Next Article in Journal
Development of a Predictive Nomogram for Sepsis in Patients with Urolithiasis-Related Obstructive Pyelonephritis
Previous Article in Journal
Smoking-Dependent Association of Serum Brain-Derived Neurotrophic Factor with Pulmonary Function Parameters in Chronic Obstructive Pulmonary Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unlocking the Potential: Angiotensin Receptor Neprilysin and Sodium Glucose Co-Transporter 2 Inhibitors for Right Ventricle Dysfunction in Heart Failure

Heart Failure and Transplant Program, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
Medicina 2024, 60(7), 1112; https://doi.org/10.3390/medicina60071112
Submission received: 17 June 2024 / Revised: 27 June 2024 / Accepted: 28 June 2024 / Published: 9 July 2024
(This article belongs to the Section Cardiology)

Abstract

:
This review article examines the mechanism of action of Angiotensin Receptor–Neprilysin Inhibitors (ARNIs) and Sodium–Glucose Co-Transporter 2 Inhibitors (SGLT2is) in managing chronic right ventricular (RV) dysfunction. Despite advancements in heart failure (HF) treatment, RV dysfunction remains a significant contributor to morbidity and mortality. This article explores the The article explores the impact of ARNIs and SGLT2is on RV function based on clinical and preclinical evidence, and the potential benefits of combined therapy. It highlights the need for further research to optimize patient outcomes and suggests that RV function should be considered in future clinical trials as part of risk stratification for HF therapies. This review underscores the importance of the early initiation of ARNIs and SGLT2is as per guideline-directed medical therapy for eligible HFrEF and HFpEF patients to improve co-existing RV dysfunction.

1. Introduction

The AHA/ACC/ESC guidelines endorse a quadruple combination therapy, including an angiotensin receptor–neprilysin inhibitor (ARNI), a beta-blocker (BB), a mineralocorticoid receptor antagonist (MRA), and a sodium–glucose co-transporter 2 inhibitor (SGLT2i), for treating heart failure (HF) in adults [1,2]. Despite the remarkable advances in pharmacological therapies, 9.1% of the total deaths in the US were due to HF in 2021 [3]. RV dysfunction remains a significant contributor to morbidity and mortality [4]. Historically, right ventricular (RV) dysfunction has been recognized as part of the HF clinical syndrome, yet it is often overlooked in the risk stratification for HF therapy. However, the advancements in invasive and non-invasive evaluations, combined with clinical and preclinical evidence, have advanced the current understanding of RV pathophysiology across the HF spectrum and reinforced the RV’s centrality in contributing to clinical outcomes. The latest universal definition of HF necessitates the inclusion of symptoms and/or signs linked to cardiac abnormalities and evidence of systemic or pulmonary congestion [5].
RV dysfunction arises from primary myocardial disease, volume overload, pressure overload, and congenital heart disease (CHD) which included RV as systemic ventricle (sRV) that maintain cardiac output [6]. Its relevance to the clinical syndrome of HF is well documented [7,8]. ARNIs and SGLT2is have become essential components of the guideline-directed medical therapy (GDMT) for HF across various ejection fractions (EF), including HFrEF, HFmEF, HFpEF, and HFimpEF [1,2]. While the impact of quadruple HF therapy on left ventricular (LV) function is well established, its effects on RV dysfunction are less well understood. The current GDMT does not explicitly address RV dysfunctioin occuring alongside HFrEF and HFpEF [9,10]. This review synthesizes the clinical trials and experimental evidence to elucidate the mechanisms of action of ARNIs and SGLT2is and their role in chronic RV dysfunction.

2. Differences between RV and LV in HF

The RV and LV exhibit significant differences, including in their morphological features, biomechanics, adaptation, cellular signaling, and calcium handling. These differences may arise from their distinct origins during early development [11]. The RV arises from the secondary heart field, while the LV originates from the primary heart field [12]. These distinct origins likely influence the orientation of myocardial fibers, the fibrous continuity between the inlet and outlet, and the responses to volume and pressure overload [13]. There are also molecular differences between the RV and LV. The gene expression varies between the RV and LV in response to pressure loading and failure, such as the increase in miRNA 28, 148a, and 93 expressions in the RV, while they decrease in the LV [14]. The RV and LV exhibit distinct responses to α1 adrenergic receptor agonists and phosphodiesterase-5 (PDE5) inhibitors [15,16]. The RV shows the stronger fibrotic response to volume loading, while the LV shows a weaker response [17].

3. Pressure–Volume (PV) Relationship between RV and LV in HF

Both the RV and LV share some features in their PV loops, but they differ in their shape, compliance, and response to loading states (Figure 1). The RV loop is triangular, lacking an isovolumetric relaxation phase, with output starting immediately during the systole. Understanding the PV relationship differences is crucial for tailored therapeutic approaches and improved patient outcomes. In HF, the LV–PV loop shifts to the right compared to the normal condition due to an increased LV end-diastolic volume (LVEDV) and end-systolic volume (LVESV), resulting in a decreased LV stroke volume (LVSV), end-systolic pressure (LVESP), and stroke work (LVSW). In HF, the RV–PV loop also shifts right due to an increased RV diastolic volume (RVEDV) and systolic volume (RVESV). Therefore, when exposed to identical volume overload, the RV response is larger compared to the LV, which may be caused by concomitant pulmonary hypertension (PH) [18]. When there is a chronically increased afterload, the RV pressure–volume relation shifts from a trapezoidal shape, reflecting its high efficiency/low impedance, to a square or rectangular shape, with well-developed isovolumic contraction and relaxation periods, indistinguishable from the normal LV pressure–volume loop.

4. RV Dysfunction with HF and Preserved Ejection Fraction (HFpEF)

In HFpEF patients, PH and RV dysfunction represent severe hemodynamic challenges [19,20]. It remains unclear whether RV dysfunction directly results from an elevated afterload or if it occurs independently of pulmonary artery systolic pressure (PASP). In an analysis of 1663 patients with HF, the patients with HFpEF, PASP > 40 mm Hg was associated with a reduced tricuspid plane systolic excursion (TAPSE) [21]. The proposed mechanisms for RV dysfunction are similar to LV dysfunction and result from autonomic nervous system activation, ischemia due to microvascular abnormality, and/or capillary rarefaction [22]. The PARAGON-HF [23] trial revealed that ARNIs did not significantly decrease mortality in HFpEF patients compared to Valsartan. However, it did reduce hospitalizations and N-terminal B-type natriuretic peptide (NT-proBNP) levels. The factors influencing RV dysfunction in HFpEF identified in the trial included a history of atrial fibrillation and flutter, LV remodeling and systolic impairment, and elevated NT-proBNP levels [24]. Notably, gender-specific differences suggest distinct mechanisms in the development of RV dysfunction in HFpEF. Gene expression profiles vary between the LV and RV and the type of LV dysfunction [25]. RV function and RV-PA coupling correlate with pulmonary congestion [26]. A small retrospective study demonstrated that ARNIs improve PH in HFpEF patients [27]. In the EMPEROR-Preserved trial, empagliflozin significantly reduced the primary endpoint of cardiovascular death or hospitalization for heart failure by 29% compared to a placebo, regardless of the presence or absence of diabetes [28,29].

5. RV Dysfunction with HF and Reduced Ejection Fraction (HFrEF)

The prevalence of RV dysfunction is up to 50% in patients with HFrEF, and the severity of RV dysfunction increases in proportion to the severity of LV dysfunction and is not related to PASP [30,31]. Furthermore, the prevalence of RV dysfunction, which results from LV dysfunction and is unaffected by LV ventricular assist device (LVAD) metrics, is up to 40% after continuous use of a LVAD in patients with HFrEF [32]. In these patients, survival is significantly worse among individuals with RV dysfunction than those without RV dysfunction [33]. In general, RV dysfunction and dilatation are significant predictors of adverse outcomes in patients with HFrEF [7,8,34]. RV dysfunction indicates HF progression, while its recovery is associated with an improved prognosis [35]. In an analysis of 1663 patients with HF, among those with HFrEF, a non-sinus rhythm, a high heart rate, ischemic etiology, and an E-wave deceleration time <140 ms were associated with a reduced TAPSE [21]. Observational studies have shown that ARNIs improve RV function and PH in HFrEF patients, independent of LV reverse remodeling [19,26]. Another retrospective study confirmed ARNIs’ efficacy in enhancing RV function, irrespective of demographic factors and LV metrics [36]. Moreover, adding SGLT2is, such as Dapagliflozin or Empagliflozin, to combination therapy has been associated with improvements in RV systolic function within 3–9 months [37,38,39,40]. The current approach to worsening RV failure includes a multifaceted and patient-centric approach to address the complex HF syndrome [41].

