1
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
3
|
Sebastian SA, Co EL, Mahtani A, Padda I, Anam M, Mathew SS, Shahzadi A, Niazi M, Pawar S, Johal G. Heart Failure: Recent Advances and Breakthroughs. Dis Mon 2024; 70:101634. [PMID: 37704531 DOI: 10.1016/j.disamonth.2023.101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Heart failure (HF) is a common clinical condition encountered in various healthcare settings with a vast socioeconomic impact. Recent advancements in pharmacotherapy have led to the evolution of novel therapeutic agents with a decrease in hospitalization and mortality rates in HF with reduced left ventricular ejection fraction (HFrEF). Lately, the introduction of artificial intelligence (AI) to construct decision-making models for the early detection of HF has played a vital role in optimizing cardiovascular disease outcomes. In this review, we examine the newer therapies and evidence behind goal-directed medical therapy (GDMT) for managing HF. We also explore the application of AI and machine learning (ML) in HF, including early diagnosis and risk stratification for HFrEF.
Collapse
Affiliation(s)
| | - Edzel Lorraine Co
- University of Santo Tomas Faculty of Medicine and Surgery, Manila, Philippines
| | - Arun Mahtani
- Richmond University Medical Center/Mount Sinai, Staten Island, New York, USA
| | - Inderbir Padda
- Richmond University Medical Center/Mount Sinai, Staten Island, New York, USA
| | - Mahvish Anam
- Deccan College of Medical Sciences, Hyderabad, India
| | | | | | - Maha Niazi
- Royal Alexandra Hospital, Edmonton, Canada
| | | | - Gurpreet Johal
- Department of Cardiology, University of Washington, Valley Medical Center, Seattle, Washington, USA
| |
Collapse
|
4
|
De Luca M, Crisci G, Armentaro G, Cicco S, Talerico G, Bobbio E, Lanzafame L, Green CG, McLellan AG, Debiec R, Caferra P, Scicali R, Cannatà A, Israr MZ, Heaney LM, Salzano A. Endothelial Dysfunction and Heart Failure with Preserved Ejection Fraction-An Updated Review of the Literature. Life (Basel) 2023; 14:30. [PMID: 38255646 PMCID: PMC10817572 DOI: 10.3390/life14010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Heart failure (HF) is a clinical syndrome consisting of typical symptoms and signs due to structural and/or functional abnormalities of the heart, resulting in elevated intracardiac pressures and/or inadequate cardiac output. The vascular system plays a crucial role in the development and progression of HF regardless of ejection fraction, with endothelial dysfunction (ED) as one of the principal features of HF. The main ED manifestations (i.e., impaired endothelium-dependent vasodilation, increased oxidative stress, chronic inflammation, leukocyte adhesion, and endothelial cell senescence) affect the systemic and pulmonary haemodynamic and the renal and coronary circulation. The present review is aimed to discuss the contribution of ED to HF pathophysiology-in particular, HF with preserved ejection fraction-ED role in HF patients, and the possible effects of pharmacological and non-pharmacological approaches. For this purpose, relevant data from a literature search (PubMed, Scopus, EMBASE, and Medline) were reviewed. As a result, ED, assessed via venous occlusion plethysmography or flow-mediated dilation, was shown to be independently associated with poor outcomes in HF patients (e.g., mortality, cardiovascular events, and hospitalization due to worsening HF). In addition, SGLT2 inhibitors, endothelin antagonists, endothelial nitric oxide synthase cofactors, antioxidants, and exercise training were shown to positively modulate ED in HF. Despite the need for future research to better clarify the role of the vascular endothelium in HF, ED represents an interesting and promising potential therapeutic target.
Collapse
Affiliation(s)
- Mariarosaria De Luca
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy
- Italian Clinical Outcome Research and Reporting Program (I-CORRP), 80131 Naples, Italy
| | - Giulia Crisci
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy
- Italian Clinical Outcome Research and Reporting Program (I-CORRP), 80131 Naples, Italy
| | - Giuseppe Armentaro
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy
| | - Sebastiano Cicco
- Internal Medicine Unit “Guido Baccelli” and Arterial Hypertension Unit “Anna Maria Pirrelli”, Department of Precision and Regenerative Medicine and Jonic Area (DiMePReJ), University of Bari Aldo Moro, Azienda Ospedaliero-Universitaria Policlinico, 70124 Bari, Italy
| | | | - Emanuele Bobbio
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Kuggen, 417 56 Gothenburg, Sweden
| | - Lorena Lanzafame
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Christopher G. Green
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| | - Abbie G. McLellan
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| | - Radek Debiec
- Department of Cardiovascular Sciences, University of Leicester, Leicester (UK), IHR Leicester Biomedical Research Centre, Groby Road, Leicester LE3 9QP, UK
| | - Paolo Caferra
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Antonio Cannatà
- Department of Cardiology, King’s College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
- Department of Cardiovascular Sciences, Faculty of Life Sciences & Medicine, King’s College, London SE1 8WA, UK
| | - Muhammad Zubair Israr
- Department of Cardiovascular Sciences, University of Leicester, Leicester (UK), IHR Leicester Biomedical Research Centre, Groby Road, Leicester LE3 9QP, UK
| | - Liam M. Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| | - Andrea Salzano
- Cardiac Unit, AORN A Cardarelli, 80131 Naples, Italy
- Cardiac Unit, University Hospital of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| |
Collapse
|
5
|
Mollace R, Scarano F, Bava I, Carresi C, Maiuolo J, Tavernese A, Gliozzi M, Musolino V, Muscoli S, Palma E, Muscoli C, Salvemini D, Federici M, Macrì R, Mollace V. Modulation of the nitric oxide/cGMP pathway in cardiac contraction and relaxation: Potential role in heart failure treatment. Pharmacol Res 2023; 196:106931. [PMID: 37722519 DOI: 10.1016/j.phrs.2023.106931] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Evidence exists that heart failure (HF) has an overall impact of 1-2 % in the global population being often associated with comorbidities that contribute to increased disease prevalence, hospitalization, and mortality. Recent advances in pharmacological approaches have significantly improved clinical outcomes for patients with vascular injury and HF. Nevertheless, there remains an unmet need to clarify the crucial role of nitric oxide/cyclic guanosine 3',5'-monophosphate (NO/cGMP) signalling in cardiac contraction and relaxation, to better identify the key mechanisms involved in the pathophysiology of myocardial dysfunction both with reduced (HFrEF) as well as preserved ejection fraction (HFpEF). Indeed, NO signalling plays a crucial role in cardiovascular homeostasis and its dysregulation induces a significant increase in oxidative and nitrosative stress, producing anatomical and physiological cardiac alterations that can lead to heart failure. The present review aims to examine the molecular mechanisms involved in the bioavailability of NO and its modulation of downstream pathways. In particular, we focus on the main therapeutic targets and emphasize the recent evidence of preclinical and clinical studies, describing the different emerging therapeutic strategies developed to counteract NO impaired signalling and cardiovascular disease (CVD) development.
Collapse
Affiliation(s)
- Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Irene Bava
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Jessica Maiuolo
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Saverio Muscoli
- Division of Cardiology, Foundation PTV Polyclinic Tor Vergata, Rome 00133, Italy
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy.
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Renato Dulbecco Institute, Lamezia Terme, Catanzaro 88046, Italy.
| |
Collapse
|
6
|
Edmonston D, Sparks M, Rajagopal S, Wolf M. Sildenafil and Kidney Function in Heart Failure with Preserved Ejection Fraction. KIDNEY360 2023; 4:631-640. [PMID: 36978225 PMCID: PMC10278830 DOI: 10.34067/kid.0000000000000103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/17/2023] [Indexed: 03/30/2023]
Abstract
Key Points Sildenafil induced an acute effect on eGFR without change in the overall eGFR slope after 24 weeks in a heart failure with preserved ejection fraction (HFpEF) cohort. N -terminal pro–brain natriuretic peptide levels and baseline diuretic use were most strongly associated with eGFR decline in this HFpEF cohort. Long-term studies are required to determine sildenafil's influence on kidney function and outcomes in HFpEF. Background CKD worsens the prognosis for people with heart failure with preserved ejection fraction (HFpEF). In the Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Diastolic HFpEF (RELAX) trial, sildenafil decreased eGFR compared with placebo despite favorable kidney effects in preclinical models. Since acute eGFR decline precedes long-term kidney benefits for select medications, we assessed the influence of sildenafil on acute and chronic eGFR slopes. Methods The RELAX trial randomized 216 participants to placebo or sildenafil and assessed 24-week changes in cardiopulmonary exercise testing, cardiovascular imaging, and laboratory data. We applied linear mixed modeling to calculate the total, acute (0–12 weeks), and chronic (3–24 weeks) eGFR slopes by treatment. Using regression modeling, we assessed respective associations between eGFR slope and baseline data and clinical end points. We repeated the analyses using a binary outcome on the basis of a substantial (≥20%) decline in eGFR. Results The mean baseline eGFR was 60.8 ml/min per 1.73 m2, and the mean eGFR slope during follow-up was −3.21 ml/min per 1.73 m2 per year. Sildenafil did not alter total eGFR slope compared with placebo (difference +0.47 ml/min per 1.73 m2 per year, 95% confidence interval [CI], −6.63 to 7.57 ml/min per 1.73 m2 per year). Sildenafil users tended to experience a more negative acute eGFR slope (difference −3.15 ml/min per 1.73 m2 per year) and more positive chronic slope (+2.06 ml/min per 1.73 m2 per year) compared with placebo, but neither difference reached statistical significance. Baseline N -terminal pro–B-type natriuretic peptide and loop diuretic use were associated with worse eGFR trajectory regardless of treatment. Substantial eGFR decline was associated with increase in endothelin-1 and a greater risk of hospitalization or death (HR, 2.34, 95% CI, 1.21 to 4.53, P =0.01). Conclusions Sildenafil induced an acute effect on eGFR without change in the overall eGFR slope after 24 weeks in an HFpEF cohort, suggesting lack of long-term risk related to early reduction in eGFR after initiating treatment. Long-term studies are needed to determine the effect of sildenafil on kidney function in HFpEF.
Collapse
Affiliation(s)
- Daniel Edmonston
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Matthew Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Renal Section, Durham VA Health Care System, Durham, North Carolina
| | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
7
|
Joshi SR, Atabay EK, Liu J, Ding Y, Briscoe SD, Alexander MJ, Andre P, Kumar R, Li G. Sotatercept analog improves cardiopulmonary remodeling and pulmonary hypertension in experimental left heart failure. Front Cardiovasc Med 2023; 10:1064290. [PMID: 36910526 PMCID: PMC9996114 DOI: 10.3389/fcvm.2023.1064290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/19/2023] [Indexed: 02/25/2023] Open
Abstract
Pulmonary hypertension due to left heart disease (PH-LHD) is the most frequent manifestation of PH but lacks any approved treatment. Activin receptor type IIA-Fc fusion protein (ActRIIA-Fc) was found previously to be efficacious in experimental and human pulmonary arterial hypertension (PAH). Here we tested the hypothesis that ActRIIA-Fc improves pulmonary vascular remodeling and alleviates PH in models of PH-LHD, specifically in subtypes of heart failure with reduced ejection fraction (PH-HFrEF) and preserved ejection fraction (PH-HFpEF). Treatment with murine ActRIIA-Fc reduced cardiac remodeling and improved cardiac function in two mouse models of left heart disease without PH, confirming that this inhibitor of activin-class ligand signaling can exert cardioprotective effects in heart failure. In a mouse model of PH-HFrEF with prolonged pressure overload caused by transverse aortic constriction, ActRIIA-Fc treatment significantly reduced pulmonary vascular remodeling, pulmonary fibrosis, and pulmonary hypertension while exerting beneficial structural, functional, and histological effects on both the left and right heart. Additionally, in an obese ZSF1-SU5416 rat model of PH-HFpEF with metabolic dysregulation, therapeutic treatment with ActRIIA-Fc normalized SMAD3 overactivation in pulmonary vascular and perivascular cells, reversed pathologic pulmonary vascular and cardiac remodeling, improved pulmonary and cardiac fibrosis, alleviated PH, and produced marked functional improvements in both cardiac ventricles. Studies in vitro revealed that treatment with ActRIIA-Fc prevents an abnormal, glucose-induced, activin-mediated, migratory phenotype in human pulmonary artery smooth muscle cells, providing a mechanism by which ActRIIA-Fc could exert therapeutic effects in experimental PH-HFpEF with metabolic dysregulation. Our results demonstrate that ActRIIA-Fc broadly corrects cardiopulmonary structure and function in experimental PH-LHD, including models of PH-HFrEF and PH-HFpEF, leading to alleviation of PH under diverse pathophysiological conditions. These findings highlight the important pathogenic contributions of activin-class ligands in multiple forms of experimental PH and support ongoing clinical evaluation of human ActRIIA-Fc (sotatercept) in patients with PH-HFpEF.
