Skip to main content
Skip main navigationClose Drawer MenuOpen Drawer Menu

Pulmonary Hypertension in HFpEF and HFrEF: JACC Review Topic of the WeekFree Access

JACC Review Topic of the Week

J Am Coll Cardiol, 76 (9) 1102–1111
Sections

Central Illustration

Abstract

Pulmonary hypertension (PH) associated with left heart disease, or Group 2 PH, includes heart failure, valvular heart diseases, and congenital heart diseases. Although it is axiomatic that in PH due to heart failure the increase in pulmonary pressure is directly related to an enhanced left atrial pressure, which is common to both heart failure with preserved ejection fraction (HFpEF) and heart failure with reduced ejection fraction (HFrEF), there has been limited attention over the years on the potential differences in terms of driving mechanisms, pathophysiology, and clinical phenotypes. Major differences between HFpEF and HFrEF are the underlying causes, associated comorbidities, and cardiac remodeling. This suggests that despite similar hemodynamic profiles, there may be some disparities in PH development. A focused knowledge on the differences between the 2 syndromes has relevant implications to seek new, personalized, and timely treatments for Group 2 PH. The purpose of the present review is to highlight the mechanisms and clinical phenotypes of PH in HFpEF and HFrEF.

Highlights

In HF, development of PH is an unfavorable clinical turning point in the natural history of the disease.

Although HFpEF and HFrEF exhibit a similar pulmonary hemodynamic profile and outcomes, there are major differences in underlying causes, cardiac remodeling, and comorbidities.

There are also different triggers and molecular pathways for pulmonary vascular injury and remodeling in HFpEF versus HFrEF.

New targets for personalized PH prevention and treatment in the 2 HF conditions are necessary.

Introduction

Pulmonary hypertension (PH) associated with left heart diseases is the most common form, classified as post-capillary or Group 2 PH (1,2). It includes heart failure (HF), valvular, and congenital heart diseases. There is great interest in the pathophysiology, clinical evolution, and therapies of left-sided PH (2). Especially in the last decade, PH and right ventricular dysfunction (RVD) have been the focus of interest in heart failure with preserved ejection fraction (HFpEF) (3–6), a condition with similar clinical manifestations and event rate to heart failure with reduced ejection fraction (HFrEF) (7,8), but different in etiology, cardiac remodeling, pathophysiology, comorbid disorders, and response to therapy (9). In HFrEF, PH has long been investigated in advanced stages (i.e., in patients waiting for left ventricular assist device [LVAD] or heart transplantation) (8). However, in HFpEF, the role of PH is under intense scrutiny (10), and a thorough recognition of putative mechanisms is demanding, given the high epidemiological impact and the lack of effective management (2).

There are, therefore, intriguing questions that deserve specific attention on how, in the 2 conditions, the molecular pathways, the pathophysiology, and the clinical manifestations of PH may differ, despite similar hemodynamic profile and outcomes. This has obvious implications in terms of prevention and treatment.

We highlight the underlying differences between PH-HFrEF and PH-HFpEF with the aim of guiding research efforts for novel therapies. Specifically, we focus on the characteristic derangements in left heart driving forces biological pathways, hemodynamics, and clinical phenotypes observed in the 2 HF conditions (Central Illustration).

Central Illustration
Central Illustration

Left Heart Driving Forces and Cellular Pathways of Left Ventricular and Left Atrial Remodeling, Pulmonary Vascular Disease, and Right Heart Failure Evolution in Heart Failure With Preserved and Reduced Ejection Fraction

In heart failure with reduced ejection fraction (HFrEF), eccentric remodeling, dynamic mitral regurgitation (MR), and greater left atrial (LA) enlargement drive PH development. In HFpEF, concentric remodeling with increased left ventricular (LV) diastolic stiffness, atrial functional MR and a stiff LA are the major driving forces to alveolar-capillary stress failure and vascular remodeling. EDPVR = end-diastolic pressure volume relationship; ESPVR = end-systolic pressure volume relationship; PAWP = pulmonary arterial wedge pressure; PH = pulmonary hypertension; TR = tricuspid regurgitation.

Definition, Characterization, and Epidemiology of Group 2 PH in HF

During the latest 2018 WHO conference, group 2 PH (2) was defined as a mean pulmonary arterial pressure (mPAP) >20 mm Hg and pulmonary arterial wedge pressure (PAWP) >15 mm Hg. Isolated post capillary (Ipc)-PH or combined pre- and post-capillary (Cpc)-PH were defined on a pulmonary vascular resistance (PVR) <3 WU or ≥3 WU, respectively.

This applies to group 2 PH at a whole, including valvular or congenital heart disease; however, PH-HFrEF and PH-HFpEF present with dissimilarities (11,12). Typically, patients with PH-HFpEF are elderly women with the features of metabolic syndrome (9,13), and when metabolic syndrome and obesity coexist, the worse PH phenotype is observed (13–15).

Additional triggers for PH-HFpEF include microvascular coronary disease, atrial fibrillation (AF), chronic kidney disease, and chronic obstructive pulmonary disease (4). PH-HFrEF is more common in middle-aged men with coronary heart disease or dilated cardiomyopathy (8).

