Review Article

Hyperkalaemia in Heart Failure

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

Hyperkalaemia has become an increasingly prevalent finding in patients with heart failure (HF), especially with renin–angiotensin–aldosterone system (RAAS) inhibitors and angiotensin–neprilysin inhibitors being the cornerstone of medical therapy. Patients living with HF often have other comorbidities, such as diabetes and chronic kidney disease, which predispose to hyperkalaemia. Until now, we have not had any reliable or tolerable therapies for the treatment of hyperkalaemia to facilitate implementation or achievement of target doses of RAAS inhibition. Patiromer sorbitex calcium and sodium zirconium cyclosilicate are two novel potassium-binding resins that have shown promise in the management of patients predisposed to developing recurrent hyperkalaemia, and their use may allow for further optimisation of guideline directed medical therapy.

Disclosure:SZ has received consulting and speaking fees from Astra Zeneca and Vifor; and is on the Cardiac Failure Review editorial board; this did not influence peer review.. All other authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Shelley Zieroth, Room Y3014, St Boniface Hospital, 409 Tache Avenue, Winnipeg, Manitoba R2H 2A6, Canada. E: szieroth@sbgh.mb.ca

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

Heart failure (HF) is defined as “a complex clinical syndrome that can result from any structural or functional cardiac disorder that impairs the ability of the ventricle to fill or eject blood”.1 Hyperkalaemia is commonly encountered in HF patients. Registry data estimate the prevalence to be between 16% and 25%.2,3 The definition of hyperkalaemia varies, but has generally been defined as a potassium level >5 mEq/l.4 It can be further subclassified into mild (5–5.5 mEq/l), moderate (5.5–6 mEq/l) and severe (>6 mEq/l).1

Hyperkalaemia poses a major obstacle in the management of HF patients. It can result in electrical issues from conduction system abnormalities to life-threatening rhythm disorders and interference with defibrillator function.5 It can also lead to the downtitration and discontinuation of renin–angiotensin–aldosterone system inhibitors (RAASi) and angiotensin–neprilysin inhibitors, which are the cornerstones of HF therapy.6,7 Furthermore, hyperkalaemia is a marker associated with an increase in hospital length of stay and mortality.8

Pathophysiology of Hyperkalaemia

Hyperkalaemia occurs due to decreased excretion or increased intake of potassium (Table 1). Potassium levels are tightly regulated in the body; under normal circumstances, 98% of potassium is intracellular, with 2% being extracellular in the plasma.9 Approximately 90% of the potassium filtered through the glomerulus will be reabsorbed in the proximal tubule and the loop of Henle, and 10% will reach the distal tubules.9 During times of renal hypoperfusion, such as in HF, renin is secreted by the juxtaglomerular cells in the kidneys; this allows angiotensinogen to be cleaved to angiotensin I, which is then converted to angiotensin II in the endothelial cells of the lungs under the influence of angiotensin-converting enzyme (ACE).10 Angiotensin II acts on the cells of the zona glomerulosa to secrete aldosterone, which in turn promotes sodium and water retention.10 Aldosterone activates the Na+/K+ pumps in the basolateral membrane of the distal tubule and collecting ducts, creating a concentration gradient that results in the reabsorption of sodium and secretion of potassium ions. It also upregulates the epithelial sodium channels and potassium efflux at the luminal surface leading to further reabsorption of sodium and excretion of potassium.10

Patients with HF often have other comorbidities, including diabetes, chronic kidney disease (CKD) and hypertension.1 Therefore, the aetiology of hyperkalaemia in patients with HF can be multifactorial, as seen in acute decompensated HF, cardiogenic shock and chronic HF.11,12 Kidney injury, which may be in the setting of cardiorenal syndrome (type I and type II), results in hyperkalaemia due to reduced renal potassium excretion.11 Some diabetic patients can develop a type IV renal tubular acidosis that can directly result in hyperkalaemia.10

In clinical practice, medications are the most common cause of hyperkalaemia in HF patients. ACE inhibitors (ACEi) and angiotensin receptor blockers (ARB) decrease the production of aldosterone in the adrenal gland, while mineralocorticoid receptor antagonists (MRA) block the receptor.13,14