Collapse
Affiliation(s)
- Sachindra R Joshi
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| | - Elif Karaca Atabay
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| | - Jun Liu
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| | - Yan Ding
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| | - Steven D Briscoe
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| | - Mark J Alexander
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| | - Patrick Andre
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| | - Ravindra Kumar
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| | - Gang Li
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Rahway, NJ, United States
| |
Collapse
|
8
|
ZHANG J, WEI X, LI X, YUAN Y, DOU Y, SHI Y, XIE P, ZHOU M, ZHAO J, LI M, ZHANG S, ZHU R, TIAN Y, TAN H, TIAN F. Shunxin decoction improves diastolic function in rats with heart failure with preserved ejection fraction induced by abdominal aorta constriction through cyclic guanosine monophosphate-dependent protein kinase Signaling Pathway. J TRADIT CHIN MED 2022; 42:764-772. [PMID: 36083484 PMCID: PMC9924685 DOI: 10.19852/j.cnki.jtcm.20220519.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/12/2021] [Indexed: 04/16/2023]
Abstract
OBJECTIVE To determine whether Shunxin decoction improves diastolic function in rats with heart failure with preserved ejection fraction (HFpEF) by regulating the cyclic guanosine monophosphate-dependent protein kinase (cGMP-PKG) signaling pathway. METHODS Except for control group 8 and sham surgery group 8, the remaining 32 male Sprague-Dawlay rats were developed into HFpEF rat models using the abdominal aorta constriction method. These rats in the HFpEF model were randomly divided into the model group, the Shunxin high-dose group, the Shunxin low-dose group, and the Qiliqiangxin capsule group. The three groups received high-dose Shunxin decoction, low-dose Shunxin decoction, and Qiliqiangxin capsule by gavage, respectively, for 14 d. After the intervention, the diastolic function of each rat was evaluated by testing E/A, heart index, hematoxylin-eosin staining, Masson, myocardial ultrastructure, and N-terminal pro-brain natriuretic peptide (NT-proBNP). The Bioinformatics Analysis Tool for Molecular Mechanism of Traditional Chinese Medicine (BATMAN-TCM) software was used to predict targets for which Shunxin decoction acts on the cGMP-PKG pathway. Natriuretic peptide receptor A (NPRA) and guanylate cyclase (GC) were detected by immunohistochemistry, and eNOS, phosphodiesterase 5A (PDE5A), and cGMP-dependent protein kinase 1(PKG I) were determined by Western blotting. RESULTS Compared to the model group, the thickness of the interventricular septum at the end of diastole (IVSd) and the thickness of the posterior wall at the end of diastole (PWd) of the Shunxin decoction high-dose group, Shunxin decoction low-dose group, and Qiliqiangxin capsule group were all significantly reduced ( < 0.01). Furthermore, Shunxin decoction high-dose group E/A value was decreased ( < 0.01). Compared to the model group, the expression of NPRA and GC increased in the Shunxin decoction low-dose group and the Qiliqiangxin capsule group ( < 0.01). Compared to the model group, the expressions of eNOS and PKG I increased ( < 0.05) in the Shunxin decoction high-dose group. The expression of PDE5A expression decreased in the myocardium of the Shunxin decoction high-dose group, Shunxin decoction low-dose group, and Qiliqiangxin capsule group compared to the model group ( < 0.01). CONCLUSIONS Shunxin decoction can improve diastolic function in rats with HFpEF. It increases the expression of NPRA, GC, and eNOS in the myocardial cell cGMP-PKG signaling pathway, upregulates cGMP expression, decreases PDE5A expression to reduce the cGMP degradation. Thus, the cGMP continually stimulates PKG I, reversing myocardial hypertrophy and improving myocardial compliance in HFpEF rats.
Collapse
Affiliation(s)
- Jiaying ZHANG
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Xiangxiang WEI
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Xuefeng LI
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Yang YUAN
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Yinghuan DOU
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Yanbin SHI
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Ping XIE
- 2 Department of Cardiology, Gansu Provincial hospital, Lanzhou 730000, China
| | - Mengru ZHOU
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Junnan ZHAO
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Miao LI
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Shuwen ZHANG
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Rui ZHU
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Ying TIAN
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Hao TAN
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| | - Feifei TIAN
- 1 School of basic medical sciences, Institute of Integrated Chinese and Western Medicine,Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
9
|
Ovchinnikov A, Potekhina A, Belyavskiy E, Ageev F. Heart Failure with Preserved Ejection Fraction and Pulmonary Hypertension: Focus on Phosphodiesterase Inhibitors. Pharmaceuticals (Basel) 2022; 15:ph15081024. [PMID: 36015172 PMCID: PMC9414416 DOI: 10.3390/ph15081024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary hypertension (PH) is common in patients with heart failure with preserved ejection fraction (HFpEF). A chronic increase in mean left atrial pressure leads to passive remodeling in pulmonary veins and capillaries and modest PH (isolated postcapillary PH, Ipc-PH) and is not associated with significant right ventricular dysfunction. In approximately 20% of patients with HFpEF, "precapillary" alterations of pulmonary vasculature occur with the development of the combined pre- and post-capillary PH (Cpc-PH), pertaining to a poor prognosis. Current data indicate that pulmonary vasculopathy may be at least partially reversible and thus serves as a therapeutic target in HFpEF. Pulmonary vascular targeted therapies, including phosphodiesterase (PDE) inhibitors, may have a valuable role in the management of patients with PH-HFpEF. In studies of Cpc-PH and HFpEF, PDE type 5 inhibitors were effective in long-term follow-up, decreasing pulmonary artery pressure and improving RV contractility, whereas studies of Ipc-PH did not show any benefit. Randomized trials are essential to elucidate the actual value of PDE inhibition in selected patients with PH-HFpEF, especially in those with invasively confirmed Cpc-PH who are most likely to benefit from such treatment.
Collapse
Affiliation(s)
- Artem Ovchinnikov
- Out-Patient Department, Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, 3-d Cherepkovskaya St., 15a, 121552 Moscow, Russia
- Department of Clinical Functional Diagnostics, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, 127473 Moscow, Russia
- Correspondence: ; Tel.: +7-(495)-414-66-12 or +7-(916)-505-79-58; Fax: +7-(495)-414-66-12
| | - Alexandra Potekhina
- Out-Patient Department, Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, 3-d Cherepkovskaya St., 15a, 121552 Moscow, Russia
| | - Evgeny Belyavskiy
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Augustenburger Platz, 13353 Berlin, Germany
| | - Fail Ageev
- Out-Patient Department, Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, 3-d Cherepkovskaya St., 15a, 121552 Moscow, Russia
| |
Collapse
|
10
|
Abstract
Chronic heart failure is one of the most common causes of hospitalization and death in industrialized countries. Demographic changes with an aging population are expected to further increase the prevalence of chronic heart failure. The associated increase in comorbidities in patients with chronic heart failure leads to a less favorable prognosis for survival. A selection of the major comorbidities discussed in this review—along with prevalence, impact on prognosis, treatment approaches, and current study status—include atrial fibrillation, arterial hypertension, coronary artery disease, coronary microvascular dysfunction, renal dysfunction, type 2 diabetes, sleep apnea, reduced lymphatic reserve, and the effects on oxygen utilization and physical activity. The complex clinical picture of heart failure with preserved ejection fraction (HFpEF) remains challenging in the nearly absence of evidence-based therapy. Except for comorbidity-specific guidelines, no HFpEF-specific treatment of comorbidities can be recommended at this time. Optimized care is becoming increasingly relevant to reducing hospitalizations through a seamless inpatient and outpatient care structure. Current treatment is focused on symptom relief and management of associated comorbidities. Therefore, prevention through early minimization of risk factors currently remains the best approach.
Collapse
|
11
|
Phosphodiesterase inhibitor for heart failure with preserved ejection fraction: a systematic review and meta-analysis. Saudi Pharm J 2022; 30:1079-1087. [PMID: 36164567 PMCID: PMC9508622 DOI: 10.1016/j.jsps.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/26/2022] [Indexed: 11/22/2022] Open
|
12
|
Grandin EW, Gulati G, Nunez JI, Kennedy K, Rame JE, Atluri P, Pagani FD, Kirklin JK, Kormos RL, Teuteberg J, Kiernan M. Outcomes With Phosphodiesterase-5 Inhibitor Use After Left Ventricular Assist Device: An STS-INTERMACS Analysis. Circ Heart Fail 2022; 15:e008613. [PMID: 35332780 PMCID: PMC9205418 DOI: 10.1161/circheartfailure.121.008613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Elevated right ventricular afterload following continuous-flow left ventricular assist device (CF-LVAD) may contribute to late right heart failure (LRHF). PDE5i (phosphodiesterase-5 inhibitors) are used to treat pulmonary hypertension and right heart dysfunction after CF-LVAD, but their impact on outcomes is uncertain. METHODS We queried Interagency Registry for Mechanically Assisted Circulatory Support from 2012 to 2017 for adults receiving a primary CF-LVAD and surviving ≥30 days from index discharge. Patients receiving early PDE5i (ePDE5i) at 1 month were propensity-matched 1:1 with controls. The primary outcome was the cumulative incidence of LRHF, defined using prevailing Interagency Registry for Mechanically Assisted Circulatory Support criteria; secondary outcomes included all-cause mortality and major bleeding. RESULTS Among 9627 CF-LVAD recipients analyzed, 2463 (25.6%) received ePDE5i and 1600 were propensity-matched 1:1 with controls. Before implant, ePDE5i patients had more severe RV dysfunction (13.1% versus 9.6%) and higher pulmonary vascular resistance (2.8±2.7 versus 2.2±2.4 WU), both P<0.001, but clinical factors were well-balanced after propensity-matching. In the unmatched cohort, ePDE5i patients had a higher 3-year cumulative incidence of LRHF, mortality, and major bleeding, but these differences were attenuated in the propensity-matched cohort: LRHF 40.8% versus 35.7% (hazard ratio, 1.14 [95% CI, 0.99-1.32]; P=0.07); mortality 38.6% versus 35.8% (hazard ratio, 0.99 [95% CI, 0.86-1.15]; P=0.93); major bleeding 51.2% versus 46.0% (hazard ratio, 1.12 [95% CI, 0.99-1.27]; P=0.06). CONCLUSIONS Compared with propensity-matched controls, adult CF-LVAD patients receiving ePDE5i had similar rates of LRHF, mortality, and major bleeding. While intrinsic patient risk factors likely account for more adverse outcomes with ePDE5i in the unmatched cohort, there is no obvious benefit of ePDE5i in the LVAD population.
