Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2732 2022-08-15 03:03:49 |
2 format correct + 2 word(s) 2734 2022-08-16 08:26:47 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Scicchitano, P.;  Iacoviello, M.;  Massari, F.;  Palo, M.D.;  Caldarola, P.;  Mannarini, A.;  Passantino, A.;  Ciccone, M.M.;  Magnesa, M. Potassium Binders for Optimizing Therapies in Heart Failure. Encyclopedia. Available online: https://encyclopedia.pub/entry/26135 (accessed on 27 July 2024).
Scicchitano P,  Iacoviello M,  Massari F,  Palo MD,  Caldarola P,  Mannarini A, et al. Potassium Binders for Optimizing Therapies in Heart Failure. Encyclopedia. Available at: https://encyclopedia.pub/entry/26135. Accessed July 27, 2024.
Scicchitano, Pietro, Massimo Iacoviello, Francesco Massari, Micaela De Palo, Pasquale Caldarola, Antonia Mannarini, Andrea Passantino, Marco Matteo Ciccone, Michele Magnesa. "Potassium Binders for Optimizing Therapies in Heart Failure" Encyclopedia, https://encyclopedia.pub/entry/26135 (accessed July 27, 2024).
Scicchitano, P.,  Iacoviello, M.,  Massari, F.,  Palo, M.D.,  Caldarola, P.,  Mannarini, A.,  Passantino, A.,  Ciccone, M.M., & Magnesa, M. (2022, August 15). Potassium Binders for Optimizing Therapies in Heart Failure. In Encyclopedia. https://encyclopedia.pub/entry/26135
Scicchitano, Pietro, et al. "Potassium Binders for Optimizing Therapies in Heart Failure." Encyclopedia. Web. 15 August, 2022.
Potassium Binders for Optimizing Therapies in Heart Failure
Edit

Heart failure (HF) is a worrisome cardiac pandemic with a negative prognostic impact on the overall survival of individuals. International guidelines recommend up-titration of standardized therapies in order to reduce symptoms, hospitalization rates, and cardiac death. Hyperkalemia (HK) has been identified in 3–18% of HF patients from randomized controlled trials and over 25% of HF patients in the “real world” setting. Pharmacological treatments and/or cardio-renal syndrome, as well as chronic kidney disease may be responsible for HK in HF patients. These conditions can prevent the upgrade of pharmacological treatments, thus, negatively impacting on the overall prognosis of patients. Potassium binders may be the best option in patients with HK in order to reduce serum concentrations of K+ and to promote correct upgrades of therapies.

patiromer sodium zirconium cyclosilicate heart failure

1. Introduction

Globally, heart failure (HF) affects approximately 23 million people worldwide including more than 15 million people in Europe [1]. Statistics have revealed a prevalence rate of 1–3% in the general adult population and an incidence equal to 1–20 cases per 1000 person-years, and mortality ranging from 2 to 3% at 30 days from hospital discharge and from 50 to 75% at five-year follow-up [2].
International guidelines [3] recommend the need to modulate the main determinants of HF progression, namely the renin–angiotensin–aldosterone system (RAAS), the autonomic system, and the natriuretic peptide system, by optimizing therapies to the maximum tolerated dose [3].
Several trials [4][5][6][7][8][9][10][11][12] have demonstrated the benefits related to RAAS inhibitors (RAASi) in counteracting the negative evolution of HF, as they have demonstrated substantial reductions in hospitalization for HF, cardiovascular mortality, and improvement in the New York Heart Association (NYHA) classification system. A network meta-analysis [13][14] substantially proved the need for an early combination of different classes of drugs that could inhibit, on parallel, the different actors of HF progression and improve the risk for all-cause mortality by at least 70%. Therefore, optimizing pharmacological treatments in HF by up-titrating doses of all HF drugs (i.e., angiotensin converting enzyme inhibitors (ACEis), angiotensin-II receptor blockers (ARBs), mineralcorticoid receptor antagonists (MRAs), angiotensin receptor-neprilysin inhibitors (ARNIs), beta-blockers, and sodium-glucose cotransporter-2 inhibitors (SGLT2is)) has the main goal to be achieved by clinicians [3].
Beyond the direct and indirect pharmacological effects of these drugs on the heart, kidneys are further, secondary targets to be affected. Compounds that can interact with RAAS or SGLT2 may alter kidney function. As kidneys are the main regulators for ionic balance, one could suppose the possible impact of such drugs on the homeostasis (secretion and excretion) of ions maintained by the kidneys. Potassium is mostly involved in such a deregulation and the kidneys are responsible for the excretion of 90% of potassium, thus, conditions that have an impact on the complex renal mechanisms involved in potassium homeostasis account for serum increases of this ion [15][16].