6. Mechanism of Action of ARNIs on RV Function

The precise mechanisms by which ARNIs benefit RV function have yet to be fully understood. Valsartan, the ARB component of ARNIs, inhibits the angiotensin II type 2 receptor (AT2R), promoting vasodilation and diuresis. Sacubitril, the neprilysin inhibitor component, is metabolized to LBQ657, which inactivates various vasoactive peptides, including natriuretic peptides (NPs), prostaglandins, bradykinin, adrenomedullin, and apelin, thereby reactivating the NO–pGC–cGMP PKG signaling pathway, which induces natriuresis, diuresis, the suppression of the renin–angiotensin–aldosterone system, and antifibrotic and anti-inflammatory actions [42]. The coadministration of Valsartan with Sacubitril can increase angiotensin levels, as angiotensin II is also a neprilysin substrate. While the specific substrates responsible for ARNIs’ benefits are unclear, studies suggest a synergistic effect of ARNIs over ACEi [43,44] and ARB [45] alone, enhancing the levels of B-type natriuretic peptide (BNP) and urinary cyclic guanosine monophosphate (cGMP) [46,47]. These changes may yield hemodynamic and biological advantages, leading to a decrease in the N-terminal brain natriuretic peptide (NT-proBNP) levels, a reliable marker of HF severity. Notably, ARNIs and BNPs are expected to increase but NT-proBNP levels decreases. Neprilysin exhibits a higher affinity for atrial-type natriuretic peptide (ANP) than BNP, resulting in a more consistent and robust increase in ANP levels [46,48].
Neprilysin has a higher affinity for atrial-type natriuretic peptide (ANP) than BNP; the ANP level is increased more consistently and robustly [39]. It may be that ANP or, indeed, other neprilysin substrates (e.g., C-type natriuretic peptide, urodilatin, bradykinin, adrenomedullin, substance P, vasointestinal peptides, calcitonin gene-related peptide, glucagon-like peptide-1, and apelin) plays a predominant role in the mechanism of action of ARNIs [49,50,51]. The ANP significantly decreases sodium reabsorption in the proximal tubule by activating the NPR-A/cGMP/PKG pathway, thus increasing sodium delivery to the distal renal tubules and natriuresis [52]. ARNIs have been shown to improve in bi-ventricular failure, and experimental models of RV dysfunction due to PH [36,40,53,54,55]. Moreover, ARNIs may cause reverse remodeling of the RV by its properties of being antiapoptotic, antifibrotic, anti-inflammatory, and antithrombotic, as well as its reendothelization and modulation of gene expression, such as transferring growth factor β1 [56,57] (Figure 2).
Certain natriuretic peptides and transcription factors were upregulated in the LV, which could be linked to the mechanisms of action of ARNIs. These findings suggest that targeting these pathways has differential effects on the RV. Emerging biomarkers for RV maladaptation in pulmonary hypertension are long non-coding RNA H19, SPARC-like protein 1 (SPARCL1), and cartilage intermediate layer protein 1 (CILP1) [58]. These biomarkers can provide valuable insights into the state of the RV, especially in the context of pulmonary hypertension and other conditions that may lead to RV failure. It is important to note that while some biomarkers may offer RV-specific information, a comprehensive assessment often requires a combination of clinical evaluation, imaging, and biomarker analysis.

7. Mechanism of Action of SGLT2is on RV Function

Sodium-glucose cotransporters (SGLT) are transmembrane proteins that facilitate sodium and glucose transport across cell membranes. The two most well-known members of the SGLT family are SGLT1 and SGLT2, members of the solute carrier 5A (SLC5A) gene family [59]. SGLT2, primarily found in renal epithelial cells and occasionally in cardiac myocytes, especially under hyperglycemic conditions [60], plays a significant role in these processes. SGLT2 inhibitors (SGLT2is), such as Dapagliflozin and Empagliflozin, exhibit a higher affinity for SGLT2 over SGLT1, influencing their therapeutic effects [61].
SGLT2is’ direct action on cardiac myocytes involve modulating the nutrient signaling pathways, leading to the increased activity of enzymes like AMPK and SIRT1, which promote cellular health and energy efficiency [62]. This results in reduced oxidative stress, endoplasmic reticulum stress, the restoration of mitochondrial health, enhanced mitochondrial biogenesis, decreased proinflammatory and profibrotic pathways, and ultimately, preserved cardiac and renal integrity (Figure 1). SGLT-2i promotes sodium and glucose excretion by inhibiting SGLT2 in the proximal renal tubule and activates Na+/K+ ATPase of the epithelial tubular cell in the kidney, leading to natriuresis and glucosuria [63]. SGLT2is differ from traditional diuretics because it does not activate the neurohormonal axis or renal sympathetic nervous system, which can exacerbate LV remodeling and worsen HF [64].
Moreover, SGLT2is also have anti-inflammatory effects and decrease meta-inflammation [65], which is described as a slow chronic inflammatory state that leads to systemic microvascular changes that lead to widespread endothelial dysfunction in cardiac myocytes by suppressing the secretion of ICAM-1, VCAM-1, TNF-α, IL-β, and IL-6 [66].
Furthermore, SGLT2is block the sodium/hydrogen exchanger 1 cotransporter at the cardiomyocyte level, triggering a signaling cascade that decreases myofilament stiffness, hypertrophy, fibrosis, and free radical production, thus protecting cardiac myocytes [67,68,69]. Their anti-inflammatory properties further contribute to reducing systemic microvascular changes and endothelial dysfunction [59].
Clinical trials have underscored the importance of SGLT2is in managing HF and chronic renal disease, irrespective of diabetes status [70,71]. Empagliflozin reduced PAP, and the reduction was amplified over time, independent of diuretics in a randomized control trial [72].
Potential biomarkers are useful in the diagnosis and prognosis of RV dysfunction in HF, and these biomarkers can offer future novel therapeutic avenues for improving the RV function alongside improving the LV [25,54]. Furthermore, the study by Frisk et al. [73] provides valuable insights into the transcriptomic differences between the LV and RV in patients with varying degrees of HF.
MicroRNAs (miRNAs) and Insulin-like Growth Factor (IGF) are two emerging biomarkers that have shown the potential to help with HF diagnosis and prognosis. Ongoing research has unearthed various potential biomolecules, including cardiac troponins (cTn), galectin-3, soluble Suppression of Tumorigenesis-2 (ST2), and growth differentiation factor 15 [74].
These biomarkers can be used alongside traditional measures to provide a more comprehensive assessment of HF, particularly in RV dysfunction. They offer a window into the underlying pathophysiological processes and could guide more personalized treatment strategies. There are several ongoing trials, and the trials are looking at the following five major issues targeting RV failure alongside LV failure (Table 1).
Furthermore, the potential roles of genetic and epigenetic factors in RV failure are complex and multifaceted. Here is a summary of the current understanding.
Genetic Factors: The RV and left ventricle (LV) have different embryologic origins, likely influencing their chamber-specific responses to physiological and pathological stress [79]. The variations in the genes involved in hypoxia signaling, metabolic regulation, and neurohormonal regulation can affect the RV’s response to hypoxia, pressure or volume overload, and surgical injury [17]. There is evidence of a recapitulation of the fetal gene pattern in dysfunctional or failing RV myocardium, with changes in the expression of myosin heavy chain genes [14].
Epigenetic Factors: DNA methylations and DNA methyl transferase (DNMT) are crucial in regulating angiogenesis, which is essential for RV function. Aberrant methylation patterns can lead to suppressed angiogenesis and contribute to RV failure. Specific microRNAs, such as miR-126, are important regulators of angiogenesis [14]. Decreased levels of miR-126 have been associated with reduced microvessel density in the RV, contributing to its failure. Histone modifications can also affect the gene expression related to RV development and function [13].
These genetic and epigenetic factors contribute to the pathogenesis of RV failure by affecting the cellular and molecular pathways that regulate the structure and function of the RV. Ongoing research continues to unravel the complexities of these contributions, which may lead to novel therapeutic approaches for RV failure.

8. Role of ARNIs or SGLT2is in Primary RV Dysfunction

The specific mechanisms of ARNIs and SGLT2is in RV cardiomyopathies, such as Arrhythmogenic RV Cardiomyopathy (ARVC), still need to be fully established. ARVC, characterized by impaired desmosome function, often leads to decreased RV contractility and failure as arrhythmias progress [80]. Notably, Matiz et al. reported symptomatic and imaging-proven RV recovery in an ARVC patient treated with ARNIs [81]. Emerging evidence suggests ARNIs’ potential antiarrhythmic effects in clinical and experimental settings [82]. Additionally, Dapagliflozin has shown promise in attenuating cardiac fibrosis and inflammation, potentially benefiting RV function indirectly [83]. Dapagliflozin reduces the vulnerability to PH-induced RV dysfunction by restoring calcium handling in rat models [84,85]. Studies have also indicated SGLT2is’ role in lowering arrhythmia risks, particularly in HFrEF patients, with the long-term follow-up showing significant improvements [86]. Many studies show the benefits of SGLT2i in transthyretin and diabetic cardiomyopathies [87,88,89,90].

9. Role of ARNIs or SGLT2is in RV Dysfunction Due to Volume Overload

The distinction between RV dysfunction due to volume versus pressure overload has yet to be fully understood. By dilating, the RV adapts to volume overload, such as tricuspid regurgitation, but chronic overload can lead to dysfunction [91]. While direct studies on ARNIs and SGLT2is’ impact on volume overload are scarce, ARNIs’ dual inhibition of neprilysin and AT2R may benefit RV function in HF patients with volume overload [92]. For conditions like tetralogy of Fallot and Ebstein’s anomaly, ARNIs could help preserve the size and function of the RV [93,94]. Observational studies suggest a link between ARNIs and RV dilatation and function improvements in HFrEF patients [36,40,55,95,96]. Similarly, SGLT2is have improved RV dysfunction, independent of LV remodeling [38,39,97]. Experimental models and small clinical trials hint at the potential benefits of SGLT2is on RV function, paving the way for future research in this area [98,99,100].

10. Role of ARNIs or SGLT2is in RV Dysfunction Due to Pressure Overload

RV dysfunction due to pressure overload, such as in PH, prompts an initial adaptive response of increased contractility and hypertrophy [91]. However, this compensatory mechanism often becomes inadequate as the pressure load escalates, leading to RV failure—the end-stage manifestation of PH [6]. Traditional pulmonary vasodilators are the cornerstone of therapy, particularly for Group 1 PH patients, as they alleviate RV afterload. Yet, not all patients, especially those with PH and HFrEF, benefit optimally from drugs like prostaglandins, endothelin receptor antagonists, or phosphodiesterase inhibitors [101].
The 2015 AHA consensus statement was conservative, recommending ARNIs and other neurohormonal therapies only for RV failure secondary to PH or left heart disease if accompanied by hypertension or coronary artery disease [91]. However, emerging evidence suggests a shift in this paradigm. Recent trials have demonstrated that SGLT2 inhibitors, when combined with standard HF medications, enhance RV function more effectively than PH drugs alone [38,39,102,103,104,105]. This is corroborated by studies indicating that adding SGLT2is to PH therapy improves RV function [105,106].
Experimental research, such as the work by Connelly et al. [107], has shown that Dapagliflozin mitigates the structural, functional, and molecular effects of RV pressure overload, evidenced by reduced remodeling in PA banding-induced models or monocrotaline (MCT)-treated PH in rats [108]. Similarly, ARNIs have been observed to decrease PH, reverse vascular remodeling, and reduce RV hypertrophy and dilatation in preclinical rat studies, suggesting its potential utility in treating PH and RV dysfunction [109,110,111].
Whether early intervention with these therapies can prevent RV failure as a result of a progression in pressure and volume overload, ongoing research and future studies will be pivotal in the answer and in shaping the therapeutic approach to RV dysfunction due to pressure overload.

