Next Article in Journal
Machine Learning Algorithms: Prediction and Feature Selection for Clinical Refracture after Surgically Treated Fragility Fracture
Next Article in Special Issue
Transversal Arch Clamping for Complete Resection of Aneurysms of the Distal Ascending Aorta without Open Anastomosis
Previous Article in Journal
Dislocation Arthropathy of the Shoulder
Previous Article in Special Issue
Prognostic Impact of Hybrid Comprehensive Telerehabilitation Regarding Diastolic Dysfunction in Patients with Heart Failure with Reduced Ejection Fraction—Subanalysis of the TELEREH-HF Randomized Clinical Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Up-to-Date Article Regarding Particularities of Drug Treatment in Patients with Chronic Heart Failure

1
Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
2
Research Center for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
3
Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
4
Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
5
Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania
6
Center for Advanced Research in Cardiovascular Pathology and Hemostasis, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(7), 2020; https://doi.org/10.3390/jcm11072020
Submission received: 3 March 2022 / Revised: 24 March 2022 / Accepted: 1 April 2022 / Published: 4 April 2022
(This article belongs to the Special Issue Advances in the Management of Cardiovascular Disease)

Abstract

:
Since the prevalence of heart failure (HF) increases with age, HF is now one of the most common reasons for the hospitalization of elderly people. Although the treatment strategies and overall outcomes of HF patients have improved over time, hospitalization and mortality rates remain elevated, especially in developed countries where populations are aging. Therefore, this paper is intended to be a valuable multidisciplinary source of information for both doctors (cardiologists and general physicians) and pharmacists in order to decrease the morbidity and mortality of heart failure patients. We address several aspects regarding pharmacological treatment (including new approaches in HF treatment strategies [sacubitril/valsartan combination and sodium glucose co-transporter-2 inhibitors]), as well as the particularities of patients (age-induced changes and sex differences) and treatment (pharmacokinetic and pharmacodynamic changes in drugs; cardiorenal syndrome). The article also highlights several drugs and food supplements that may worsen the prognosis of HF patients and discusses some potential drug–drug interactions, their consequences and recommendations for health care providers, as well as the risks of adverse drug reactions and treatment discontinuation, as an interdisciplinary approach to treatment is essential for HF patients.

1. Introduction

Heart failure is defined by the European Society of Cardiology Guidelines as a clinical syndrome derived from structural and/or functional cardiac abnormalities [1]. This syndrome is characterized by common symptoms (such as fatigue, breathlessness, or ankle swelling) and typical signs (such as peripheral edema, elevated jugular venous pressure, or pulmonary crackles), all leading to reduced cardiac output and/or high intracardiac pressure (at rest or during stress periods) [2]. Thus, heart failure (HF) can be defined as the inability of the heart to ensure optimal blood flow, which is necessary for the organs to maintain the metabolic and functional processes of all organs. Currently, several HF definitions are available, which differ in the function of the setting (from the medical literature to medical practice, including the current guidelines) [3]. Moreover, several classification frameworks also exist, which aim to properly characterize different subsets of HF (from NYHA classification to EF categories or HF etiology) [3].
Drug therapy is a well-established strategy in treating heart failure (HF) and there are guidelines that cover most of the long-standing and recent research, although specific situations cannot be extensively analyzed [2].
HF affects a wide range of patients and thus occurs in several forms that are widely based on the assessment of left ventricular (LV) ejection fraction (EF): heart failure with preserved ejection fraction (HFpEF), with mildly reduced ejection fraction (HFmrEF), and with reduced ejection fraction (HFrEF) [4]. HFpEF is considered in patients with normal LVEF (commonly considered to be ≥50%, with symptoms and signs, elevated levels of natriuretic peptides and 1 additional criterion of relevant structural heart disease/diastolic dysfunction at least) [3]. The HFrEF designation is typically applied to patients presenting with less than 40% EF. Patients with LVEF between 40 and 49% represent a gray area, nowadays referred to as HFmrEF [5].
The diagnosis of HFpEF is relatively demanding, as patients usually have a normal LV, with a certain degree of wall hypertrophy and/or increased left atrial (LA) volume. Diagnosis requires proof of increased LV filling pressure or impaired LV filling, which explains the terminology of diastolic HF/dysfunction. However, diastolic dysfunction is also found in most HFrEF and HFmrEF patients (previously referred to as systolic HF) [2].
It seems that the prevalence of HF significantly varies with age, starting from approximately 1–2% among adults and increasing strikingly to more than 10% in people older than 70 years, and is more common in men than in women [2,6]. Since HF incidence and prevalence increase with age, HF is nowadays one of the most common reasons for hospitalization of elderly people [7]. Although treatment strategies and overall outcomes of HF patients have improved over time, hospitalization and mortality rates still remain elevated, especially in developed countries where the population is aging, which represents an economic burden for healthcare budgets [7].
Except for the pathology itself, HF is also associated with co-morbidities such as prior stroke or myocardial infarction, atrial fibrillation, diabetes mellitus, chronic lung disease, osteoarthritis, thyroid disease, dementia, depression, and chronic renal/hepatic failure, and all of these pathologies require additional treatment strategies [8].
Moreover, several aspects, such as changes in the mechanisms (neuroendocrine, inflammatory, immunological, or metabolic) involved in the physiopathogenesis of HF [7], the presence of co-morbidities, the use of polypharmacy in HF patients, and altered pharmacokinetics and pharmacodynamics of drugs in elderly people [9,10], require a patient-centered approach in order to avoid inappropriate medical prescriptions, drug interactions, exacerbation of adverse drug effects [11], and low adherence to pharmacological treatment, with altered prognosis of HF patients being a major consequence [12,13,14].
Moreover, because the worldwide population is aging and the number of people ≥80 years old will triple by 2050, it is extremely important to decrease the prevalence and incidence of cardiovascular pathologies in order to decrease multi-morbidity and health care costs [15].
Herein, we discuss the main important aspects regarding pharmacological approaches, treatment strategies, and the particularities of patients and treatment that should absolutely be taken into account in order to improve treatment outcomes for HF patients.

2. General Considerations Regarding HF Treatment

The pharmacological treatment of HF is oriented towards the following: long-term management of the pathology and improvement in survival (e.g., ACEIs (angiotensin-converting enzyme inhibitors), ARBs (angiotensin receptor blockers), beta-blockers, MRA (mineralocorticoid receptor antagonists), ARNI (angiotensin receptor-neprilysin inhibitors), SGLT2 (sodium-glucose cotransporter-2) inhibitors and ivabradine, an If channel blocker, highly selective for sinoatrial node pacemaker current) and symptom relief medication (e.g., administration of diuretics, nitrates or digoxin). Except for loop diuretics and digoxin, all of these options for treatment have been shown to improve symptoms, reduce hospitalization rates and/or prolong survival, in large randomized controlled trials [16,17].
Drug selection for HF depends on the type of HF and on the personal characteristics of the patients, the most important goals being as already mentioned: to reduce mortality, to improve clinical status and functional capacity and to prevent hospitalization [16,17].

2.1. Mechanisms of Action of the Classical Therapy in Chronic HF Patients

Later, we summarize the mechanisms of actions and the benefits of the main classes of drugs/pharmaceutical substances used in the treatment of HF.
Beta-blockers bind to β adrenergic receptors (β1-receptors located in the heart and kidneys; β2-receptors located in the vessels, lungs, gastrointestinal tract, liver, uterus, and skeletal muscle; β3-receptors located in the adipocytes). By binding toβ1-receptors, they block the deleterious actions of catecholamines: noradrenaline and adrenaline [18]. As a result, the heart rate and the contractility decrease and thus, the cardiac output and blood pressure will also decrease. As the heart rate will decrease, this will allow a longer time for diastolic filling, without typically reducing the stroke volume. Moreover, certain beta-blockers (cardioselective ones) will also reduce rennin secretion (via the blockade of β1 receptors in the juxtaglomerular apparatus), thus decreasing the severity of angiotensin II-induced vasoconstriction and aldosterone-induced volume expansion [19]. They are classified into noncardioselective beta-blockers (e.g., propranolol, carvedilol, and labetalol) and cardioselective beta-blockers (β1-selective, e.g., atenolol, metoprolol, bisoprolol and nebivolol). Certain beta-blockers are associated with vasodilating properties (nebivolol improves nitric oxide release, whereas carvedilol and labetalol block the α1-receptor). The vasodilating properties are beneficial because they decrease the peripheral vascular resistance, thus improving stroke volume, left ventricular function and therefore, cardiac output [18].
ACEIs selectively inhibit the angiotensin-converting enzyme leading to decreased angiotensin II production and, therefore, limit its negative effects, such as vasoconstriction, antidiuretic hormones, and aldosterone secretion. Moreover, ACEIs will increase the levels of the potent vasoactive peptide bradykinin, an endogenous vasodilator. Thus, ACEIs will induce vasodilatation, decreasing the total peripheral resistance (both arterial and venous) and blood pressure. In this way, they decrease the left ventricular afterload, thus increasing cardiac output and decreasing filling pressures (both left and right), which will improve pulmonary and systemic venous congestion [20].
ARBs work on the same angiotensin pathway, the difference being the fact that they bind to AT1 receptors located on the vascular smooth muscle, as well as in other tissues (e.g., heart) and thus, they block the damaging actions of angiotensin II. They induce less vasoconstriction and antidiuretic hormone and aldosterone secretion and lower blood pressure. Therefore, as well as ACEIs, they prevent damage to the vasculature, heart and kidneys [20].
As in some cases, the ACEIs or ARBs do not suppress the excessive formation of aldosterone sufficiently, patients with moderate to severe heart failure can also benefit from aldosterone antagonists. MRAs work by competitively blocking the binding of aldosterone to the mineralocorticoid receptor, thus decreasing the reabsorption of sodium and water, as well as decreasing the excretion of potassium, leading to cardioprotective effects [21].
Loop diuretics act by inhibiting the luminal sodium-potassium chloride cotransporter located in the thick ascending limb of the loop of Henle, where approximately 20–30% of the filtration of sodium occurs. Therefore, compared with other diuretics, loop diuretics reduce the reabsorption of a much greater proportion of sodium, leading to the excretion of it, alongside water. This will decrease the plasma volume, cardiac workload, and oxygen demand, thereby relieving the signs and symptoms of volume excess. They are currently used to relieve symptoms associated with pulmonary congestion and peripheral edema in HF patients [22].
If the patient is intolerant to ACEIs/ARBs/ARNIs, other vasodilators can be used, such as isosorbide dinitrate or hydralazine. Isosorbide dinitrate acts by releasing nitric oxide into the vascular smooth muscle cell, which activates guanylyl cyclase (an enzyme that catalyzes the formation of cyclic guanosine monophosphate-cGMP from guanosine triphosphate–GTP). Therefore, the increased intracellular cGMP will activate a series of reactions, which will decrease the intracellular calcium and thus, the contractility of vascular smooth muscle, leading to smooth muscle relaxation and vasodilatation. Hydralazine also acts on the vascular smooth muscle, with multiple effects such as the stimulation of nitric oxide release from the vascular endothelium (with cGMP production and low-intracellular calcium concentration), opening of potassium channels and inhibition of calcium release from the sarcoplasmic reticulum, thus inducing smooth muscle relaxation and subsequent vasodilatation [23].
Digoxin increases cardiac muscle cells’ contractility by inhibiting Na+/K+/ATPaze pump in the cardiac muscle, a pump responsible for moving sodium ions out of the cells and bringing potassium ions into the cells. When sodium concentrations in the cardiac cell increases, another electrolyte mover known as the sodium-calcium exchanger pushes the excess of the sodium ions out, while bringing additional calcium ions in. Therefore, the intracellular calcium increases, which will later increase the force of contraction and thus the cardiac output. Cardiac output increases followed by a decrease in ventricular filling pressures. Moreover, it inhibits the atrio-ventricular node, by stimulating the parasympathetic nervous system. Therefore, it diminishes the electrical conduction in the AV node and thus the heart rate. However, it has not been shown to reduce mortality [24].
Ivabradine acts by blocking the If current channel, responsible for the cardiac peacemaker, which regulates the heart rate. In this way, it prolongs the diastolic time and decreases the heart rate without affecting myocardial contraction/relaxation or ventricular repolarization [25].

2.2. New Approaches in HF Pharmacological Treatment

As several pharmacological classes of drugs have emerged in recent years with proven long-term benefits, in the following, we describe some of the most important aspects, as they are currently underused.

2.2.1. Sacubitril/Valsartan

The combination of sacubitril and valsartan is the first from the class of angiotensin receptor–neprilysin inhibitors (ARNI). Agents in this new therapeutic class (sacubitril/valsartan) act at the level of RAAS and the neutral endopeptidase system. Sacubitril acts by inhibiting neprilysin and slowing down the degradation of natriuretic peptides, bradykinin, adrenomedullin, and other peptides [26]. It is indicated in chronic symptomatic heart failure with reduced ejection fraction [27].
Sacubitril/valsartan also improves symptom severity and heart functionality in patients with HFpEF, reducing the serum levels of the biomarker NT-pro BNP (and increasing BNP), an indicator of heart failure severity, and improves quality of life after 24 weeks [28].
One of the largest HF trials ever performed (PARADIGM-HF trial) compared enalapril with sacubitril/valsartan. In this trial, 8442 patients with HFrEF with FEVS ≤ 40% were enrolled and randomly received enalapril or sacubitril/valsartan twice daily. The trial was stopped early after 27 months because sacubitril/valsartan met the pre-specified stopping endpoint for an overwhelming benefit. All of the outcomes showed a 20% lower event rate in favor of sacubitril/valsartan; even the death rate from any cause was 16% lower in the group receiving sacubitril/valsartan [29]. ARNIs have been associated with improvements in diastolic function, left ventricular function, quality of life and decrease in ventricular arrythmias [30,31].
In the PROVE-HF and EVALUATE-HF trials, sacubitril/valsartan showed efficacy in improving the structural and functional changes that occur during heart failure. It improves cardiac remodeling and decreases the biomarker NT-pro BNP, so the drug reverses the damage to the heart in HFrEF patients [32].
Sacubitril/valsartan is recommended to replace ACE inhibitors when HFrEF patients are still symptomatic after optimal therapy. When initiating therapy with sacubitril/valsartan, there are some safety issues, including symptomatic hypotension, angioedema, and risk of hyperkalemia, so monitoring blood pressure levels, kidney function, and kalemia is extremely important [27,33]. Although the new combination was approved for the market starting from 2015, it is currently still underused, despite its proven benefits [34].
Figure 1 presents the mechanism of action of sacubitril/valsartan association and its consequences [27,28,29,30,31,32,33,34].