Incidence and Significance of Hyperkalaemia in the Heart Failure Population

Clinical trials involving RAASi have demonstrated varying rates of hyperkalaemia due to variability in the definition of hyperkalaemia used. In SOLVD, there was a 7.8% occurrence of hyperkalaemia (defined as a potassium level >5.5 mEq/l) in the enalapril arm compared with placebo.15 In CHARM, the incidence of hyperkalaemia was 5.2%.16 In PARADIGM-HF, the incidence of hyperkalaemia (potassium level >5.5 mEq/l) was similar between the sacubitril/valsartan and the enalapril arms, but the incidence of severe hyperkalaemia was lower in the sacubitril/valsartan arm (4.3% versus 5.6%).17 Interestingly, a follow-up analysis of PARADIGM-HF showed that sacubitril/valsartan had no effect on potassium levels overall, and in fact, those on therapy had a slight reduction in potassium levels compared with enalapril, especially when the latter was combined with an MRA.18

Common Aetiologies for Hyperkalaemia in Heart Failure Patients

Article image

In EMPHASIS-HF, hyperkalaemia (defined as a potassium level >5.5 mEq/l) occurred in 11.8% of patients receiving eplerenone, whereas serious hyperkalaemia (potassium level >6 mEq/l) occurred in 2.5% of patients in the eplerenone group.19 In RALES, where an ACEi was combined with spironolactone, the incidence of hyperkalaemia (potassium level >5.5 mEq/l) was 13%, whereas serious hyperkalaemia (potassium level >6 mEq/l) was seen in only 2% of patients.20 Following the publication of RALES, data from Ontario, Canada showed a higher incidence of serious hyperkalaemia and higher rates of hospitalisation and in-hospital mortality. Those findings were later attributed to the widespread inappropriate prescription of MRAs.21,22

More recently, in January 2020, Rossignol et al. used the EPHESUS data to develop a risk prediction score that was then validated in the EMPHASIS cohort.23 The risk model incorporated multiple variables, including MRA use and time-dependent potassium levels. They found this model to be better than the MAGGIC score to describe the risk of cardiovascular events.23

A strong relationship between potassium levels and mortality in patients with chronic HF has been demonstrated in multiple trials.3,24,25 This may lead the clinician to consider withdrawal of therapies with proven mortality benefit, which may also lead to higher mortality.3

Beusekamp et al. analysed patients in the PROTECT trial to look for associations between hyperkalaemia and mortality in patients admitted for HF exacerbations.26 Potassium levels were followed daily until discharge or day 7 of admission. RAASi use was common, with 76% of patients on an ACEi and 46% on an MRA. Incident hyperkalaemia (defined as any potassium level >5 mEq/l) was seen in 35% of patients. Although incident hyperkalaemia was not associated with increased mortality at 180 days, it was associated with downtitration or even discontinuation of RAASi, especially MRAs, a variable that was independently associated with increased mortality.26

These findings are further supported by real-world data published from a large observational cohort of more than 114,000 patients (most had CKD and 13,113 had HF), where potassium levels >5 mEq/l led to downtitration of RAASi. This change was independently associated with major fatal and non-fatal cardiac events, as well as overall mortality.27 For MRAs in particular, registry analyses show that many other factors are associated with their underutilisation, including CKD, higher ejection fraction, male sex, older age and lower income.28

A Swedish nationwide registry found that the association between potassium levels and mortality in HF patients was U-shaped, and suggested that the optimal potassium level in HF patients is 4.2 mEq/l, with higher and lower levels associated with higher mortality.29 Interestingly, another analysis of the Swedish registry noted that the incidence of hyperkalaemia was higher in patients with HF with preserved ejection fraction, including severe hyperkalaemia.30 The 2016 European Society of Cardiology Guidelines recommend short-term cessation of RAASi and MRAs to manage acute hyperkalaemia, followed by careful reintroduction of the discontinued drug(s).4 These guidelines were published prior to the trials discussed above.

