Heart Failure





Introduction


Drug therapies for heart failure (HF) differ depending on the phenotype of presentation. Distinctive phenotypes of presentation have different management targets, ranging from acute decompensated HF (ADHF) to chronic HF with reduced ejection fraction (HFrEF) or preserved ejection fraction (HFpEF). For the purposes of this discussion, we shall focus on drug therapy for specific phenotypic syndromes including ADHF, HFrEF, and HFpEF. Preventive care, self-management, cardiac rehabilitation, device-based therapies, and surgical management of HF are discussed elsewhere.


Acute Decompensated Heart Failure


Introduction


Acute decompensated HF is a life-threatening condition generally defined as a first occurrence ( de novo ) or, more frequently a presentation of acute worsening of preexisting chronic HF with prior reported admissions for decompensation. It affects primarily the elderly and often requires urgent evaluation and hospitalization ( Table 3.1 ).



Table 3.1

New York Heart Association (NYHA) functional classification and American College of Cardiology/American Heart Association (ACC/AHA) stages of heart failure























NYHA functional classification



  • Class I—Patients with heart disease without resulting limitation of physical activity. Ordinary physical activity does not cause heart failure symptoms such as fatigue or dyspnea.




  • Class II—Patients with heart disease resulting in slight limitation of physical activity. Symptoms of heart failure develop with ordinary activity but there are no symptoms at rest.




  • Class III—Patients with heart disease resulting in marked limitation of physical activity. Symptoms of heart failure develop with less than ordinary physical activity but there are no symptoms at rest.




  • Class IV—Patients with heart disease resulting in inability to carry on any physical activity without discomfort. Symptoms of heart failure may occur even at rest.

ACC/AHA stages of heart failure



  • Stage A—At high risk for heart failure but without structural heart disease or symptoms of heart failure.




  • Stage B—Structural heart disease but without signs or symptoms of heart failure. This stage includes patients in NYHA class I with no prior or current symptoms or signs of heart failure.




  • Stage C—Structural heart disease with prior or current symptoms of heart failure. This stage includes patients in any NYHA class with prior or current symptoms of heart failure.




  • Stage D—Refractory heart failure requiring specialized interventions. This stage includes patients in NYHA class IV with refractory heart failure.



Patients with de novo ADHF usually have a sudden presentation, with pulmonary edema or cardiogenic shock, while those with acute decompensation of chronic HF tend to show progressive signs and symptoms before acute presentation. These differences are explained by compensatory mechanisms, the capacity of the pulmonary lymphatic system and preexisting use of diuretic therapy. Also, although acute decompensation of chronic HF can happen without known precipitants, most patients have identifiable triggers such as infection, arrhythmia, anemia, pulmonary embolism, acute coronary syndrome (ACS), and nonadherence.


Although prehospital management is recommended, it should not delay transfer to an appropriate medical environment. The assessment includes discrimination of severity and adjudication of the relative contribution of either congestion or perfusion to the bedside clinical profile. In the absence of cardiogenic shock, immediate echocardiography is not mandatory, since initial treatment of ADHF is similar for patients with HFrEF or HFpEF, which centers around decongestion with diuretic therapy and maintenance of adequate peripheral perfusion using either vasodilators or in selected situations, inotropic support. There is some controversy as to whether chronic therapies for HF should be reduced or stopped during acute decompensations. Current guidelines suggest that patients should continue long-term HF therapies in the setting of ADHF, except in the presence of hemodynamic instability, hyperkalemia or severely impaired renal function. The clinical supposition in such cases is that the neurohormonal activation may be necessary during acute decompensation to maintain cardiorenal hemodynamics. In these cases, daily dosages of neurohormonal antagonists may be down-titrated or withheld temporarily until stabilization, with rapid reexposure once the acute state abates. The choice of drug therapy will need to take prognostic variables into account and parameters such as an elevated blood urea nitrogen (BUN) level ≥ 43 mg/dL, low systolic blood pressure (BP) < 115 mmHg, high serum creatinine level ≥ 2.75 mg/dL, and increased troponin I level signify elevated risk. Permanent signs of congestion and renal dysfunction have been shown to be among the most important prognostic variables. The routine use of a pulmonary artery catheter is not recommended and should be restricted to hemodynamically unstable patients with an unknown mechanism of deterioration and influences the therapy which in such cases tends to be targeted towards the hemodynamic aberration. A clinical approach that may act as a surrogate to invasive hemodynamic parameters has been widely used to classify patients into therapeutic categories ( Fig. 3.1 ). Patients with a “wet” profile have correlated higher pulmonary capillary wedge pressure (PCWP ≥ 18 mmHg) while those with a “cold” profile have lower cardiac indexes (CI ≤ 2.2 L/min/m 2 ). Fig. 3.2 summarizes the 2016 European Society of Cardiology (ESC) algorithm for management of patients admitted with ADHF based on clinical profiles.




Fig. 3.1


Clinical profiles based on the presence of symptoms and signs of congestion and peripheral hypoperfusion. CI , Cardiac index; PCWP , pulmonary capillary wedge pressure; S3 , third heart sound; SVR , systemic vascular resistance.

(Based on data from Nohria A, Tsang SW, Fang JC, et al. Clinical assessment identifies hemodynamic profiles that predict outcomes in patients admitted with heart failure. J Am Coll Cardiol 2003;41(10):1797–1804.)



Fig. 3.2


Algorithm for management of patients admitted with acute heart failure (AHF) based on clinical profiles.

(Data from 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 Heart J 2016.)


Pharmacotherapy


Diuretics


Congestion is the leading cause of hospitalization in patients with ADHF. Although robust evidence is lacking, clinical experience has demonstrated that diuretics effectively improve hemodynamics and congestive symptoms, even in the setting of de novo ADHF, in which pulmonary edema may occur without significant volume overload. During ADHF hospitalization, net changes in weight and fluid balance are commonly used to assess response to decongestive therapies.


Diuretics are classified according to their major site of action, chemical structure, or type of diuresis. Table 3.2 summarizes the doses and site of action of most diuretics commonly used in patients with acute and chronic HF. The optimal regimen is often influenced by renal function, preexisting diuretic therapy and urgency of decongestion. Loop diuretics, such as furosemide, torsemide, and bumetanide, have the most potent diuretic effect and remain the diuretic of choice for treating ADHF. These agents act by directly inhibiting the sodium–potassium–chloride cotransporter (NKCC) at the thick ascending limb of the loop of Henle. They also inhibit a second cotransporter isoform that is widely expressed throughout the body, including the ear, which probably explains the side effect of ototoxicity associated with their use.



Table 3.2

Doses of diuretics commonly used in patients with heart failure

Modified from Mullens W, Damman K, Harjola VP, Mebazaa A, Brunner-La Rocca HP, Martens P, et al. The use of diuretics in heart failure with congestion – a position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2019;21(2):137–155.

























































































Diuretic class Site Starting dose Maintenance dose Half-life
Carbonic anhydrase inhibitors *
Acetazolamide Proximal tubule 250–375 mg orally/IV once a day One dose every other day or once daily for 2 days alternating with a day of rest 2.4–5.4 hours
Loop diuretics a
Bumetanide b Ascending loop of Henle Oral: 0.5–2 mg once or twice
IV or IM: 0.5–1 mg once or twice
Oral doses may be titrated every 4 hours IV doses may be titrated every 2–3 hours Maximum recommended dose is 10 mg/day 1–1.5 hours
Furosemide b Oral: 20–80 mg once or twice
IV or IM: 20–40 mg once or twice
Maximum dose: 600 mg/day 30–120 min (normal renal function)
9 hr (end-stage renal disease)
Torsemide b Oral or IV: 10–20 mg once 200 mg 3.5 hours
Thiazide-like diuretics c
Chlorothiazide Distal convoluted tubule Oral or IV: 500–1000 mg once or twice 1000 mg once or twice 45–120 minutes
Chlorthalidone 12.5–50 mg once 100 mg 45–60 hours
Hydrochlorothiazide 12.5–50 mg once or twice 200 mg 6–15 hours
Metolazone 2.5–10 mg once 20 mg 6–20 hours
Potassium-sparing diuretics
Amiloride Collecting duct 5–10 mg once or twice 20 mg 6–9 hours
Mineralocorticoid-receptor antagonists c,d
Spironolactone Collecting duct 12.5–25 mg once 50–100 mg e 1–2 hours
Eplerenone 25–50 mg once 50–100 mg e 3–6 hours
Sequential nephron blockade
Metolazone 2.5–10.0 mg once + loop diuretic
Hydrochlorothiazide 25–100 mg once or twice + loop diuretic
Chlorothiazide (IV) 500–1000 mg once + loop diuretic

IV, Intravenous.

* Carbonic anhydrase inhibitors can be used to treat the edema of congestive heart failure but are not commonly used for this purpose.


a Dose of intravenous and oral loop diuretics are similar


b Equivalent doses: 40 mg furosemide = 1 mg bumetanide = 20 mg torsemide


c Do not use if estimated glomerular filtration rate is < 30 mL/min/1.73 m 2


d Minimal diuretic effect


e Doses up to 400 mg may be used in hepatology



Loop diuretics are organic anions that are heavily bound to proteins as they circulate, reaching the tubular lumen predominantly by active secretion rather than glomerular filtration and thus maintain efficacy unless renal function is severely impaired. While structurally similar as a class, these agents have substantial differences in pharmacokinetics. The oral bioavailability of furosemide ranges from 10% to 90% and is determined by absorption from the gastrointestinal tract into the bloodstream, which is decreased in patients with severe ADHF-associated bowel edema. In contrast, the oral bioavailability of torsemide and bumetanide remain high (> 90%), making oral and intravenous (IV) doses similar. In addition, torsemide has a longer half-life when compared to furosemide or bumetanide. In healthy individuals, an oral dose of a loop diuretic may be as effective as an IV dose because the diuretic bioavailability that is above the natriuretic threshold is approximately equal. However, the natriuretic threshold increases in patients with HF and the oral dose may not provide a high enough serum level to elicit a significant natriuresis. Therefore, IV therapy is recommended in patients with ADHF to achieve a higher and consistent bioavailability, allowing rapid onset of action within few minutes.


Diuretic dosing should be individualized and titrated according to hemodynamic response. Patients on chronic loop diuretic therapy may need higher doses in the setting of acute decompensation of chronic HF. The DOSE trial prospectively compared diuretic strategies in patients with ADHF and suggested that 2.5 times the usual home dose is associated with better symptom improvement than low dose, at the cost of some renal impairment. The DOSE trial also showed no difference in outcomes between continuous IV infusion and intermittent bolus strategies, suggesting that both approaches could be considered when managing significant volume overload or diuretic resistance. Clinical experience, however, has demonstrated that when the effect of high-dose loop diuretic is suboptimal, a continuous infusion may be superior to reduce toxicity and maintain stable serum drug levels. Given the steep dose-response curve of loop diuretics, prompt doubling of dose at 2-hour intervals might allow the attainment of a ceiling dose earlier. Increasing the dose above the ceiling can lead to additional natriuresis by extending the time during which serum drug levels exceeds the natriuretic threshold, which makes it appear as if a ceiling does not exist.


Impaired diuretic response is a common complication in patients with ADHF and is associated with increased rehospitalization and mortality. Although not fully understood, diuretic resistance is thought to result from a complex interplay between cardiac and renal dysfunction, specific renal adaptation, and escape mechanisms. In fact, the magnitude of natriuresis following a given dose of diuretics declines over time as a result of the braking phenomenon, an appropriate homeostatic response that prevents excessive volume depletion during continued diuretic therapy. However, in patients with secondary hyperaldosteronism, such as those with HF, this phenomenon can be pronounced, leading to an increased reabsorption of sodium and contributing to diuretic resistance. Furthermore, chronic treatment with a loop diuretic results in compensatory hypertrophy of the distal tubular cells, which bypasses the proximal effect of the loop diuretic.


The coexistence of HF and impaired renal function is another common cause of diuretic resistance, as decreased renal blood flow and impaired tubular secretion may lead to insufficient therapeutic urinary drug concentrations. Among these patients, it is important to distinguish between underlying kidney disease and cardiorenal syndrome, which is being increasingly recognized as a complication of ADHF. The pathways to these distinct conditions involve not only hemodynamic deterioration but also neurohormonal, inflammatory, and intrinsic renal mechanisms. It is hypothesized that the cardiorenal syndrome may represent a backward failure, with elevated right-sided filling pressures contributing more to renal dysfunction than low cardiac output (CO). Traditional therapy with diuretics can contribute to cardiorenal syndrome, probably by further neurohormonal activation and worsening intrarenal hemodynamics, while vasodilator or inotropic therapy has not been shown to help. In most cases, however, worsening of renal function in the setting of aggressive diuresis may reflect hemodynamic or functional changes in glomerular filtration rather than actual renal tubular injury. Because residual clinical congestion is a predictor of poor outcomes in HF, a small to moderate increase in the BUN/creatinine ratio should not prevent further diuretic therapy if clinical evidence of congestion is still present in patients with ADHF.


A useful approach for overcoming diuretic resistance is by sequential nephron blockade, which is the concurrent use of diuretics acting upon different segments of the nephron to produce an additive or synergistic diuretic response. There is some evidence that a stepped pharmacologic strategy with early assessment of urinary output and sequential nephron blockade might result in equal decongestion when compared to mechanical ultrafiltration (UF), without significant renal impairment. A post hoc analysis of the CARRESS-HF, DOSE, and ROSE-ADHF trials compared the efficacy of a urine-output guided diuretic adjustment versus standard high-dose loop diuretics therapy for the management of cardiorenal syndrome in patients with ADHF. Compared with standard therapy, the stepped pharmacologic algorithm dosed to maintain urine output between 3 and 5 L/day resulted in greater weight change and more net fluid loss after 24 hours with slight improvement in renal function. A practical stepped pharmacologic algorithm to diuretic assessment in ADHF is reflected in Fig. 3.3 .




