Skip to main content

Lack of durable natriuresis and objective decongestion following SGLT2 inhibition in randomized controlled trials of patients with heart failure

Abstract

Patients with heart failure have increased cardiac filling pressures, circulating natriuretic peptides, and physical signs of fluid retention, which are related to sodium retention by the kidneys and are alleviated by conventional diuretics. Sodium-glucose cotransporter 2 (SGLT2) inhibitors interfere with sodium and glucose reabsorption in the proximal renal tubule, but they evoke a marked counterregulatory activation of sodium and water reabsorption in distal nephron segments, which opposes and negates any diuretic effect. Nevertheless, it has been postulated that SGLT2 inhibitors modulate the volume set point, leading selectively to decongestion in patients with fluid overload. This hypothesis was tested in a review of 15 randomized controlled trials of SGLT2 inhibitors in patients with heart failure, with 7 trials focusing on urinary volume within the first week, and 8 trials focusing on objective decongestion at 12 weeks. In trials < 1 week, SGLT2 inhibition increased urine volume in the first 24 h, but typically without a change in urinary sodium excretion, and this diuresis was not sustained. In 8 trials of 12 weeks’ duration, none reported alleviation of edema, ascites or pulmonary rales. The 2 trials that evaluated changes in left ventricular filling pressure noted no or small changes (1–2 mm Hg); the two trials that measured interstitial lung water or total blood volume found no effect; and 6 of the 7 trials found no decrease in circulating natriuretic peptides. Therefore, randomized controlled trials do not indicate that SGLT2 inhibitors produce a durable natriuresis or objective decongestion in patients with heart failure.

Patients with acutely decompensated or chronic heart failure experience dyspnea at rest or exertion, which is typically accompanied by increased cardiac filling pressures and circulating levels of natriuretic peptides as well as physical signs of fluid retention (i.e., edema, ascites, pulmonary rales or pleural effusion). Many of these clinical and physiological abnormalities are related to the retention of sodium and water by the kidneys, and accordingly, treatment with loop diuretics or aquaretics are accompanied by symptomatic improvement, a reduction in cardiac filling pressures and natriuretic peptides, and alleviation of physical signs of congestion [1,2,3,4]. Increases in right and left ventricular filling pressures can be substantially reduced by loop diuretic therapy (typically by > 10 mm Hg) in patients with or without overt physical signs of fluid retention (i.e., jugular venous distension, ascites or peripheral edema) [4].

Renal tubular actions of SGLT2 inhibitors

Conventional diuretics and aquaretics act on the loop of Henle and more distal segments of the nephron. In contrast, sodium-glucose cotransporter 2 (SGLT2) inhibitors act in the most proximal segments of the renal tubule (i.e., the S1 and S2 segments) and inhibit not only SGLT2, but also sodium-hydrogen exchanger 3 [5, 6]. This combined action increases tubular sodium, chloride and glucose, but it also evokes a marked counterregulatory activation of sodium and water reabsorption in the loop of Henle and more distal nephron segments, which is related to upregulation of vasopressin, aldosterone, a-ketoglutarate, uromodulin and carbonic anhydrase [7,8,9,10]. These compensatory mechanisms oppose and negate the natriuretic and osmotic diuretic response to SGLT2 inhibitors, leading to marked attenuation and truncation of any short-term increase in urine volume or sodium excretion [11, 12]. Nevertheless, it has been postulated that SGLT2 inhibitors modulate the volume set point, [13] leading selectively to a meaningful natriuresis and objective evidence of decongestion in patients with fluid overload, i.e., those with heart failure.

In order to evaluate this possibility, we evaluated all randomized controlled trials of SGLT2 inhibitors in patients with chronic or acutely decompensated heart failure for (1) evidence of a meaningful natriuresis; and (2) objective evidence of decongestion, assessed by physical signs and physiological testing.

