagliflozin resulted in a median serum sodium increase of 4.1 mmol/l compared to placebo (P = 0.004) which was achieved at the end of first week and remained stable until the end of the study period (12). Following four-week empagliflozin administration, median urine osmolality was 601 mOsm/kg, significantly higher than 494 mOsm/kg in placebo group (P < 0.001) (12). Empagliflozin was well tolerated with no events of hypoglycemia, hypotension, overly sodium correction or acute kidney injury. In addition, empagliflozin led to a small improvement in neurocognitive function, especially when normonatremia was restored, but without any significant improvement in gait (12). In total, this study reported that a 4-week treatment with empagliflozin led to a significant, albeit modest, increase in serum sodium levels in outpatients with chronic SIAD (12).

Secondary analysis of both RCTs has shown that empagliflozin-induced correction of hyponatremia in patients with SIAD is associated with a statistically significant increase in Procollagen type 1 N-terminal propeptide (P1NP), a surrogate marker of bone formation, but without a significant effect on C-terminal telopeptide (CTX), a marker of bone resorption (41) (42). In total, bone formation index, the ratio of P1NP to CTX, increases, indicating that empagliflozin-induced increase in serum sodium levels over a short duration of 4 days (41) or 4 weeks (42) has a positive impact on bone metabolism through activation of osteoblast function, while the long-term effect on bone turnover warrants further study (41, 42).

Empagliflozin-induced increase of serum sodium in patients with SIAD cannot be extrapolated to patients without SIAD since analysis of pooled data from several RCTs does not show any changes in serum sodium concentration in empagliflozin-treated patients with type 2 DM and normonatremia (43, 44). A retrospective analysis of hospitalized patients with type 2 DM has shown that long-term therapy with SGLT2 inhibitors does not alter the prevalence of hyponatremia on hospital admission (45). Therefore, SGLT2 inhibitors do not prevent the development of hyponatremia and should not be used as prophylaxis for hyponatremia in at-risk individuals (45). Additionally, data about the effect of empagliflozin on hypervolemic hyponatremia due to heart failure or cirrhosis are lacking.

Pros and cons of empagliflozin for SIAD

Empagliflozin may represent a promising therapeutic option for SIAD, having the following advantages: (i) rapid onset of action since difference in serum sodium compared to placebo was already noted after 12 hours (11), (ii) efficacy in increasing serum sodium concentration compared to placebo in both hospitalized patients (11) and outpatients with SIAD (12) as well as in combination with both standard fluid restriction < 1000 ml/day (11) and limitation of fluid intake to 1500-1600 ml/day (12), (iii) being well tolerated with no events of hypoglycemia or hypotension in SIAD studies (11, 12), (iv) favorable safety profile in general based on numerous RCTs and post-marketing surveillance in patients with type 2 DM (17) (v) cardiovascular benefits in individuals with type 2 DM and established cardiovascular disease (17, 46), cardioprotective properties in those with heart failure (19) (20) and nephroprotective effects in those with type 2 DM (47) or CKD (21) (48), (vi) widespread availability, clinicians’ familiarity and ease of use in comparison to the alternative drug therapies for SIAD (vii) relatively low cost, being around 1/40th of tolvaptan cost in the UK.

Empagliflozin as a novel drug agent for SIAD carries some disadvantages: (i) modest efficacy, as suggested by a mean placebo-subtracted serum sodium increase of 3 mmol/l after 4 days (11) and 4.1 mmol/l after 4 weeks (12), (ii) limited evidence base supporting its efficacy, based on its use in 43 patients over 4 days (11) and 14 individuals over 4 weeks (12), (iii) lack of data evaluating its efficacy and safety in combination with tight fluid restriction, such as below 1000 or 500 ml/day, (iv) potential risk of excessive hyponatremia correction and renal deterioration (11).