Fig. 3.3


A practical stepped pharmacologic algorithm to diuretic assessment in acute heart failure.

Total loop diuretic dose can be administered either as continuous infusion or bolus infusion. SGLT-2i , Sodium-glucose cotransporter 2 inhibitor; UF , ultrafiltration; UO , urine output.

(Modified from Mullens W, Damman K, Harjola VP, et al. The use of diuretics in heart failure with congestion—a position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2019;21(2):137–155.)


Combined diuretic therapy using any of several thiazide-type diuretics can more than double daily natriuresis, although care must be taken for symptomatic hypotension, renal dysfunction, and electrolyte abnormalities. Both IV chlorothiazide and oral metolazone have been shown to provide significant increases in urine-output when added to furosemide monotherapy, with chlorothiazide inducing a larger diuretic effect but at higher costs and greater need for potassium replacement. There are no randomized controlled trials to establish whether a preferable thiazide exists in the treatment of ADHF.


Potassium-sparing diuretics, such as spironolactone, eplerenone, amiloride, and triamterene, should be considered to prevent hypokalemia associated with loop and thiazide diuretics use. Spironolactone and eplerenone are synthetic mineralocorticoid-receptor antagonists (MRAs) with a strong recommendation for patients with symptomatic chronic HFrEF, while amiloride and triamterene are epithelial sodium channel blockers that do not affect the mineralocorticoid receptor.


Carbonic anhydrase inhibitors, such as acetazolamide, potently inhibit sodium bicarbonate reabsorption in the proximal tubules, resulting in increased sodium levels in the loop of Henle where loop diuretics may exert their natriuretic action. These agents may be particularly useful during concomitant metabolic alkalosis, but, when used repeatedly, can lead to metabolic acidosis and hypokalemia. Other potential drugs for further decongestion, such as the arginine vasopressin (AVP) antagonists, have been investigated as adjuncts to standard HF therapies and will be addressed in other sections.


Other approaches that may also be helpful in patients with diuretic resistance are UF or hypertonic saline solution (HSS 3%) administration. The potential benefit of veno-venous UF is the removal of isotonic fluid without neurohormonal activation seen with diuretics. A trial comparing UF to diuretic therapy in patients with ADHF showed that UF was associated with greater weight loss by 48 hours but no difference in dyspnea score when compared to the control group. UF was associated with a reduction in rehospitalization and unscheduled office visits. By contrast, other trials have observed a higher frequency of adverse events in patients treated with UF, including worsening of renal function in the CARRESS-HF and venous access complications in the AVOID-HF, which was not completed by the sponsor. Based on these concerns, current HF guidelines do not recommend routine use of UF, which should be restricted to patients with congestion not responding to a stepped-care diuretic strategy. HSS 3% administration along with high-dose furosemide in selected hyponatremic patients with systemic congestion and renal dysfunction may be associated with greater clinical response and renal function preservation. However, routine use of HSS 3% is controversial and further studies are needed before use can be endorsed.


Vasodilators


Although there is no robust evidence to support their routine use, IV vasodilators are the second most commonly used agents to relieve congestive symptoms in patients with ADHF. Some vasodilators may act primarily on arterial resistance, leading to a decrease in afterload, while others act predominantly on venous capacitance with consequent reduction in preload. Most vasodilators, however, have balanced action on both afterload and preload.


Vasodilators are indicated in situations of pulmonary and systemic edema with or without hypoperfusion and are particularly helpful during hypertensive ADHF. However, they must be used with caution in patients who are preload or afterload dependent, such as those with severe diastolic dysfunction or aortic stenosis, since these agents can cause significant hypotension. Moreover, the use of vasodilators requires intensive BP monitoring and dose titration, and they should be avoided in patients with hypotension, hypovolemia, and recent use of phosphodiesterase (PDE)-5 inhibitors such as sildenafil, vardenafil, and tadalafil.


As a class, traditional direct-acting vasodilators, such as organic nitrates, sodium nitroprusside, and nesiritide ( Table 3.3 ), act as an exogenous source of nitric oxide (NO) to activate soluble guanylate cyclase (sGC), producing cyclic guanosine monophosphate (cGMP) and consequent vascular smooth muscle relaxation ( Fig. 3.4 ). Conversely, novel vasodilator agents targeting new pathways have been developed, including serelaxin, natriuretic peptides, neurohormonal antagonists, and sGC stimulators and sGC activators.



Table 3.3

Intravenous vasodilators used to treat acute heart failure

Modified from 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 Heart J 2016;18(8):891–975.








































Vasodilator Initial dose Dose range Main side effects Other
Nitroglycerin 10–20 μg/min 40–400 μg/min Hypotension, headache Tolerance on continuous use
Isosorbide dinitrate 1 mg/h 2–10 mg/h Hypotension, headache Tolerance on continuous use
Sodium nitroprusside 0.3 μg/kg/min 0.3–5 μg/kg/min Hypotension, isocyanate toxicity Light sensitive
Nesiritide Bolus 2 μg/kg 0.01–0.03 μg/kg/min Hypotension
Infusion 0.01 μg/kg/min



Fig. 3.4


Nitric oxide, nitroprusside, and nesiritide stimulate guanylate cyclase to form cyclic guanosine monophosphate (cGMP) with vasodilatory properties. GTP, guanosine-5′-triphosphate; LDL , low-density lipoprotein; LVH, left ventricular hypertrophy; SH , Sulfhydryl.

(Figure © L.H. Opie, 2012.)


Organic Nitrates


Organic nitrates, such as nitroglycerin, isosorbide dinitrate, and isosorbide mononitrate, reduce ventricular preload primarily via venodilation. At higher doses, especially in the presence of vasoconstriction, these agents can decrease systemic vascular resistance (SVR) and left ventricular (LV) afterload, consequently increasing stroke volume (SV) and CO. Nitrates are also partially selective for epicardial coronary arteries, thus justifying use in ADHF associated with ACS.


Nitrate indication is largely based upon hemodynamic response and expert opinion, as evidence is scarce and has relatively limited methodological quality. Intravenous administration is recommended for greater bioavailability and ease of titration. Low initial doses of nitroglycerin (NTG) are recommended, with rapid up-titration increments every 5 minutes as required and tolerated. Aggressive early up-titration of NTG is associated with a significant reduction in pulmonary capillary wedge pressure (PCWP) that reaches a maximum effect at 2 to 3 hours and declines subsequently within the following 24 hours due to tachyphylaxis. Tachyphylaxis is described as a rapid and significant attenuation of hemodynamic effects, limiting NTG usefulness in the treatment of patients with ADHF. The most common side effects of NTG are hypotension, headache, and nausea.


Sodium Nitroprusside


In contrast to NTG, sodium nitroprusside (SNP) causes balanced arterial and venous dilation, therefore reducing both the preload and the afterload. The use of SNP in patients with ADHF results in a significant decrease in systemic BP, right atrial pressure (RAP), pulmonary arterial pressure (PAP), PCWP, SVR, and pulmonary vascular resistance (PVR). SNP has a very short half-life, of seconds to a few minutes, and is particularly effective in the setting of ADHF secondary to elevated afterload such as acute aortic or mitral regurgitation, ventricular septal rupture, or hypertensive emergency.


Administration of SNP requires close hemodynamic monitoring due to its potent hemodynamic effects. Although severe hypotension is rare and resolves quickly, significant vasodilation of the intramyocardial vasculature may cause a “coronary steal” phenomenon. Therefore, SNP is not recommended in the setting of ACS. Also, sudden withdrawal may cause a “rebound hypertension” effect and gradual tapering is advised, ideally transitioning to oral vasodilators.


The major limitation to the use of SNP is its metabolism to cyanide. When doses > 400 μg/min are used for long periods of time, especially in patients with renal and/or hepatic dysfunction, the accumulation of SNP metabolites can lead to the development of cyanide, or rarely thiocyanate, toxicity, which may be fatal. The first sign of cyanide toxicity is lactic acidosis, and the most common side effects of thiocyanate toxicity are mental status changes, nausea, and abdominal pain. Thiocyanate can be removed with dialysis, and cyanide toxicity has been successfully managed with infusions of thiosulfate, sodium nitrate, and hydroxycobalamin. There are no randomized clinical trials of SNP in patients with ADHF and, as with NTG, their indication is based upon hemodynamic response and expert opinion.


Nesiritide


Nesiritide is a synthetic analogue form of the human B-type natriuretic peptide (BNP), manufactured from Escherichia coli with recombinant DNA technology. It causes balanced arterial and venous dilation, resulting in significant reductions in filling pressures and mild increases in CO.


In patients with ADHF, the administration of nesiritide has been investigated more extensively than both NTG and SNP. The VMAC trial tested the safety and efficacy of nesiritide in patients with ADHF and reported a significantly greater decrease in PCWP compared to both NTG and placebo, with no evidence of tachyphylaxis. Nesiritide, but not NTG, was also associated with significant improvements in patient-reported dyspnea at 3 hours. The ASCEND-HF trial tested nesiritide in a broad population of patients with ADHF and demonstrated a modest improvement in dyspnea when compared to placebo, but no difference in the composite outcome of death or HF hospitalization at 30 days. While there was no increase in renal failure with nesiritide, the incidence of symptomatic hypotension was higher with nesiritide. Those findings were consistent with the latter ROSE-ADHF trial, which assessed the effects of low-dose nesiritide in patients with ADHF and showed no benefits on urine output, congestion, renal function, or clinical outcomes, but more symptomatic hypotension.


Because of its high cost and lack of clear clinical benefit beyond other vasodilator therapies, such as NTG or SNP, nesiritide is not recommended as a first-line drug for patients with ADHF. Neither should it be used for the indication of replacing diuretic therapy, enhancing natriuresis, preventing cardiorenal syndrome, or improving survival. However, in selected patients who remain symptomatic despite standard therapy, a trial of nesiritide may be helpful.


Novel Vasodilators


Given the central role of vasodilator therapy in ADHF, there has been considerable enthusiasm for developing other types of vasodilator therapies. Serelaxin is a recombinant form of the human peptide relaxin-2, a hormone that regulates CV and renal adaptation to improve arterial compliance during pregnancy in humans. It acts primarily via receptor-based increase of intracellular NO, with additional activation of matrix metalloproteinases, upregulation of endothelin type-B receptors, and expression of vascular endothelial growth factor (VEGF). Low-dose serelaxin decreases RAP and PCWP, while high-dose increases CO. The RELAX-ADHF trial tested the efficacy of serelaxin by randomizing patients with ADHF to a 48-hour infusion of either serelaxin or placebo within 16 hours of hospital presentation. Serelaxin improved short-term dyspnea and was associated with reduced worsening signs or symptoms of HF, shorter hospital length of stay (LOS), and improved biomarkers of end-organ dysfunction including troponin, N-terminal pro-BNP (NT-proBNP), creatinine, and transaminases. Following a similar protocol, RELAX-ADHF-2 trial aimed to confirm the beneficial effects of serelaxin on CV mortality and worsening HF, but failed to meet either of its primary outcomes. These seemingly contradictory results suggest that serelaxin decongests the system rapidly, but with no improvement in CV outcomes.


Ularitide is a synthetic form of urodilatin, another natriuretic peptide, which is produced in the distal renal tubules with vasodilating, natriuretic, and diuretic effects. The TRUE-ADHF trial randomized patients with ADHF to continuous infusion of ularitide or placebo for 48 hours and found no difference in the risk of long-term CV mortality between the groups despite ularitide improving PCWP, SVR, CI, and dyspnea. Additionally, ularitide increased creatinine associated with a doubling of hypotension.


Neurohormonal antagonism started aggressively during ADHF hospitalization and on top of more established therapy has been tested in large-scale trials, with mostly disappointing results. The ASTRONAUT trial found that the addition of aliskiren, a direct renin inhibitor (DRI), to standard ADHF therapy did not reduce mortality or hospitalization outcomes and was associated with higher rates of hyperkalemia, renal dysfunction, and hypotension. Likewise, there was no benefit with tezosentan, a nonselective endothelin antagonist, or TRV027, an angiotensin type-1 (AT-1) receptor antagonist, over matching placebo in the VERITAS and BLAST-ADHF trials, respectively. In contrast, the PIONEER-HF trial showed that sacubitril–valsartan, an angiotensin receptor-neprilysin inhibitor (ARNI), reduced NT-proBNP to a greater degree than enalapril among eligible patients admitted with acute decompensated HF. Of note, side effects including hyperkalemia and hypotension were similar between the two drugs. Nevertheless, because reduction in biomarkers is a surrogate outcome, a larger trial powered for clinical outcomes is warranted.


The sGC stimulators and the sGC activators have a mechanism of action similar to that of organic nitrates, since both classes of drugs activate the sGC in smooth muscle cells, thus leading to the synthesis of cGMP and subsequent vasodilation. However, unlike traditional nitrates, sGC stimulators and sGC activators can induce sGC in its NO-insensitive state. Intravenous cinaciguat showed dose-dependent improvement in hemodynamic parameters but failed to demonstrate effectiveness in clinical trials for ADHF due to a high incidence of treatment induced hypotensive events requiring emergency intervention. Subsequently, oral vericiguat was evaluated in two dose-finding studies within the SOCRATES program. In the SOCRATES-REDUCED trial, vericiguat showed a significant reduction in NT-proBNP and a trend toward fewer hospitalizations in patients with HFrEF. In the SOCRATES-PRESERVED trial, it did not change NT-proBNP but was associated with better quality-of-life assessments in patients with HFpEF. Recently, the oral sGC stimulator vericiguat was tested in the VICTORIA trial, which randomized patients with NYHA class II-IV HF, LVEF ≤ 45%, and a recent decompensation event to receive vericiguat or placebo. Over a median follow-up of 10.8 months, the incidence of CV death or HF hospitalization was significantly lower among patients receiving vericiguat, but there was no difference in all-cause mortality. Vericiguat was safe and well tolerated and did not require monitoring of renal function or electrolytes.