11. Role of ARNIs or SGLT2is in Systemic RV Dysfunction

Systemic right ventricle (sRV) dysfunction, as seen in transposition of the great arteries (d-TGA) after the Senning or Mustard procedures, the single RV Fontan procedure, and congenitally corrected transposition of the great arteries (ccTGA), poses a high risk of maladaptation to the systemic cardiac output. This can lead to progressive enlargement, dysfunction, and worsening tricuspid regurgitation (TR), along with atrial and ventricular arrhythmias [112,113]. The gene expression in the sRV differs due to its unique functional demands, affecting contractile proteins, ion channels, and remodeling factor [12]. The mechanisms contributing to sRV dysfunction include a compromised coronary flow reserve, perfusion defects, myocardial fibrosis, TR, mechanical dyssynchrony, and arrhythmias [114]. Although ARNIs and SGLT2is are not standard treatments for sRV, their cardiovascular benefits could improve the function and prevent myocardial damage [115]. Studies have shown that ARNIs [116,117,118,119,120,121,122,123,124] and SGLT2is [99,125,126,127,128,129,130,131] have positive effects on cardiac remodeling, inflammation, and fibrosis in both pediatric and adult patients with congenital heart disease (ACHD) and sRV or single RV Fontan. The impact of quadruple combination therapy on reverse remodeling and sRV failure warrants further investigation.

12. Role of Combined ARNI and SGLT2i Therapy in RV Dysfunction

The mechanism of action of the combined use of ARNIs and SGLT2is in HFrEF or HFpEF has been synergistic and promising for bi-ventricular failure (Figure 2). Meta-analyses have highlighted the superiority of this combination over individual therapies in reducing cardiovascular death and all-cause mortality, albeit with an increased risk of volume depletion [132,133]. Direct comparison trials are needed to determine if one therapy significantly outperforms the other in reducing HF risk and preserving RV function. Some evidence suggests a preference for SGLT2is due to their renal protective benefits in diabetic patients, with effects on myocardial infarction, ischemic stroke, and heart remodeling comparable to ARNIs [134]. A meta-analysis indicates that a combination of SGLT2is, ARNIs, BBs, and MRAs is highly effective in reducing cardiovascular outcomes in HFrEF patients [135]. Such combination therapy could improve the RV function in both HFrEF and HFpEF patients. However, high medication costs and limited evidence often limit the simultaneous initiation of both drugs.
The treatment landscape for HF is rapidly evolving, with ongoing research and clinical trials heralding a new era of innovative therapies. Looking ahead, a plethora of promising pharmacological agents are on the horizon for treating aHFrEF in adults. Currently, there are 119 trials underway exploring treatments for HF. A suite of novel drugs at the forefront of HF therapy is emerging. Yet, their role in RV failure remains to be explored, primarily due to a shortage of clinical trials that focus specifically on RV failure. Among these potential new treatments are Omecamtiv mecarbil, Serelaxin, Istaroxime, Ularitide, Elamipretide, Finerenone, APD418 (a β3-Adrenergic Receptor Antagonist), IONIS-AGT-LRx, and JK07, among others. As the field advances, these agents are poised to revolutionize the management of RV failure in adults. Notably, Vericiguat and Omecamtiv have been utilized based on clinical experience and consensus statements for treating RV failure.
Limitations of current GDMT for HF to treat RV failure: The current GDMT [1,2] for HF primarily focuses on LV dysfunction, and there are several drawbacks when applying these guidelines to treat RV failure. There is a lack of clinical trials specifically targeting RV failure [41]. This means that the efficacy and safety of these therapies for RV failure need to be well established. RV failure has a different pathophysiology compared to LV failure. For instance, RV failure is often associated with PH, which is not the primary concern in LV failure [58]. Therefore, the treatments that are effective for LV failure may not be directly applicable or beneficial for RV failure. Certain medications used in GDMT, such as beta-blockers, can worsen RV function due to their negative inotropic effects, especially in patients with pre-existing RV dysfunction. The patients with significant RV dysfunction are often underrepresented in HF clinical trials, leading to a gap in knowledge about the optimal management of RV failure. RV failure usually requires a more complex management strategy that addresses the underlying cause, such as PH. It may involve targeted therapies that are not part of the standard GDMT [41]. The biomarkers and clinical endpoints used to monitor LV failure may be less effective for monitoring RV failure, necessitating the development of RV-specific markers and endpoints [74]. These drawbacks highlight the need for more RV-specific research and new guidelines that address the unique aspects of RV failure management.
Future directions: The future direction for the treatment of RV failure is multifaceted, focusing on medical and mechanical interventions tailored to the unique physiology of the RV [41]. It is important to enhance the timely identification of RV failure through advanced diagnostic tools like magnetic resonance and radionuclide imaging. These tools are crucial for assessing RV function and guiding treatment. The medical treatment of RV failure typically involves two approaches: perfusion pressure restoration and myocardial contractility improvement [76]. Future therapies may include novel pharmacological agents targeting the molecular pathways involved in RV dysfunction [73]. The use of mechanical assist devices, such as mechanical circulatory support (MCS), is being explored for managing acute RV failure, especially where there is insufficient pharmacotherapy. These devices can improve hemodynamics and support a failing RV [75,77]. In cases where recovery is not observed with medical management or mechanical support, heart transplantation remains a definitive treatment option. Future strategies may improve the selection and management of candidates for transplantation.