2.2.2. Sodium Glucose Co-Transporter-2 Inhibitors

It is known that patients with type 2 diabetes mellitus (T2DM) are prone to developing cardiovascular events and heart failure, which can lead to high rates of hospitalization and premature mortality [35].
A new class of antidiabetics, sodium glucose co-transporter-2 (SGLT2) inhibitors, has also been found to have beneficial effects in patients with cardiac diseases [36,37]. The compounds in this class are represented by empagliflozin, dapagliflozin, canagliflozin, and ertugliflozin [38]. They act by inhibiting glucose transport in the proximal tube of the kidney, resulting in glucosuria and, as a result, lower blood glucose levels [35].
Aside from the direct mechanism of action on glucose control, other indirect mechanisms are taken into account regarding possible cardiovascular benefits [39].
In Figure 2, we summarize the possible mechanisms involved, their actions, and their effect on the heart [39,40].
The main trials reporting the benefits of SGLT2 inhibitors in HF patients with reduced EF, more precisely of dapagliflozin and empagliflozin, are as follows: DAPA-HF [41], DEFINE-HF [42] and EMPEROR-reduced [43].
The DAPA-HF trial evaluated the long-term effects of dapagliflozin on the incidence of cardiovascular death or HF hospitalization, regardless of the presence of diabetes. It was a phase 3 randomized placebo-controlled study, enrolling 4744 patients suffering from chronic HFrEF (NYHA class II-IV, LVEF ≤ 40% in addition to the recommended HF therapy, NT-proBNP high and eGFR ≥ 30 mL/min/1.73 m2) and having a median period of 18 months. The obtained results were as follows: a reduction in all-cause mortality and HF symptom aggravation, and the improvement in physical condition and overall quality of life. Their excellent benefits were seen very soon after starting the treatment with dapagliflozin. Regarding the incidence of adverse effects, they were attributed to volume depletion, renal dysfunction or hypoglycemia, but they did not differ between the studied groups [41].
The DEFINE-HF trial assessed the effect of dapagliflozin on the symptoms and biomarker plasmatic concentration of HFrEF patients (NYHA class II-III, LVEF ≤ 40%, eGFR ≥ 30 mL/min/1.73 m2 and with elevated natriuretic peptides). In total, 263 patients were included (taking either dapagliflozin 10 mg/once, daily, or a placebo, for a period of 12 weeks, in addition to the recommended HF therapy). Dapagliflozin induced an improvement in the patients’ health conditions or in their natriuretic peptides’ plasmatic concentrations [42].
The EMPEROR-reduced clinical trial evaluated the outcome of empagliflozin in patients with chronic HFrEF (NYHA class II-IV, LVEF ≤ 40%, eGFR ≥ 20 mL/min/1.73 m2). It was a double-blind clinical trial involving 3730 patients who received either empagliflozin (10 mg/once daily) or a placebo, in addition to the recommended HF therapy, for a median period of 18 months. Cardiovascular death and hospitalization rates (due to the worsening of HF) were reduced by empagliflozin, regardless of the presence of diabetes mellitus. The annual decline in the renal filtration rate was reduced, as well as the severity of renal complications. Non-complicated fungal infections of the genital tract were reported more often in patients taking empagliflozin [43].
Therefore, both substances are included as recommend treatments for HFrEF patients by the American and European guidelines [1,17].
The EMPEROR-preserved study assessed the effects of empagliflozin in patients with chronic HFpEF (NYHA class II-IV, LVEF ≥ 40%, eGFR ≥ 20 mL/min/1.73 m2). In total, 5988 patients were included, who were randomized 1:1 and received either empagliflozin (10 mg/once daily) or a placebo, in addition to their classical HF therapy. Over a period of 26.2 months, the primary outcome was obtained (decreased risk of hospitalization in HF patients, regardless of the presence/absence of diabetes). Beneficial effects were also seen in eGFR, without considering the renal outcomes by themselves. It is important to note the fact that the most used medicines for HFrEF have not shown benefits in patients with HFpEF; therefore, empagliflozin is superior in improving HF outcomes even in patients with HFpEF, which are symptomatic and stable [44,45].
In Table 1, we summarize the indications, contra-indications and cautions worth considering for ARNI and SGLT2 inhibitors [17].

3. Treatment Strategies in HF Patients

For the treatment of heart failure with preserved ejection fraction or with mildly reduced ejection fraction (LVEF ≥ 50% or LVEF between 40 and 49), the guidelines recommend the prescription of diuretics, as first line therapy [1]. The other drugs (ACEI or ARB, beta-blockers or MRA) may be considered as a second alternative [1].
The treatment strategy also focuses on treating co-morbidities such as: hypertension, atrial fibrillation, cardiac ischemic disease, pulmonary hypertension, diabetes mellitus, chronic kidney disease, COPD (chronic obstructive pulmonary disease), anemia and obesity. The optimal management of co-morbidities has been shown to improve symptoms and to improve the patient’s quality of life [2].
In the case of congestion, diuretics will be very effective and will improve the symptomatology. There is proof that nebivolol, candesartan, digoxin and spironolactone might reduce hospitalization for patients with HFpEF in sinus rhythm [46]. Moreover, besides empagliflozin, none of other drugs consistently met their primary endpoint in the clinical trials that were performed, and none reduced mortality and morbidity [44,45].
For patients in atrial fibrillation, the prescription of an anticoagulant is very important for reducing thrombo-embolic events [47]. For the control of heart rate, the use of digoxin, beta-blockers or verapamil/diltiazem is recommended, targeting an optimal rate control between 60 and 100 bpm [48].
Amiodarone and non-dihydropyridine calcium-channel blockers (CCB) are able to reduce heart rate, but due to their adverse effects profile, they should be replaced, if possible. In the case of a fast ventricular rate and symptoms, it might be appropriate to consider AV node ablation, and if there are indications for ICD (implantable cardioverter-defibrillator), AV node ablation with the implantation of CRT-D (cardiac resynchronization therapy–defibrillator) might be preferred. The rhythm control strategy has not been shown to be superior to the rate control strategy. Urgent cardioversion is indicated if atrial fibrillation is life threatening [49].
Regarding HFrEF treatment, the evidence base for drug treatment in HF is for HFrEF. Either an ACEI/ARB/ARNI or a beta-blocker should be started (sometimes also ACEI/ARB/ARNI and beta-blocker at the same time), with doses up-titrated to the maximum tolerated/targeted dose every 2 weeks. ACEI, beta-blockers and MRA proved to improve survival and are recommended for the treatment of every patient with HFrEF. The new ARNI (sacubitril/valsartan) has been shown to be superior to ACEI in reducing the risk of death and hospitalization. Thus, ARNI is recommended to replace ACEI in cases of HFrEF patients if they are symptomatic despite optimal therapy [26].
In the case of decompensated patients, beta-blockers should not be initiated or if already initiated but patients develop worsening of HF symptoms (e.g., fatigue, dyspnea, dizziness or erectile dysfunction) caution should be applied regarding their prescription. Moreover, in the case of frailty or other complications (e.g., marginal hemodynamics), a longer period of time may be required for dose up-titration [17].
ARNI can be prescribed as an alternative to ACEI/ARB intolerance (e.g., angioedema) or in the absence of hypotension, electrolyte or renal imbalance. It is recommended to avoid the association of an ARNI with an ACEI and if previously administered ACEI, to ensure a 36 h washout period before the initiation of an ARNI, due to the high risk of angioedema [50]. This delay is not required when switching from ARB to ARNI. When up-titrating ARNI/ACEI/ARB (every 2 weeks or more), the monitoring of the potassium level, renal function and blood pressure is required. Lower loop diuretic doses may be necessary for the optimal titration of ARNI/ACEI/ARB and caution regarding the potassium concentration is required, as well as the dietary restriction of/supplementation with potassium, as the kaliuretic effect of loop diuretics might no longer be present [17].
If the patients have LVEF ≤ 35%, the guidelines recommend the use of MRAs to reduce mortality and hospitalization. MRAs (e.g., spironolactone or eplerenone) are added in patients with symptomatic chronic HFrEF, as a triple therapy (ACEI/ARB/ARNI + beta-blockers + MRA), in the absence of contra-indications. It is essential to achieve the targeted dose of other drugs before initiating the treatment with an aldosterone antagonist and to monitor the potassium levels and renal function under the treatment [17].
SGLT2 inhibitors can also be added, as part of the quadruple therapy (ACEI/ARB/ARNI + beta-blocker + MRA + SGLT2 inhibitor), in the absence of contra-indications. There is no need to achieve targeted doses of other drugs before adding SGLT2 inhibitors, although the loop diuretic dose might require adjustments based on the close monitoring of symptoms and weight [17].
Isosorbide dinitrate/Hydralazine could be prescribed especially for African American patients once the targeted dose of ACEI/ARB/ARNI + beta-blockers + MRA has been achieved [17].
The If channel inhibitor ivabradine is recommended in patients with symptomatic HFrEF or LVEF ≤ 35%, in sinus rhythm and heart rate ≥ 70 bpm, and in patients that have been hospitalized for HF in the last year, despite receiving beta-blockers at the maximum tolerated dose, ACEI and an MRA. The titration of the dose should be performed every 2 weeks in order to decrease the heart rate. In the case of patients ≥ 75 years old or in those with a history of conduction defects, the recommended initial dose is 2.5 mg twice daily, administered with meals [17].

4. Particularities of Patients

4.1. Age-Induced Changes

4.1.1. Cardiovascular Structure and Function

A reduction in the response after beta adrenergic stimulation was observed (due to impaired coupling of G-protein receptors to adenyl cyclase and a decrease in adenyl cyclase concentration), which damages the capacity of the aging heart to increase cAMP as a response to the stimulation of beta receptors [7,51]. Thus, age-related cardiovascular changes are associated with a reduction in chronotropic and inotropic responses, which decline with age (peak contractility and heart rate decline almost linearly with age) [52].
The filling of left ventricular diastole is impaired by the aging process, as it is a process that depends on energy and active myocardial relaxation. Altered calcium release by the cardiomyocytes, with resulting prolonged contractile period of the heart, was also observed in elderly people [7,53].
The high deposits of collagen, amyloid, and lipofuscin in the interstitial space and myocyte hypertrophy seen in older people increase cardiac stiffness and decrease cardiac compliance, altering cardiac filling, especially in critical situations [7,54,55,56].
The increased vascular (arterial) stiffness (due to collagen deposition and cross-linking in the vascular media and to fragmentation of arterial elastin), together with impaired endothelium-dependent vasorelaxation (a consequence of vascular inflammation and altered endothelial nitric oxide synthesis) observed in aging lead to a higher afterload and a predisposition to systolic hypertension in the elderly [57,58].
Inadequate mitochondrial synthesis of adenosine triphosphate in response to stress will lead to altered energy release, thus altered cellular reactions, such as gene expression, chromatin remodeling, intra/extra-cellular signaling, ion homeostasis, muscle contraction, protein and hormone synthesis and secretion, and neurotransmitter release and reuptake [59].

4.1.2. Other Organs

Age-associated modifications in the glomerular filtration rate and electrolyte imbalances [60,61] often seen in the elderly (due to dehydration, diuretic use, etc.) can raise the risk of HF decompensation and exacerbate the risk of drug side effects, with dangerous consequences, especially if the patient also has chronic kidney disease [62].
The aging of the respiratory system can lead to decreased compliance in pulmonary function. Moreover, the presence of chronic lung disease or sleep-related breathing disorders can increase the risk of pulmonary hypertension, exacerbate the sensation of dyspnea, and decrease biventricular filling [7,63,64].
The aging of the autonomic nervous system is characterized by sympathetic hyperreactivity and increased plasma concentrations of catecholamines, but a reduced sympathetic response is observed due to the diminished response of catecholamine receptors. Thus, tachycardia is felt less in elderly than middle-aged adults [65].

4.2. Sex Differences in HF

Regarding sex differences in heart failure, it seems that a large percentage of women tend to develop HRpEF, with the etiology of HF being either hypertension, diastolic dysfunction, or valvular pathology, whereas men tend to develop HFrEF or HFmrEF (HF with mid-range ejection fraction), with the etiology usually being an ischemic condition [66]. Moreover, it seems that women with HF are usually older and present with increased EF and more frequent symptoms linked to HF. Although they tend to also have multiple comorbidities compared to men, a meta-analysis showed that they have a better prognostic rate regarding hospitalization and mortality risk, regardless of EF [67].
The cardioprotection found in women seems to be due to the secretion of 17β-estradiol, an estrogen with a very clear established role in counteracting ischemic, hypertrophic, apoptotic, and cytotoxic impulses related to the heart [66,68,69].
Animal studies have shown that the cardiomyocytes of female models had a higher rate of survival after they were exposed to oxidative stress, which led to cell death, with the explanation relying on the fact that highly expressed estrogen receptor alpha (ER-α) can mediate the inhibition of pro-apoptotic pathways and the activation of the Akt signaling pathway [66,70].
Other differences regarding plasma B-type natriuretic peptide (BNP) levels, left ventricular mass index, left ventricular ejection fraction, and peak oxygen consumption between the sexes were also noted, suggesting that men are more susceptible to HF development than women [66,71,72].
Concerning treatment, it seems that although angiotensin-converting enzyme (ACE) inhibitors decrease the morbidity and mortality rates in both men and women, their effect seems to be more pronounced in men [73]. On the contrary, angiotensin II receptor blockers (ARBs) seem to have a higher mortality reduction rate in women than in men, although no difference was observed between the two classes of drugs (ACE inhibitors and ARBs) in terms of reduced mortality rates [74]. All of these aspects could be due to the action of estrogen on the receptor expression of angiotensin II by the ACE2 gene, located on chromosome X, and to the higher incidence of coughing and thus higher rate of discontinuation of ACE inhibitors in women [75].
No sex-related differences were observed in terms of treatment outcomes in patients under treatment with beta blockers or mineralocorticoid receptor antagonist [66].
Differences between the sexes were also noted in the case of digoxin treatment; women with a digoxin plasma concentration of 1.2–2.0 ng/mL had a higher mortality rate than men, although plasma levels of 0.5–0.9 ng/mL in men were associated with reduced mortality, but not any effect in women [76].

5. Particularities of Treatment

5.1. Pharmacokinetic Considerations and Their Consequences in HF Patients

Reduced blood flow to the gastrointestinal tract causes decreased absorption of drugs [77]. In the case of medicines with low permeability into the intestinal tissue, edema in the intestinal mucosa may affect their transport into the intestine [13,78].
Intestinal wall dysfunction secondary to hypoperfusion can, over time, induce chronic enteral inflammation and malnutrition. On the other hand, increased intestinal permeability in patients with HF can stimulate the transfer of drugs from the gastrointestinal tract to the portal blood [13].
Decreased blood perfusion in the central and peripheral organs results in an irregular tissue distribution of drugs [79]. Differences in the body’s water load can also affect the distribution of drugs [80,81].
Plasma protein binding may also be affected, especially after a myocardial infarction (production of α1-glycoproteins in the liver increases tissue necrosis and inflammatory reactions in the myocardium) or in patients with cachexia [82].
Reduced hepatic and renal blood flow induces an altered metabolism and elimination of administered drugs and their metabolites. In addition to an irregular distribution of drugs in the liver (a consequence of poor hepatic blood infusion), hepatic congestion and/or hypoxia (as a major consequence) and hepatocellular lesions may occur, manifested by hepatocytolysis (and thus increased liver transaminases) and disorders affecting enzymatic activity [83,84].
Since the concentration of active substances at the site of action cannot yet be directly determined, plasma concentration is often measured as a surrogate marker of the drug effect, depending on the concentration of active substances at the site of action [80].
Practically, changes in pharmacokinetics have been only observed in patients with renal and/or hepatic complications [13,84,85].
Increased action of the following drugs was observed after oral administration in patients with decompensated HF: captopril, enalapril, perindopril, carvedilol, felodipine, candesartan, furosemide, milrinone, and enoximone [13,84,85].
Since most studies to date (clinical trials) have not included patients with decompensated HF or major renal or hepatic problems (which involve more severe changes in PK and PD), the pharmacokinetic and pharmacodynamic parameters are currently under-studied in patients with HF. Thus, we recommend paying more attention to monitoring the efficacy and safety of drugs used in HF. Furthermore, the progressive titration of drugs should be implemented and the benefit/risk ratio should be periodically evaluated [13,84,85].

5.2. Pharmadynamic Considerations

The pharmacodynamics of drugs, as well as their tolerability, may also be affected by several neuronal and endocrinological compensatory mechanisms in HF, including the activation of the renin-angiotensin (RAA) and sympathetic system. Moreover, nodal activity and baroreceptor sensitivity are affected, and peripheral vascular resistance is increased; these are aspects that could cause an altered response to administered drugs [13,84,85].
The activation of the sympathetic nervous system can alter the perfusion of the viscera, especially the splenic organs (liver, gastrointestinal tract, kidneys), to maintain the perfusion of vital organs (brain and heart), resulting in hypoperfusion in the liver and kidneys. Furthermore, increased central pressure in patients with right HF causes hepatic congestion and dilation of the central vein in the hepatic acini, inducing hepatocellular ischemia and necrosis, and reducing the activity of microsomal enzymes [13,84,85,86].
Therefore, it is advisable to consider all changes that might occur in heart failure patients (Figure 3) [13,84,85,86].