Several other agents can reduce afterload and improve hemodynamic parameters, such as nitroxyl donors, short-acting calcium channel blockers, and potassium channel activators. However, robust randomized clinical trials for such agents are lacking.


Inotropes


Inotropic agents should be considered selectively in patients with advanced HF and low CO syndrome resulting in hypoperfusion, hypotension (relative and absolute), and end-organ dysfunction to prevent hemodynamic collapse and stabilize the clinical situation. They are also a reasonable pharmacologic bridge to cardiac transplantation in patients with end-stage HF.


The mechanism of action for most inotropes involves their ability to increase intracellular calcium, either by increasing influx into the cell or stimulating release from the sarcoplasmic reticulum. However, there is long-standing concern that even short-term use of IV inotropes might lead to hypotension, atrial or ventricular arrhythmias, and death. In particular, patients with ischemic heart disease (IHD) may be at higher risk of further myocardial ischemia due to reduced coronary perfusion and increased oxygen consumption. In any case, inotropic therapy should be started at very low doses, up-titrated under close monitoring, and discontinued as soon as appropriate organ perfusion is reestablished ( Table 3.4 ). Inotropes are not indicated in patients with HFPEF, and there is limited evidence to suggest that one particular agent is better than another.



Table 3.4

Positive inotropes and/or vasopressors used to treat acute heart failure

From Farmakis D, Agostoni P, Baholli L, et al. A pragmatic approach to the use of inotropes for the management of acute and advanced heart failure: An expert panel consensus. Int J Cardiol . 2019;297:83–90. https://doi-org.easyaccess2.lib.cuhk.edu.hk/10.1016/j.ijcard.2019.09.005 .








































































































Adrenergic receptors agonists Calcium sensitizer PDE III inhibitor
Agents Dopamine Dobutamine Norepinephrine Epinephrine Levosimendan Milrinone
Mechanism of action Dopa > β
HD, α
β1 > β2 > α α > β1 > β2 β1 = β2 > α Calcium sensitization
HD, PDE III inhibition
PDE III inhibition
Inotropic effect ↑↑ ↑↑ (↑) ↑↑
Arterial vasodilatation ↑↑ (renal, LD) 0 ↑↑ ↑↑↑
Vasoconstriction ↑↑ (HD) ↑ (HD) ↑↑ ↑ (HD) 0 0
Pulmonary vasodilatation ↑ or 0 ↓ or 0 (at high PVR) ↓ or 0 (at high PVR) ↑↑ ↑↑
Elimination t1/2 2 min 2.4 min 3 min 2 min 1.3h (active metabolite, 80h) 2.5 h
Infusion dose < 3 μg/kg/min: renal vasodilation
3–5 μg/kg/min: inotropic
> 5 μg/kg/min: vasoconstrictor
1–20 μg/kg/min 0.02–10 μg/kg/min 0.05–0.5 μg/kg/min 0.05–0.2 μg/kg/min 0.375–0.75 μg/kg/min
Bolus dose No No No 1 mg during CPR every 3–5 min 6–12 μg/kg over 10 min
(optional)
25–75 μg/kg over
10–20 min

CPR , Cardiopulmonary resuscitation; Dopa , dopaminergic receptors; HD , high dose; LD , low dose; PDE , phospodiesterase; PVR , pulmonary vascular resistances.


β-Adrenergic Agonists


Dobutamine


Dobutamine is the most widely used β-adrenergic agent for inotropic support. It is a synthetic analog of dopamine, with strong β 1 agonist activity as well as minor effects on β 2 -and α 1 -receptors. It has been suggested that the inotropic activity of dobutamine results from combined β 1 and α 1 stimulation in the myocardium. β 1 -stimulation enhances cardiac contractility through increases in intracellular cyclic adenosine monophosphate (cAMP) and calcium. At low doses, β2-stimulation generally offsets α1-adrenergic activity, resulting in peripheral artery vasodilation that occasionally may lead to symptomatic hypotension. At higher doses, though, peripheral vasoconstriction predominates through vascular α 1 -receptor stimulation. Since it has no dopaminergic effects, dobutamine is less prone to induce hypertension than is dopamine.


Dobutamine provides hemodynamic support with a dose-dependent increase in SV and CO and modest decreases in SVR and PCWP, referred to as an inodilatory effect. Lower doses might improve perfusion in patients with cardiogenic shock, but higher doses are generally recommended for more profound hypoperfusion states.


Dobutamine may induce serious atrial and ventricular arrhythmias at any infusion dose, particularly in the context of myocarditis and electrolyte imbalance. It also increases heart rate (HR) and myocardial oxygen demand and should be used cautiously in patients with recent myocardial ischemia. Hypersensitivity to dobutamine is a rare and unrecognized cause of eosinophilic myocarditis after prolonged infusion. Also, β-receptors may be downgraded or therapeutically blocked in patients with advanced HF so that intolerance to dobutamine may ensue. In the absence of cardiogenic shock, either levosimendan or milrinone could be alternatives for treating ADHF when β-blockade is thought to be contributing to hypoperfusion.


Clinical trials and HF registries have documented excess mortality in patients receiving intermittent or continuous dobutamine infusion, despite its beneficial hemodynamic effects. Thus, dobutamine should be restricted to HF patients with pulmonary congestion and low CO syndrome. In this setting, dobutamine appears to be as effective as milrinone.


Dopamine


Dopamine is a catecholamine-like agent, with complex effects that vary greatly with dose. At low doses, dopamine acts primarily on dopamine-1 receptors to dilate renal, splanchnic, and cerebral arteries. Although it has been proposed that dopamine might improve renal blood flow promoting natriuresis through direct distal tubular effects, data supporting such a potential benefit are limited. The DAD-HF study suggested that a combination of low-dose furosemide and low-dose dopamine as a continuous infusion for 8 hours was equally effective as high-dose furosemide but associated with improved potassium homeostasis and preservation of renal function. The follow-up DAD-HF II trial, however, found no differences in mortality rates, HF hospitalization, or overall dyspnea relief with the combined therapy. These findings are consistent with the ROSE-ADHF trial, which found that neither low-dose dopamine nor low-dose nesiritide enhanced decongestion or improved renal function in patients with ADHF. Notably, both DAD-HF II and ROSE-ADHF studies showed a higher incidence of tachycardia in the dopamine group.


At intermediate doses, dopamine acts as a precursor in the synthesis of norepinephrine (NE), an agonist of both adrenergic and dopaminergic receptors, and an inhibitor of NE reuptake, increasing SV and CO with variable effects on HR. Both the β 1 -stimulation and the rapid release of NE can precipitate tachycardia as well as atrial and ventricular arrhythmias. Tachyphylaxis to the inotropic effects of dopamine may develop, in part, because myocardial NE stores often become depleted in patients with advanced HF.


At higher doses, dopamine also stimulates α-receptors leading to pulmonary and peripheral artery vasoconstriction. In the treatment of shock, dopamine compares similarly to NE as the first-line vasopressor agent with respect to 28-day mortality but is associated with an increased risk of arrhythmias. Also, vasoconstriction dosages carry significant risk of precipitating limb and end-organ ischemia and should be used with caution. Discontinuation from high infusion rates should be done gradually to no less than 3 μg/kg/min, to minimize potential hypotensive response of low-dose dopamine.


Epinephrine


Epinephrine is the first-line vasopressor for cardiac arrest and anaphylactic shock. It acts as a complete β-receptor agonist, with dose-dependent α-agonism effect at higher doses. At lower doses, epinephrine acts predominantly on β 1 -receptors, with less prominent effects on β 2 and α 1 , resulting in an overall increase in CO with balanced vasodilator and vasoconstrictor effects. At higher doses, it increases SVR and BP, with combined inotropic and vasopressor effect. Common side effects, particularly at high doses, include tachycardia, arrhythmias, poor peripheral perfusion, headaches, anxiety, cerebral hemorrhage, and pulmonary edema. There is also a risk of local tissue necrosis with extravasation.


Isoproterenol


This relatively pure β-stimulant should be considered in cardiogenic shock secondary to bradycardia or when excessive β-blockade is thought to be contributing to hypoperfusion. It increases inotropy and chronotropy through β 1 -stimulation, with a variable response on BP, depending upon the degree of concomitant β 2 -vasodilator stimulation. The cardiac effects of isoproterenol may lead to palpitations, sinus tachycardia, and more serious arrhythmias. Other common side effects are hypotension, angina pectoris, flushing, headache, restlessness, and sweating. Patients with IHD may be at higher risk of further myocardial ischemia due to increased oxygen consumption. Also, there are some concerns regarding cost effectiveness of isoproterenol when compared to significantly cheaper alternative chronotropic agents.


Phosphodiesterase Inhibitors


Intravenous PDE-3 inhibitors, such as milrinone and enoximone (also available for oral use), decrease the rate of cAMP degradation, leading to enhanced inotropy, chronotropy, and lusitropy in cardiomyocytes. They also cause significant peripheral and pulmonary vasodilation via inhibition of vascular PDE, reducing preload and afterload while increasing contractility ( Fig. 3.5 ).




Fig. 3.5


Inotropic dilators (“inodilators”).

Increase of cyclic adenosine monophosphate in vascular smooth muscle (top) and in myocardium (bottom). α 1 , α 1 -Adrenergic stimulation; A-II , Angiotensin-II; cAMP, cyclic adenosine monophosphate; PDE , phosphodiesterase; SR , Sarcoplasmic reticulum; VP , vasopressin.

(Figure © L.H. Opie, 2012.)


Since PDE-3 inhibitors do not act via β-receptor stimulation, their effects are not offset by concomitant β-blocker therapy as are those of dobutamine or dopamine. Additionally, this independence of adrenergic pathways also allows for synergistic effects with the β-agonist inotropes. Nevertheless, while the hemodynamic effects of PDE-3 inhibitors can be helpful for short-term support, the increased levels of myocardial cAMP predispose to life-threatening arrhythmias, and the routine use of these agents for periods longer than 48 hours is not recommended. Milrinone is widely available for clinical use. Intravenous infusions may be started with a slow bolus over 10–20 minutes, which is often omitted due to hypotensive effects. The dose requires adjustments in the presence of renal dysfunction.


Major side effects include hypotension and atrial and ventricular arrhythmias. The OPTIME-CHF trial compared the effects of short-term IV milrinone versus placebo in patients with acute decompensation of chronic HF not requiring inotropic support. It showed that routine use of IV milrinone for ADHF was not associated with a reduction in hospital-based resource utilization at 60 days but increased the risk of sustained hypotension and atrial arrhythmias. In addition, a later subanalysis revealed increased mortality and rehospitalization in patients with IHD receiving milrinone when compared to placebo. Retrospective analysis of the ADHERE registry also found an increased in-hospital mortality associated with dobutamine or milrinone when compared to NTG or nesiritide. However, given that sicker patients were treated with inotropes, it is difficult to know if the adjustments were sufficient.


Calcium Sensitizers


Calcium sensitizers increase the sensitivity of troponin C fibers to ionic calcium that is already available in the cytoplasm, improving myocardial contractility with no additional calcium overload and minimal increase in oxygen demand. The most widely studied calcium sensitizer is levosimendan, while pimobendan is primarily used as a veterinary medication. Neither levosimendan nor pimobendan, however, are pure calcium sensitizers, as both share some PDE-3 inhibitor activity that may be partially responsible for their inotropic and vasodilator properties. Levosimendan exerts additional pleiotropic effects through the opening of potassium-dependent adenosine triphosphate (ATP) channels in vascular smooth muscle cells and mitochondria.


Although not approved for any use in the United States, levosimendan is currently available worldwide, including some countries in Europe and South America. It is indicated as short-term therapy for patients with ADHF who need inotropic support in the absence of severe hypotension. Intravenous infusions may be given with a bolus over 10 minutes, which is usually omitted due to the risk of significant hypotension. Due to an active long-acting metabolite, the hemodynamic effects of levosimendan can last for up to at least a week after stopping the infusion.


Despite a dose-dependent improvement in indices of cardiac performance, including a reduction in PCWP and afterload and an increase in CO, there is limited evidence of clinical benefit from levosimendan therapy. Initial clinical studies, such as LIDO and RUSSLAN, suggested a survival advantage from levosimendan when compared to dobutamine or placebo, respectively. The sequential more definitive trials REVIVE I and REVIVE II demonstrated a significant improvement in symptoms, BNP level, and hospital LOS with levosimendan therapy. However, there were more episodes of hypotension and cardiac arrhythmias in the levosimendan group, as well as a nonsignificant increase in early mortality when compared to placebo. The SURVIVE trial demonstrated no survival difference between levosimendan and dobutamine during long-term follow-up of patients with ADHF despite evidence for an early reduction of plasma BNP level with levosimendan. Levosimendan therapy was also associated with more episodes of atrial fibrillation (AFib) and hypokalemia.