Right heart catheterization (RHC) remains the gold standard for a correct PH diagnosis and for timing heart transplantation (HTx) or LVAD support (2).

RHC is also valuable for investigating the hemodynamic response during exercise, discriminating PH-HFpEF from PAH (2) and monitoring interventions (16,17). However, PH prevalence is derived from a nonuniform application of RHC diagnostic standards and observations obtained in different stages of the disease, explaining the wide variability (i.e., between 40% and 75% for PH-HFrEF [18] and 36% to 83% for PH-HFpEF [3]). Some reports also refer to old diagnostic criteria or to the use of pulmonary artery systolic pressure (PASP) estimated by echo Doppler (1). For example, in the recent subanalysis of the PARAGON-HF (Prospective Comparison of ARNI [Angiotensin Receptor-Neprilysin Inhibitor] with ARB [Angiotensin Receptor Blocker] Global Outcomes in HF with Preserved Ejection Fraction) trial, the rate of PH-HFpEF was 31% (6). Nonetheless, these numbers would consistently change based on the HFpEF phenogroup subdivision described by Shah et al. (15), where the true PH phenotype exhibited higher PVR, right atrial pressure, and combined kidney disease, as major PH contributory mechanisms.

There is also some variability in the expected Cpc-PH prevalence. In a large sample of PH-HFrEF, Miller et al. (8) documented a 55% Cpc-PH rate, using PVR criteria. In a retrospective analysis of 2,587 PH-HFpEF patients, the prevalence varied from 8.8% to 3.5% by using a diastolic pressure gradient (DPG) >7 mm Hg or a PVR >3 WU as hemodynamic criteria (19). Another report, merging retrospective and prospective data, found a Cpc-PH rate of 22.6% and 18.8%, for HFpEF and HFrEF, respectively, using a DPG >7 mm Hg (18). For a similar DPG cutoff, a higher Cpc-PH rate (38%) was observed in PH-HFpEF compared with PH-HFrEF (17%) (11).

Overall, these data suggest that Cpc-PH may be more frequent in HFpEF.

Left Heart Driving Forces and Hemodynamics

In both HFrEF and HFpEF, the primary hemodynamic driver of PH is an impaired LV relaxation and filling, which yields backward transmission of elevated left atrial pressure (LAP) to the venous system, capillaries, and arteries and, ultimately, to the right heart. LAP elevation results from the interaction between the type of LV remodeling and left atrial (LA) adaptation in dimensions and dynamics. Indeed, the type of LV myocyte hypertrophy and the amount of reactive and replacement fibrosis well separate HFpEF from HFrEF (20).

Impaired LV filling and cardiac remodeling

In HFpEF, LV concentric hypertrophy and increased diastolic stiffness are common in hypertension and obesity (14,21). Approximately one-third of patients may have diabetes and 10% to 15% (9) may experience infiltrative disease such as cardiac amyloidosis, which is, however, a condition that needs to be diagnosed and excluded from a typical HFpEF phenotype, as indicated by the recent HFA-PEFF score (22). Comorbid conditions exert negative effects on the myocardium, stimulating oxidative stress and hypertrophic pathways (8). On this background, Paulus and Tschope (23) proposed a unifying hypothesis based on the central role of proinflammatory pathways to alter passive tension myocyte properties by increasing collagen proliferation.

High wall stiffness is sustained by increased phosphorylation of titin protein, overexpression of growth factor ß signaling, reduced expression of elastases, high mitochondrial oxidative stress, and epigenetic changes definitively impairing cellular Ca+ homeostasis (9).

In HFrEF, LV morphology and myocyte adaptations are driven by excessive wall stress, in most cases due to ischemia and a myocyte genetic background favoring chamber dilation and eccentric hypertrophy. The cardiomyocyte loss alters the balance between collagen deposition and degradation, and patchy areas of fibrosis develop through replacement of dead cardiomyocytes (20). Alterations within the extracellular matrix further contribute to LV enlargement and promote impairment in LV compliance and late increase in stiffness. Abnormalities in LV filling correlate with symptoms better than LVEF and predict outcome (24). At variance with HFrEF, increased LV filling pressures with altered LV passive chamber properties develop early and are prominent in HFpEF (9).

An elevated arterial elastance is an additional pathogenetic substrate for LV stiffness in HFpEF related to aging (25) and menopause estrogen deficiency (26). Accordingly, PH-HFpEF may be exposed to a negative LV mismatch when receiving vasodilators (27).

An enhanced diastolic ventricular interaction is common in PH-HFrEF; RV enlargement and pericardial restraint further impair LV filling, a phenomenon observed in approximately 40% of HFrEF (1). In HFpEF, an unfavorable diastolic ventricular interaction is unmasked during exercise even in the earlier stages (28) and becomes a primary mechanism of PAWP increase in the obese phenotype (14).

Abnormalities in LA dynamics and their determinants

The LA is extremely sensitive to volume and pressure overload and remodels quite rapidly (29). An impaired LA dynamics is fundamental in PH, becoming the major contributor to the elevation in pulsatile loading (1). LA remodeling reflects abnormalities in LV filling pressure and diastolic function unless AF, high-output states, and mitral regurgitation (MR) coexist (30).