13. Conclusions

This review underscores the complex interplay of ventricular function within the HF continuum. An RV failure when it occurs alongside an LV failure, whether the RV failure is a separate entity or due to an LV failure, requires further clarification. Pending LV- or RV-specific therapies, the early initiation of ARNIs and SGLT2is in line with GDMT for eligible HFrEF and HFpEF patients is vital to address RV dysfunction. Large-scale, multicenter randomized controlled trials are imperative to ascertain the efficacy and safety of ARNIs/SGLT2is and to determine whether an improvement in the RV function is independent of the LV function enhancement. RV dysfunction merits a focus in future HF clinical trials, representing a significant gap in current management strategies. Incorporating the RV function into risk stratification and trial endpoints could optimize patient outcomes, particularly in selecting HFrEF candidates for LVAD and tailoring individualized drug therapies.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Heidenreich, P.A.; Bozkurt, B.; Aguilar, D.; Allen, L.A.; Byun, J.J.; Colvin, M.M.; Deswal, A.; Drazner, M.H.; Dunlay, S.M.; Evers, L.R.; et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2022, 145, E895–E1032. [Google Scholar] [CrossRef] [PubMed]
  2. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2023 Focused Update of the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. J. Heart Fail. 2024, 26, 5–17. [Google Scholar] [CrossRef] [PubMed]
  3. Martin, S.S.; Aday, A.W.; Almarzooq, Z.I.; Anderson, C.A.M.; Arora, P.; Avery, C.L.; Baker-Smith, C.M.; Barone Gibbs, B.; Beaton, A.Z.; Boehme, A.K.; et al. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024, 149, e347–e913. [Google Scholar] [CrossRef]
  4. De Groote, P.; Millaire, A.; Foucher-Hossein, C.; Nugue, O.; Marchandise, X.; Ducloux, G.; Lablanche, J.-M. Right ventricular ejection fraction is an independent predictor of survival in patients with moderate heart failure. J. Am. Coll. Cardiol. 1998, 32, 948–954. [Google Scholar] [CrossRef] [PubMed]
  5. 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. Eur. J. Heart Fail. 2021, 23, 352–380. [Google Scholar] [CrossRef] [PubMed]
  6. Houston, B.A.; Taichman, D.B.; Brittain, E.L.; Tedford, R.J. Right Ventricular Failure. N. Engl. J. Med. 2023, 388, 1111–1125. [Google Scholar] [CrossRef] [PubMed]
  7. Dini, F.L.; Carluccio, E.; Simioniuc, A.; Biagioli, P.; Reboldi, G.; Galeotti, G.G.; Raineri, C.; Gargani, L.; Scelsi, L.; Mandoli, G.E.; et al. Right ventricular recovery during follow-up is associated with improved survival in patients with chronic heart failure with reduced ejection fraction. Eur. J. Heart Fail. 2016, 18, 1462–1471. [Google Scholar] [CrossRef] [PubMed]
  8. Ghio, S.; Gavazzi, A.; Campana, C.; Inserra, C.; Klersy, C.; Sebastiani, R.; Arbustini, E.; Recusani, F.; Tavazzi, L. Independent and additive prognostic value of right ventricular systolic function and pulmonary artery pressure in patients with chronic heart failure. J. Am. Coll. Cardiol. 2001, 37, 183–188. [Google Scholar] [CrossRef]
  9. Arora, S.; Patel, P.; Lahewala, S.; Patel, N.; Patel, N.J.; Thakore, K.; Amin, A.; Tripathi, B.; Kumar, V.; Shah, H.; et al. Etiologies, Trends, and Predictors of 30-Day Readmission in Patients With Heart Failure. Am. J. Cardiol. 2017, 119, 760–769. [Google Scholar] [CrossRef]
  10. Halliday, B.P.; Owen, R.; Gregson, J.; Vassiliou, V.S.; Chen, X.; Wage, R.; Lota, A.S.; Khalique, Z.; Tayal, U.; Hammersley, D.J.; et al. Myocardial remodelling after withdrawing therapy for heart failure in patients with recovered dilated cardiomyopathy: Insights from TRED-HF. Eur. J. Heart Fail. 2021, 23, 293–301. [Google Scholar] [CrossRef]
  11. Friedberg, M.K.; Redington, A.N. Right Versus Left Ventricular Failure. Circulation 2014, 129, 1033–1044. [Google Scholar] [CrossRef] [PubMed]
  12. Taverne, Y.J.H.J.; Sadeghi, A.; Bartelds, B.; Bogers, A.J.J.C.; Merkus, D. Right ventricular phenotype, function, and failure: A journey from evolution to clinics. Heart Fail. Rev. 2021, 26, 1447–1466. [Google Scholar] [CrossRef] [PubMed]
  13. Mittal, S. Genetic Variation and Outcomes in Right Ventricular Congenital Heart Disease. In Right Ventricular Physiology, Adaptation and Failure in Congenital and Acquired Heart Disease; Friedberg, M., Redington, A., Eds.; Springer: Cham, Switzerland, 2018. [Google Scholar] [CrossRef]
  14. Reddy, S.; Zhao, M.; Hu, D.Q.; Fajardo, G.; Hu, S.; Ghosh, Z.; Rajagopalan, V.; Wu, J.C.; Bernstein, D. Dynamic microRNA expression during the transition from right ventricular hypertrophy to failure. Physiol Genom. 2012, 44, 562–575. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, G.Y.; McCloskey, D.T.; Turcato, S.; Swigart, P.M.; Simpson, P.C.; Baker, A.J. Contrasting inotropic responses to alpha1-adrenergic receptor stimulation in left versus right ventricular myocardium. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H2013–H2017. [Google Scholar] [CrossRef] [PubMed]
  16. Nagendran, J.; Archer, S.L.; Soliman, D.; Gurtu, V.; Moudgil, R.; Haromy, A.; St. Aubin, C.; Webster, L.; Rebeyka, I.M.; Ross, D.B.; et al. Phosphodiesterase Type 5 Is Highly Expressed in the Hypertrophied Human Right Ventricle, and Acute Inhibition of Phosphodiesterase Type 5 Improves Contractility. Circulation 2007, 116, 238–248. [Google Scholar] [CrossRef]
  17. Modesti, P.A.; Vanni, S.; Bertolozzi, I.; Cecioni, I.; Lumachi, C.; Perna, A.M.; Boddi, M.; Gensini, G.F. Different Growth Factor Activation in the Right and Left Ventricles in Experimental Volume Overload. Hypertension 2004, 43, 101–108. [Google Scholar] [CrossRef]
  18. Havlenova, T.; Skaroupkova, P.; Miklovic, M.; Behounek, M.; Chmel, M.; Jarkovska, D.; Sviglerova, J.; Stengl, M.; Kolar, M.; Novotny, J.; et al. Right versus left ventricular remodeling in heart failure due to chronic volume overload. Sci. Rep. 2021, 11, 17136. [Google Scholar] [CrossRef] [PubMed]
  19. Bosch, L.; Lam, C.S.P.; Gong, L.; Chan, S.P.; Sim, D.; Yeo, D.; Jaufeerally, F.; Leong, K.T.G.; Ong, H.Y.; Ng, T.P.; et al. Right ventricular dysfunction in left-sided heart failure with preserved versus reduced ejection fraction. Eur. J. Heart Fail. 2017, 19, 1664–1671. [Google Scholar] [CrossRef] [PubMed]
  20. Sanders, J.L.; Koestenberger, M.; Rosenkranz, S.; Maron, B.A. Right ventricular dysfunction and long-term risk of death. Cardiovasc. Diagn. Ther. 2020, 10, 1646–1658. [Google Scholar] [CrossRef]
  21. Ghio, S.; Guazzi, M.; Scardovi, A.B.; Klersy, C.; Clemenza, F.; Carluccio, E.; Temporelli, P.L.; Rossi, A.; Faggiano, P.; Traversi, E.; et al. Different correlates but similar prognostic implications for right ventricular dysfunction in heart failure patients with reduced or preserved ejection fraction. Eur. J. Heart Fail. 2017, 19, 873–879. [Google Scholar] [CrossRef]
  22. Baandrup, J.D.; Markvardsen, L.H.; Peters, C.D.; Schou, U.K.; Jensen, J.L.; Magnusson, N.E.; Ørntoft, T.F.; Kruhøffer, M.; Simonsen, U. Pressure Load: The Main Factor for Altered Gene Expression in Right Ventricular Hypertrophy in Chronic Hypoxic Rats. PLoS ONE 2011, 6, e15859. [Google Scholar] [CrossRef] [PubMed]
  23. Solomon, S.D.; McMurray, J.J.V.; Anand, I.S.; Ge, J.; Lam, C.S.P.; Maggioni, A.P.; Martinez, F.; Packer, M.; Pfeffer, M.A.; Pieske, B.; et al. Angiotensin–Neprilysin Inhibition in Heart Failure with Preserved Ejection Fraction. N. Engl. J. Med. 2019, 381, 1609–1620. [Google Scholar] [CrossRef] [PubMed]
  24. Lu, H.; Inciardi, R.M.; Abanda, M.; Shah, A.M.; Cikes, M.; Claggett, B.; Prasad, N.; Lam, C.S.; Redfield, M.M.; McMurray, J.J.; et al. Abstract 13706: Determinants of Right Ventricular Dysfunction in Patients With Heart Failure With Preserved Ejection Fraction: The PARAGON-HF Trial Experience. Circulation 2023, 148, A13706. [Google Scholar] [CrossRef]
  25. Frisk, C.; Das, S.; Eriksson, M.J.; Walentinsson, A.; Corbascio, M.; Hage, C.; Kumar, C.; Ekström, M.; Maret, E.; Persson, H.; et al. Cardiac biopsies reveal differences in transcriptomics between left and right ventricle in patients with or without diagnostic signs of heart failure. Sci. Rep. 2024, 14, 5811. [Google Scholar] [CrossRef] [PubMed]
  26. Anastasiou, V.; Papazoglou, A.S.; Moysidis, D.V.; Daios, S.; Barmpagiannos, K.; Gossios, T.; Efthimiadis, G.K.; Karamitsos, T.; Ziakas, A.; Kamperidis, V. The prognostic impact of right ventricular-pulmonary arterial coupling in heart failure: A systematic review and meta-analysis. Heart Fail. Rev. 2023, 29, 13–26. [Google Scholar] [CrossRef]
  27. Brockmöller, J.; Ivanoski, S.; Hundack, L.; Blohm, J.-H.; Januszewski, M.; Remppis, B.A.; Burgdorf, C. Clinical and Hemodynamic Improvement in Pulmonary Hypertension After Switching to Sacubitril/Valsartan in Patients With Heart Failure With Preserved Ejection Fraction. J. Cardiovasc. Pharmacol. 2023, 82, 419–425. [Google Scholar] [CrossRef] [PubMed]