2. Hyperkalemia in HF: Definition, Prevalence, and Prognosis

Hyperkalemia is defined when serum potassium (K+) levels exceed 5 mEq/L [17]. It can be classified hyperkalemia (HK) into: “mild” when serum K+ levels range between 5 and 5.5 mEq/L, “moderate” when serum K+ levels range between 5.5 and 6 mEq/L, and “severe” when serum K+ levels are higher than 6 mEq/L [17].
The prevalence of HK in the setting of HF is well-established [18]. The evaluation of results from randomized controlled trials (RCTs) dealing with HF in all its forms revealed an overall prevalence in any type of HK ranging from 3 to 18% [18]. The identification of this higher number of cases of HK in RCTs accounted for discontinuing HF therapies in 0.6–3.5% of cases [18].
Current real-world percentages are not dissimilar to those from RCTs. Data from 2,270,635 U.S. patients (2010–2014) highlighted a 1.57% prevalence rate of HK in the overall population and a 6.35% prevalence rate in those with chronic kidney disease (CKD) and/or heart failure, while the prevalence rate of CKD and/or HF was 48.43% in patients with HK [19]. Similar results have been reported in the Medicare population, with a 2.6–2.7% prevalence rate of HK in the overall population and a 8.9–9.3% prevalence rate of HK among patients with CKD and/or HF [20]. The data from Europe are not dissimilar to those from the USA, for example, in Italy, the prevalence rate of HK has been calculated to range from 6 to 10% in patients with HF [18]. The data from a recent Danish population-based cohort study revealed the prevalence rate of HK, in patients with congestive HF but normal kidney function (defined as estimated glomerular filtration rate (eGFR) to be >60 mL/min/1.73 m2), to br about 35% (mild HK), 13% (moderate HK), and 8% (severe HK) in relation to HK degree [21]. Indeed, in patients with congestive HF and severe CKD (eGFR 15–29 mL/min/1.73 m2), the prevalence of HK ranged from about 70% (mild HK) to 25% (severe HK) [21]. According to the SwedeHF (Swedish Heart Failure) Registry, about a quarter of patients with any type of HF (with reduced ejection fraction (HFrEF), mildly reduced ejection fraction (HFmrEF), and preserved ejection fraction (HFpEF)) suffer HK, thus, negatively impacting on the overall survival rate of these patients [22].
Higher plasma K+ levels could effectively impact on the prognosis of patients, and those suffering from HF in particular. The relationship between serum K+ concentration and the prognosis of patients with HF is similar to a U wave: lower and higher values than normal range negatively impact on the health of patients [23]. Indeed, the higher the plasma K+ concentration, the higher the incidence of all-cause mortality, cardiovascular death, death due to HF, and sudden cardiac death even after adjusting for confounding factors [23]. Although the data from the SwedeHF Registry have revealed mortality rates higher than 20% [22][23] in patients with HK and HF, the return to normokalemia might definitely improve the cumulative probability of all-cause mortality [23].
Many conditions can be considered to be independent predictors of high potassium levels, i.e., advancing age, history of diabetes, and decrease in eGFR. Indeed, drugs, such as ACEi/ARB or spironolactone, might be responsible for a 51% and 47% increase in serum K+ levels > 5.5 mEq/L [24]. A subanalysis from the ESC-EORP-HFA Heart Failure Long-Term Registry revealed that HK could prevent persistence on treatment or avoidance of target of the recommended dose of RAASi in patients with HFrEF, independently from the baseline value of K+ [25].
The main consequences of such data are related to the net increase in 1-year mortality and re-admission for HF: HF patients who discontinued or did not start RAASi showed a cumulative incidence in mortality at 1-year follow-up higher than 41%, while the rate of re-admission for HF decompensation at 1-year could exceed 64% [26]. Lisi et al. [27] observed a 75% mortality rate at 65-month follow-up when MRA was discontinued in patients with HFrEF.