Novel Inotropes


Omecamtiv Mecarbil


Omecamtiv mecarbil is a direct cardiac myosin activator that enhances effective actin-myosin cross-bridge formation and creates a force-producing state that is not associated with cytosolic calcium accumulation. Thus, omecamtiv mecarbil is believed to act as a calcium-sensitizer with pure inotropy action and no pleiotropic effects. Earlier studies have shown that it is safe, well tolerated, and produces dose-dependent increases in systolic ejection time, SV, EF, and fractional shortening. In the ATOMIC-AHF trial, IV omecamtiv mecarbil did not meet the primary outcome of dyspnea improvement in patients with ADHF compared to placebo, except in the higher-dose group. It did, however, increase systolic ejection time and decrease LV end-systolic diameter.


Istaroxime


Istaroxime is an investigational drug that mediates lusitropism through inhibition of sodium-potassium ATPase and stimulation of the sarcoendoplasmic reticulum calcium ATPase type 2a (SERCA2a). In the phase 2 HORIZON-HF trial, the addition of istaroxime to standard therapy lowered PCWP and HR and increased systolic BP in patients with ADHF. Also, higher doses of istaroxime appeared to be associated with more improvement in diastolic function. The role of this agent remains uncertain at this time.


Vasopressors


Vasopressor therapy should be reserved for patients with persistent hypotension, especially in the management of cardiogenic shock when hypoperfusion is evident despite optimization of filling pressures. Vasopressors increase vasoconstriction, which leads to increased SVR and mean arterial pressure (MAP), improving end-organ perfusion at the cost of peripheral perfusion and increased afterload. Among patients without preexisting cardiac dysfunction under vasopressor therapy, CO is either maintained or actually increased. However, the impact on CO in HF patients will depend on the balance between contractility improvement and afterload increase. Therefore, it is generally recommended to begin vasopressor therapy at very low doses, often in combination with inotropes.


The most often used vasopressors are NE, high-dose dopamine, high-dose epinephrine, vasopressin, and phenylephrine ( Table 3.4 ). NE has a high affinity for α 1 -receptors and moderate affinity for β-adrenergic receptors ( Fig. 3.6 ), resulting in marked vasoconstriction with mild to modest increase in HR, CO, and myocardial oxygen demand. In general, NE is the vasopressor of choice for generalized shock. The SOAP II trial evaluated first-line vasopressor selection in patients with generalized shock and showed that, among the prespecified cardiogenic shock subgroup, dopamine was associated with a higher risk of death and arrhythmia when compared to NE. Although the SOAP II trial was the largest study in patients with generalized shock, a scientific statement from the American Heart Association (AHA) raised questions about the external validity and applicability of the findings in the subgroup with cardiogenic shock. Therefore, NE might be the vasopressor of choice in many patients with shock, especially those presenting with arrhythmias, but whether it the optimal first-line vasoactive medication to treat cardiogenic shock remains unclear. Side effects include tachycardia, reflex bradycardia, anxiety, pulmonary edema, headache, and hypertension. As with all catecholamines and vasodilators, there is risk of local tissue necrosis with extravasation.




Fig. 3.6


Neuromodulation of vascular tone.

Upper panel, terminal neuron; lower panel, vascular smooth muscle (VSM) . Adrenergic sympathetic depolarization (top left) leads to release of norepinephrine (NE) from the storage granules of the terminal neurons into the synaptic cleft that separates the terminals from the arterial wall to act on postsynaptic vasoconstrictive β 1 -receptors. NE also stimulates presynaptic β 2 -receptors to invoke feedback inhibition of its own release, to modulate excess release of NE. Vagal cholinergic stimulation releases nitric oxide (NO) , which acts on muscarinic receptors (subtype two, M2 ) to inhibit the release of NE, thereby indirectly causing vasodilation. Circulating epinephrine (EPI) stimulates vascular vasodilatory β 2 -receptors but also presynaptic receptors on the nerve terminal that promote release of NE. Angiotensin-II (A-II) formed in response to renin released from the kidneys in shock-like states is also powerfully vasoconstrictive, acting both by inhibition of NE release (presynaptic receptors, schematically shown to the left of the terminal neuron) and also directly on arteriolar receptors.

(Figure © L.H. Opie, 2012.)


Phenylephrine is a selective α 1 -receptor agonist with potent arterial vasoconstrictor effect and minimal cardiac inotropy or chronotropy. It is particularly useful in patients with severe hypotension related to systemic vasodilation, such as septic shock, rather than to a decrease in CO. Thus, it should be reserved for patients in whom NE is contraindicated due to arrhythmias or who have failed other vasopressors.


As noted above, both dopamine and epinephrine may also be used for their vasoconstrictor properties. Dopamine is a precursor of NE and epinephrine, which acts in a dose-dependent fashion on dopaminergic receptors as well as α- and β-receptors. Epinephrine has essentially α 1 – and β- activity, increasing SVR, HR, CO, and BP ( Fig. 3.6 ).


Angiotensin II is a naturally occurring hormone secreted as part of the renin-angiotensin system that results in systemic vasoconstriction. In the ATHOS-3 trial, IV angiotensin II at a rate of 20 ng/kg/min adjusted to increase the mean arterial BP to at least 75 mmHg effectively increased BP and reduced vasopressor needs in patients with refractory vasodilatory shock. Although angiotensin II has been better studied in vasodilatory shock, it may be beneficial in both cardiogenic shock and cardiac arrest as well.


Vasopressin is an endogenous vasopressor stored mainly in the posterior lobe of the pituitary gland and myocardium. In a retrospective study of 36 patients who developed cardiogenic shock after myocardial infarction (MI), IV vasopressin therapy increased mean arterial BP without adversely affecting PCWP, CI, urine output, or other inotropic requirements. In a prospective randomized clinical trial of 48 patients, the combined infusion of vasopressin and NE proved to be superior to infusion of NE alone in the treatment of cardiocirculatory failure in catecholamine-resistant vasodilatory shock. A meta-analysis of randomized clinical trials compared vasopressin or terlipressin, which is a vasopressin analog not available in the United States, with supportive care in vasodilatory shock and showed no difference in short-term mortality. Currently, vasopressin is used as a second-line agent in refractory vasodilatory shock, particularly septic shock or anaphylaxis that is unresponsive to epinephrine.


Special Situations


Acute Coronary Syndromes


The acute onset of severe myocardial ischemia, with or without infarction, can result in decreased CO and/or elevated filling pressures. The coexistence of ADHF within ACS identifies a high-risk cohort, for whom aggressive therapy aiming at reperfusion should be rapidly instituted. Primary angioplasty is the strategy of choice for patients with ST-elevation MI. Inopressor therapy can offer pharmacological support to improve hemodynamics during cardiogenic shock but should be administered at the lowest possible doses to avoid further ischemia. Inodilator agents are not recommended in the absence of shock.


Hypertensive Emergency


Hypertensive ADHF accounts for approximately 10% of all ADHF cases, being most common in patients with HFpEF. ADHF precipitates in the setting of markedly elevated afterload, and typically manifests as acute pulmonary edema. Aggressive BP reduction with IV vasodilators and diuretics is recommended. Calcium channel blockers (CCBs) without significant myocardial depressant effects, such as nicardipine and clevidipine, may be helpful in patients presenting with hypertensive ADHF. Intravenous clevidipine has a rapid onset of action, high clearance, and no effect on ventricular contractility or central venous pressure (CVP), and is currently approved for the acute management of severe hypertension. Intravenous esmolol, a β-blocker, may be also used, but it is contraindicated in severe bradycardia heart block, cardiogenic shock, and overt HF.


Right Ventricular Heart Failure


Isolated right-sided or right ventricular (RV) HF is generally caused by acute RV infarction, severe pulmonary hypertension, or acute pulmonary embolism. Acute RV infarction should be approached as any other ACS, including early reperfusion and primary angioplasty when indicated. However, patients with RV infarction are very preload-dependent and can develop severe hypotension in response to nitrates or other preload-reducing agents. Hemodynamic stabilization with careful volume loading to a goal CVP of 10–12 mmHg can be beneficial, especially in patients with MAP < 60 mmHg. After ensuring adequate filling pressures, inotropic support can further augment RV CO. However, these principals do not apply to chronic right-sided HF.


The RV is also sensitive to increases in afterload, which is affected by the PVR. Selective pulmonary artery vasodilation includes treatment with NO, prostacyclin analogues, endothelin receptor antagonists, or, less frequently, certain CCBs. Hemodynamically unstable patients due to acute pulmonary embolism should be treated with immediate reperfusion either with thrombolysis, catheter-based approach, or surgical embolectomy.


Cardiogenic Shock


Cardiogenic shock is defined as a state of sustained hypotension (systolic BP < 90 mmHg for ≥ 30 min) and a reduced CI (< 2.2 L/min/m 2 ) in the presence of normal or elevated PCWP (> 15 mmHg) and inadequate tissue perfusion (e.g., elevation in lactate levels). Acute MI with LV dysfunction is the most frequent cause of cardiogenic shock. In patients with a recent ACS, mechanical complications such as papillary muscle rupture, ventricular septal defect, and free wall rupture may present as cardiogenic shock within 24 hours of hospitalization. Also, around 2%–6% patients may develop postcardiotomy cardiogenic shock following cardiac surgery. Therefore, immediate echocardiography is mandatory in all individuals presenting with cardiogenic shock. Other, less common etiologies include advanced valvular heart disease, arrhythmias, cardiac tamponade, cardiac constriction, pulmonary embolism, peripartum cardiomyopathy, acute coronary dissection, acute myocarditis, and drug poisoning.


Fluid challenge for up to 30 minutes, either with saline or ringer lactate, is recommended as the first-line treatment if there is no sign of overt fluid overload. Pharmacological therapy with inotropic and vasopressor agents aims to improve organ perfusion by increasing CO and BP. Despite their frequent use, few clinical outcome data are available to guide the initial selection of such therapies in patients with cardiogenic shock. Dobutamine is the most used inotrope, while levosimendan may be an alternative to patients already on oral β-blockade. Norepinephrine is recommended as the first-line vasopressor agent in the treatment of shock, as the use of dopamine is associated with a greater number of adverse events, such as arrhythmia. Short-term MCS, including extracorporeal membrane oxygenation (ECMO), may be considered as a bridge to heart transplantation or to other mechanical intervention in refractory cases. Currently, the intra-aortic balloon pump (IABP) is still the most widely used MCS device in cardiogenic shock. However, among patients with MI and cardiogenic shock, the use of an IABP did not improve mortality or any long-term secondary outcome in the IABP-SHOCK II trial. Therefore, routine use of an IABP cannot be recommended.


Heart Failure Patients With Reduced Ejection Fraction


Introduction


There has been substantial progress in the pharmacological management of chronic HF over the past three decades. The positive results of successive landmark clinical trials are reflected in clinical practice guidelines, which, in turn, have become standard of HF care. In general, the goals of treatment are to improve symptoms, functional capacity, and general quality of life, prevent disease progression and recurrent admissions, and prolong survival. To accomplish these goals, HFrEF should be viewed as a continuum that comprises four interrelated stages with incremental therapy at each stage aimed at modifying risk factors (A), treating structural heart disease (B-D), and reducing morbidity and mortality.


Currently, the recommendation for patients with stage A HF is largely preventive and should focus on risk factor modification and treatment of atherosclerotic vascular disease. There is robust evidence showing that the onset of HF may be delayed or prevented through interventions aimed at modifying risk factors or treating asymptomatic LV systolic dysfunction. The SPRINT trial showed that intensive BP control in subjects with a high CV risk was associated with a 38% relative risk reduction in the development of overt HF. In patients with stable coronary artery disease (CAD) without LV dysfunction, meta-analyses of randomized clinical trials have shown a modest benefit of angiotensin converting–enzyme (ACE) inhibitors, with a reduction in major CV outcomes, including HF. The antidiabetic sodium-glucose cotransporter 2 (SGLT-2) inhibitors reduced the composite risk of CV death or HF hospitalization in patients with type 2 diabetes mellitus (T2DM), regardless of established CV disease or HF. There is also reasonable evidence that intensive statin therapy, but not aspirin or other antiplatelet agents, may prevent or delay the onset of HF after ACS, with the most gain in patients with elevated levels of BNP.


Several mechanisms contribute to the progression of HF, such as neurohormonal activation, endothelial dysfunction, venous congestion, and myocardial remodeling. Once patients have established structural heart disease, the choice of pharmacotherapy will depend on their NYHA functional class. For patients with structural heart disease but without signs or symptoms of HF (stage B), the therapeutic goal should be the prevention of further cardiac remodeling. In asymptomatic patients with chronically reduced LV ejection fraction (LVEF), an ACE inhibitor should be used to prevent the risk of HF hospitalization. In all patients with a recent or remote history of MI or ACS, β-blockers and ACE inhibitors should be used, regardless of LVEF. It is noteworthy that β-blocker therapy should only be initiated after acute MI has resolved to avoid the risk of cardiogenic shock.


Symptomatic patients (stages C and D) have a worse prognosis, particularly after hospital admission for ADHF. The major goals of treatment in patients with structural heart disease and previous or current symptoms (stage C) are to alleviate symptoms and reduce morbidity by reversing or slowing the cardiac and peripheral dysfunction. Neurohormonal antagonists, such as ACE inhibitors, angiotensin receptor blockers (ARBs), β-blockers, MRAs, and ARNIs, have been shown to improve outcomes in HFrEF and are recommended, unless contraindicated or not tolerated. More recently, the SGLT-2 inhibitor dapagliflozin was also associated with improved clinical outcomes among patients with symptomatic HFrEF, irrespective of T2DM status, signaling a new approach in the treatment of HF.


Additional pharmacological agents, such as hydralazine plus nitrate, ivabradine, and digoxin, should be considered in selected patients. Patients with refractory end-stage HF (stage D) may be eligible for specialized, advanced treatments, such as LV assist devices (LVADs) and heart transplantation.


