1
|
Rendell M. Lessons learned from early-stage clinical trials for diabetic nephropathy. Expert Opin Investig Drugs 2024; 33:287-301. [PMID: 38465470 DOI: 10.1080/13543784.2024.2326025] [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] [Received: 08/13/2023] [Accepted: 02/28/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION The evolution of treatment for diabetic nephropathy illustrates how basic biochemistry and physiology have led to new agents such as SGLT2 inhibitors and mineralocorticoid blockers. Conversely, clinical studies performed with these agents have suggested new concepts for investigational drug development. We reviewed currently available treatments for diabetic nephropathy and then analyzed early clinical trials of new agents to assess the potential for future treatment modalities. AREAS COVERED We searched ClinicalTrials.gov for new agents under study for diabetic nephropathy in the past decade. Once we have identified investigation trials of new agents, we then used search engines and Pubmed.gov to find publications providing insight on these drugs. Current treatments have shown benefit in both cardiac and renal disease. In our review, we found 51 trials and 43 pharmaceuticals in a number of drug classes: mineralocorticoid blockers, anti-inflammatory, anti-fibrosis, nitric oxide stimulatory, and podocyte protection, and endothelin inhibitors. EXPERT OPINION It is difficult to predict which early phase treatments will advance to confirmatory clinical trials. Current agents are thought to improve hemodynamic function. However, the coincident benefit of both myocardial function and the glomerulus argues for primary effects at the subcellular level, and we follow the evolution of agents which modify fundamental cellular processes.
Collapse
Affiliation(s)
- Marc Rendell
- The Association of Diabetes Investigators, Newport Coast, CA, USA
- The Rose Salter Medical Research Foundation, Newport Coast, CA, USA
| |
Collapse
|
2
|
Vang S, Cochran P, Sebastian Domingo J, Krick S, Barnes JW. The Glycobiology of Pulmonary Arterial Hypertension. Metabolites 2022; 12:metabo12040316. [PMID: 35448503 PMCID: PMC9026683 DOI: 10.3390/metabo12040316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disease of complex etiology. Cases of PAH that do not receive therapy after diagnosis have a low survival rate. Multiple reports have shown that idiopathic PAH, or IPAH, is associated with metabolic dysregulation including altered bioavailability of nitric oxide (NO) and dysregulated glucose metabolism. Multiple processes such as increased proliferation of pulmonary vascular cells, angiogenesis, apoptotic resistance, and vasoconstriction may be regulated by the metabolic changes demonstrated in PAH. Recent reports have underscored similarities between metabolic abnormalities in cancer and IPAH. In particular, increased glucose uptake and altered glucose utilization have been documented and have been linked to the aforementioned processes. We were the first to report a link between altered glucose metabolism and changes in glycosylation. Subsequent reports have highlighted similar findings, including a potential role for altered metabolism and aberrant glycosylation in IPAH pathogenesis. This review will detail research findings that demonstrate metabolic dysregulation in PAH with an emphasis on glycobiology. Furthermore, this report will illustrate the similarities in the pathobiology of PAH and cancer and highlight the novel findings that researchers have explored in the field.
Collapse
|
3
|
Durgin BG, Wood KC, Hahn SA, McMahon B, Baust JJ, Straub AC. Smooth muscle cell CYB5R3 preserves cardiac and vascular function under chronic hypoxic stress. J Mol Cell Cardiol 2022; 162:72-80. [PMID: 34536439 PMCID: PMC8766905 DOI: 10.1016/j.yjmcc.2021.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/01/2021] [Accepted: 09/08/2021] [Indexed: 01/03/2023]
Abstract
Chronic hypoxia is a major driver of cardiovascular complications, including heart failure. The nitric oxide (NO) - soluble guanylyl cyclase (sGC) - cyclic guanosine monophosphate (cGMP) pathway is integral to vascular tone maintenance. Specifically, NO binds its receptor sGC within vascular smooth muscle cells (SMC) in its reduced heme (Fe2+) form to increase intracellular cGMP production, activate protein kinase G (PKG) signaling, and induce vessel relaxation. Under chronic hypoxia, oxidative stress drives oxidation of sGC heme (Fe2+→Fe3+), rendering it NO-insensitive. We previously showed that cytochrome b5 reductase 3 (CYB5R3) in SMC is a sGC reductase important for maintaining NO-dependent vasodilation and conferring resilience to systemic hypertension and sickle cell disease-associated pulmonary hypertension. To test whether CYB5R3 may be protective in the context of chronic hypoxia, we subjected SMC-specific CYB5R3 knockout mice (SMC CYB5R3 KO) to 3 weeks hypoxia and assessed vascular and cardiac function using echocardiography, pressure volume loops and wire myography. Hypoxic stress caused 1) biventricular hypertrophy in both WT and SMC CYB5R3 KO, but to a larger degree in KO mice, 2) blunted vasodilation to NO-dependent activation of sGC in coronary and pulmonary arteries of KO mice, and 3) decreased, albeit still normal, cardiac function in KO mice. Overall, these data indicate that SMC CYB5R3 deficiency potentiates bilateral ventricular hypertrophy and blunts NO-dependent vasodilation under chronic hypoxia conditions. This implicates that SMC CYB5R3 KO mice post 3-week hypoxia have early stages of cardiac remodeling and functional changes that could foretell significantly impaired cardiac function with longer exposure to hypoxia.