3. Treatment of Hyperkalemia: Advances in Therapies

3.1. Sodium Polystyrene Sulfonate

Sodium polystyrene sulfonate (SPS) is a cation-exchange resin; its chemical structure is formed by benzene, diethenyl-polymer, with ethenylbenzene-sulfonated sodium salt [28][29]. The Food and Drug Administration (FDA) approved this compound in 1958. It can be orally or rectal taken, and the compound mainly acts at the level of the colon where it exchanges sodium for potassium. Indeed, SPS is a non-specific cation-exchange: the sodium ions in the compound may be displaced by calcium (Ca2+) and/or magnesium (Mg2+), thus, accounting for the lack of specificity of the drug [28][29]. This also accounts for the variability in the drug’s action onset, while 33% is the exchange skill of SPS bolus [28][29].
The literature contains scant data on the effectiveness and safety of SPS employment in clinical practice.
Batterink et al. [30] evaluated the impact of SPS on patients with HK (K+ level between 5.0 and 5.9 mEq/L) as compared with a placebo. SPS reduced serum K+ levels by 0.14 mEq/L, but no significantly clinical effect was observed [30].When administered for 7 days in patients with CKD and mild hyperkalemia (5.0–5.9 mEq/L) at a dose of 30 g orally o.d., SPS was effectively able to significantly reduce serum K+ levels as compared with a placebo, with no significant increase in side effects [31]. Indeed, a dose of 60 g o.d. might be more effective than the 30/15 g o.d. or the use of a rectal dose of 30 g o.d. [32][33]. Specifically, it seems that the higher the serum K+ levels, the higher the absolute reduction in the plasma levels of the ions within 24 h after the administration, although conflicting results are in the literature about this datum [34]. Indeed, data from small groups of patients who suffered CKD, used RAASi, and showed at least one episode of HK, low dose (15 mg o.d.) SPS might effectively reduce serum K+ levels when administered for long-term follow-up [35][36].
Nevertheless, SPS has demonstrated adverse serious side effects [37]. Case reports have reported gastrointestinal injuries such as colitis and necrosis; however, a recent analysis in the literature by Holleck et al. [37] did not find significantly higher rates in intestinal necrosis, although the composite outcome of severe gastrointestinal adverse events was significantly increased. The FDA has provided a warning about the use of sorbitol in concomitance with SPS as it can increase the risk for gastrointestinal necrotic events [28][29].

3.2. Sodium Zirconium Cyclosilicate (SZC)

SZC is an inorganic cation exchanger which is composed of a uniform crystalline structure with micropores that are responsible for the entrapment of the cations [38]. Specifically, the chemical structure of SZC can preferentially address an exchange between hydrogen and sodium monovalent cations such as potassium or ammonium [38]. The engagement with Ca2+ and Mg2+ is less favourable: this might be related to the ionic dimension of the K+ and ammonium and the complex architecture of the crystalline structure [38]. Experimental studies have demonstrated that SZC interacts with K+ 25-fold more selectively than Ca2+ or Mg2+ (SPS has a 0.2–0.3-fold selectivity for both the di-cations) [38]. Laboratory data have revealed that SZC has a 9.3-fold higher affinity for K+ than SPS and a 125-fold more selectivity for K+ than SPS [38].
The compound is not orally absorbed and it is completely eliminated via feces, thus, there is no systemic dissemination for SZC. Its specific action is performed in the gastrointestinal tract as a whole; therefore, the drug reduces the absorption of K+ and eliminates it via feces [38].
Packham et al. [39] considered patients with HK, as defined by serum K+ levels between 5.0 and 6.5 mEq/L, to identify a correct dose for SZC in this setting.
Patients were randomized to receive 1.25 g, 2.5 g, 5 g, or 10 g of ZS-9 or a placebo t.i.d. during the initial 48 h (initial phase); then, those who reached serum K+ levels between 3.5 and 4.9 mEq/L were randomized 1:1 to the placebo or the corresponding SZC dose o.d. (maintenance phase). All of the dosages succeeded in reducing serum K+ levels to normal values within 48 h, but only 5 g and 10 g doses maintained K+ levels in the normal range. Interestingly, the 10 g dose was able to significantly reduce K+ levels after 1 h from administration [39]. In the HARMONIZE trial [40], patients with serum K+ levels > 5.1 mEq/L were randomized to 10 g t.i.d. within 48 h, then to 5, 10, and 15 g o.d. for the following 28 days: 10 g t.i.d. reduced serum K+ levels to a normal range in 84% of patients within 24 h and in 98% of patients within 48 h, while all the regimes maintained K+ levels in a normal range during the 28 days after. Similar results were obtained from the HARMONIZE Global trial [41]. Indeed, when extending the administration of SZC to 337 days, as in the HARMONIZE extension trial [42], 84.3% of patients maintained K+ levels < 5.1 mEq/L and 98% of patients maintained serum K+ levels < 5.5 mEq/L. Such data were independent from kidney function. Roger et al. [43] demonstrated that 82% and 90% of patients with baseline eGFR < 30 and ≥30 mL/min/1.73 m2 succeeded in maintaining normokalemia after 10 g three times daily for 24–72 h, followed by once daily SZC 5 g for ≤12 months. SZC was also able to return K+ levels to normokalemia even in end-stage renal disease (ESRD) patients who were on hemodalysis. More than 40% of patients were able to return to a normal range of K+ with a 5 g administration which could be increased to 15 g in the DIALIZE trial [44].
Indeed, there are little data are on the efficacy of SZC in patients with HF who cannot optimize their pharmacological regimen due to HK conditions. A subanalysis from the HARMONIZE trial involved patients with HF history, with or without RAASi therapy, who had to continue their pharmacological treatments [45]. The results pointed out that 83%, 89%, and 92% of patients with HF and in therapy with 5 g, 10 g, and 15 g SZC, respectively, reached normal K+ levels during the maintenance phase [45]. Nevertheless, the small sample size (n = 94), the lack of differentiation of HF subtype, as well as the reduced number of patients with optimized therapy (only 69% of patients were on RAASi) reduced the evaluation of the impact of SZC in patients with HF. Imamura et al. [46] retrospectively evaluated 24 patients with left ventricle ejection fraction (LVEF) < 50% who were treated with SZC for 3 months (5 g, 10 g, and 15 g o.d.). They observed reductions in plasma levels of K+, improvement in the pharmacological treatments with RAASi, and amelioration in LVEF after 3 months [46]. Interesting insights are expected from the results of the ongoing Lokelma for RAAS Maximisation in CKD & Heart Failure (LIFT) trial [47]. The LIFT trial will include patients with LVEF < 40%, NYHA class II–IV, with serum K+ levels of 5.0–5.5 mEq/L, CKD (as defined by eGFR < 60 mL/min/1.73 m2), with none or a submaximal dose of ACEi/ARB and/or MRA [47]. Patients will undergo treatments with SZC 10 g t.i.d. for 48 h, then 5 or 10 g o.d. in agreement with serum K+ at each visit; meanwhile, RAASi therapy will be upgraded to the maximum tolerated dose. This trial is in the recruiting phase [48]. Further studies are needed in order to better evaluate the impact of SZC in HF patients with HK and the impact on the clinical outcomes of these individuals.