5.3. HF Treatment in Patients with Cardiorenal Syndrome

It is well known that the acute/chronic dysfunction of one organ could induce the acute/chronic dysfunction of the other organ [87], therefore, cardiorenal syndrome has been defined as a spectrum of diseases involving the heart and the kidneys. This syndrome implies a “hemodynamic cross-talk” between the injured heart and the kidneys’ responses and vice versa [87,88]. Several mechanisms underline this cardiorenal syndrome, such as the hemodynamic interactions between the heart and kidneys in HF patients; cytokine production; the impact of atherosclerotic disease on both organs; biochemical perturbations due to the installation of chronic kidney disease; and the structural changes that appear in the heart, which are due to kidney disease progression [87,89].
In summary, the drop in cardiac output induces the activation of the sympathetic nervous system which will increase the stroke volume and the heart rate, as a compensatory mechanism. Sympathetic nervous system activation will also stimulate the release of renin from the kidneys, with the consequence of RAAS activation. Moreover, the drop in cardiac output will also induce a decreased perfusion of the kidneys, leading to kidney injuries (the beginning of cardiorenal syndrome). A reduced perfusion in the kidneys will stimulate renin release, RAAS activation and thus sodium and water retention (due to aldosterone secretion and antidiuretic hormone release), which will later increase the mean arterial pressure and the preload and decrease the cardiac output. RAAS activation will also cause vasoconstriction, which will contribute to reduced renal perfusion [87,89,90].
Moreover, chronic kidney disease (CKD) can lead to cardiovascular dysfunction, as a low glomerular filtration rate activates RAAS, which will lead, in time, to cardiac remodeling and left ventricular hypertrophy. CKD also implies a reduction in erythropoietin production over time, leading to anemia, which will increase the risk of ischemic events in the heart. Moreover, CKD induces a decrease in vitamin D production and parathormone stimulation, leading to an increase in calcium and phosphate levels and thus, increased risk of coronary and vessel calcification, augmenting the high risk of ischemic events [91]. Electrolyte imbalances are also observed in CKD patients, more precisely, hyperkalemia, which can increase the risk of cardiovascular complications [87].
Therefore, the management of cardiorenal syndrome is challenging and must be directed towards the specific pathophysiologic mechanism involved. The volume overload can be either addressed by prescribing diuretics (usually loop diuretics, as they are the most potent diuretics e.g., furosemide, torsemide and bumetanide) or using ultrafiltration methods. The addition of a thiazide diuretic to a loop diuretic may be preferred in the case of diuretic resistance, as an initial approach to restore euvolemia [87,89]. Regarding HF treatment in patients with renal disease, the renal function and potassium level should be checked within 1–2 weeks of the initiation or up-titration of an ACEI/ARB/ARN. Regarding aldosterone antagonists (MRA), in patients with preserved renal function or mild to moderate impairment, potassium levels and renal function should be checked within 2–3 days after the initiation of the therapy, followed by a check after 7 days of treatment, and at least monthly for the first 3 months and then, every 3 months [17].
In patients with severe renal impairment (eGFR < 30 mL/min/1.73 m2), the ARBs/ACEIs are considered safe. The starting dose of ARNI (Sacubitril/Valsartan) should be reduced to 24/26 mg, twice a day, in patients with severe renal impairment. The dose of ARNI might also need to be reduced in the case of hypotension or hyperkalemia. MRAs are contraindicated in patients with severe renal impairment, creatinine > 2.5 mg/dL in men, creatinine > 2 mg/dL in women or potassium > 5.0 mEq/L. As for SGLT2 inhibitors, there is currently no evidence regarding dose adjustments in patients with eGFR < 30 mL/min/1.73 m2 for dapagliflozin and eGFR < 20 mL/min/1.73 m2 for empagliflozin [17]. As a general rule, a decrease in eGFR of more than 30% or the apparition of hyperkalemia should alert the clinician to adjust (decrease) the doses of HF drugs [17].

5.4. HF Treatment in Pregnancy and Lactation

During pregnancy, the increased physiological requirements are partially fulfilled through changes in the physiology of the cardiovascular system, which has to adapt to the extra metabolic demands of the fetus and of the other organ systems. Therefore, the augmentation of the size and activity of the uterus, as well as the increase in blood flow in the choriodecidual space, represents extra work for the cardiovascular system. Moreover, during pregnancy, the skin and kidneys have an increased perfusion which allows them to disperse heat and retain sodium and water [92,93].
Symptoms of HF are more likely to appear in the second trimester as a consequence of an increased cardiac output and of intravascular volume (during pregnancy, the plasma volume increases by 40% and the cardiac output by 30–50%) [94]. Therefore, the therapeutical management of HF during pregnancy will be adapted to the clinical setting and the severity of the pathology. For cases in which the oral administration of drugs is sufficient, diuretics, betablockers, hydralazine or nitrates can be recommended. Usually, diuretics represent the first line treatment for pregnant HF women due to the increased preload associated with pregnancy (therefore, reducing preload will diminish the left side filling pressure and the pulmonary capillary pressure, and thus, it will allow the resorption of the pulmonary interstitial fluid). Currently, there is no evidence that diuretics are directly responsible for fetal growth restrictions. Betablockers decrease the heart rate and allow a greater filling during diastole. Beta-1-selective blockers (for example metoprolol succinate or bisoprolol) are preferred and better tolerated [1,92].
Regarding the management of HF before pregnancy, ACEIs, ARBs, ARNI, MRA and SGLT2 inhibitors, as well as ivabradine should all be avoided and stopped prior to conception due to an increased risk of fetal harm. It is recommended that the pregnancy be planned and closely monitored by a multidisciplinary team of specialists in order to avoid HF decompensation and fetal harm (induced by either the pathology or the treatment) [1].
Hydralazine, methyldopa, or oral nitrates can also be recommended during pregnancy [1].
In patients with atrial fibrillation, low-molecular-weight heparins are the first choice of anticoagulant treatment (NOAC should be avoided due to insufficient data regarding their safety) [1].
Concerning the breastfeeding period in HF women, it is important to know that the mother’s treatment prevail over breastfeeding compatibility, and that the benefits of breastfeeding are important for both the mother and child [95,96]. Enalapril is among the preferred options of ACEIs (as it has the most assuring safety data) and can be used from birth. [95]. As a second option, among the ARBs, losartan can be a good choice, due to its extensive first-pass metabolism and thus low systemic concentration, but breastfeeding should be performed with caution [95]. From the betablockers, metoprolol succinate or propranolol are the preferred choices (favorable PK profile and assuring data). Additionally, carvedilol or bisoprolol can be seen as a second option of treatment. Sacubitril/valsartan association should be avoided due to lack of data regarding their use during pregnancy, as well as SGLT2 inhibitors. Moreover, there is good evidence for digoxin, hydralazine and spironolactone use during breastfeeding period. The monitorization of babies exposed to either betablockers or ACEIs is recommended for hypotension (especially in neonates), lethargy, drowsiness, bradycardia, poor feeding, or weight gain [95].

6. Drugs and Food Supplements That Can Aggravate HF

The treatment of HF patients is very complex and includes not only lifestyle changes but also multiple pharmacological therapies, as well as the presence of co-morbidities and individual pharmacological strategies; this leads to polypharmacy in HF patients, generating increased iatrogenic risks [97,98].
In Appendix A we summarize the main/most used drugs and food supplements that can worsen the prognosis of heart failure patients; thus, it is recommended to avoid them by this category of patients [99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170].

7. Potential Drug–Drug Interactions in HF Patients

Several studies have shown a strong association between the number of drugs taken by HF patients (usually more than five) and the occurrence of potential drug–drug interactions, leading to the conclusion that the incidence of drug–drug interactions in HF patients is extremely high [97,171,172].
The coordinated efforts of a multidisciplinary team of healthcare providers also involving a clinical pharmacist could reduce the medication-related problems and improve the efficacy, tolerability, and safety of the pharmacological strategies implemented by physicians [171,173,174].
Appendix B summarizes the main important drug–drug interactions that should be considered in HF patients, their consequences, and some recommendations regarding their management [174,175,176,177,178,179,180,181,182,183,184,185].

8. Adverse Drug Reactions in HF Patients

Adverse drug reactions (ADRs) have been estimated to account for approximatively 10–20% of hospital admissions in geriatric units [186]. Moreover, an observational study performed in 1996 highlighted that the iatrogenic problems accounted for nearly 7% of HF admissions and were associated with higher mortality and prolonged hospital stays compared with those of non-iatrogenic causes [187,188]. Thus, the decompensation of HF patients due to iatrogenic conditions is a well-known and documented problem, which leads to increased morbidity and mortality rates. Therefore, good management of all prescribed drugs is mandatory for HF patients.
It seems that there are also sex-related differences between men and women regarding ADRs. Although women are underrepresented in all phases of clinical trials and little is known about this aspect, several meta-analyses concluded that women are more susceptible (1.5–1.7×) to developing ADRs than men and are also at higher risk of hospitalization due to the severity of ADRs [189,190].
As women usually present HFpEF with additional risk factors (co-morbidities and advanced age) compared with other types of HF, there seems to be a high incidence of polypharmacy, as they tend to take more drugs than men (including over-the-counter drugs and food supplements); thus, they have an increased risk of iatrogenic events (due to ADRs and drug interactions) and low adherence to treatment. Other explanations may underline this problem of high iatrogenic risk. Sex differences in the pharmacokinetics and pharmacodynamics of administered drugs (regarding distribution volume, hepatic/renal clearance, sex hormones, alterations in drug target expression and signal transduction pathways, immunological conditions, etc.) predispose women to a higher probability of overdosing than men [190,191,192,193,194]. Drug-induced ventricular arrhythmia (torsade de pointes) is more often encountered in women, as women have longer QTc intervals, probably due to the sex hormone modulation of Ca2+ and K+ channels implicated in ventricular repolarization [190,195]. Differences in prescribing habits for men and women compared with the recommended guidelines is another reason supporting the high incidence of ADRs in women, as well as the overall poor quality of life observed in women HF patients [189,190].
All of the aforementioned sex-related differences in female patients predispose women to a higher probability of drug-induced complications such as bleeding problems (e.g., under antithrombotics), electrolyte abnormalities (e.g., under diuretics), cough and increased creatinine (under treatment with ACE inhibitors), myopathy (under statin treatment), hepatotoxicity, skin diseases, etc. [190,196,197].
Thus, it is important to adjust the drug dosage as a function of total body weight/size or glomerular filtration rate and titrate it to the required clinical effect, especially in those with a narrow therapeutic index, in order to avoid the incidence of ADRs [189,190].

9. Discontinuation of Drugs in HF Patients

Several articles also discuss the negative outcomes of HF patients after discontinuing chronic HF treatment [2,187,198,199,200].
It was observed that RAAS (renin-angiotensin-aldosterone system) inhibitors provide the most beneficial outcomes in terms of mortality reduction in patients with HFrEF, although renal function is affected at baseline [201]. The cessation of these drugs in patients with HFrEF was associated with increased mortality and re-hospitalization admissions after 1 month, 3 months, and 1 year, which led to the conclusion that RAAS inhibitors should not be discontinued in patients with moderate to several renal dysfunction if the benefits outweigh the risks [202].
Regarding beta blocker discontinuation, although they are associated with a risk of negative inotropic effects and hypotension, ESC has recommended not to disrupt beta blocker treatment unless severe hypotension is present, due to the risk of rebound effects (such as rebound tachycardia, aggravation of angina pectoris, risk of ventricular arrhythmia) and the correlation with increased mortality and readmissions rates after cessation of treatment [2,199,203]. Several trials highlighted that continuous administration of beta blockers in patients with decompensated HF reduced the mortality and readmission rates [203,204,205].

10. Conclusions

In order to reduce exacerbations, hospital readmission rates, morbidity, and mortality and to improve the overall quality of life, an interdisciplinary approach to treatment strategies is mandatory for HF patients. The treatment strategy must be individualized for each HF patient, periodically monitored and reviewed by the healthcare team. Moreover, patient education, including topics such as dietary counseling, healthy lifestyle habits, regular exercise in a tolerable amount, alcohol and smoking cessation; moreover, understanding the alarming signs and symptoms of HF decompensation (shortness of breath, fatigue, ankle swelling, sudden weight modification) is another extremely important action that needs to be urgently implemented by societies with aging populations.