Collapse
Affiliation(s)
- E. Wilson Grandin
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston MA
- Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston MA
| | - Gaurav Gulati
- Cardiovascular Center, Tufts Medical Center, Boston MA
| | - Jose I Nunez
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston MA
| | - Kevin Kennedy
- Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston MA
| | - J Eduardo Rame
- Division of Cardiology, Jefferson Heart Institute, Philadelphia, PA
| | - Pavan Atluri
- Division of Cardiothoracic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Francis D Pagani
- Division of Cardiothoracic Surgery, University of Michigan School of Medicine, Ann Arbor, MI
| | - James K Kirklin
- Division of Cardiothoracic Surgery, University of Alabama Birmingham School of Medicine, Birmingham, AL
| | - Robert L Kormos
- Division of Cardiothoracic Surgery, University of Pittsburgh; Abbott Laboratories, Austin, TX
| | - Jeffrey Teuteberg
- Division of Cardiovascular Medicine, Stanford University Medical Center, Stanford, CA
| | | |
Collapse
|
13
|
Weerts J, Mourmans SGJ, Barandiarán Aizpurua A, Schroen BLM, Knackstedt C, Eringa E, Houben AJHM, van Empel VPM. The Role of Systemic Microvascular Dysfunction in Heart Failure with Preserved Ejection Fraction. Biomolecules 2022; 12:biom12020278. [PMID: 35204779 PMCID: PMC8961612 DOI: 10.3390/biom12020278] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 02/06/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a condition with increasing incidence, leading to a health care problem of epidemic proportions for which no curative treatments exist. Consequently, an urge exists to better understand the pathophysiology of HFpEF. Accumulating evidence suggests a key pathophysiological role for coronary microvascular dysfunction (MVD), with an underlying mechanism of low-grade pro-inflammatory state caused by systemic comorbidities. The systemic entity of comorbidities and inflammation in HFpEF imply that patients develop HFpEF due to systemic mechanisms causing coronary MVD, or systemic MVD. The absence or presence of peripheral MVD in HFpEF would reflect HFpEF being predominantly a cardiac or a systemic disease. Here, we will review the current state of the art of cardiac and systemic microvascular dysfunction in HFpEF (Graphical Abstract), resulting in future perspectives on new diagnostic modalities and therapeutic strategies.
Collapse
Affiliation(s)
- Jerremy Weerts
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
- Correspondence: ; Tel.: +31-43-387-7097
| | - Sanne G. J. Mourmans
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Arantxa Barandiarán Aizpurua
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Blanche L. M. Schroen
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Christian Knackstedt
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| | - Etto Eringa
- Department of Physiology, CARIM School for Cardiovascular Diseases, Maastricht University, 6211 LK Maastricht, The Netherlands;
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Alfons J. H. M. Houben
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands;
| | - Vanessa P. M. van Empel
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands; (S.G.J.M.); (A.B.A.); (B.L.M.S.); (C.K.); (V.P.M.v.E.)
| |
Collapse
|
14
|
Inampudi C, Silverman D, Simon MA, Leary PJ, Sharma K, Houston BA, Vachiéry JL, Haddad F, Tedford RJ. Pulmonary Hypertension in the Context of Heart Failure With Preserved Ejection Fraction. Chest 2021; 160:2232-2246. [PMID: 34391755 PMCID: PMC8727853 DOI: 10.1016/j.chest.2021.08.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is the most common form of heart failure and frequently is associated with pulmonary hypertension (PH). HFpEF associated with PH may be difficult to distinguish from precapillary forms of PH, although this distinction is crucial because therapeutic pathways are divergent for the two conditions. A comprehensive and systematic approach using history, clinical examination, and noninvasive and invasive evaluation with and without provocative testing may be necessary for accurate diagnosis and phenotyping. After diagnosis, HFpEF associated with PH can be subdivided into isolated postcapillary pulmonary hypertension (IpcPH) and combined postcapillary and precapillary pulmonary hypertension (CpcPH) based on the presence or absence of elevated pulmonary vascular resistance. CpcPH portends a worse prognosis than IpcPH. Despite its association with reduced functional capacity and quality of life, heart failure hospitalizations, and higher mortality, therapeutic options focused on PH for HFpEF associated with PH remain limited. In this review, we aim to provide an updated overview on clinical definitions and hemodynamically characterized phenotypes of PH, pathophysiologic features, therapeutic strategies, and ongoing challenges in this patient population.
Collapse
Affiliation(s)
- Chakradhari Inampudi
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Daniel Silverman
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Marc A Simon
- Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco
| | - Peter J Leary
- Department of Medicine, University of Washington, Seattle, WA
| | - Kavita Sharma
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD
| | - Brian A Houston
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Jean-Luc Vachiéry
- Département de Cardiologie Cliniques, Universitaires de Bruxelles-Hôpital Erasme, Brussels, Belgium
| | - Francois Haddad
- Division of Cardiovascular Medicine and Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
| | - Ryan J Tedford
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC.
| |
Collapse
|
15
|
Wan S, McKie PM, Slusser JP, Burnett JC, Hodge DO, Chen HH. Effects of phosphodiesterase V inhibition alone and in combination with BNP on cardiovascular and renal response to volume load in human preclinical diastolic dysfunction. Physiol Rep 2021; 9:e14974. [PMID: 34405565 PMCID: PMC8371344 DOI: 10.14814/phy2.14974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/05/2021] [Indexed: 01/22/2023] Open
Abstract
Preclinical diastolic dysfunction (PDD) results in impaired cardiorenal response to volume load (VL) which may contribute to the progression to clinical heart failure with preserved ejection fraction (HFpEF). The objective was to evaluate if phosphodiesterase V inhibition (PDEVI) alone or combination PDEVI plus B-type natriuretic peptide (BNP) administration will correct the impaired cardiorenal response to VL in PDD. A randomized double-blinded placebo-controlled cross-over study was conducted in 20 subjects with PDD, defined as left ventricular ejection fraction (LVEF) >50% with moderate or severe diastolic dysfunction by Doppler echocardiography and without HF diagnosis or symptoms. Effects of PDEVI with oral tadalafil alone and tadalafil plus subcutaneous (SC) BNP, administered prior to acute volume loading, were assessed. Tadalafil alone did not result in improvement in cardiac response to VL, as measured by LVEF, LV end diastolic volume, left atrial volume (LAV), or right ventricular systolic pressure (RVSP). Tadalafil plus SC BNP resulted in improved cardiac response to VL, with increased LVEF (4.1 vs. 1.8%, p = 0.08) and heart rate (4.3 vs. 1.6 bpm, p = 0.08), and reductions in both LAV (-4.3 ± 10.4 vs. 2.8 ± 6.6 ml, p = 0.03) and RVSP (-4.0 ± 3.0 vs. 2.1 ± 6.0 mmHg, p < 0.01) versus tadalafil alone. Plasma and urinary cyclic guanosine monophosphate (cGMP) excretion levels were higher (11.3 ± 12.3 vs. 1.7 ± 3.8 pmol/ml, 1851.0 ± 1386.4 vs. 173.4 ± 517.9 pmol/min, p < 0.01) with tadalafil plus SC BNP versus tadalafil alone. There was no improvement in renal response as measured by GFR, renal plasma flow, sodium excretion, and urine flow with tadalafil plus SC BNP compared to tadalafil alone. In subjects with PDD, tadalafil alone resulted in no improvement in cardiac adaptation, while tadalafil and SC BNP resulted in enhanced cardiac adaptation to VL. TRIAL REGISTRATION: ClinicalTrials.gov NCT01544998.
Collapse
Affiliation(s)
- Siu‐Hin Wan
- Division of CardiologyDepartment of Internal MedicineUniversity of Texas—Southwestern Medical CenterDallasTXUSA
| | - Paul M. McKie
- Department of Cardiovascular DiseasesCardiorenal Research LaboratoryMayo Clinic and FoundationRochesterMNUSA
| | - Joshua P. Slusser
- Department of Health Sciences ResearchMayo Clinic and FoundationRochesterMNUSA
| | - John C. Burnett
- Department of Cardiovascular DiseasesCardiorenal Research LaboratoryMayo Clinic and FoundationRochesterMNUSA
| | - David O. Hodge
- Department of Health Sciences ResearchMayo Clinic and FoundationRochesterMNUSA
| | - Horng H. Chen
- Department of Cardiovascular DiseasesCardiorenal Research LaboratoryMayo Clinic and FoundationRochesterMNUSA
| |
Collapse
|
16
|
Egbe AC, Miranda WR, Anderson JH, Borlaug BA. Hemodynamic and Clinical Implications of Impaired Pulmonary Vascular Reserve in the Fontan Circulation. J Am Coll Cardiol 2021; 76:2755-2763. [PMID: 33272370 DOI: 10.1016/j.jacc.2020.10.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/11/2020] [Accepted: 10/05/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Pulmonary vascular disease, pulmonary endothelial dysfunction, liver fibrosis, renal disease, and exercise intolerance are common in adults with Fontan physiology. Although the pathophysiologic mechanisms linking these phenomena have been studied, certain aspects are not well understood. OBJECTIVES This study hypothesized that impaired pulmonary vascular reserve (VR) plays a central role linking these abnormalities, and that patients with abnormal pulmonary VR with exercise, compared with patients with normal VR, would display poorer pulmonary endothelial function, greater liver stiffness, more renal dysfunction, and poorer exercise capacity. METHODS Symptomatic adults with the Fontan palliation (n = 29) underwent invasive cardiopulmonary exercise testing, echocardiography, and assessment of microvascular function. Abnormal pulmonary VR was defined by the slope of increase in pulmonary pressure relative to cardiac output with exercise >3 mm Hg/l/min. Pulmonary endothelial function was assessed using reactive hyperemia index. End-organ function was assessed using magnetic resonance elastography-derived liver stiffness, glomerular filtration rate, N-terminal pro-B-type natriuretic peptide, and peak oxygen consumption (Vo2). RESULTS Compared with individuals with normal VR (n = 8), those with abnormal VR (n = 21) displayed higher central and pulmonary venous pressures, and more severely impaired cardiac output and stroke volume responses to exertion, but similar pulmonary vascular resistance at rest. Patients with abnormal VR displayed more severely impaired reactive hyperemia index, increased liver stiffness, lower glomerular filtration rate, higher N-terminal pro-B-type natriuretic peptide, and lower peak Vo2. As compared to pulmonary vascular resistance at rest, slope of increase in pulmonary pressure relative to cardiac output displayed stronger correlations with reactive hyperemia index (r = -0.63 vs. r = -0.31; Meng test p = 0.009), magnetic resonance elastography-derived liver stiffness (r = 0.47 vs. r = 0.29; Meng test p = 0.07), glomerular filtration rate (r = -0.52 vs. r = -0.24; Meng test p = 0.03), N-terminal pro-B-type natriuretic peptide (r = 0.56 vs. r = 0.17; Meng test p = 0.02), and peak Vo2 (r = -0.63 vs. r = -0.26; Meng test p = 0.02). CONCLUSIONS Pulmonary vascular limitations in Fontan physiology are related to pulmonary endothelial and end-organ dysfunction, suggesting a mechanistic link between these commonly observed findings, and these abnormalities are more apparent during exercise testing, with little relationship at rest.