  28. Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Bocchi, E.; Böhm, M.; Brunner–La Rocca, H.-P.; Choi, D.-J.; Chopra, V.; Chuquiure-Valenzuela, E.; et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N. Engl. J. Med. 2021, 385, 1451–1461. [Google Scholar] [CrossRef] [PubMed]
  29. 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]
  30. Rodríguez, M.A.R.; Alonso, D.; Martín, J.M.; López-Benito, M.L.-B.; Garrote, C.; Olalla-Gómez, C.O.-G.; Iglesias-Garriz, I. Contribution of Right Ventricular Dysfunction to Heart Failure Mortality: A Meta-Analysis. Rev. Cardiovasc. Med. 2012, 13, 62–69. [Google Scholar] [CrossRef]
  31. Surkova, E.; Kovács, A.; Tokodi, M.; Lakatos, B.K.; Merkely, B.; Muraru, D.; Ruocco, A.; Parati, G.; Badano, L.P. Contraction Patterns of the Right Ventricle Associated with Different Degrees of Left Ventricular Systolic Dysfunction. Circ. Cardiovasc. Imaging 2021, 14, 982–994. [Google Scholar] [CrossRef]
  32. Tran, J.S.; Havakuk, O.; McLeod, J.M.; Hwang, J.; Kwong, H.Y.; Shavelle, D.; Zile, M.R.; Elkayam, U.; Fong, M.W.; Grazette, L.P. Acute pulmonary pressure change after transition to sacubitril/valsartan in patients with heart failure reduced ejection fraction. ESC Heart Fail. 2021, 8, 1706–1710. [Google Scholar] [CrossRef] [PubMed]
  33. Lampert, B.C.; Teuteberg, J.J. Right ventricular failure after left ventricular assist devices. J. Heart Lung Transplant. 2015, 34, 1123–1130. [Google Scholar] [CrossRef] [PubMed]
  34. Damy, T.; Kallvikbacka-Bennett, A.; Goode, K.; Khaleva, O.; Lewinter, C.; Hobkirk, J.; Nikitin, N.P.; Dubois-Randé, J.-L.; Hittinger, L.; Clark, A.L.; et al. Prevalence of, Associations With, and Prognostic Value of Tricuspid Annular Plane Systolic Excursion (TAPSE) Among Out-Patients Referred for the Evaluation of Heart Failure. J. Card. Fail. 2012, 18, 216–225. [Google Scholar] [CrossRef]
  35. Linde, C.; Ekström, M.; Eriksson, M.J.; Maret, E.; Wallén, H.; Lyngå, P.; Wedén, U.; Cabrera, C.; Löfström, U.; Stenudd, J.; et al. Baseline characteristics of 547 new onset heart failure patients in the PREFERS heart failure study. ESC Heart Fail. 2022, 9, 2125–2138. [Google Scholar] [CrossRef] [PubMed]
  36. Correale, M.; Mallardi, A.; Mazzeo, P.; Tricarico, L.; Diella, C.; Romano, V.; Ferraretti, A.; Leopizzi, A.; Merolla, G.; Di Biase, M.; et al. Sacubitril/valsartan improves right ventricular function in a real-life population of patients with chronic heart failure: The Daunia Heart Failure Registry. Int. J. Cardiol. Heart Vasc. 2020, 27, 100486. [Google Scholar] [CrossRef] [PubMed]
  37. Yenerçağ, M.; Arslan, U.; Dereli, S.; Çoksevim, M.; Doğduş, M.; Kaya, A. Effects of angiotensin receptor neprilysin inhibition on pulmonary arterial stiffness in heart failure with reduced ejection fraction. Int. J. Cardiovasc. Imaging 2021, 37, 165–173. [Google Scholar] [CrossRef] [PubMed]
  38. Mustapic, I.; Bakovic, D.; Susilovic Grabovac, Z.; Borovac, J.A. Impact of SGLT2 Inhibitor Therapy on Right Ventricular Function in Patients with Heart Failure and Reduced Ejection Fraction. J. Clin. Med. 2022, 12, 42. [Google Scholar] [CrossRef]
  39. Axelsen, J.S.; Nielsen-Kudsk, A.H.; Schwab, J.; Ringgaard, S.; Nielsen-Kudsk, J.E.; De Man, F.S.; Andersen, A.; Andersen, S. Effects of empagliflozin on right ventricular adaptation to pressure overload. Front. Cardiovasc. Med. 2023, 10, 1302265. [Google Scholar] [CrossRef] [PubMed]
  40. Melenovsky, V.; Hwang, S.-J.; Lin, G.; Redfield, M.M.; Borlaug, B.A. Right heart dysfunction in heart failure with preserved ejection fraction. Eur. Heart J. 2014, 35, 3452–3462. [Google Scholar] [CrossRef]
  41. D’amato, A.; Prosperi, S.; Severino, P.; Myftari, V.; Francia, A.L.; Cestiè, C.; Pierucci, N.; Marek-Iannucci, S.; Mariani, M.V.; Germanò, R.; et al. Current Approaches to Worsening Heart Failure: Pathophysiological and Molecular Insights. Int. J. Mol. Sci. 2024, 25, 1574. [Google Scholar] [CrossRef]
  42. Vardeny, O.; Claggett, B.; Packer, M.; Zile, M.R.; Rouleau, J.; Swedberg, K.; Teerlink, J.R.; Desai, A.S.; Lefkowitz, M.; Shi, V.; et al. Efficacy of sacubitril/valsartan vs. enalapril at lower than target doses in heart failure with reduced ejection fraction: The PARADIGM-HF trial. Eur. J. Heart Fail. 2016, 18, 1228–1234. [Google Scholar] [CrossRef] [PubMed]
  43. Hechter, S.J.; Fredriksen, P.M.; Liu, P.; Veldtman, G.; Merchant, N.; Freeman, M.; Therrien, J.; Benson, L.; Siu, S.; Webb, G. Angiotensin-converting enzyme inhibitors in adults after the Mustard procedure. Am. J. Cardiol. 2001, 87, 660–663. [Google Scholar] [CrossRef] [PubMed]
  44. Tutarel, O.; Meyer, G.P.; Bertram, H.; Wessel, A.; Schieffer, B.; Westhoff-Bleck, M. Safety and efficiency of chronic ACE inhibition in symptomatic heart failure patients with a systemic right ventricle. Int. J. Cardiol. 2012, 154, 14–16. [Google Scholar] [CrossRef] [PubMed]
  45. Lester, S.J.; McElhinney, D.B.; Viloria, E.; Reddy, G.P.; Ryan, E.; Tworetzky, W.; Schiller, N.B.; Foster, E. Effects of losartan in patients with a systemically functioning morphologic right ventricle after atrial repair of transposition of the great arteries. Am. J. Cardiol. 2001, 88, 1314–1316. [Google Scholar] [CrossRef] [PubMed]
  46. Januzzi, J.L.; Prescott, M.F.; Butler, J.; Felker, G.M.; Maisel, A.S.; McCague, K.; Camacho, A.; Piña, I.L.; Rocha, R.A.; Shah, A.M.; et al. Association of Change in N-Terminal Pro–B-Type Natriuretic Peptide Following Initiation of Sacubitril-Valsartan Treatment With Cardiac Structure and Function in Patients With Heart Failure With Reduced Ejection Fraction. JAMA 2019, 322, 1085–1095. [Google Scholar] [CrossRef] [PubMed]
  47. Packer, M.; McMurray, J.J.; Desai, A.S.; Gong, J.; Lefkowitz, M.P.; Rizkala, A.R.; Rouleau, J.L.; Shi, V.C.; Solomon, S.D.; Swedberg, K.; et al. Angiotensin receptor neprilysin inhibition compared with enalapril on the risk of clinical progression in surviving patients with heart failure. Circulation 2015, 131, 54–61. [Google Scholar] [CrossRef] [PubMed]
  48. Murphy, S.P.; Prescott, M.F.; Camacho, A.; Iyer, S.R.; Maisel, A.S.; Felker, G.M.; Butler, J.; Pina, I.L.; Ibrahim, N.E.; Abbas, C.; et al. Atrial Natriuretic Peptide and Treatment With Sacubitril/Valsartan in Heart Failure With Reduced Ejection Fraction. JACC Heart Fail 2021, 9, 127–136. [Google Scholar] [CrossRef] [PubMed]
  49. Zile, M.R.; Claggett, B.L.; Prescott, M.F.; McMurray, J.J.; Packer, M.; Rouleau, J.L.; Swedberg, K.; Desai, A.S.; Gong, J.; Shi, V.C.; et al. Prognostic Implications of Changes in N-Terminal Pro-B-Type Natriuretic Peptide in Patients With Heart Failure. J. Am. Coll. Cardiol. 2016, 68, 2425–2436. [Google Scholar] [CrossRef] [PubMed]
  50. 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]
  51. Pieske, B.; Wachter, R.; Shah, S.J.; Baldridge, A.; Szeczoedy, P.; Ibram, G.; Shi, V.; Zhao, Z.; Cowie, M.R.; Investigators, P.; et al. Effect of Sacubitril/Valsartan vs Standard Medical Therapies on Plasma NT-proBNP Concentration and Submaximal Exercise Capacity in Patients With Heart Failure and Preserved Ejection Fraction: The PARALLAX Randomized Clinical Trial. JAMA 2021, 326, 1919–1929. [Google Scholar] [CrossRef]
  52. Vives, D.; Farage, S.; Motta, R.; Lopes, A.G.; Caruso-Neves, C. Atrial natriuretic peptides and urodilatin modulate proximal tubule Na(+)-ATPase activity through activation of the NPR-A/cGMP/PKG pathway. Peptides 2010, 31, 903–908. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, J.; Du, L.; Qin, X.; Guo, X. Effect of Sacubitril/Valsartan on the Right Ventricular Function and Pulmonary Hypertension in Patients With Heart Failure With Reduced Ejection Fraction: A Systematic Review and Meta-Analysis of Observational Studies. J. Am. Heart Assoc. 2022, 11, e024449. [Google Scholar] [CrossRef]
  54. Alcidi, G.; Pugliese, R.; Ioannoni, S.; Romano, M.; Palmieri, G.; Tabella, E.; Correale, M.; Di Biase, M.; Brunetti, N.D.; Iacoviello, M. Improvement in Left and Right Ventricular Function after Introduction of SGLT2 Inhibitors in Heart Failure Outpatients with Reduced Ejection Fraction. Clin. Pract. 2023, 13, 1303–1312. [Google Scholar] [CrossRef] [PubMed]
  55. Imamura, T.; Hori, M.; Ueno, H.; Kinugawa, K. Impact of Sacubitril/Valsartan on Right Heart Failure. Int. Heart J. 2021, 62, 932–934. [Google Scholar] [CrossRef]
  56. Bazgir, F.; Nau, J.; Nakhaei-Rad, S.; Amin, E.; Wolf, M.J.; Saucerman, J.J.; Lorenz, K.; Ahmadian, M.R. The Microenvironment of the Pathogenesis of Cardiac Hypertrophy. Cells 2023, 12, 1780. [Google Scholar] [CrossRef]
  57. Winter, M.M.; Bouma, B.J.; Groenink, M.; Konings, T.C.; Tijssen, J.G.P.; Van Veldhuisen, D.J.; Mulder, B.J.M. Latest insights in therapeutic options for systemic right ventricular failure: A comparison with left ventricular failure. Heart 2009, 95, 960–963. [Google Scholar] [CrossRef] [PubMed]