Unanswered questions and research gaps

The evidence to date, limited to two small RCTs, highlights the need for large-scale, prospective studies, evaluating the long-term durability of response and safety profile. The efficacy and safety of empagliflozin for treatment of SIAD should be assessed across different levels of baseline serum sodium and at different degrees of fluid restriction, while it remains to be tested whether relaxation of fluid restriction is required prior to empagliflozin initiation in order to prevent decrease in renal function, especially in the elderly. Another safety concern to be addressed is whether hospitalized patients treated with empagliflozin for SIAD are at an increased risk for euglycemic diabetic ketoacidosis (DKA), a well-known association in inpatients with type 2 DM treated with SGLT2 inhibitors (49). Future studies should incorporate as secondary end points the association of various baseline parameters, including biochemical variables, cause of SIAD and level of renal function, with the therapeutic potency and safety of empagliflozin in order to identify predictors of response and facilitate personalized decision-making. In addition, head-to-head studies are warranted to directly compare the efficacy and safety of empagiflozin with other drug options for SIAD, such as urea and tolvaptan. Taking into account the similar efficacy and safety profiles of all SGLT2 inhibitors (50), the role of dapagliflozin and canagliflozin as effective therapies for SIAD also warrants evaluation. A prospective, multicentric, randomized, double-blind placebo-controlled trial (the EMPOWER study; NCT04447911) is currently underway, evaluating the effect of empagliflozin not only in in euvolemic, but also in hypervolemic hyponatremia over a 30-day treatment period. The results of this study are eagerly awaited, especially with respect to elucidating the effect of empagliflozin on natremia in patients with heart failure and liver cirrhosis (51). Without generating high-quality data, empagliflozin may remain a promising agent with limited evidence base, used in an ‘‘off-label’’ manner only in selected cases, following the example of urea. For this reason, the scientific community should urge the pharmaceutical industry to undertake large RCTs, enabling application to regulatory authorities for this new indication.

Current and future role of empagliflozin

Could empagliflozin be another example of successful drug repurposing? Evidence to date suggests that, at present, empagliflozin combined with fluid restriction should be primarily used in outpatients with mild to moderate, chronic, paucisymptomatic hyponatremia due to SIAD, leading to a significant, but modest, sodium increase and possibly allowing relaxation of fluid restriction. However, the lack of long-term data except for a single case report (52) should limit its use to 4 weeks. The current role of empagliflozin is limited, if any, and merits further investigation in the following three scenarios:

i. In hospitalized patients since current guidelines recommend withholding SGLT2 inhibitors during admission for acute serious medical illness or major surgical procedures in view of the increased risk for DKA (53) (54), unless it is proven that this excess risk does not apply to patients without DM.

ii. In profound hyponatremia with serum sodium below 120 mmol/l, since therapeutic options with greater potency, such as hypertonic saline or tolvaptan, would be required to provide the desired increment of sodium rise.

iii. In hypervolemic hyponatremia due to heart failure or liver cirrhosis since all data on empagliflozin-related sodium increase are limited to SIAD. In the setting of acute heart failure, the data about empagliflozin are limited to a single study which examined its effects on renal glucose and sodium handling, reporting that empagliflozin significantly increased urinary output and produced osmotic diuresis through glycosuria, with these effects occurring within first 24 hours and remaining significant throughout a 30-day period (55). Despite empagliflozin-related osmotic diuresis, an effect of empagliflozin on serum sodium levels was not observed at any time point which might be explained by the fact that the majority of participants were eunatremic at the baseline, with the lowest serum sodium on admission being 129 mmol/l (55). Conflicting data are available about the effect of dapagliflozin, another SGLT2 inhibitor, on serum sodium concentration in patients with heart failure. On the one hand, findings from a large trial suggested that dapagliflozin did not result in statistically significant increase of serum sodium in those with established hyponatremia (56). On the other hand, a small study of hospitalized patients with acute heart failure demonstrated that dapagliflozin administration for 48 hours was associated with a statistically significant mean increase in serum sodium by 3 mmol/l compared to the control group and a higher resolution rate of hyponatremia (57). In total, the effect of empagliflozin on serum sodium in patients with heart failure and established hyponatremia has not been determined yet.