Acute Management of Hyperkalaemia

The most dreaded consequence of hyperkalaemia is the development of electrical abnormalities.31 As such, it is of paramount importance to stabilise the myocardial membrane with intravenous calcium, either in the form of calcium gluconate or calcium chloride, both of which act within 5 minutes.31 Potential side-effects include vasodilation-related hypotension and bradycardia. Calcium itself does not lower potassium levels in the bloodstream.31 Calcium chloride contains three times more elemental calcium than calcium gluconate. Therefore, in acute situations, calcium chloride is preferred. However, in less critical cases, the preferred agent is the gluconate salt, because it is less likely to cause tissue necrosis if it extravasates.32

To acutely lower the potassium levels in the blood, agents that shift potassium from the extracellular to the intracellular space are utilised (Figure 1A). The most commonly used agents are short-acting insulins in combination with 25–50g of dextrose (unless hyperglycaemic). Insulin shifts potassium into the intracellular compartment via the action of the Na+/K+ ATPase pump. It works rapidly within 15 minutes. β2 agonists work similarly. Salbutamol is usually given at a dose of 10 mg and acts within 15–30 minutes. Side-effects include tachycardia, anxiety and headaches. Sodium bicarbonate can also be considered if the acutely hyperkalaemic patient is acidotic.33

After stabilisation, the next step is elimination. This is typically accomplished with loop diuretics, commonly furosemide. Furosemide works by inhibiting the Na+/K+/2Cl cotransporter in the loop of Henle, and thus blocking sodium and potassium reabsorption. Increased sodium delivery to the distal nephron stimulates potassium excretion. Loop diuretics can precipitate hypokalaemia and are typically reserved for hyperkalaemic patients who are volume overloaded. Dialysis is recommended in cases of hyperkalaemia refractory to medical management (Figures 1B and 1C).34

The final step of hyperkalaemia management is maintenance. This is usually achieved on a long-term basis by eliminating the precipitants, minimising intake in the diet and reducing supplementation. Finally, binding resins can also be used if recurrent hyperkalaemia becomes an issue.5,35

Chronic Management of Hyperkalaemia in HF

Sodium Polystyrene Sulfate

Sodium polystyrene sulfate (SPS) is the oldest binding resin available. It is a non-specific binder that works to bind potassium ions in the gut in exchange for sodium (Figure 1D). As it is non-specific, it can also bind to calcium and magnesium. The potassium-bound SPS is eventually eliminated from the body in the faeces.36 The initial evidence for the use of SPS, culminating in its approval in hyperkalaemia, comes from two small trials published in the early 1960s.37,38

Mechanism of Action of Drugs for Hyperkalaemia

Article image

SPS is combined with sorbitol to relieve its constipating effects. It can be administered either as an oral (15 g 1–4 times/day) or rectal formulation (30–50 g every 6 hours). SPS use for hyperkalaemia is hampered by the fact that it has not been studied in large randomised controlled trials.39 One small trial of 33 outpatients with CKD evaluated the effect of SPS on mild hyperkalaemia (5–5.9 mmol/l) at a dose of 30 g for 7 days. The average reduction in potassium was 1.25 mEq/l. Normokalaemia was achieved in 73% of the patients on SPS.40

The main side-effects of SPS are gastrointestinal (GI), and include nausea, vomiting, constipation and diarrhoea. Serious side-effects have been reported with SPS, including colonic perforation and necrosis.41,42 This has been mainly attributed to the sorbitol added to SPS, which led to a black box warning in 2009 by the Food and Drug Administration. More relevant to a HF population, SPS can lead to worsening HF symptoms, as it exchanges potassium ions for sodium ions.43

Sodium Zirconium Cyclosilicate

Sodium zirconium cyclosilicate (SZC) is a novel binding resin. It is a non-absorbable, inorganic polymer of zirconium silicate, with the ion channels formed by its atomic structure mimicking those of endogenous potassium channels.44 It preferentially captures potassium ions, in exchange for hydrogen and sodium ions, through a selectivity filter (Figure 1D).

SZC works in the entirety of the GI tract; is not systemically absorbed and is excreted in the faeces. In vitro studies have shown that SZC can achieve a potassium equilibrium in as early as 20 minutes.45 As such, it can be used for acute (10 mg orally three-times a day for up to 48 hours) and chronic hyperkalaemia (5–15 mg orally daily).