Natriuretic effect of SGLT2 inhibitors in patients with heart failure

The natriuretic effect of drugs is generally assessed by the measurement of urinary sodium excretion. However, urinary sodium measurements can be made on 24-hour collection or on spot urine collections, with the former being subject to less sampling error. Nevertheless, even 24-hour urine collections are influenced by changes in dietary sodium or concomitant medications. These confounding influences are reduced if the data are collected in randomized controlled clinical trials. Body weight is typically used to assess the magnitude of a diuretic effect over short periods of time (e.g., < 1 week), but this metric cannot be used to assess the natriuretic effect of SGLT2 inhibitors over longer periods, because glycosuria causes weight loss through caloric loss in the urine [14]. Furthermore, because SGLT2 inhibitors induce erythropoiesis, increases in hemoglobin and hematocrit cannot be used to infer the occurrence of hemoconcentration and intravascular decongestion, even during treatment periods as short as 7 days [15, 16].

Seven randomized controlled trials have evaluated the effect of SGLT2 inhibitors on urinary sodium and volume in patients with chronic heart failure, Table 1 [17,18,19,20,21,22,23,24]. All patients had elevated levels of natriuretic peptides, implying the high likelihood of elevated cardiac filling pressures and fluid overload. Three trials evaluated patients with stable chronic heart failure, whereas four trials studied patients who had been hospitalized for worsening symptoms of heart failure. Most of the trials evaluated the effect of these drugs over short periods, typically less than 1–4 weeks, and all enrolled fewer than 100 patients.

In patients with acute or chronic heart failure, SGLT2 inhibition produced increases in urine volume in the first 24 h, but typically without a change in urinary sodium excretion. This increase may have been related to an osmotic diuresis secondary to enhanced glycosuria, but the diuresis was generally not sustained beyond 24 h, presumably related to the activation of compensatory mechanisms, i.e., vasopressin [10]. There were no consistent decreases in body weight, circulating natriuretic peptides or dyspnea scores during observation periods of < 1 week. Only one study reported a persistent natriuretic effect accompanied by a decline in body weight after 2 weeks, [17] but interestingly, this study enrolled clinically euvolemic patients who had the lowest levels of circulating natriuretic peptides among all the trials in Table 1. Although some have proposed that SGLT2 inhibitors may potentiate the effects of loop diuretics, the natriuretic potentiation effect with SGLT2 inhibitors (if any) was less marked than with metolazone [24]. There was little evidence that SGLT2 inhibitors were particularly likely to produce a natriuresis in patients with notable fluid overload.

Table 1 Effect of SGLT2 Inhibitors on Urinary Sodium Excretion and Objective Evidence of Congestion in Randomized Controlled Trials of Patients With Heart Failure

Effect of SGLT2 inhibitors on objective signs of decongestion

Many physicians consider dyspnea (at rest or during exertion) as evidence of symptomatic congestion. The effects of dyspnea or effort tolerance and health status can be assessed by formal exercise testing (e.g., a 6-minute walk distance or cardiopulmonary exercise testing) or by the Kansas City Cardiomyopathy Questionnaire (KCCQ), but these assessments may not be concordant [25]. Furthermore, by improving ventricular performance or by effects on the peripheral circulation or skeletal muscle performance, many drugs for heart failure (e.g., beta-blockers and systemic vasodilators) can alleviate dyspnea and improve exercise capacity in the absence of any effect on the kidney to promote urinary sodium and water excretion [26, 27]. Therefore, this review did not regard a reduction in the symptoms of heart failure (e.g., an improvement in KCCQ scores) as providing reliable evidence of decongestion. Instead, the current analysis focused its attention on objective evidence for the alleviation of fluid retention. Objective decongestion was defined as the alleviation of physical signs of fluid retention (i.e., edema, ascites, pulmonary rales or pleural effusion). Whenever available, the assessment of objective decongestion was supported by changes in left ventricular filling pressure or natriuretic peptides, even though it is understood that these variables can improve markedly without an effect of treatment on urinary sodium or water excretion [26,27,28,29].