Collapse
Affiliation(s)
- Alexander C Egbe
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.
| | - William R Miranda
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jason H Anderson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
17
|
Wu C, Cheng J, Huang C, Chen Z, Chen S, Lin L. Iloprost and exercise haemodynamics in heart failure with preserved ejection fraction—the ILO‐HOPE randomised controlled trial. Br J Clin Pharmacol 2021. [DOI: 10.1111/bcp.14484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Cho‐Kai Wu
- Division of Cardiology, Department of Internal Medicine National Taiwan University College of Medicine and Hospital Taipei Taiwan
| | - Jen‐Fang Cheng
- Division of Cardiology, Department of Internal Medicine National Taiwan University College of Medicine and Hospital Taipei Taiwan
- Division of Cardiology, Department of Internal Medicine Pingtung Hospital, Ministry of Health and Welfare Pingtung Taiwan
- Graduate Institute of Clinical Medicine National Taiwan University College of Medicine and National Taiwan University Hospital Taipei Taiwan
| | - Chen‐Yu Huang
- Division of Cardiology, Department of Internal Medicine National Taiwan University College of Medicine and Hospital Taipei Taiwan
- Division of Cardiology, Department of Internal Medicine Kinmen Hospital, Ministry of Health and Welfare Kinmen Taiwan
| | - Zheng‐Wei Chen
- Division of Cardiology, Department of Internal Medicine National Taiwan University College of Medicine and Hospital Taipei Taiwan
- Division of Cardiology, Department of Internal Medicine National Taiwan University College of Medicine and Hospital, Yun‐Lin Branch Dou‐Liu Taiwan
| | - Ssu‐Yuan Chen
- Department of Physical Medicine & Rehabilitation Fu Jen Catholic University Hospital and Fu Jen Catholic University School of Medicine New Taipei City Taiwan
- Department of Physical Medicine & Rehabilitation National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Lian‐Yu Lin
- Division of Cardiology, Department of Internal Medicine National Taiwan University College of Medicine and Hospital Taipei Taiwan
| |
Collapse
|
18
|
Monzo L, Reichenbach A, Al-Hiti H, Borlaug BA, Havlenova T, Solar N, Tupy M, Ters J, Kautzner J, Melenovsky V. Acute Unloading Effects of Sildenafil Enhance Right Ventricular-Pulmonary Artery Coupling in Heart Failure. J Card Fail 2020; 27:224-232. [PMID: 33232820 DOI: 10.1016/j.cardfail.2020.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/02/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Phosphodiesterase-5A inhibitors (PDE5i) are sometimes used in patients with advanced heart failure with reduced ejection fraction before heart transplant or left ventricular assist device implantation to decrease right ventricular (RV) afterload and mitigate the risk of right heart failure. Conflicting evidence exists regarding the impact of these drugs on RV contractility. The aim of this study was to explore the acute effects of PDE5i on ventricular-vascular coupling and load-independent RV contractility. METHODS Twenty-two patients underwent right heart catheterization and gated equilibrium blood pool single photon emission computed tomography, before and after 20 mg intravenous sildenafil. Single photon emission computed tomography and right heart catheterization-derived data were used to calculate RV loading and contractility. RESULTS PDE5i induced a decrease in the right atrial pressure (-43%), pulmonary artery (PA) mean pressure (-26%), and PA wedge pressure (PAWP; -23%), with favorable reductions in pulmonary vascular resistance (-41%) and PA elastance (-40%), and increased cardiac output (+13%) (all P < 0.01). The RV ejection fraction increased with sildenafil (+20%), with no change of RV contractility (P = 0.74), indicating that the improvement in the RV ejection fraction was related to enhanced RV-PA coupling (r = 0.59, P = 0.004) by a decrease in the ventricular load. RV diastolic compliance increased with sildenafil. The decrease in the PAWP correlated with RV end-diastolic volume decrease; no relationship was observed with the change in LV transmural pressure, suggesting decreased pericardial constraint. CONCLUSIONS Acute PDE5i administration has profound RV afterload-reducing effects, improves the RVEF, decreases RV volumes, and decreases the PAWP, predominantly through relief of pericardial constraint, without effects on RV chamber contractility. These findings support further study of PDE5i in protection of RV function in advanced heart failure with reduced ejection fraction who are at risk of RV failure.
Collapse
Affiliation(s)
- Luca Monzo
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic; Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Adrian Reichenbach
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Hikmet Al-Hiti
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Tereza Havlenova
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Nevenka Solar
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Marek Tupy
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Jiri Ters
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Josef Kautzner
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Vojtech Melenovsky
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic.
| |
Collapse
|
19
|
Barbagallo F, Campolo F, Franceschini E, Crecca E, Pofi R, Isidori AM, Venneri MA. PDE5 Inhibitors in Type 2 Diabetes Cardiovascular Complications. ENDOCRINES 2020; 1:90-101. [DOI: 10.3390/endocrines1020009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
Pharmacological inhibition of Phosphodiesterase type 5 (PDE5) proved its efficacy treating several pathological conditions, such as erectile dysfunction and pulmonary hypertension. Nowadays, its benefits on cardiovascular diseases are well documented, particularly in the treatment of type 2 diabetes (T2DM)-related cardiovascular complications. In this context, treatment of T2DM with PDE5 inhibitors, such as sildenafil, tadalafil or vardenafil ameliorates endothelial dysfunction both in patients and animal models through an augmented flow mediated dilation rate and an up-regulation of endothelial markers; it also reduces the inflammatory state by down-regulating inflammatory cytokines expression and improves diabetic cardiomyopathy and ischemia-reperfusion injury mainly through the activation of NO-cGMP-PKG pathway. The present review summarizes the state of art on PDE5 inhibition in the treatment of cardiovascular complications in T2DM.
Collapse
|
20
|
Patel RB, Mehta R, Redfield MM, Borlaug BA, Hernandez AF, Shah SJ, Dubin RF. Renal Dysfunction in Heart Failure With Preserved Ejection Fraction: Insights From the RELAX Trial. J Card Fail 2020; 26:233-242. [PMID: 31931098 DOI: 10.1016/j.cardfail.2020.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/17/2019] [Accepted: 01/03/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Patients with heart failure with preserved ejection fraction (HFpEF) and chronic kidney disease (CKD) represent a high-risk phenotype. The Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Heart Failure with Preserved Ejection Fraction (RELAX) trial enrolled a high proportion of CKD participants, allowing investigation into differences in HFpEF by CKD status. METHODS AND RESULTS Among 212 participants, we investigated the associations of CKD with biomarkers, cardiac structure, and exercise capacity, and identified predictors of change in estimated glomerular filtration rate (eGFR) over trial follow-up. CKD participants (eGFR ≤60 mL/min/1.73m2) were older, had more comorbidities, and had worse diastolic function. Lower eGFR was associated with higher levels of endothelin-1, N-terminal pro-B-type natriuretic peptide, aldosterone, uric acid, and biomarkers of fibrosis (P < .05 for all). Whereas lower eGFR was associated with worse peak oxygen consumption (VO2) after adjustment for demographics, clinical comorbidities, exercise modality, ejection fraction, and chronotropic index (β coefficient per 1 SD decrease in eGFR: -0.61, 95% CI: -1.01, -0.22, P = .002), this association was attenuated after further adjustment for hemoglobin (β coefficient: -0.26, 95% CI: -0.68, 0.16, P = .22). Hemoglobin mediated 35% of the association between eGFR and peak VO2. Sildenafil therapy was independently associated with worsening eGFR over the trial (β coefficient: -2.79, 95% CI: -5.34, -0.24, P = .03). CONCLUSION Renal dysfunction in HFpEF is characterized by echocardiographic and biomarker profiles indicative of more advanced disease, and reduced hemoglobin is a strong mediator of the association between renal dysfunction and low exercise capacity. Sildenafil therapy was associated with worsening of renal function in RELAX.
Collapse
Affiliation(s)
- Ravi B Patel
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Barry A Borlaug
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Adrian F Hernandez
- Division of Cardiology, Duke University Medical Center, Durham, North Carolina
| | - Sanjiv J Shah
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ruth F Dubin
- Division of Nephrology, San Francisco VA Medical Center/University of California, San Francisco, California
| |
Collapse
|
21
|
Cyclic nucleotide phosphodiesterases: New targets in the metabolic syndrome? Pharmacol Ther 2020; 208:107475. [PMID: 31926200 DOI: 10.1016/j.pharmthera.2020.107475] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Metabolic diseases have a tremendous impact on human morbidity and mortality. Numerous targets regulating adenosine monophosphate kinase (AMPK) have been identified for treating the metabolic syndrome (MetS), and many compounds are being used or developed to increase AMPK activity. In parallel, the cyclic nucleotide phosphodiesterase families (PDEs) have emerged as new therapeutic targets in cardiovascular diseases, as well as in non-resolved pathologies. Since some PDE subfamilies inactivate cAMP into 5'-AMP, while the beneficial effects in MetS are related to 5'-AMP-dependent activation of AMPK, an analysis of the various controversial relationships between PDEs and AMPK in MetS appears interesting. The present review will describe the various PDE families, AMPK and molecular mechanisms in the MetS and discuss the PDEs/PDE modulators related to the tissues involved, thus supporting the discovery of original molecules and the design of new therapeutic approaches in MetS.
Collapse
|
22
|
Reddy YNV, Obokata M, Koepp KE, Egbe AC, Wiley B, Borlaug BA. The β-Adrenergic Agonist Albuterol Improves Pulmonary Vascular Reserve in Heart Failure With Preserved Ejection Fraction. Circ Res 2019; 124:306-314. [PMID: 30582447 DOI: 10.1161/circresaha.118.313832] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
RATIONALE Pulmonary vascular resistance fails to decrease appropriately during exercise in patients with heart failure with preserved ejection fraction (HFpEF). Interventions that enhance pulmonary vasodilation might be beneficial in this cohort but could also worsen left atrial hypertension, exacerbating lung congestion. Intravenous β-agonists reduce pulmonary vascular resistance but are not suitable for chronic use. OBJECTIVE We hypothesized that the inhaled β-adrenergic agonist albuterol would improve pulmonary vasodilation during exercise in patients with HFpEF, without increasing left heart filling pressures. METHODS AND RESULTS We performed a randomized, double-blind, placebo-controlled trial testing the effects of inhaled albuterol on resting and exercise hemodynamics in subjects with HFpEF using high-fidelity micromanometer catheters and expired gas analysis. The primary end point was pulmonary vascular resistance during exercise. Subjects with HFpEF (n=30) underwent resting and exercise hemodynamic assessment and were then randomized 1:1 to inhaled, nebulized albuterol or placebo. Rest and exercise hemodynamic testing was then repeated. Albuterol improved the primary end point of exercise pulmonary vascular resistance as compared with placebo (-0.6±0.5 versus +0.1±0.7 WU; P=0.003). Albuterol enhanced cardiac output reserve and right ventricular pulmonary artery coupling, reduced right atrial and pulmonary artery pressures, improved pulmonary artery compliance, and enhanced left ventricular transmural distending pressure (all P <0.01), with no increase in pulmonary capillary hydrostatic pressures. CONCLUSIONS Albuterol improves pulmonary vascular reserve in patients with HFpEF without worsening left heart congestion. Further study is warranted to evaluate the chronic efficacy of β-agonists in HFpEF and other forms of pulmonary hypertension. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov . Unique identifier: NCT02885636.
Collapse
Affiliation(s)
- Yogesh N V Reddy
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Masaru Obokata
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Katlyn E Koepp
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Alexander C Egbe
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Brandon Wiley
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Barry A Borlaug
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
23
|
Phosphodiesterase-5 Inhibitor Therapy for Pulmonary Hypertension in the United States. Actual versus Recommended Use. Ann Am Thorac Soc 2019; 15:693-701. [PMID: 29485908 DOI: 10.1513/annalsats.201710-762oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RATIONALE Care of patients with pulmonary hypertension is complex. Although pulmonary vasodilators are effective for Group 1 pulmonary hypertension, clinical guidelines and the Choosing Wisely Campaign recommend against routine use for Groups 2 and 3 pulmonary hypertension (the most common types of pulmonary hypertension) because of a lack of benefit, potential for harm, and high cost ($10,000-$13,000 per patient per year treated). Little is known about how these medications are used in practice. OBJECTIVES To determine national patterns of phosphodiesterase-5 inhibitor prescribing for pulmonary hypertension in the Veterans Health Administration. METHODS Retrospective analysis of Veterans prescribed phosphodiesterase-5 inhibitor for pulmonary hypertension between 2005 and 2012 at any Veterans Health Administration site. Patients were identified by presence of an International Classification of Diseases, Ninth Revision, Clinical Modification diagnosis code for pulmonary hypertension and one or more outpatient prescriptions for daily phosphodiesterase-5 inhibitor therapy. We developed and validated, using gold-standard chart abstraction, an International Classification of Diseases, Ninth Revision, Clinical Modification-based algorithm to assign pulmonary hypertension group. Our primary outcome was the proportion of patients who received potentially inappropriate phosphodiesterase-5 inhibitor, as determined by guideline recommendations (Group 1 pulmonary hypertension: appropriate; Groups 2/3: potentially inappropriate; Groups 4/5: uncertain value), among all patients prescribed phosphodiesterase-5 inhibitor for pulmonary hypertension. Secondary outcomes included proportion of treated patients who received guideline-recommended right heart catheterization. RESULTS Among 108,777 Veterans with pulmonary hypertension, 2,790 (2.6% [95% confidence interval, 2.5-2.7%]) received daily phosphodiesterase-5 inhibitor therapy. Among treated patients, 541 (19.4% [95% confidence interval, 18.0-20.9%]) received appropriate treatment, 1,711 (61.3% [95% confidence interval, 59.5-63.1%]) potentially inappropriate treatment, and 358 (12.8% [95% confidence interval, 11.6-14.1%]) treatment of uncertain value. The number of potentially inappropriately treated patients per year increased substantially over the study period (53 in 2005, 748 in 2012). On the basis of chart abstraction in a randomly selected subset of patients treated with phosphodiesterase-5 inhibitor, half (110 of 230, 47.8% [95% confidence interval, 41.3-54.5%]) had documented right heart catheterization to confirm presence or type of pulmonary hypertension. After factoring presence of and data from right heart catheterization into our treatment appropriateness algorithm, only 11.7% (95% confidence interval, 8.0-16.8%) received clearly appropriate treatment. CONCLUSIONS Most Veterans with pulmonary hypertension do not receive phosphodiesterase-5 inhibitor therapy. However, among treated Veterans, almost two-thirds of phosphodiesterase-5 inhibitor prescriptions are inconsistent with pulmonary hypertension guidelines, exposing patients to potential harm and creating a financial burden on the healthcare system. Further study is warranted to clarify the effects of these prescription patterns on pulmonary hypertension outcomes.