Neurohormonal Activation


HFrEF classically develops after an index event that could be an acute injury to the heart, a long-standing hemodynamic overload, or a response to genetic variations that disrupt contractile function or lead to myocyte death. The circulatory changes that arise from impaired cardiac pumping and/or filling trigger a series of compensatory mechanisms referred to as neurohormonal activation. This historical term reflects early observations that many of the molecules that are elaborated in response to HFrEF are produced by the neuroendocrine system. However, most neurohormones such as NE and angiotensin II are now known to be synthesized directly within the myocardium and, therefore, act in an autocrine or paracrine manner.


The sympathetic nervous system (SNS) and the renin–angiotensin–aldosterone system (RAAS) are the major neurohormonal systems, working collectively to maintain CO through increased retention of salt and water, peripheral vasoconstriction, and increased contractility. The activation of inflammatory mediators also contributes to maintain early CV homeostasis through cardiac repair and remodeling. However, sustained and chronic neurohormonal activation has deleterious effects and leads to progressive ventricular remodeling and worsening HF. Further activation of the natriuretic peptide (NP) system briefly offsets these harmful effects but is unable to sustain compensation for chronic neurohormonal activation over time.


Neuroendocrine modulation with β-blockers targeting the SNS and ACE inhibitors, ARBs, and MRAs targeting the RAAS have become the cornerstone of medical therapy to reduce morbidity and mortality in patients with chronic HFrEF. Most recently, the ARNIs further improved clinical outcomes by targeting both the RAAS and the NP system.


Pharmacotherapy


β -Blockers


SNS activation is one of the earliest responses to a decrease in CO, resulting in both increased release and decreased reuptake of catecholamines at adrenergic nerve endings. It plays a complex role in HF, with both beneficial and adverse effects. In the short term, catecholamine-mediated chronotropic and inotropic responses help to maintain CO while increased SVR and venous tone preserve BP and preload. NE and angiotensin II also mediate constriction of the efferent arterioles, allowing for stable glomerular filtration rate (GFR) despite a low renal perfusion, and stimulate sodium retention and volume expansion, improving SV via the Frank–Starling law.


Over time, however, these responses become detrimental, resulting in disruptions of β-adrenergic signaling and impaired mobilization of intracellular calcium. As a consequence, increased SNS activation has been implicated in the development and progression of HF through multiple mechanisms, involving cardiac, renal, and vascular function. In the heart, it may lead to downregulation and functional desensitization of β-adrenergic receptors, cardiomyocyte hypertrophy, necrosis, apoptosis, and fibrosis. In the kidneys, SNS activation stimulates salt and water retention, attenuates response to natriuretic factors, and activates the RAAS, which in turn promotes a positive feedback loop that adversely affects hemodynamic and cardiac remodeling. In the peripheral vessels, it also mediates neurogenic vasoconstriction and vascular hypertrophy.


The association between SNS activation, reflected by an increase in the plasma NE concentration, and mortality in patients with HFrEF raised the possibility of therapeutic efficacy for sympathetic inhibition. The potential mechanisms by which β-blockers improve outcomes in HFrEF are likely related to reducing the detrimental effects of sustained SNS activation by competitively antagonizing β-adrenergic receptors. Particular benefits might include decreased myocyte death from catecholamine-induced necrosis, antiarrhythmic and HR-lowering effects, upregulation of β 1 -receptors, and inhibition of renin secretion. In addition, β-blockers directly reduce myocardial oxygen consumption and BP, revert production of fetal protein isoforms, and prevent proapoptotic and cardiotoxic effects of cAMP-mediated calcium overload.


β-Blockers also strongly modulate cardiac remodeling, improve symptoms, reduce hospitalizations, and prolong survival in patients with HFrEF. Recommendations for their use are mainly based on the outcomes of large randomized placebo-controlled trials. The three β-blockers that have been shown to reduce the risk of death in patients with chronic HFrEF are carvedilol, bisoprolol, and extended-release metoprolol succinate (CR/XL). Nebivolol was also associated with a reduction in the composite outcome of mortality or CV hospitalizations, but it did not reduce mortality alone and is not approved by the US Food a Drug Administration (FDA) for the treatment of HFrEF.


The MDC was the first randomized clinical trial testing a β-blocker, the short-acting metoprolol tartrate, in subjects with idiopathic dilated cardiomyopathy (DCM) and NYHA class II-III. At 12 months, the metoprolol group had significant improvement in quality-of-life assessments, LVEF, and exercise capacity. There was also a nonsignificant 34% relative risk reduction in the combined outcome of death or need for cardiac transplantation, which was driven entirely by the reduction in transplantation, since there was no difference in all-cause mortality. The following MERIT-HF trial also investigated the impact of metoprolol on mortality in subjects with symptomatic HFrEF. However, it was larger, included patients with IHD as well as idiopathic DCM, and used the metoprolol succinate, which has a better pharmacological profile than metoprolol tartrate because of its extended-release formulation (CR/XL) and longer half-life. The MERIT-HF trial was stopped early at a mean follow-up of 12 months after an interim analysis demonstrated a 34% relative risk reduction in all-cause mortality in subjects with NYHA class II–IV HF and LVEF ≤ 40% with metoprolol CR/XL compared to placebo. Additional secondary outcomes showed a reduction in all-cause hospitalization and CV events. Of note, metoprolol CR/XL reduced mortality from both sudden cardiac death and progressive pump failure while nearly 95% of the study population was already taking a concomitant ACE inhibitor or ARB at baseline.


The first study performed with bisoprolol in HFrEF was the CIBIS trial, which showed a nonsignificant trend toward 20% lower mortality and 30% fewer HF hospitalizations at a mean follow-up of 1.9 years in the bisoprolol group. The subsequent follow-up CIBIS-II trial had greater statistical power to detect a mortality benefit and included approximately four times as many subjects as did CIBIS. The CIBIS-II was stopped early, at about 16 months, after the second interim analysis demonstrated a 34% relative risk reduction in all-cause mortality in patients with NYHA class III–IV HF and LVEF ≤ 35% with bisoprolol compared to placebo. At the time of CIBIS-II publishing, standard HFrEF therapy included diuretics, ACE inhibitors, nitrates, and digoxin. Other benefits of bisoprolol therapy, such as a reduction in the risk of sudden cardiac death, HF hospitalizations, and all-cause hospitalizations, were observed regardless of the HF etiology.


Of the three FDA-approved β-blockers for the treatment of HFrEF, carvedilol has been the most studied. The US Carvedilol Heart Failure Program was designed as a stratified clinical program consisting of four component trials to evaluate nonfatal outcomes in subjects with mild to severe HF and LVEF ≤ 35%. Although mortality was not a predefined primary outcome for the combined trials, it was prospectively measured by a safety committee, which prematurely stopped the program at a mean follow-up of 6.5 months due to a highly significant 65% relative risk reduction in mortality associated with the use of carvedilol across all four trials. The following ANZ-Carvedilol trial was conducted primarily to determine the effect of carvedilol therapy on LVEF and exercise tolerance in subjects with HF due to ischemic heart disease. The study concluded that carvedilol therapy for 12 months reduced LV volumes and preserved exercise performance at a lower rate-pressure product. At 19 months, there was also a significant 26% relative risk reduction in the clinical composite of death or hospitalization with carvedilol compared to placebo. However, the ANZ-Carvedilol trial was not powered to determine statistically significant effects on mortality, and therefore, the 24% relative risk reduction in mortality alone did not achieve statistical significance.


Taken together, the outcomes of the US Carvedilol HF Program and the ANZ-Carvedilol trial provided a strong rationale for other large, randomized mortality studies, such as the COPERNICUS and the CAPRICORN trials. The COPERNICUS trial was specifically designed to evaluate the impact of carvedilol in a population with more advanced HF than was studied in previous trials, including subjects with NYHA class III–IV and severely reduced LVEF < 25%. After a mean follow-up of 10.4 months, carvedilol was associated with a 34% relative risk reduction in annual mortality rates, as well as reductions in rehospitalizations, hospital length of stay, and cardiogenic shock when compared to placebo. In addition, subjects in the carvedilol group felt better and were less likely to develop any serious adverse event. The CAPRICORN trial tested the effects of carvedilol in patients with acute MI and LV dysfunction with LVEF ≤ 40% and showed no difference in the prespecified primary composite outcome of mortality and CV hospitalization when compared to placebo. However, all-cause mortality was reduced by 23% in the carvedilol group, a magnitude nearly identical to the results of the BHAT trial, which tested propranolol in patients with acute MI and was published two decades before. Importantly, almost all subjects in CAPRICORN were taking an ACE inhibitor, while approximately one-half had received thrombolysis and/or primary angioplasty.


Differences in β-blockers clinically used in HFrEF are associated to β 1 -receptor selectivity, presence of α 1 -receptor antagonism, antioxidant properties, and vasodilating effects. Both metoprolol and bisoprolol are second-generation agents that block β 1 -receptors to a much greater extent than β 2 -receptors, without antioxidant properties or vasodilating effects. Metoprolol is 75-fold selective, whereas bisoprolol has approximately 120-fold higher affinity for human β 1 -versus β 2 -receptors. This is important because their effects are mainly restricted to areas containing β 1 receptors, especially the heart and part of the kidney. Also, side effects linked to β 2 -blockade, such as bronchospasm, peripheral vasoconstriction, and abnormalities of glucose and lipid metabolism, are less common with β 1 -selective agents, although receptor selectivity weakens at higher doses. Conversely, carvedilol is a third-generation nonselective β-blocker that competitively blocks β 1 -, β 2 -, and α-receptors with some antioxidant properties. Approximately 80% of adrenergic receptors in the normal myocardium are β 1 -receptors. However, due to β 1 downregulation in HF, β 2 may rise up to 40% of the total adrenergic receptors. Interestingly, β 2 also mediates the effects of catecholamines in the heart, and as a result, it becomes a relatively important mediator of inotropic support and arrhythmogenic effects of sympathetic stimulation in patients with HF. The human heart also expresses α 1 -receptors, although at much lower levels than β-receptors. The importance of cardiac α 1 -receptors in the pathophysiology of HF is still controversial, but their role in the vasoconstriction of major arteries, such as the aorta, pulmonary arteries, mesenteric vessels, and coronary arteries, is well known. Consequently, carvedilol blockade of α 1 -receptors causes vasodilation of blood vessels and might help to improve afterload while posing a higher risk for hypotension.


The hypothesis that multiple adrenergic blockade is more effective than selective β 1 -adrenergic blockade was tested in the COMET trial, which remains the largest and most well-designed head-to-head trial of β-blockers in HF. The COMET trial randomized subjects with NYHA class II–IV HF and LVEF ≤ 35% to carvedilol or short-acting metoprolol tartrate in addition to other standard therapies and showed that carvedilol was more effective in reducing all-cause mortality. The generalizability of these findings is limited given that the degree of β-blockade may not have been equivalent in the two arms of COMET. The mean dose of metoprolol achieved in COMET was lower than the ones given in the MDC and MERIT-HF trials, which resulted in significantly lesser HR reductions when compared to carvedilol. Another potential concern is that metoprolol tartrate differs from the extended-release metoprolol succinate (CR/XL) used in MERIT-HF, the main trial showing a survival benefit of metoprolol compared with placebo in HFrEF patients. Metoprolol tartrate produces a less sustained β 1 -blockade because of its shorter half-life and has not been shown to reduce mortality in the MDC trial. There have been no trials directly comparing outcomes on carvedilol and extended-release metoprolol succinate (CR/XL).


The COMET trial supports the hypothesis that meaningful differences exist in the clinical effects of different β-blockers, suggesting that their beneficial effects in HFrEF may not necessarily be a class effect. For instance, some β-blockers with intrinsic sympathomimetic activity, such as xamoterol, have been shown to increase, rather than reduce, mortality in patients with HFrEF. Other β-blockers, such as bucindolol and nebivolol, had a neutral effect on overall mortality rates in the BEST and SENIORS trials. Bucindolol is a third-generation nonselective β-blocker, with additional weak α-blocking properties and some intrinsic sympathomimetic activity. Although it failed to reduce total mortality in the BEST trial, a prespecified subanalysis suggested a survival benefit restricted to white participants that might be secondary to a polymorphism in β 1 -receptors in that population. Nebivolol is a selective β 1 -receptor antagonist with some vasodilatory effects that are partially mediated by NO production via activation of β 3 -adrenergic receptors. It is approximately 3.5 times more β 1 -selective than bisoprolol and, along with carvedilol, is one of the few β-blockers to cause vasodilation. The SENIORS trial tested the effects of nebivolol on mortality and morbidity in patients ≥ 70 years old with stable HF. At a mean follow-up of 21 months, nebivolol therapy resulted in a significant 14% relative risk reduction in the primary composite outcome of all-cause mortality or CV hospitalizations compared to placebo but did not reduce all-cause mortality. In the United States, nebivolol is only approved for the treatment of hypertension, while in Europe it is registered for use in hypertension and HF as well.


Overall, the results of the COMET, BEST, and SENIORS trials highlight the importance of using doses and formulations of β-blockers with proven benefit in clinical trials. In accordance with clinical practice guidelines recommendations, β-blockers should be up-titrated to reach maximally tolerated doses whenever possible. Evidence-based doses for recommended β-blockers are listed in Table 3.5 . However, the abrupt withdrawal of adrenergic support may lead to exacerbation of congestive HF and significant negative chronotropy. Thus, therapy with β-blockers is recommended for clinically stable patients and should be initiated at very low doses and up-titrated gradually, with dose doubling no earlier than every 2 weeks as tolerated. Nevertheless, patients may experience some fluid retention within the first 3–5 days of treatment and should be advised to weigh themselves on a daily basis. Fluid retention can be managed by increasing the diuretic dose until weight returns to baseline and should not prevent future up-titration of the β-blocker.