The HFrEF phenotype exhibits LA enlargement and eccentric remodeling secondary to severe MR, whereas in HFpEF, the LA becomes stiffer earlier, dilates more slowly, and shows a peculiar cellular substrate favoring AF (9).

Comorbidities and inflammatory mediators also play a main role in atrial dysfunction, rendering the atrium stiff, a condition that begets AF (31). The AF predominant HFpEF syndrome is a unique phenotype associated with MR, higher PVR, and worse outcome (31).

Irrespective of AF, MR coexistence represents the leading hemodynamic mechanism to PH (30,32). Interestingly, data from the ATTEND registry showed that HFpEF may develop MR at a similar rate compared with HFrEF, bearing similar prognostic impact (33). Nonetheless, the observed differences were a higher frequency of mild MR in HFpEF versus severe degree in HFrEF. At variance with HFrEF, in which MR develops on the imbalance between tethering and closing forces, HFpEF exhibits the so called atrial functional MR (34), i.e., mitral annulus (MA) dilatation induced by AF and/or, irrespective of AF, secondary to increased LV end-diastolic pressure and MA dilation (34). AF and HFpEF share pathophysiological grounds (31), making it problematic to differentiate what comes first.

Both HFrEF and HFpEF exhibit impaired LA dynamics with deranged LA reservoir and contractile function tightly connected with PH and RV dysfunction (35). Interestingly, in HFpEF, an abnormal reservoir dynamics combines with elevated PAWP, even when LA volume is normal and functional abnormalities in LA dynamics and hemodynamics are better characterized on exercise. In a study of HFrEF or HFpEF and of healthy control subjects, changes in LA strain and LA pump function (strain rate) were assessed at rest, during exercise, and during the recovery phase. Either HFpEF and HFrEF showed a markedly depressed response (Figures 1A and 1B), with a steep increase in the PASP/tricuspid annular plane systolic excursion relationship for lower LA strain, pointing to the role of the LA in the RV to pulmonary circulation (Pc) uncoupling (Figure 1C) (32).

Figure 1
Figure 1

Example of Speckle Tracking Analysis of LA Dynamics (LA Strain and Strain Rate Analysis) at Rest, During Exercise, and in Recovery Phase in Patients With HFpEF, HFrEF, and Control Subjects

Average changes in left atrial (LA) strain (A) and strain rate (B) during exercise and recovery phase and rest to exercise variation in LA strain pulmonary artery systolic pressure (PASP)/tricuspid annular plane systolic excursion (TAPSE) relationship (C). HFpEF = heart failure with preserved ejection fraction; HFrEF = heart failure with reduced ejection fraction.

Increased LAP and pulmonary hemodynamic phenotypes

Any increase in LAP, even mild, perturbates pulmonary hemodynamics and exacerbates symptoms (36). The longitudinal changes in pulmonary hemodynamics at different LAP stages are undefined; however, an analysis by Adir et al. (11) comparing the pulmonary hemodynamics in response to long-lasting elevation in LAP in PH-HFrEF and PH-HFpEF provided intriguing differences. For similar average PAWP, PH-HFpEF had higher DPG and PVR, similar arterial compliance and lower MR rate, indicating a more pronounced vascular disease under similar pulsatile loading and likely reflecting a worse lung microvascular pathology secondary to inflammation.

The thorough assessment of pulmonary hemodynamics during exercise defines distinct pathophysiological features that help to unmask peculiar hemodynamic patterns (37).

Although exercise-induced MR has been recently documented in HFpEF (30), it remains the main trigger of PH in HFrEF (32). There is established evidence that Cpc-PH combines with exercise ventilatory inefficiency similarly in PH-HFpEF and PH-HFrEF (37).

Pathobiology of Lung Vascular Disease and Remodeling

Irrespective of LVEF, endothelial injury is central to the microvascular dysfunction of PH (Figure 2). Studies on the lung vasculature in humans are limited, but preclinical evidence and genetic/epigenetic blood-omic analyses are generating a considerable amount of data (38).

Figure 2
Figure 2

Mediators of Pulmonary Vein and Arteriolar/Capillary Remodeling

The main trigger for remodeling is the raised left atrial pressure (LAP). Veins and capillaries remodel similarly, with variable dense fibrosis versus loose fibrosis/hyalinosis. The only documented pathway for vein remodeling is the urokinase plasminogen activator receptor (uPAR). The increased capillary pressure impairs endothelial function and permeability by increasing the caveolin family and altering the NO, ET1, and natriuretic peptide pathways. Remodeling ensues through a progressive inflammatory cascade promoted by growth factors and smooth muscle cells (SMCs) proliferation. The HFpEF phenotype with metabolic syndrome and obesity shows a parallel and additive way of endothelial injury induced by molecule adhesion, reactive oxygen species (ROS) activation, and reduced adiponectin concentration. Angio = angiotensin; cGMP = cyclic guanosine 3', 5'-monophosphate; EC = endothelial cell; EMT = epithelial-mesenchymal transition; ET = endothelin; IL = interleukin; MCP = monocyte chemoattractant activating protein; MYF = myofibroblast; NO = nitric oxide; NP = natriuretic peptide; PKG = cGMP-dependent protein kinase; STAT = signal transducer and activator of transcription; TGF = transforming growth factor; TNF = tumor necrosis factor; other abbreviations as in Figure 1.