3.3. Patiromer

Patiromer was approved by the FDA in 2015 [49] and by the European Medicines Agency (EMA) in 2017 [50]. The complex chemical structure of the compound is formed by a carboxylic acid compound substituted at the alpha position with fluorine, comprising a copolymer of 2-fluoroacrylic acid, divinylbenzene, and 1,7-octadiene [51]. Sorbitol is added to patiromer but, differently from SPS, its concentration is 5- to 10-fold lower than SPS [51]. It has been calculated that patiromer has 1.5- to 2.5-fold higher skill in binding K+ than other polymers [51]. Calcium represents the cation for the exchange with K+ and this exchange is mostly performed in the large intestine. There is no absorption of this compound, thus, it can not pass into the systemic fluids.
Patiromer has been included in clinical studies in order to evaluate its impact on HK and, to some extent, in the HF setting.
The OPAL HK study [52] evaluated the impact of patiromer (initial dose 4.2 g or 8.4 g twice a day for 4 weeks, then those with normokalemia were randomized to patiromer or placebo for 8 weeks) in patients with CKD, receiving RAASi, and with serum K+ levels between 5.1 and 6.5 mEq/L. Seventy-six percent of patients showed normal K+ levels after 4 weeks of treatment; during the maintenance phase, only 15% of patients showed HK (vs. 60% of the placebo group) [52]. The effects of patiromer on serum K+ levels have been shown to remain when the compound was continued to be administered for a long period of time. The AMETHYST-DN trial just demonstrated lower K+ levelswhen patiromer was administered for 52 weeks [53].
The need for implementing therapies in those with HF has been provisionally considered in the PEARL HF trial [54]. Although the trial was not set for different types of HF, the aim was to implement therapies in those with chronic HF and HK, who discontinued RAASi or had CKD (eGFR < 60 mL/min/1.72 m2). Patients also underwent implementation of MRA therapy; 91% of patients reached the goal target of spironolactone with K+ levels maintained in a normal range [54]. A subanalysis of the AMETHYST-DN trial pointed out that the use of patiromer was safe and well-tolerated in patients with mild HF and on RAASi therapy [55]. Indeed, the DIAMOND trial will shed light on the efficacy of patiromer in patients with HF and LVEF ≤ 40% who will be started or continued on MRA titrated to 50 mg/day and other RAASi therapy to ≥50% target dose [56]. Meanwhile, a recent pooled analysis on 653 patients (214 with and 439 without HF) on patiromer treatment, coming from RCT populations, has demonstrated a consistent decrease in serum K+ level during therapy, with mild-to-moderate adverse events in one third of patients [57]. A retrospective analysis did not observe a significant increase in hospitalization rate for heart failure between patiromer and SZC users [58]. Furthermore, retrospective studies [59][60] have demonstrated contrasting results in the overall performance of patiromer, although more structured trials should be designed in order to address gaps in evidences.