Eight randomized controlled trials have evaluated the effect of SGLT2 inhibitors on symptoms of heart failure, exercise capacity or health status — together with measures of left ventricular filling pressure, natriuretic peptides or objective evidence of congestion — typically in multicenter trials of < 600 patients, studied for 12 weeks [30,31,32,33,34,35,36,37,38,39,40,41]. Although many of these trials reported an improvement in KCCQ scores, no trial reported alleviation of physical signs of congestion, i.e., edema, ascites, pulmonary rales or pleural effusion. One trial reported a lessening of “clinical congestion” in patients with acutely decompensated heart failure after 15 days, [39,40,41] but in this trial, clinical congestion was assessed entirely by symptoms, and the investigators did not report changes in physical signs of fluid retention, even though these were apparent at the time of randomization. The two trials that evaluated changes in pulmonary capillary wedge pressure or pulmonary arterial diastolic pressure noted no or small changes (i.e., 1–2 mm Hg) [31, 34]. The one trial that measured total blood volume noted no treatment effect [32]. Six of the seven trials that evaluated changes in circulating natriuretic peptides found no effect of SGLT2 inhibition. One trial assessed changes in lung fluid volume after 12 weeks using noninvasive impedance measurements and found no difference between patients receiving an SGLT2 inhibitor and those receiving placebo [38]. Reported changes in body weight and hematocrit in some trials could be readily attributable to urinary caloric loss [14] (rather than diuresis) or to erythropoiesis [15, 16] (rather than hemoconcentration).

Conclusions

Conventional diuretics produce a natriuresis that acts to alleviate objective signs of fluid retention in patients with heart failure, thus minimizing jugular venous distension, pulmonary rales, ascites and peripheral edema [1,2,3,4]. In contrast, the action of SGLT2 inhibitors in the proximal renal tubules elicits a vigorous counterregulatory response that minimizes any natriuretic or osmotic diuretic effect. As a result, SGLT2 inhibitors have not been shown to produce a durable natriuretic effect or to alleviate objective signs of congestion in randomized controlled trials (Table 1). In these trials, SGLT2 inhibitors did not generally produce meaningful changes in pulmonary wedge pressure, interstitial lung fluid or circulating natriuretic peptides during treatment periods lasting up to 12 weeks. Therefore, the effect of SGLT2 inhibitors to improve health status (i.e., KCCQ scores) and cardiac structure and function as well as decreasing the risk of cardiovascular death or hospitalization for heart failure [42, 43] are likely due to effects that are unrelated to a discernable action of SGLT2 inhibitors in the kidney during the first 3 months of treatment [44]. This conclusion is supported by observations that SGLT2 inhibitors produce cardioprotective effects in isolated cardiomyocytes (that do not express SGLT2) and in animal models in which SGLT2 has been knocked out [44, 45].

Data Availability

There are no original data in this paper.

References

  1. Patterson JH, Adams KF Jr, Applefeld MM, Corder CN, Masse BR. Oral torsemide in patients with chronic congestive heart failure: effects on body weight, edema, and electrolyte excretion. Torsemide Investigators Group Pharmacotherapy. 1994;14:514–21.

    CAS  PubMed  Google Scholar 

  2. Gheorghiade M, Gattis WA, O’Connor CM, Adams KF Jr, Elkayam U, Barbagelata A, Ghali JK, Benza RL, McGrew FA, Klapholz M, Ouyang J, Orlandi C. Acute and Chronic Therapeutic Impact of a Vasopressin antagonist in congestive heart failure (ACTIV in CHF) investigators. Effects of tolvaptan, a vasopressin antagonist, in patients hospitalized with worsening heart failure: a randomized controlled trial. JAMA. 2004;291:1963–71.

    Article  CAS  PubMed  Google Scholar 

  3. Westheim AS, Bostrøm P, Christensen CC, Parikka H, Rykke EO, Toivonen L. Hemodynamic and neuroendocrine effects for candoxatril and frusemide in mild stable chronic heart failure. J Am Coll Cardiol. 1999;34:1794–801.