Author Contributions

Conceptualization, V.B., A.P., D.C., S.N. and L.P.; methodology, V.B., A.P., C.R. and C.D.; resources, D.E.M., M.S., M.A., C.D. and S.C.; data curation, L.P., S.C. and S.N.; writing—original draft preparation, V.B., A.P., A.S., D.E.M., D.C. and C.R.; writing—review and editing, M.A., M.S., L.P. and C.A.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Drugs and food supplements that can worsen HF prognosis.
Table A1. Drugs and food supplements that can worsen HF prognosis.
Drugs [99]Possible Mechanism InvolvedResultsReferences
NSAIDsInhibition of cyclooxygenase enzyme
Inhibition of renal prostaglandin synthesis
Sodium and water retention
Higher systemic vascular resistance
Reduction in
renal perfusion, glomerular filtration rate, sodium excretion
[100,101,102,103,104,105,106]
Alpha-1 blockers (e.g., doxazosin)Beta-1 receptor stimulation
Stimulation of renin and aldosterone release
Chronic alfa1 antagonism
Stimulation of heart fibrosis factor galectin-3 expression
Edema
Tachyphylaxis
Cardiomyocyte apoptosis
Myocardial hypertrophy
[107]
Calcium channel blockers (e.g., verapamil, diltiazem)Negative inotrope
Calcium channel blockade
Cardiac depression
Atrioventricular conduction block
[103,106]
Moxonidine (centrally acting α-adrenergic drug)Possible sympathetic withdrawalMyocardial depression
Hypotension
Rebound norepinephrine increase
[108]
Class I antiarrhythmic (e.g., flecainide, disopyramide)Negative inotrope
Pro-arrhythmic stimulation
Myocardial infarction
Premature ventricular beats
Myocardial depressant effects
[106,109]
Class III antiarrhythmic (e.g., sotalol)Beta inhibition
Pro-arrhythmic stimulation
Potassium channel blockade
Bradycardia
Prolonged QT interval
Torsades de pointes
T-wave abnormalities
[109,110,111]
Inhibitors of dipeptidyl peptidase 4 (e.g., sitagliptin, saxagliptin)Dipeptidyl peptidase 4 enzyme interference
Direct interaction in myocytes
Calcium channel interference
Interference in substance P degradation
Sympathetic nervous
system stimulation
Myocardial infarction
Stroke
[103,112,113,114,115]
Thiazolidinediones (e.g., rosiglitazone, pioglitazone)Possible calcium channel blockade
Interference with mitochondrial respiration or oxidative stress
Sodium and water retention
Peripheral edema
Myocardial infarction
Stroke Transient ischemic attacks
[106,115,116,117]
ItraconazoleNegative inotropic effect
Mitochondrial dysfunction
Inhibition of 11 beta-hydroxysteroid dehydrogenase 2
Cytochrome P450 inhibition
Peripheral edema
Hypertension
Prolonged QT interval
Cardiac depression
Excess mineralocorticoid
Myofibroblast damage
[118,119,120]
Amphotericin BUnknownCardiotoxicity
Dilated cardiomyopathy
[121]
Carbamazepine(overdose)Negative inotropic and chronotropic effects
Depression of phase 2 repolarization
Direct toxic effect on myocardial fibers
Anticholinergic action
Increased automaticity of ectopic pacemakers
Sodium channel blockade
Left ventricular dysfunction
Suppressed sinus nodal activity
Atrioventricular conduction disturbances
Hypotension
[122,123,124]
PregabalinAlterations in cardiac renin angiotensin system (RAS)
L-type calcium channel blockade
Peripheral edema
Decreased calcium influx in cardiomyocytes
Left ventricular deterioration
[125,126,127]
Tricyclic antidepressantsNegative inotrope
Pro-arrhythmic stimulation
Norepinephrine and serotonin reuptake blockade
Sodium channel blockade
Suppression of potassium channels in myocytes
Vasoconstriction of cerebral arteries
Arrhythmias
Impaired heart conduction
Prolonged intraventricular conduction
Prolonged QT interval Hemorrhagic stroke
Ischemic stroke
[128,129]
CitalopramInhibition of depolarizing current mediated by L-type calcium channels
Antagonistic effects on myocardial potassium channels
Prolonged QT interval
Episodes of torsades de pointes
Arrhythmias
[130,131]
Pergolide, cabergoline, pramipexolePotent agonists at cardiac myocyte 5-
HT2B
serotonin receptors
Induction of fibroblast activation
Valvular damage
Cardiac valvular regurgitation
Pulmonary arterial hypertension
Peripheral edema
[132,133,134]
ClozapineCalcium channel blockade
Ig-E mediated hypersensitivity
Reduced left ventricular function
Myocarditis
Cardiomyopathy
Prolonged QT interval
Elevated troponin
[135,136,137,138]
LithiumAltered acetylcholinesterase activity
Direct myofibril degeneration
Induction of oxidative stress
Interference with calcium ion influx
Cardiac fibrosis
Cardiomyocyte apoptosis
Rhythm disturbances
Edema, ascites
Complete heart block and first-degree AV block
[139,140,141,142,143]
β2 adrenergic agonists (e.g., salbutamol)Decreased β-receptor responsiveness
Small positive inotropic and chronotropic effects
Activation of Gs/cAMP/PKA
Inhibition of Gi/PDE
Arrhythmias
Prolonged QT interval
[144,145]
Tumor necrosis factor-α (TNF-α) inhibitorsCytokine mediation
Sympathetic excitation
Inflammation and renin-angiotensin system upregulation
Peripheral inflammation
Cardiac dysfunction
[146,147,148]
Topical beta-blockers (e.g., timolol)Hemodynamic effects due to beta blockadeArrhythmias
Myocardial ischemia
Hypotension
Pulmonary edema
[99]
Food supplements [149]Possible mechanism involvedResultsReference
Aconitum spp. (Monkshood)Alkaloids block potassium channelsVentricular fibrillation
Bradycardia
Hypotension
[150]
Aesculus hippocastanum L.
(Horse chestnut)
Antiplatelet effectIncreased risk of bleeding when associated with anticoagulant drugs[151]
Allium sativum L. (Garlic)Inhibition of platelet aggregation (dose-dependent)Increased risk of bleeding when associated with anti-thrombotic drugs[152]
Aloe barbadensis Mill.
(Aloe vera)
Laxative effectRisk of hypokalemia with increased toxicity of cardiotonic glycosides or antiarrhythmia drugs[153]
Angelica sinensis (Oliv.) Diels
(Angelica)
Antiplatelet and anticoagulant effectIncreased anticoagulant effect[154]
Cassia senna L.
(Senna)
Laxative effectRisk of hypokalemia with increased toxicity of digitalis or antiarrhythmia drugs[153]
Citrus paradisi Macfad.
(Grapefruit)
Inhibition of CYP3A4 enzymeIncreased effects (therapeutic or toxic) of co-administered drugs (e.g., calcium channel blockers, antiarrhythmia drugs)
Inefficacy of pro-drugs metabolized by CYP3A4
[155,156]
Cratageus spp.
(Hawthorn)
Increases digitalis toxicity
(incompletely elucidated)
Risk of digitalis intoxication if co-administered[157,158]
Ephedra sinica Stapf
(Chinese ephera)
Alkaloids stimulate adrenergic receptors
Indirect agonist stimulation and noradrenaline release
Tachycardia Hypertension Arrythmias
Heart attack
Stroke
[159]
Ginkgo biloba L.
(Ginkgo)
Antiplatelet effectIncreased risk of bleeding when co-administered with antithrombotic drugs[160,161]
Glycyrrhiza glabra L.
(Licorice)
Hypokalemia
Reduced sodium and water excretion
Increased toxicity of digitalis or antiarrhythmic drugs
Decreased effect of diuretics
[153]
Harpagophytum procumbens Burch.
(Devil’s claw)
Inhibition of CYP1A2 and CYP2D6Increased effects of diuretics, antihypertensives, statins, and anticoagulants[162,163]
Hypericum perforatum L.
(St. John’s Wort)
Induction of CYP3A4 isoenzyme activityDecreases plasma levels of co-administered drugs metabolized by this enzyme[164,165,166]
Leonurus cardiaca L.
(Motherwort)
Antiplatelet effectIncreased risk of bleeding when co-administered with antithrombotic drugs[152]
Oenothera biennis L.
(Evening primrose)
Inhibition of platelet activating factorIncreased risk of bleeding when co-administered with antithrombotic drugs[167,168]
Panax ginseng C.A. Meyer
(Asian ginseng)
Decreased prothrombin timeDecreased warfarin effect and increased risk of thrombo-embolic events[152]
Stephania tetrandra S. MooreCalcium channel blockadeCardiac depression[169]
Zingiber officinale Roscoe
(Ginger)
Thromboxane synthase inhibition
Prostacyclin agonist
Increased risk of bleeding when co-administered with antithrombotic drugs
Increased effects of antihypertensive drugs
[170]

Appendix B

Table A2. Drug-drug interactions in HF.
Table A2. Drug-drug interactions in HF.
Main Drug for HFCo-Administered DrugsConsequencesRecommendations
ACE inhibitorsARBs/aliskiren
(angiotensin II receptor blockers)
Increased risk of impaired renal function, acute renal failure, hyperkalemia, hypotension, syncope and falls, thus increased risk of fractures in the elderlyAvoid association
SacubitrilHigh risk of angioedemaAvoid association
NSAIDs
(nonsteroidal anti-inflammatory drugs)
Risk of acute renal failure due to decreased glomerular filtration rate (decreased synthesis of renal vasodilating prostaglandins), especially if patient is elderly, dehydrated, or under diuretic treatmentIf possible, avoid association
If association is needed, proper hydration is recommended, monitoring of renal function, administering the lowest therapeutic NSAID dose and for the shortest period of time
Spironolactone, amiloride, triamtereneHigh risk of hyperkalemia, especially in patients with chronic renal failureEvaluate renal function before beginning of treatment (determine creatinine clearance), administer in therapeutically effective minimum doses and periodically check potassium
AllopurinolHigher risk of hypersensitivity reactions (Steven-Johnson syndrome)If associated, ensure clinical supervision and adjust dose [177]
GliptinsIncreased risk of angioedema through decreased DPPIV by gliptinAvoid association
If associated, ensure clinical supervision and adjust dose
InsulinHigh risk of hypoglycemiaMonitor blood glucose and adjust insulin dosage
Hypoglycemic sulfonamidesHypoglycemic risk through improved glucose tolerance and decreased hypoglycemic sulfonamide dose requirementsMonitor blood glucose and adjust dosage of hypoglycemic sulfonamides
RacecadotrilHigh risk of allergic side effects (angioneurotic edema)Avoid association
If associated, ensure clinical supervision and adjust dose
LithiumIncreased lithium plasma concentration through decreased eliminationAvoiding association
If associated, ensure clinical supervision and adjust lithium dose
ARBsACE inhibitors
NSAIDs
Spironolactone
Lithium
Same as for ACE inhibitors
Sacubitril/valsartanStatinsIncreased effects of statinsAdjust statin dose [178]
SildenafilAdditional blood pressure reductionUse caution when associated and adjust dose of sildenafil [179]
Beta
blockers
(carvedilol, bisoprolol, metoprolol, nebivolol)
AmiodaroneCardiac conduction disorders, bradycardia, atrioventricular blockPreferably avoid association, or adapt drug dosages and conduct patient monitoring (ECG, heart rate)
Verapamil
Diltiazem
Cardiac depression, HF decompensation, AV blockPreferably avoid association
Antidiabetic drugsRisk of masking signs of hypoglycemia (palpitations, tachycardia, tremor of extremities)Preferably avoid association or closely monitor dosage of antidiabetic drugs
DigitalisAutomatic disorders (bradycardia, sinus arrest), AV blockPreferably avoid association or adjust dosages
NSAIDsDecreased antihypertensive effect due to inhibition of renal vasodilating prostaglandin synthesis by NSAIDsPreferably avoid association or adjust dosages
MexiletineNegative inotropic effect
Automation disorders
Risk of cardiac decompensation
Preferably avoid association
Central antihypertensivesDecreased central sympathetic tone and vasodilating effect of central blood-lowering drugsPreferably avoid association
Imipramine antidepressants (e.g., amitriptyline)Intensification of vasodilating effect and risk of orthostatic hypotensionAvoid association or adapt beta blocker dosage
NeurolepticsVasodilator effect
Risk of orthostatic hypotension
Monitor blood pressure and adapt dosages if needed
AnticholinesterasesExcessive bradycardiaAvoid association or monitor heart rate with adjustment of beta blocker dosage
DiureticsNSAIDsDecreased diuretic effect and risk of kidney failureAvoid association if possible
CarbamazepineIncreased risk of hyponatremiaHydrate patient and correct electrolyte imbalances
LithiumDecreased renal elimination of lithium with high risk of accumulationAvoid association if possible or adapt lithium dosage
SGLT2 inhibitors
Dapagliflozin Empagliflozin
Thiazide diuretics/loop diureticsIncreased diuretic effectAdjust dosage
Monitor the blood pressure.
Hydrate patients and monitor the electrolyte balance
NitratesSildenafilIncreased risk of hypotension, blood pressure collapseAvoid association or adjust dosage
HeparinsIncreased excretion of heparinsAdjust dosage
DigoxinAmiodarone
Propafenone
Quinidine
Clarithromycin
Hypokalemic diuretics
Digoxin toxicityAvoid association or adjust dosage
Carbamazepine
Dronedarone
Decreased plasma concentration of digoxin
Cardiac deprivation
Increased digoxinemia
Therapeutic supervision
Therapeutic supervision
(clinical and ECG)
Reduce digoxin dosage by half
AmiodaroneVerapamil/
Diltiazem
Cardiac deprivation with high risk of bradycardia and atrioventricular blockAvoid intravenous administration,
use ECG surveillance when administered orally
Levofloxacin/
moxifloxacin
Ventricular rhythm disorders
(risk of torsades des pointes)
Therapeutic supervision
(clinical and ECG)
StatinsIncreased effects of statinsAdjust statin dose (maximum 20 mg/day for simvastatin)
IvabradineVerapamil/
diltiazem
Increased ivabradine plasma concentration with increased risk of side effects
Marked bradycardia
Avoid association
AzithromycinVentricular rhythm disorders
(risk of torsades des pointes)
Therapeutic supervision
(clinical and ECG)
AVKAmiodaroneIncreased AVK effects
Hemorrhagic risk
INR (International Normalized Ratio) control
Adjust dosage (up to 4 weeks after stopping amiodarone treatment)
AllopurinolIncreased hemorrhagic riskINR surveillance and adjust AVK dosage up to 8 days after stopping allopurinol treatment [180]
Cefamandole/
cefazolin/
ceftriaxone
Increased AVK plasma concentration with high hemorrhagic riskINR surveillance and adjust AVK dosage
FluoroquinolonesIncreased AVK plasma concentration with high hemorrhagic riskINR surveillance and adjust AVK dosage
FenofibrateIncreased AVK plasma concentration with high hemorrhagic riskINR surveillance and adjust AVK dosage
ParacetamolIncreased AVK plasma concentration with high hemorrhagic risk when given paracetamol in high dosage (>4 g/day), >4 daysINR surveillance and adjust AVK and paracetamol dosage
Thiamazole
(methimazole)
Increased risk of bleeding due to hypoprothrombinemia caused by methimazoleIf possible, avoid association or conduct INR surveillance and adjust AVK dosage [181]
NSAIDsIncreased AVK plasmatic concentration with high hemorrhagic riskIf possible, avoid association or conduct INR surveillance and adjust AVK dosage
NOAC
(New Oral Anticoagulants)
RifampicinDecreased NOAC efficacy and increased thromboembolic riskClinical supervision
Adjust NOAC dose up to 8 days after stopping rifampicin treatment [180]
Itraconazole/
ketoconazole/
voriconazole
Increased NOAC plasma concentration and efficacy with high risk of bleedingClinical surveillance and adjust dose of NOAC
Carbamazepine/
levetiracetam/
phenobarbital/
valproic acid
Decreased NOAC efficacy and increased thromboembolic riskClinical supervision and adjust NOAC dose
DabigatranAmiodaroneHigh plasma concentration of dabigatran and increased risk of bleedingClinical supervision and adjust dabigatran dose (maximum 150 mg/day) [182]
DronedaroneHigh plasma concentration of dabigatran (also rivaroxaban) with increased risk of bleedingClinical supervision and adjust dabigatran/rivaroxaban dose
QuinidineHigh plasma concentration of dabigatran with increased risk of bleedingAvoid association
If associated, clinical supervision and adjust dabigatran dose
Fluconazole/
itraconazole/
ketoconazole
High plasma concentration of dabigatran with increased risk of bleedingAvoid association
If associated, clinical supervision and adjust dabigatran dose
ApixabanDiltiazemIncreased plasma concentration of apixaban with increased risk of bleeding [182,183]Clinical supervision and adjust apixaban dose
Clarithromycin/
Erythromycin
High plasma concentration of apixaban/rivaroxaban with increased risk of bleedingClinical supervision and adjust apixaban/rivaroxaban dose
FluconazoleHigh plasma concentration of apixaban/rivaroxaban with increased risk of bleedingAvoid association
If associated, clinical supervision and adjust apixaban dose
Antiplatelet agentsNSAIDsIncreased risk of bleeding (especially gastro-intestinal)Avoid association
If associated, clinical supervision and adjust dose
Heparins/
oral anticoagulants
Increased risk of bleedingAvoid association
If associated, clinical supervision and adjust dose
Selective serotonin reuptake inhibitors (SSRIs)Increased risk of bleedingAvoid association
If associated, clinical supervision and adjust dose
Antidepressants with mixed adrenergic–serotoninergic mechanismIncreased risk of bleedingAvoid association
If associated, clinical supervision and adjust dose
PentoxifyllineIncreased risk of bleedingClinical supervision and dose adjustments
ClopidogrelProton pump inhibitors (PPIs)High thromboembolic riskAvoid association [184]
RepaglinideIncreased plasma concentration of oral antidiabetic with intensified side effectsAdjust repaglinide dose
TicagrelorDabigatranHigh plasma concentration of dabigatran and increased risk of bleedingAvoid association
If associated, clinical supervision and adjust dabigatran dose
Diltiazem/
verapamil
High plasma concentration of ticagrelor and increased risk of bleedingAvoid association
If associated, clinical supervision and adjust ticagrelor dose
AtorvastatinIncreased plasma concentration of statinAdjust statin dosage (maximum 40 mg/day) [178,185]