References

  1. Savarese, G.; Lund, L.H. Global Public Health Burden of Heart Failure. Card. Fail. Rev. 2017, 3, 7–11.
  2. Savarese, G.; Becher, P.M.; Lund, L.H.; Seferovic, P.; Rosano, G.M.C.; Coats, A. Global Burden of Heart Failure: A Comprehensive and Updated Review of Epidemiology. Cardiovasc. Res. 2022; Epub ahead of print.
  3. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: Developed by the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). With the special contribution of the Heart Failure Association (HFA) of the ESC. Eur. J. Heart Fail. 2022, 24, 4–131.
  4. Heart Outcomes Prevention Evaluation (HOPE) Study Investigators. Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: Results of the HOPE study and MICRO-HOPE substudy. Heart Outcomes Prevention Evaluation Study Investigators. Lancet 2000, 355, 253–259.
  5. Yusuf, S.; Pitt, B.; Davis, C.E.; Hood, W.B.; Cohn, J.N. 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.
  6. Desai, A.S.; Swedberg, K.; McMurray, J.J.; Granger, C.B.; Yusuf, S.; Young, J.B.; Dunlap, M.E.; Solomon, S.D.; Hainer, J.W.; Olofsson, B.; et al. CHARM Program Investigators. Incidence and predictors of hyperkalemia in patients with heart failure: An analysis of the CHARM Program. J. Am. Coll. Cardiol. 2007, 50, 1959–1966.
  7. Lewis, E.J.; Hunsicker, L.G.; Clarke, W.R.; Berl, T.; Pohl, M.A.; Lewis, J.B.; Ritz, E.; Atkins, R.C.; Rohde, R.; Raz, I.; et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N. Engl. J. Med. 2001, 345, 851–860.
  8. Brenner, B.M.; Cooper, M.E.; de Zeeuw, D.; Keane, W.F.; Mitch, W.E.; Parving, H.H.; Remuzzi, G.; Snapinn, S.M.; Zhang, Z.; Shahinfar, S.; et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N. Engl. J. Med. 2001, 345, 861–869.
  9. Pitt, B.; Zannad, F.; Remme, W.J.; Cody, R.; Castaigne, A.; Perez, A.; Palensky, J.; Wittes, J.; Randomized Aldactone Evaluation Study Investigators. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. N. Engl. J. Med. 1999, 341, 709–717.
  10. Zannad, F.; McMurray, J.J.; Krum, H.; van Veldhuisen, D.J.; Swedberg, K.; Shi, H.; Vincent, J.; Pocock, S.J.; Pitt, B.; EMPHASIS-HF Study Group. Eplerenone in patients with systolic heart failure and mild symptoms. N. Engl. J. Med. 2011, 364, 11–21.
  11. McMurray, J.J.; Packer, M.; Desai, A.S.; Gong, J.; Lefkowitz, M.P.; Rizkala, A.R.; Rouleau, J.L.; Shi, V.C.; Solomon, S.D.; Swedberg, K.; et al. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N. Engl. J. Med. 2014, 371, 993–1004.
  12. Solomon, S.D.; McMurray, J.J.V.; Anand, I.S.; Ge, J.; Lam, C.S.P.; Maggioni, A.P.; Martinez, F.; Packer, M.; Pfeffer, M.A.; Pieske, B.; et al. Angiotensin-Neprilysin Inhibition in Heart Failure with Preserved Ejection Fraction. N. Engl. J. Med. 2019, 381, 1609–1620.
  13. Burnett, H.; Earley, A.; Voors, A.A.; Senni, M.; McMurray, J.J.; Deschaseaux, C.; Cope, S. Thirty Years of Evidence on the Efficacy of Drug Treatments for Chronic Heart Failure With Reduced Ejection Fraction: A Network Meta-Analysis. Circ. Heart Fail. 2017, 10, e003529.
  14. Tromp, J.; Ouwerkerk, W.; van Veldhuisen, D.J.; Hillege, H.L.; Richards, A.M.; van der Meer, P.; Anand, I.S.; Lam, C.S.P.; Voors, A.A. A Systematic Review and Network Meta-Analysis of Pharmacological Treatment of Heart Failure with Reduced Ejection Fraction. JACC Heart Fail. 2022, 10, 73–84.
  15. Raebel, M.A. Hyperkalemia associated with use of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers. Cardiovasc. Ther. 2012, 30, e156–e166.
  16. Greenlee, M.; Wingo, C.S.; McDonough, A.A.; Youn, J.H.; Kone, B.C. Narrative review: Evolving concepts in potassium homeostasis and hypokalemia. Ann. Intern. Med. 2009, 150, 619–625.