Table 3.5

Drugs for the prevention and treatment of chronic heart failure with reduced ejection fraction

Modified from 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 Heart J 2016;18(8):891–975.


































































































Drug Starting dose Target dose
β-Blockers
Bisoprolol 1.25 mg once 10 mg once
Carvedilol 3.125 mg twice 25 mg twice a
Metoprolol succinate 12.5–25 mg once 200 mg once
Angiotensin-converting enzyme inhibitors
Captopril 6.25 mg 3 times 50 mg 3 times
Enalapril 2.5 mg twice 10–20 mg twice
Lisinopril 2.5–5.0 mg once 20–35 mg once
Ramipril 1.25–2.5 mg once 10 mg once
Trandolapril 0.5 mg once 4 mg once
Angiotensin receptor blocker
Candesartan 4–8 mg once 32 mg once
Losartan 25 mg once 150 mg once
Valsartan 40 mg twice 160 mg twice
Angiotensin receptor neprilysin inhibitor
Sacubitril–valsartan 24 mg/26 mg twice 97 mg/103 mg twice
Mineralocorticoid receptor antagonist
Eplerenone 25 mg once 50–100 mg once
Spironolactone 12.5 mg once 50–100 mg once
Sodium-glucose cotransporter-2 inhibitors b
Canagliflozin 100 mg once 300 mg once
Dapagliflozin c 5 mg once 10 mg once
Empagliflozin 10 mg once 25 mg once
Additional therapy
Ivabradine 5 mg twice daily 7.5 mg twice
Fixed H-ISDN d
Digoxin
37.5 mg/20 mg 3 times
0.125 mg once
75 mg/40 mg 3 times
Higher doses (0.375–0.5 mg) are rarely recommended

a A maximum dose of 50 mg twice daily can be administered to patients weighing over 85 kg.


b No current guideline recommendation.


c Dapagliflozin is the only FDA-approved SGLT-2 inhibitor for patients with HFrEF with and without T2DM.


d Fixed combination of hydralazine plus isosorbide dinitrate (BiDil).



Other commonly mentioned noncardiac side effects of β-blockers include exacerbation of reactive airway disease, increased peripheral vascular resistance, depression, fatigue, insomnia, and sexual dysfunction. Hence, despite extensive evidence of clinical efficacy, several HF registries have shown that the use and dose of β-blockers is often suboptimal, possibly due to side effects or intolerance. Reasons for β-blocker discontinuation have differed across registries, but generally include worsening of HF symptoms, bradycardia, hypotension, dizziness, and fatigue.


β-Βlocker selection may affect tolerability, as patients with reactive airway disease may benefit from β 1 -selective blockers, while those with peripheral vascular disease may benefit from carvedilol, given its vasodilatory effects. The CIBIS-ELD trial was designed to compare the tolerability of bisoprolol and carvedilol during attempted titration to guideline-recommended target doses after 12 weeks of treatment in elderly subjects with chronic HF. It showed that adverse bradycardia was more common with bisoprolol, whereas pulmonary events were more common with carvedilol. Lipophilic β-blockers, such as metoprolol, can cause sleep disturbances, insomnia, vivid dreams, and nightmares, due to the high penetration across the blood–brain barrier. Furthermore, intolerance may not be a class effect, as 80% of subjects considered intolerant to one β-blocker might be successfully changed to another. Actually, contrary to early reports, β-blocker therapy is well tolerated by 90% of HF patients, including those with diabetes mellitus, chronic obstructive lung disease, and peripheral vascular disease.


Severe bradycardia and asthma with active bronchospasm remain the most important contraindications to β-blockade. The dose of β-blocker should be adjusted if HR decreases to < 50 beats/min or in the setting of second- or third-degree heart block. Symptomatic hypotension can be managed by decreasing the diuretic dose. Finally, continuation, but not introduction, of β-blocker treatment during an episode of ADHF is safe, although dose reduction may be necessary.


Angiotensin-Converting Enzyme Inhibitors


The RAAS plays a critical role in the pathophysiology of HF, with effects on cardiac remodeling, vascular tone, endothelial function, sodium retention, oxidative stress, fibrosis, sympathetic tone, and inflammation. The likely mechanisms for RAAS activation include renal hypoperfusion, reduced filtered sodium, and increased SNS stimulation of the kidney, leading to increased renin release from juxtaglomerular apparatus.


ACE inhibitors have many short- and long-term biological effects that are beneficial in patients with chronic HFrEF, and the evidence supporting their efficacy has been consistently demonstrated in large randomized clinical trials. These agents prevent the conversion of angiotensin I to angiotensin II, which is formed by the proteolytic action of renin on circulating angiotensinogen. Angiotensin II is a peptide hormone that causes vasoconstriction, modulates SNS activation, and mediates aldosterone and vasopressin secretion, both associated with abnormal fluid retention and volume regulation in HF. It also promotes a prothrombotic state and abnormal cellular growth. Through RAAS modulation, ACE inhibitors have a consistent effect in increasing plasma renin while decreasing angiotensin II, aldosterone, NE, epinephrine, and vasopressin. Therefore, they reduce SNS activity and improve arterial tone, endothelial function and ventricular compliance, contributing to a decrease in ventricular afterload and cardiac remodeling. In addition, because ACE is identical to kininase II, ACE inhibitors may also lead to the upregulation of bradykinin, an inflammatory mediator with vasodilator properties. These actions may potentially enhance the effects of angiotensin II suppression and further contribute to the vasodilatory effects of ACE inhibitors.


In the era before ACE inhibitors, the V-HeFT trial demonstrated that a combined vasodilator therapy with hydralazine and isosorbide dinitrate (H-ISDN) conferred a nonsignificant survival benefit in subjects with HFrEF. Subsequently, several prospective, randomized pivotal trials have demonstrated a significant survival benefit with ACE inhibitors in that population. The CONSENSUS trial randomized subjects with NYHA class IV HFrEF to enalapril or placebo in addition to standard medical therapy, which until the late 1980s focused primarily on symptom control through use of digitalis, diuretics, and nitrates. The intervention group had a significant 40% relative risk reduction in 6-month mortality. Enalapril was also associated with significant reductions in NYHA class and is the requirement for further HF therapies.


The SOLVD trial randomized subjects with NYHA class II–IV HFrEF to enalapril or placebo and showed a 16% relative risk reduction in the 4-year mortality in the enalapril group, predominantly due to a reduction in the risk of HF mortality. There was also a reduction in the risk of CV hospitalizations and in the end-diastolic LV volume index, which began a recurring observation that some disease-modifying therapies in HFrEF also cause reverse remodeling. Published at the same time as the SOLVD trial, the V-HeFT II showed that enalapril was superior to H-ISDN in patients with NYHA class II–III HFrEF, providing evidence that ACE inhibition improves HF outcomes through additional mechanisms than just vasodilation.


Enalapril is the only ACE inhibitor that has been used in placebo-controlled mortality trials in HFrEF. However, similar favorable effects were found when captopril, ramipril, and trandolapril were started at least 3 days following MI in the SAVE, AIRE, and TRACE trials, respectively. Additionally, the GISSI-3 trial also showed a mortality benefit with lisinopril after MI regardless of subsequent LV function. Taken together, these observations suggest a class effect of ACE inhibitors in the treatment of HFrEF. Thus, ACE inhibitors are strongly recommended to all patients with HFrEF, unless contraindicated or not tolerated. Evidence-based doses for ACE inhibitor are listed in Table 3.5 .


Most side effects of ACE inhibitors are primarily related to interfering with the conversion of angiotensin I to angiotensin II and degradation of bradykinin. Clinical evaluation prior to ACE inhibitor initiation includes assessment of BP, renal function, and electrolytes. It should be emphasized that patients with hypotension or impaired renal function were excluded from most of the clinical trials, and the efficacy of ACE inhibitors for such patients is less well established. Hypotension, early decrease in renal function, and hyperkalemia are dose-dependent side effects that may be avoided by slow dose titration. Symptomatic hypotension and modest reduction in GFR may be initially managed by reducing the dose of diuretics in the absence of significant fluid retention. However, GFR at reduced renal perfusion pressure ultimately depends upon the postglomerular efferent arteriolar vasoconstrictor effects of angiotensin II. Hence, in subjects with renal-artery stenosis or reduced intravascular volume, RAAS blockade might be capable of reducing filtration pressure at critical levels of kidney perfusion, worsening renal function. Progressive loss of GFR can sometimes recover by withholding ACE inhibitors. An ACE inhibitor therapy should be avoided in nondialysis patients with serum creatinine > 3.5 mg/dL or estimated GFR < 20 mL/min/1.73 m 2 , while dialysis patients can be treated with ACE inhibitors. Hyperkalemia can be prevented by prescription of a low-potassium diet, loop diuretics, and prior discontinuation or dose reduction of other medications that raise serum potassium, such as potassium supplements or a potassium-sparing diuretics. A serum potassium level of up to 5.5 mmol/L is acceptable as long as it is stable. In patients with uncontrollable hyperkalemia and an increase of more than 20% to 30% in creatinine level within a week, the ACE inhibitor should be discontinued or down-titrated. Frequent monitoring is advised, although ACE inhibitors may rarely cause severe hyperkalemia that requires emergency management.


Other side effects, as dry cough and angioneurotic edema, are related to the accumulation of bradykinin. Bradykinin is not only a vasodilator, but it also increases prostaglandin concentrations and vascular permeability with fluid extravasation. This clinical distinction is important as side effects related to reduced angiotensin II, but not those related to increased kinins, are also seen with the ARBs. Persistent dry cough, a recognized side effect that occurs in 5% to 20% of subjects on ACE inhibitors, may be dose-related and is not a true allergic effect. When the ACE inhibitor is discontinued, improvement often begins within 4–7 days. Angioneurotic edema is rare (< 1%) and resolves without complications in most cases. However, because endotracheal intubation or emergent tracheostomy may be necessary and fatalities have been reported, an ACE inhibitor should not be prescribed for patients with idiopathic or prior angioneurotic edema. The incidence of ACE inhibitor–induced angioneurotic edema is up to five times greater in individuals of African descent. Actually, there is conflicting evidence on the efficacy of ACE inhibitors in people of African descent, which may be genetically explained by a low-renin system. However, given the available evidence, ACE inhibitor therapy recommendations are the same for all HFrEF patients, regardless of race. Allergic reactions include skin rash, neutropenia, dysgeusia, or anaphylactoid reactions. Both ACE inhibitors and ARBs can cause fetal abnormalities and are absolutely contraindicated in pregnancy in the second and third trimesters.


Angiotensin-converting Enzyme Inhibitors or β -Blockers First


Although the combination of an ACE inhibitor and a β-blocker is more effective in HFrEF than monotherapy with either, the order of initiation is important in clinical practice because patients often cannot tolerate optimum doses of both agents. There are theoretical considerations suggesting it may be more beneficial to initiate the treatment with a β-blocker, as the SNS is systemically activated at an earlier stage than is the RAAS. The CIBIS-III trial addressed this important question and showed that initiating treatment with the selective β 1 -receptor blocker bisoprolol is as effective and well tolerated as beginning treatment with the ACE inhibitor enalapril.


In clinical practice, drug initiation and up-titration could be tailored to individual patients and their unique clinical circumstances. The initial treatment strategy with β-blockers first may be better for subjects with ischemic cardiomyopathies and tachycardia, while the strategy of using an ACE inhibitor first should be indicated for patients with hypertension and fluid overload. Current clinical practice guidelines recommend starting with an ACE inhibitor, followed by the addition of a β-blocker. In most cases, β-blockers are then up-titrated to their maximum recommended dose before further increase in the dose of ACE inhibitors.


Angiotensin Receptor Blockers


Long-term treatment with ACE inhibitors leads to a gradual return of circulating angiotensin II concentrations through non-ACE-dependent pathways. Any angiotensin II that is produced through such alternative pathways remains available for binding with two types of angiotensin receptors, AT-1 and AT-2. The AT-1 receptors are abundant in the vessels, brain, heart, kidney, adrenal, and nerves, while AT-2 are only available in small amounts in the adult kidney, adrenal, heart, brain, uterus, and ovary. Rather than preventing the conversion of angiotensin I to angiotensin II, the ARBs selectively block the binding of angiotensin II to the AT-1 receptors. Activation of AT-1 leads to increased vasoconstriction, aldosterone and catecholamine release, sodium resorption in the proximal tubules of the kidney, and cell growth in the arteries and heart. Thus, antagonizing AT-1 causes a reduction in both cardiac afterload and preload. Also, ARBs may be better tolerated, since they do not interfere with the degradation of bradykinin and do not appear to induce cough and possibly other side effects that force some patients to discontinue treatment with ACE inhibitors.