References

  1. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef] [PubMed]
  2. Ponikowski, P.; Voors, A.A.; Anker, S.D.; Bueno, H.; Cleland, J.G.; Coats, A.J.; Falk, V.; González-Juanatey, J.R.; Harjola, V.P.; Jankowska, E.A.; et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur. J. Heart Fail. 2016, 18, 891–975. [Google Scholar] [CrossRef]
  3. Bozkurt, B.; Coats, A.J.S.; Tsutsui, H.; Abdelhamid, C.M.; Adamopoulos, S.; Albert, N.; Anker, S.D.; Atherton, J.; Böhm, M.; Butler, J.; et al. Universal definition and classification of heart failure: A report of the Heart Failure Society of America, Heart Failure Association of the European Society of Cardiology, Japanese Heart Failure Society and Writing Committee of the Universal Definition of Heart Failure: Endorsed by the Canadian Heart Failure Society, Heart Failure Association of India, Cardiac Society of Australia and New Zealand, and Chinese Heart Failure Association. Eur. J. Heart Fail. 2021, 23, 352–380. [Google Scholar] [CrossRef]
  4. Andronic, A.A.; Mihaila, S.; Cinteza, M. Heart Failure with Mid-Range Ejection Fraction—A New Category of Heart Failure or Still a Gray Zone. Maedica 2016, 11, 320–324. [Google Scholar] [PubMed]
  5. Delepaul, B.; Robin, G.; Delmas, C.; Moine, T.; Blanc, A.; Fournier, P.; Roger-Rollé, A.; Domain, G.; Delon, C.; Uzan, C.; et al. Who are patients classified within the new terminology of heart failure from the 2016 ESC guidelines? ESC Heart Fail. 2017, 4, 99–104. [Google Scholar] [CrossRef]
  6. Mosterd, A.; Hoes, A.W. Clinical epidemiology of heart failure. Heart 2007, 93, 1137–1146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Dharmarajan, K.; Rich, M.W. Epidemiology, Pathophysiology, and Prognosis of Heart Failure in Older Adults. Heart Fail. Clin. 2017, 13, 417–426. [Google Scholar] [CrossRef] [Green Version]
  8. Snipelisky, D.; Chaudhry, S.P.; Stewart, G.C. The Many Faces of Heart Failure. Card. Electrophysiol. Clin. 2019, 11, 11–20. [Google Scholar] [CrossRef]
  9. Crooks, J.; O’Malley, K.; Stevenson, I.H. Pharmacokinetics in the elderly. Clin. Pharmacokinet. 1976, 1, 280–296. [Google Scholar] [CrossRef] [PubMed]
  10. Turnheim, K. When drug therapy gets old: Pharmacokinetics and pharmacodynamics in the elderly. Exp. Gerontol. 2003, 38, 843–853. [Google Scholar] [CrossRef] [PubMed]
  11. Salwe, K.J.; Kalyansundaram, D.; Bahurupi, Y. A Study on Polypharmacy and Potential Drug-Drug Interactions among Elderly Patients Admitted in Department of Medicine of a Tertiary Care Hospital in Puducherry. J. Clin. Diagn. Res. 2016, 10, FC06. [Google Scholar] [CrossRef] [PubMed]
  12. Mastromarino, V.; Casenghi, M.; Testa, M.; Gabriele, E.; Coluccia, R.; Rubattu, S.; Volpe, M. Polypharmacy in heart failure patients. Curr. Heart Fail. Rep. 2014, 11, 212–219. [Google Scholar] [CrossRef] [PubMed]
  13. Lainscak, M.; Vitale, C.; Seferovic, P.; Spoletini, I.; Cvan Trobec, K.; Rosano, G.M. Pharmacokinetics and pharmacodynamics of cardiovascular drugs in chronic heart failure. Int. J. Cardiol. 2016, 224, 191–198. [Google Scholar] [CrossRef] [PubMed]
  14. Lainscak, M.; Vitale, C. Biological and chronological age in heart failure: Role of immunosenescence. J. Cardiovasc. Med. 2016, 17, 857–859. [Google Scholar] [CrossRef]
  15. United Nations. Population Ageing. 2015. Available online: WPA2015_Report.pdf(un.org) (accessed on 25 January 2022).
  16. Yancy, C.W.; Jessup, M.; Bozkurt, B.; Butler, J.; Casey, D.E., Jr.; Drazner, M.H.; Fonarow, G.C.; Geraci, S.A.; Horwich, T.; Januzzi, J.L.; et al. 2013 ACCF/AHA guideline for the management of heart failure: A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J. Am. Coll. Cardiol. 2013, 62, e147–e239. [Google Scholar] [CrossRef] [Green Version]
  17. Writing Committee; Maddox, T.M.; Januzzi, J.L., Jr.; Allen, L.A.; Breathett, K.; Butler, J.; Davis, L.L.; Fonarow, G.C.; Ibrahim, N.E.; Lindenfeld, J.; et al. 2021 Update to the 2017 ACC Expert Consensus Decision Pathway for Optimization of Heart Failure Treatment: Answers to 10 Pivotal Issues About Heart Failure with Reduced Ejection Fraction: A Report of the American College of Cardiology Solution Set Oversight Committee. J. Am. Coll. Cardiol. 2021, 77, 772–810. [Google Scholar] [CrossRef]
  18. Oliver, E.; Mayor, F., Jr.; D’Ocon, P. Beta-blockers: Historical Perspective and Mechanisms of Action. Rev. Española Cardiol. 2019, 72, 853–862, (In English and Spanish). [Google Scholar] [CrossRef]
  19. Bie, P.; Mølstrøm, S.; Wamberg, S. Normotensive sodium loading in conscious dogs: Regulation of renin secretion during beta-receptor blockade. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2009, 296, R428–R435. [Google Scholar] [CrossRef] [Green Version]
  20. Sayer, G.; Bhat, G. The renin-angiotensin-aldosterone system and heart failure. Cardiol. Clin. 2014, 32, 21–32. [Google Scholar] [CrossRef]
  21. Bruno, N.; Sinagra, G.; Paolillo, S.; Bonomi, A.; Corrà, U.; Piepoli, M.; Veglia, F.; Salvioni, E.; Lagioia, R.; Metra, M.; et al. Mineralocorticoid receptor antagonists for heart failure: A real-life observational study. ESC Heart Fail. 2018, 5, 267–274. [Google Scholar] [CrossRef] [Green Version]
  22. Casu, G.; Merella, P. Diuretic Therapy in Heart Failure-Current Approaches. Eur. Cardiol. 2015, 10, 42–47. [Google Scholar] [CrossRef] [PubMed]
  23. Levine, T.B. Role of vasodilators in the treatment of congestive heart failure. Am. J. Cardiol. 1985, 55, 32A–35A. [Google Scholar] [CrossRef]
  24. David, M.N.V.; Shetty, M. Digoxin Toxicity. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2021. Available online: https://www.ncbi.nlm.nih.gov/books/NBK556025/ (accessed on 25 January 2022).
  25. Reed, M.; Kerndt, C.C.; Nicolas, D. Ivabradine. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2021. Available online: https://www.ncbi.nlm.nih.gov/books/NBK507783/ (accessed on 25 January 2022).
  26. Yancy, C.; Jessup, M.; Butler, J.; Butler, J.; Casey, D.E., Jr.; Colvin, M.M.; Drazner, M.H.; Filippatos, G.S.; Fonarow, G.C.; Givertz, M.M.; et al. 2017 ACC/AHA/HFSA Focused Update of the 2013 ACCF/AHA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Failure Society of America. Circulation 2017, 136, e137–e161. [Google Scholar] [CrossRef] [PubMed]
  27. Sauer, A.J.; Cole, R.; Jensen, B.C.; Pal, J.; Sharma, N.; Yehya, A.; Vader, J. Practical guidance on the use of sacubitril/valsartan for heart failure. Heart Fail. Rev. 2019, 24, 167–176. [Google Scholar] [CrossRef] [Green Version]
  28. Chandra, A.; Lewis, E.; Claggertt, B.; Desai, A.S.; Packer, M.; Zile, M.R.; Swedberg, K.; Rouleau, J.L.; Shi, V.C.; Lefkowitz, M.P.; et al. The Effects of Sacubitril/Valsartan on Physical and Social Activity Limitations in Heart Failure Patients: The PARADIGM-HF Trial. JAMA Cardiol. 2018, 3, 498–505. [Google Scholar] [CrossRef] [Green Version]
  29. Velazquez, E.; Morrow, D.; DeVore, A.; Duffy, C.I.; Ambrosy, A.P.; McCague, K.; Rocha, R.; Braunwald, E.; PIONEER-HF Investigators. Angiotensin-Neprilysin Inhibition in Acute Decompensated Heart Failure. N. Engl. J. Med. 2019, 380, 539–548. [Google Scholar] [CrossRef]
  30. Myhre, P.L.; Vaduganathan, M.; Claggett, B.; Packer, M.; Desai, A.S.; Rouleau, J.L.; Zile, M.R.; Swedberg, K.; Lefkowitz, M.; Shi, V.; et al. B-type natriuretic peptide during treatment with sacubitril/valsartan: The PARADIGM-HF trial. J. Am. Coll. Cardiol. 2019, 73, 1264–1272. [Google Scholar] [CrossRef]
  31. Lewis, E.F.; Claggett, B.L.; McMurray, J.J.V.; Packer, M.; Lefkowitz, M.P.; Rouleau, J.L.; Liu, J.; Shi, V.C.; Zile, M.R.; Desai, A.S.; et al. Health-related quality of life outcomes in PARADIGM-HF. Circ. Heart Fail. 2017, 10, e003430. [Google Scholar] [CrossRef] [Green Version]
  32. Seferovic, P.; Ponikowski, P.; Anker, S.; Bauersachs, J.; Chioncel, O.; Cleland, J.G.F.; de Boer, R.A.; Drexel, H.; Ben Gal, T.; Hill, L.; et al. Clinical practice update on heart failure 2019: Pharmacotherapy, procedures, devices and patient management. An expert consensus meeting report of The Heart Failure Association of the European Society of Cardiology. Eur. J. Heart Fail. 2019, 21, 1169–1186. [Google Scholar] [CrossRef]
  33. Gillette, M.; Bozkurt, B. Ins and Outs: Perspectives of Inpatient Prescribing for Sacubitril/Valsartan. Ann. Pharmacother. 2020, 55, 805–813. [Google Scholar] [CrossRef]
  34. Byrne, D.; Fahey, T.; Moriarty, F. Efficacy and safety of sacubitril/valsartan in the treatment of heart failure: Protocol for a systematic review incorporating unpublished clinical study reports. HRB Open Res. 2020, 3, 5. [Google Scholar] [CrossRef] [Green Version]
  35. Scheen, A.J. Sodium-glucose cotransporter type 2 inhibitors for the treatment of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2020, 16, 556–577. [Google Scholar] [CrossRef]
  36. Al Hamed, F.A.; Elewa, H. Potential Therapeutic Effects of Sodium Glucose-linked Cotransporter 2 Inhibitors in Stroke. Clin. Ther. 2020, 42, e242–e249. [Google Scholar] [CrossRef] [PubMed]
  37. Ling, A.W.; Chan, C.C.; Chen, S.W.; Kao, Y.W.; Huang, C.Y.; Chan, Y.H.; Chu, P.H. The risk of new-onset atrial fibrillation in patients with type 2 diabetes mellitus treated with sodium glucose cotransporter 2 inhibitors versus dipeptidyl peptidase-4 inhibitors. Cardiovasc. Diabetol. 2020, 19, 188. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, H.C.; Shiou, Y.L.; Jhuo, S.J.; Chang, C.Y.; Liu, P.L.; Jhuang, W.J.; Dai, Z.K.; Chen, W.Y.; Chen, Y.F.; Lee, A.S. The sodium-glucose co-transporter 2 inhibitor empagliflozin attenuates cardiac fibrosis and improves ventricular hemodynamics in hypertensive heart failure rats. Cardiovasc. Diabetol. 2019, 18, 45. [Google Scholar] [CrossRef] [PubMed]
  39. GrubićRotkvić, P.; CigrovskiBerković, M.; Bulj, N.; Rotkvić, L. Minireview: Are SGLT2 inhibitors heart savers in diabetes? Heart Fail. Rev. 2020, 25, 899–905. [Google Scholar] [CrossRef] [PubMed]
  40. Lopaschuk, G.D.; Verma, S. Mechanisms of Cardiovascular Benefits of Sodium Glucose Co-Transporter 2 (SGLT2) Inhibitors: A State-of-the-Art Review. JACC Basic Transl. Sci. 2020, 5, 632–644. [Google Scholar] [CrossRef]
  41. McMurray, J.J.V.; Solomon, S.D.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; Anand, I.S.; Bělohlávek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008. [Google Scholar] [CrossRef] [Green Version]
  42. Nassif, M.E.; Windsor, S.L.; Tang, F.; Khariton, Y.; Husain, M.; Inzucchi, S.E.; McGuire, D.K.; Pitt, B.; Scirica, B.M.; Austin, B.; et al. Dapagliflozin Effects on Biomarkers, Symptoms, and Functional Status in Patients with Heart Failure with Reduced Ejection Fraction: The DEFINE-HF Trial. Circulation 2019, 140, 1463–1476. [Google Scholar] [CrossRef]
  43. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. EMPEROR-Reduced Trial Investigators. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef]
  44. Anker, S.D.; Butler, J.; Filippatos, G.; Shahzeb Khan, M.; Ferreira, J.P.; Bocchi, E.; Böhm, M.; Brunner-La Rocca, H.P.; Choi, D.J.; EMPEROR-Preserved Trial Committees and Investigators; et al. Baseline characteristics of patients with heart failure with preserved ejection fraction in the EMPEROR-Preserved trial. Eur. J. Heart Fail. 2020, 22, 2383–2392. [Google Scholar] [CrossRef] [PubMed]
  45. Packer, M.; Butler, J.; Zannad, F.; Filippatos, G.; Ferreira, J.P.; Pocock, S.J.; Carson, P.; Anand, I.; Doehner, W.; Haass, M.; et al. Effect of Empagliflozin on Worsening Heart Failure Events in Patients with Heart Failure and Preserved Ejection Fraction: EMPEROR-Preserved Trial. Circulation 2021, 144, 1284–1294. [Google Scholar] [CrossRef] [PubMed]
  46. Gazewood, J.D.; Turner, P.L. Heart failure with preserved ejection fraction: Diagnosis and management. Am. Fam. Physician 2017, 96, 582–588. [Google Scholar]
  47. Mulder, B.A.; Schnabel, R.B.; Rienstra, M. Predicting the future in patients with atrial fibrillation: Who develops heart failure? Eur. J. Heart Fail. 2013, 15, 366–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Ouyang, A.J.; Lv, Y.N.; Zhong, H.L.; Wen, J.H.; Wei, X.H.; Peng, H.W.; Zhou, J.; Liu, L.L. Meta-analysis of digoxin use and risk of mortality in patients with atrial fibrillation. Am. J. Cardiol. 2015, 115, 901–906. [Google Scholar] [CrossRef] [PubMed]
  49. Van Gelder, I.C.; Haegeli, L.M.; Brandes, A.; Heidbuchel, H.; Aliot, E.; Kautzner, J.; Szumowski, L.; Mont, L.; Morgan, J.; Willems, S.; et al. Rationale and current perspective for early rhythm control therapy in atrial fibrillation. Europace 2011, 13, 1517–1525. [Google Scholar] [CrossRef] [PubMed]
  50. Packer, M.; Califf, R.M.; Konstam, M.A.; Krum, H.; McMurray, J.J.; Rouleau, J.L.; Swedberg, K. Comparison of omapatrilat and enalapril in patients with chronic heart failure: The Omapatrilat Versus Enalapril Randomized Trial of Utility in Reducing Events (OVERTURE). Circulation 2002, 106, 920–926. [Google Scholar] [CrossRef] [PubMed]
  51. Fleg, J.L.; Strait, J. Age-associated changes in cardiovascular structure and function: A fertile milieu for future disease. Heart Fail. Rev. 2012, 17, 545–554. [Google Scholar] [CrossRef] [Green Version]
  52. Lakatta, E.G. Diminished beta-adrenergic modulation of cardiovascular function in advanced age. Cardiol. Clin. 1986, 4, 185–200. [Google Scholar] [CrossRef]