Veins

Intriguing isolated findings have shown that veins undergo a remarkable remodeling process in both HFpEF and HFrEF (39), consisting of luminal narrowing with increased neointimal thickening and medial hypertrophy. Intimal thickening and PASP correlated even after adjusting for arterial medial and intimal thickening, and a loss in gas diffusion was reported. In few patients with sequential lung biopsy at the time of removal of the LVADs, high levels of urokinase plasminogen activator receptor expression have been detected, representing the only pathway so far identified in venous remodeling process (40).

Capillaries and small arteries

The raised backward pressure causes lung capillary and small artery stress failure, a barotrauma that breaks the endothelial layer and promotes fluid and protein swelling in the interstitium. Fluid triggers a cascade of molecular mediators of vascular remodeling, whose pathogenesis is attractive for new targeted therapeutic intervention (41). Edema activates inflammatory mediators, inhibits nitric oxide and natriuretic peptide activities, and increases endothelin (ET)-1 expression leading to fibroblasts/myofibroblast proliferation, luminal occlusion, and thickening of the alveolar septa (1,36). The transition from alveolar-capillary stress failure to remodeling is critical and, in clinical practice, is reflected by a reduced alveolar membrane gas diffusion and high levels of surfactant protein type B (36).

A genetic analysis of vascular remodeling in PH-HFrEF demonstrated an enrichment in genes related to cytoskeleton structure and immune function involving biologically plausible pathways including actin binding, extracellular matrix, basement membrane, transferase activity, pre-ribosome structure, and the major histocompatibility Class II protein complex (42).

A reduced gas diffusion in HFrEF has long been documented (1), and there has also been a recent focus on PH-HFpEF (43).

Evidence on abnormal insulin metabolism as a mediator of pulmonary vascular disease in HFpEF is increasing (44). Activation of macrophage and leukocyte adhesion (45) as well as an overexpression of the ET-1 pathway (46) have been observed in the capillaries of diabetic rats. Increased reactive oxygen species production in diabetic mice is also implicated in the loss of pulmonary endothelial permeability, also sustained by a higher caveolae number.

In models of metabolic syndrome and PH, interleukin (IL)-6 overexpression induces pulmonary arterial smooth muscle cells (SMCs) proliferation and remodeling through STAT3 activation (44).

In a human ex vivo study, high glucose treatment induced SMCs proliferation via reactive oxygen species generation (47), whereas in obese models, SMCs proliferation was found to depend on the imbalance of adiponectin versus proinflammatory adipokines.

The Right Heart and its Coupling with the Pulmonary Circulation

Adaptations of the RV to chronic pressure overload have been detailed elsewhere (36). Briefly, they consist of hypertrophy, progression to chamber dilatation, functional tricuspid incompetence, and RV failure. Padang et al. (48) recently investigated a cohort of 1,299 patients with newly diagnosed severe RV failure and different etiologies, to examine survival at mid- and long-term. Over a 2-year mean follow-up, severe RV failure was associated with an extremely high mortality rate (62%) and similar outcome for HFpEF and HFrEF. However, the determinants of RVD and the clinical manifestations over time may differ because in HFrEF, the RVD generally develops on an intrinsic myopathy and functional tricuspid regurgitation is independently associated with PH and more severe HF (49). Accordingly, in a large observational study, a substantial proportion of HFrEF patients had low tricuspid annular plane systolic excursion with normal pulmonary pressure, whereas RVD was negligible when pulmonary pressure was normal in HFpEF (12). In HFpEF, RV diastolic dysfunction and increased stiffness occur before systolic dysfunction with a slower progression to dilation and functional deterioration (50).

AF and RV pacing are common backgrounds that, combined with the RV inflammatory injury, well explain the RV diffuse fibrosis as peculiar of HFpEF (51). RV to Pc uncoupling may occur earlier in the PH-HFpEF evolving stages (5).

Hemodynamic Phenotyping and New Pathways for Targeted Treatments of PH

There is no established treatment for Group 2 PH, and guidelines warn against the use of pulmonary vasodilators (2). However, given the burden of PH in HF, the search for new targeted therapies is demanding and pharmacological trials are underway. Early experiences with PAH therapies in this setting were disappointing because they were carried out with the aim to reduce pulmonary pressures rather than targeting the biological vascular properties.

Hemodynamic phenotyping by machine learning is advocated by many as the most likely way to identify the subset of patients that may benefit of treatment (52).

Therefore, there appear to be 2 major unmet needs in group 2 PH treatment: first, the thorough identification of the hemodynamic phenotypes; and second, the definition of pathways to be targeted based on solid experimental evidence (36). A herewith hemodynamically oriented way is proposed referring to the pulmonary arterial compliance versus PVR exponential relationship. As exemplified in Figure 3A, there are some subjects (case A, Ipc-PH) who just benefit from moving pulmonary arterial compliance upward and rightward by reducing PAWP; other subjects (case B, Cpc-PH) get an advantage from moving leftward and upward by reducing PVR and shifting out of the flat portion of the curve.