  17. Rosano, G.M.C.; Tamargo, J.; Kjeldsen, K.P.; Lainscak, M.; Agewall, S.; Anker, S.D.; Ceconi, C.; Coats, A.J.S.; Drexel, H.; Filippatos, G.; et al. Expert consensus document on the management of hyperkalaemia in patients with cardiovascular disease treated with renin angiotensin aldosterone system inhibitors: Coordinated by the Working Group on Cardiovascular Pharmacotherapy of the European Society of Cardiology. Eur. Heart J. Cardiovasc. Pharmacother. 2018, 4, 180–188.
  18. Tromp, J.; van der Meer, P. Hyperkalaemia: Aetiology, epidemiology, and clinical significance. Eur. Heart J. Suppl. 2019, 21, A6–A11.
  19. Betts, K.A.; Woolley, J.M.; Mu, F.; McDonald, E.; Tang, W.; Wu, E.Q. The prevalence of hyperkalemia in the United States. Curr. Med. Res. Opin. 2018, 34, 971–978.
  20. Mu, F.; Betts, K.A.; Woolley, J.M.; Dua, A.; Wang, Y.; Zhong, J.; Wu, E.Q. Prevalence and economic burden of hyperkalemia in the United States Medicare population. Curr. Med. Res. Opin. 2020, 36, 1333–1341.
  21. Thomsen, R.W.; Nicolaisen, S.K.; Hasvold, P.; Garcia-Sanchez, R.; Pedersen, L.; Adelborg, K.; Egfjord, M.; Egstrup, K.; Sørensen, H.T. Elevated Potassium Levels in Patients With Congestive Heart Failure: Occurrence, Risk Factors, and Clinical Outcomes: A Danish Population-Based Cohort Study. J. Am. Heart Assoc. 2018, 7, e008912.
  22. Savarese, G.; Xu, H.; Trevisan, M.; Dahlström, U.; Rossignol, P.; Pitt, B.; Lund, L.H.; Carrero, J.J. 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.
  23. Núñez, J.; Bayés-Genís, A.; Zannad, F.; Rossignol, P.; Núñez, E.; Bodí, V.; Miñana, G.; Santas, E.; Chorro, F.J.; Mollar, A.; et al. Long-Term Potassium Monitoring and Dynamics in Heart Failure and Risk of Mortality. Circulation 2018, 137, 1320–1330.
  24. Hoss, S.; Elizur, Y.; Luria, D.; Keren, A.; Lotan, C.; Gotsman, I. Serum Potassium Levels and Outcome in Patients With Chronic Heart Failure. Am. J. Cardiol. 2016, 118, 1868–1874.
  25. Crespo-Leiro, M.G.; Barge-Caballero, E.; Segovia-Cubero, J.; González-Costello, J.; López-Fernández, S.; García-Pinilla, J.M.; Almenar-Bonet, L.; de Juan-Bagudá, J.; Roig-Minguell, E.; Bayés-Genís, A.; et al. Hyperkalemia in heart failure patients in Spain and its impact on guidelines and recommendations: ESC-EORP-HFA Heart Failure Long-Term Registry. Rev. Esp. Cardiol. (Engl. Ed.) 2020, 73, 313–323.
  26. Gilstrap, L.G.; Fonarow, G.C.; Desai, A.S.; Liang, L.; Matsouaka, R.; DeVore, A.D.; Smith, E.E.; Heidenreich, P.; Hernandez, A.F.; Yancy, C.W.; et al. Initiation, Continuation, or Withdrawal of Angiotensin-Converting Enzyme Inhibitors/Angiotensin Receptor Blockers and Outcomes in Patients Hospitalized With Heart Failure With Reduced Ejection Fraction. J. Am. Heart Assoc. 2017, 6, e004675.
  27. Lisi, F.; Parisi, G.; Gioia, M.I.; Amato, L.; Bellino, M.C.; Grande, D.; Massari, F.; Caldarola, P.; Ciccone, M.M.; Iacoviello, M. Mineralcorticoid Receptor Antagonist Withdrawal for Hyperkalemia and Mortality in Patients with Heart Failure. Cardiorenal Med. 2020, 10, 145–153.
  28. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/011287s023lbl.pdf (accessed on 31 May 2022).
  29. Available online: https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-fda-recommends-separating-dosing-potassium-lowering-drug-sodium (accessed on 31 May 2022).
  30. Batterink, J.; Lin, J.; Au-Yeung, S.H.; Cessford, T. Effectiveness of Sodium Polystyrene Sulfonate for Short-Term Treatment of Hyperkalemia. Can. J. Hosp. Pharm. 2015, 68, 296–303.
  31. Lepage, L.; Dufour, A.C.; Doiron, J.; Handfield, K.; Desforges, K.; Bell, R.; Vallée, M.; Savoie, M.; Perreault, S.; Laurin, L.P.; et al. Randomized Clinical Trial of Sodium Polystyrene Sulfonate for the Treatment of Mild Hyperkalemia in CKD. Clin. J. Am. SocNephrol. 2015, 10, 2136–2142.