Collapse
Affiliation(s)
- Brittany G Durgin
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Katherine C Wood
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Scott A Hahn
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Brenda McMahon
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Jeffrey J Baust
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Microvascular Research, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
4
|
McCarty MF, DiNicolantonio JJ, Lerner A. Review - Nutraceuticals Can Target Asthmatic Bronchoconstriction: NADPH Oxidase-Dependent Oxidative Stress, RhoA and Calcium Dynamics. J Asthma Allergy 2021; 14:685-701. [PMID: 34163181 PMCID: PMC8214517 DOI: 10.2147/jaa.s307549] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/21/2021] [Indexed: 12/17/2022] Open
Abstract
Activation of various isoforms of NADPH oxidase contributes to the pathogenesis of asthma at multiple levels: promoting hypercontractility, hypertrophy, and proliferation of airway smooth muscle; enabling lung influx of eosinophils via VCAM-1; and mediating allergen-induced mast cell activation. Free bilirubin, which functions physiologically within cells as a feedback inhibitor of NADPH oxidase complexes, has been shown to have a favorable impact on each of these phases of asthma pathogenesis. The spirulina chromophore phycocyanobilin (PhyCB), a homolog of bilirubin's precursor biliverdin, can mimic the inhibitory impact of biliverdin/bilirubin on NADPH oxidase activity, and spirulina's versatile and profound anti-inflammatory activity in rodent studies suggests that PhyCB may have potential as a clinical inhibitor of NADPH oxidase. Hence, spirulina or PhyCB-enriched spirulina extracts merit clinical evaluation in asthma. Promoting biosynthesis of glutathione and increasing the expression and activity of various antioxidant enzymes - as by supplementing with N-acetylcysteine, Phase 2 inducers (eg, lipoic acid), selenium, and zinc - may also blunt the contribution of oxidative stress to asthma pathogenesis. Nitric oxide (NO) and hydrogen sulfide (H2S) work in various ways to oppose pathogenic mechanisms in asthma; supplemental citrulline and high-dose folate may aid NO synthesis, high-dose biotin may mimic and possibly potentiate NO's activating impact on soluble guanylate cyclase, and NAC and taurine may boost H2S synthesis. The amino acid glycine has a hyperpolarizing effect on airway smooth muscle that is bronchodilatory. Insuring optimal intracellular levels of magnesium may modestly blunt the stimulatory impact of intracellular free calcium on bronchoconstriction. Nutraceutical regimens or functional foods incorporating at least several of these agents may have utility as nutraceutical adjuvants to standard clinical management of asthma.