SZC demonstrated significant efficacy in reducing K+ levels when used for hyperkalaemia in an emergency department setting compared with placebo, regardless of aetiology.46 When used as maintenance for chronic hyperkalaemia, SZC can be continued as long as it is clinically indicated.44 As it is not systemically absorbed following oral administration, SZC is thought to be safe in pregnant and lactating women.47

Studies that Evaluated Sodium Zirconium Cyclosilicate in Hyperkalaemia

Article image

ZS-003 was a phase III trial that randomised 753 hyperkalaemic patients to receive SZC or placebo three times daily for 48 hours (see Table 2).44 Patients who achieved normokalaemia were then reassigned to receive placebo or SZC once daily for 2 weeks. Serum potassium was significantly lower in patients receiving the 5 and 10 g doses of SZC compared with placebo. The most common side-effects were GI related and occurred in 12.9% of patients on SZC.

HARMONIZE enrolled 258 ambulatory patients with hyperkalaemia. In the first 48 hours, they received 10 g SZC three-times a day (see Table 2). Nearly all were normokalaemic at 48 hours.48 This was followed by a maintenance phase, in which patients who achieved normokalaemia were randomised to receive SZC or placebo daily for 28 days. Normokalaemia was higher in all treatment groups versus placebo. Oedema was more common in SZC (2–6%). GI side-effects occurred in 2–9% of the patients on SZC, and these were dose dependent. HARMONIZE-Global was a follow-up trial that enrolled patients with a more diverse geographical distribution. Results were congruent with HARMONIZE, with 77.3% of the SZC group achieving normokalaemia compared with only 24% in the placebo group.49 In the subgroup of HF patients (n=94) from HARMONIZE, all three doses of SZC were effective in lowering and maintaining normokalaemia, including in those taking RAASi (n=60) .50

More recently, Spinowitz et al. assessed SZC in a single-arm study. SZC was able to acutely achieve normokalaemia during the first 72 hours of administration in nearly all patients (see Table 2).51 Following a 12-month maintenance period, 88% were normokalaemic and this enabled the initiation and/or uptitration of RAASi.51

Patiromer Sorbitex Calcium

Patiromer sorbitex calcium (patiromer) is a 100 µm bead organic polymer that preferentially uses calcium ions to exchange potassium ions (Figure 1D), although it can bind to magnesium and sodium. Patiromer is not systemically absorbed and is excreted in the faeces.52 It can be given as a starting dose of 8.4 g, and is increased in 4.2 g increments up to a dose of 25.2 g. It has a slow onset of action of approximately 7 hours, and therefore is only approved for chronic hyperkalaemia. Patiromer can be continued as long as it is clinically indicated.53 As it is not systemically absorbed following oral administration, SZC is thought to be safe in pregnant and lactating women.54

OPAL-HK was a study of 237 CKD patients with hyperkalaemia who were on RAASi (see Table 3).55 The study was divided into two phases: an initial 4-week phase where everyone received patiromer twice daily, followed by an 8-week withdrawal phase, in which patients were randomly assigned to continue the initial dose or placebo. By the end of the first phase, 76% of the study population were normokalaemic. During the withdrawal phase, there was a 15% incidence of hyperkalaemia in the treatment group compared with 60% in the placebo group. In the placebo group, 50% of patients were taken off RAASi in the withdrawal phase, whereas this occurred in only 6% of the patients in the patiromer group. The reported side-effects in this trial were constipation (11%), diarrhoea (8%), hypomagnesemia (8%), and hypokalaemia (3%).

A subgroup analysis of the OPAL-HK trial considered the effects of patiromer in HF (ejection fraction <35%) patients. Of the 102 patients with HF (42% of the study population), 76% were normokalaemic after the first 4 weeks. In the withdrawal phase, hyperkalaemia occurred in 52% of patients taking placebo and 8% of those on patiromer.55

PEARL-HF studied patiromer in patients with chronic HF (see Table 3). Patients included in the study either had stage III CKD or a history of hyperkalaemia necessitating the discontinuation of RAASi.56 A total of 105 patients were initiated on 25 mg/day of spironolactone, and were randomised to treatment with patiromer or placebo for 4 weeks. At 4 weeks, 92.7% of the patients in the patiromer group achieved normokalaemia compared with 75.5% in the placebo group. The patiromer group was more likely to have had spironolactone increased to 50 mg/day (91% versus 74%). Patiromer resulted in a 6% incidence of hypokalaemia in the trial, whereas hypomagnesemia occurred in 24% of the patients.