Figure 3
Figure 3

Case Examples of Hemodynamic Phenotyping (PAC-PVR Relationship) for Potential Therapeutic Use of Pulmonary Vasodilators

(A) Case A (Ipc-PH) would just benefit by moving upward and rightward the pulmonary arterial compliance (PAC) by simply reducing pulmonary arterial wedge pressure (PAWP); case B (Cpc-PH) would get advantage from moving leftward and upward from the flat portion of the curve by reducing pulmonary vascular resistance (PVR) and improving PAC. Case A would take advantage from pharmacological therapies especially diuretic agents, valve replacement, mitral clip, or TAVR when appropriate, whereas Case B would get benefit from the same approach plus LVAD or heart transplantation (HTx) when indicated. The Cpc PH-HFpEF case, who generally is not a recipient of LVAD or HTx treatment, might likely benefit from a pulmonary vasodilator, well tolerated and selective. (B) Case C moves back in the curve remaining in the flat PVR portion; case D is a nonresponder. LVAD = left ventricular assist device; TAVR = transcatheter aortic valve replacement; other abbreviations as in Figure 1.

Indeed, case A sees an advantage from pharmacological therapies, especially diuretic agents, valve replacement, mitral clip, or TAVR when appropriate, whereas case B benefits from the same approach for case A plus LVAD or transplantation, when indicated. There might be a benefit from a pulmonary vasodilator for PH-HFpEF candidates who generally are not recipients of LVAD or HTx. Tolerability and safety are important prerequisites, and considering the potential volume overload/preload sensitivity of the LV, pulmonary selectivity should be looked at. Case C (Figure 3B) is a responder without benefits at all by treatment, i.e., moving back along the flat curve, and case D is a nonresponder to pulmonary vasodilators. These phenotypes should be viewed as different entities in a continuum of iterative relationship between disease duration and response to therapy.

In a study of Cpc PH-HFpEF, sildenafil was effective in a long-term follow-up to improve hemodynamics and gas diffusion (17), whereas a study of Ipc-PH did not show any benefit (53). The PASSION trial is underway and will scrutinize the long-term effects of tadalafil in Cpc-PH-HFpEF patients on event/hospitalization. The ongoing SPHERE-HF (β3 Agonist Treatment in Chronic Pulmonary Hypertension Secondary to Heart Failure), a phase II randomized, double-blind clinical trial (54), will test the effects of a β3 adrenergic receptor agonist, mirabegron, on PVR and arteriolar remodeling based on pilot small number trials showing that albuterol may improve exercise pulmonary vascular reserve (55).

Along with more advanced molecular phenotyping in humans with Cpc-PH, increasingly preclinical reports are providing directions on specific pathways to target. Findings by Lai et al. (56) have paved the way to a connection between metabolic syndrome features and lung vasculopathy. More recently, Ranchoux et al. (44) have identified a role of inflammatory signaling through STAT3 and IL-6 in the microvascular remodeling of PH-HFpEF, demonstrating that the pulmonary vascular phenotype could be rescued by either IL-6 antibody or metformin.

PBI 40-50, a first in class antifibrotic agent, showed a modulating activity on mRNA protein expression of fibrogenic markers in lung tissue of post-MI PH-HFrEF (41). Fasudil, a rho kinase signaling inhibitor, reversed microvascular remodeling by inhibiting ET-1 and ETA expression (57). These compounds are waiting for phase II human studies, and trials are underway to test the effects of improving insulin sensitivity ( NCT03617458, NCT03069716, and NCT03629340).

Conclusions and Perspectives

Despite their similar hemodynamic categorization, there is a growing appreciation of evolving differences between PH-HFrEF and PH-HFpEF. Left hemodynamic drivers for PH-HFrEF are LV dilatation, secondary MR, and LA enlargement. Predominant mechanisms in PH-HFpEF are diastolic stiffness, atrial myopathy more susceptible to evolve to AF, and LA functional MR. Vascular stress failure and remodeling similarly affect veins, capillaries, and small arteries, but a main differential trait is the metabolic injury superimposed to the pressure-induced one, typical of HFpEF with metabolic syndrome. An intrinsic RV pathology is frequently observed in PH-HFrEF, making RV to Pc uncoupling an earlier occurrence.

Further definition in the trajectory of left-sided PH and identification of additional differential pathways for targeting vascular pathology are warranted.

Abbreviations and Acronyms

AF

atrial fibrillation

Cpc

combined pre- and post-capillary

HFpEF

heart failure with preserved ejection fraction

HFrEF

heart failure with reduced ejection fraction

Ipc

isolated post capillary

LAP

left atrial pressure

MR

mitral regurgitation

PASP

pulmonary artery systolic pressure

PH

pulmonary hypertension

PVR

pulmonary vascular resistance

RVD

right ventricular dysfunction

References

  • 1. Guazzi M., Borlaug B.A. "Pulmonary hypertension due to left heart disease". Circulation 2012;126:975-990.

    CrossrefMedlineGoogle Scholar
  • 2. Vachiery J.L., Tedford R.J., Rosenkranz S., et al. "Pulmonary hypertension due to left heart disease". Eur Respir J 2019;53:1801897.

    CrossrefMedlineGoogle Scholar
  • 3. Lam C.S., Roger V.L., Rodeheffer R.J., Borlaug B.A., Enders F.T., Redfield M.M. "Pulmonary hypertension in heart failure with preserved ejection fraction: a community-based study". J Am Coll Cardiol 2009;53:1119-1126.