  53. Loffredo, F.S.; Nikolova, A.P.; Pancoast, J.R.; Lee, R.T. Heart failure with preserved ejection fraction: Molecular pathways of the aging myocardium. Circ. Res. 2014, 115, 97–107. [Google Scholar] [CrossRef] [Green Version]
  54. Olivetti, G.; Melissari, M.; Capasso, J.M.; Anversa, P. Cardiomyopathy of the aging human heart. Myocyte loss and reactive cellular hypertrophy. Circ. Res. 1991, 68, 1560–1568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Burgess, M.L.; McCrea, J.C.; Hedrick, H.L. Age-associated changes in cardiac matrix and integrins. Mech. Ageing Dev. 2001, 122, 1739–1756. [Google Scholar] [CrossRef]
  56. Eghbali, M.; Eghbali, M.; Robinson, T.F.; Seifter, S.; Blumenfeld, O.O. Collagen accumulation in heart ventricles as a function of growth and aging. Cardiovasc. Res. 1989, 23, 723–729. [Google Scholar] [CrossRef] [PubMed]
  57. Lakatta, E.G.; Levy, D. Arterial and cardiac aging: Major shareholders in cardiovascular disease enterprises: Part I: Aging arteries: A “set up” for vascular disease. Circulation 2003, 107, 139–146. [Google Scholar] [CrossRef] [Green Version]
  58. Lakatta, E.G.; Levy, D. Arterial and cardiac aging: Major shareholders in cardiovascular disease enterprises: Part II: The aging heart in health: Links to heart disease. Circulation 2003, 107, 346–354. [Google Scholar] [CrossRef] [Green Version]
  59. Picard, M.; McEwen, B.S. Psychological Stress and Mitochondria: A Conceptual Framework. Psychosom. Med. 2018, 80, 126–140. [Google Scholar] [CrossRef]
  60. Liamis, G.; Milionis, H.J.; Elisaf, M. A review of drug-induced hypernatraemia. NDT Plus 2009, 2, 339–346. [Google Scholar] [CrossRef] [Green Version]
  61. Miller, M. Fluid and electrolyte homeostasis in the elderly: Physiological changes of ageing and clinical consequences. Baillière’s Clin. Endocrinol. Metab. 1997, 11, 367–387. [Google Scholar] [CrossRef]
  62. Peeters, L.E.J.; Kester, M.P.; Feyz, L.; Van Den Bemt, P.M.L.A.; Koch, B.C.P.; Van Gelder, T.; Versmissen, J. Pharmacokinetic and pharmacodynamic considerations in the treatment of the elderly patient with hypertension. Expert Opin. Drug Metab. Toxicol. 2019, 15, 287–297. [Google Scholar] [CrossRef]
  63. Barr, R.G.; Bluemke, D.A.; Ahmed, F.S.; Carr, J.J.; Enright, P.L.; Hoffman, E.A.; Jiang, R.; Kawut, S.M.; Kronmal, R.A.; Lima, J.A.; et al. Percent emphysema, airflow obstruction, and impaired left ventricular filling. N. Engl. J. Med. 2010, 362, 217–227. [Google Scholar] [CrossRef] [Green Version]
  64. Petrescu, C.; Schlink, U.; Richter, M.; Suciu, O.; Ionovici, R.; Herbarth, O. Risk assessment of the respiratory health effects due to air pollution and meteorological factors in a population from Drobeta Turnu Severin, Romania. In Proceedings of the 17th European Symposium on Computer Aided Process Engineering, Cluj, Romania, 27–30 May 2007; Volume 24, pp. 1205–1210. [Google Scholar]
  65. Monfredi, O.; Lakatta, E.G. Complexities in cardiovascular rhythmicity: Perspectives on circadian normality, ageing and disease. Cardiovasc. Res. 2019, 115, 1576–1595. [Google Scholar] [CrossRef] [PubMed]
  66. Savarese, G.; D’Amario, D. Sex Differences in Heart Failure. Adv. Exp. Med. Biol. 2018, 1065, 529–544. [Google Scholar] [CrossRef] [PubMed]
  67. Aimo, A.; Vergaro, G.; Barison, A.; Maffei, S.; Borrelli, C.; Morrone, D.; Cameli, M.; Palazzuoli, A.; Ambrosio, G.; Coiro, S.; et al. Sex-related differences in chronic heart failure. Int. J. Cardiol. 2018, 255, 145–151. [Google Scholar] [CrossRef] [PubMed]
  68. Luo, T.; Kim, J.K. The Role of Estrogen and Estrogen Receptors on Cardiomyocytes: An Overview. Can. J. Cardiol. 2016, 32, 1017–1025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Bouma, W.; Noma, M.; Kanemoto, S.; Matsubara, M.; Leshnower, B.G.; Hinmon, R.; Gorman, J.H., 3rd; Gorman, R.C. Sex-related resistance to myocardial ischemia-reperfusion injury is associated with high constitutive ARC expression. Am. J. Physiol. Heart Circ. Physiol. 2010, 298, H1510–H1517. [Google Scholar] [CrossRef] [Green Version]
  70. Jia, M.; Dahlman-Wright, K.; Gustafsson, J.Å. Estrogen receptor alpha and beta in health and disease. Best Pract. Res. Clin. Endocrinol. Metab. 2015, 29, 557–568. [Google Scholar] [CrossRef]
  71. Tasevska-Dinevska, G.; Kennedy, L.M.; Cline-Iwarson, A.; Cline, C.; Erhardt, L.; Willenheimer, R. Gender differences in variables related to B-natriuretic peptide, left ventricular ejection fraction and mass, and peak oxygen consumption, in patients with heart failure. Int. J. Cardiol. 2011, 149, 364–371. [Google Scholar] [CrossRef]
  72. Elmariah, S.; Goldberg, L.R.; Allen, M.T.; Kao, A. Effects of gender on peak oxygen consumption and the timing of cardiac transplantation. J. Am. Coll. Cardiol. 2006, 47, 2237–2242. [Google Scholar] [CrossRef] [Green Version]
  73. vanDeursen, V.M.; Urso, R.; Laroche, C.; Damman, K.; Dahlström, U.; Tavazzi, L.; Maggioni, A.P.; Voors, A.A. Co-morbidities in patients with heart failure: An analysis of the European Heart Failure Pilot Survey. Eur. J. Heart Fail. 2014, 16, 103–111. [Google Scholar] [CrossRef]
  74. Hudson, M.; Rahme, E.; Behlouli, H.; Sheppard, R.; Pilote, L. Sex differences in the effectiveness of angiotensin receptor blockers and angiotensin converting enzyme inhibitors in patients with congestive heart failure—A population study. Eur. J. Heart Fail. 2007, 9, 602–609. [Google Scholar] [CrossRef]
  75. Ghali, J.K.; Lindenfeld, J. Sex differences in response to chronic heart failure therapies. Expert Rev. Cardiovasc. Ther. 2008, 6, 555–565. [Google Scholar] [CrossRef]
  76. Adams, K.F., Jr.; Patterson, J.H.; Gattis, W.A.; O’Connor, C.M.; Lee, C.R.; Schwartz, T.A.; Gheorghiade, M. Relationship of serum digoxin concentration to mortality and morbidity in women in the digitalis investigation group trial: A retrospective analysis. J. Am. Coll. Cardiol. 2005, 46, 497–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Valentova, M.; von Haehling, S. An overview of recent developments in the treatment of heart failure: Update from the ESC Congress 2013. Expert Opin. Investig. Drugs 2014, 23, 573–578. [Google Scholar] [CrossRef] [PubMed]
  78. Arutyunov, G.P.; Kostyukevich, O.I.; Serov, R.A.; Rylova, N.V.; Bylova, N.A. Collagen accumulation and dysfunctional mucosal barrier of the small intestine in patients with chronic heart failure. Int. J. Cardiol. 2008, 125, 240–245. [Google Scholar] [CrossRef] [PubMed]
  79. Sandek, A.; Bauditz, J.; Swidsinski, A.; Buhner, S.; Weber-Eibel, J.; von Haehling, S.; Schroedl, W.; Karhausen, T.; Doehner, W.; Rauchhaus, M.; et al. Altered intestinal function in patients with chronic heart failure. J. Am. Coll. Cardiol. 2007, 50, 1561–1569. [Google Scholar] [CrossRef] [Green Version]
  80. Schwartz, J.B. The current state of knowledge on age, sex, and their interactions on clinical pharmacology. Clin. Pharmacol. Ther. 2007, 82, 87–96. [Google Scholar] [CrossRef] [PubMed]
  81. Sica, D.A.; Wood, M.; Hess, M. Gender and its effect in cardiovascular pharmacotherapeutics: Recent considerations. Congest. Heart Fail. 2005, 11, 163–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ogawa, R.; Stachnik, J.M.; Echizen, H. Clinical pharmacokinetics of drugs in patients with heart failure: An update (part 1, drugs administered intravenously). Clin. Pharmacokinet. 2013, 52, 169–185. [Google Scholar] [CrossRef]
  83. Valentová, M.; von Haehling, S.; Doehner, W.; Murín, J.; Anker, S.D.; Sandek, A. Liver dysfunction and its nutritional implications in heart failure. Nutrition 2013, 29, 370–378. [Google Scholar] [CrossRef]
  84. Ogawa, R.; Stachnik, J.M.; Echizen, H. Clinical pharmacokinetics of drugs in patients with heart failure: An update (part 2, drugs administered orally). Clin. Pharmacokinet. 2014, 53, 1083–1114. [Google Scholar] [CrossRef]
  85. Mangoni, A.A.; Jarmuzewska, E.A. The influence of heart failure on the pharmacokinetics of cardiovascular and non-cardiovascular drugs: A critical appraisal of the evidence. Br. J. Clin. Pharmacol. 2019, 85, 20–36. [Google Scholar] [CrossRef] [PubMed]
  86. Bader, F.; Atallah, B.; Brennan, L.F.; Rimawi, R.H.; Khalil, M.E. Heart failure in the elderly: Ten peculiar management considerations. Heart Fail. Rev. 2017, 22, 219–228. [Google Scholar] [CrossRef] [PubMed]
  87. Rangaswami, J.; Bhalla, V.; Blair, J.E.A.; Chang, T.I.; Costa, S.; Lentine, K.L.; Lerma, E.V.; Mezue, K.; Molitch, M.; Mullens, W.; et al. Cardiorenal Syndrome: Classification, Pathophysiology, Diagnosis, and Treatment Strategies: A Scientific Statement From the American Heart Association. Circulation 2019, 139, e840–e878. [Google Scholar] [CrossRef] [PubMed]
  88. Rangaswami, J.; Mathew, R.O. Pathophysiological Mechanisms in Cardiorenal Syndrome. Adv. Chronic. Kidney Dis. 2018, 25, 400–407. [Google Scholar] [CrossRef]
  89. Kousa, O.; Mullane, R.; Aboeata, A. Cardiorenal Syndrome. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2021. Available online: https://www.ncbi.nlm.nih.gov/books/NBK542305/ (accessed on 25 January 2022).
  90. Damman, K.; Testani, J.M. The kidney in heart failure: An update. Eur. Heart J. 2015, 36, 1437–1444. [Google Scholar] [CrossRef] [Green Version]
  91. Ronco, C.; Chionh, C.Y.; Haapio, M.; Anavekar, N.S.; House, A.; Bellomo, R. The cardiorenal syndrome. Blood Purif. 2009, 27, 114–126. [Google Scholar] [CrossRef]
  92. Anthony, J.; Sliwa, K. Decompensated Heart Failure in Pregnancy. Card Fail. Rev. 2016, 2, 20–26. [Google Scholar] [CrossRef] [Green Version]
  93. Stergiopoulos, K.; Lima, F.V.; Butler, J. Heart Failure in Pregnancy: A Problem Hiding in Plain Sight. J. Am. Heart Assoc. 2019, 8, e012905. [Google Scholar] [CrossRef] [Green Version]
  94. Dorn, G.W., 2nd. The fuzzy logic of physiological cardiac hypertrophy. Hypertension 2007, 49, 962–970. [Google Scholar] [CrossRef] [Green Version]
  95. Kearney, L.; Wright, P.; Fhadil, S.; Thomas, M. Postpartum Cardiomyopathy and Considerations for Breastfeeding. Card Fail. Rev. 2018, 4, 112–118. [Google Scholar] [CrossRef]
  96. Tschiderer, L.; Seekircher, L.; Kunutsor, S.K.; Peters, S.A.E.; O’Keeffe, L.M.; Willeit, P. Breastfeeding Is Associated with a Reduced Maternal Cardiovascular Risk: Systematic Review and Meta-Analysis Involving Data from 8 Studies and 1 192 700 Parous Women. J. Am. Heart Assoc. 2022, 11, e022746. [Google Scholar] [CrossRef] [PubMed]
  97. Von Lueder, T.G.; Atar, D. Comorbidities and polypharmacy. Heart Fail. Clin. 2014, 10, 367–372. [Google Scholar] [CrossRef] [PubMed]
  98. Brockhattingen, K.K.; Anru, P.L.; Masud, T.; Petrovic, M.; Ryg, J. Association between number of medications and mortality in geriatric inpatients: A Danish nationwide register-based cohort study. Eur. Geriatr. Med. 2020, 11, 1063–1071. [Google Scholar] [CrossRef] [PubMed]
  99. Page, R.L., 2nd; O’Bryant, C.L.; Cheng, D.; Dow, T.J.; Ky, B.; Stein, C.M.; Spencer, A.P.; Trupp, R.J.; Lindenfeld, J.; American Heart Association Clinical Pharmacology and Heart Failure and Transplantation Committees of the Council on Clinical Cardiology; et al. Drugs That May Cause or Exacerbate Heart Failure: A Scientific Statement from the American Heart Association. Circulation 2016, 134, e32–e69. [Google Scholar] [CrossRef] [PubMed]
  100. Sunaga, T.; Yokoyama, A.; Nakamura, S.; Miyamoto, N.; Watanabe, S.; Tsujiuchi, M.; Nagumo, S.; Nogi, A.; Maezawa, H.; Mizukami, T.; et al. Association of Potentially Inappropriate Medications with All-Cause Mortality in the Elderly Acute Decompensated Heart Failure Patients: Importance of Nonsteroidal Anti-Inflammatory Drug Prescription. Cardiol. Res. 2020, 11, 239–246. [Google Scholar] [CrossRef] [PubMed]
  101. Jödicke, A.M.; Burden, A.M.; Zellweger, U.; Tomka, I.T.; Neuer, T.; Roos, M.; Kullak-Ublick, G.A.; Curkovic, I.; Egbring, M. Medication as a risk factor for hospitalization due to heart failure and shock: A series of case-crossover studies in Swiss claims data. Eur. J. Clin. Pharmacol. 2020, 76, 979–989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Huerta, C.; Varas-Lorenzo, C.; Castellsague, J.; García Rodríguez, L.A. Non-steroidal anti-inflammatory drugs and risk of first hospital admission for heart failure in the general population. Heart 2006, 92, 1610–1615. [Google Scholar] [CrossRef] [Green Version]
  103. Silva Almodóvar, A.; Nahata, M.C. Potentially Harmful Medication Use among Medicare Patients with Heart Failure. Am. J. Cardiovasc. Drugs 2020, 20, 603–610. [Google Scholar] [CrossRef]
  104. Arfè, A.; Scotti, L.; Varas-Lorenzo, C.; Zambon, A.; Kollhorst, B.; Schink, T.; Garbe, E.; Herings, R.; Straatman, H.; Schade, R.; et al. Safety of Non-steroidal Anti-inflammatory Drugs (SOS) Project Consortium. Non-steroidal anti-inflammatory drugs and risk of heart failure in four European countries: Nested case-control study. BMJ 2016, 354, i4857. [Google Scholar] [CrossRef] [Green Version]
  105. Alvarez, P.A.; Putney, D.; Ogunti, R.; Puppala, M.; Ganduglia, C.; Torre-Amione, G.; Schutt, R.; Wong, S.T.C.; Estep, J.D. Prevalence of in-hospital nonsteroidal antiinflammatory drug exposure in patients with a primary diagnosis of heart failure. Cardiovasc. Ther. 2017, 35, e12256. [Google Scholar] [CrossRef] [PubMed]
  106. Alvarez, P.A.; Gao, Y.; Girotra, S.; Mentias, A.; Briasoulis, A.; Vaughan Sarrazin, M.S. Potentially harmful drug prescription in elderly patients with heart failure with reduced ejection fraction. ESC Heart Fail. 2020, 7, 1862–1871. [Google Scholar] [CrossRef] [PubMed]