Collapse
|
24
|
Blanton R, Gulati G, Kiernan M. Response by Blanton et al to Letter Regarding Article, "Preimplant Phosphodiesterase-5 Inhibitor Use Is Associated With Higher Rates of Severe Early Right Heart Failure After LVAD Implantation: An INTERMACS Analysis". Circ Heart Fail 2019; 12:e006465. [PMID: 31514516 DOI: 10.1161/circheartfailure.119.006465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Robert Blanton
- Cardiovascular Center, Division of Cardiology, Tufts Medical Center, Boston, MA (R.B., G.G., M.K.).,Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (R.B.)
| | - Gaurav Gulati
- Cardiovascular Center, Division of Cardiology, Tufts Medical Center, Boston, MA (R.B., G.G., M.K.)
| | - Michael Kiernan
- Cardiovascular Center, Division of Cardiology, Tufts Medical Center, Boston, MA (R.B., G.G., M.K.)
| |
Collapse
|
25
|
van de Wouw J, Broekhuizen M, Sorop O, Joles JA, Verhaar MC, Duncker DJ, Danser AHJ, Merkus D. Chronic Kidney Disease as a Risk Factor for Heart Failure With Preserved Ejection Fraction: A Focus on Microcirculatory Factors and Therapeutic Targets. Front Physiol 2019; 10:1108. [PMID: 31551803 PMCID: PMC6737277 DOI: 10.3389/fphys.2019.01108] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) and chronic kidney disease (CKD) co-exist, and it is estimated that about 50% of HF patients suffer from CKD. Although studies have been performed on the association between CKD and HF with reduced ejection fraction (HFrEF), less is known about the link between CKD and heart failure with preserved ejection fraction (HFpEF). Approximately, 50% of all patients with HF suffer from HFpEF, and this percentage is projected to rise in the coming years. Therapies for HFrEF are long established and considered quite successful. In contrast, clinical trials for treatment of HFpEF have all shown negative or disputable results. This is likely due to the multifactorial character and the lack of pathophysiological knowledge of HFpEF. The typical co-existence of HFpEF and CKD is partially due to common underlying comorbidities, such as hypertension, dyslipidemia and diabetes. Macrovascular changes accompanying CKD, such as hypertension and arterial stiffening, have been described to contribute to HFpEF development. Furthermore, several renal factors have a direct impact on the heart and/or coronary microvasculature and may underlie the association between CKD and HFpEF. These factors include: (1) activation of the renin-angiotensin-aldosterone system, (2) anemia, (3) hypercalcemia, hyperphosphatemia and increased levels of FGF-23, and (4) uremic toxins. This review critically discusses the above factors, focusing on their potential contribution to coronary dysfunction, left ventricular stiffening, and delayed left ventricular relaxation. We further summarize the directions of novel treatment options for HFpEF based on the contribution of these renal drivers.
Collapse
Affiliation(s)
- Jens van de Wouw
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Michelle Broekhuizen
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Division of Neonatology, Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
26
|
Hutchings DC, Trafford AW, Callan PD. Letter by Hutchings et al Regarding Article, “Preimplant Phosphodiesterase-5 Inhibitor Use Is Associated With Higher Rates of Severe Early Right Heart Failure After Left Ventricular Assist Device Implantation : An INTERMACS Analysis”. Circ Heart Fail 2019; 12:e006410. [DOI: 10.1161/circheartfailure.119.006410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- David C. Hutchings
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, United Kingdom (D.C.H., A.W.T.)
- Department of Advanced Heart Failure and Cardiac Transplantation, Manchester University NHS Foundation Trust, United Kingdom (D.C.H., P.D.C.)
| | - Andrew W. Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, United Kingdom (D.C.H., A.W.T.)
| | - Paul D. Callan
- Department of Advanced Heart Failure and Cardiac Transplantation, Manchester University NHS Foundation Trust, United Kingdom (D.C.H., P.D.C.)
| |
Collapse
|
27
|
Alem MM. Endothelial Dysfunction in Chronic Heart Failure: Assessment, Findings, Significance, and Potential Therapeutic Targets. Int J Mol Sci 2019; 20:E3198. [PMID: 31261886 PMCID: PMC6651535 DOI: 10.3390/ijms20133198] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/13/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure (CHF) is a complex syndrome that results from structural and functional disturbances that affect the ability of the heart to supply oxygen to tissues. It largely affects and reduces the patient's quality of life, socio-economic status, and imposes great costs on health care systems worldwide. Endothelial dysfunction (ED) is a newly discovered phenomenon that contributes greatly to the pathophysiology of numerous cardiovascular conditions and commonly co-exists with chronic heart failure. However, the literature lacks clarity as to which heart failure patients might be affected, its significance in CHF patients, and its reversibility with pharmacological and non-pharmacological means. This review will emphasize all these points and summarize them for future researchers interested in vascular pathophysiology in this particular patient population. It will help to direct future studies for better characterization of these two phenomena for the potential discovery of therapeutic targets that might reduce future morbidity and mortality in this "at risk" population.
Collapse
Affiliation(s)
- Manal M Alem
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia.
| |
Collapse
|
28
|
Silverman DN, Shah SJ. Treatment of Heart Failure With Preserved Ejection Fraction (HFpEF): the Phenotype-Guided Approach. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2019; 21:20. [DOI: 10.1007/s11936-019-0709-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
D'Andrea S, Barbonetti A, Martorella A, Necozione S, Francavilla F, Francavilla S. Effect of prolonged treatment with phosphodiesterase-5-inhibitors on endothelial dysfunction in vascular diseases and vascular risk conditions: A systematic review analysis and meta-analysis of randomized double-blind placebo-controlled trials. Int J Clin Pract 2019; 73:e13296. [PMID: 30471172 DOI: 10.1111/ijcp.13296] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/15/2018] [Accepted: 11/18/2018] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE To challenge the argument that continuous use of phosphodiesterase-5-selective inhibitors may reduce endothelial cell dysfunction in patients with vascular diseases or vascular risk conditions. DESIGN This study included systematic reviews and meta-analysis of randomized double-blind placebo-controlled trials dealing with the prolonged use of phosphodiesterase-5-selective inhibitors. The risk of bias and quality of trials were assessed by the Cochrane algorithm. Fixed or random effect models, standardised mean differences and heterogeneity were estimated in the study. DATA SOURCES Systematic search for randomized double-blind placebo-controlled trials was done in PubMed, Scopus, CINAHL, Science direct and the Cochrane Library. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Randomized double-blind placebo-controlled trials reporting measures of endothelial cell dysfunction and/or endothelial cell activation were included. RESULTS On the whole, 469 subjects were allocated to the phosphodiesterase-5-selective inhibitor group, while 463 were assigned to the placebo group in 13 randomized double-blind placebo-controlled trials. Flow-mediated dilation of the brachial artery was found to improve after the administration of phosphodiesterase-5-selective inhibitors (P < 0.0001). The results were questioned by the elevated and uncorrectable heterogeneity (I2 = 92%) and the asymmetry of the funnel plot suggested a publication bias. Phosphodiesterase-5-selective inhibitors have no effect on endothelial cell dysfunction, as assessed in the resistance vessels by digital arterial tonometry. The blood level of endothelin-1 was observed to be decreased in phosphodiesterase-5-selective inhibitors arm (P = 0.03), although the effect disappeared once the publication bias and heterogeneity were corrected. The effect of phosphodiesterase-5-selective inhibitors on biomarkers of endothelial cell activation was found to be inconsistent. CONCLUSIONS The results on the benefits of a prolonged use of phosphodiesterase-5-selective inhibitors, with the objective of lowering endothelial cell dysfunction in patients with vascular diseases or vascular risk conditions are not convincing. This is because of the overall low quality of evidence, giving an unclear scientific support to this treatment. Systematic review registration: PROSPERO registration: CRD42017055399.
Collapse
Affiliation(s)
- Settimio D'Andrea
- Department of Life, Health and Environmental Sciences, Andrology Unit, University of L'Aquila, L'Aquila, Italy
| | - Arcangelo Barbonetti
- Department of Life, Health and Environmental Sciences, Andrology Unit, University of L'Aquila, L'Aquila, Italy
| | - Alessio Martorella
- Department of Life, Health and Environmental Sciences, Andrology Unit, University of L'Aquila, L'Aquila, Italy
| | | | - Felice Francavilla
- Department of Life, Health and Environmental Sciences, Andrology Unit, University of L'Aquila, L'Aquila, Italy
| | - Sandro Francavilla
- Department of Life, Health and Environmental Sciences, Andrology Unit, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
30
|
Alogna A, Schwarzl M, Manninger M, Hamdani N, Zirngast B, Kloth B, Steendijk P, Verderber J, Zweiker D, Westermann D, Blankenberg S, Maechler H, Tschöpe C, Linke WA, Marsche G, Pieske BM, Post H. Acute stimulation of the soluble guanylate cyclase does not impact on left ventricular capacitance in normal and hypertrophied porcine hearts in vivo. Am J Physiol Heart Circ Physiol 2018; 315:H669-H680. [PMID: 29727215 DOI: 10.1152/ajpheart.00510.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Experimental data indicate that stimulation of the nitric oxide-soluble guanylate cyclase(sGC)-cGMP-PKG pathway can increase left ventricular (LV) capacitance via phosphorylation of the myofilamental protein titin. We aimed to test whether acute pharmacological sGC stimulation with BAY 41-8543 would increase LV capacitance via titin phosphorylation in healthy and deoxycorticosteroneacetate (DOCA)-induced hypertensive pigs. Nine healthy Landrace pigs and 7 pigs with DOCA-induced hypertension and LV concentric hypertrophy were acutely instrumented to measure LV end-diastolic pressure-volume relationships (EDPVRs) at baseline and during intravenous infusion of BAY 41-8543 (1 and 3 μg·kg-1·min-1 for 30 min, respectively). Separately, in seven healthy and six DOCA pigs, transmural LV biopsies were harvested from the beating heart to measure titin phosphorylation during BAY 41-8543 infusion. LV EDPVRs before and during BAY 41-8543 infusion were superimposable in both healthy and DOCA-treated pigs, whereas mean aortic pressure decreased by 20-30 mmHg in both groups. Myocardial titin phosphorylation was unchanged in healthy pigs, but total and site-specific (Pro-Glu-Val-Lys and N2-Bus domains) titin phosphorylation was increased in DOCA-treated pigs. Bicoronary nitroglycerin infusion in healthy pigs ( n = 5) induced a rightward shift of the LV EDPVR, demonstrating the responsiveness of the pathway in this model. Acute systemic sGC stimulation with the sGC stimulator BAY 41-8543 did not recruit an LV preload reserve in both healthy and hypertrophied LV porcine myocardium, although it increased titin phosphorylation in the latter group. Thus, increased titin phosphorylation is not indicative of increased in vivo LV capacitance. NEW & NOTEWORTHY We demonstrate that acute pharmacological stimulation of soluble guanylate cyclase does not increase left ventricular compliance in normal and hypertrophied porcine hearts. Effects of long-term soluble guanylate cyclase stimulation with oral compounds in disease conditions associated with lowered myocardial cGMP levels, i.e., heart failure with preserved ejection fraction, remain to be investigated.