Discussion

Beyond the established role of empagliflozin as a glucose-lowering agent and the recently added indications for heart failure and CKD, the emerging question is whether empagliflozin could gain a fourth indication for the management of euvolemic hyponatremia. Recent prospective trials have provided evidence in favor of its efficacy and safety for treatment of SIAD, both in the inpatient (11) and outpatient setting (12), mediated by increased free renal water excretion as a result of glycosuria-induced osmotic diuresis. However, the small sample size and short duration of treatment limit the generalizability of these findings which warrant further exploration in large prospective trials, with several research questions remaining to be answered. Moving forward, skepticism should balance enthusiasm. Even if future studies confirm the favorable efficacy-safety profile of empagliflozin, the small magnitude of sodium increase would not render it the new wonder drug for SIAD, but rather a valuable addition to the current treatment options for SIAD. Its primary use would be as an adjunct to fluid restriction in those with mild to moderate chronic euvolemic hyponatremia, but its administration as add-on to oral urea for those with inadequate response to it may also be an effective treatment option.

Author contributions

PT: Conceptualization, Data curation, Formal analysis, Methodology, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Cuesta M, Garrahy A, Slattery D, Gupta S, Hannon AM, McGurren K, et al. Mortality rates are lower in siad, than in hypervolaemic or hypovolaemic hyponatraemia; results of a prospective observational study. Clin Endocrinol (Oxf). (2017) 87(4):400–6. doi: 10.1111/cen.2017.87.issue-4

PubMed Abstract | Crossref Full Text | Google Scholar

2. Cuesta M, Thompson CJ. The syndrome of inappropriate antidiuresis (SIAD). Best Pract Res Clin Endocrinol Metab. (2016) 30:175–87. doi: 10.1016/j.beem.2016.02.009

PubMed Abstract | Crossref Full Text | Google Scholar

3. Garrahy A, Galloway I, Hannon AM, Dineen R, O’Kelly P, Torney WP, et al. Fluid restriction therapy for chronic SIAD; results of a prospective randomized controlled trial. J Clin Endocrinol Metab. (2020) 105(12):e4360–e4369. doi: 10.1210/clinem/dgaa619

Crossref Full Text | Google Scholar

4. Cuesta MI, Ortol Aacute A, Garrahy A, Luis Calle Pascual A, Runkle I, Thompson CJ. Predictors of failure to respond to fluid restriction in siad in clinical practice; time to re-evaluate clinical guidelines? QJM. (2017) 110(8):489–92. doi: 10.1093/qjmed/hcx036

PubMed Abstract | Crossref Full Text | Google Scholar

5. Verbalis JG, Greenberg A, Burst V, Haymann JP, Johansson G, Peri A, et al. Diagnosing and treating the syndrome of inappropriate antidiuretic hormone secretion. Am J Med. (2016) 129:537.e9– e23. doi: 10.1016/j.amjmed.2015.11.005

PubMed Abstract | Crossref Full Text | Google Scholar

6. Rondon-Berrios H, Tandukar S, Mor MK, Ray EC, Bender FH, Kleyman TR, et al. Urea for the treatment of hyponatremia. Clin J Am Soc Nephrol. (2018) 13:1627–32. doi: 10.2215/CJN.04020318

PubMed Abstract | Crossref Full Text | Google Scholar

7. Sterns RH, Silver SM, Hix JK. Urea for hyponatremia? Kidney Int. (2015) 87:268–70. doi: 10.1038/ki.2014.320

PubMed Abstract | Crossref Full Text | Google Scholar

8. Schrier RW, Gross P, Gheorghiade M, Berl T, Verbalis JG, Czerwiec FC, et al. Tolvaptan, a selective oral vasopressin V2-receptor antagonist, for hyponatremia. N Engl J Med. (2006) 355:2099–112. doi: 10.1056/NEJMoa065181

PubMed Abstract | Crossref Full Text | Google Scholar

9. Tzoulis P, Waung JA, Bagkeris E, Carr H, Khoo B, Cohen M, et al. Real-life experience of tolvaptan use in the treatment of severe hyponatraemia due to syndrome of inappropriate antidiuretic hormone secretion. Clin Endocrinol (Oxf). (2016) 84:620–6. doi: 10.1111/cen.2016.84.issue-4

PubMed Abstract | Crossref Full Text | Google Scholar

10. Tzoulis P, Kaltsas G, Baldeweg SE, Bouloux PM, Grossman AB. Tolvaptan for the treatment of the syndrome of inappropriate antidiuresis (SIAD). Ther Adv Endocrinol Metab. (2023) 14:20420188231173327. doi: 10.1177/20420188231173327