    Article  CAS  PubMed  Google Scholar 

  4. Chomsky DB, Lang CC, Rayos G, Wilson JR. Treatment of subclinical fluid retention in patients with symptomatic heart failure: effect on exercise performance. J Heart Lung Transplant. 1997;16:846–53.

    CAS  PubMed  Google Scholar 

  5. Borges-Júnior FA, Silva Dos Santos D, Benetti A, Polidoro JZ, Wisnivesky ACT, Crajoinas RO, Antônio EL, Jensen L, Caramelli B, Malnic G, Tucci PJ, Girardi ACC. Empagliflozin inhibits proximal tubule NHE3 activity, preserves GFR, and restores euvolemia in nondiabetic rats with induced heart failure. J Am Soc Nephrol. 2021;321616–29.

  6. Onishi A, Fu Y, Patel R, Darshi M, Crespo-Masip M, Huang W, Song P, Freeman B, Kim YC, Soleimani M, Sharma K, Thomson SC, Vallon V. A role for tubular Na+/H + exchanger NHE3 in the natriuretic effect of the SGLT2 inhibitor empagliflozin. Am J Physiol Renal Physiol. 2020;319:F712–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Layton AT, Vallon V, Edwards A. Modeling oxygen consumption in the proximal tubule: effects of NHE and SGLT2 inhibition. Am J Physiol Renal Physiol. 2015;308:F1343–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zannad F, Ferreira JP, Butler J, Filippatos G, Januzzi JL, Sumin M, Zwick M, Saadati M, Pocock SJ, Sattar N, Anker SD, Packer M. Effect of empagliflozin on circulating proteomics in heart failure: mechanistic insights from the EMPEROR program. Eur Heart J. 2022;43:4991–5002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bahena-Lopez JP, Rojas-Vega L, Chávez-Canales M, Bazua-Valenti S, Bautista-Pérez R, Lee JH, Madero M, Vazquez-Manjarrez N, Alquisiras-Burgos I, Hernandez-Cruz A, Castañeda-Bueno M, Ellison DH, Gamba G. Glucose/fructose delivery to the distal nephron activates the sodium-chloride cotransporter via the calcium-sensing receptor. J Am Soc Nephrol. 2023;34:55–72.

    Article  PubMed  Google Scholar 

  10. Masuda T, Ohara K, Vallon V, Nagata D. SGLT2 inhibitor and loop diuretic induce different vasopressin and fluid homeostatic responses in nondiabetic rats. Am J Physiol Renal Physiol. 2022;323:F361–9.

    Article  CAS  PubMed  Google Scholar 

  11. Wilcox CS, Shen W, Boulton DW, Leslie BR, Griffen SC. Interaction between the sodium-glucose-linked transporter 2 inhibitor dapagliflozin and the loop diuretic bumetanide in normal human subjects. J Am Heart Assoc. 2018;7(4):e007046. https://doi.org/10.1161/JAHA.117.007046.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Heise T, Jordan J, Wanner C, Heer M, Macha S, Mattheus M, Lund SS, Woerle HJ, Broedl UC. Acute pharmacodynamic effects of empagliflozin with and without diuretic agents in patients with type 2 diabetes mellitus. Clin Ther. 2016;38:2248–64.

    Article  CAS  PubMed  Google Scholar 

  13. Borlaug BA, Testani JM. SGLT2 inhibitors and diuretics in heart failure: clicking reset on the renal volume setpoint? Eur Heart J. 2023 May;23:ehad345. https://doi.org/10.1093/eurheartj/ehad345.