Collapse
Affiliation(s)
- Alessio Alogna
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Berlin , Germany.,Berlin Institute of Health , Berlin , Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung E.V.-Partner Site Berlin, Berlin, Germany
| | - Michael Schwarzl
- Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf , Hamburg , Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung E.V.-Partner Site Hamburg/Kiel/Lübeck, Hamburg , Germany
| | - Martin Manninger
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz , Graz , Austria
| | - Nazha Hamdani
- Institute of Physiology II, University of Muenster , Muenster , Germany
| | - Birgit Zirngast
- Department of Cardiothoracic Surgery, Medical University of Graz , Graz , Austria
| | - Benjamin Kloth
- Department of Cardiovascular Surgery, University Heart Center Hamburg-Eppendorf , Hamburg , Germany
| | - Paul Steendijk
- Department of Cardiology, Leiden University Medical Center , Leiden , The Netherlands
| | - Jochen Verderber
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz , Graz , Austria
| | - David Zweiker
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz , Graz , Austria
| | - Dirk Westermann
- Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf , Hamburg , Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung E.V.-Partner Site Hamburg/Kiel/Lübeck, Hamburg , Germany
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf , Hamburg , Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung E.V.-Partner Site Hamburg/Kiel/Lübeck, Hamburg , Germany
| | - Heinrich Maechler
- Department of Cardiothoracic Surgery, Medical University of Graz , Graz , Austria
| | - Carsten Tschöpe
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Berlin , Germany.,Berlin Institute of Health , Berlin , Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung E.V.-Partner Site Berlin, Berlin, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster , Muenster , Germany
| | - Gunther Marsche
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz , Graz , Austria
| | - Burkert M Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Berlin , Germany.,Berlin Institute of Health , Berlin , Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung E.V.-Partner Site Berlin, Berlin, Germany
| | - Heiner Post
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Berlin , Germany.,Berlin Institute of Health , Berlin , Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung E.V.-Partner Site Berlin, Berlin, Germany
| |
Collapse
|
31
|
Tampakakis E, Shah SJ, Borlaug BA, Leary PJ, Patel HH, Miller WL, Kelemen BW, Houston BA, Kolb TM, Damico R, Mathai SC, Kasper EK, Hassoun PM, Kass DA, Tedford RJ. Pulmonary Effective Arterial Elastance as a Measure of Right Ventricular Afterload and Its Prognostic Value in Pulmonary Hypertension Due to Left Heart Disease. Circ Heart Fail 2018; 11:e004436. [PMID: 29643065 PMCID: PMC5901761 DOI: 10.1161/circheartfailure.117.004436] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 03/09/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Patients with combined post- and precapillary pulmonary hypertension due to left heart disease have a worse prognosis compared with isolated postcapillary. However, it remains unclear whether increased mortality in combined post- and precapillary pulmonary hypertension is simply a result of higher total right ventricular load. Pulmonary effective arterial elastance (Ea) is a measure of total right ventricular afterload, reflecting both resistive and pulsatile components. We aimed to test whether pulmonary Ea discriminates survivors from nonsurvivors in patients with pulmonary hypertension due to left heart disease and if it does so better than other hemodynamic parameters associated with combined post- and precapillary pulmonary hypertension. METHODS AND RESULTS We combined 3 large heart failure patient cohorts (n=1036) from academic hospitals, including patients with pulmonary hypertension due to heart failure with preserved ejection fraction (n=232), reduced ejection fraction (n=335), and a mixed population (n=469). In unadjusted and 2 adjusted models, pulmonary Ea more robustly predicted mortality than pulmonary vascular resistance and the transpulmonary gradient. Along with pulmonary arterial compliance, pulmonary Ea remained predictive of survival in patients with normal pulmonary vascular resistance. The diastolic pulmonary gradient did not predict mortality. In addition, in a subset of patients with echocardiographic data, Ea and pulmonary arterial compliance were better discriminators of right ventricular dysfunction than the other parameters. CONCLUSIONS Pulmonary Ea and pulmonary arterial compliance more consistently predicted mortality than pulmonary vascular resistance or transpulmonary gradient across a spectrum of left heart disease with pulmonary hypertension, including patients with heart failure with preserved ejection fraction, heart failure with reduced ejection fraction, and pulmonary hypertension with a normal pulmonary vascular resistance.
Collapse
Affiliation(s)
- Emmanouil Tampakakis
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Sanjiv J Shah
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Barry A Borlaug
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Peter J Leary
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Harnish H Patel
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Wayne L Miller
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Benjamin W Kelemen
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Brian A Houston
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Todd M Kolb
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Rachel Damico
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Stephen C Mathai
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Edward K Kasper
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Paul M Hassoun
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - David A Kass
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.)
| | - Ryan J Tedford
- Division of Cardiology (E.T., B.W.K., E.K.K., D.A.K., R.J.T.) and Division of Pulmonary and Critical Care Medicine (T.M.K., R.D., S.C.M., P.M.H.), Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD. Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S., H.H.P.). Division of Cardiology, Mayo Clinic, Rochester, MN (B.A.B., W.L.M.). Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle (P.J.L.). Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (B.A.H., R.J.T.).
| |
Collapse
|
32
|
Impaired Alveolar Capillary Membrane Diffusion: A Recently Recognized Contributor to Exertional Dyspnea in Heart Failure With Preserved Ejection Fraction. JACC-HEART FAILURE 2018; 4:499-501. [PMID: 27256753 DOI: 10.1016/j.jchf.2016.03.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 11/21/2022]
|
33
|
Abstract
Novel cardioprotective agents are needed in both heart failure (HF) and myocardial infarction. Increasing evidence from cellular studies and animal models indicate protective effects of phosphodiesterase-5 (PDE5) inhibitors, drugs usually reserved as treatments of erectile dysfunction and pulmonary arterial hypertension. PDE5 inhibitors have been shown to improve contractile function in systolic HF, regress left ventricular hypertrophy, reduce myocardial infarct size and suppress ischaemia-induced ventricular arrhythmias. Underpinning these actions are complex but increasingly understood cellular mechanisms involving the cyclic GMP activation of protein kinase-G in both cardiac myocytes and the vasculature. In clinical trials, PDE5 inhibitors improve symptoms and ventricular function in systolic HF, and accumulating epidemiological data indicate a reduction in cardiovascular events and mortality in PDE5 inhibitor users at high cardiovascular risk. Here, we focus on the translation of underpinning basic science to clinical studies and report that PDE5 inhibitors act through a number of cardioprotective mechanisms, including a direct myocardial action independent of the vasculature. We conclude that future clinical trials should be designed with these mechanisms in mind to identify patient subsets that derive greatest treatment benefit from these novel cardioprotective agents.
Collapse
Affiliation(s)
- David Charles Hutchings
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Simon George Anderson
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jessica L Caldwell
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
34
|
|
35
|
Ramu B, Houston BA, Tedford RJ. Pulmonary Vascular Disease: Hemodynamic Assessment and Treatment Selection—Focus on Group II Pulmonary Hypertension. Curr Heart Fail Rep 2018; 15:81-93. [DOI: 10.1007/s11897-018-0377-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
36
|
Abstract
Nitric oxide (NO) signalling has pleiotropic roles in biology and a crucial function in cardiovascular homeostasis. Tremendous knowledge has been accumulated on the mechanisms of the nitric oxide synthase (NOS)-NO pathway, but how this highly reactive, free radical gas signals to specific targets for precise regulation of cardiovascular function remains the focus of much intense research. In this Review, we summarize the updated paradigms on NOS regulation, NO interaction with reactive oxidant species in specific subcellular compartments, and downstream effects of NO in target cardiovascular tissues, while emphasizing the latest developments of molecular tools and biomarkers to modulate and monitor NO production and bioavailability.
Collapse
Affiliation(s)
- Charlotte Farah
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| |
Collapse
|
37
|
Kaye DM, Silvestry FE, Gustafsson F, Cleland JG, van Veldhuisen DJ, Ponikowski P, Komtebedde J, Nanayakkara S, Burkhoff D, Shah SJ. Impact of atrial fibrillation on rest and exercise haemodynamics in heart failure with mid-range and preserved ejection fraction. Eur J Heart Fail 2017; 19:1690-1697. [DOI: 10.1002/ejhf.930] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/01/2017] [Accepted: 06/04/2017] [Indexed: 11/09/2022] Open
Affiliation(s)
- David M. Kaye
- Alfred Hospital and Baker IDI Heart & Diabetes Institute; Melbourne Australia
| | | | | | - John G. Cleland
- National Heart & Lung Institute; Royal Brompton and Harefield Hospitals, Imperial College; London UK
| | | | | | | | - Shane Nanayakkara
- Alfred Hospital and Baker IDI Heart & Diabetes Institute; Melbourne Australia
| | | | | |
Collapse
|
38
|
Reddy YNV, Lewis GD, Shah SJ, LeWinter M, Semigran M, Davila-Roman VG, Anstrom K, Hernandez A, Braunwald E, Redfield MM, Borlaug BA. INDIE-HFpEF (Inorganic Nitrite Delivery to Improve Exercise Capacity in Heart Failure With Preserved Ejection Fraction): Rationale and Design. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.117.003862. [PMID: 28476756 DOI: 10.1161/circheartfailure.117.003862] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/29/2017] [Indexed: 02/06/2023]
Abstract
Approximately half of patients with heart failure have preserved ejection fraction. There is no proven treatment that improves outcome. The pathophysiology of heart failure with preserved ejection fraction is complex and includes left ventricular systolic and diastolic dysfunction, pulmonary vascular disease, endothelial dysfunction, and peripheral abnormalities. Multiple lines of evidence point to impaired nitric oxide (NO)-cGMP bioavailability as playing a central role in each of these abnormalities. In contrast to traditional organic nitrate therapies, an alternative strategy to restore NO-cGMP signaling is via inorganic nitrite. Inorganic nitrite, previously considered to be an inert byproduct of NO metabolism, functions as an important in vivo reservoir for NO generation, particularly under hypoxic and acidosis conditions. As such, inorganic nitrite becomes most active at times of greater need for NO signaling, as during exercise when left ventricular filling pressures and pulmonary artery pressures increase. Herein, we present the rationale and design for the INDIE-HFpEF trial (Inorganic Nitrite Delivery to Improve Exercise Capacity in Heart Failure with Preserved Ejection Fraction), which is a multicenter, randomized, double-blind, placebo-controlled cross-over study assessing the effect of inhaled inorganic nitrite on peak exercise capacity, conducted in the National Heart, Lung, and Blood Institute-sponsored Heart Failure Clinical Research Network. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT02742129.
Collapse
Affiliation(s)
- Yogesh N V Reddy
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Gregory D Lewis
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Sanjiv J Shah
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Martin LeWinter
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Marc Semigran
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Victor G Davila-Roman
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Kevin Anstrom
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Adrian Hernandez
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Eugene Braunwald
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Margaret M Redfield
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.)
| | - Barry A Borlaug
- From the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.N.V.R., M.M.R., B.A.B.); Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston (G.D.L., M.S.); Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Cardiology Unit, University of Vermont College of Medicine, Burlington (M.L.W.); Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (V.G.D.-R.); Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (K.A., A.H.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.B.).
| |
Collapse
|
39
|
Nanayakkara S, Kaye DM. Targets for Heart Failure With Preserved Ejection Fraction. Clin Pharmacol Ther 2017; 102:228-237. [PMID: 28466986 DOI: 10.1002/cpt.723] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/21/2017] [Indexed: 12/19/2022]
Abstract
Heart failure (HF) with preserved ejection fraction (HFPEF) is responsible for half of all HF cases and will be the most common form of HF within the next 5 years. Previous studies of pharmacological agents in HFPEF have proved neutral or negative, in part due to phenotypic heterogeneity and complex underlying mechanisms. This review summarizes the key molecular and cellular pathways characterized in HFPEF as well as current and future therapies that target these mechanisms.