Collapse
Affiliation(s)
| | - James J DiNicolantonio
- Department of Preventive Cardiology, Saint Luke's Mid America Heart Institute, Kansas, MO, USA
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, 5262000, Israel
| |
Collapse
|
5
|
Chaumais MC, Djessas MRA, Thuillet R, Cumont A, Tu L, Hebert G, Gaignard P, Huertas A, Savale L, Humbert M, Guignabert C. Additive protective effects of sacubitril/valsartan and bosentan on vascular remodelling in experimental pulmonary hypertension. Cardiovasc Res 2021; 117:1391-1401. [PMID: 32653925 DOI: 10.1093/cvr/cvaa200] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/03/2020] [Accepted: 07/03/2020] [Indexed: 01/26/2023] Open
Abstract
AIMS Although right ventricular (RV) function is an important determinant of morbidity and mortality in patients with pulmonary arterial hypertension (PAH), there is no treatment targeting directly the RV. We evaluate the efficacy of sacubitril/valsartan (LCZ 696) as add-on therapy to bosentan in rats with severe pulmonary hypertension (PH). METHODS AND RESULTS Combination therapy of LCZ 696 and bosentan has additive vascular protective effects against the pulmonary vascular remodelling and PH in two preclinical models of severe PH. Compared with monotherapy, co-treatment of LCZ 696 (30 or 68 mg/kg/day for 2 weeks, per os) and bosentan (100 mg/kg/day for 2 weeks, per os) started 7 days after monocrotaline (MCT) injection substantially reduces pulmonary pressures, vascular remodelling, and RV hypertrophy and fibrosis in rats. Consistent with these observations, co-treatment of rats with established PH induced by sugen/hypoxia (SuHx) with LCZ 696 (30 mg/kg/day for 3 weeks, per os) and bosentan (100 mg/kg/day for 3 weeks, per os) started 5 weeks after Sugen injection partially attenuate total pulmonary vascular resistance and cardiovascular structures. We also obtained evidence showing that LCZ 696 has anti-proliferative effect on cultured human pulmonary artery smooth muscle cells derived from patients with idiopathic PAH, an effect that is more pronounced in presence of bosentan. Finally, we found that the plasma levels of atrial natriuretic peptide (ANP) and cyclic guanosine monophosphate (cGMP) are higher in rats co-treated with LCZ 696 (30 mg/kg/day) and bosentan (100 mg/kg/day) than in MCT and SuHx rats treated with vehicle. CONCLUSION Dual therapy with LCZ 696 plus bosentan proved significantly superior beneficial effect to LCZ 696 or bosentan alone on vascular remodelling and severity of experimental PH.
Collapse
MESH Headings
- Aminobutyrates/pharmacology
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Atrial Natriuretic Factor/blood
- Biphenyl Compounds/pharmacology
- Bosentan/pharmacology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclic GMP/blood
- Disease Models, Animal
- Disease Progression
- Drug Combinations
- Drug Therapy, Combination
- Endothelin Receptor Antagonists/pharmacology
- Familial Primary Pulmonary Hypertension/drug therapy
- Familial Primary Pulmonary Hypertension/metabolism
- Familial Primary Pulmonary Hypertension/physiopathology
- Humans
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neprilysin/antagonists & inhibitors
- Protease Inhibitors/pharmacology
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Rats, Wistar
- Valsartan/pharmacology
- Vascular Remodeling/drug effects
- Rats
Collapse
Affiliation(s)
- Marie-Camille Chaumais
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Pharmacie, 92290, Châtenay-Malabry, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pharmacie, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Mohamed Reda Amar Djessas
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Raphaël Thuillet
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Amélie Cumont
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Guillaume Hebert
- Hôpital Marie Lannelongue, Service pharmacie, 92350, Le Plessis-Robinson, France
| | - Pauline Gaignard
- Université Paris-Saclay, Faculté de Pharmacie, 92290, Châtenay-Malabry, France
- AP-HP, Laboratoire de biochimie, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Alice Huertas
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Laurent Savale
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| |
Collapse
|
6
|
Stam K, Clauss S, Taverne YJHJ, Merkus D. Chronic Thromboembolic Pulmonary Hypertension - What Have We Learned From Large Animal Models. Front Cardiovasc Med 2021; 8:574360. [PMID: 33937352 PMCID: PMC8085273 DOI: 10.3389/fcvm.2021.574360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Chronic thrombo-embolic pulmonary hypertension (CTEPH) develops in a subset of patients after acute pulmonary embolism. In CTEPH, pulmonary vascular resistance, which is initially elevated due to the obstructions in the larger pulmonary arteries, is further increased by pulmonary microvascular remodeling. The increased afterload of the right ventricle (RV) leads to RV dilation and hypertrophy. This RV remodeling predisposes to arrhythmogenesis and RV failure. Yet, mechanisms involved in pulmonary microvascular remodeling, processes underlying the RV structural and functional adaptability in CTEPH as well as determinants of the susceptibility to arrhythmias such as atrial fibrillation in the context of CTEPH remain incompletely understood. Several large animal models with critical clinical features of human CTEPH and subsequent RV remodeling have relatively recently been developed in swine, sheep, and dogs. In this review we will discuss the current knowledge on the processes underlying development and progression of CTEPH, and on how animal models can help enlarge understanding of these processes.