  58. Llucià-Valldeperas, A.; van Wezenbeek, J.; Goumans, M.-J.; de Man, F.S. The battle of new biomarkers for right heart failure in pulmonary hypertension: Is the queen of hearts NT-proBNP defeated at last? Eur. Respir. J. 2021, 57, 2004277. [Google Scholar] [CrossRef]
  59. Sano, R.; Shinozaki, Y.; Ohta, T. Sodium–glucose cotransporters: Functional properties and pharmaceutical potential. J. Diabetes Investig. 2020, 11, 770–782. [Google Scholar] [CrossRef]
  60. Quagliariello, V.; De Laurentiis, M.; Rea, D.; Barbieri, A.; Monti, M.G.; Carbone, A.; Paccone, A.; Altucci, L.; Conte, M.; Canale, M.L.; et al. The SGLT-2 inhibitor empagliflozin improves myocardial strain, reduces cardiac fibrosis and pro-inflammatory cytokines in non-diabetic mice treated with doxorubicin. Cardiovasc. Diabetol. 2021, 20, 150. [Google Scholar] [CrossRef]
  61. Pawlos, A.; Broncel, M.; Woźniak, E.; Gorzelak-Pabiś, P. Neuroprotective Effect of SGLT2 Inhibitors. Molecules 2021, 26, 7213. [Google Scholar] [CrossRef]
  62. Packer, M. Critical Reanalysis of the Mechanisms Underlying the Cardiorenal Benefits of SGLT2 Inhibitors and Reaffirmation of the Nutrient Deprivation Signaling/Autophagy Hypothesis. Circulation 2022, 146, 1383–1405. [Google Scholar] [CrossRef] [PubMed]
  63. Zelniker, T.A.; Braunwald, E. Mechanisms of Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors. J. Am. Coll. Cardiol. 2020, 75, 422–434. [Google Scholar] [CrossRef]
  64. Wilcox, C.S. Antihypertensive and Renal Mechanisms of SGLT2 (Sodium-Glucose Linked Transporter 2) Inhibitors. Hypertension 2020, 75, 894–901. [Google Scholar] [CrossRef]
  65. Schiattarella, G.G.; Rodolico, D.; Hill, J.A. Metabolic inflammation in heart failure with preserved ejection fraction. Cardiovasc. Res. 2021, 117, 423–434. [Google Scholar] [CrossRef]
  66. Giannattasio, S.; Citarella, A.; Trocchianesi, S.; Filardi, T.; Morano, S.; Lenzi, A.; Ferretti, E.; Crescioli, C. Cell-Target-Specific Anti-Inflammatory Effect of Empagliflozin: In Vitro Evidence in Human Cardiomyocytes. Front. Mol. Biosci. 2022, 9, 879522. [Google Scholar] [CrossRef]
  67. Benes, J.; Kotrc, M.; Wohlfahrt, P.; Kroupova, K.; Tupy, M.; Kautzner, J.; Melenovsky, V. Right ventricular global dysfunction score: A new concept of right ventricular function assessment in patients with heart failure with reduced ejection fraction (HFrEF). Front. Cardiovasc. Med. 2023, 10, 1194174. [Google Scholar] [CrossRef] [PubMed]
  68. Iborra-Egea, O.; Santiago-Vacas, E.; Yurista, S.R.; Lupón, J.; Packer, M.; Heymans, S.; Zannad, F.; Butler, J.; Pascual-Figal, D.; Lax, A.; et al. Unraveling the Molecular Mechanism of Action of Empagliflozin in Heart Failure With Reduced Ejection Fraction With or Without Diabetes. JACC Basic Transl. Sci. 2019, 4, 831–840. [Google Scholar] [CrossRef] [PubMed]
  69. Xie, Y.; Wei, Y.; Li, D.; Pu, J.; Ding, H.; Zhang, X. Mechanisms of SGLT2 Inhibitors in Heart Failure and Their Clinical Value. J. Cardiovasc. Pharmacol. 2023, 81, 4–14. [Google Scholar] [CrossRef] [PubMed]
  70. 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]
  71. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef]
  72. Nassif, M.E.; Qintar, M.; Windsor, S.L.; Jermyn, R.; Shavelle, D.M.; Tang, F.; Lamba, S.; Bhatt, K.; Brush, J.; Civitello, A.; et al. Empagliflozin Effects on Pulmonary Artery Pressure in Patients With Heart Failure. Circulation 2021, 143, 1673–1686. [Google Scholar] [CrossRef]
  73. Gorr, M.W.; Sriram, K.; Chinn, A.M.; Muthusamy, A.; Insel, P.A. Transcriptomic profile reveals differences etween the right and left ventricle in normoxia and hypoxia. Ohysiol. Rep. 2020, 8, e14344. [Google Scholar] [CrossRef]
  74. Alzaabi, M.A.; Abdelsalam, A.; Alhamaadi, M.; Hani, H.B.; Almheiri, A.; Matrooshi, N.A.; Al Zaman, K. Evaluating biomarkers as tools for early detection and prognosis of heart failure: A comprehensive review. Card. Fail. Rev. 2024, 10, e06. [Google Scholar] [CrossRef] [PubMed]
  75. Salam, M.F.; Gorgis, S.; Basir, M.B. Current and Emerging Strategies for RV Shock Management in the Setting of RV Infarct. JACC. Available online: https://www.acc.org/latest-in-cardiology/articles/2021/11/01/12/41/current-and-emerging-strategies-for-rv-shock-management (accessed on 26 June 2024).
  76. Mansoor, T.; Khalid, S.N.; Bilal, M.I.; Ijaz, S.H.; Fudim, M.; Greene, S.J.; Warraich, H.J.; Nambi, V.; Virani, S.S.; Fonarow, G.C.; et al. Ongoing and Future Clinical Trials of Pharmacotherapy for Heart Failure. Am. J. Cardiovasc. Drugs 2024. [Google Scholar] [CrossRef] [PubMed]
  77. Yuriditsky, E.; Chonde, M.; Friedman, O.; Horowitz, J.M. Medical and Mechanical Circulatory Support of the Failing Right Ventricle. Curr. Cardiol. Rep. 2024, 26, 23–34. [Google Scholar] [CrossRef] [PubMed]
  78. Diaz, J.C.; Duque, M.; Aristizabal, J.; Marin, J.; Niño, C.; Bastidas, O.; Ruiz, L.M.; Matos, C.D.; Hoyos, C.; Hincapie, D.; et al. The Emerging Role of Left Bundle Branch Area Pacing for Cardiac Resynchronisation Therapy. Arrhythmia Electrophysiol. Rev. 2023, 12, e29. [Google Scholar] [CrossRef]
  79. Lippmann, M.R.; Maron, B.A. The Right Ventricle: From Embryologic Development to RV Failure. Curr. Heart Fail. Rep. 2022, 19, 325–333. [Google Scholar] [CrossRef]
  80. Chang, P.C.; Lin, S.F.; Chu, Y.; Wo, H.T.; Lee, H.L.; Huang, Y.C.; Wen, M.S.; Chou, C.C. LCZ696 Therapy Reduces Ventricular Tachyarrhythmia Inducibility in a Myocardial Infarction-Induced heart failure rat model. Cardiovasc Ther. 2019, 2019, 6032631. [Google Scholar] [CrossRef]
  81. Maitz, T.N.; Gupta, R.; Persin, K.; Sundlof, D.W. Utilization of sacubitril-valsartan for right ventricular failure in a patient with arrhythmogenic right ventricular cardiomyopathy. Future Cardiol. 2022, 18, 547–552. [Google Scholar] [CrossRef]
  82. Zhou, Y.; Rui, S.; Tang, S.; Ju, C. Exploration of Mechanisms of Sacubitril/Valsartan in the Treatment of Cardiac Arrhythmias Using a Network Pharmacology Approach. Front. Cardiovasc. Med. 2022, 9, 829484. [Google Scholar] [CrossRef]
  83. Yang, Z.; Li, T.; Xian, J.; Chen, J.; Huang, Y.; Zhang, Q.; Lin, X.; Lu, H.; Lin, Y. SGLT2 inhibitor dapagliflozin attenuates cardiac fibrosis and inflammation by reverting the HIF-2α signaling pathway in arrhythmogenic cardiomyopathy. FASEB J. 2022, 36, e22410. [Google Scholar] [CrossRef] [PubMed]
  84. Qin, T.; Kong, B.; Dai, C.; Xiao, Z.; Fang, J.; Shuai, W.; Huang, H. Protective effects of Dapagliflozin on the vulnerability of ventricular arrhythmia in rats with pulmonary artery hypertension induced by monocrotaline. Bioengineered 2022, 13, 2697–2709. [Google Scholar] [CrossRef] [PubMed]
  85. Wu, J.; Liu, T.; Shi, S.; Fan, Z.; Hiram, R.; Xiong, F.; Cui, B.; Su, X.; Chang, R.; Zhang, W.; et al. Dapagliflozin reduces the vulnerability of rats with pulmonary arterial hypertension-induced right heart failure to ventricular arrhythmia by restoring calcium handling. Cardiovasc. Diabetol. 2022, 21, 197. [Google Scholar] [CrossRef] [PubMed]
  86. Liao, J.; Ebrahimi, R.; Ling, Z.; Meyer, C.; Martinek, M.; Sommer, P.; Futyma, P.; Di Vece, D.; Schratter, A.; Acou, W.-J.; et al. Effect of SGLT-2 inhibitors on arrhythmia events: Insight from an updated secondary analysis of > 80,000 patients (the SGLT2i—Arrhythmias and Sudden Cardiac Death). Cardiovasc. Diabetol. 2024, 23, 78. [Google Scholar] [CrossRef] [PubMed]
  87. Gonzalez-Montes, M.; Maccallini, M.; Barge-Caballero, G.; Enriquez-Vazquez, D.; Barge-Caballero, E.; Crespo-Leiro, M.G. Use of SGLT2i in patients with transthyretin amyloid cardiomyopathy: Prevalence and safety in a Spanish prospective cohort. Rev. Esp. Cardiol. 2024, 77, 349–352. [Google Scholar] [CrossRef] [PubMed]
  88. Lang, F.M.; Teruya, S.; Weinsaft, A.; Cuomo, M.; Santos, A.M.; Nalbandian, A.; Bampatsias, D.; Maurer, M.S. Sodium-glucose cotransporter 2 inhibitors for transthyretin amyloid cardiomyopathy: Analyses of short-term efficacy and safety. Eur. J. Heart Fail. 2024, 26, 938–947. [Google Scholar] [CrossRef]
  89. Li, N.; Zhou, H. SGLT2 Inhibitors: A Novel Player in the Treatment and Prevention of Diabetic Cardiomyopathy. Drug Des. Dev. Ther. 2020, 14, 4775–4788. [Google Scholar] [CrossRef] [PubMed]
  90. Patoulias, D.; Katsimardou, A.; Toumpourleka, M.; Papadopoulos, C.; Doumas, M. Time to assess the effects of sodium–glucose co-transporter-2 inhibitors on the ‘forgotten’ right ventricle? ESC Heart Fail. 2019, 7, 334–335. [Google Scholar] [CrossRef] [PubMed]
  91. Lopez, L.; Cohen, M.S.; Anderson, R.H.; Redington, A.N.; Nykanen, D.G.; Penny, D.J.; Deanfield, J.E.; Eidem, B.W. Unnatural history of the right ventricle in patients with congenitally malformed hearts. Cardiol. Young 2010, 20 (Suppl. 3), 107–112. [Google Scholar] [CrossRef]