Given their different mechanisms, earlier rationale postulated that ARB therapy would provide an advantage over ACE inhibitor therapy. However, despite the theoretical benefits, clinical evidence of ARB effectiveness in HFrEF patients is less robust than that of ACE inhibitors. In the Val-HeFT trial, the addition of valsartan to standard HFrEF therapy that included ACE inhibitors did not improve survival but reduced the incidence of the composite outcome of morbidity and mortality, largely through a decrease in HF hospitalizations. Moreover, a retrospective subanalysis of the Val-HeFT suggested that valsartan might also reduce all-cause mortality in the small subgroup of subjects not receiving an ACE inhibitor. The CHARM program consisted of three independent, parallel, placebo-controlled trials evaluating candesartan in different symptomatic HF populations with the same primary composite outcome of CV death or HF hospitalization. HFrEF subjects who received candesartan had significant better outcomes than those who received placebo in both the CHARM-Alternative and CHARM-Added component trials. Specifically, the CHARM-Alternative trial showed that candesartan was well tolerated in symptomatic HFrEF patients who had previously failed an ACE inhibitor, resulting in a 20% relative risk reduction in CV mortality and fewer HF hospitalization. While similar findings were shown in the CHARM-Added trial, the combination of candesartan with an ACE inhibitor and a β-blocker resulted in higher rates of adverse outcomes including increased creatinine and hyperkalemia. Additionally, a later Cochrane meta-analysis showed that the combination of ARBs and ACE inhibitors increased the risk of withdrawals due to side effects, especially renal dysfunction and hyperkalemia, and did not reduce total mortality or hospitalizations when compared to ACE inhibitors alone. It is noteworthy that only 17% of subjects in the CHARM-Added trial were on an MRA, which might be the preferred next agent to add on to the combination of an ACE inhibitor and a β-blocker in patients with HFrEF. Combined use of MRA, ACE inhibitor, and ARB should be avoided because of concerns about hyperkalemia and lack of evidence of efficacy.


Regarding dosing of ARBs, the HEAAL trial showed that high-dose losartan was associated with a significant reduction in HF admissions when compared to low-dose losartan. Thus, they should be started at low doses and up-titrated to the maximum recommended and tolerated dose. Evidence-based doses for ARBs are listed in Table 3.5 . Long-term therapy with ARBs in patients with HFrEF produces hemodynamic, neurohormonal, and clinical effects consistent with those expected after interference with the RAAS. However, controversial results from different meta-analysis cannot confirm their superiority over placebo, particularly when compared to ACE inhibitors. A direct comparison of ACE inhibitors and ARBs was assessed in the ELITE-II trial, which did not observe any mortality benefit of losartan over captopril in elderly subjects with HFrEF. Furthermore, the VALIANT trial showed that valsartan was noninferior to captopril on all-cause mortality in subjects with post-MI LV dysfunction. Although both classes might have similar effects, the general consensus is that an ARB should only be recommended as an alternative for patients with prior or current symptoms of chronic HFrEF who are intolerant to ACE inhibitors due to cough, skin rash, or angioneurotic edema. It should be stated, however, that other side effects such as hypotension, renal dysfunction, and hyperkalemia appear to be similar for ACE inhibitors and ARBs. Additionally, angioedema has also been reported in some patients taking ARBs, although much less frequently than with ACE inhibitors.


ARBs are also reasonable choices as first-line drugs for HFrEF patients who are already on ARB therapy for other indications, like hypertension, and those who are unable to tolerate an MRA for other reasons than renal dysfunction and hyperkalemia. Both ACE inhibitors and ARBs can cause fetal abnormalities and are absolutely contraindicated in pregnancy in the second and third trimesters.


Angiotensin Receptor Neprilysin Inhibitors


In the context of normal cardiac physiology, both the RAAS and SNS are balanced by the NP system to maintain BP and fluid homeostasis. The NP system consists of structurally similar peptides, such as the atrial, B-type, and C-type NPs, that act through receptor-based generation of cGMP, enhancing diuresis, natriuresis, and vasodilation while countering SNS and RAAS overstimulation. As HFrEF progresses, the effects of the NP system become attenuated by several mechanisms, including reduced availability of active forms of NPs, diminished organ responsiveness, and overactivation of counter-regulatory neurohormones. Therefore, pharmacological approaches to enhance the functional effectiveness of the NP system in chronic HFrEF have been proposed.


Initial efforts focused on the IV administration of synthetic forms of NPs, such as nesiritide and ularitide, in the setting of ADHF, but did not improve clinical outcomes. Similar disappointing findings were seen with the lone inhibition of neprilysin (NEP), a neutral endopeptidase responsible for the breakdown of several vasodilator peptides, such as NPs, bradykinin, and adrenomedullin, as well as vasoconstrictors peptides, as angiotensin and endothelin-1. By blocking regulators of opposite actions of vasodilation and vasoconstriction, lone NEP inhibition essentially neutralize each effect and further increase RAAS activation stimulated by upregulation of angiotensin II. Later studies tested omapatrilat, a compound that inhibited both ACE and NEP, to further inhibit angiotensin II in addition to NEP. The OVERTURE trial found that the use of omapatrilat in HFrEF patients was not superior to enalapril alone in reducing mortality and hospitalization but was associated with a higher incidence of angioedema (2.17% versus 0.68%). The increased incidence of this life-threatening complication was attributed to the synergism between the ACE and NEP inhibition on the breakdown of bradykinin. In addition, omapatrilat also inhibits a third enzyme involved in the degradation of bradykinin, the aminopeptidase P. Further clinical research on the entire class of ACE-NEP inhibitors were halted.


The strategy that ultimately proved successful in improving HFrEF outcomes was the molecular combination of sacubitril, a NEP inhibitor, with valsartan, an ARB, resulting in the first-in-class ARNI sacubitril–valsartan. The PARADIGM-HF trial showed a highly significant 20% relative risk reduction in the primary composite outcome of CV death or HF hospitalization of sacubitril–valsartan over enalapril in subjects with NYHA class II–IV HFrEF. Sacubitril–valsartan also reduced secondary outcomes of CV death by 20%, first hospitalization for worsening HF by 21%, all-cause mortality by 16%, and prevented deterioration of symptoms and quality of life.


Since all participants of the PARADIGM-HF were required to first tolerate a run-in phase of enalapril, most HFrEF guideline-issuing societies suggested that sacubitril–valsartan should be initiated only in subjects tolerating full-dose ACE inhibitor or ARB. However, given the positive results of the subsequent PIONEER-HF trial, it is now considered a reasonable strategy to initiate sacubitril–valsartan as a first-line component of the long-term RAAS blockade in hemodynamically stable patients with ADHF. To facilitate initiation and titration, the approved ARNI is available in three doses, including a lower one that was not tested in the PARADIGM-HF trial ( Table 3.5 ).


In the PARADIGM-HF trial, symptomatic hypotension was more common in patients receiving sacubitril–valsartan, although there was no increase in the rate of discontinuation. Measures to avoid hypotension include adjusting the dose of diuretics or other concomitant antihypertensive drugs, correcting volume depletion prior to starting sacubitril–valsartan, and starting at a lower dose. Hyperkalemia and renal dysfunction were reported as adverse events in both treatment groups in PARADIGM-HF, although clinically important increases in serum creatinine were less frequent in the sacubitril–valsartan than in the enalapril group. In addition, among MRA-treated patients, severe hyperkalemia was also more likely with enalapril. Nonetheless, the same precautions for renal impairment and hyperkalemia should be applied to all subjects receiving RAAS inhibitors, including careful screening of baseline renal function and serum potassium concentration, followed by close periodic monitoring. Recruiting only subjects who tolerated both enalapril and sacubitril–valsartan during the active run-in phase reduced the risk of angioneurotic edema in the PARADIGM-HF. However, because of the previous experience with Omapatrilat, a 36-hour washout period when switching from an ACE inhibitor to sacubitril–valsartan is advised. Combined treatment with sacubitril–valsartan and an ACE inhibitor, or a lone ARB, is contraindicated. If none of these three agents is tolerated, the combination of H-ISDN is a potential alternative therapy for patients with HFrEF. Sacubitril–valsartan is also contraindicated during pregnancy due to concerns about risk of teratogenicity with ARBs.


There are also additional concerns about the effects of NEP inhibition on the degradation of β-amyloid peptide in the brain, which could theoretically accelerate amyloid deposition, leading to impaired cognitive function. A subanalysis found no increase in dementia-related adverse events in PARADIGM-HF, but long-term follow-up may be necessary to detect such a signal.


Mineralocorticoid-Receptor Antagonists


Aldosterone is a mineralocorticoid hormone produced primarily by the adrenal cortex but also by endothelial and vascular smooth muscle cells in the blood vessels and myocardium in response to angiotensin II, hyperkalemia, and corticotropin. In addition to its classic mineralocorticoid properties, which can lead to hypokalemia and hypomagnesemia, aldosterone has other adverse effects that can contribute to the pathophysiology of HF, such as inflammation, vascular stiffening, collagen formation, and myocardial necrosis. Therefore, chronically elevated aldosterone is associated with coronary and renovascular remodeling, endothelial and baroreceptor dysfunction, myocardial hypertrophy, and reduced HR variability.


Since angiotensin II prevents renin release by negative feedback, a large increase in plasma renin activity occurs with the administration of ACE inhibitors and ARBs. Moreover, plasma aldosterone returns to pretreatment levels after several weeks of therapy in up to 30% to 40% of patients, either through non-ACE-dependent pathways, or high serum potassium concentrations. MRAs, often referred to as aldosterone antagonists, block receptors that bind aldosterone and, with different degrees of affinity, other steroid hormone receptors such as corticosteroids and androgens. It has been shown that MRAs provide a more complete inhibition of the RAAS when added to standard HFrEF therapy, preventing many of the maladaptive effects of aldosterone. Most of their benefits are mediated by antifibrotic mechanisms, which slows or reverse cardiac remodeling and reduce arrhythmogenesis. In addition, MRAs also preserve serum potassium levels, countering the risk of hypokalemia and further associated arrhythmic risk induced by other diuretics, such as loop diuretics.


The combination of an MRA with standard HFrEF regimen improves survival and reduces morbidity in patients with symptomatic chronic HFrEF, as well as those with LV systolic dysfunction after MI. Such major clinical benefits have been demonstrated in a few well-conducted randomized clinical trials. The first evidence was demonstrated by the RALES trial, which tested spironolactone versus placebo in subjects with NYHA class III–IV HFrEF receiving a background of loop diuretics, ACE inhibitors, and, in most cases, digoxin. RALES demonstrated a 30% relative risk reduction in all-cause mortality with spironolactone, without a significant increase in the risk of serious hyperkalemia or renal failure as hypothesized. HF hospitalization was also 35% lower in the MRA group.


However, although generally well tolerated in RALES, spironolactone was associated with dose-dependent reports of gynecomastia or breast tenderness in 10% of men, compared with 1% in the placebo group. Gynecomastia is clinically defined as benign enlargement of the glandular tissue of male breast due to periductal fibrosis, stromal hyalinization, and increased subareolar fat, while breast pain is caused by inflammatory infiltration of the periductal tissue. Its pathophysiological process usually involves an imbalance between free estrogen and free androgen actions in the breast tissue. Spironolactone induces gynecomastia by decreasing testosterone production in the testicles, increasing its peripheral conversion to estradiol and displacing more estrogen from sex hormone-binding globulin, increasing the bioavailability of estrogen to a greater extent than androgen. Other distressing endocrine side effects of spironolactone include menstrual irregularities in premenopausal females and impotence and decreased libido in men.


Shortly after publication of the RALES trial, the MRA eplerenone became available for clinical evaluation. It was postulated that due to its greater specificity for the mineralocorticoid receptor, eplerenone would have fewer antiandrogenic effects than spironolactone, as a greater fraction of free testosterone could tightly bind to sex hormone-binding globulin. This prompted the subsequent EPHESUS trial, which investigated eplerenone in the treatment of LV systolic dysfunction and clinical evidence of HF or diabetes after an MI, as well as the EMPHASIS-HF trial, investigating eplerenone in the treatment of NYHA class II HFrEF. Both were positive trials, reinforcing the benefits of MRAs in improving survival and hospitalizations in patients with HFrEF, but with fewer endocrine side effects in comparison to spironolactone. Importantly, both trials were performed in the era of widespread β-blocker use, in contrast to RALES in which only 10% subjects were on β-blocker therapy. In EPHESUS, particularly, eplerenone was beneficial on top of all of the existing therapies for MI at that time, including aspirin, reperfusion, and statin. Prior to EMPHASIS-HF, no clinical trial had evaluated the benefits of aldosterone blockade in HFrEF subjects with mild symptoms, as RALES and EPHESUS randomized a population with higher NYHA classes. Therefore, an MRA is recommended for all patients with HFrEF who remain symptomatic despite treatment with an ACE inhibitor (or ARB or ARNI) and a β-blocker, to reduce the risk of HF hospitalization and death, unless contraindicated. Evidence-based doses for spironolactone and eplerenone are listed in Table 3.5 .


The major side effect of MRAs is the development of life-threatening hyperkalemia. Caution should be taken in patients with serum potassium concentrations > 5.0 mmol/L or serum creatinine > 2.5 mg/dL. In RALES, EPHESUS, and EMPHASIS-HF, hyperkalemia was more common in the active treatment groups, although serum potassium concentration ≥ 6 mmol/L was rare. The risks of severe hyperkalemia can be mitigated through appropriate patient selection and education, dose adjustments, careful monitoring of serum potassium levels and renal function, and closer follow-up. The development of worsening renal function should prompt therapy discontinuation.


Although eplerenone is associated with fewer endocrine adverse effects, spironolactone is the most widely prescribed MRA, presumably because of its lower cost. Generally, discontinuation of spironolactone results in resolution of such effects. Thus, it may be reasonable to start MRA therapy with spironolactone, and eventually switch to eplerenone case endocrine side effects occurs. Novel, nonsteroidal MRAs, such as the finerenone, combine the potency of spironolactone with the selectivity of eplerenone, resulting in less hyperkalemia and greater decrease in BNP and NT-proBNP levels. Exploratory analysis of the phase 2 ARTS-HF trial suggested a more favorable effect of finerenone on CV mortality and HF hospitalizations when compared to eplerenone. The relative effectiveness of finerenone over eplerenone remains to be determined in a large outcomes trial.