  14. Ferrannini G, Hach T, Crowe S, Sanghvi A, Hall KD, Ferrannini E. Energy balance after sodium-glucose cotransporter 2 inhibition. Diabetes Care. 2015;38:1730–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yamada T, Sakaguchi K, Okada Y, Miura H, Otowa-Suematsu N, So A, Komada H, Hirota Y, Ohara T, Kuroki Y, Hara K, Matsuda T, Kishi M, Takeda A, Yokota K, Tamori Y, Ogawa W. Analysis of time-dependent alterations of parameters related to erythrocytes after ipragliflozin initiation. Diabetol Int. 2020;12:197–206.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Aberle J, Menzen M, Schmid SM, Terkamp C, Jaeckel E, Rohwedder K, Scheerer MF, Xu J, Tang W, Birkenfeld AL. Dapagliflozin effects on haematocrit, red blood cell count and reticulocytes in insulin-treated patients with type 2 diabetes. Sci Rep. 2020;10(1):22396. https://doi.org/10.1038/s41598-020-78734-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Griffin M, Rao VS, Ivey-Miranda J, Fleming J, Mahoney D, Maulion C, Suda N, Siwakoti K, Ahmad T, Jacoby D, Riello R, Bellumkonda L, Cox Z, Collins S, Jeon S, Turner JM, Wilson FP, Butler J, Inzucchi SE, Testani JM. Empagliflozin in heart failure: diuretic and cardiorenal effects. Circulation. 2020;142:1028–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kolwelter J, Kannenkeril D, Linz P, Jung S, Nagel AM, Bosch A, Ott C, Bramlage P, Nöh L, Schiffer M, Uder M, Achenbach S, Schmieder RE. The SGLT2 inhibitor empagliflozin reduces tissue sodium content in patients with chronic heart failure: results from a placebo-controlled randomised trial. Clin Res Cardiol. 2023;112:134–44.

    Article  CAS  PubMed  Google Scholar 

  19. Mordi NA, Mordi IR, Singh JS, McCrimmon RJ, Struthers AD, Lang CC. Renal and cardiovascular effects of SGLT2 inhibition in combination with loop diuretics in patients with type 2 diabetes and chronic heart failure: the RECEDE-CHF trial. Circulation. 2020;142:1713–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Schulze PC, Bogoviku J, Westphal J, Aftanski P, Haertel F, Grund S, von Haehling S, Schumacher U, Möbius-Winkler S, Busch M. Effects of early empagliflozin initiation on diuresis and kidney function in patients with acute decompensated heart failure (EMPAG-HF). Circulation. 2022;146:289–98.

    Article  CAS  PubMed  Google Scholar 

  21. Boorsma EM, Beusekamp JC, Ter Maaten JM, Figarska SM, Danser AHJ, van Veldhuisen DJ, van der Meer P, Heerspink HJL, Damman K, Voors AA. Effects of empagliflozin on renal sodium and glucose handling in patients with acute heart failure. Eur J Heart Fail. 2021;23:68–78.

    Article  CAS  PubMed  Google Scholar 

  22. Damman K, Beusekamp JC, Boorsma EM, Swart HP, Smilde TDJ, Elvan A, van Eck JWM, Heerspink HJL, Voors AA. Randomized, double-blind, placebo-controlled, multicentre pilot study on the effects of empagliflozin on clinical outcomes in patients with acute decompensated heart failure (EMPA-RESPONSE-AHF). Eur J Heart Fail. 2020;22:713–22.

    Article  CAS  PubMed  Google Scholar 

  23. Tamaki S, Yamada T, Watanabe T, Morita T, Furukawa Y, Kawasaki M, Kikuchi A, Kawai T, Seo M, Abe M, Nakamura J, Yamamoto K, Kayama K, Kawahira M, Tanabe K, Fujikawa K, Hata M, Fujita Y, Umayahara Y, Taniuchi S, Sanada S, Shintani A, Fukunami M. Effect of empagliflozin as an add-on therapy on decongestion and renal function in patients with diabetes hospitalized for acute decompensated heart failure: a prospective randomized controlled study. Circ Heart Fail. 2021;14(3):e007048. https://doi.org/10.1161/CIRCHEARTFAILURE.120.007048.