Studies that Evaluated Patiromer in Hyperkalaemia

Article image

AMBER evaluated the effect of patiromer versus placebo on the rates of discontinuation of spironolactone in 295 patients with stage IIIb CKD and resistant hypertension over a 12-week period (see Table 3). All patients initially had a potassium level of <5.1 mmol/l, and were on either ACEi or ARB at baseline and were initiated on spironolactone. At the end of the 12 weeks, 86% of the patients in the patiromer arm remained on spironolactone compared with 66% of the placebo group. In the subgroup analysis of the patients with HF in the study (45% of the population), 84.1% of HF patients in the patiromer group remained on spironolactone compared with 68.1% in the placebo group. Hyperkalaemia was the most common cause of discontinuation, occurring in 23% of the placebo group versus 7% in the spironolactone group.57

Economic Impact

Hyperkalaemia poses a significant financial burden on the healthcare system. Mu et al. analysed a 5% random sample of Medicare beneficiaries in the US between 2010 and 2014 and randomised them based on the presence of hyperkalaemia (potassium level >5 mEq/l).58 Those with hyperkalaemia had more inpatient admissions, outpatient and emergency department visits and skilled nursing care facility admissions. Patients with hyperkalaemia and concomitant HF and/or CKD had even higher rates of hospital visits and longer inpatient stays. The subgroup of patients with HF and hyperkalaemia incurred a significant cost to the healthcare system at an average of US$11,750 over 30 days and US$49,244 over 1 year per patient compared with US$2,600 and US$23,936 in their normokalaemic counterparts.

Newer agents for hyperkalaemia remain expensive, but may overall save costs to the healthcare system. SZC 10 mg is US$749 for a 30-day supply, whereas patiromer 8.4 g is US$947 per month.59,60 In the British National Formulary, these prices are £427.20 and £172.50, respectively.61

Conclusion

Hyperkalaemia is a frequent issue encountered in patients with HF and is associated with increased mortality. RAASi interfere with potassium homeostasis, resulting in hyperkalaemia and lead to dose reduction or discontinuation of these agents, which have proven mortality benefits.

The novel potassium binding resins, SZC and patiromer, have shown positive short- and long-term results in counteracting hyperkalaemia and maintaining normokalaemia in various populations, including HF patients, and thus may aid in the optimisation of RAASi therapy and long-term maintenance.