Collapse
Affiliation(s)
- S Nanayakkara
- Alfred Hospital and Baker Heart & Diabetes Institute, Melbourne, Australia
| | - D M Kaye
- Alfred Hospital and Baker Heart & Diabetes Institute, Melbourne, Australia
| |
Collapse
|
40
|
Hwang IC, Kim YJ, Park JB, Yoon YE, Lee SP, Kim HK, Cho GY, Sohn DW. Pulmonary hemodynamics and effects of phosphodiesterase type 5 inhibition in heart failure: a meta-analysis of randomized trials. BMC Cardiovasc Disord 2017; 17:150. [PMID: 28606099 PMCID: PMC5468951 DOI: 10.1186/s12872-017-0576-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/22/2017] [Indexed: 01/08/2023] Open
Abstract
Background Previous studies suggested that phosphodiesterase 5 inhibitors (PDE5i) have a beneficial effect in patients with heart failure (HF), although the results were inconsistent. We performed a meta-analysis to evaluate the effect of PDE5i in HF patients, and investigated the relationship between PDE5i effects and pulmonary hemodynamics. Method We searched PubMed, EMBASE and the Cochrane Library for randomized controlled trials (RCTs) that compared PDE5i with placebo in HF with reduced ejection fraction (HFrEF) or HF with preserved EF (HFpEF). PDE5i effects were interpolated according to baseline pulmonary arterial pressure (PAP) or according to changes in PAP after PDE5i treatment. Results Thirteen RCTs enrolling 898 HF patients, and two sub-analysis studies with different study outcomes, were included in the meta-analysis. Among patients with HFrEF, PDE5i improved peak VO2 (mean difference [MD], 3.76 mL/min/kg; 95% confidence interval [CI], 3.27 to 4.25; P < 0.00001), VE/VCO2 slope (MD, −6.04; 95% CI, −7.45 to −4.64; P < 0.00001), LVEF (MD, 4.30%; 95% CI, 2.18 to 6.42; P < 0.0001), and pulmonary vascular resistance (MD, −80.74 dyn·sec/cm5; 95% CI, −110.69 to −50.79; P < 0.00001). The effects of PDE5i in patients with HFpEF were heterogeneous. Meta-regression analyses indicated that the beneficial effect of PDE5i was related to the baseline PAP as well as the extent of PDE5i-mediated PAP decrease. Conclusion PDE5i improved pulmonary hemodynamics and exercise capacity in patients with HFrEF, but not in HFpEF. The relationship between the benefits by PDE5i with the baseline PAP and the changes in PAP indicates the therapeutic potential of PDE5i in HF according to pulmonary hemodynamics. Electronic supplementary material The online version of this article (doi:10.1186/s12872-017-0576-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- In-Chang Hwang
- Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Yong-Jin Kim
- Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea.
| | - Jun-Bean Park
- Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Yeonyee E Yoon
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seung-Pyo Lee
- Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Hyung-Kwan Kim
- Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Goo-Yeong Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Dae-Won Sohn
- Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| |
Collapse
|
41
|
De Keulenaer GW, Segers VFM, Zannad F, Brutsaert DL. The future of pleiotropic therapy in heart failure. Lessons from the benefits of exercise training on endothelial function. Eur J Heart Fail 2017; 19:603-614. [PMID: 28105791 DOI: 10.1002/ejhf.735] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/15/2016] [Accepted: 11/24/2016] [Indexed: 12/14/2022] Open
Abstract
A novel generation of drugs is introduced in the treatment of heart failure (HF). These drugs, including phosphodiesterase-5 inhibitors, guanylate cyclase stimulators and activators, share the feature that their action is either endothelial-mediated or substitutes for endothelial pathways, in particular the nitric oxide-cyclic guanosine monophosphate pathway, thereby influencing homeostatic balances in virtually each organ system in a pleiotropic fashion. Unfortunately, recent clinical trials with some of these drugs have shown disappointing results, at least in the setting of HF with a preserved ejection fraction. This suggests that their clinical use may require approaches that diverge from traditional pharmacological approaches, the latter often titrated on the effects of drugs on haemodynamic parameters or single biomarkers. In this paper we preconize that HF drugs with an endothelial profile should be applied conform to principles of endothelial physiology and systems pharmacology. This type of drug therapy should be viewed as a systems physio-pharmacological intervention and its clinical use accustomed to systems pharmacological principles, comparable to the systemic endothelial-mediated benefits induced by exercise training in HF. We will review the actions of these drugs and define criteria to which trials with these drugs should comply in order to increase chances of success.
Collapse
Affiliation(s)
- Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium.,Department of Cardiology, Middelheim Hospital, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Faiez Zannad
- CHU Nancy, Pôle de Cardiologie, Institut Lorrain du Cœur et des Vaisseaux, Vandoeuvre-lès-Nancy, France
| | - Dirk L Brutsaert
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
42
|
Patel HC, Hayward C, Keegan J, Gatehouse PD, Rajani R, Khattar RS, Mohiaddin RH, Rosen SD, Lyon AR, di Mario C. Effects of renal denervation on vascular remodelling in patients with heart failure and preserved ejection fraction: A randomised control trial. JRSM Cardiovasc Dis 2017; 6:2048004017690988. [PMID: 28228942 PMCID: PMC5308435 DOI: 10.1177/2048004017690988] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To assess the effect of renal denervation (RDT) on micro- and macro-vascular function in patients with heart failure with preserved ejection fraction (HFpEF). DESIGN A prospective, randomised, open-controlled trial with blinded end-point analysis. SETTING A single-centre London teaching hospital. PARTICIPANTS Twenty-five patients with HFpEF who were recruited into the RDT-PEF trial. MAIN OUTCOME MEASURES Macro-vascular: 24-h ambulatory pulse pressure, aorta distensibilty (from cardiac magnetic resonance imaging (CMR), aorta pulse wave velocity (CMR), augmentation index (peripheral tonometry) and renal artery blood flow indices (renal MR). Micro-vascular: endothelial function (peripheral tonometry) and urine microalbuminuria. RESULTS At baseline, 15 patients were normotensive, 9 were hypertensive and 1 was hypotensive. RDT did not lower any of the blood pressure indices. Though there was evidence of abnormal vascular function at rest, RDT did not affect these at 3 or 12 months follow-up. CONCLUSIONS RDT did not improve markers of macro- and micro-vascular function.
Collapse
Affiliation(s)
- Hitesh C Patel
- Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial College, London, UK
| | - Carl Hayward
- Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial College, London, UK
| | - Jennifer Keegan
- Department of Cardiac MRI, Royal Brompton Hospital, London, UK
| | | | - Ronak Rajani
- Department of Cardiology, St Thomas' Hospital, London, UK
| | - Rajdeep S Khattar
- Department of Non-Invasive Cardiology, Royal Brompton Hospital, London, UK
| | - Raad H Mohiaddin
- Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial College, London, UK
| | - Stuart D Rosen
- Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial College, London, UK; Department of Cardiology, Ealing Hospital, Southall, UK
| | - Alexander R Lyon
- Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial College, London, UK
| | - Carlo di Mario
- Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial College, London, UK
| |
Collapse
|
43
|
Lin S, Wang J, Wang L, Wen J, Guo Y, Qiao W, Zhou J, Xu G, Zhi F. Phosphodiesterase-5 inhibition suppresses colonic inflammation-induced tumorigenesis via blocking the recruitment of MDSC. Am J Cancer Res 2017; 7:41-52. [PMID: 28123846 PMCID: PMC5250679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 11/01/2016] [Indexed: 06/06/2023] Open
Abstract
Phosphodiesterase 5 (PDE-5) is a major isoform of cGMP phosphodiesterase in diverse tissues and plays a critical role in regulating intracellular cGMP concentrations. However, the distribution and expression of PDE-5 in colitis-related colon cancer was still unclear, not even the function and mechanism. Western blotting and ELISA were performed to detect colonic PDE-5 expression in AOM/DSS-induced tumorigenesis model. Sildenafil, a specific PDE-5 inhibitor, was used to treat AOM/DSS-induced and AOM-induced colonic tumorigenesis model and DSS-induced colitis model. The leukocyte infiltration in colonic tissue was examined by flow cytometry and immunofluorescence staining. Further matrigel-based invasion assay was employed to determine the effects of Sildenafil on myeloid-derived suppressor cell (MDSC) in vitro. We first demonstrated the upregulation of colonic PDE-5 expression and the prevention role of PDE-5 inhibition in AOM/DSS-induced tumorigenesis model. More importantly, PDE-5 inhibitor Sildenafil inhibited colonic tumorigenesis dependent on inflammation and suppressed DSS-induced colitis. Molecular mechanism investigation indicated that Sildenafil regulated inflammation microenvironment via directly inhibiting MDSC infiltration in colonic tissue. The study provides solid evidence for the use of PDE-5 inhibitor in preventing and treating colonic inflammation-related tumorigenesis.
Collapse
Affiliation(s)
- Shiyong Lin
- Guangdong Provincial Key Laboratory of Gastroenterology; Inst. of Gastroenterology of Guangdong Province; Department of Gastroenterology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, Guangdong Province, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer MedicineGuangzhou 510060, Guangdong Province, China
| | - Jing Wang
- Guangdong Provincial Key Laboratory of Gastroenterology; Inst. of Gastroenterology of Guangdong Province; Department of Gastroenterology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, Guangdong Province, China
| | - Lihui Wang
- Guangdong Provincial Key Laboratory of Gastroenterology; Inst. of Gastroenterology of Guangdong Province; Department of Gastroenterology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, Guangdong Province, China
| | - Jing Wen
- Guangdong Provincial Key Laboratory of Gastroenterology; Inst. of Gastroenterology of Guangdong Province; Department of Gastroenterology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, Guangdong Province, China
| | - Yandong Guo
- Guangdong Provincial Key Laboratory of Gastroenterology; Inst. of Gastroenterology of Guangdong Province; Department of Gastroenterology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, Guangdong Province, China
| | - Weiguang Qiao
- Guangdong Provincial Key Laboratory of Gastroenterology; Inst. of Gastroenterology of Guangdong Province; Department of Gastroenterology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, Guangdong Province, China
| | - Jieqiong Zhou
- Guangdong Provincial Key Laboratory of Gastroenterology; Inst. of Gastroenterology of Guangdong Province; Department of Gastroenterology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, Guangdong Province, China
| | - Guoliang Xu
- Department of Endoscopy, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer MedicineGuangzhou 510060, Guangdong Province, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology; Inst. of Gastroenterology of Guangdong Province; Department of Gastroenterology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, Guangdong Province, China
| |
Collapse
|
44
|
Abstract
A key feature of chronic heart failure (HF) is the sustained activation of endogenous neurohormonal systems in response to impaired cardiac pumping and/or filling properties. The clinical use of neurohormonal blockers has revolutionised the care of HF patients over the past three decades. Drug therapy that is active against imbalance in both the autonomic and renin-angiotensin-aldosterone systems consistently reduces morbidity and mortality in chronic HF with reduced left ventricular ejection fraction and in sinus rhythm. This article provides an assessment of the major neurohormonal systems and their therapeutic blockade in patients with chronic HF.
Collapse
Affiliation(s)
- Thomas G von Lueder
- Department of Cardiology, Oslo University Hospital UllevÅl, Oslo, Norway.,Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia and Alfred Hospital, Melbourne, Australia
| | - Dipak Kotecha
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia and Alfred Hospital, Melbourne, Australia.,University of Birmingham Institute of Cardiovascular Sciences, Birmingham, UK
| | - Dan Atar
- Department of Cardiology, Oslo University Hospital UllevÅl, Oslo, Norway
| | - Ingrid Hopper
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia and Alfred Hospital, Melbourne, Australia
| |
Collapse
|
45
|
Abstract
Pulmonary hypertension associated with left heart disease is the most common form of pulmonary hypertension. Although its pathophysiology remains incompletely understood, it is now well recognized that the presence of pulmonary hypertension is associated with a worse prognosis. Right ventricular failure has independent and additive prognostic value over pulmonary hypertension for adverse outcomes in left heart disease. Recently, several new terminologies have been introduced to better define and characterize the nature and severity of pulmonary hypertension. Several new treatment options including the use of pulmonary arterial hypertension specific therapies are being considered, but there is lack of evidence. Here, we review the recent advances in this field and summarize the diagnostic and therapeutic modalities of use in the management of pulmonary hypertension associated with left heart disease.