PubMed Abstract | Crossref Full Text | Google Scholar

11. Refardt J, Imber C, Sailer CO, Jeanloz N, Potasso L, Kutz A, et al. A randomized trial of empagliflozin to increase plasma sodium levels in patients with the syndrome of inappropriate antidiuresis. J Am Soc Nephrol. (2020) 31:615–24. doi: 10.1681/ASN.2019090944

PubMed Abstract | Crossref Full Text | Google Scholar

12. Refardt J, Imber C, Nobbenhuis R, Sailier CO, Haslbauer A, Monnerat S, et al. Treatment effect of the SGLT2 inhibitor empagliflozin on chronic syndrome of inappropriate antidiuresis: results of a randomized, double-blind, placebo-controlled, crossover trial. J Am Soc Nephrol. (2023) 34:322–32. doi: 10.1681/ASN.2022050623

PubMed Abstract | Crossref Full Text | Google Scholar

13. Haring HU, Merker L, Seewaldt-Becker E, Weimer M, Meinicke T, Broedl UC, et al. Empagliflozin as add-on to metformin in patients with type 2 diabetes: a 24-week, randomized, double-blind, placebo-controlled trial. Diabetes Care. (2014) 37:1650–9. doi: 10.2337/dc13-2105

PubMed Abstract | Crossref Full Text | Google Scholar

14. Haring HU, Merker L, Seewaldt-Becker E, Weimer M, Meinicke T, Woerle HJ, et al. Empagliflozin as add-on to metformin plus sulfonylurea in patients with type 2 diabetes: a 24-week, randomized, double-blind, placebo-controlled trial. Diabetes Care. (2013) 36:3396–404. doi: 10.2337/dc12-2673

PubMed Abstract | Crossref Full Text | Google Scholar

15. Terauchi Y, Utsunomiya K, Yasui A, Seki T, Cheng G, Shiki K, et al. Safety and efficacy of empagliflozin as add-on therapy to GLP-1 receptor agonist (Liraglutide) in Japanese patients with type 2 diabetes mellitus: A randomised, double-blind, parallel-group phase 4 study. Diabetes Ther. (2019) 10:951–63. doi: 10.1007/s13300-019-0604-8

PubMed Abstract | Crossref Full Text | Google Scholar

16. Rosenstock J, Jelaska A, Zeller C, Klm G, Broedl UC, Woerle HJ, et al. Impact of empagliflozin added on to basal insulin in type 2 diabetes inadequately controlled on basal insulin: a 78-week randomized, double-blind, placebo-controlled trial. Diabetes Obes Metab. (2015) 17:936–48. doi: 10.1111/dom.2015.17.issue-10

PubMed Abstract | Crossref Full Text | Google Scholar

17. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantle S, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. (2015) 373:2117–28. doi: 10.1056/NEJMoa1504720

PubMed Abstract | Crossref Full Text | Google Scholar

18. Davies MJ, Aroda VR, Collins BS, Gabbay RA, Green J, Maruthur NM, et al. Management of hyperglycaemia in type 2 diabetes, 2022. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia. (2022) 65:1925–66. doi: 10.1007/s00125-022-05787-2

PubMed Abstract | Crossref Full Text | Google Scholar

19. Packer M, Anker SD, Butler J, Filipattos G, Pocock SJ, Carson P, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med. (2020) 383:1413–24. doi: 10.1056/NEJMoa2022190

PubMed Abstract | Crossref Full Text | Google Scholar

20. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Bohm M, et al. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med. (2021) 385:1451–61. doi: 10.1056/NEJMoa2107038

PubMed Abstract | Crossref Full Text | Google Scholar

21. The E-KCG, Herrington WG, Staplin N, Wanner C, Green JB, Hauske SJ, et al. Empagliflozin in patients with chronic kidney disease. N Engl J Med. (2023) 388:117–27. doi: 10.1056/NEJMoa2204233

PubMed Abstract | Crossref Full Text | Google Scholar

22. Zelniker TA, Braunwald E. Mechanisms of cardiorenal effects of sodium-glucose cotransporter 2 inhibitors: JACC state-of-the-art review. J Am Coll Cardiol. (2020) 75:422–34. doi: 10.1016/j.jacc.2019.11.031