References

  1. Roger VL. Epidemiology of heart failure. Circ Res 2013;113:646–59.
    Crossref | PubMed
  2. Jain N, Kotla S, Little BB, et al. Predictors of hyperkalemia and death in patients with cardiac and renal disease. Am J Cardiol 2012;109:1510–3.
    Crossref | PubMed
  3. Rossignol P, Lainscak M, Crespo-Leiro MG, et al. Unravelling the interplay between hyperkalaemia, renin-angiotensin-aldosterone inhibitor use and clinical outcomes. Data from 9222 chronic heart failure patients of the ESC-HFA-EORP Heart Failure Long-Term Registry. Eur J Heart Fail 2020;22:1378–89.
    Crossref | PubMed
  4. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail 2016;18:891–975.
    Crossref | PubMed
  5. Sterns RH, Grieff M, Bernstein PL. Treatment of hyperkalemia: something old, something new. Kidney Int 2016;89:546–54.
    Crossref | PubMed
  6. Komajda M, Anker SD, Cowie MR, et al. Physicians’ adherence to guideline-recommended medications in heart failure with reduced ejection fraction: data from the QUALIFY global survey. Eur J Heart Fail 2016;18:514–22.
    Crossref | PubMed
  7. Ouwerkerk W, Voors AA, Anker SD, et al. Determinants and clinical outcome of uptitration of ACE-inhibitors and beta-blockers in patients with heart failure: a prospective European study. Eur Heart J 2017;38:1883–90.
    Crossref | PubMed
  8. Núñez J, Bayés-Genís A, Zannad F, et al. Long-term potassium monitoring and dynamics in heart failure and risk of mortality. Circulation 2018;137:1320–30.
    Crossref | PubMed
  9. López Vilella R, Morillas Climent H, Plaza-López D, et al. Hyperkalemia in heart failure patients: current challenges and future prospects. Research Reports in Clinical Cardiology 2015;7:1–8.
    Crossref
  10. Sarwar CM, Papadimitriou L, Pitt B, et al. Hyperkalemia in heart failure. J Am Coll Cardiol 2016;68:1575–89.
    Crossref | PubMed
  11. Ferreira JP, Butler J, Rossignol P, et al. Abnormalities of potassium in heart failure: JACC state-of-the-art review. J Am Coll Cardiol 2020;75:2836–50.
    Crossref | PubMed
  12. Dunn JD, Benton WW, Orozco-Torrentera E, et al. The burden of hyperkalemia in patients with cardiovascular and renal disease. Am J Manag Care 2015;21(15 Suppl):307–15.
    PubMed
  13. Lehnhardt A, Kemper MJ. Pathogenesis, diagnosis and management of hyperkalemia. Pediatr Nephrol 2011;26:377–84.
    Crossref | PubMed
  14. Weir MR, Rolfe M. Potassium homeostasis and renin-angiotensin-aldosterone system inhibitors. Clin J Am Soc Nephrol 2010;5:531–48.
    Crossref | PubMed
  15. SOLVD Investigators. Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. N Engl J Med 1991;325:293–302.
    Crossref | PubMed
  16. Desai AS, Swedberg K, McMurray JJ, et al. Incidence and predictors of hyperkalemia in patients with heart failure: an analysis of the CHARM Program. J Am Coll Cardiol 2007;50:1959–66.
    Crossref | PubMed
  17. McMurray JJ, Packer M, Desai AS, et al. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med 2014;371:993–1004.
    Crossref | PubMed
  18. Ferreira JP, Mogensen UM, Jhund PS, et al. Serum potassium in the PARADIGM-HF trial. Eur J Heart Fail 2020;22:2056–64.
    Crossref | PubMed
  19. Zannad F, McMurray JJ, Krum H, et al. Eplerenone in patients with systolic heart failure and mild symptoms. N Engl J Med 2011;364:11–21.
    Crossref | PubMed
  20. Pitt B, Zannad F, Remme WJ, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med 1999;341:709–17.
    Crossref | PubMed
  21. Juurlink DN, Mamdani MM, Lee DS, et al. Rates of hyperkalemia after publication of the Randomized Aldactone Evaluation Study. N Engl J Med 2004;351:543–51.
    Crossref | PubMed
  22. Dev S, Lacy ME, Masoudi FA, et al. Temporal trends and hospital variation in mineralocorticoid receptor antagonist use in veterans discharged with heart failure. J Am Heart Assoc 2015;4:e002268.
    Crossref | PubMed
  23. Rossignol P, Duarte K, Girerd N, et al. Cardiovascular risk associated with serum potassium in the context of mineralocorticoid receptor antagonist use in patients with heart failure and left ventricular dysfunction. Eur J Heart Fail 2020;22:1402–11.