  92. Xu, Y.; Zhang, M.; Chen, Y.; Chen, X.; Song, W.; Zhu, L.; Liu, L.; Gong, X.; Zhang, Y.; Xu, Z. Does sacubitril/valsartan work in children with heart failure?—A pilot study. Front. Cardiovasc. Med. 2023, 10, 1274990. [Google Scholar] [CrossRef]
  93. Stout, K.K.; Broberg, C.S.; Book, W.M.; Cecchin, F.; Chen, J.M.; Dimopoulos, K.; Everitt, M.D.; Gatzoulis, M.; Harris, L.; Hsu, D.T.; et al. Chronic Heart Failure in Congenital Heart Disease. Circulation 2016, 133, 770–801. [Google Scholar] [CrossRef] [PubMed]
  94. Thandavarayan, R.A.; Chitturi, K.R.; Guha, A. Pathophysiology of Acute and Chronic Right Heart Failure. Cardiol. Clin. 2020, 38, 149–160. [Google Scholar] [CrossRef]
  95. Ciepłucha, A.; Trojnarska, O.; Kociemba, A.; Łanocha, M.; Barczynski, M.; Rozmiarek, S.; Kramer, L.; Pyda, M. Clinical aspects of myocardial fibrosis in adults with Ebstein’s anomaly. Heart Vessels 2018, 33, 1076–1085. [Google Scholar] [CrossRef] [PubMed]
  96. Polito, M.V.; Silverio, A.; Rispoli, A.; Vitulano, G.; Auria, F.D.; De Angelis, E.; Loria, F.; Gigantino, A.; Bonadies, D.; Citro, R.; et al. Clinical and echocardiographic benefit of Sacubitril/Valsartan in a real-world population with HF with reduced ejection fraction. Sci. Rep. 2020, 10, 6665. [Google Scholar] [CrossRef] [PubMed]
  97. Fusco, F.; Scognamiglio, G.; Sorice, D.; Abbate, M.; Altobelli, I.; Sarubbi, B. Biventricular performance in adults with a systemic right ventricle: New insights from myocardial work analysis. Int. J. Cardiovasc. Imaging 2024, 40, 1067–1079. [Google Scholar] [CrossRef]
  98. Agasthi, P.; Van Houten, H.K.; Yao, X.; Jain, C.C.; Egbe, A.; Warnes, C.A.; Miranda, W.R.; Dunlay, S.M.; Stephens, E.H.; Johnson, J.N.; et al. Mortality and Morbidity of Heart Failure Hospitalization in Adult Patients With Congenital Heart Disease. J. Am. Heart Assoc. 2023, 12, e030649. [Google Scholar] [CrossRef]
  99. Fusco, F.; Scognamiglio, G.; Abbate, M.; Merola, A.; Grimaldi, N.; Ciriello, G.D.; Sarubbi, B. Dapagliflozin in Patients With a Failing Systemic Right Ventricle: Results From the DAPA-SERVE Trial. JACC Heart Fail. 2024, 12, 789–791. [Google Scholar] [CrossRef]
  100. Joki, Y.; Konishi, H.; Takasu, K.; Minamino, T. Tofogliflozin, a sodium-glucose cotransporter 2 inhibitor, improves pulmonary vascular remodeling due to left heart disease in mice. J. Cardiol. 2023, 81, 347–355. [Google Scholar] [CrossRef] [PubMed]
  101. Konstam, M.A.; Kiernan, M.S.; Bernstein, D.; Bozkurt, B.; Jacob, M.; Kapur, N.K.; Kociol, R.D.; Lewis, E.F.; Mehra, M.R.; Pagani, F.D.; et al. Evaluation and Management of Right-Sided Heart Failure: A Scientific Statement From the American Heart Association. Circulation 2018, 137, e578–e622. [Google Scholar] [CrossRef]
  102. Clements, R.T.; Vang, A.; Fernandez-Nicolas, A.; Kue, N.R.; Mancini, T.J.; Morrison, A.R.; Mallem, K.; McCullough, D.J.; Choudhary, G. Treatment of Pulmonary Hypertension With Angiotensin II Receptor Blocker and Neprilysin Inhibitor Sacubitril/Valsartan. Circ. Heart Fail. 2019, 12, e005819. [Google Scholar] [CrossRef]
  103. Jariwala, P.; Pramod, G. Impact of SGLT2 Inhibitor Therapy, EMPAgliflozin on RIGHT Ventricular Function in Patients with Right Heart Failure and Severe Pulmonary Hypertension in Chronic Pulmonary Disorders (EMPA-RIGHT Study). Indian Heart J. 2023, 75, S11. [Google Scholar] [CrossRef]
  104. Sarak, B.; Verma, S.; David Mazer, C.; Teoh, H.; Quan, A.; Gilbert, R.E.; Goodman, S.G.; Bami, K.; Coelho-Filho, O.R.; Ahooja, V.; et al. Impact of empagliflozin on right ventricular parameters and function among patients with type 2 diabetes. Cardiovasc. Diabetol. 2021, 20, 200. [Google Scholar] [CrossRef] [PubMed]
  105. Zhao, Q.H.; Chen, J.; Chen, F.D.; Ruan, H.Y.; Zhang, W.; Zhou, Y.L.; Wang, Q.Q.; Xu, X.L.; Feng, K.F.; Guo, J.Z.; et al. Evaluating the efficacy and safety of oral triple sequential combination therapy for treating patients with pulmonary arterial hypertension: A multicenter retrospective study. Pulm. Circ. 2024, 14, e12351. [Google Scholar] [CrossRef] [PubMed]
  106. Chaumais, M.-C.; Djessas, M.R.A.; Thuillet, R.; Cumont, A.; Tu, L.; Hebert, G.; Gaignard, P.; Huertas, A.; Savale, L.; Humbert, M.; et al. Additive protective effects of sacubitril/valsartan and bosentan on vascular remodelling in experimental pulmonary hypertension. Cardiovasc. Res. 2021, 117, 1391–1401. [Google Scholar] [CrossRef] [PubMed]
  107. Connelly, K.A.; Wu, E.; Visram, A.; Friedberg, M.K.; Batchu, S.N.; Yerra, V.G.; Thai, K.; Nghiem, L.; Zhang, Y.; Kabir, G.; et al. The SGLT2i Dapagliflozin Reduces RV Mass Independent of Changes in RV Pressure Induced by Pulmonary Artery Banding. Cardiovasc. Drugs Ther. 2022, 38, 57–68. [Google Scholar] [CrossRef] [PubMed]
  108. Chowdhury, B.; Luu, A.Z.; Luu, V.Z.; Kabir, M.G.; Pan, Y.; Teoh, H.; Quan, A.; Sabongui, S.; Al-Omran, M.; Bhatt, D.L.; et al. The SGLT2 inhibitor empagliflozin reduces mortality and prevents progression in experimental pulmonary hypertension. Biochem. Biophys. Res. Commun. 2020, 524, 50–56. [Google Scholar] [CrossRef]
  109. Andersen, S.; Axelsen, J.B.; Ringgaard, S.; Nyengaard, J.R.; Hyldebrandt, J.A.; Bogaard, H.J.; de Man, F.S.; Nielsen-Kudsk, J.E.; Andersen, A. Effects of combined angiotensin II receptor antagonism and neprilysin inhibition in experimental pulmonary hypertension and right ventricular failure. Int. J. Cardiol. 2019, 293, 203–210. [Google Scholar] [CrossRef]
  110. Liu, S.; Wang, Y.; Lu, S.; Hu, J.; Zeng, X.; Liu, W.; Wang, Y.; Wang, Z. Sacubitril/valsartan treatment relieved the progression of established pulmonary hypertension in rat model and its mechanism. Life Sci. 2021, 266, 118877. [Google Scholar] [CrossRef] [PubMed]
  111. Wang, J.; Ma, Y.-R.; Chang, Y.-E.; Duo, D.-L.; Duan, K.-K.; Zhao, N.; Cui, W.-L.; Huan, Z.-L.; Wang, Y.-F. Preventive effect of LCZ696 on hypoxic pulmonary hypertension in rats via regulating the PI3K/AKT signaling pathway. Pulm. Pharmacol. Ther. 2023, 82, 102229. [Google Scholar] [CrossRef]
  112. Miranda, W.R.; Jain, C.C.; Egbe, A.C.; Reddy, Y.N.; Dearani, J.A.; Hagler, D.J.; Connolly, H.M. Hemodynamics in Adults with Systemic Right Ventricles: Differences Between Congenitally Corrected and Complete Transposition of the Great Arteries. Pediatr. Cardiol. 2024. [Google Scholar] [CrossRef]
  113. Ladouceur, M.; Segura de la Cal, T.; Gaye, B.; Valentin, E.; Ly, R.; Iserin, L.; Legendre, A.; Mousseaux, E.; Li, W.; Rafiq, I.; et al. Effect of medical treatment on heart failure incidence in patients with a systemic right ventricle. Heart 2021, 107, 1384–1389. [Google Scholar] [CrossRef] [PubMed]
  114. Lopez-Candales, A.; Norgard, N.B. Right here and right now: Time to incorporate measuring right ventricular function when assessing left heart failure clinical outcomes. J. Card. Fail. 2022, 28, 1741–1742. [Google Scholar] [CrossRef] [PubMed]
  115. Chaix, M.-A.; Dore, A.; Mondésert, B.; Mongeon, F.-P.; Roy, V.; Guertin, M.-C.; White, M.; Ibrahim, R.; O’Meara, E.; Rouleau, J.-L.; et al. Design of the Prospective Comparison of Angiotensin Receptor-Neprilysin Inhibitor Versus Placebo in Patients With Congenital Systemic Right Ventricle Heart Failure (PARACYS-RV) Trial. CJC Open 2023, 5, 537–544. [Google Scholar] [CrossRef] [PubMed]
  116. Andi, K.; Abozied, O.; Miranda, W.R.; Anderson, J.H.; Connolly, H.M.; Jain, C.C.; Burchill, L.J.; Egbe, A.C. Clinical benefits of angiotensin receptor-Neprilysin inhibitor in adults with congenital heart disease. Int. J. Cardiol. 2023, 387, 131152. [Google Scholar] [CrossRef] [PubMed]
  117. Appadurai, V.; Thoreau, J.; Malpas, T.; Nicolae, M. Sacubitril/Valsartan in Adult Congenital Heart Disease Patients With Chronic Heart Failure—A Single Centre Case Series and Call for an International Registry. Heart Lung Circ. 2020, 29, 137–141. [Google Scholar] [CrossRef] [PubMed]
  118. Ephrem, G.; McCollum, J.C.; Green-Hess, D.; Guglin, M.E.; Sawada, S.G.; Rao, R.A. Subjective and Objective Impact of Angiotensin Receptor-Neprilysin Inhibitors on Systemic Right Ventricle Patients. Heart Lung Circ. 2022, 31, 964–973. [Google Scholar] [CrossRef] [PubMed]
  119. Fusco, F.; Scognamiglio, G.; Merola, A.; Iannuzzi, A.; Palma, M.; Grimaldi, N.; Sarubbi, B. Safety and Efficacy of Sacubitril/Valsartan in Patients With a Failing Systemic Right Ventricle: A Prospective Single-Center Study. Circ. Heart Fail. 2023, 16, e009848. [Google Scholar] [CrossRef] [PubMed]
  120. Lluri, G.; Lin, J.; Reardon, L.; Miner, P.; Whalen, K.; Aboulhosn, J. Early Experience With Sacubitril/Valsartan in Adult Patients With Congenital Heart Disease. World J. Pediatr. Congenit. Heart Surg. 2019, 10, 292–295. [Google Scholar] [CrossRef]
  121. Maurer, S.J.; Pujol Salvador, C.; Schiele, S.; Hager, A.; Ewert, P.; Tutarel, O. Sacubitril/valsartan for heart failure in adults with complex congenital heart disease. Int. J. Cardiol. 2020, 300, 137–140. [Google Scholar] [CrossRef]