PubMed Abstract | Crossref Full Text | Google Scholar

23. Alicic RZ, Neumiller JJ, Johnson EJ, Dieter B, Tuttle KR. Sodium-glucose cotransporter 2 inhibition and diabetic kidney disease. Diabetes. (2019) 68:248–57. doi: 10.2337/dbi18-0007

PubMed Abstract | Crossref Full Text | Google Scholar

24. Braunwald E. Gliflozins in the management of cardiovascular disease. N Engl J Med. (2022) 386:2024–34. doi: 10.1056/NEJMra2115011

PubMed Abstract | Crossref Full Text | Google Scholar

25. Marton A, Kaneko T, Kovalik JP, Yasui A, Nishiyama A, Kitada K, et al. Organ protection by SGLT2 inhibitors: role of metabolic energy and water conservation. Nat Rev Nephrol. (2021) 17:65–77. doi: 10.1038/s41581-020-00350-x

PubMed Abstract | Crossref Full Text | Google Scholar

26. Tang H, Xu C, Zhang P, Luo T, Huang Y, Yang X. A profile of SGLT-2 inhibitors in hyponatremia: The evidence to date. Eur J Pharm Sci. (2023) 184:106415. doi: 10.1016/j.ejps.2023.106415

PubMed Abstract | Crossref Full Text | Google Scholar

27. Tanaka H, Takano K, Iijima H, Kubo K, Maruama N, Hashimoto T, et al. Factors affecting canagliflozin-induced transient urine volume increase in patients with type 2 diabetes mellitus. Adv Ther. (2017) 34:436–51. doi: 10.1007/s12325-016-0457-8

PubMed Abstract | Crossref Full Text | Google Scholar

28. Zanchi A, Burnier M, Muller ME, Ghajarzadeh-Wurzner A, Mailard M, Loncle N, et al. Acute and chronic effects of SGLT2 inhibitor empagliflozin on renal oxygenation and blood pressure control in nondiabetic normotensive subjects: A randomized, placebo-controlled trial. J Am Heart Assoc. (2020) 9:e016173. doi: 10.1161/JAHA.119.016173

PubMed Abstract | Crossref Full Text | Google Scholar

29. Onishi A, Fu Y, Patel R, Darshi M, Crespo-Masip M, Huang W, et al. A role for tubular Na(+)/H(+) exchanger NHE3 in the natriuretic effect of the SGLT2 inhibitor empagliflozin. Am J Physiol Renal Physiol. (2020) 319:F712–F28. doi: 10.1152/ajprenal.00264.2020

PubMed Abstract | Crossref Full Text | Google Scholar

30. Heise T, Jordan J, Wanner C, Heer M, Macha S, Mattheus M, et al. Pharmacodynamic effects of single and multiple doses of empagliflozin in patients with type 2 diabetes. Clin Ther. (2016) 38:2265–76. doi: 10.1016/j.clinthera.2016.09.001

PubMed Abstract | Crossref Full Text | Google Scholar

31. Sarafidis P, Ferro CJ, Morales E, Ortiz A, Malyszko J, Hojs R, et al. SGLT-2 inhibitors and GLP-1 receptor agonists for nephroprotection and cardio protection in patients with diabetes mellitus and chronic kidney disease. A consensus statement by the EURECA-m and the DIABESITY working groups of the ERA-EDTA. Nephrol Dial Transplant. (2019) 34:208–30. doi: 10.1093/ndt/gfy407

PubMed Abstract | Crossref Full Text | Google Scholar

32. Chung S, Kim S, Son M, Kim M, Koh ES, Shin SJ, et al. Empagliflozin contributes to polyuria via regulation of sodium transporters and water channels in diabetic rat kidneys. Front Physiol. (2019) 10:271. doi: 10.3389/fphys.2019.00271

PubMed Abstract | Crossref Full Text | Google Scholar

33. Refardt J, Winzeler B, Meienberg F, Vogt DR, Christ-Crain M. Empagliflozin increases short-term urinary volume output in artificially induced syndrome of inappropriate antidiuresis. Int J Endocrinol. (2017) 2017:7815690. doi: 10.1155/2017/7815690