    Crossref | PubMed
  24. Collins AJ, Pitt B, Reaven N, et al. Association of serum potassium with all-cause mortality in patients with and without heart failure, chronic kidney disease, and/or diabetes. Am J Nephrol 2017;46:213–21.
    Crossref | PubMed
  25. Aldahl M, Jensen AC, Davidsen L, et al. Associations of serum potassium levels with mortality in chronic heart failure patients. Eur Heart J 2017;38:2890–6.
    Crossref | PubMed
  26. Beusekamp JC, Tromp J, Cleland JGF, et al. Hyperkalemia and treatment with RAAS inhibitors during acute heart failure hospitalizations and their association with mortality. JACC Heart Fail 2019;7:970–9.
    Crossref | PubMed
  27. Linde C, Bakhai A, Furuland H, et al. Real-world associations of renin-angiotensin-aldosterone system inhibitor dose, hyperkalemia, and adverse clinical outcomes in a cohort of patients with new-onset chronic kidney disease or heart failure in the United Kingdom. J Am Heart Assoc 2019;8:e012655.
    Crossref | PubMed
  28. Savarese G, Carrero JJ, Pitt B, et al. Factors associated with underuse of mineralocorticoid receptor antagonists in heart failure with reduced ejection fraction: an analysis of 11 215 patients from the Swedish Heart Failure Registry. Eur J Heart Fail 2018;20:1326–34.
    Crossref | PubMed
  29. Cooper LB, Benson L, Mentz RJ, et al. Association between potassium level and outcomes in heart failure with reduced ejection fraction: a cohort study from the Swedish Heart Failure Registry. Eur J Heart Fail 2020;22:1390–8.
    Crossref | PubMed
  30. Savarese G, Xu H, Trevisan M, et al. Incidence, predictors, and outcome associations of dyskalemia in heart failure with preserved, mid-range, and reduced ejection fraction. JACC Heart Fail 2019;7:65–76.
    Crossref | PubMed
  31. Dépret F, Peacock WF, Liu K,D et al. Management of hyperkalemia in the acutely ill patient. Ann Intensive Care 2019;9:32.
    Crossref | PubMed
  32. Semple P, Booth C. Calcium chloride; a reminder. Anaesthesia 1996;51:93.
    Crossref | PubMed
  33. Liu M, Rafique Z. Acute management of hyperkalemia. Curr Heart Fail Rep 2019;16:67–74.
    Crossref | PubMed
  34. Fordjour K, Walton, T, Doran JJ. Management of hyperkalemia in hospitalized patients. Am J Med Sci 2014;347:93–100.
    Crossref | PubMed
  35. Sterns RH, Rojas M, Bernstein P, et al. Ion-exchange resins for the treatment of hyperkalemia: are they safe and effective? J Am Soc Nephrol 2010;21:733–5.
    Crossref | PubMed
  36. Beccari MV, Meaney CJ. Clinical utility of patiromer, sodium zirconium cyclosilicate, and sodium polystyrene sulfonate for the treatment of hyperkalemia: an evidence-based review. Core Evid 2017;12:11–24.
    Crossref | PubMed
  37. Flinn RB, Merrill JP, Welzant WR. Treatment of the oliguric patient with a new sodium-exchange resin and sorbitol; a preliminary report. N Engl J Med 1961;264:111–5.
    Crossref | PubMed
  38. Scherr L, Ogden DA, Mead AW, et al. Management of hyperkalemia with a cation-exchange resin. N Engl J Med 1961;264:115–9.
    Crossref | PubMed
  39. Kessler C, Ng J, Valdez K, et al. The use of sodium polystyrene sulfonate in the inpatient management of hyperkalemia. J Hosp Med 2011;6:136–40.
    Crossref | PubMed
  40. Lepage L, Dufour AC, Doiron J, et al. Randomized clinical trial of sodium polystyrene sulfonate for the treatment of mild hyperkalemia in CKD. Clin J Am Soc Nephrol 2015;10:2136–42.
    Crossref | PubMed
  41. Scott TR, Graham SM, Schweitzer EJ, et al. Colonic necrosis following sodium polystyrene sulfonate (Kayexalate)-sorbitol enema in a renal transplant patient. Report of a case and review of the literature. Dis Colon Rectum 1993;36:607–9.
    Crossref | PubMed
  42. Yuan CM, Nee R, Little DJ, Abbott KC. Incidence of sodium polystyrene sulfonate-associated colonic necrosis. Am J Med 2013;126:e13.
    Crossref | PubMed