    View ArticleGoogle Scholar
  • 4. Guazzi M. "Pulmonary hypertension in heart failure preserved ejection fraction: prevalence, pathophysiology, and clinical perspectives". Circ Heart Fail 2014;7:367-377.

    CrossrefMedlineGoogle Scholar
  • 5. Guazzi M., Dixon D., Labate V., et al. "RV contractile function and its coupling to pulmonary circulation in heart failure with preserved ejection fraction: stratification of clinical phenotypes and outcomes". J Am Coll Cardiol Img 2017;10:1211-1221.

    View ArticleGoogle Scholar
  • 6. Shah A.M., Cikes M., Prasad N., et al. "Echocardiographic features of patients with heart failure and preserved left ventricular ejection fraction". J Am Coll Cardiol 2019;74:2858-2873.

    View ArticleGoogle Scholar
  • 7. Lam C.S.P., Gamble G.D., Ling L.H., et al. "Mortality associated with heart failure with preserved vs. reduced ejection fraction in a prospective international multi-ethnic cohort study". Eur Heart J 2018;39:1770-1780.

    CrossrefMedlineGoogle Scholar
  • 8. Miller W.L., Grill D.E., Borlaug B.A. "Clinical features, hemodynamics, and outcomes of pulmonary hypertension due to chronic heart failure with reduced ejection fraction: pulmonary hypertension and heart failure". J Am Coll Cardiol HF 2013;1:290-299.

    Google Scholar
  • 9. Shah S.J., Borlaug B.A., Kitzman D.W., et al. "Research priorities for heart failure with preserved ejection fraction: national heart, lung, and blood institute working group summary". Circulation 2020;141:1001-1026.

    CrossrefMedlineGoogle Scholar
  • 10. Bosch L., Lam C.S.P., Gong L., et al. "Right ventricular dysfunction in left-sided heart failure with preserved versus reduced ejection fraction". Eur J Heart Fail 2017;19:1664-1671.

    CrossrefMedlineGoogle Scholar
  • 11. Adir Y., Guazzi M., Offer A., Temporelli P.L., Cannito A., Ghio S. "Pulmonary hemodynamics in heart failure patients with reduced or preserved ejection fraction and pulmonary hypertension: similarities and disparities". Am Heart J 2017;192:120-127.

    CrossrefMedlineGoogle Scholar
  • 12. Ghio S., Guazzi M., Scardovi A.B., 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.

    CrossrefMedlineGoogle Scholar
  • 13. Gopal D.M., Santhanakrishnan R., Wang Y.C., et al. "Impaired right ventricular hemodynamics indicate preclinical pulmonary hypertension in patients with metabolic syndrome". J Am Heart Assoc 2015;4:e001597.

    CrossrefMedlineGoogle Scholar
  • 14. Obokata M., Reddy Y.N.V., Pislaru S.V., Melenovsky V., Borlaug B.A. "Evidence supporting the existence of a distinct obese phenotype of heart failure with preserved ejection fraction". Circulation 2017;136:6-19.

    CrossrefMedlineGoogle Scholar
  • 15. Shah S.J., Katz D.H., Selvaraj S., et al. "Phenomapping for novel classification of heart failure with preserved ejection fraction". Circulation 2015;131:269-279.

    CrossrefMedlineGoogle Scholar
  • 16. Vachiery J.L., Delcroix M., Al-Hiti H., et al. "Macitentan in pulmonary hypertension due to left ventricular dysfunction". Eur Respir J 2018;51:1701886.

    CrossrefMedlineGoogle Scholar
  • 17. Guazzi M., Vicenzi M., Arena R., Guazzi M.D. "Pulmonary hypertension in heart failure with preserved ejection fraction: a target of phosphodiesterase-5 inhibition in a 1-year study". Circulation 2011;124:164-174.

    CrossrefMedlineGoogle Scholar
  • 18. Gerges M., Gerges C., Pistritto A.M., et al. "Pulmonary hypertension in heart failure. epidemiology, right ventricular function, and survival". Am J Respir Crit Care Med 2015;192:1234-1246.

    CrossrefMedlineGoogle Scholar
  • 19. Vanderpool R.R., Saul M., Nouraie M., Gladwin M.T., Simon M.A. "Association between hemodynamic markers of pulmonary hypertension and outcomes in heart failure with preserved ejection fraction". JAMA Cardiol 2018;3:298-306.

    CrossrefMedlineGoogle Scholar
  • 20. van Heerebeek L., Hamdani N., Handoko M.L., et al. "Diastolic stiffness of the failing diabetic heart: importance of fibrosis, advanced glycation end products, and myocyte resting tension". Circulation 2008;117:43-51.

    CrossrefMedlineGoogle Scholar
  • 21. Hieda M., Sarma S., Hearon C.M., et al. "Increased myocardial stiffness in patients with high risk left ventricular hypertrophy: the hallmark of stage-B HFpEF". Circulation 2019;141:115-123.

    CrossrefMedlineGoogle Scholar
  • 22. Pieske B., Tschope C., de Boer R.A., et al. "How to diagnose heart failure with preserved ejection fraction: the HFA-PEFF diagnostic algorithm: a consensus recommendation from the Heart Failure Association (HFA) of the European Society of Cardiology (ESC)". Eur Heart J 2019;40:3297-3317.