    Article  CAS  PubMed  Google Scholar 

  24. Yeoh SE, Osmanska J, Petrie MC, Brooksbank KJM, Clark AL, Docherty KF, Foley PWX, Guha K, Halliday CA, Jhund PS, Kalra PR, McKinley G, Lang NN, Lee MMY, McConnachie A, McDermott JJ, Platz E, Sartipy P, Seed A, Stanley B, Weir RAP, Welsh P, McMurray JJV, Campbell RT. Dapagliflozin versus metolazone in heart failure resistant to loop diuretics. Eur Heart J. 2023 May;21:ehad341. https://doi.org/10.1093/eurheartj/ehad341.

  25. Michelis KC, Grodin JL, Zhong L, Pandey A, Toto K, Ayers CR, Thibodeau JT, Drazner MH. Discordance between severity of heart failure as determined by patient report versus cardiopulmonary exercise testing. J Am Heart Assoc. 2021;10(13):e019864. https://doi.org/10.1161/JAHA.120.019864.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Krum H, Sackner-Bernstein JD, Goldsmith RL, Kukin ML, Schwartz B, Penn J, Medina N, Yushak M, Horn E, Katz SD, Levin HL, Neuberg GW, DeLong G, Packer M. Double-blind, placebo-controlled study of the long-term efficacy of carvedilol in patients with severe chronic heart failure. Circulation. 1995;92:1499–506.

    Article  CAS  PubMed  Google Scholar 

  27. Packer M, Narahara KA, Elkayam U, Sullivan JM, Pearle DL, Massie BM, Creager MA. Double-blind, placebo-controlled study of the efficacy of flosequinan in patients with chronic heart failure. Principal investigators of the REFLECT Study. J Am Coll Cardiol. 1993;22:65–72.

    Article  CAS  PubMed  Google Scholar 

  28. Burgdorf C, Brockmöller J, Strampe H, Januszewski M, Remppis BA. Reduction of pulmonary hypertension after transition to sacubitril/valsartan in patients with heart failure with preserved ejection fraction. Front Cardiovasc Med. 2021;8:734697. https://doi.org/10.3389/fcvm.2021.734697.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Myhre PL, Prescott MF, Murphy SP, Fang JC, Mitchell GF, Ward JH, Claggett B, Desai AS, Solomon SD, Januzzi JL. Early B-type natriuretic peptide change in HFrEF patients treated with sacubitril/valsartan: a pooled analysis of EVALUATE-HF and PROVE-HF. JACC Heart Fail. 2022;10:119–28.

    Article  PubMed  Google Scholar 

  30. Jensen J, Omar M, Kistorp C, Poulsen MK, Tuxen C, Gustafsson I, Køber L, Gustafsson F, Faber J, Fosbøl EL, Bruun NE, Brønd JC, Forman JL, Videbæk L, Møller JE, Schou M. Twelve weeks of treatment with empagliflozin in patients with heart failure and reduced ejection fraction: a double-blinded, randomized, and placebo-controlled trial. Am Heart J. 2020;228:47–56.

    Article  CAS  PubMed  Google Scholar 

  31. Omar M, Jensen J, Frederiksen PH, Kistorp C, Videbæk L, Poulsen MK, Möller S, Ali M, Gustafsson F, Køber L, Borlaug BA, Schou M, Møller JE. Effect of empagliflozin on hemodynamics in patients with heart failure and reduced ejection fraction. J Am Coll Cardiol. 2020;76:2740–51.

    Article  CAS  PubMed  Google Scholar 

  32. Omar M, Jensen J, Burkhoff D, Frederiksen PH, Kistorp C, Videbæk L, Poulsen MK, Gustafsson F, Køber L, Borlaug BA, Schou M, Møller JE. Effect of empagliflozin on blood volume redistribution in patients with chronic heart failure and reduced ejection fraction: an analysis from the Empire HF randomized clinical trial. Circ Heart Fail. 2022;15(3):e009156.