Collapse
Affiliation(s)
- Kelly Stam
- Department of Cardiology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sebastian Clauss
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany.,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Yannick J H J Taverne
- Department of Cardiothoracic Surgery, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Daphne Merkus
- Department of Cardiology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands.,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
7
|
Boutou AK, Pitsiou G. Treatment of pulmonary hypertension with riociguat: a review of current evidence and future perspectives. Expert Opin Pharmacother 2020; 21:1145-1155. [PMID: 32089012 DOI: 10.1080/14656566.2020.1727446] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Pulmonary arterial hypertension (PAH) is still a chronic disorder characterized by high morbidity and mortality. Chronic thromboembolic pulmonary hypertension (CTEPH) is another form of pulmonary hypertension (PH) for which pulmonary endarterectomy (PEA) is the treatment of choice. However, not all patients are operable, while PH is often recurrent or persistent. Thus, for both disorders novel treatment options are urgently needed. AREAS COVERED This review describes the mechanism of action of riociguat, a soluble guanylate cyclase (sGC) stimulator, with a dual mode of action. The most relevant publications are presented regarding the efficacy of riociguat in PAH and CTEPH, and also data regarding its potential effect on other forms of PH. EXPERT OPINION Riociguat is a first-in-class drug approved for the treatment of PAH as a monotherapy or added to endothelin-receptor antagonists as a sequential combination therapy, and for the treatment of inoperable CTEPH or persistent/recurrent PH after PEA. As it can stimulate sGC independently of NO, it could be beneficial in PAH patients with inadequate response to phosphodiesterase 5 inhibitors (PDE5i). Future studies are needed to evaluate whether drug switching is beneficial in PAH and which baseline markers could guide the optimal initial treatment selection.
Collapse
Affiliation(s)
- Afroditi K Boutou
- Department of Respiratory Medicine, G. Papanikolaou Hospital , Thessaloniki, Greece
| | - Georgia Pitsiou
- Department of Respiratory Failure, Aristotle University of Thessaloniki , Thessaloniki, Greece
| |
Collapse
|
8
|
Barnes JW, Patel RP. Things We "kNOw" and Do Not "kNOw" about Pulmonary Hypertension. Am J Respir Crit Care Med 2019; 198:151-152. [PMID: 29590535 DOI: 10.1164/rccm.201803-0424ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Jarrod W Barnes
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine University of Alabama at Birmingham Birmingham, Alabama.,2 Lerner Research Institute Cleveland Clinic Cleveland, Ohio and
| | - Rakesh P Patel
- 3 Department of Pathology University of Alabama at Birmingham Birmingham, Alabama
| |
Collapse
|
9
|
Toxvig AK, Wehland M, Grimm D, Infanger M, Krüger M. A focus on riociguat in the treatment of pulmonary arterial hypertension. Basic Clin Pharmacol Toxicol 2019; 125:202-214. [PMID: 31206240 DOI: 10.1111/bcpt.13272] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/03/2019] [Indexed: 01/24/2023]
Abstract
Current treatment of pulmonary arterial hypertension (PAH) targets three signalling pathways: the nitric oxide (NO) pathway, the endothelin pathway and the prostacyclin pathway. Riociguat is a soluble guanylate cyclase stimulator, acting via the NO pathway in a new way: unlike other common drugs targeting this pathway (eg tadalafil and sildenafil), riociguat acts independently of endogenous NO. This MiniReview focuses on PAH treatment with riociguat and on its advantages and disadvantages compared with other drugs targeting the NO pathway. In the PATENT-1 trial (NCT00810693), riociguat improved significantly the 6-minute walking distance in patients suffering from PAH, with a mean difference (MD) of 36 m compared with a placebo group. The results are comparable to those found for its competitors tadalafil (MD of 33 m) and sildenafil (MD of 50 m) in the PHIRST-1 trial (NCT00125918) and the SUPER-1 trial (NCT00644605). No obvious advantages were found regarding pharmacokinetic features and adverse events. In the RESPITE study (NCT02007629), patients with PAH with insufficient response to treatment with tadalafil or sildenafil were switched to riociguat. These results indicate that riociguat might be superior regarding efficacy to PDE-5 inhibitors in a patient group, where endogenous NO production might be insufficient. This finding was further examined in the REPLACE study (NCT02891850). Moreover, riociguat has shown promising anti-proliferative, anti-inflammatory and anti-fibrotic effects in animal models. Further investigations are needed to determine whether this applies also to human beings. Taken together, riociguat induces vasodilation of the pulmonary arteries and leads to an improvement in the ability to carry out physical activity.