PubMed Abstract | Crossref Full Text | Google Scholar

34. Berton AM, Parasiliti-Caprino M, Prencipe N, Bioletto F, Lopez C, Bona C, et al. Copeptin adaptive response to SGLT2 inhibitors in patients with type 2 diabetes mellitus: The GliRACo study. Front Neurosci. (2023) 17:1098404. doi: 10.3389/fnins.2023.1098404

PubMed Abstract | Crossref Full Text | Google Scholar

35. Llorens-Cortes C, Moos F. Apelin and vasopressin: two work better than one. J Neuroendocrinol. (2012) 24:1085–6. doi: 10.1111/j.1365-2826.2012.02316.x

PubMed Abstract | Crossref Full Text | Google Scholar

36. Galanth C, Hus-Citharel A, Li B, Llorens-Cortes C. Apelin in the control of body fluid homeostasis and cardiovascular functions. Curr Pharm Des. (2012) 18:789–98. doi: 10.2174/138161212799277770

PubMed Abstract | Crossref Full Text | Google Scholar

37. Girault-Sotias PE, Gerbier R, Flahault A, de Mota N, Llorens-Cortes C. Apelin and vasopressin: the yin and yang of water balance. Front Endocrinol (Lausanne). (2021) 12:735515. doi: 10.3389/fendo.2021.735515

PubMed Abstract | Crossref Full Text | Google Scholar

38. Monnerat S, Drivakos N, Chapman FA, Dhaun N, Refardt J, Christ-Crain M. Apelin and copeptin levels in patients with chronic SIAD treated with empagliflozin. J Endocr Soc. (2024) 8:bvae106. doi: 10.1210/jendso/bvae106

PubMed Abstract | Crossref Full Text | Google Scholar

39. Nicco C, Wittner M, DiStefano A, Jounier S, Bankir L, Bouby N. Chronic exposure to vasopressin upregulates ENaC and sodium transport in the rat renal collecting duct and lung. Hypertension. (2001) 38:1143–9. doi: 10.1161/hy1001.092641

PubMed Abstract | Crossref Full Text | Google Scholar

40. Warren AM, Grossmann M, Christ-Crain M, Russell N. Syndrome of inappropriate antidiuresis: from pathophysiology to management. Endocr Rev. (2023) 44:819–61. doi: 10.1210/endrev/bnad010

PubMed Abstract | Crossref Full Text | Google Scholar

41. Potasso L, Refardt J, Meier C, Christ-Crain M. Effect of hyponatremia normalization on osteoblast function in patients with SIAD. Eur J Endocrinol. (2021) 186:1–8. doi: 10.1530/EJE-21-0604

PubMed Abstract | Crossref Full Text | Google Scholar

42. Monnerat S, Refardt J, Potasso L, Meier C, Christ-Crain M. An increase in plasma sodium levels is associated with an increase in osteoblast function in chronic SIAD. J Clin Endocrinol Metab. (2023) 108:e1027–e33. doi: 10.1210/clinem/dgad238

PubMed Abstract | Crossref Full Text | Google Scholar

43. Kohler S, Salsali A, Hantel S, Kaspers S, Woerle HJ, Kim G, et al. Safety and tolerability of empagliflozin in patients with type 2 diabetes. Clin Ther. (2016) 38:1299–313. doi: 10.1016/j.clinthera.2016.03.031

PubMed Abstract | Crossref Full Text | Google Scholar

44. Kohler S, Zeller C, Iliev H, Kaspers S. Safety and tolerability of empagliflozin in patients with type 2 diabetes: pooled analysis of phase I-III clinical trials. Adv Ther. (2017) 34:1707–26. doi: 10.1007/s12325-017-0573-0

PubMed Abstract | Crossref Full Text | Google Scholar

45. Monnerat S, Atila C, Refardt J, Christ-Crain M. Prevalence of admission hyponatremia in patients with diabetes treated with and without an SGLT2 inhibitor. J Endocr Soc. (2023) 7:bvad011. doi: 10.1210/jendso/bvad011