    Article  CAS  PubMed  Google Scholar 

  33. Nassif ME, Windsor SL, Borlaug BA, Kitzman DW, Shah SJ, Tang F, Khariton Y, Malik AO, Khumri T, Umpierrez G, Lamba S, Sharma K, Khan SS, Chandra L, Gordon RA, Ryan JJ, Chaudhry SP, Joseph SM, Chow CH, Kanwar MK, Pursley M, Siraj ES, Lewis GD, Clemson BS, Fong M, Kosiborod MN. The SGLT2 inhibitor dapagliflozin in heart failure with preserved ejection fraction: a multicenter randomized trial. Nat Med. 2021;27:1954–60.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Nassif ME, Qintar M, Windsor SL, Jermyn R, Shavelle DM, Tang F, Lamba S, Bhatt K, Brush J, Civitello A, Gordon R, Jonsson O, Lampert B, Pelzel J, Kosiborod MN. Empagliflozin effects on pulmonary artery pressure in patients with heart failure: results from the EMBRACE-HF trial. Circulation. 2021;143:1673–86.

    Article  CAS  PubMed  Google Scholar 

  35. Spertus JA, Birmingham MC, Nassif M, Damaraju CV, Abbate A, Butler J, Lanfear DE, Lingvay I, Kosiborod MN, Januzzi JL. The SGLT2 inhibitor canagliflozin in heart failure: the CHIEF-HF remote, patient-centered randomized trial. Nat Med. 2022;28:809–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Abraham WT, Lindenfeld J, Ponikowski P, Agostoni P, Butler J, Desai AS, Filippatos G, Gniot J, Fu M, Gullestad L, Howlett JG, Nicholls SJ, Redon J, Schenkenberger I, Silva-Cardoso J, Störk S, Krzysztof Wranicz J, Savarese G, Brueckmann M, Jamal W, Nordaby M, Peil B, Ritter I, Ustyugova A, Zeller C, Salsali A, Anker SD. Effect of empagliflozin on exercise ability and symptoms in heart failure patients with reduced and preserved ejection fraction, with and without type 2 diabetes. Eur Heart J. 2021;42:700–10.

    Article  CAS  PubMed  Google Scholar 

  37. Nassif ME, Windsor SL, Tang F, Khariton Y, Husain M, Inzucchi SE, McGuire DK, Pitt B, Scirica BM, Austin B, Drazner MH, Fong MW, Givertz MM, Gordon RA, Jermyn R, Katz SD, Lamba S, Lanfear DE, LaRue SJ, Lindenfeld J, Malone M, Margulies K, Mentz RJ, Mutharasan RK, Pursley M, Umpierrez G, Kosiborod M. Dapagliflozin effects on biomarkers, symptoms, and functional status in patients with heart failure with reduced ejection fraction: the DEFINE-HF trial. Circulation. 2019;140:1463–76.

    Article  CAS  PubMed  Google Scholar 

  38. Nassif ME, Windsor SL, Tang F, Husain M, Inzucchi SE, McGuire DK, Pitt B, Scirica BM, Austin B, Fong MW, LaRue SJ, Umpierrez G, Hartupee J, Khariton Y, Malik AO, Ogunniyi MO, Wenger NK, Kosiborod MN. Dapagliflozin effects on lung fluid volumes in patients with heart failure and reduced ejection fraction: results from the DEFINE-HF trial. Diabetes Obes Metab. 2021;23:1426–30.

    Article  CAS  PubMed  Google Scholar 

  39. Voors AA, Angermann CE, Teerlink JR, Collins SP, Kosiborod M, Biegus J, Ferreira JP, Nassif ME, Psotka MA, Tromp J, Borleffs CJW, Ma C, Comin-Colet J, Fu M, Janssens SP, Kiss RG, Mentz RJ, Sakata Y, Schirmer H, Schou M, Schulze PC, Spinarova L, Volterrani M, Wranicz JK, Zeymer U, Zieroth S, Brueckmann M, Blatchford JP, Salsali A, Ponikowski P. The SGLT2 inhibitor empagliflozin in patients hospitalized for acute heart failure: a multinational randomized trial. Nat Med. 2022;28:568–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Biegus J, Voors AA, Collins SP, Kosiborod MN, Teerlink JR, Angermann CE, Tromp J, Ferreira JP, Nassif ME, Psotka MA, Brueckmann M, Salsali A, Blatchford JP, Ponikowski P. Impact of empagliflozin on decongestion in acute heart failure: the EMPULSE trial. Eur Heart J. 2023;44:41–50.