Collapse
Affiliation(s)
- Bhavadharini Ramu
- Cardiovascular Division, Section of Advanced Heart Failure and Pulmonary Hypertension, Lillehei Heart Institute, University of Minnesota, 420 Delaware Street SE, MMC 508, Minneapolis, MN, 55455, USA
| | - Thenappan Thenappan
- Cardiovascular Division, Section of Advanced Heart Failure and Pulmonary Hypertension, Lillehei Heart Institute, University of Minnesota, 420 Delaware Street SE, MMC 508, Minneapolis, MN, 55455, USA.
| |
Collapse
|
46
|
Shah SJ, Kitzman DW, Borlaug BA, van Heerebeek L, Zile MR, Kass DA, Paulus WJ. Phenotype-Specific Treatment of Heart Failure With Preserved Ejection Fraction: A Multiorgan Roadmap. Circulation 2016; 134:73-90. [PMID: 27358439 DOI: 10.1161/circulationaha.116.021884] [Citation(s) in RCA: 720] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (EF; HFpEF) accounts for 50% of HF cases, and its prevalence relative to HF with reduced EF continues to rise. In contrast to HF with reduced EF, large trials testing neurohumoral inhibition in HFpEF failed to reach a positive outcome. This failure was recently attributed to distinct systemic and myocardial signaling in HFpEF and to diversity of HFpEF phenotypes. In this review, an HFpEF treatment strategy is proposed that addresses HFpEF-specific signaling and phenotypic diversity. In HFpEF, extracardiac comorbidities such as metabolic risk, arterial hypertension, and renal insufficiency drive left ventricular remodeling and dysfunction through systemic inflammation and coronary microvascular endothelial dysfunction. The latter affects left ventricular diastolic dysfunction through macrophage infiltration, resulting in interstitial fibrosis, and through altered paracrine signaling to cardiomyocytes, which become hypertrophied and stiff because of low nitric oxide and cyclic guanosine monophosphate. Systemic inflammation also affects other organs such as lungs, skeletal muscle, and kidneys, leading, respectively, to pulmonary hypertension, muscle weakness, and sodium retention. Individual steps of these signaling cascades can be targeted by specific interventions: metabolic risk by caloric restriction, systemic inflammation by statins, pulmonary hypertension by phosphodiesterase 5 inhibitors, muscle weakness by exercise training, sodium retention by diuretics and monitoring devices, myocardial nitric oxide bioavailability by inorganic nitrate-nitrite, myocardial cyclic guanosine monophosphate content by neprilysin or phosphodiesterase 9 inhibition, and myocardial fibrosis by spironolactone. Because of phenotypic diversity in HFpEF, personalized therapeutic strategies are proposed, which are configured in a matrix with HFpEF presentations in the abscissa and HFpEF predispositions in the ordinate.
Collapse
Affiliation(s)
- Sanjiv J Shah
- From Division of Cardiology, Department of Medicine, and the Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC (D.W.K.); Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, (B.A.B.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (L.v.H., W.J.P.); Department of Cardiology, Onze Lieve Vrouw Gasthuis, Amsterdam, The Netherlands (L.v.H.); Department of Medicine, Medical University of South Carolina (MUSC) and the RHJ Department of Veterans Affairs Medical Center, Charleston (M.R.Z.); and Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (D.A.K.)
| | - Dalane W Kitzman
- From Division of Cardiology, Department of Medicine, and the Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC (D.W.K.); Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, (B.A.B.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (L.v.H., W.J.P.); Department of Cardiology, Onze Lieve Vrouw Gasthuis, Amsterdam, The Netherlands (L.v.H.); Department of Medicine, Medical University of South Carolina (MUSC) and the RHJ Department of Veterans Affairs Medical Center, Charleston (M.R.Z.); and Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (D.A.K.)
| | - Barry A Borlaug
- From Division of Cardiology, Department of Medicine, and the Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC (D.W.K.); Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, (B.A.B.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (L.v.H., W.J.P.); Department of Cardiology, Onze Lieve Vrouw Gasthuis, Amsterdam, The Netherlands (L.v.H.); Department of Medicine, Medical University of South Carolina (MUSC) and the RHJ Department of Veterans Affairs Medical Center, Charleston (M.R.Z.); and Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (D.A.K.)
| | - Loek van Heerebeek
- From Division of Cardiology, Department of Medicine, and the Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC (D.W.K.); Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, (B.A.B.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (L.v.H., W.J.P.); Department of Cardiology, Onze Lieve Vrouw Gasthuis, Amsterdam, The Netherlands (L.v.H.); Department of Medicine, Medical University of South Carolina (MUSC) and the RHJ Department of Veterans Affairs Medical Center, Charleston (M.R.Z.); and Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (D.A.K.)
| | - Michael R Zile
- From Division of Cardiology, Department of Medicine, and the Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC (D.W.K.); Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, (B.A.B.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (L.v.H., W.J.P.); Department of Cardiology, Onze Lieve Vrouw Gasthuis, Amsterdam, The Netherlands (L.v.H.); Department of Medicine, Medical University of South Carolina (MUSC) and the RHJ Department of Veterans Affairs Medical Center, Charleston (M.R.Z.); and Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (D.A.K.)
| | - David A Kass
- From Division of Cardiology, Department of Medicine, and the Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC (D.W.K.); Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, (B.A.B.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (L.v.H., W.J.P.); Department of Cardiology, Onze Lieve Vrouw Gasthuis, Amsterdam, The Netherlands (L.v.H.); Department of Medicine, Medical University of South Carolina (MUSC) and the RHJ Department of Veterans Affairs Medical Center, Charleston (M.R.Z.); and Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (D.A.K.)
| | - Walter J Paulus
- From Division of Cardiology, Department of Medicine, and the Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.); Sections on Cardiovascular Medicine and Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC (D.W.K.); Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, (B.A.B.); Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (L.v.H., W.J.P.); Department of Cardiology, Onze Lieve Vrouw Gasthuis, Amsterdam, The Netherlands (L.v.H.); Department of Medicine, Medical University of South Carolina (MUSC) and the RHJ Department of Veterans Affairs Medical Center, Charleston (M.R.Z.); and Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (D.A.K.).
| |
Collapse
|
47
|
Mehta S, Vachiéry JL. Pulmonary hypertension: the importance of correctly diagnosing the cause. Eur Respir Rev 2016; 25:372-380. [DOI: 10.1183/16000617.0104-2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/18/2016] [Indexed: 01/22/2023] Open
Abstract
Pulmonary hypertension (PH) is a complex condition that can occur as a result of a wide range of disorders, including left heart disease, lung disease and chronic pulmonary thromboembolism. Contemporary PH patients are older and frequently have a multitude of comorbidities that may contribute to or simply coincide with their PH. Identifying the cause of PH in these complicated patients can be challenging but is essential, given that the aetiology of the disease has a significant impact on the management options available. In this article, we present two cases that highlight the difficulties involved in obtaining a precise diagnosis of the cause of PH within the setting of multiple comorbidities. The importance of performing a comprehensive, multidimensional diagnostic work-up is demonstrated, in addition to the need to specifically consider cardiopulmonary haemodynamic data in the context of the wider clinical picture. The article also illustrates why achieving an accurate diagnosis is necessary for optimal patient management. This may involve treatment of comorbidities as a priority, which can ameliorate the severity of PH, obviating the need to consider PH-targeted medical treatment.
Collapse
|
48
|
Reddy YNV, Borlaug BA. Sildenafil, unbridled optimism, and heart failure with preserved ejection fraction. Eur J Heart Fail 2016; 19:126-128. [PMID: 27873389 DOI: 10.1002/ejhf.690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/12/2016] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yogesh N V Reddy
- Division of Cardiovascular Diseases, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN, 55905, USA
| | - Barry A Borlaug
- Division of Cardiovascular Diseases, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
49
|
Liu LC, Hummel YM, van der Meer P, Berger RM, Damman K, van Veldhuisen DJ, Voors AA, Hoendermis ES. Effects of sildenafil on cardiac structure and function, cardiopulmonary exercise testing and health‐related quality of life measures in heart failure patients with preserved ejection fraction and pulmonary hypertension. Eur J Heart Fail 2016; 19:116-125. [DOI: 10.1002/ejhf.662] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/22/2016] [Accepted: 08/22/2016] [Indexed: 12/28/2022] Open
Affiliation(s)
- Licette C.Y. Liu
- Department of Cardiology, University of Groningen University Medical Centre Groningen Hanzeplein 1 9713 GZ Groningen the Netherlands
| | - Yoran M. Hummel
- Department of Cardiology, University of Groningen University Medical Centre Groningen Hanzeplein 1 9713 GZ Groningen the Netherlands
| | - Peter van der Meer
- Department of Cardiology, University of Groningen University Medical Centre Groningen Hanzeplein 1 9713 GZ Groningen the Netherlands
| | - Rolf M.F. Berger
- Department of Pediatric and Congenital Cardiology, University of Groningen University Medical Centre Groningen Groningen the Netherlands
| | - Kevin Damman
- Department of Cardiology, University of Groningen University Medical Centre Groningen Hanzeplein 1 9713 GZ Groningen the Netherlands
| | - Dirk J. van Veldhuisen
- Department of Cardiology, University of Groningen University Medical Centre Groningen Hanzeplein 1 9713 GZ Groningen the Netherlands
| | - Adriaan A. Voors
- Department of Cardiology, University of Groningen University Medical Centre Groningen Hanzeplein 1 9713 GZ Groningen the Netherlands
| | - Elke S. Hoendermis
- Department of Cardiology, University of Groningen University Medical Centre Groningen Hanzeplein 1 9713 GZ Groningen the Netherlands
| |
Collapse
|
50
|
Pilot Study of Endothelin Receptor Blockade in Heart Failure with Diastolic Dysfunction and Pulmonary Hypertension (BADDHY-Trial). Heart Lung Circ 2016; 26:433-441. [PMID: 27816421 DOI: 10.1016/j.hlc.2016.09.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/09/2016] [Accepted: 09/02/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND In this multi-centre, randomised, placebo-controlled pilot trial, we investigated the clinical and haemodynamic effects of the endothelin-receptor blocker Bosentan in patients with heart failure, preserved ejection fraction and pulmonary hypertension (PH-HFpEF). MATERIALS AND METHODS Eligible patients received either 12 weeks of Bosentan therapy, or a placebo drug. Patients were thereafter followed for a further period of 12 weeks without the study medication. At three points during the study (study Commencement, Week 12 and Week 24), a six-minute walk test (6MWT), echocardiographic and laboratory assessments were performed, as well as a quality of life survey. Right heart catheterisation (RHC) was undertaken at commencement only. The study was aborted early, after an interim analysis favoured the placebo. RESULTS Six-minute walk distance (6MWD) did not change in the Bosentan group (309.7±96.3m (Commencement), 317.0±126.1m (Week 12), 307.0±84.4m (Week 24); p=0.86), but almost reached statistical significance in the placebo group from 328.8±79.6m, to 361.6±98.2m and 384.0±74.9m (Week 24); p=0.075. In the placebo group, estimated systolic pulmonary artery pressure (measured via echocardiography) significantly decreased (from 62.3±16.7mmHg [Commencement], 45.3±13.9mmHg [Week 12], to 44.6±14.5mmHg [Week 24]; p=0.014) as did right atrial pressure (13.1±5.3 [Commencement], 10.0±3.8 [Week 12], to 9.4±3.2 [Week 24]; p=0.046). CONCLUSION Despite this study's limited sample size and premature cessation, it nevertheless suggests that endothelin receptor blockade in patients with PH-HFpEF may have no beneficial effects and could even be detrimental in comparison to a placebo.
Collapse
|