  122. Nederend, M.; Kies, P.; Regeer, M.V.; Vliegen, H.W.; Mertens, B.J.; Robbers-Visser, D.; Bouma, B.J.; Tops, L.F.; Schalij, M.J.; Jongbloed, M.R.M.; et al. Tolerability and beneficial effects of sacubitril/valsartan on systemic right ventricular failure. Heart 2023, 109, 1525–1532. [Google Scholar] [CrossRef]
  123. Yan, L.; Loh, J.K.; Tan, J.L. Sacubitril/valsartan for heart failure in patients with complex adult congenital heart disease—Experience from a tertiary centre in Singapore. Int. J. Cardiol. Congenit. Heart Dis. 2021, 6, 100268. [Google Scholar] [CrossRef]
  124. Zandstra, T.E.; Nederend, M.; Jongbloed, M.R.M.; Kies, P.; Vliegen, H.W.; Bouma, B.J.; Tops, L.F.; Schalij, M.J.; Egorova, A.D. Sacubitril/valsartan in the treatment of systemic right ventricular failure. Heart 2021, 107, 1725–1730. [Google Scholar] [CrossRef]
  125. Neijenhuis, R.M.L.; MacDonald, S.T.; Zemrak, F.; Mertens, B.J.A.; Dinsdale, A.; Hunter, A.; Walker, N.L.; Swan, L.; Reddy, S.; Rotmans, J.I.; et al. Effect of Sodium-Glucose Cotransporter 2 Inhibitors in Adults With Congenital Heart Disease. J. Am. Coll. Cardiol. 2024, 83, 1403–1414. [Google Scholar] [CrossRef] [PubMed]
  126. Gupta, R.; Maitz, T.; Egeler, D.; Mehta, A.; Nyaeme, M.; Hajra, A.; Goel, A.; Sreenivasan, J.; Patel, N.; Aronow, W.S. SGLT2 inhibitors in hypertension: Role beyond diabetes and heart failure. Trends Cardiovasc. Med. 2023, 33, 479–486. [Google Scholar] [CrossRef]
  127. Egorova, A.D.; Nederend, M.; Tops, L.F.; Vliegen, H.W.; Jongbloed, M.R.M.; Kies, P. The first experience with sodium-glucose cotransporter 2 inhibitor for the treatment of systemic right ventricular failure. ESC Heart Fail. 2022, 9, 2007–2012. [Google Scholar] [CrossRef]
  128. Muneuchi, J.; Sugitani, Y.; Kobayashi, M.; Ezaki, H.; Yamada, H.; Watanabe, M. Feasibility and Safety of Sodium Glucose Cotransporter-2 Inhibitors in Adults with Heart Failure after the Fontan Procedure. Case Rep. Cardiol. 2022, 2022, 5243594. [Google Scholar] [CrossRef] [PubMed]
  129. Kheiwa, A.; Ssembajjwe, B.; Chatta, P.; Nageotte, S.; Abramov, D. Safety of SGLT-2 inhibitors in the management of heart failure in the adult congenital heart disease patient population. Int. J. Cardiol. Congenit. Heart Dis. 2024, 15, 100495. [Google Scholar] [CrossRef]
  130. Saef, J.; Sundaravel, S.; Ortega-Legaspi, J.; Vaikunth, S. Safety and Treatment Experience With Sodium/glucose Cotransporter-2 Inhibitors in Adult Patients With Congenital Heart Disease. J. Card. Fail. 2023, 29, 974–975. [Google Scholar] [CrossRef] [PubMed]
  131. Karnakoti, S.; Andi, K.; Miranda, W.R.; Jain, C.C.; Burchill, L.J.; Jokhadar, M.; Connolly, H.M.; Egbe, A.C. Outcomes of Sodium-Glucose Cotransporter 2 Inhibitor Use in Adults With Congenital Heart Disease. CJC Pediatr. Congenit. Heart Dis. 2024, 3, 115–116. [Google Scholar] [CrossRef]
  132. Lin, Y.; Zhang, H.; Zhao, S.; Chen, L.; Li, J.; Wang, X.; Tian, W. The Efficacy and Safety of the Combined Therapy of Sodium-Glucose Co-Transporter-2 Inhibitors and Angiotensin Receptor-Neprilysin Inhibitor in Patients With Heart Failure With Reduced Ejection Fraction: A Meta-Analysis of the EMPEROR-Reduced and DAPA-HF Sub-Analysis. Front. Cardiovasc. Med. 2022, 9, 882089. [Google Scholar] [CrossRef]
  133. Mo, X.; Lu, P.; Yang, X. Efficacy of sacubitril-valsartan and SGLT2 inhibitors in heart failure with reduced ejection fraction: A systematic review and meta-analysis. Clin. Cardiol. 2023, 46, 1137–1145. [Google Scholar] [CrossRef] [PubMed]
  134. Tsai, M.-L.; Lin, Y.; Lin, M.-S.; Tsai, T.-H.; Yang, N.-I.; Wang, C.-Y.; Hsieh, I.C.; Hung, M.-J.; Chen, T.-H. Comparing angiotensin receptor–neprilysin inhibitors with sodium–glucose cotransporter 2 inhibitors for heart failure with diabetes mellitus. Diabetol. Metab. Syndr. 2023, 15, 110. [Google Scholar] [CrossRef] [PubMed]
  135. Diaz Acevedo, J.; Castro, A.; Moreno, P.M. Hierarchical ranking of novel therapies combinations in heart failure with reduced ejection fraction: Network meta-analysis. JACC Basic Transl. Sci. 2022, 79, 337. [Google Scholar] [CrossRef]
Figure 1. A conceptual representation of the pathophysiological pressure–volume (PV) loop. The left ventricle (LV) PV loop in normal hemodynamic condition (in black), with a rectangular PV loop, illustrating distinct isovolumetric relaxation and contraction phases, and the change in HF (in red). Right ventricle (RV) PV loop in normal hemodynamic condition (in black), with a triangular shape due to the absence of isovolumetric relaxation or contraction phases, and the change in HF (in red) (Figure represents original artwork). (ESP: end-systolic pressure; EDP: end-diastolic pressure; ESV: end-systolic volume; EDV: end-diastolic volume; ESPVR: end-systolic pressure volume relationship; EDPVR: end-diastolic pressure volume relationship; ELV: effective LV elastance; ERV: effective RV elastance; and Ea: effective arterial elastance).
Figure 1. A conceptual representation of the pathophysiological pressure–volume (PV) loop. The left ventricle (LV) PV loop in normal hemodynamic condition (in black), with a rectangular PV loop, illustrating distinct isovolumetric relaxation and contraction phases, and the change in HF (in red). Right ventricle (RV) PV loop in normal hemodynamic condition (in black), with a triangular shape due to the absence of isovolumetric relaxation or contraction phases, and the change in HF (in red) (Figure represents original artwork). (ESP: end-systolic pressure; EDP: end-diastolic pressure; ESV: end-systolic volume; EDV: end-diastolic volume; ESPVR: end-systolic pressure volume relationship; EDPVR: end-diastolic pressure volume relationship; ELV: effective LV elastance; ERV: effective RV elastance; and Ea: effective arterial elastance).
Medicina 60 01112 g001
Figure 2. Mechanism of action of combined ARNI and SGLT2i therapy in chronic RV Failure (Figure is original artwork). (NHE1: Sodium-hydrogen exchange isoform 1; NHE3: Sodium-hydrogen exchange isoform 3; PH: Pulmonary hypertension; BNP: B-type natriuretic peptide; CNP: C-type natriuretic peptide; cGMP: cyclic guanosine monophosphate; Na+: Sodium; Gluc: Glucose; H2O: water; R-A-S: Renin-angiotensin-aldosterone; AT1R: Angiotensin receptor 1; ATP: angiotensin receptor 1 antagonist; ATP: adenosine triphosphate; AMPK: adenosine monophosphate kinase; PGC1∝: Peroxisome proliferator-activated receptor Ÿ 1-œ; and ROS: reactive oxygen species).
Figure 2. Mechanism of action of combined ARNI and SGLT2i therapy in chronic RV Failure (Figure is original artwork). (NHE1: Sodium-hydrogen exchange isoform 1; NHE3: Sodium-hydrogen exchange isoform 3; PH: Pulmonary hypertension; BNP: B-type natriuretic peptide; CNP: C-type natriuretic peptide; cGMP: cyclic guanosine monophosphate; Na+: Sodium; Gluc: Glucose; H2O: water; R-A-S: Renin-angiotensin-aldosterone; AT1R: Angiotensin receptor 1; ATP: angiotensin receptor 1 antagonist; ATP: adenosine triphosphate; AMPK: adenosine monophosphate kinase; PGC1∝: Peroxisome proliferator-activated receptor Ÿ 1-œ; and ROS: reactive oxygen species).
Medicina 60 01112 g002
Table 1. Summarizes the goals of ongoing trials.
Table 1. Summarizes the goals of ongoing trials.
AuthorsStudyWhat the Study Entails
Salam et al. [75]RV Shock ManagementStrategies for managing RV shock, particularly in the setting of RV infarct
Mansoor et al. [76]Pharmacotherapy for HFTrials looking at novel medications, as well as currently approved drugs
Yuriditsky et al. [77]Medical Optimization of Acute RV FailureTailored volume administration, ideal vasopressor selection, inotropes to restore contractility, and pulmonary vasodilators to improve afterload
Diaz JC et al. [78]Left Bundle Branch Area Pacing (LBBAP)Investigation of LBBAP in the context of RV depolarization provided by the right bundle branch
Lippman et al. [79]Pathogenesis of RV DysfunctionNew insights into the role of fibrosis, inflammation, myocyte contraction, and mitochondrial dynamics in RV dysfunction
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Das, B.B. Unlocking the Potential: Angiotensin Receptor Neprilysin and Sodium Glucose Co-Transporter 2 Inhibitors for Right Ventricle Dysfunction in Heart Failure. Medicina 2024, 60, 1112. https://doi.org/10.3390/medicina60071112

AMA Style

Das BB. Unlocking the Potential: Angiotensin Receptor Neprilysin and Sodium Glucose Co-Transporter 2 Inhibitors for Right Ventricle Dysfunction in Heart Failure. Medicina. 2024; 60(7):1112. https://doi.org/10.3390/medicina60071112

Chicago/Turabian Style

Das, Bibhuti B. 2024. "Unlocking the Potential: Angiotensin Receptor Neprilysin and Sodium Glucose Co-Transporter 2 Inhibitors for Right Ventricle Dysfunction in Heart Failure" Medicina 60, no. 7: 1112. https://doi.org/10.3390/medicina60071112

Article Metrics

Back to TopTop