    CrossrefMedlineGoogle Scholar
  • 23. Paulus W.J., Tschope C. "A novel paradigm for heart failure with preserved ejection fraction: comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation". J Am Coll Cardiol 2013;62:263-271.

    View ArticleGoogle Scholar
  • 24. Benfari G., Miller W.L., Antoine C., et al. "Diastolic determinants of excess mortality in heart failure with reduced ejection fraction". JACC Heart Fail 2019;7:808-817.

    View ArticleGoogle Scholar
  • 25. Reddy Y.N.V., Andersen M.J., Obokata M., et al. "Arterial stiffening with exercise in patients with heart failure and preserved ejection fraction". J Am Coll Cardiol 2017;70:136-148.

    View ArticleGoogle Scholar
  • 26. Sabbatini A.R., Kararigas G. "Menopause-related estrogen decrease and the pathogenesis of HFpEF: JACC review topic of the week". J Am Coll Cardiol 2020;75:1074-1082.

    View ArticleGoogle Scholar
  • 27. Schwartzenberg S., Redfield M.M., From A.M., Sorajja P., Nishimura R.A., Borlaug B.A. "Effects of vasodilation in heart failure with preserved or reduced ejection fraction implications of distinct pathophysiologies on response to therapy". J Am Coll Cardiol 2012;59:442-451.

    View ArticleGoogle Scholar
  • 28. Parasuraman S.K., Loudon B.L., Lowery C., et al. "Diastolic ventricular interaction in heart failure with preserved ejection fraction". J Am Heart Assoc 2019;8:e010114.

    CrossrefMedlineGoogle Scholar
  • 29. Zakeri R., Moulay G., Chai Q., et al. "Left atrial remodeling and atrioventricular coupling in a canine model of early heart failure with preserved ejection fraction". Circ Heart Fail 2016;9:e003238.

    CrossrefMedlineGoogle Scholar
  • 30. Tamargo M., Obokata M., Reddy Y.N.V., et al. "Functional mitral regurgitation and left atrial myopathy in heart failure with preserved ejection fraction". Eur J Heart Fail 2020;22:489-498.

    CrossrefMedlineGoogle Scholar
  • 31. Packer M., Lam C.S.P., Lund L.H., Redfield M.M. "Interdependence of atrial fibrillation and heart failure with a preserved ejection fraction reflects a common underlying atrial and ventricular myopathy". Circulation 2020;141:4-6.

    CrossrefMedlineGoogle Scholar
  • 32. Sugimoto T., Bandera F., Generati G., et al. "Left atrial dynamics during exercise in mitral regurgitation of primary and secondary origin: pathophysiological insights by exercise echocardiography combined with gas exchange analysis". J Am Coll Cardiol Img 2020;13:25-40.

    View ArticleGoogle Scholar
  • 33. Kajimoto K., Sato N., Takano T. "Investigators of the Acute Decompensated Heart Failure Syndromes (ATTEND) Registry. Functional mitral regurgitation at discharge and outcomes in patients hospitalized for acute decompensated heart failure with a preserved or reduced ejection fraction". Eur J Heart Fail 2016;18:1051-1059.

    CrossrefMedlineGoogle Scholar
  • 34. Deferm S., Bertrand P.B., Verbrugge F.H., et al. "Atrial functional mitral regurgitation: JACC review topic of the week". J Am Coll Cardiol 2019;73:2465-2476.

    View ArticleGoogle Scholar
  • 35. Melenovsky V., Hwang S.J., Redfield M.M., Zakeri R., Lin G., Borlaug B.A. "Left atrial remodeling and function in advanced heart failure with preserved or reduced ejection fraction". Circ Heart Fail 2015;8:295-303.

    CrossrefMedlineGoogle Scholar
  • 36. Guazzi M., Naeije R. "Pulmonary hypertension in heart failure: pathophysiology, pathobiology, and emerging clinical perspectives". J Am Coll Cardiol 2017;69:1718-1734.

    View ArticleGoogle Scholar
  • 37. Ho J.E., Zern E.K., Lau E.S., et al. "Exercise pulmonary hypertension predicts clinical outcomes in patients with dyspnea on effort". J Am Coll Cardiol 2020;75:17-26.

    View ArticleGoogle Scholar
  • 38. Tang W.H.W., Wilcox J.D., Jacob M.S., et al. "Comprehensive diagnostic evaluation of cardiovascular physiology in patients with pulmonary vascular disease: insights from the PVDOMICS Program". Circ Heart Fail 2020;13:e006363.

    CrossrefGoogle Scholar
  • 39. Fayyaz A.U., Edwards W.D., Maleszewski J.J., et al. "Global pulmonary vascular remodeling in pulmonary hypertension associated with heart failure and preserved or reduced ejection fraction". Circulation 2018;137:1796-1810.

    CrossrefMedlineGoogle Scholar
  • 40. Hunt J.M., Bethea B., Liu X., et al. "Pulmonary veins in the normal lung and pulmonary hypertension due to left heart disease". Am J Physiol Lung Cell Mol Physiol 2013;305:L725-L736.