Collapse
Affiliation(s)
| | - Markus Wehland
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Daniela Grimm
- Department of Biomedicine, Pharmacology, Aarhus University, Aarhus C, Denmark.,Department of Microgravity and Translational Regenerative Medicine, Faculty of Medicine and Mechanical Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Manfred Infanger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Marcus Krüger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
10
|
Budas GR, Boehm M, Kojonazarov B, Viswanathan G, Tian X, Veeroju S, Novoyatleva T, Grimminger F, Hinojosa-Kirschenbaum F, Ghofrani HA, Weissmann N, Seeger W, Liles JT, Schermuly RT. ASK1 Inhibition Halts Disease Progression in Preclinical Models of Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2019; 197:373-385. [PMID: 28910144 DOI: 10.1164/rccm.201703-0502oc] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Progression of pulmonary arterial hypertension (PAH) is associated with pathological remodeling of the pulmonary vasculature and the right ventricle (RV). Oxidative stress drives the remodeling process through activation of MAPKs (mitogen-activated protein kinases), which stimulate apoptosis, inflammation, and fibrosis. OBJECTIVES We investigated whether pharmacological inhibition of the redox-sensitive apical MAPK, ASK1 (apoptosis signal-regulating kinase 1), can halt the progression of pulmonary vascular and RV remodeling. METHODS A selective, orally available ASK1 inhibitor, GS-444217, was administered to two preclinical rat models of PAH (monocrotaline and Sugen/hypoxia), a murine model of RV pressure overload induced by pulmonary artery banding, and cellular models. MEASUREMENTS AND MAIN RESULTS Oral administration of GS-444217 dose dependently reduced pulmonary arterial pressure and reduced RV hypertrophy in PAH models. The therapeutic efficacy of GS-444217 was associated with reduced ASK1 phosphorylation, reduced muscularization of the pulmonary arteries, and reduced fibrotic gene expression in the RV. Importantly, efficacy was observed when GS-444217 was administered to animals with established disease and also directly reduced cardiac fibrosis and improved cardiac function in a model of isolated RV pressure overload. In cellular models, GS-444217 reduced phosphorylation of p38 and JNK (c-Jun N-terminal kinase) induced by adenoviral overexpression of ASK1 in rat cardiomyocytes and reduced activation/migration of primary mouse cardiac fibroblasts and human pulmonary adventitial fibroblasts derived from patients with PAH. CONCLUSIONS ASK1 inhibition reduced pathological remodeling of the pulmonary vasculature and the right ventricle and halted progression of pulmonary hypertension in rodent models. These preclinical data inform the first description of a causal role of ASK1 in PAH disease pathogenesis.
Collapse
Affiliation(s)
| | - Mario Boehm
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Baktybek Kojonazarov
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Gayathri Viswanathan
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Xia Tian
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Swathi Veeroju
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Tatyana Novoyatleva
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Friedrich Grimminger
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | | | - Hossein A Ghofrani
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Norbert Weissmann
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Werner Seeger
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and.,3 Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Ralph T Schermuly
- 2 Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany; and
| |
Collapse
|
11
|
Stam K, van Duin RW, Uitterdijk A, Krabbendam-Peters I, Sorop O, Danser AHJ, Duncker DJ, Merkus D. Pulmonary microvascular remodeling in chronic thrombo-embolic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2018; 315:L951-L964. [PMID: 30260284 DOI: 10.1152/ajplung.00043.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pulmonary vascular remodeling in pulmonary arterial hypertension involves perturbations in the nitric oxide (NO) and endothelin-1 (ET-1) pathways. However, the implications of pulmonary vascular remodeling and these pathways remain unclear in chronic thrombo-embolic pulmonary hypertension (CTEPH). The objective of the present study was to characterize changes in microvascular morphology and function, focussing on the ET-1 and NO pathways, in a CTEPH swine model. Swine were chronically instrumented and received up to five pulmonary embolizations by microsphere infusion, whereas endothelial dysfunction was induced by daily administration of the endothelial NO synthase inhibitor Nω-nitro-l-arginine methyl ester until 2 wk before the end of study. Swine were subjected to exercise, and the pulmonary vasculature was investigated by hemodynamic, histological, quantitative PCR, and myograph experiments. In swine with CTEPH, the increased right-ventricular afterload, decreased cardiac index, and mild ventilation-perfusion-mismatch were exacerbated during exercise. Pulmonary microvascular remodeling was evidenced by increased muscularization, which was accompanied by an increased maximal vasoconstriction. Although ET-1-induced vasoconstriction was increased in CTEPH pulmonary small arteries, the ET-1 sensitivity was decreased. Moreover, the contribution of the ETA receptor to ET-1 vasoconstriction was increased, whereas the contribution of the ETB receptor was decreased and the contribution of Rho-kinase was lost. A reduction in endogenous NO production was compensated in part by a decreased phosphodiesterase 5 (PDE5) activity resulting in an apparent increased NO sensitivity in CTEPH pulmonary small arteries. These findings suggest that pulmonary microvascular remodeling with a reduced activity of PDE5 and Rho-kinase may contribute to the lack of therapeutic efficacy of PDE5 inhibitors and Rho-kinase inhibitors in CTEPH.