  107. Qian, X.; Li, M.; Wagner, M.B.; Chen, G.; Song, X. Doxazosin Stimulates Galectin-3 Expression and Collagen Synthesis in HL-1 Cardiomyocytes Independent of Protein Kinase C Pathway. Front. Pharmacol. 2016, 7, 495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Edwards, L.P.; Brown-Bryan, T.A.; McLean, L.; Ernsberger, P. Pharmacological properties of the central antihypertensive agent, moxonidine. Cardiovasc. Ther. 2012, 30, 199–208. [Google Scholar] [CrossRef] [PubMed]
  109. Valembois, L.; Audureau, E.; Takeda, A.; Jarzebowski, W.; Belmin, J.; Lafuente-Lafuente, C. Antiarrhythmics for maintaining sinus rhythm after cardioversion of atrial fibrillation. Cochrane Database Syst. Rev. 2019, 9, CD005049. [Google Scholar] [CrossRef]
  110. Frommeyer, G.; Milberg, P.; Witte, P.; Stypmann, J.; Koopmann, M.; Lücke, M.; Osada, N.; Breithardt, G.; Fehr, M.; Eckardt, L. A new mechanism preventing proarrhythmia in chronic heart failure: Rapid phase-III repolarization explains the low proarrhythmic potential of amiodarone in contrast to sotalol in a model of pacing-induced heart failure. Eur. J. Heart Fail. 2011, 13, 1060–1069. [Google Scholar] [CrossRef]
  111. Finks, S.W.; Rogers, K.C.; Manguso, A.H. Assessment of sotalol prescribing in a community hospital: Opportunities for clinical pharmacist involvement. Int. J. Pharm. Pract. 2011, 19, 281–286. [Google Scholar] [CrossRef]
  112. Kongwatcharapong, J.; Dilokthornsakul, P.; Nathisuwan, S.; Phrommintikul, A.; Chaiyakunapruk, N. Effect of dipeptidyl peptidase-4 inhibitors on heart failure: A meta-analysis of randomized clinical trials. Int. J. Cardiol. 2016, 211, 88–95. [Google Scholar] [CrossRef] [Green Version]
  113. Savarese, G.; Schrage, B.; Cosentino, F.; Lund, L.H.; Rosano, G.M.C.; Seferovic, P.; Butler, J. Non-insulin antihyperglycaemic drugs and heart failure: An overview of current evidence from randomized controlled trials. ESC Heart Fail. 2020, 7, 3438–3451. [Google Scholar] [CrossRef]
  114. Patel, K.V.; Sarraju, A.; Neeland, I.J.; McGuire, D.K. Cardiovascular Effects of Dipeptidyl Peptidase-4 Inhibitors and Glucagon-Like Peptide-1 Receptor Agonists: A Review for the General Cardiologist. Curr. Cardiol. Rep. 2020, 22, 105. [Google Scholar] [CrossRef]
  115. Nassif, M.E.; Kosiborod, M. A Review of Cardiovascular Outcomes Trials of Glucose-Lowering Therapies and Their Effects on Heart Failure Outcomes. Am. J. Cardiol. 2019, 124 (Suppl. S1), S12–S19. [Google Scholar] [CrossRef] [Green Version]
  116. Hantson, P. Mechanisms of toxic cardiomyopathy. Clin. Toxicol. 2019, 57, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Wallach, J.D.; Wang, K.; Zhang, A.D.; Cheng, D.; Grossetta Nardini, H.K.; Lin, H.; Bracken, M.B.; Desai, M.; Krumholz, H.M.; Ross, J.S. Updating insights into rosiglitazone and cardiovascular risk through shared data: Individual patient and summary level meta-analyses. BMJ 2020, 368, l7078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Teaford, H.R.; Abu Saleh, O.M.; Villarraga, H.R.; Enzler, M.J.; Rivera, C.G. The Many Faces of Itraconazole Cardiac Toxicity. Mayo Clin. Proc. Innov. Qual. Outcomes 2020, 4, 588–594. [Google Scholar] [CrossRef] [PubMed]
  119. Abraham, A.O.; Panda, P.K. Itraconazole Induced Congestive Heart Failure, A Case Study. Curr. Drug Saf. 2018, 13, 59–61. [Google Scholar] [CrossRef] [PubMed]
  120. Paul, V.; Rawal, H. Cardiotoxicity with Itraconazole. BMJ Case Rep. 2017, 2017, bcr2017219376. [Google Scholar] [CrossRef]
  121. Soares, J.R.; Nunes, M.C.; Leite, A.F.; Falqueto, E.B.; Lacerda, B.E.; Ferrari, T.C. Reversible dilated cardiomyopathy associated with amphotericin B therapy. J. Clin. Pharm. Ther. 2015, 40, 333–335. [Google Scholar] [CrossRef]
  122. Mégarbane, B.; Leprince, P.; Deye, N.; Guerrier, G.; Résière, D.; Bloch, V.; Baud, F.J. Extracorporeal life support in a case of acute carbamazepine poisoning with life-threatening refractory myocardial failure. Intensive Care Med. 2006, 32, 1409–1413. [Google Scholar] [CrossRef]
  123. Faisy, C.; Guerot, E.; Diehl, J.L.; Rezgui, N.; Labrousse, J. Carbamazepine-associated severe left ventricular dysfunction. J. Toxicol. Clin. Toxicol. 2000, 38, 339–342. [Google Scholar] [CrossRef]
  124. Takamiya, M.; Aoki, Y.; Niitsu, H.; Saigusa, K. A case of carbamazepine overdose with focal myocarditis. Leg. Med. 2006, 8, 243–247. [Google Scholar] [CrossRef]
  125. Lund, M.; Poulsen, G.; Pasternak, B.; Worm Andersson, N.; Melbye, M.; Svanström, H. Use of Pregabalin and Worsening Heart Failure: A Nationwide Cohort Study. Drug Saf. 2020, 43, 1035–1044. [Google Scholar] [CrossRef]
  126. Awwad, Z.M.; El-Ganainy, S.O.; ElMallah, A.I.; Khedr, S.M.; Khattab, M.M.; El-Khatib, A.S. Assessment of Pregabalin-Induced Cardiotoxicity in Rats: Mechanistic Role of Angiotensin 17–7. Cardiovasc. Toxicol. 2020, 20, 301–311. [Google Scholar] [CrossRef] [PubMed]
  127. Ho, J.M.; Tricco, A.C.; Perrier, L.; Chen, M.; Juurlink, D.N.; Straus, S.E. Risk of heart failure and edema associated with the use of pregabalin: A systematic review. Syst. Rev. 2013, 2, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Nezafati, M.H.; Vojdanparast, M.; Nezafati, P. Antidepressants and cardiovascular adverse events: A narrative review. ARYA Atheroscler. 2015, 11, 295–304. [Google Scholar] [PubMed]
  129. Biffi, A.; Rea, F.; Scotti, L.; Lucenteforte, E.; Vannacci, A.; Lombardi, N.; Chinellato, A.; Onder, G.; Vitale, C.; Cascini, S.; et al. Antidepressants and the Risk of Cardiovascular Events in Elderly Affected by Cardiovascular Disease: A Real-Life Investigation From Italy. J. Clin. Psychopharmacol 2020, 40, 112–121. [Google Scholar] [CrossRef]
  130. Deshmukh, A.; Ulveling, K.; Alla, V.; Abuissa, H.; Airey, K. Prolonged QTc interval and torsades de pointes induced by citalopram. Tex. Heart Inst. J. 2012, 39, 68–70. [Google Scholar]
  131. Assimon, M.M.; Brookhart, M.A.; Flythe, J.E. Comparative Cardiac Safety of Selective Serotonin Reuptake Inhibitors among Individuals Receiving Maintenance Hemodialysis. J. Am. Soc. Nephrol. 2019, 30, 611–623. [Google Scholar] [CrossRef]
  132. Tran, T.; Brophy, J.M.; Suissa, S.; Renoux, C. Risks of Cardiac Valve Regurgitation and Heart Failure Associated with Ergot- and Non-Ergot-Derived Dopamine Agonist Use in Patients with Parkinson’s Disease: A Systematic Review of Observational Studies. CNS Drugs 2015, 29, 985–998. [Google Scholar] [CrossRef]
  133. Montastruc, F.; Moulis, F.; Araujo, M.; Chebane, L.; Rascol, O.; Montastruc, J.L. Ergot and non-ergot dopamine agonists and heart failure in patients with Parkinson’s disease. Eur. J. Clin. Pharmacol. 2017, 73, 99–103. [Google Scholar] [CrossRef]
  134. Renoux, C.; Dell’Aniello, S.; Brophy, J.M.; Suissa, S. Dopamine agonist use and the risk of heart failure. Pharmacoepidemiol. Drug Saf. 2012, 21, 34–41. [Google Scholar] [CrossRef]
  135. Patuszynski, D.; Applegate, P.M. Suspected Clozapine-Induced Cardiomyopathy and Heart Failure with Reduced Ejection Fraction. Fed. Pract. 2017, 34, 20–22. [Google Scholar]
  136. Whiskey, E.; Yuen, S.; Khosla, E.; Piper, S.; O’Flynn, D.; Taylor, D. Resolution without discontinuation: Heart failure during clozapine treatment. Ther. Adv. Psychopharmacol. 2020, 10, 2045125320924786. [Google Scholar] [CrossRef]
  137. Garg, A.; Bath, A.S.; Kalavakunta, J.K. Non-ischemic Cardiomyopathy: A Rare Adverse Effect of Clozapine. Cureus 2020, 12, e7901. [Google Scholar] [CrossRef] [PubMed]
  138. Chow, V.; Yeoh, T.; Ng, A.C.; Pasqualon, T.; Scott, E.; Plater, J.; Whitwell, B.; Hanzek, D.; Chung, T.; Thomas, L.; et al. Asymptomatic left ventricular dysfunction with long-term clozapine treatment for schizophrenia: A multicentre cross-sectional cohort study. Open Heart 2014, 1, e000030. [Google Scholar] [CrossRef] [PubMed]
  139. Salimi, A.; Gholamifar, E.; Naserzadeh, P.; Hosseini, M.J.; Pourahmad, J. Toxicity of lithium on isolated heart mitochondria and cardiomyocyte: A justification for its cardiotoxic adverse effect. J. Biochem. Mol. Toxicol. 2017, 31, e21836. [Google Scholar] [CrossRef] [PubMed]
  140. Mezni, A.; Aoua, H.; Khazri, O.; Limam, F.; Aouani, E. Lithium induced oxidative damage and inflammation in the rat’s heart: Protective effect of grape seed and skin extract. Biomed. Pharmacother. 2017, 95, 1103–1111. [Google Scholar] [CrossRef]
  141. Asim, K.; Selman, Y.; Suleyman, Y.; Ozgur, K.; Ozlem, B.; Gokhan, E. Heart Attack in the Course of Lithium Overdose. Iran. Red Crescent Med. J. 2016, 18, e21731. [Google Scholar] [CrossRef] [Green Version]
  142. Acharya, S.; Siddiqui, A.H.; Anwar, S.; Habib, S.; Anwar, S. Lithium-induced Cardiotoxicity: A Rare Clinical Entity. Cureus 2020, 12, e7286, Erratum in Cureus 2020, 12, c33. [Google Scholar] [CrossRef] [Green Version]
  143. Ataallah, B.; Al-Zakhari, R.; Sharma, A.; Tofano, M.; Haggerty, G. A Rare but Reversible Cause of Lithium-Induced Bradycardia. Cureus 2020, 12, e8600. [Google Scholar] [CrossRef]
  144. Wang, Y.; Yuan, J.; Qian, Z.; Zhang, X.; Chen, Y.; Hou, X.; Zou, J. β2 adrenergic receptor activation governs cardiac repolarization and arrhythmogenesis in a guinea pig model of heart failure. Sci. Rep. 2015, 5, 7681. [Google Scholar] [CrossRef] [Green Version]
  145. Say, B.; Degirmencioglu, H.; Kutman, H.; Uras, N.; Dilmen, U. Supraventricular tachycardia after nebulized salbutamol therapy in a neonate: Case report. Arch. Argent. Pediatr. 2015, 113, e98–e100, (In English and Spanish). [Google Scholar] [CrossRef]
  146. Yu, Y.; Wei, S.G.; Weiss, R.M.; Felder, R.B. TNF-α receptor 1 knockdown in the subfornical organ ameliorates sympathetic excitation and cardiac hemodynamics in heart failure rats. Am. J. Physiol. Heart Circ. Physiol. 2017, 313, H744–H756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Yu, Y.; Cao, Y.; Bell, B.; Chen, X.; Weiss, R.M.; Felder, R.B.; Wei, S.G. Brain TACE (Tumor Necrosis Factor-α-Converting Enzyme) Contributes to Sympathetic Excitation in Heart Failure Rats. Hypertension 2019, 74, 63–72. [Google Scholar] [CrossRef] [PubMed]
  148. Schumacher, S.M.; Naga Prasad, S.V. Tumor Necrosis Factor-α in Heart Failure: An Updated Review. Curr. Cardiol. Rep. 2018, 20, 117. [Google Scholar] [CrossRef] [PubMed]
  149. Suroowan, S.; Mahomoodally, F. Common phyto-remedies used against cardiovascular diseases and their potential to induce adverse events in cardiovascular patients. Clin. Phytoscience 2015, 1, 1. [Google Scholar] [CrossRef] [Green Version]
  150. Kim, E.J.Y.; Chen, Y.; Huang, J.Q.; Li, K.M.; Razmovski-Naumovski, V.; Poon, J.; Li, K.M.; Razmovski-Naumovski, V.; Poon, J.; Chan, K.; et al. Evidence-based toxicity evaluation and scheduling of Chinese herbal medicines. J. Ethnopharmacol. 2013, 146, 40–61. [Google Scholar] [CrossRef]
  151. Alternative Medicine Review. Aesculus hippocacastanum. 2009, 14, 278–283.
  152. Tachjian, A.; Maria, V.; Jahangir, A. Use of herbal products and potential interactions in patients with cardiovascular diseases. J. Am. Coll. Cardiol. 2010, 55, 515–525. [Google Scholar] [CrossRef] [Green Version]
  153. World Health Organization. Aloe. Folium Sennae, Fructus Sennae. Radix Glycyrrhizae. WHO Monograph on Selected Medicinal Plants—Volume 1. ISBN: 9241545178. Available online: http://apps.who.int/medicinedocs/en/d/Js2200e/5.html (accessed on 25 January 2022)ISBN 9241545178.
  154. World Health Organization. Radix Angelicae Sinensis. WHO Monograph on Selected Medicinal Plants—Volume 2. Available online: http://apps.who.int/medicinedocs/en/d/Js4927e/5.html (accessed on 25 January 2022).
  155. Agosti, S.; Casalino, L.; Bertero, G.; Barsotti, A.; Brunelli, C.; Morelloni, S. A dangerous fruit juice. Am. J. Emerg. Med. 2012, 30, 248.e5–248.e8. [Google Scholar] [CrossRef]
  156. Papandreou, D.; Phily, A. An updated mini review on grapefruit: Interactions with drugs, obesity and cardiovascular Risk factors. Food Nutr. Sci. 2014, 5, 376–381. [Google Scholar] [CrossRef] [Green Version]
  157. Tassell, M.; Kingston, R.; Gilroy, D.; Lehane, M.; Furey, A. Hawthorn (Crataegus spp.) in the treatment of cardiovascular disease. Pharmacogn. Rev. 2010, 4, 32. [Google Scholar]
  158. Pittler, M.H.; Schmidt, K.; Ernst, E. Hawthorn extract for treating chronic heart failure: Meta-analysis of randomized trials. Am. J Med 2003, 114, 665–674. [Google Scholar] [CrossRef]
  159. Chen, W.L.; Tsai, T.H.; Yang, C.C.H.; Kuo, T.B.J. Effects of ephedra on autonomic nervous modulation in healthy young adults. J. Ethnopharmacol. 2010, 130, 563–568. [Google Scholar] [CrossRef] [PubMed]
  160. Koch, E. Inhibition of platelet activating factor (PAF)-induced aggregation of human thrombocytes by ginkgolides: Considerations on possible bleeding complications after oral intake of Ginkgo biloba extracts. Phytomed 2005, 12, 10–16. [Google Scholar] [CrossRef] [PubMed]
  161. Bone, K.M. Potential interaction of Ginkgo biloba leaf with antiplatelet or anticoagulant drugs: What is the evidence? Mol. Nutr. Food Res. 2008, 52, 764–771. [Google Scholar] [CrossRef]
  162. Pengelly, A. Harpagophytum procumbens. Altern. Med. Rev. 2008, 13, 248–252. [Google Scholar]
  163. Calitz, C.; Steenekamp, J.H.; Steyn, J.D.; Gouws, C.; Viljoen, J.M.; Hamman, J.H. Impact of traditional African medicine on drug metabolism and transport. Expert Opin. Drug Metab. Toxicol. 2014, 10, 991–1003. [Google Scholar] [CrossRef]