PubMed Abstract | Crossref Full Text | Google Scholar

46. Ingelfinger JR, Rosen CJ. Cardiovascular risk and sodium-glucose cotransporter 2 inhibition in type 2 diabetes. N Engl J Med. (2015) 373:2178–9. doi: 10.1056/NEJMe1512602

PubMed Abstract | Crossref Full Text | Google Scholar

47. Wanner C, Inzucchi SE, Zinman B. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med. (2016) 375:1801–2. doi: 10.1056/NEJMoa1515920

PubMed Abstract | Crossref Full Text | Google Scholar

48. August P. Chronic kidney disease - another step forward. N Engl J Med. (2023) 388:179–80. doi: 10.1056/NEJMe2215286

PubMed Abstract | Crossref Full Text | Google Scholar

49. Fralick M, Schneeweiss S, Patorno E. Risk of diabetic ketoacidosis after initiation of an SGLT2 inhibitor. N Engl J Med. (2017) 376:2300–2. doi: 10.1056/NEJMc1701990

PubMed Abstract | Crossref Full Text | Google Scholar

50. Kani R, Watanabe A, Miyamoto Y, Ejiri K, Iwagami M, Tagaki H, et al. Comparison of effectiveness among different sodium-glucose cotransoporter-2 inhibitors according to underlying conditions: A network meta-analysis of randomized controlled trials. J Am Heart Assoc. (2024) 13:e031805. doi: 10.1161/JAHA.123.031805

PubMed Abstract | Crossref Full Text | Google Scholar

51. Siafarikas C, Kapelios CJ, Papatheodoridi M, Vlachogiannakos J, Tentolouris N, Papatheodoridis G. Sodium-glucose linked transporter 2 inhibitors in liver cirrhosis: Beyond their antidiabetic use. Liver Int. (2024) 44:884–93. doi: 10.1111/liv.15851

PubMed Abstract | Crossref Full Text | Google Scholar

52. Bioletto F, Varaldo E, Prencipe N, Benso A, Berton AM. Long-term efficacy of empagliflozin as an add-on treatment for chronic SIAD: a case report and literature review. Hormones (Athens). (2023) 22:343–7. doi: 10.1007/s42000-023-00430-0

PubMed Abstract | Crossref Full Text | Google Scholar

53. 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) 104:3077–87. doi: 10.1210/jc.2019-00139

PubMed Abstract | Crossref Full Text | Google Scholar

54. 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) 22:753–62. doi: 10.4158/EP161292.PS

PubMed Abstract | Crossref Full Text | Google Scholar

55. Boorsma EM, Beusekamp JC, Ter Maaten JM, Figarska SM, Danser AHJ, Van Veldhuisen DJ, et al. Effects of empagliflozin on renal sodium and glucose handling in patients with acute heart failure. Eur J Heart Fail. (2021) 23:68–78. doi: 10.1002/ejhf.v23.1

PubMed Abstract | Crossref Full Text | Google Scholar

56. Yeoh SE, Docherty KF, Jhund PS, Petrie MS, Inzucchi SE, Kober L, et al. Relationship of dapagliflozin with serum sodium: findings from the DAPA-HF trial. JACC Heart Fail. (2022) 10:306–18. doi: 10.1016/j.jchf.2022.01.019

PubMed Abstract | Crossref Full Text | Google Scholar

57. Charaya K, Shchekochikhin D, Agadzhanyan A, Vashkevich M, Chashkina M, Kulikov V, et al. Impact of dapagliflozin treatment on serum sodium concentrations in acute heart failure. Cardiorenal Med. (2023) 13:101–8. doi: 10.1159/000529614

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: empagliflozin, SGLT 2 inhibitor, hyponatremia, SIAD(H), AVP

Citation: Tzoulis P (2024) Empagliflozin: a wonder drug for the treatment of SIAD? Front. Endocrinol. 15:1453159. doi: 10.3389/fendo.2024.1453159

Received: 22 June 2024; Accepted: 20 September 2024;
Published: 07 October 2024.

Edited by:

Andras Balla, Semmelweis University, Hungary

Reviewed by:

Alessandro Maria Berton, University Hospital of the City of Health and Science of Turin, Italy

Copyright © 2024 Tzoulis. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ploutarchos Tzoulis, ptzoulis@yahoo.co.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.