Collapse
Affiliation(s)
- Kelly Stam
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, Rotterdam , The Netherlands
| | - Richard W van Duin
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, Rotterdam , The Netherlands
| | - André Uitterdijk
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, Rotterdam , The Netherlands
| | - Ilona Krabbendam-Peters
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, Rotterdam , The Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, Rotterdam , The Netherlands
| | - A H Jan Danser
- Department of Pharmacology, Erasmus Medical Center , Rotterdam , The Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, Rotterdam , The Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, Rotterdam , The Netherlands
| |
Collapse
|
12
|
Wilck N, Markó L, Balogh A, Kräker K, Herse F, Bartolomaeus H, Szijártó IA, Gollasch M, Reichhart N, Strauss O, Heuser A, Brockschnieder D, Kretschmer A, Lesche R, Sohler F, Stasch JP, Sandner P, Luft FC, Müller DN, Dechend R, Haase N. Nitric oxide-sensitive guanylyl cyclase stimulation improves experimental heart failure with preserved ejection fraction. JCI Insight 2018; 3:96006. [PMID: 29467337 DOI: 10.1172/jci.insight.96006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 01/11/2018] [Indexed: 12/17/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) can arise from cardiac and vascular remodeling processes following long-lasting hypertension. Efficacy of common HF therapeutics is unsatisfactory in HFpEF. Evidence suggests that stimulators of the nitric oxide-sensitive soluble guanylyl cyclase (NOsGC) could be of use here. We aimed to characterize the complex cardiovascular effects of NOsGC stimulation using NO-independent stimulator BAY 41-8543 in a double-transgenic rat (dTGR) model of HFpEF. We show a drastically improved survival rate of treated dTGR. We observed less cardiac fibrosis, macrophage infiltration, and gap junction remodeling in treated dTGR. Microarray analysis revealed that treatment of dTGR corrected the dysregulateion of cardiac genes associated with fibrosis, inflammation, apoptosis, oxidative stress, and ion channel function toward an expression profile similar to healthy controls. Treatment reduced systemic blood pressure levels and improved endothelium-dependent vasorelaxation of resistance vessels. Further comprehensive in vivo phenotyping showed an improved diastolic cardiac function, improved hemodynamics, and less susceptibility to ventricular arrhythmias. Short-term BAY 41-8543 application in isolated untreated transgenic hearts with structural remodeling significantly reduced the occurrence of ventricular arrhythmias, suggesting a direct nongenomic role of NOsGC stimulation on excitation. Thus, NOsGC stimulation was highly effective in improving several HFpEF facets in this animal model, underscoring its potential value for patients.