    Article  CAS  PubMed  Google Scholar 

  41. Kosiborod MN, Angermann CE, Collins SP, Teerlink JR, Ponikowski P, Biegus J, Comin-Colet J, Ferreira JP, Mentz RJ, Nassif ME, Psotka MA, Tromp J, Brueckmann M, Blatchford JP, Salsali A, Voors AA. Effects of empagliflozin on symptoms, physical limitations, and quality of life in patients hospitalized for acute heart failure: results from the EMPULSE trial. Circulation. 2022;146:279–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lee MMY, Brooksbank KJM, Wetherall K, Mangion K, Roditi G, Campbell RT, Berry C, Chong V, Coyle L, Docherty KF, Dreisbach JG, Labinjoh C, Lang NN, Lennie V, McConnachie A, Murphy CL, Petrie CJ, Petrie JR, Speirits IA, Sourbron S, Welsh P, Woodward R, Radjenovic A, Mark PB, McMurray JJV, Jhund PS, Petrie MC, Sattar N. Effect of empagliflozin on left ventricular volumes in patients with type 2 diabetes, or prediabetes, and heart failure with reduced ejection fraction (SUGAR-DM-HF). Circulation. 2021;143:516–25.

    Article  CAS  PubMed  Google Scholar 

  43. Vaduganathan M, Docherty KF, Claggett BL, Jhund PS, de Boer RA, Hernandez AF, Inzucchi SE, Kosiborod MN, Lam CSP, Martinez F, Shah SJ, Desai AS, McMurray JJV, Solomon SD. SGLT-2 inhibitors in patients with heart failure: a comprehensive meta-analysis of five randomised controlled trials. Lancet. 2022;400:757–67.

    Article  CAS  PubMed  Google Scholar 

  44. Packer M. Critical reanalysis of the mechanisms underlying the cardiorenal benefits of SGLT2 inhibitors and reaffirmation of the nutrient deprivation signaling/autophagy hypothesis. Circulation. 2022;146:1383–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chen S, Wang Q, Christodoulou A, Mylonas N, Bakker D, Nederlof R, Hollmann MW, Weber NC, Coronel R, Wakker V, Christoffels VM, Andreadou I, Zuurbier CJ. Sodium glucose cotransporter-2 inhibitor empagliflozin reduces infarct size independently of sodium glucose cotransporter-2. Circulation. 2023;147:276–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

Milton Packer conceived of the work, carried out a comprehensive literature review, extracted the data, synthesized the concepts, and wrote the original draft and revised subsequent drafts of the paper.

Corresponding author

Correspondence to Milton Packer.

Ethics declarations

Competing interests

During the past three years, Dr. Packer reports personal fees for consulting from 89bio, Abbvie, Actavis, Amarin, Amgen, AstraZeneca, Boehringer Ingelheim, Caladrius, Casana, CSL Behring, Cytokinetics, Imara, Lilly, Moderna, Novartis, Pharmacocosmos, Reata, Relypsa, SalamandraDr. Packer was the chair of the Executive Committee for the EMPEROR Trial Program, which evaluated the effect of empagliflozin in patients with heart failure and a reduced or preserved ejection fraction (EMPEROR-Reduced and EMPEROR-Preserved, respectively. This work is not related to any of the competing interests, described above.

Ethics approval and consent to participate

Not applicable. Not applicable.

Consent for publication

There is only one author who takes responsibility for the entire work.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Packer, M. Lack of durable natriuresis and objective decongestion following SGLT2 inhibition in randomized controlled trials of patients with heart failure. Cardiovasc Diabetol 22, 197 (2023). https://doi.org/10.1186/s12933-023-01946-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12933-023-01946-w

Keywords