  32. Mistry, M.; Shea, A.; Giguère, P.; Nguyen, M.L. Evaluation of Sodium Polystyrene Sulfonate Dosing Strategies in the Inpatient Management of Hyperkalemia. Ann. Pharmacother. 2016, 50, 455–462.
  33. Kessler, C.; Ng, J.; Valdez, K.; Xie, H.; Geiger, B. The use of sodium polystyrene sulfonate in the inpatient management of hyperkalemia. J. Hosp. Med. 2011, 6, 136–140.
  34. Sandal, S.; Karachiwala, H.; Noviasky, J.; Wang, D.; Elliott, W.C.; Lehmann, D.F. To bind or to let loose: Effectiveness of sodium polystyrene sulfonate in decreasing serum potassium. Int. J. Nephrol. 2012, 2012, 940320.
  35. Chernin, G.; Gal-Oz, A.; Ben-Assa, E.; Schwartz, I.F.; Weinstein, T.; Schwartz, D.; Silverberg, D.S. Secondary prevention of hyperkalemia with sodium polystyrene sulfonate in cardiac and kidney patients on renin-angiotensin-aldosterone system inhibition therapy. Clin. Cardiol. 2012, 35, 32–36.
  36. Georgianos, P.I.; Liampas, I.; Kyriakou, A.; Vaios, V.; Raptis, V.; Savvidis, N.; Sioulis, A.; Liakopoulos, V.; Balaskas, E.V.; Zebekakis, P.E. Evaluation of the tolerability and efficacy of sodium polystyrene sulfonate for long-term management of hyperkalemia in patients with chronic kidney disease. Int. Urol. Nephrol. 2017, 49, 2217–2221.
  37. Holleck, J.L.; Roberts, A.E.; Marhoffer, E.A.; Grimshaw, A.A.; Gunderson, C.G. Risk of Intestinal Necrosis With Sodium Polystyrene Sulfonate: A Systematic Review and Meta-analysis. J. Hosp. Med. 2021, 16, 489–494.
  38. Stavros, F.; Yang, A.; Leon, A.; Nuttall, M.; Rasmussen, H.S. Characterization of structure and function of ZS-9, a K+ selective ion trap. PLoS ONE 2014, 9, e114686.
  39. Packham, D.K.; Rasmussen, H.S.; Lavin, P.T.; El-Shahawy, M.A.; Roger, S.D.; Block, G.; Qunibi, W.; Pergola, P.; Singh, B. Sodium zirconium cyclosilicate in hyperkalemia. N. Engl. J. Med. 2015, 372, 222–231.
  40. Kosiborod, M.; Rasmussen, H.S.; Lavin, P.; Qunibi, W.Y.; Spinowitz, B.; Packham, D.; Roger, S.D.; Yang, A.; Lerma, E.; Singh, B. Effect of sodium zirconium cyclosilicate on potassium lowering for 28 days among outpatients with hyperkalemia: The HARMONIZE randomized clinical trial. JAMA 2014, 312, 2223–2233.
  41. Zannad, F.; Hsu, B.G.; Maeda, Y.; Shin, S.K.; Vishneva, E.M.; Rensfeldt, M.; Eklund, S.; Zhao, J. Efficacy and safety of sodium zirconium cyclosilicate for hyperkalaemia: The randomized, placebo-controlled HARMONIZE-Global study. ESC Heart Fail. 2020, 7, 54–64.
  42. Roger, S.D.; Spinowitz, B.S.; Lerma, E.V.; Singh, B.; Packham, D.K.; Al-Shurbaji, A.; Kosiborod, M. Efficacy and Safety of Sodium Zirconium Cyclosilicate for Treatment of Hyperkalemia: An 11-Month Open-Label Extension of HARMONIZE. Am. J. Nephrol. 2019, 50, 473–480.
  43. Roger, S.D.; Lavin, P.T.; Lerma, E.V.; McCullough, P.A.; Butler, J.; Spinowitz, B.S.; von Haehling, S.; Kosiborod, M.; Zhao, J.; Fishbane, S.; et al. Long-term safety and efficacy of sodium zirconium cyclosilicate for hyperkalaemia in patients with mild/moderate versus severe/end-stage chronic kidney disease: Comparative results from an open-label, Phase 3 study. Nephrol. Dial. Transplant. 2021, 36, 137–150.
  44. Fishbane, S.; Ford, M.; Fukagawa, M.; McCafferty, K.; Rastogi, A.; Spinowitz, B.; Staroselskiy, K.; Vishnevskiy, K.; Lisovskaja, V.; Al-Shurbaji, A.; et al. A Phase 3b, Randomized, Double-Blind, Placebo-Controlled Study of Sodium Zirconium Cyclosilicate for Reducing the Incidence of Predialysis Hyperkalemia. J. Am. Soc. Nephrol. 2019, 30, 1723–1733.