Collapse
Affiliation(s)
- Nicola Wilck
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Lajos Markó
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - András Balogh
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Kristin Kräker
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - István A Szijártó
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Nadine Reichhart
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Olaf Strauss
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Arnd Heuser
- Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | | - Ralf Lesche
- Bayer AG, Drug Discovery, Wuppertal & Berlin, Germany
| | | | | | - Peter Sandner
- Bayer AG, Drug Discovery, Wuppertal & Berlin, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany.,HELIOS-Klinikum, Berlin, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
13
|
Effect of Riociguat and Sildenafil on Right Heart Remodeling and Function in Pressure Overload Induced Model of Pulmonary Arterial Banding. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3293584. [PMID: 29511676 PMCID: PMC5817266 DOI: 10.1155/2018/3293584] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/04/2017] [Accepted: 11/16/2017] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by remodeling of the pulmonary vasculature and a rise in right ventricular (RV) afterload. The increased RV afterload leads to right ventricular failure (RVF) which is the reason for the high morbidity and mortality in PAH patients. The objective was to evaluate the therapeutic efficacy and antiremodeling potential of the phosphodiesterase type 5 (PDE5) inhibitor sildenafil and the soluble guanylate cyclase stimulator riociguat in a model of pressure overload RV hypertrophy induced by pulmonary artery banding (PAB). Mice subjected to PAB, one week after surgery, were treated with either sildenafil (100 mg/kg/d, n = 5), riociguat (30 mg/kg/d, n = 5), or vehicle (n = 5) for 14 days. RV function and remodeling were assessed by right heart catheterization, magnetic resonance imaging (MRI), and histomorphometry. Both sildenafil and riociguat prevented the deterioration of RV function, as determined by a decrease in RV dilation and restoration of the RV ejection fraction (EF). Although both compounds did not decrease right heart mass and cellular hypertrophy, riociguat prevented RV fibrosis induced by PAB. Both compounds diminished TGF-beta1 induced collagen synthesis of RV cardiac fibroblasts in vitro. Treatment with either riociguat or sildenafil prevented the progression of pressure overload-induced RVF, representing a novel therapeutic approach.
Collapse
|
14
|
Barnes JW, Tonelli AR, Heresi GA, Newman JE, Mellor NE, Grove DE, Dweik RA. Novel methods in pulmonary hypertension phenotyping in the age of precision medicine (2015 Grover Conference series). Pulm Circ 2016; 6:439-447. [PMID: 28090286 PMCID: PMC5210071 DOI: 10.1086/688847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 12/24/2022] Open
Abstract
Among pulmonary vascular diseases, pulmonary hypertension (PH) is the best studied and has been the focus of our work. The current classification of PH is based on a relatively simple combination of patient characteristics and hemodynamics. This leads to inherent limitations, including the inability to customize treatment and the lack of clarity from a more granular identification based on individual patient phenotypes. Accurate phenotyping of PH can be used in the clinic to select therapies and determine prognosis and in research to increase the homogeneity of study cohorts. Rapid advances in the mechanistic understanding of the disease, improved imaging methods, and innovative biomarkers now provide an opportunity to define novel PH phenotypes. We have recently shown that altered metabolism may affect nitric oxide levels and protein glycosylation, the peripheral circulation (which may provide insights into the response to therapy), and exhaled-breath analysis (which may be useful in disease evaluation). This review is based on a talk presented during the 2015 Grover Conference and highlights the relevant literature describing novel methods to phenotype pulmonary arterial hypertension patients by using approaches that involve the pulmonary and systemic (peripheral) vasculature. In particular, abnormalities in metabolism, the pulmonary and peripheral circulation, and exhaled breath in PH may help identify phenotypes that can be the basis for a precision-medicine approach to PH management. These approaches may also have a broader scope and may contribute to a better understanding of other diseases, such as asthma, diabetes, and cancer.
Collapse
Affiliation(s)
- Jarrod W. Barnes
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Adriano R. Tonelli
- Department of Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Gustavo A. Heresi
- Department of Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jennie E. Newman
- Department of Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Noël E. Mellor
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - David E. Grove
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Raed A. Dweik
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
15
|
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare but life-threatening form of pulmonary artery hypertension that is defined as a mean arterial pulmonary pressure greater than 25mmHg that persists for more than 6 months following anticoagulation therapy in the setting of pulmonary emboli. CTEPH is categorized by the World Health Organization as group IV pulmonary hypertension and is thought to be due to unresolved thromboemboli in the pulmonary artery circulation. Among the 5 classes of pulmonary hypertension, CTEPH is unique in that it is potentially curable with the use of pulmonary thromboendarterectomy surgery. Despite an increasing array of medical and surgical treatment options for patients with CTEPH over the past 2 decades, patients commonly present with advanced disease and carry a poor prognosis, thus, the need for early diagnosis and appropriate referral to an expert center. This review article first highlights the epidemiology, pathophysiology, and clinical presentation of CTEPH. The article then provides diagnostic and therapeutic algorithms for the management of the patient with suspected CTEPH.
Collapse
|