  43. Chaitman M, Dixit D, Bridgeman MB. Potassium-binding agents for the clinical management of hyperkalemia. P T 2016;41:43–50.
    PubMed
  44. Packham DK, Rasmussen HS, Lavin PT, et al. Sodium zirconium cyclosilicate in hyperkalemia. N Engl J Med 2015;372:222–31.
    Crossref | PubMed
  45. Stavros F, Yang A, Leon A, et al. Characterization of structure and function of ZS-9, a K+ selective ion trap. PLoS One 2014;9:e114686.
    Crossref | PubMed
  46. Peacock WF, Rafique Z, Vishnevskiy K, et al. Emergency potassium normalization treatment including sodium zirconium cyclosilicate: a phase ii, randomized, double-blind, placebo-controlled study (ENERGIZE). Acad Emerg Med 2020;27:475–86.
    Crossref | PubMed
  47. FDA. Lokelmatm (sodium zirconium cyclosilicate) for oral suspension. Prescribing Information. FDA. May 2018.
    https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/207078s000lbl.pdf (accessed 31 December 2020).
  48. Kosiborod M, Rasmussen HS, Lavin P, et al. Effect of sodium zirconium cyclosilicate on potassium lowering for 28 days among outpatients with hyperkalemia: the HARMONIZE randomized clinical trial. JAMA 2014;312:2223–33.
    Crossref | PubMed
  49. Zannad F, Hsu BG, Maeda Y, et al. Efficacy and safety of sodium zirconium cyclosilicate for hyperkalaemia: the randomized, placebo-controlled HARMONIZE-Global study. ESC Heart Fail 2020;7:54–64.
    Crossref | PubMed
  50. Anker SD, Kosiborod M, Zannad F, et al. Maintenance of serum potassium with sodium zirconium cyclosilicate (ZS-9) in heart failure patients: results from a phase 3 randomized, double-blind, placebo-controlled trial. Eur J Heart Fail 2015;17:1050–6.
    Crossref | PubMed
  51. Spinowitz BS, Fishbane S, Pergola PE, et al. Sodium zirconium cyclosilicate among individuals with hyperkalemia: a 12-month phase 3 study. Clin J Am Soc Nephrol 2019;14:798–809.
    Crossref | PubMed
  52. Li L, Harrison SD, Cope MJ, et al. Mechanism of action and pharmacology of patiromer, a nonabsorbed cross-linked polymer that lowers serum potassium concentration in patients with hyperkalemia. J Cardiovasc Pharmacol Ther 2016;21:456–65.
    Crossref | PubMed
  53. Bakris GL, Pitt B, Weir MR, et al. Effect of patiromer on serum potassium level in patients with hyperkalemia and diabetic kidney disease: the AMETHYST-DN randomized clinical trial. JAMA 2015;314:151–61.
    Crossref | PubMed
  54. FDA. Veltassa (patiromer) for oral suspension. FDA. October 2015. https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/205739s000lbl.pdf (accessed 31 December 2020).
  55. Pitt B, Bakris GL, Bushinsky DA, et al. Effect of patiromer on reducing serum potassium and preventing recurrent hyperkalaemia in patients with heart failure and chronic kidney disease on RAAS inhibitors. Eur J Heart Fail 2015;17:1057–65.
    Crossref | PubMed
  56. Pitt B, Anker SD, Bushinsky DA, et al. Evaluation of the efficacy and safety of RLY5016, a polymeric potassium binder, in a double-blind, placebo-controlled study in patients with chronic heart failure (the PEARL-HF) trial. Eur Heart J 2011;32:820–8.
    Crossref | PubMed
  57. Agarwal R, Rossignol P, Romero A, et al. Patiromer versus placebo to enable spironolactone use in patients with resistant hypertension and chronic kidney disease (AMBER): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet 2019;394:1540–50.
    Crossref | PubMed
  58. Mu F, Betts KA, Woolley JM, et al. Prevalence and economic burden of hyperkalemia in the United States Medicare population. Curr Med Res Opin 2020;36:1333–41.
    Crossref | PubMed
  59. Drugs.com. Lokelma Prices, Coupons and Patient Assistance Programs. https://www.drugs.com/price-guide/lokelma (accessed 11 March 2021).
  60. Drugs.com. Veltassa Prices, Coupons and Patient Assistance Programs. https://www.drugs.com/price-guide/veltassa (accessed 11 March 2021).
  61. British National Formulary. London: Royal Pharmaceutical Society; 2021.
  62. Sidhu K, Sanjanwala R, Zieroth S. Hyperkalemia in heart failure. Curr Opin Cardiol 2020;35:150–5.
    Crossref | PubMed