  45. Anker, S.D.; Kosiborod, M.; Zannad, F.; Piña, I.L.; McCullough, P.A.; Filippatos, G.; van der Meer, P.; Ponikowski, P.; Rasmussen, H.S.; Lavin, P.T.; 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, 1050–1056.
  46. Imamura, T.; Oshima, A.; Narang, N.; Kinugawa, K. Clinical Implications of Sodium Zirconium Cyclosilicate Therapy in Patients with Systolic Heart Failure and Hyperkalemia. J. Clin. Med. 2021, 10, 5523.
  47. Murphy, D.; Ster, I.C.; Kaski, J.C.; Anderson, L.; Banerjee, D. The LIFT trial: Study protocol for a double-blind, randomised, placebo-controlled trial of K+-binder Lokelma for maximisation of RAAS inhibition in CKD patients with heart failure. BMC Nephrol. 2021, 22, 254.
  48. Available online: https://clinicaltrials.gov/ct2/show/NCT05004363 (accessed on 2 June 2022).
  49. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/205739Orig1s000TOC.cfm (accessed on 2 June 2022).
  50. Available online: https://www.ema.europa.eu/en/documents/product-information/veltassa-epar-product-information_en.pdf (accessed on 2 June 2022).
  51. Li, L.; Harrison, S.D.; Cope, M.J.; Park, C.; Lee, L.; Salaymeh, F.; Madsen, D.; Benton, W.W.; Berman, L.; Buysse, J. 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, 456–465.
  52. Weir, M.R.; Bakris, G.L.; Bushinsky, D.A.; Mayo, M.R.; Garza, D.; Stasiv, Y.; Wittes, J.; Christ-Schmidt, H.; Berman, L.; Pitt, B.; et al. Patiromer in patients with kidney disease and hyperkalemia receiving RAAS inhibitors. N. Engl. J. Med. 2015, 372, 211–221.
  53. Bakris, G.L.; Pitt, B.; Weir, M.R.; Freeman, M.W.; Mayo, M.R.; Garza, D.; Stasiv, Y.; Zawadzki, R.; Berman, L.; Bushinsky, D.A.; 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, 151–161.
  54. Pitt, B.; Anker, S.D.; Bushinsky, D.A.; Kitzman, D.W.; Zannad, F.; Huang, I.Z.; PEARL-HF Investigators. 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, 820–828.
  55. Pitt, B.; Bakris, G.L.; Weir, M.R.; Freeman, M.W.; Lainscak, M.; Mayo, M.R.; Garza, D.; Zawadzki, R.; Berman, L.; Bushinsky, D.A. Long-term effects of patiromer for hyperkalaemia treatment in patients with mild heart failure and diabetic nephropathy on angiotensin-converting enzymes/angiotensin receptor blockers: Results from AMETHYST-DN. ESC Heart Fail. 2018, 5, 592–602.
  56. Butler, J.; Anker, S.D.; Siddiqi, T.J.; Coats, A.J.S.; Dorigotti, F.; Filippatos, G.; Friede, T.; Göhring, U.M.; Kosiborod, M.N.; Lund, L.H.; et al. Patiromer for the management of hyperkalaemia in patients receiving renin-angiotensin-aldosterone system inhibitors for heart failure: Design and rationale of the DIAMOND trial. Eur. J. Heart Fail. 2022, 24, 230–238.
  57. Piña, I.L.; Yuan, J.; Ackourey, G.; Ventura, H. Effect of patiromer on serum potassium in hyperkalemic patients with heart failure: Pooled analysis of 3 randomized trials. Prog. Cardiovasc. Dis. 2020, 63, 656–661.
  58. Zhuo, M.; Kim, S.C.; Patorno, E.; Paik, J.M. Risk of Hospitalization for Heart Failure in Patients With Hyperkalemia Treated With Sodium Zirconium Cyclosilicate vs. Patiromer. J. Card. Fail. 2022; Online ahead of print.
  59. Kovesdy, C.P.; Rowan, C.G.; Conrad, A.; Spiegel, D.M.; Fogli, J.; Oestreicher, N.; Connaire, J.J.; Winkelmayer, W.C. Real-World Evaluation of Patiromer for the Treatment of Hyperkalemia in Hemodialysis Patients. Kidney Int. Rep. 2018, 4, 301–309.
  60. Kovesdy, C.P.; Gosmanova, E.O.; Woods, S.D.; Fogli, J.J.; Rowan, C.G.; Hansen, J.L.; Sauer, B.C. Real-world management of hyperkalemia with patiromer among United States Veterans. Postgrad. Med. 2020, 132, 176–183.
More
Information
Subjects: Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , ,
View Times: 249
Revisions: 2 times (View History)
Update Date: 16 Aug 2022
1000/1000
Video Production Service