    CrossrefMedlineGoogle Scholar
  • 41. Nguyen Q.T., Jalloul Nsaibia M., Sirois M.G., et al. "PBI-4050 reduces pulmonary hypertension, lung fibrosis and right ventricular dysfunction in heart failure". Cardiovasc Res 2020;116:171-182.

    MedlineGoogle Scholar
  • 42. Assad T.R., Hemnes A.R., Larkin E.K., et al. "Clinical and biological insights into combined post- and pre-capillary pulmonary hypertension". J Am Coll Cardiol 2016;68:2525-2536.

    View ArticleGoogle Scholar
  • 43. Olson T.P., Johnson B.D., Borlaug B.A. "Impaired pulmonary diffusion in heart failure with preserved ejection fraction". J Am Coll Cardiol HF 2016;4:490-498.

    Google Scholar
  • 44. Ranchoux B., Nadeau V., Bourgeois A., et al. "Metabolic syndrome exacerbates pulmonary hypertension due to left heart disease". Circ Res 2019;125:449-466.

    CrossrefMedlineGoogle Scholar
  • 45. Moral-Sanz J., Lopez-Lopez J.G., Menendez C., et al. "Different patterns of pulmonary vascular disease induced by type 1 diabetes and moderate hypoxia in rats". Exp Physiol 2012;97:676-686.

    CrossrefMedlineGoogle Scholar
  • 46. Cayir A., Ugan R.A., Albayrak A., et al. "The lung endothelin system: a potent therapeutic target with bosentan for the amelioration of lung alterations in a rat model of diabetes mellitus". J Endocrinol Invest 2015;38:987-998.

    CrossrefMedlineGoogle Scholar
  • 47. Clemmer J.S., Xiang L., Lu S., Mittwede P.N., Hester R.L. "Hyperglycemia-mediated oxidative stress increases pulmonary vascular permeability". Microcirculation 2016;23:221-229.

    CrossrefMedlineGoogle Scholar
  • 48. Padang R., Chandrashekar N., Indrabhinduwat M., et al. "Aetiology and outcomes of severe right ventricular dysfunction". Eur Heart J 2020;41:1273-1282.

    CrossrefMedlineGoogle Scholar
  • 49. Benfari G., Antoine C., Miller W.L., et al. "Excess mortality associated with functional tricuspid regurgitation complicating heart failure with reduced ejection fraction". Circulation 2019;140:196-206.

    CrossrefMedlineGoogle Scholar
  • 50. Obokata M., Reddy Y.N.V., Melenovsky V., Pislaru S., Borlaug B.A. "Deterioration in right ventricular structure and function over time in patients with heart failure and preserved ejection fraction". Eur Heart J 2019;40:689-697.

    CrossrefMedlineGoogle Scholar
  • 51. Patel R.B., Li E., Benefield B.C., et al. "Diffuse right ventricular fibrosis in heart failure with preserved ejection fraction and pulmonary hypertension". ESC Heart Fail 2020;7:253-263.

    MedlineGoogle Scholar
  • 52. Hoeper M.M., Lam C.S.P., Vachiery J.L., et al. "Pulmonary hypertension in heart failure with preserved ejection fraction: a plea for proper phenotyping and further research". Eur Heart J 2017;38:2869-2873.

    MedlineGoogle Scholar
  • 53. Hoendermis E.S., Liu L.C., Hummel Y.M., et al. "Effects of sildenafil on invasive haemodynamics and exercise capacity in heart failure patients with preserved ejection fraction and pulmonary hypertension: a randomized controlled trial". Eur Heart J 2015;36:2565-2573.

    CrossrefMedlineGoogle Scholar
  • 54. Garcia-Lunar I., Blanco I., Fernandez-Fiera L., et al. "Design of the b3-Adrenergic Agonist Treatment in Chronic Pulmonary Hypertension Secondary to Heart Failure Trial". J Am Coll Cardiol Basic to Transl Sci 2020;4:317-337.

    Google Scholar
  • 55. Reddy Y.N.V., Obokata M., Koepp K.E., Egbe A.C., Wiley B., Borlaug B.A. "The beta-adrenergic agonist albuterol improves pulmonary vascular reserve in heart failure with preserved ejection fraction". Circ Res 2019;124:306-314.

    CrossrefMedlineGoogle Scholar
  • 56. Lai Y.C., Tabima D.M., Dube J.J., et al. "SIRT3-AMP-activated protein kinase activation by nitrite and metformin improves hyperglycemia and normalizes pulmonary hypertension associated with heart failure with preserved ejection fraction". Circulation 2016;133:717-731.

    CrossrefMedlineGoogle Scholar
  • 57. Zhuang R., Wu J., Lin F., et al. "Fasudil preserves lung endothelial function and reduces pulmonary vascular remodeling in a rat model of end-stage pulmonary hypertension with left heart disease". Int J Mol Med 2018;42:1341-1352.

    MedlineGoogle Scholar

Footnotes

This investigation was supported by a grant from the Monzino Foundation (to Dr. Guazzi). The authors have reported that they have no relationships relevant to the contents of this paper to disclose.

The authors attest they are in compliance with human studies committees and animal welfare regulations of the authors’ institutions and Food and Drug Administration guidelines, including patient consent where appropriate. For more information, visit the JACC author instructions page.

Listen to this manuscript's audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org.