Sodium-Glucose Cotransporter-2 Inhibitors


The SGLT-2 inhibitors, also called “gliflozins,” are a class of medications that reduce glucose reabsorption by inhibiting the high-capacity sodium-glucose cotransporter-2 in the proximal tubule of the nephron. This leads to an increased concentration of chloride in the distal tubule and a resetting of the tubulo-glomerular feedback mechanism, which results in a contraction of the plasma volume without activation of the SNS. Empagliflozin, canagliflozin, and dapagliflozin have been shown to reduce the risk of HF hospitalization in three large clinical trials in T2DM patients at high cardiovascular risk: the EMPA-REG Outcomes, the CANVAS Program, and the DECLARE-TIMI 58 trial. The DAPA-HF trial was the first outcomes trial with an SGLT-2 inhibitor investigating the treatment of HFREF irrespective of T2DM status. The DAPA-HF trial randomized subjects with NYHA class II–IV HFrEF to dapagliflozin 10 mg or placebo, in addition to standard care, and showed a statistically significant 26% reduction in the risk of the primary composite outcome of worsening HF or CV death. When analyzed separately, worsening HF was reduced by 30% and the risk of CV death was reduced by 18%. Notably, the treatment effect was consistent across all prespecified subgroups, including patients without baseline T2DM and regardless of hemoglobulin A1c levels. In addition, dapagliflozin reduced the risk for all-cause death by 17% and showed a significant improvement in patient-reported HF symptoms. Adverse events related to volume depletion, renal adverse events, major hypoglycemia, lower limb amputation, and fracture were comparable to placebo, with low rates of discontinuation in both groups (< 5%). The potential mechanisms of benefit from gliflozins in HFrEF are likely multifactorial, involving not only glycosuria or osmotic diuresis but also cardio-renal protection through metabolic and hemodynamic effects.


Additional Therapy


Diuretics


Diuretics are the cornerstone of therapy for the treatment of volume overload in patients with HF, relieving clinical symptoms and signs more rapidly than any other drug. They can reduce CVP, pulmonary congestion, peripheral edema, and body weight within hours or days of initiation of therapy, whereas the clinical effects of digoxin, ACE inhibitors, or β-blockers may require weeks or months to become apparent. Additionally, in the intermediate term, diuretics have also been shown to improve cardiac function and exercise tolerance in HF patients. However, the effects of diuretic therapy with long-term follow-up in chronic HFrEF have not been studied in large prospective randomized controlled trials. A Cochrane meta-analysis suggested that in patients with chronic HF, conventional diuretic therapy might reduce the risk of death and worsening of HF compared to placebo and appear to improve exercise capacity compared to active control. However, this meta-analysis included only small-size studies with limited follow-up and cannot be used as formal evidence to recommend the use of diuretics to reduce HF mortality.


Patients at higher risk for fluid overload would benefit from long-term therapy with oral loop diuretics. However, the use of loop diuretics might associate with electrolyte disturbances, arrhythmias, neurohormonal activation, accelerated renal function decline, and hypotension. The latter might be particularly relevant, as it could limit optimal target doses of neurohormonal antagonists. Therefore, it is advised to use the lowest possible dose of loop diuretic, which should be adjusted individually. As previously stated, diuretics do not have a smooth dose-response curve, and natriuresis will only occur when the threshold rate of drug excretion, which is often higher in HF patients, is attained. Hence, if there is little or no response to the initial dose, it is recommended to double it rather than giving the same dose twice a day. Additionally, for patients presenting with an ADHF episode while previously taking a loop diuretic, a higher dose at discharge should be considered.


Thiazide diuretics are widely used for the management of hypertension. Despite structural variation among the heterogeneous group of agents with similar physiological properties, including the thiazide-like diuretics, the term “thiazide diuretic” comprises all diuretics believed to have a primary action in the distal convoluted tubule. Metolazone, a quinazoline sulfonamide, is a thiazide-like diuretic that can be used in combination with furosemide in patients with diuretic resistance. The chronic use of sequential nephron blockade with thiazide diuretics should be avoided in stable patients, as it often induces electrolyte disorders that could go unseen in the ambulatory setting.


Since sodium retention occurs in more proximal tubular sites, potassium-sparing diuretics are ineffective in achieving a net negative sodium balance when given alone in HF patients. Besides, the benefit of MRAs in patients with HF is thought to be relatively independent of their diuretic properties and may be due to their ability to antagonize the RAAS.


Vasopressin receptor antagonists modulate the renal actions of AVP by directly blocking its receptors V1A, V1B, and V2. The inappropriate elevation of AVP in HF plays a key role in mediating vasoconstriction, water retention, and electrolyte imbalance. Combined V1A and V2 antagonism causes a decrease in SVR and prevents dilutional hyponatremia. The AVP antagonists, or “vaptans,” can either selectively block the V2-receptor or nonselectively block both the V1A- and the V2-receptors. The EVEREST-Outcomes trial randomized subjects admitted with ADHF to the selective V2-receptor antagonist tolvaptan or placebo and showed no difference in long-term mortality or HF-related morbidity at a median follow-up of 9.9 months. Currently, conivaptan and tolvaptan are FDA-approved for the treatment of clinically significant hypervolemic and euvolemic hyponatremia, but they are not officially approved for HF. Although it is reasonable to use these agents after traditional measures to hyponatremia have failed, including fluid restriction and optimal target doses of angiotensin inhibitors, their widespread use might be limited by high costs.


Patients receiving diuretics should be monitored regularly for complications, including electrolyte and metabolic disturbances, volume depletion, and worsening renal function. Even when successful in controlling symptoms and fluid retention, diuretics alone are unable to maintain the clinical stability of patients with chronic HF for long periods of time. Therefore, they should be combined with standard guideline recommended HFrEF therapies, including neurohormonal antagonists.


Combination of Hydralazine Plus Isosorbide Dinitrate


Until the 1970s, standard medical therapy for HF was limited to digitalis and diuretics. A hemodynamic rationale for the combined use of a venous and arterial vasodilator therapy is to reduce both preload and afterload by improving venous capacitance while decreasing SVR. Early pivotal studies found that the combination of two oral vasodilators, hydralazine and isosorbide dinitrate, in patients with NYHA class III–IV HF results in a better hemodynamic response than either drug individually. Additionally, since nitrates are NO donors and hydralazine has antioxidant properties, it is possible that the NO-mediated effects of H-ISDN might also play a complex role in the maintenance of CV health.


The V-HeFT trial was the first major randomized, placebo-controlled HF study, which compared H-ISDN, prazosin, or placebo in subjects with symptomatic HF and LVEF < 45%. At a mean follow-up of 2.3 years, there was no statistically significant difference in mortality between the groups, although H-ISDN was associated with a trend toward improved survival when compared to placebo. H-ISDN also improved LVEF at 8 weeks and 1 year, while prazosin was associated neither with mortality nor LVEF improvement. The subsequent V-HeFT II trial was undertaken to compare H-ISDN with enalapril in a similar population to that of V-HeFT and showed a survival benefit with enalapril at the primary specified time point of 2 years but not during the entire follow-up of the trial. The survival benefit was largely driven by a reduction in sudden cardiac death, but there were no significant differences in rates of hospitalizations between the two groups.


Of interest, post hoc subgroup analysis from both V-HeFT and V-HeFT II trials suggested improved survival benefit with H-ISDN among self-identified black subjects, which was the basis of the A-HeFT trial. Based on the rationale that persons of African descent may have less RAAS activity, rendering ACE inhibitors less effective, the A-HeFT trial randomized self-identified black subjects with NYHA class III–IV HFrEF to either placebo or a fixed combination of H-ISDN. The trial was stopped early at a mean follow-up of 10 months due to a significant 43% relative risk reduction in all-cause mortality associated with the use of H-ISDN added to standard therapy with diuretics, ACE inhibitors, β-blockers, digoxin, and spironolactone. H-ISDN was also associated with a lower rate of first and recurrent HF hospitalizations and a significant improvement in quality-of-life assessments. Importantly, the A-HeFT trial tested a fixed-combination single-pill equivalent to the generic H-ISDN, called BiDil, but it is probable that the same benefits can be achieved with a combination of the separate formulations. BiDil has provoked controversy as the first FDA-approved drug marketed for a single racial-ethnic group—those of African descent—in the treatment of HFrEF.


Guideline-issuing professional societies recommend the combination of H-ISDN for self-identified black patients with NYHA class III–IV HFrEF who remain symptomatic despite concomitant use of ACE inhibitors, β-blockers, and MRAs. The results of the A-HeFT trial are difficult to translate to other racial or ethnic origins, and there is less robust evidence supporting the use of H-ISDN as a contemporary first-line therapy for nonblack HFrEF subjects. Nonetheless, based on the V-HeFT trial, which recruited patients who received only digoxin and diuretics, H-ISDN may be considered in symptomatic HFrEF patients who cannot be given an ACE inhibitor, ARB, or ARNI because of drug intolerance, hypotension, or renal dysfunction. Evidence-based doses for H-ISDN are listed in Table 3.5 . Concomitant use of any form of nitrate with PDE-5 inhibitors or sGC stimulators is contraindicated due to the increased risk of refractory hypotension.


If Channel Inhibitor


An elevated HR in patients with HFrEF reflects, in part, an imbalance between sympathetic overstimulation and parasympathetic suppression, which are components of the neurohumoral activation. It has been shown that increased HR not only predicts CV death and HF hospitalization but may also be a therapeutic target in patients with chronic HFrEF. Several classes of pharmacological agents can modulate HR, including β-blockers, ivabradine, digoxin, amiodarone, and nondihydropyridine CCBs, such as diltiazem and verapamil. While some agents are associated with clinical benefits in patients with HFrEF, such as β-blockers and ivabradine, others provide no direct benefit and should generally be avoided, such as most CCBs. Choice will depend on heart rhythm, comorbidities, and disease phenotype.


Although the conventional definition of tachycardia is a resting HR over 100 beats/min, the risk of adverse CV outcomes in HFrEF patients appears to increase progressively with HR ≥ 70 beats/min. β-Blockers reduce HR by antagonizing the adverse effects of the SNS and have become a cornerstone for HFrEF therapy. In some cases, however, recommended target doses cannot be tolerated or are contraindicated, and other drugs should be considered for adequate HR control. Ivabradine is a selective blocker of the cardiac pacemaker If (“funny”) channel current that controls the spontaneous diastolic depolarization of the sinoatrial node, approved for the treatment of HFrEF in the United States. Specific blockade of the If channels prolongates the slow depolarization phase, causing a dose-dependent reduction in HR. As a consequence, ivabradine does not affect contractility or SVR.


Initially developed and approved as an antianginal agent in Europe, ivabradine was also shown to improve outcomes in subjects with HFrEF. The BEAUTIFUL trial randomized subjects with IHD to ivabradine or placebo and did not meet its primary outcome of reducing CV death, MI, or HF hospitalization. However, in a post hoc analysis, ivabradine was shown to reduce rates of MI and coronary revascularization in the subgroup with a HR ≥ 70 beats/min, yielding insights into the potential benefits of additional HR reduction in patients with HFrEF. The SHIFT trial randomized symptomatic HFrEF patients with LVEF ≤ 35%, sinus rhythm, and resting HR ≥ 70 beats/min despite maximally tolerated β-blocker therapy to either ivabradine or matching placebo. The primary composite outcome of HF hospitalization or mortality was reduced by 18% in the ivabradine group, driven primarily by 26% fewer HF hospitalizations, since there was no decrease in CV or all-cause deaths. Remarkably, only 26% of subjects in SHIFT were taking optimal target doses of β-blockers and only 56% were on at least half-target doses at inclusion. Given that ivabradine lowered HR by approximately 10 beats/min, and that its benefits were somewhat attenuated in the subgroup of subjects on at least half-target doses of β-blocker therapy, it is possible that titrating β-blockers to recommended doses may have reduced HF hospitalizations to a similar degree.


Overall, ivabradine was well tolerated in the SHIFT trial, with only a modest increase in bradycardia. Transient visual disturbances secondary to effects on the hyperpolarization-activated current within the retina, referred to as phosphenes, were the most common non-CV side effect, but they frequently improved over time. Current HF guidelines recommend considering ivabradine in HFrEF patients with LVEF ≤ 35%, sinus rhythm, and resting HR ≥ 70 beats/min who remain symptomatic despite β-blocker therapy at maximum tolerated dose. It should be noted that the off-label use of ivabradine for inappropriate sinus tachycardia or other electrophysiological disorders lacks evidence support and remains unapproved. Evidence-based doses for ivabradine are listed in Table 3.5 .


Ivabradine is contraindicated in patients with ADHF, hypotension, sinus node dysfunction, sinus bradycardia with a HR < 60 beats/min, and sinoatrial (SA) or third-degree atrioventricular (AV) block, unless a functioning demand pacemaker is present. Other contraindications for ivabradine are severe hepatic dysfunction, pacemaker dependence, and known atrial or ventricular arrhythmias. In the SIGNIFY trial, subjects with CAD treated with ivabradine had an increase in relative risk for AF, which is consistent with the findings of a later meta-analysis. Ivabradine also has the potential to cause fetal toxicity, and women of reproductive age should therefore be advised.


Digoxin


Digoxin is a purified cardiac glycoside derived from the foxglove plant that has been used for more than 200 years to treat HF. It acts by inhibiting the sodium–potassium (Na-K) ATPase pump in cell membranes, reducing the transport of sodium from the intracellular to the extracellular space in both cardiac and noncardiac cells and contributing to hemodynamic, neurohumoral, and electrophysiologic effects ( Fig. 3.7 ).


Jan 3, 2021 | Posted by in CARDIOLOGY | Comments Off on Heart Failure

Full access? Get Clinical Tree

Get Clinical Tree app for offline access