  164. Johne, A.; Brockmoller, J.; Bauer, S.; Maurer, A.; Langheinrich, M.; Roots, I. Pharmacokinetic interactionof digoxin with an herbal extract from St John’s wort (Hypericum perforatum). Clin. Pharmacol. Ther. 1999, 66, 338–345. [Google Scholar] [CrossRef]
  165. Yue, Q.Y.; Bergquist, C.; Gerden, B. Safety of St John’s wort (Hypericum perforatum). Lancet 2002, 355, 576–577. [Google Scholar] [CrossRef]
  166. Henderson, L.; Yue, Q.Y.; Bergquist, C.; Gerden, B.; Arlett, P. St John’s wort (Hypericum perforatum): Drug interactions and clinical outcomes. Br. J. Clin. Pharmacol. 2002, 54, 349–356. [Google Scholar] [CrossRef] [Green Version]
  167. World Health Organization. Oleum OenotheraeBiennis. WHO Monograph on Selected Medicinal Plants—Volume 2. Available online: http://apps.who.int/medicinedocs/en/d/Js4927e/22.html (accessed on 25 January 2022).
  168. Fecker, R.; Buda, V.; Alexa, E.; Avram, S.; Pavel, I.Z.; Muntean, D.; Cocan, I.; Watz, C.; Minda, D.; Dehelean, C.A.; et al. Phytochemical and biological screening of Oenothera biennis L. Hydroalcoholic extract. Biomolecules 2020, 10, 818. [Google Scholar] [CrossRef]
  169. Frishman, W.H.; Beravol, P.; Carosella, C. Alternative and complementary medicine for preventing and treating cardiovascular disease. Dis. Mon. 2009, 55, 121–192. [Google Scholar] [CrossRef] [PubMed]
  170. World Health Organization. RhizomaZingiberis. WHO Monograph on Selected Medicinal Plants–Volume 1. Available online: http://apps.who.int/medicinedocs/en/d/Js2200e/30.html (accessed on 25 January 2022).
  171. Georgiev, K.D.; Hvarchanova, N.; Georgieva, M.; Kanazirev, B. The role of the clinical pharmacist in the prevention of potential drug interactions in geriatric heart failure patients. Int. J. Clin. Pharm. 2019, 41, 1555–1561. [Google Scholar] [CrossRef] [PubMed]
  172. Bhagat, A.A.; Greene, S.J.; Vaduganathan, M.; Fonarow, G.C.; Butler, J. Initiation, Continuation, Switching, and Withdrawal of Heart Failure Medical Therapies During Hospitalization. JACC Heart Fail. 2019, 7, 1–12. [Google Scholar] [CrossRef] [PubMed]
  173. Anderson, S.L.; Marrs, J.C. A Review of the Role of the Pharmacist in Heart Failure Transition of Care. Adv. Ther. 2018, 35, 311–323. [Google Scholar] [CrossRef] [Green Version]
  174. Investigators of the MAGIC-PHARM Study; Khazaka, M.; Laverdière, J.; Li, C.C.; Correal, F.; Mallet, L.; Poitras, M.; Nguyen, P.V. Medication appropriateness on an acute geriatric care unit: The impact of the removal of a clinical pharmacist. Age Ageing 2020, 50, afaa175. [Google Scholar] [CrossRef]
  175. Association Nationale Des Enseignants de Pharmacie Clinique; Limat, S.; Dupuis, A.; Fagnoni, P.; Deamore, B.; Fernandez, C.; Aulagner, G.; Cazin, J.L. Pharmacie Clinique et Therapeutique; Elsevier: Amsterdam, The Netherlands, 2018; ISBN 9782294750779. [Google Scholar]
  176. Preston, C.L. Stockley’s Drug Interactions, 12th ed.; Pharmaceutical Press: London, UK, 2019; ISBN 978-0-85-711347-4. [Google Scholar]
  177. Sica, D.A. Angiotensin-converting enzyme inhibitors side effects—Physiologic and non-physiologic considerations. J. Clin. Hypertens 2004, 6, 410–416. [Google Scholar] [CrossRef]
  178. Wiggins, B.S.; Saseen, J.J.; Page, R.L., 2nd; Reed, B.N.; Sneed, K.; Kostis, J.B.; Lanfear, D.; Virani, S.; Morris, P.B.; American Heart Association Clinical Pharmacology Committee of the Council on Clinical Cardiology; et al. Recommendations for Management of Clinically Significant Drug-Drug Interactions With Statins and Select Agents Used in Patients With Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation 2016, 134, e468–e495. [Google Scholar] [CrossRef] [Green Version]
  179. Hsiao, H.L.; Langenickel, T.H.; Petruck, J.; Kode, K.; Ayalasomayajula, S.; Schuehly, U.; Greeley, M.; Pal, P.; Zhou, W.; Prescott, M.F.; et al. Evaluation of Pharmacokinetic and Pharmacodynamic Drug-Drug Interaction of Sacubitril/Valsartan (LCZ696) and Sildenafil in Patients with Mild-to-Moderate Hypertension. Clin. Pharmacol. Ther. 2018, 103, 468–476. [Google Scholar] [CrossRef] [Green Version]
  180. Referentiel National des Interactions Medicamenteuses—ANSM. Thesaurus des Interactions Medicamenteuses 2019. Available online: https://ansm.sante.fr/var/ansm_site/storage/original/application/0002510e4ab3a9c13793a1fdc0d4c955.pdf (accessed on 25 January 2022).
  181. Singh, G.; Correa, R. Methimazole. StatPearls [Internet]. 2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK545223/ (accessed on 25 January 2022).
  182. Steffel, J.; Verhamme, P.; Potpara, T.S.; Albaladejo, P.; Antz, M.; Desteghe, L.; Haeusler, K.G.; Oldgren, J.; Reinecke, H.; Roldan-Schilling, V.; et al. The 2018 European Heart Rhythm Association Practical Guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation. Eur. Heart J. 2018, 39, 1330–1393. [Google Scholar] [CrossRef] [Green Version]
  183. Frost, C.E.; Byon, W.; Song, Y.; Wang, J.; Schuster, A.E.; Boyd, R.A.; Zhang, D.; Yu, Z.; Dias, C.; Shenker, A.; et al. Effect of ketoconazole and diltiazem on the pharmacokinetics of apixaban, an oral direct factor Xa inhibitor. Br. J. Clin. Pharmacol. 2015, 79, 838–846. [Google Scholar] [CrossRef] [Green Version]
  184. Bundhun, P.K.; Teeluck, A.R.; Bhurtu, A.; Huang, W.Q. Is the concomitant use of clopidogrel and Proton Pump Inhibitors still associated with increased adverse cardiovascular outcomes following coronary angioplasty? A systematic review and meta-analysis of recently published studies (2012–2016). BMC Cardiovasc. Disord. 2017, 17, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Kariyanna, P.T.; Haseeb, S.; Chowdhury, Y.S.; Jayarangaiah, A.; Maryniak, A.; Mo, G.; Hegde, S.; Marmur, J.D.; McFarlane, I.M. Ticagrelor and Statin Interaction Induces Rhabdomyolysis and Acute Renal Failure: Case reports and Scoping Review. Am. J. Med. Case Rep. 2019, 7, 337–341. [Google Scholar] [CrossRef] [PubMed]
  186. Oscanoa, T.J.; Lizaraso, F.; Carvajal, A. Hospital admissions due to adverse drug reactions in the elderly. A meta-analysis. Eur. J. Clin. Pharmacol. 2017, 73, 759–770. [Google Scholar] [CrossRef] [PubMed]
  187. Tran, P.; Banerjee, P. Iatrogenic Decompensated Heart Failure. Curr. Heart Fail. Rep. 2020, 17, 21–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Rich, M.W.; Shah, A.S.; Vinson, J.M.; Freedland, K.E.; Kuru, T.; Sperry, J.C. Iatrogenic congestive heart failure in older adults: Clinical course and prognosis. J. Am. Geriatr. Soc. 1996, 44, 638–643. [Google Scholar] [CrossRef]
  189. Bots, S.H.; Groepenhoff, F.; Eikendal, A.L.M.; Tannenbaum, C.; Rochon, P.A.; Regitz-Zagrosek, V.; Miller, V.M.; Day, D.; Asselbergs, F.W.; den Ruijter, H.M. Adverse Drug Reactions to Guideline-Recommended Heart Failure Drugs in Women: A Systematic Review of the Literature. JACC Heart Fail. 2019, 7, 258–266. [Google Scholar] [CrossRef]
  190. Tamargo, J.; Rosano, G.; Walther, T.; Duarte, J.; Niessner, A.; Kaski, J.C.; Ceconi, C.; Drexel, H.; Kjeldsen, K.; Savarese, G.; et al. Gender differences in the effects of cardiovascular drugs. Eur. Heart J. Cardiovasc. Pharmacother. 2017, 3, 163–182. [Google Scholar] [CrossRef] [Green Version]
  191. Soldin, O.P.; Chung, S.; Mattison, D.R. Sex differences in drug disposition. J. Biomed. Biotechnol. 2011, 2011, 187103. [Google Scholar] [CrossRef]
  192. Soldin, O.P.; Mattison, D.R. Sex differences in pharmacokinetics and pharmacodynamics. Clin. Pharmacokinet. 2009, 48, 143–158. [Google Scholar] [CrossRef] [Green Version]
  193. Drici, M.D.; Clement, N. Is gender a risk factor for adverse drug reactions? The example of drug-induced long QT syndrome. Drug Saf. 2001, 24, 575–585. [Google Scholar] [CrossRef]
  194. Yap, Y.G.; Camm, A.J. Drug induced QT prolongation and torsades de pointes. Heart 2003, 89, 1363–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Pratt, C.M.; Camm, A.J.; Cooper, W.; Friedman, P.L.; MacNeil, D.J.; Moulton, K.M.; Pitt, B.; Schwartz, P.J.; Veltri, E.P.; Waldo, A.L.; et al. Mortality in the Survival WithORal D-Sotalol (SWORD) trial: Why did patients die? Am. J. Cardiol. 1998, 81, 869–876. [Google Scholar] [CrossRef]
  196. Jochmann, N.; Stangl, K.; Garbe, E.; Baumann, G.; Stangl, V. Female-specific aspects in the pharmacotherapy of chronic cardiovascular diseases. Eur. Heart J. 2005, 26, 1585–1595. [Google Scholar] [CrossRef] [PubMed]
  197. Capodanno, D.; Angiolillo, D.J. Impact of race and gender on antithrombotic therapy. Thromb. Haemost. 2012, 104, 471–484. [Google Scholar] [CrossRef]
  198. Gilstrap, L.G.; Fonarow, G.C.; Desai, A.S.; Fonarow, G.C.; Butler, J. Initiation, Continuation, or Withdrawal of Angiotensin-Converting Enzyme Inhibitors/Angiotensin Receptor Blockers and Outcomes in Patients Hospitalized with Heart Failure with Reduced Ejection Fraction. J. Am. Heart Assoc. 2017, 6, e004675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. O’Brien, E.T. Beta-blockade withdrawal. Lancet 1975, 2, 819. [Google Scholar] [CrossRef]
  200. Miller, R.R.; Olson, H.G.; Amsterdam, E.A.; Mason, D.T. Propranolol-withdrawal rebound phenomenon. Exacerbation of coronary events after abrupt cessation of antianginal therapy. N. Engl. J. Med. 1975, 293, 416–418. [Google Scholar] [CrossRef]
  201. Clark, H.; Krum, H.; Hopper, I. Worsening renal function during renin-angiotensin-aldosterone system inhibitor initiation and long-term outcomes in patients with left ventricular systolic dysfunction. Eur. J. Heart Fail. 2014, 16, 41–48. [Google Scholar] [CrossRef]
  202. Clark, A.L.; Kalra, P.R.; Petrie, M.C.; Mark, P.B.; Tomlinson, L.A.; Tomson, C.R. Change in renal function associated with drug treatment in heart failure: National guidance. Heart 2019, 105, 904–910. [Google Scholar] [CrossRef]
  203. Jondeau, G.; Neuder, Y.; Eicher, J.C.; Jourdain, P.; Fauveau, E.; Galinier, M.; Jegou, A.; Bauer, F.; Trochu, J.N.; Bouzamondo, A.; et al. B-CONVINCED: Beta-blocker CONtinuationVs. INterruption in patients with Congestive heart failure hospitalizED for a decompensation episode. Eur. Heart J. 2009, 30, 2186–2192. [Google Scholar] [CrossRef]
  204. Fonarow, G.C.; Abraham, W.T.; Albert, N.M.; Stough, W.G.; Gheorghiade, M.; Greenberg, B.H.; O’Connor, C.M.; Nunez, E.; Yancy, C.W.; Young, J.B. A smoker’s paradox in patients hospitalized for heart failure: Findings from OPTIMIZE-HF. Eur. Heart J. 2008, 29, 1983–1991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Halliday, B.P.; Wassall, R.; Lota, A.S.; Khalique, Z.; Gregson, J.; Newsome, S.; Jackson, R.; Rahneva, T.; Wage, R.; Smith, G.; et al. Withdrawal of pharmacological treatment for heart failure in patients with recovered dilated cardiomyopathy (TRED-HF): An open-label, pilot, randomized trial. Lancet 2019, 393, 61–73. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Neurohumoral imbalance in heart failure.
Figure 1. Neurohumoral imbalance in heart failure.
Jcm 11 02020 g001
Figure 2. Mechanisms, actions and effects of SGLT2 inhibitors on the heart.
Figure 2. Mechanisms, actions and effects of SGLT2 inhibitors on the heart.
Jcm 11 02020 g002
Figure 3. Particularities in heart failure.
Figure 3. Particularities in heart failure.
Jcm 11 02020 g003
Table 1. The indications, contra-indications and cautions for ARNI and SGLT2 inhibitors.
Table 1. The indications, contra-indications and cautions for ARNI and SGLT2 inhibitors.
ARNISGLT2 Inhibitors
Indications▪ HFrEF (≤40%)
▪ NYHA class II-IV
▪ Alternative of ACEI/ARB
▪ HFrEF (≤40%) ± diabetes mellitus
▪ NYHA class II-IV
Contra-indications- hypersensitivity to the active substances
- history of angioedema
- severe hepatic impairment
- ≤36 h of the last ACEI dose
- hypersensitivity to the active substance
- type I diabetes
- dialysis
- eGFR < 30 mL/min/1.73 m2 (dapagliflozin)
- eGFR < 20 mL/min/1.73 m2 (empagliflozin)
Cautions◊ severe renal impairment (starting dose: 24/26 mg × 2/day)
◊ moderate hepatic impairment (starting dose: 24/26 mg × 2/day)
◊ SBP < 100 mmHg
◊ volume depletion
◊ renal artery stenosis
◊ pregnancy/lactation
◊ high risk of genital infections (especially mycotic) and urinary infections
◊ hypovolemia
◊ ketoacidosis
◊ acute renal impairment
◊ necrotizing fasciitis of the perineum (Fournier gangrene)
◊ bladder cancer
◊ pregnancy
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Buda, V.; Prelipcean, A.; Cozma, D.; Man, D.E.; Negres, S.; Scurtu, A.; Suciu, M.; Andor, M.; Danciu, C.; Crisan, S.; et al. An Up-to-Date Article Regarding Particularities of Drug Treatment in Patients with Chronic Heart Failure. J. Clin. Med. 2022, 11, 2020. https://doi.org/10.3390/jcm11072020

AMA Style

Buda V, Prelipcean A, Cozma D, Man DE, Negres S, Scurtu A, Suciu M, Andor M, Danciu C, Crisan S, et al. An Up-to-Date Article Regarding Particularities of Drug Treatment in Patients with Chronic Heart Failure. Journal of Clinical Medicine. 2022; 11(7):2020. https://doi.org/10.3390/jcm11072020

Chicago/Turabian Style

Buda, Valentina, Andreea Prelipcean, Dragos Cozma, Dana Emilia Man, Simona Negres, Alexandra Scurtu, Maria Suciu, Minodora Andor, Corina Danciu, Simina Crisan, and et al. 2022. "An Up-to-Date Article Regarding Particularities of Drug Treatment in Patients with Chronic Heart Failure" Journal of Clinical Medicine 11, no. 7: 2020. https://doi.org/10.3390/jcm11072020

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop