1
|
Bajpai J, Saxena M, Pradhan A, Kant S. Sotatercept: A novel therapeutic approach for pulmonary arterial hypertension through transforming growth factor-β signaling modulation. World J Methodol 2025; 15:102688. [DOI: 10.5662/wjm.v15.i3.102688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 03/06/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease marked by degeneration of the lung’s blood vessels. As the disease progresses, the resistance to blood flow in the pulmonary arteries increases, putting a strain on the right side of the heart as it pumps blood through the lungs. PAH is characterized by changes in the structure of blood vessels and excessive cell growth. Untreated PAH leads to irreversible right-sided heart failure, often despite medical intervention. Patients experience a gradual decline in function until they are unable to perform daily activities. Advances in treatment have improved the prognosis for many PAH patients. Currently approved therapies target the prostacyclin, endothelin, nitric oxide, or phosphodiesterase pathways to slow the progression of the disease. To address the unmet need for effective PAH therapies, research efforts are focused on identifying new targets and developing therapies that specifically address the underlying disease mechanisms and restore vascular wall homeostasis. Among these, sotatercept, a fusion protein that targets the transforming growth factor-β superfamily signaling pathway, has emerged as a promising therapeutic option. In this review, we examine the available evidence from clinical trials to assess the potential of sotatercept as a treatment for PAH.
Collapse
Affiliation(s)
- Jyoti Bajpai
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Mehul Saxena
- Gandhi Memorial Hospital, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Akshyaya Pradhan
- Department of Cardiology, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Surya Kant
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| |
Collapse
|
2
|
Gao J, Pan H, Guo X, Huang Y, Luo JY. Endothelial Krüppel-like factor 2/4: Regulation and function in cardiovascular diseases. Cell Signal 2025; 130:111699. [PMID: 40023301 DOI: 10.1016/j.cellsig.2025.111699] [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: 12/02/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
This review presents an overview of the regulation, function, disease-relevance and pharmacological regulation of the critical endothelial transcription factors KLF2/4 in vasculature. The regulatory mechanisms of KLF2/4 expression and activity in vascular endothelium in response to hemodynamic forces and biochemical stimuli are depicted. The functional effects mediated by direct or indirect target genes of KLF2/4 in endothelial cells are systematically summarized. The contributory roles that dysregulated KLF2/4 play in relevant cardiovascular pathologies, such as atherosclerotic vascular lesions, pulmonary arterial hypertension and vascular complications of diabetes were reviewed. Moreover, this review also discusses the pharmacological regulation of KLF2/4 by drugs used in clinics and therapeutic possibility by directly targeting these two transcription factors for treating atherosclerotic cardiovascular diseases. Finally, prospective opinions on the gaps in disclosing novel vascular function mediated by KLF2/4 and future research needs are expressed.
Collapse
Affiliation(s)
- Jing Gao
- Department of Cardiology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Hongjie Pan
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Jiang-Yun Luo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Sakurai M, Hosokawa S, Yamaguchi Y, Kirimura S, Ihara K, Ohashi K, Furukawa T, Sasano T, Kashimada K, Ishii T. Cyclopamine Attenuates Pulmonary Arterial Hypertension Development: Implications of Hedgehog Signaling Involvement for the Pathophysiology. FASEB J 2025; 39:e70628. [PMID: 40353829 DOI: 10.1096/fj.202403350r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/24/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Pulmonary arterial hypertension (PAH) is one of the most severe pulmonary diseases. Although combination therapies of the key drugs have improved survival rates, unmet needs remain in its management. We examined the effects of cyclopamine, a Hedgehog (HH) signaling inhibitor, as a potential novel therapeutic approach for PAH. C57BL/6J male mice were exposed to 10% oxygen for 3 weeks to induce pulmonary hypertension. One week after hypoxia exposure, these mice were treated with cyclopamine or vehicle. Cyclopamine significantly attenuated right ventricular (RV) systolic pressure (H + C: 31 mmHg vs. H: 38 mmHg, p < 0.01) and RV hypertrophy (H + C: 0.28 vs. H: 0.37, p < 0.01). The fully muscularized small pulmonary arteries significantly decreased with cyclopamine (H + C: 30% vs. H: 80%, p < 0.01), suggesting a mediation by vascular remodeling inhibition. In vitro, human pulmonary arterial smooth muscle cells (HPASMC) exposed to hypoxia revealed that the inhibitory action of cyclopamine was limited to hypoxia-promoted cell proliferation. In single-cell RNA sequencing analysis of mice lungs treated with cyclopamine, the signaling pathways of vascular smooth muscle contraction and cGMP-PKG, that is, key regulators in PAH development through vascular remodeling, were suppressed in cells with the characteristics of vascular endothelial and smooth muscle cells. RNA sequencing analysis of hypoxia-exposed hPASMCs revealed that the pathways related to extracellular matrix regulation were particularly recovered. Our animal model-based data revealed that HH signaling inhibition would improve PAH development by suppressing pulmonary vascular remodeling.
Collapse
MESH Headings
- Animals
- Veratrum Alkaloids/pharmacology
- Hedgehog Proteins/metabolism
- Male
- Mice
- Signal Transduction/drug effects
- Mice, Inbred C57BL
- Humans
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/drug therapy
- Hypoxia
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/drug therapy
- Vascular Remodeling/drug effects
- Cell Proliferation/drug effects
Collapse
Affiliation(s)
- Makito Sakurai
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
| | - Susumu Hosokawa
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
- Department of Pediatrics, Japanese Red Cross Musashino Hospital, Tokyo, Japan
| | - Yohei Yamaguchi
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
| | - Susumu Kirimura
- Department of Pathology, Institute of Science Tokyo, Tokyo, Japan
| | - Kensuke Ihara
- Department of Cardiovascular Medicine, Institute of Science Tokyo, Tokyo, Japan
| | - Kenichi Ohashi
- Department of Human Pathology, Institute of Science Tokyo, Tokyo, Japan
| | - Tetsushi Furukawa
- Department of Bio-Informational Pharmacology, Medical Research Institute, Institute of Science Tokyo, Tokyo, Japan
| | - Tetsuo Sasano
- Department of Cardiovascular Medicine, Institute of Science Tokyo, Tokyo, Japan
| | - Kenichi Kashimada
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
- Department of Pediatric Endocrinology, National Center for Child Health and Development, Tokyo, Japan
| | - Taku Ishii
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Flores CV, Chan SY. Therapeutic targets for pulmonary arterial hypertension: insights into the emerging landscape. Expert Opin Ther Targets 2025:1-17. [PMID: 40368635 DOI: 10.1080/14728222.2025.2507034] [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: 02/24/2025] [Revised: 04/21/2025] [Accepted: 05/13/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a progressive, life-threatening disease driven by vascular remodeling, right ventricular (RV) dysfunction, and metabolic and inflammatory dysregulation. Current therapies primarily target vasodilation to relieve symptoms but do not reverse disease progression. The recent approval of sotatercept, which modulates BMP/TGF-β signaling, marks a shift toward anti-remodeling therapies. Building on this, recent preclinical advances have identified promising therapeutic targets and potentially disease-modifying treatments. AREAS COVERED This review synthesizes the evolving preclinical landscape of emerging PAH therapeutic targets and drugs, highlighting innovative approaches aimed at addressing the underlying mechanisms of disease progression. Additionally, we discuss novel therapeutic strategies under development. EXPERT OPINION Recent advances in PAH research have identified novel therapeutic targets beyond vasodilators, including modulation of BMP/TGF-β signaling, metabolic programs, epigenetics, cancer-related signaling, the extracellular matrix, and immune pathways, among others. Sotatercept represents a significant advance in therapies that go beyond vasodilation, and long-term safety, efficacy, and durability are being assessed. Future treatment strategies will focus on precision approaches, noninvasive technologies, and regenerative biology to improve outcomes and reverse vascular remodeling.
Collapse
Affiliation(s)
- Christopher V Flores
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Cracowski JL, Khouri C. Sotatercept in pulmonary arterial hypertension. Trends Pharmacol Sci 2025:S0165-6147(25)00074-4. [PMID: 40382242 DOI: 10.1016/j.tips.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Affiliation(s)
- Jean-Luc Cracowski
- Université Grenoble Alpes, HP2 Laboratory, Inserm U1300, F-38000 Grenoble, France; Grenoble Alpes University Hospital, Pharmacovigilance Unit, F-38000 Grenoble, France.
| | - Charles Khouri
- Université Grenoble Alpes, HP2 Laboratory, Inserm U1300, F-38000 Grenoble, France; Grenoble Alpes University Hospital, Pharmacovigilance Unit, F-38000 Grenoble, France
| |
Collapse
|
6
|
Rothman AMK, Florentin A, Zink F, Quigley C, Bonneau O, Hemmig R, Hachey A, Rejtar T, Thaker M, Jain R, Huang SM, Sutton D, Roger J, Zhang JH, Weiler S, Cotesta S, Ottl J, Srivastava S, Kordonsky A, Avishid R, Yariv E, Rathi R, Khvalevsky O, Troxler T, Binmahfooz SK, Kleifeld O, Morrell NW, Humbert M, Thomas MJ, Jarai G, Beckwith REJ, Cobb JS, Smith N, Ostermann N, Tallarico J, Shaw D, Guth-Gundel S, Prag G, Rowlands DJ. Therapeutic potential of allosteric HECT E3 ligase inhibition. Cell 2025; 188:2603-2620.e18. [PMID: 40179885 PMCID: PMC12087876 DOI: 10.1016/j.cell.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 12/03/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025]
Abstract
Targeting ubiquitin E3 ligases is therapeutically attractive; however, the absence of an active-site pocket impedes computational approaches for identifying inhibitors. In a large, unbiased biochemical screen, we discover inhibitors that bind a cryptic cavity distant from the catalytic cysteine of the homologous to E6-associated protein C terminus domain (HECT) E3 ligase, SMAD ubiquitin regulatory factor 1 (SMURF1). Structural and biochemical analyses and engineered escape mutants revealed that these inhibitors restrict an essential catalytic motion by extending an α helix over a conserved glycine hinge. SMURF1 levels are increased in pulmonary arterial hypertension (PAH), a disease caused by mutation of bone morphogenetic protein receptor-2 (BMPR2). We demonstrated that SMURF1 inhibition prevented BMPR2 ubiquitylation, normalized bone morphogenetic protein (BMP) signaling, restored pulmonary vascular cell homeostasis, and reversed pathology in established experimental PAH. Leveraging this deep mechanistic understanding, we undertook an in silico machine-learning-based screen to identify inhibitors of the prototypic HECT E6AP and confirmed glycine-hinge-dependent allosteric activity in vitro. Inhibiting HECTs and other glycine-hinge proteins opens a new druggable space.
Collapse
Affiliation(s)
- Alexander M K Rothman
- University of Sheffield, Sheffield, UK; Novartis BioMedical Research (NBR), Cambridge, MA, USA.
| | - Amir Florentin
- School of Neurobiology, Biochemistry and Biophysics, The Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Florence Zink
- Novartis BioMedical Research (NBR), Basel, Switzerland
| | | | | | - Rene Hemmig
- Novartis BioMedical Research (NBR), Basel, Switzerland
| | - Amanda Hachey
- Novartis BioMedical Research (NBR), Cambridge, MA, USA
| | - Tomas Rejtar
- Novartis BioMedical Research (NBR), Cambridge, MA, USA
| | - Maulik Thaker
- Novartis BioMedical Research (NBR), Cambridge, MA, USA
| | - Rishi Jain
- Novartis BioMedical Research (NBR), Cambridge, MA, USA
| | | | - Daniel Sutton
- Novartis Institutes for BioMedical Research (NIBR), Horsham, UK
| | - Jan Roger
- Novartis Institutes for BioMedical Research (NIBR), Horsham, UK
| | - Ji-Hu Zhang
- Novartis BioMedical Research (NBR), Cambridge, MA, USA
| | - Sven Weiler
- Novartis BioMedical Research (NBR), Cambridge, MA, USA
| | | | - Johannes Ottl
- Novartis BioMedical Research (NBR), Basel, Switzerland
| | | | - Alina Kordonsky
- School of Neurobiology, Biochemistry and Biophysics, The Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Reut Avishid
- School of Neurobiology, Biochemistry and Biophysics, The Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Elon Yariv
- School of Neurobiology, Biochemistry and Biophysics, The Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Ritu Rathi
- School of Neurobiology, Biochemistry and Biophysics, The Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Oshrit Khvalevsky
- School of Neurobiology, Biochemistry and Biophysics, The Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Thomas Troxler
- School of Neurobiology, Biochemistry and Biophysics, The Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel; Novartis BioMedical Research (NBR), Basel, Switzerland
| | - Sarah K Binmahfooz
- University of Sheffield, Sheffield, UK; Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Oded Kleifeld
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Nicholas W Morrell
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Marc Humbert
- Université Paris-Saclay, INSERM UMR_S 999 (HPPIT), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre (Assistance Publique Hôpitaux de Paris), Le Kremlin Bicêtre, France
| | | | - Gabor Jarai
- Novartis Institutes for BioMedical Research (NIBR), Horsham, UK
| | | | | | - Nichola Smith
- Novartis BioMedical Research (NBR), Cambridge, MA, USA
| | | | | | - Duncan Shaw
- Novartis BioMedical Research (NBR), Cambridge, MA, USA
| | | | - Gali Prag
- School of Neurobiology, Biochemistry and Biophysics, The Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| | | |
Collapse
|
7
|
Constant B, Kamzolas I, Yang X, Guo J, Rodriguez-Fdez S, Mali I, Rodriguez-Cuenca S, Petsalaki E, Vidal-Puig A, Li W. Distinct signalling dynamics of BMP4 and BMP9 in brown versus white adipocytes. Sci Rep 2025; 15:15971. [PMID: 40335635 PMCID: PMC12059129 DOI: 10.1038/s41598-025-99122-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Adipocyte dysfunction contributes to lipotoxicity and cardiometabolic diseases. Bone morphogenetic protein 4 (BMP4) is expressed in white adipocytes and remodels white adipose tissue, while liver-derived BMP9, a key circulating BMP, influences adipocyte lipid metabolism. The gene sets regulated by BMP4 and BMP9 signalling in mature adipocytes remain unclear. Here, we directly compare BMP4 and BMP9 signalling in mature brown and white adipocytes. While both BMPs showed comparable potency across adipocyte types, RNA sequencing analysis revealed extensive gene regulation, with many more differentially expressed genes and suppression of critical metabolic pathways in white adipocytes. Although BMP4 and BMP9 induced inhibitors of BMP and GDF signalling in both adipocytes, they selectively upregulated several TGF-β family receptors and BMP4 expression only in white adipocytes. These findings underscore a central role of BMP signalling in adipocyte homeostasis and suggest both BMP4 and BMP9 as regulators of white adipocyte plasticity with potential therapeutic implications.
Collapse
Affiliation(s)
- Benjamin Constant
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Ioannis Kamzolas
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Xudong Yang
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Jingxu Guo
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Sonia Rodriguez-Fdez
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Iman Mali
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Sergio Rodriguez-Cuenca
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Antonio Vidal-Puig
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK.
- CIBERDEN, Centro de Investigacion Principe Felipe, Valencia, Spain.
| | - Wei Li
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| |
Collapse
|
8
|
Madonna R, Ghelardoni S. Sotatercept in pulmonary hypertension and beyond. Eur J Clin Invest 2025; 55:e14386. [PMID: 39825683 DOI: 10.1111/eci.14386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025]
Abstract
Sotatercept binds free activins by mimicking the extracellular domain of the activin receptor type IIA (ACTRIIA). Additional ligands are BMP/TGF-beta, GDF8, GDF11 and BMP10. The binding with activins leads to the inhibition of the signalling pathway and the deactivation of the bone morphogenic protein (BMP) receptor type 2. In this way, sotatercept activates an antiproliferative signalling to the cells of the pulmonary arteries and arterioles with the aim of rebalancing the proliferative and antiproliferative pathway that characterizes the pulmonary arterial hypertension (PAH). Sotatercept is indicated for the treatment of group 1 PAH in combination with drugs that act through the endothelin receptor, nitric oxide or prostacyclin. Its effects, demonstrated in the STELLAR study, are the improvement of exercise capacity and the FC-WHO functional class, together with the reduction of the risk of clinical worsening events. In addition to its antiremodeling effects on the pulmonary circulation, sotatercept has several haematological effects that could suggest its use in the treatment of some blood disorders other than PAH. In this review, we will discuss the effects of the drug on PAH and in parallel provide an in-depth overview of its application in haematological disorders, focusing on clinical and preclinical studies.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy
| | - Sandra Ghelardoni
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Laboratory of Biochemistry, University of Pisa, Pisa, Italy
| |
Collapse
|
9
|
Zhang H, Lu X, Guo Z, Jiang X, Zhang W, Wang S, Liu Q, Dong X, Li Y, Guo L, Zhang Y, Liu J, Zhang Z, Xie W, Song W, Zhang H, Zhai Z, Yang P. Development of a clinically relevant rat model of chronic thromboembolic pulmonary hypertension by combining splenectomy with pulmonary thromboembolism. Thromb Res 2025; 249:109310. [PMID: 40132406 DOI: 10.1016/j.thromres.2025.109310] [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: 12/31/2024] [Revised: 03/04/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Chronic thromboembolic pulmonary hypertension (CTEPH) is a severe condition resulting from unresolved thrombi in the pulmonary arteries, leading to increased pulmonary vascular resistance and right heart failure. Currently, the scarcity of clinically relevant animal models of CTEPH significantly hampers mechanistic studies and drug development. METHODS This study aimed to establish a rat model of CTEPH by combining splenectomy with thrombus injection, simulating key clinical risk factors associated with the disease. Rats underwent splenectomy and subsequent intravenous administration of thrombi, followed by hemodynamic and histological measurements as well as lung tissue RNA sequencing. RESULTS Splenectomized rats exhibited significant increases in platelets and delayed thrombolysis. Five weeks after splenectomy and thrombus injection, the rats exhibited thrombus retention in large pulmonary arteries, increased right ventricular systolic pressure, and pulmonary vascular remodeling, which were characteristic of CTEPH. Transcriptomic analysis revealed increased expression of inflammatory cytokines Ccl2 and Ccl3, as well as the B cell marker Cd79a, which was confirmed as an increase in CD79A+ B cells in the lung tissue. CONCLUSIONS Overall, this novel approach of combining splenectomy with thrombus injection provides a clinically relevant model for studying CTEPH pathophysiology and evaluating potential therapeutic interventions.
Collapse
Affiliation(s)
- Haobing Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Xiaoxuan Lu
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Zhuangjie Guo
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Xuehan Jiang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Wensi Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Shuang Wang
- School of Basic Medicine, Inner Mongolia Medical University, Inner Mongolia, Hohhot 010000, China
| | - Qiwei Liu
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Xiaotong Dong
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yishan Li
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Lina Guo
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yu Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jixiang Liu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Zhu Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wanmu Xie
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wanlu Song
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Hong Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| | - Zhenguo Zhai
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
10
|
Lordan J, Weatherald J. Pulmonary arterial hypertension and targeting pulmonary vascular remodelling: are we there yet? Eur Respir J 2025; 65:2500322. [PMID: 40335086 DOI: 10.1183/13993003.00322-2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 03/26/2025] [Indexed: 05/09/2025]
Affiliation(s)
- James Lordan
- Pulmonary Vascular Unit, Freeman Hospital, Newcastle upon Tyne, UK
| | - Jason Weatherald
- Department of Medicine, Division of Pulmonary Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
11
|
Hoeper MM, Gomberg-Maitland M, Badesch DB, Gibbs JSR, Grünig E, Kopeć G, McLaughlin VV, Meyer G, Olsson KM, Preston IR, Rosenkranz S, Souza R, Waxman AB, Perchenet L, Strait J, Xing A, Manimaran S, Wang X, Miller B, Cornell AG, de Oliveira Pena J, Ghofrani HA, Humbert M. Efficacy and safety of the activin signalling inhibitor, sotatercept, in a pooled analysis of PULSAR and STELLAR studies. Eur Respir J 2025; 65:2401424. [PMID: 39884760 PMCID: PMC12056246 DOI: 10.1183/13993003.01424-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025]
Abstract
INTRODUCTION Pulmonary arterial hypertension is a progressive disease associated with significant morbidity and mortality. Sotatercept is a first-in-class activin signalling inhibitor that acts to restore the balance between the growth-promoting and growth-inhibiting signalling pathways. METHODS This post hoc, exploratory, pooled analysis combines data from the double-blind placebo periods of the phase 2 PULSAR (NCT03496207) and phase 3 STELLAR (NCT04576988) studies. Both studies were international, multicentre, randomised, double-blind, placebo-controlled trials in patients with pulmonary arterial hypertension. Efficacy and safety parameters common to both studies were analysed. RESULTS A total of 429 patients were randomised and treated; 237 received sotatercept and 192 received placebo. Adding sotatercept to background pulmonary arterial hypertension therapy for 24 weeks improved exercise capacity (as assessed by 6-min walk distance), pulmonary vascular resistance and World Health Organization functional class, and delayed time to first occurrence of death or clinical worsening event. There were clinically important reductions in both pulmonary and right heart pressures; improvements in right ventricle size during both systole and diastole; and enhancements in right ventricle contractility and right ventricular-pulmonary artery coupling. The number of patients who experienced at least one adverse event of interest or special interest (increased haemoglobin, thrombocytopenia, bleeding events (mostly epistaxis), increased blood pressure and telangiectasia) was higher in the sotatercept group than the placebo group. CONCLUSION This pooled analysis confirms that sotatercept delivers therapeutic benefit across a range of efficacy end-points and has favourable safety in patients with pulmonary arterial hypertension. Increased duration of follow-up will provide further insight into long-term outcomes of sotatercept in patients with pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Marius M Hoeper
- Hannover Medical School and the German Center for Lung Research, Hannover, Germany
| | | | - David B Badesch
- University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - J Simon R Gibbs
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ekkehard Grünig
- Thoraxklinik-Heidelberg and the German Center for Lung Research, Heidelberg, Germany
| | - Grzegorz Kopeć
- The Pulmonary Circulation Center, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, St. John Paul II Hospital in Krakow, Krakow, Poland
| | | | - Gisela Meyer
- Irmandade da Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, Brazil
| | - Karen M Olsson
- Hannover Medical School and the German Center for Lung Research, Hannover, Germany
| | | | - Stephan Rosenkranz
- Department of Cardiology, Heart Center, University Hospital Cologne, and Cologne Cardiovascular Research Center (CCRC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Rogerio Souza
- Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | - H Ardeschir Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Marc Humbert
- Université Paris-Saclay, INSERM Unité Mixte de Recherche en Santé 999 (HPPIT), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre (Assistance Publique-Hôpitaux de Paris), Le Kremlin-Bicêtre, France
| |
Collapse
|
12
|
Sun J, Lin J, Yin D, Pan Z, Ye Y, Wang Y, Wang X, Guo Q. Androgen receptor inhibitor ameliorates pulmonary arterial hypertension by enhancing the apoptosis level through suppressing the Notch3/Hes5 pathway. Front Pharmacol 2025; 16:1572489. [PMID: 40356960 PMCID: PMC12067419 DOI: 10.3389/fphar.2025.1572489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Background Pulmonary arterial hypertension (PAH) exhibits significant gender differences in prognosis, with male patients typically showing worse outcomes than females. These disparities may stem from differences in androgen receptor expression and activity. Clinical studies suggest that the androgen receptor plays a crucial role in the pathophysiology of PAH, influencing disease progression and treatment response. Despite the lack of targeted therapies for PAH, these findings have spurred investigations into the potential therapeutic role of androgen receptors. This study explores the role of androgen receptors in PAH and evaluates their therapeutic potential. Methods PAH was induced in rats via intraperitoneal injection of monocrotaline (MCT). Following model establishment, Enzalutamide was administered every 3 days at 10 mg/kg once for a total of 7 times (21 days). A mouse model of PAH was developed by subcutaneously injecting SU5416 and exposing the mice to hypoxia. Androgen receptor knockout (AR-/-) mice were also utilized to investigate the role of androgen receptors in disease progression. Key indicators were compared across groups. The in vivo mechanisms through which androgen receptors influence PAH were examined in both rat and mouse models. Additionally, mouse pulmonary artery endothelial cells (PAECs) were cultured under hypoxic conditions to create an in vitro model of PAH, facilitating further investigation into the role of androgen receptors in disease pathogenesis. Results Compared to the normal group, the model group exhibited significantly increased androgen receptor expression in rats, mice, and mPAECs. This was accompanied by pronounced pulmonary artery wall thickening, right ventricular hypertrophy, pulmonary fibrosis, elevated pulmonary artery pressure, and a reduced level of apoptosis both in vivo and in vitro. Furthermore, activation of the Notch3/Hes5 signaling pathway was observed. However, treatment with androgen receptor inhibitors or gene knockout significantly ameliorated these pathological changes. Apoptosis levels increased both in vivo and in vitro, and the activation of the Notch3/Hes5 signaling pathway was effectively inhibited. Conclusion Our findings suggest that in both animal models and the hypoxic mPAECs, inhibition of androgen receptor expression leads to increased apoptosis via suppression of the Notch3/Hes5 signaling pathway. This mechanism likely contributes to the therapeutic effects observed, providing insights for potential treatment strategies targeting androgen receptors in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Jiayan Sun
- Medical College of Soochow Universuty, Soochow University, Suzhou, Jiangsu, China
- Medical Center of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu, China
| | - Jiancheng Lin
- Medical College of Soochow Universuty, Soochow University, Suzhou, Jiangsu, China
- Medical Center of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu, China
| | - Di Yin
- Medical College of Soochow Universuty, Soochow University, Suzhou, Jiangsu, China
- Medical Center of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu, China
| | - Zetao Pan
- Medical College of Soochow Universuty, Soochow University, Suzhou, Jiangsu, China
- Medical Center of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu, China
| | - Yuheng Ye
- Medical College of Soochow Universuty, Soochow University, Suzhou, Jiangsu, China
- Medical Center of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu, China
| | - Yi Wang
- Medical College of Soochow Universuty, Soochow University, Suzhou, Jiangsu, China
- Medical Center of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu, China
| | - Xiaowan Wang
- Medical College of Soochow Universuty, Soochow University, Suzhou, Jiangsu, China
- Medical Center of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu, China
| | - Qiang Guo
- Medical College of Soochow Universuty, Soochow University, Suzhou, Jiangsu, China
- Medical Center of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu, China
- The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
13
|
Weng Y, Wang X, Tang Y, Du C, Li X, Zhu K, Bao Y, Zeng W, Cai C, Jia B, Yang Z, Tang L. Inhibition of bone morphogenetic protein 4 alleviates angiotensin II-induced abdominal aortic aneurysm by reducing inflammation and endothelial-mesenchymal transition. Atherosclerosis 2025; 403:119134. [PMID: 40081251 DOI: 10.1016/j.atherosclerosis.2025.119134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 01/21/2025] [Accepted: 02/15/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND AND AIMS Abdominal aortic aneurysm (AAA) is one of the most common fatal macrovascular diseases worldwide which pathogenesis is still not well clarified. In this study, we systematically investigated the alternations of endothelial cell (ECs) functions and phenotypes by single-cell RNA sequencing in angiotensin (Ang) II-induced AAA mice models. METHOD AND RESULTS According to 10 × single-cell sequencing analysis, we revealed that ECs inflammation and endothelial-mesenchymal transition (EndoMT) were involved in the progress of Ang II-induced AAA. Three types of ECs, including Mature ECs (uninjured ECs), EndoMT ECs and Injury & inflammation ECs successively emerged during the progression of AAA. By using pseudotime-trajectory analysis, we speculated bone morphogenetic protein 4 (BMP4) as a candidate gene, participating in Ang II-induced AAA by regulating EndoMT and vascular inflammation. We found that inhibition of BMP4 ameliorated EndoMT and vascular inflammation in Ang II-induced AAA in vivo. In addition, we found that exogenous BMP4 directly promoted the phenotypic transition, inflammation, cell migration and invasion of mouse aortic endothelial cells via PI3K/AKT/mTOR pathways in vitro. Finally, Protein-protein interaction (PPI) analysis and co-immunoprecipitation (Co-IP) revealed that biglycan (BGN) directly combined with BMP4 and promoted the conversion of EndoMT. CONCLUSION Our findings firstly revealed a critical role of BMP4 in AAA progression, which promoted disease progression by inducing EndoMT and reprogramming ECs from anti-inflammatory to proinflammatory phenotype.
Collapse
Affiliation(s)
- Yingzheng Weng
- Department of Medicine, The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310013, China; Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Xihao Wang
- Department of Medicine, The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310013, China
| | - Yimin Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Changqing Du
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Xinyao Li
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Kefu Zhu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Yizhong Bao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Wenping Zeng
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Changhong Cai
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Bingbing Jia
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China.
| | - Zhouxin Yang
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China.
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China.
| |
Collapse
|
14
|
Dunmore BJ, Kikuchi N, Li W, Upton PD, Morrell NW. Activin-A Regulates Bone Morphogenetic Protein Signaling in Pulmonary Endothelial Cells Without Affecting Bone Morphogenetic Protein Type-II Receptor Expression. Pulm Circ 2025; 15:e70095. [PMID: 40330556 PMCID: PMC12052755 DOI: 10.1002/pul2.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
Activin-A is elevated in pulmonary arterial hypertension (PAH) patients, and reportedly suppresses BMPR-II. This suggests one mechanism of action for PAH drug, sotatercept, an activin-ligand trap. However, we were unable to confirm that activin-A reduces BMPR-II in pulmonary endothelial cells. Thus, it seems unlikely that sotatercept influences BMPR-II or PAH via this mechanism.
Collapse
Affiliation(s)
- Benjamin J. Dunmore
- Victor Phillip Dahdaleh Heart and Lung Research InstituteUniversity of CambridgeCambridgeUK
- Department of Medicine, School of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Nobuhiro Kikuchi
- Victor Phillip Dahdaleh Heart and Lung Research InstituteUniversity of CambridgeCambridgeUK
- Department of Medicine, School of Clinical MedicineUniversity of CambridgeCambridgeUK
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Wei Li
- Victor Phillip Dahdaleh Heart and Lung Research InstituteUniversity of CambridgeCambridgeUK
- Department of Medicine, School of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Paul D. Upton
- Victor Phillip Dahdaleh Heart and Lung Research InstituteUniversity of CambridgeCambridgeUK
- Department of Medicine, School of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Nicholas W. Morrell
- Victor Phillip Dahdaleh Heart and Lung Research InstituteUniversity of CambridgeCambridgeUK
- Department of Medicine, School of Clinical MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
15
|
Gomberg-Maitland M, Badesch DB, Gibbs JSR, Grünig E, Hoeper MM, Humbert M, Kopeć G, McLaughlin VV, Meyer G, Olsson KM, Preston IR, Rosenkranz S, Souza R, Waxman AB, Perchenet L, Strait J, Xing A, Johnson-Levonas AO, Cornell AG, de Oliveira Pena J, Ardeschir Ghofrani H. Efficacy and safety of sotatercept across ranges of cardiac index in patients with pulmonary arterial hypertension: A pooled analysis of PULSAR and STELLAR. J Heart Lung Transplant 2025; 44:609-624. [PMID: 39645016 DOI: 10.1016/j.healun.2024.11.037] [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: 08/23/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND This analysis examined the effects of the activin signaling inhibitor, sotatercept, in pulmonary arterial hypertension (PAH) subgroups stratified by baseline cardiac index (CI). METHODS Pooled data from PULSAR (N = 106; NCT03496207) and STELLAR (N = 323; NCT04576988) were analyzed using 2 different CI thresholds, <2.0 and ≥2.0 liter/min/m2 as well as <2.5 and ≥2.5 liter/min/m2. Median changes from baseline at week 24 were evaluated using Hodges-Lehmann estimator and least squares (LS) means, with 95% confidence intervals and p-values (significance: p = 0.05). Categorial endpoints and time-to-clinical worsening were analyzed by Cochran-Mantel-Haenszel and Cox model respectively. RESULTS Of 429 participants, 51 had CI <2.0 and 378 ≥2.0 liter/min/m2, while 179 had CI <2.5 and 250 ≥2.5 liter/min/m2. Sotatercept significantly improved median 6-minute walk distance (range: 33.9 to 63.7 m: p < 0.001), pulmonary vascular resistance (range: -202.8 to -395.4 dyn•s•cm-5; p ≤ 0.002), and N-terminal pro-B-type natriuretic peptide (range: -317.3 to -1,041.2 pg/ml; p < 0.001) across subgroups. LS means showed reductions in pulmonary and right atrial pressures, decreased right ventricular size, and improved tricuspid annular plane systolic excursion/systolic pulmonary artery pressure. Sotatercept delayed time to first occurrence of death or a worsening event for CI ≥2.5 (hazard ratio [HR] 0.12; p < 0.001), ≥2.0 (HR 0.13; p < 0.001), and <2.5 (HR 0.21; p < 0.001) liter/min/m2. Improvements were observed in WHO functional class (all p < 0.050) and ESC/ERS risk scores (all p < 0.001). CONCLUSIONS Sotatercept demonstrated consistent efficacy and safety across CI subgroups, supporting its use in PAH patients irrespective of baseline cardiac hemodynamics.
Collapse
Affiliation(s)
- Mardi Gomberg-Maitland
- Division of Cardiovascular Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia.
| | - David B Badesch
- Pulmonary Hypertension Center, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - J Simon R Gibbs
- Department of Cardiology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Ekkehard Grünig
- Center for Pulmonary Hypertension, Translational Lung Research Center Heidelberg (TLRC), Thoraxklinik-Heidelberg and the German Center for Lung Research, Heidelberg, Germany
| | - Marius M Hoeper
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School and the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH), Hannover, Germany
| | - Marc Humbert
- Université Paris-Saclay, INSERM Unité Mixte de Recherche en Santé 999, Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique (HPPIT), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre (Assistance Publique-Hôpitaux de Paris), Le Kremlin-Bicêtre, France
| | - Grzegorz Kopeć
- Department of Cardiac and Vascular Diseases, St. John Paul II Hospital in Krakow, Krakow, Poland; Pulmonary Circulation Centre, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Vallerie V McLaughlin
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gisela Meyer
- Departamento de Circulação Pulmonar, Irmandade da Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, Brazil
| | - Karen M Olsson
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School and the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH), Hannover, Germany
| | - Ioana R Preston
- Pulmonary and Critical Care Medicine, Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - Stephan Rosenkranz
- Department of Cardiology, Cologne Cardiovascular Research Center (CCRC), Heart Center, University Hospital Cologne, Cologne, Germany
| | - Rogerio Souza
- Divisão de Pneumologia, Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Aaron B Waxman
- Division of Pulmonary and Critical Care Medicine, Brigham and Woman's Hospital, Boston, Massachusetts
| | | | | | - Aiwen Xing
- MRL, Merck & Co., Inc., Rahway, New Jersey
| | | | | | | | - H Ardeschir Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
16
|
Ma W, Ma Y, Bai Y, Su X. Changes in Macrophages in Pulmonary Hypertension: A Focus on High-altitude Pulmonary Hypertension. Anatol J Cardiol 2025; 29:210-221. [PMID: 40062372 PMCID: PMC12053306 DOI: 10.14744/anatoljcardiol.2025.5013] [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: 11/14/2024] [Accepted: 01/23/2025] [Indexed: 05/08/2025] Open
Abstract
High-altitude pulmonary hypertension (HAPH) is a condition characterized by elevated pulmonary arterial pressure exceeding normal physiological values, resulting from a combination of high-altitude low-pressure, hypoxic environments, genetic susceptibility, immune dysfunction, and neurogenic disturbances. This condition predominantly manifests as right heart failure, severely impacting quality of life and life expectancy. Macrophages, as one of the most prevalent innate immune cells, have been increasingly recognized for their crucial role in the pathogenesis of HAPH. The low-pressure and hypoxic environment, along with other etiological factors, lead to metabolic abnormalities in tissue cells and the microenvironment. This results in increased secretion of chemokines, cytokines, and growth factors in the microenvironment, which promote the proliferation of tissue-resident macrophages and the differentiation of monocytes recruited from the blood into macrophages. This exacerbates the inflammatory cascade, further promoting cell proliferation, tissue repair, and inhibition of apoptosis. These processes contribute to the migration and proliferation of pulmonary arterial smooth muscle cells, endothelial cells, and fibroblasts, leading to vascular remodeling and ultimately the development of pulmonary arterial hypertension. This review examines the role of macrophage-mediated immune responses in high-altitude pulmonary arterial hypertension, with a focus on hypoxia as a key feature.
Collapse
Affiliation(s)
- Wende Ma
- Qinghai University, Xining, Qinghai, China
- Compact Medical Service Community in Menyuan County, Menyuan, Qinghai, China
| | - Yumei Ma
- Department of Digestive, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Yuting Bai
- Department of Cardiology, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Xiaoling Su
- Department of Cardiology, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| |
Collapse
|
17
|
Sharma M, Paudyal V, Syed SK, Thapa R, Kassam N, Surani S. Management of Pulmonary Arterial Hypertension: Current Strategies and Future Prospects. Life (Basel) 2025; 15:430. [PMID: 40141775 PMCID: PMC11943839 DOI: 10.3390/life15030430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Primary pulmonary hypertension (PPH), now known as pulmonary arterial hypertension (PAH), has induced significant treatment breakthroughs in the past decade. Treatment has focused on improving patient survival and quality of life, and delaying disease progression. Current therapies are categorized based on targeting different pathways known to contribute to PAH, including endothelin receptor antagonists (ERAs), phosphodiesterase-5 inhibitors (PDE-5 inhibitors), prostacyclin analogs, soluble guanylate cyclase stimulators, and activin signaling inhibitors such as Sotatercept. The latest addition to treatment options is soluble guanylate cyclase stimulators, such as Riociguat, which directly stimulates the nitric oxide pathway, facilitating vasodilation. Looking to the future, advancements in PAH treatment focus on precision medicine involving the sub-stratification of patients through a deep characterization of altered Transforming Growth Factor-β(TGF-β) signaling and molecular therapies. Gene therapy, targeting specific genetic mutations linked to PAH, and cell-based therapies, such as mesenchymal stem cells, are under investigation. Besides prevailing therapies, emerging PH treatments target growth factors and inflammation-modulating pathways, with ongoing trials assessing their long-term benefits and safety. Hence, this review explores current therapies that delay progression and improve survival, as well as future treatments with curative potential.
Collapse
Affiliation(s)
- Munish Sharma
- Division of Pulmonary, Critical Care and Sleep Medicine, Baylor Scott and White, Temple, TX 76508, USA;
| | - Vivek Paudyal
- Department of General Practice and Emergency Medicine, Karnali Academy of Health Sciences, Chandannath 21200, Jumla, Nepal; (V.P.); (R.T.)
| | - Saifullah Khalid Syed
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Rubi Thapa
- Department of General Practice and Emergency Medicine, Karnali Academy of Health Sciences, Chandannath 21200, Jumla, Nepal; (V.P.); (R.T.)
| | - Nadeem Kassam
- Department of Medicine, Aga Khan University, Nairobi 30270, Kenya;
| | - Salim Surani
- Department of Medicine and Pharmacy, Texas A&M, College Station, TX 77840, USA
| |
Collapse
|
18
|
Guglielmi G, Dimopoulos K, Wort SJ. New therapies in pulmonary arterial hypertension: Recent insights. INTERNATIONAL JOURNAL OF CARDIOLOGY CONGENITAL HEART DISEASE 2025; 19:100571. [PMID: 39991439 PMCID: PMC11847046 DOI: 10.1016/j.ijcchd.2025.100571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Pulmonary Arterial Hypertension (PAH) is a complex and progressive disease characterized by elevated pulmonary vascular resistance and right heart failure. Current therapies primarily focus on pulmonary vasodilation; however, novel approaches that target the underlying pathophysiological mechanisms-such as TGF-β signalling, epigenetic alterations, growth factors, inflammation, and extracellular matrix remodelling-are promising alternatives for improving treatment outcomes. This is a review of recent advances in the development of innovative therapeutic strategies for PAH. The first section of this paper explores approaches targeting TGF-β signalling, both acting directly on receptors through drugs like Sotatercept and exogenous BMP9, and indirectly, inhibiting the degradation of key receptors, such as BMPR2. Subsequent sections describe treatments that target epigenetic regulators, e.g. poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors and direct BRD4 antagonists, tyrosine kinase inhibitors (Seralutinib), and therapies aimed at inflammation, such as IL-6 inhibitors, CD-20 inhibitors, and monoclonal antibodies that prevent macrophage migration. Finally, strategies that target the serotonin pathway, and other metabolic and hormonal pathways are described. This review includes both preclinical and clinical trial data that support efficacy, safety and the future potential of such therapies. Collectively, these therapeutic approaches can be valuable in treating PAH by targeting multiple aspects of its pathogenesis, potentially resulting in improved clinical outcomes for patients affected by this debilitating, life-limiting condition.
Collapse
Affiliation(s)
- Giulia Guglielmi
- Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, United Kingdom
| | - Konstantinos Dimopoulos
- Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, United Kingdom
- National Pulmonary Hypertension Service, Royal Brompton Hospital, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - S. John Wort
- National Pulmonary Hypertension Service, Royal Brompton Hospital, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
19
|
Miranda AC, Cornelio CK, Tran BAC, Fernandez J. Sotatercept: A First-In-Class Activin Signaling Inhibitor for Pulmonary Arterial Hypertension. J Pharm Technol 2025:87551225251317957. [PMID: 39995630 PMCID: PMC11847314 DOI: 10.1177/87551225251317957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025] Open
Abstract
Objective: The objective of the study is to review the characteristics, efficacy, safety, and clinical relevance of sotatercept in pulmonary arterial hypertension (PAH). Data Sources: A literature search containing search terms related to sotatercept and PAH was conducted. Embase via Elsevier, MEDLINE via Ovid, the medRxiv preprint server, Cochrane Library CENTRAL trials registry, and ClinicalTrials.gov were searched from inception through October 31, 2024. The package insert was utilized to obtain drug information and additional data. Study Selection and Data Extraction: Phase II-III clinical trials investigating sotatercept for PAH were included. Articles written in English were extracted while animal studies and phase I clinical trials were excluded. Data Synthesis: In patients with WHO Group 1, functional class II-III PAH, adding sotatercept to background therapy increased 6-minute walk distance in phase II-III trials. Pooled analysis from PULSAR (phase II) and STELLAR (phase III) showed improvements in pulmonary vascular resistance and NT-proBNP. Exploratory data from PULSAR revealed that BMPR2 genetic variant status was not associated with significant differences in treatment effects. SPECTRA (phase IIb) demonstrated improved right ventricular structure and function. Interim analysis from SOTERIA showed that treatment effects persist at 1 year. Conclusions: Sotatercept is a viable add-on therapy for patients with PAH Group 1 and functional class II-III. Additional data are needed to assess long-term outcomes among treatment-naïve patients and those with the most severe symptomatology.
Collapse
Affiliation(s)
- Aimon C. Miranda
- Department of Pharmacotherapeutics and Clinical Research, Taneja College of Pharmacy, University of South Florida Health, Tampa, FL, USA
| | - Cyrille K. Cornelio
- Department of Pharmacotherapeutics and Clinical Research, Taneja College of Pharmacy, University of South Florida Health, Tampa, FL, USA
| | - Bao Anh C. Tran
- Department of Pharmacotherapeutics and Clinical Research, Taneja College of Pharmacy, University of South Florida Health, Tampa, FL, USA
| | - Joel Fernandez
- Internal Medicine, Morsani College of Medicine, University of South Florida Health, Tampa, FL, USA
| |
Collapse
|
20
|
Paudyal V, Thapa R, Basnet S, Sharma M, Surani S, Varon J. Updates on Pulmonary Hypertension. Open Respir Med J 2025; 19:e18743064344024. [PMID: 40322494 PMCID: PMC12046238 DOI: 10.2174/0118743064344024250203101417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/06/2024] [Accepted: 01/01/2025] [Indexed: 05/08/2025] Open
Abstract
Pulmonary Arterial Hypertension (PAH) is an uncommon condition with high mortality. It is an underrecognized condition both in developing and developed countries, especially in developing countries, due to a lack of advanced healthcare facilities and resources for timely diagnosis. More than half of the individuals diagnosed with PAH live less than five years after diagnosis. In recent years, tremendous advancements have been made in diagnostic and therapeutic strategies for PAH patients. Phosphodiesterase 5 (PDE5) inhibitors, endothelin receptor antagonists, and prostacyclin inhibitors in various forms (oral, inhaled, intravenous, or subcutaneous) have been the cornerstone of medical treatment. Atrial septostomy, heart and lung transplant, balloon pulmonary angioplasty, and pulmonary thromboendarterectomy are existing therapeutic options currently available. There has been a continuous effort to introduce newer therapies to improve life expectancy and modify disease. Newer therapies have shown promising results but require future data to guarantee long-term safety and efficacy. We aim to discuss a few of these critical updates in the constantly evolving field of PAH.
Collapse
Affiliation(s)
- Vivek Paudyal
- Department of General Practice and Emergency Medicine, Karnali Academy of Health Sciences, Jumla, Nepal
| | - Rubi Thapa
- Department of General Practice and Emergency Medicine, Karnali Academy of Health Sciences, Jumla, Nepal
| | - Sagarika Basnet
- Department of Internal Medicine, Kathmandu Medical College and Teaching Hospital, Kathmandu, Nepal
| | - Munish Sharma
- Department of Medicine, Baylor College of Medicine, Texas, TX, United States
| | - Salim Surani
- Department of Medicine, Texas A & M University, Texas, TX, United States
| | - Joseph Varon
- College of Medicine, University of Houston, Houston, United States
| |
Collapse
|
21
|
Huang S, Wu Y, Chen M, Shen J, Zhu J, Yu H. GDF11 improves cardiac repair after myocardial infarction by reducing Macrophage infiltration and attenuating their inflammatory Properties. Int Immunopharmacol 2025; 147:113994. [PMID: 39765001 DOI: 10.1016/j.intimp.2024.113994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/29/2025]
Affiliation(s)
- Shushi Huang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China
| | - Yuling Wu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China
| | - Mingyao Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China
| | - Jiahua Shen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China
| | - Jinyun Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China.
| | - Hong Yu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China; Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| |
Collapse
|
22
|
Ghofrani HA, Gomberg-Maitland M, Zhao L, Grimminger F. Mechanisms and treatment of pulmonary arterial hypertension. Nat Rev Cardiol 2025; 22:105-120. [PMID: 39112561 DOI: 10.1038/s41569-024-01064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 11/28/2024]
Abstract
Substantial progress has been made in the management of pulmonary arterial hypertension (PAH) in the past 25 years, but the disease remains life-limiting. Established therapies for PAH are mostly limited to symptomatic relief by correcting the imbalance of vasoactive factors. The tyrosine kinase inhibitor imatinib, the first predominantly non-vasodilatory drug to be tested in patients with PAH, improved exercise capacity and pulmonary haemodynamics compared with placebo but at the expense of adverse events such as subdural haematoma. Given that administration by inhalation might reduce the risk of systemic adverse effects, inhaled formulations of tyrosine kinase inhibitors are currently in clinical development. Other novel therapeutic approaches for PAH include suppression of activin receptor type IIA signalling with sotatercept, which has shown substantial efficacy in clinical trials and was approved for use in the USA in 2024, but the long-term safety of the drug remains unclear. Future advances in the management of PAH will focus on right ventricular function and involve deep phenotyping and the development of a personalized medicine approach. In this Review, we summarize the mechanisms underlying PAH, provide an overview of available PAH therapies and their limitations, describe the development of newer, predominantly non-vasodilatory drugs that are currently being tested in phase II or III clinical trials, and discuss future directions for PAH research.
Collapse
Affiliation(s)
- Hossein-Ardeschir Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany.
| | - Mardi Gomberg-Maitland
- George Washington University School of Medicine and Health Sciences, Department of Medicine, Washington, DC, USA
| | - Lan Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Friedrich Grimminger
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
23
|
Hashemi A, Kwak MJ, Goyal P. Pharmacologic Management of Heart Failure with Preserved Ejection Fraction (HFpEF) in Older Adults. Drugs Aging 2025; 42:95-110. [PMID: 39826050 DOI: 10.1007/s40266-024-01165-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2024] [Indexed: 01/20/2025]
Abstract
There are several pharmacologic agents that have been touted as guideline-directed medical therapy for heart failure with preserved ejection fraction (HFpEF). However, it is important to recognize that older adults with HFpEF also contend with an increased risk for adverse effects from medications due to age-related changes in pharmacokinetics and pharmacodynamics of medications, as well as the concurrence of geriatric conditions such as polypharmacy and frailty. With this review, we discuss the underlying evidence for the benefits of various treatments in HFpEF and incorporate key considerations for older adults, a subpopulation that may be at higher risk for adverse drug events. Key considerations for older adults include: the use of loop diuretics, mineralocorticoid receptor antagonists (MRAs), and sodium glucose co-transporter-2 (SGLT2) inhibitors for most; angiotensin receptor blockers/ angiotensin receptor-neprilysin inhibitors (ARB/ARNIs) and glucagon-like peptide-1 receptor agonists (GLP-1RAs) as add-on therapies for some, though risk of geriatric conditions such as falls, malnutrition, and/or sarcopenia must be considered; and beta blockers for a smaller subset of patients (with consideration of deprescribing for some, though data are lacking on this approach). Naturally, when making clinical decisions for older adults with cardiovascular disease, it is critical to consider the complexity of their conditions, including cognitive and physical function and social and environmental factors, and ensure alignment of care plans with the patient's health goals and priorities.
Collapse
Affiliation(s)
- Ashkan Hashemi
- Program for the Care and Study of the Aging Heart, Department of Medicine, Weill Cornell Medicine, 420 East 70th St, New York, NY, LH-36510063, USA
| | - Min Ji Kwak
- Division of Geriatric and Palliative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, USA
| | - Parag Goyal
- Program for the Care and Study of the Aging Heart, Department of Medicine, Weill Cornell Medicine, 420 East 70th St, New York, NY, LH-36510063, USA.
| |
Collapse
|
24
|
Castro C, Delwarde C, Shi Y, Roh J. Geroscience in heart failure: the search for therapeutic targets in the shared pathobiology of human aging and heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2025; 5:10.20517/jca.2024.15. [PMID: 40297496 PMCID: PMC12036312 DOI: 10.20517/jca.2024.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Age is a major risk factor for heart failure, but one that has been historically viewed as non-modifiable. Emerging evidence suggests that the biology of aging is malleable, and can potentially be intervened upon to treat age-associated chronic diseases, such as heart failure. While aging biology represents a new frontier for therapeutic target discovery in heart failure, the challenges of translating Geroscience research to the clinic are multifold. In this review, we propose a strategy that prioritizes initial target discovery in human biology. We review the rationale for starting with human omics, which has generated important insights into the shared (patho)biology of human aging and heart failure. We then discuss how this knowledge can be leveraged to identify the mechanisms of aging biology most relevant to heart failure. Lastly, we provide examples of how this human-first Geroscience approach, when paired with rigorous functional assessments in preclinical models, is leading to early-stage clinical development of gerotherapeutic approaches for heart failure.
Collapse
Affiliation(s)
- Claire Castro
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Constance Delwarde
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Yanxi Shi
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jason Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
25
|
Anand SC, Furqan M, Tonelli AR, Brady D, Levine A, Rosenzweig EB, Frishman WH, Aronow WS, Lanier GM. Sotatercept: A New Era in Pulmonary Arterial Hypertension. Cardiol Rev 2025:00045415-990000000-00397. [PMID: 39773755 DOI: 10.1097/crd.0000000000000837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by proliferative remodeling and obliterative narrowing of the pulmonary vasculature. While outcomes have improved with existing treatments targeting 3 main pathways, there remains a critical need for novel therapies that address different and novel mechanisms of PAH. Sotatercept, recently Food and Drug Administration (FDA) approved, is a groundbreaking fusion protein that binds to activin and growth differentiation factors, rebalancing antiproliferative and pro-proliferative signals to reverse remodeling in both the pulmonary vasculature and the right ventricle. This review highlights current evidence exploring the safety and efficacy of sotatercept in the 2 landmark trials, phase 2 Pulmonary Arterial Hypertension and Sotatercept Trial and Research and phase 3 Sotatercept Treatment in Expansion of Long-term Learning and Assessment in PAH trial, which were instrumental in securing FDA approval for adult PAH patients with WHO functional class II or III symptoms already receiving background pulmonary hypertension therapy. Overall, sotatercept represents a landmark advancement in PAH treatment, offering hope for patients and the potential to delay or avoid lung transplantation. Importantly, this marks the beginning of an era of targeted therapies aimed at reverse remodeling in PAH while improving outcomes.
Collapse
Affiliation(s)
- Suneesh C Anand
- From the Department of Pulmonary and Critical Care Medicine, Integrated Hospital Care Institute, Hillcrest Hospital, Cleveland Clinic, OH
- Department of Medicine, New York Medical College, Valhalla, NY
| | - Muhammad Furqan
- Department of Medicine, New York Medical College, Valhalla, NY
| | - Adriano R Tonelli
- Department of Pulmonary, Allergy, and Critical Care Medicine, Integrated Hospital Care Institute, Cleveland Clinic, Cleveland, OH
| | - Daniela Brady
- Department of Pediatrics and Medicine, Maria Fareri Children's Hospital of Westchester Medical Center, New York Medical College, Valhalla, NY
| | - Avi Levine
- Department of Medicine, New York Medical College, Valhalla, NY
- Department of Medicine Westchester Medical Center, Valhalla, NY
- Department of Cardiology, Westchester Medical Center, Valhalla, NY
| | - Erika B Rosenzweig
- Department of Pediatrics and Medicine, Maria Fareri Children's Hospital of Westchester Medical Center, New York Medical College, Valhalla, NY
| | | | - Wilbert S Aronow
- Department of Medicine, New York Medical College, Valhalla, NY
- Department of Medicine Westchester Medical Center, Valhalla, NY
- Department of Cardiology, Westchester Medical Center, Valhalla, NY
| | - Gregg M Lanier
- Department of Medicine, New York Medical College, Valhalla, NY
- Department of Medicine Westchester Medical Center, Valhalla, NY
- Department of Cardiology, Westchester Medical Center, Valhalla, NY
| |
Collapse
|
26
|
Forbes LM, Bauer N, Bhadra A, Bogaard HJ, Choudhary G, Goss KN, Gräf S, Heresi GA, Hopper RK, Jose A, Kim Y, Klouda T, Lahm T, Lawrie A, Leary PJ, Leopold JA, Oliveira SD, Prisco SZ, Rafikov R, Rhodes CJ, Stewart DJ, Vanderpool RR, Yuan K, Zimmer A, Hemnes AR, de Jesus Perez VA, Wilkins MR. Precision Medicine for Pulmonary Vascular Disease: The Future Is Now (2023 Grover Conference Series). Pulm Circ 2025; 15:e70027. [PMID: 39749110 PMCID: PMC11693987 DOI: 10.1002/pul2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Pulmonary vascular disease is not a single condition; rather it can accompany a variety of pathologies that impact the pulmonary vasculature. Applying precision medicine strategies to better phenotype, diagnose, monitor, and treat pulmonary vascular disease is increasingly possible with the growing accessibility of powerful clinical and research tools. Nevertheless, challenges exist in implementing these tools to optimal effect. The 2023 Grover Conference Series reviewed the research landscape to summarize the current state of the art and provide a better understanding of the application of precision medicine to managing pulmonary vascular disease. In particular, the following aspects were discussed: (1) Clinical phenotypes, (2) genetics, (3) epigenetics, (4) biomarker discovery, (5) application of precision biology to clinical trials, (6) the right ventricle (RV), and (7) integrating precision medicine to clinical care. The present review summarizes the content of these discussions and the prospects for the future.
Collapse
Affiliation(s)
- Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
| | - Natalie Bauer
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | - Aritra Bhadra
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Center for Lung BiologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
| | - Harm J. Bogaard
- Department of Pulmonary MedicineAmsterdam UMCAmsterdamNetherlands
| | - Gaurav Choudhary
- Division of CardiologyWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island and Miriam HospitalsProvidenceRhode IslandUSA
- Department of CardiologyProvidence VA Medical CenterProvidenceRhode IslandUSA
| | - Kara N. Goss
- Department of Medicine and PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Stefan Gräf
- Division of Computational Genomics and Genomic Medicine, Department of MedicineUniversity of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research InstituteCambridgeUK
| | | | - Rachel K. Hopper
- Department of PediatricsStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Arun Jose
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Yunhye Kim
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Timothy Klouda
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
- Division of Pulmonary, Critical Care, and Sleep MedicineNational Jewish HealthDenverColoradoUSA
- Pulmonary and Critical Care SectionRocky Mountain Regional VA Medical CenterDenverColoradoUSA
| | - Allan Lawrie
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Peter J. Leary
- Departments of Medicine and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Suellen D. Oliveira
- Department of Anesthesiology, Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sasha Z. Prisco
- Division of CardiovascularLillehei Heart Institute, University of MinnesotaMinneapolisMinnesotaUSA
| | - Ruslan Rafikov
- Department of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Duncan J. Stewart
- Ottawa Hospital Research InstituteFaculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Ke Yuan
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Alexsandra Zimmer
- Department of MedicineBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island HospitalProvidenceRhode IslandUSA
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | |
Collapse
|
27
|
Estrada RA, Sahay S, Tonelli AR. Treatment of pulmonary hypertension after seven world symposia. Ther Adv Respir Dis 2025; 19:17534666251342898. [PMID: 40405724 DOI: 10.1177/17534666251342898] [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: 05/24/2025] Open
Abstract
This review focuses on the advancements in the treatment of pulmonary hypertension (PH), especially after the Food and Drug Administration (FDA) approval of sotatercept and the advances in treatment recommendations after seven World Symposia on PH. PH, a complex and progressive condition defined hemodynamically by a mean pulmonary artery pressure >20 mmHg, encompasses multiple PH groups, each with distinct pathophysiological characteristics and treatment implications. Diagnosing PH can be challenging because symptoms like shortness of breath, fatigue, and chest pain are nonspecific. Contemporary treatment of pulmonary arterial hypertension aims to improve outcomes, symptoms, and overall quality of life, with a primary focus on preventing and treating right ventricular failure. Comprehensive risk stratification remains crucial, aiding in personalized therapy adjustments that improve patients' outcomes. This review also touches upon the limited treatment options for other PH groups, like PH associated with left heart disease, parenchymal lung diseases, and chronic thromboembolic PH, underscoring the need for expanded therapeutic options. Despite advances, challenges remain: diagnostic delays, misdiagnosis, absence of head-to-head clinical trials, and the timing of introducing newer treatments such as sotatercept are discussed, emphasizing an integrated approach that transcends vasodilation to target underlying disease mechanisms. Future directions envision a comprehensive risk stratification incorporating right ventricular function and a mechanism-based treatment paradigm, encouraging a tailored therapeutic approach in PH management.
Collapse
Affiliation(s)
- Rodolfo A Estrada
- Division of Pulmonary Diseases and Critical Care Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Sandeep Sahay
- Division of Pulmonary, Critical Care & Sleep Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Adriano R Tonelli
- Pulmonary and Critical Care Divisions, Integrated Hospital Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
28
|
Guignabert C. From basic scientific research to the development of new drugs for pulmonary arterial hypertension: insights from activin-targeting agents. Breathe (Sheff) 2025; 21:240116. [PMID: 39845439 PMCID: PMC11747879 DOI: 10.1183/20734735.0116-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/10/2024] [Indexed: 01/24/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disorder of the pulmonary vasculature leading to right ventricular failure. This pulmonary vascular remodelling leads to increased pulmonary vascular resistance and high pulmonary arterial pressures. Despite the development of new therapies, many patients continue to experience significant morbidity and mortality. This review offers a comprehensive overview of the current understanding of PAH pathophysiology, with a focus on key mechanisms that contribute to pulmonary endothelial cell dysfunction and the pathological accumulation of pulmonary artery smooth muscle cells, mesenchymal cells and inflammatory cells in the walls of remodelled small pulmonary vessels, three processes central to the progression of PAH. In particular, it highlights recent developments in targeting the activin signalling pathway, a novel therapeutic approach that shows promise in modulating these pathological processes. The review also addresses the ongoing challenges in translating preclinical findings into effective clinical treatments, emphasising the importance of integrating human data with preclinical models and adopting innovative strategies to bridge the gap between research and clinical practice.
Collapse
Affiliation(s)
- Christophe Guignabert
- Université Paris-Saclay, INSERM UMR_S 999, Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique (HPPIT), Faculté de Médecine, Le Kremlin-Bicêtre, France
| |
Collapse
|
29
|
Fujiwara T, Ishii S, Minatsuki S, Hatano M, Takeda N. Exploring Novel Therapeutics for Pulmonary Arterial Hypertension. Int Heart J 2025; 66:3-12. [PMID: 39894550 DOI: 10.1536/ihj.24-615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by progressive obliteration of pulmonary arteries. Dysregulated bone morphogenetic protein (BMP) signaling pathway contributes to the development of PAH, and pulmonary vasodilators including endothelin receptor antagonists, phosphodiesterase 5 inhibitors, prostaglandins and soluble guanylate cyclase stimulators, dramatically improve the long-term prognosis. However, there still exist refractory patients who require continuous catecholamine support or lung transplantation, and the development of new treatment strategies targeting molecular mechanisms of PAH is highly anticipated. Sotatercept, a first-in-class activin signaling inhibitor, has recently been approved for the treatment of PAH, and it targets and restores an imbalance in activin-growth differentiation factor and BMP pathway signaling. In addition, treatment strategies targeting peroxisome proliferator-activated receptor-γ signaling, inflammatory and immune systems, DNA damage response and cellular senescence, and growth factor receptors including vascular endothelial growth factor and platelet-derived growth factor receptors, are being devised. In this review, we briefly summarize the recent advances in basic research paving the way for the development of more effective treatments for PAH and their potential in clinical therapeutic applications.
Collapse
Affiliation(s)
- Takayuki Fujiwara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
- Department of Computational Diagnostic Radiology and Preventive Medicine, Graduate School of Medicine, The University of Tokyo
- Center for Molecular Medicine, Jichi Medical University
| | - Satoshi Ishii
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Shun Minatsuki
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Masaru Hatano
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
- Advanced Medical Center for Heart Failure, The University of Tokyo Hospital
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| |
Collapse
|
30
|
Jiang D, Wang J, Wang R, Wu Y. Comprehensive Insights into Mechanisms for Ventricular Remodeling in Right Heart Failure. Rev Cardiovasc Med 2024; 25:426. [PMID: 39742244 PMCID: PMC11683703 DOI: 10.31083/j.rcm2512426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 01/03/2025] Open
Abstract
Ventricular remodeling in right heart failure is a complex pathological process involving interactions between multiple mechanisms. Overactivation of the neuro-hormonal pathways, activation of the oxidative stress response, expression of cytokines, apoptosis of cardiomyocytes, and alterations of the extracellular matrix (ECM) are among the major mechanisms involved in the development of ventricular remodeling in right heart failure. These mechanisms are involved in ventricular remodeling, such as myocardial hypertrophy and fibrosis, leading to the deterioration of myocardial systolic and diastolic function. A deeper understanding of these mechanisms can help develop more effective therapeutic strategies in patients with right heart failure (RHF) to improve patient survival and quality of life. Despite the importance of ventricular remodeling in RHF, there are a limited number of studies in this field. This article explores in-depth historical and current information about the specific mechanisms in ventricular remodeling in RHF, providing a theoretical rationale for recognizing its importance in health and disease.
Collapse
Affiliation(s)
- Dongmei Jiang
- Department of General Medicine, First Affiliated Hospital of Xinjiang Medical University, 830011 Urumchi, Xinjiang, China
| | - Jie Wang
- Department of Pharmacy, First Affiliated Hospital of Xinjiang Medical University, 830011 Urumchi, Xinjiang, China
| | - Rui Wang
- Department of General Medicine, First Affiliated Hospital of Xinjiang Medical University, 830011 Urumchi, Xinjiang, China
| | - Yun Wu
- Department of General Medicine, First Affiliated Hospital of Xinjiang Medical University, 830011 Urumchi, Xinjiang, China
| |
Collapse
|
31
|
Madonna R, Biondi F. Sotatercept: New drug on the horizon of pulmonary hypertension. Vascul Pharmacol 2024; 157:107442. [PMID: 39571875 DOI: 10.1016/j.vph.2024.107442] [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: 10/10/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Sotatercept (brand name WINREVAIR, developed by Merck) is an activin receptor type IIA-Fc (ActRIIA-Fc), working by sequestering free activins. Sotatercept restores the balance between the activin proliferative pathway and the bone morphogenic protein (BMP) antiproliferative pathway in the pulmonary arterial cirulation. Sotatercept recently received approval in the USA and in Europe for the treatment of adults with pulmonary arterial hypertension (PAH) Group 1, on top of background PAH therapy to increase exercise capacity, improve WHO functional class and reduce the risk of clinical worsening events. Nevertheless, several studies are ongoing to investigate the potential adverse reactions of the drug especially at the haematological level. We provide an overview of the clinical and preclinical evidence of the targeting the activing pathway through sotatercept on the treatment of PAH. We also discuss what other possibilities there are for the application of sotatercept in the setting of pulmonary hypertension other than PAH Group 1.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy.
| | - Filippo Biondi
- Department of Surgical, Medical and Molecular Pathology and Critical Area, Cardiology Division, University of Pisa, Pisa, Italy
| |
Collapse
|
32
|
Tan JS, Wei Y, Chong L, Yang Y, Hu S, Wang Y. SGLT2 inhibitors as a potential therapeutic option for pulmonary hypertension: mechanisms and clinical perspectives. Crit Rev Clin Lab Sci 2024; 61:709-725. [PMID: 38847284 DOI: 10.1080/10408363.2024.2361012] [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: 04/03/2024] [Revised: 04/29/2024] [Accepted: 05/24/2024] [Indexed: 11/27/2024]
Abstract
Pulmonary arterial hypertension (PAH), one subtype of pulmonary hypertension (PH), is a life-threatening condition characterized by pulmonary arterial remodeling, elevated pulmonary vascular resistance, and blood pressure in the pulmonary arteries, leading to right heart failure and increased mortality. The disease is marked by endothelial dysfunction, vasoconstriction, and vascular remodeling. The role of Sodium-Glucose Co-Transporter-2 (SGLT2) inhibitors, a class of medications originally developed for diabetes management, is increasingly being explored in the context of cardiovascular diseases, including PAH, due to their potential to modulate these pathophysiological processes. In this review, we systematically examine the burgeoning evidence from both basic and clinical studies that describe the effects of SGLT2 inhibitors on cardiovascular health, with a special emphasis on PAH. By delving into the complex interactions between these drugs and the potential pathobiology that underpins PH, this study seeks to uncover the mechanistic underpinnings that could justify the use of SGLT2 inhibitors as a novel therapeutic approach for PAH. We collate findings that illustrate how SGLT2 inhibitors may influence the normal function of pulmonary arteries, possibly alleviating the pathological hallmarks of PAH such as inflammation, oxidative stress, aberrant cellular proliferation, and so on. Our review thereby outlines a potential paradigm shift in PAH management, suggesting that these inhibitors could play a crucial role in modulating the disease's progression by targeting the potential dysfunctions that drive it. This comprehensive synthesis of existing research underscores the imperative need for further clinical trials to validate the efficacy of SGLT2 inhibitors in PAH and to integrate them into the therapeutic agents used against this challenging disease.
Collapse
Affiliation(s)
- Jiang-Shan Tan
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Clinical Research Center of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yixiao Wei
- Peking University Health Science Center, Beijing, China
| | - Lingtao Chong
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanmin Yang
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Clinical Research Center of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Song Hu
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yimeng Wang
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Clinical Research Center of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
Du Q, Zhang C, Qu T, Zhou X, Liu Y, Chen Z, Shen Z, Chen P, Zhang R. Methyltransferase-Like 3-Driven N6-Methyladenosine Modification of Recombination Signal Binding Protein for Immunoglobulin Kappa J Region Promotes Vascular Remodeling in Pulmonary Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2252-2271. [PMID: 39222906 DOI: 10.1016/j.ajpath.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/08/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The dysregulation of N6-methyladenosine (m6A) RNA modification is widely recognized for its crucial roles in various diseases, including pulmonary hypertension (PH). Prior studies have highlighted the significant role of methyltransferase-like 3 (METTL3) in the pathogenesis of PH. Nevertheless, the potential and underlying mechanisms of METTL3 and its inhibitors as targets for PH treatment require further elucidation. In this study, increased levels of METTL3 were observed in various rodent models of PH. In vitro studies revealed that METTL3 silencing or treatment with STM2457, a specific METTL3 inhibitor, attenuated the proliferation and migration of pulmonary artery smooth muscle cells stimulated by platelet-derived growth factor-BB or hypoxia. Moreover, in vivo experiments using adeno-associated virus 9-mediated METTL3 silencing or STM2457 inhibition demonstrated improvement in SU5416/hypoxia-induced PH in mice. Additionally, m6A RNA immunoprecipitation analysis identified recombination signal binding protein for immunoglobulin kappa J region (RBPJ) regulated by METTL3 in rodent models of PH. Loss-of-function studies showed that silencing RBPJ could attenuate the changes in the proliferation and migration of pulmonary artery smooth muscle cells induced by platelet-derived growth factor-BB or hypoxia. Further studies indicated that METTL3 and YTH N6-methyladenosine RNA binding protein F1 (YTHDF1) regulated RBPJ mRNA expression in an m6A-dependent manner. These findings indicated that targeting METTL3 may be a promising therapeutic strategy for treating PH, and modulation of RBPJ could offer a potential intervention mechanism.
Collapse
Affiliation(s)
- Qiang Du
- Department of Respiratory Medicine, Zhongda Hospital of Southeast University, Nanjing, China
| | - Chun Zhang
- Department of Respiratory Medicine, Zhongda Hospital of Southeast University, Nanjing, China
| | - Tianyu Qu
- Department of Respiratory Medicine, Zhongda Hospital of Southeast University, Nanjing, China
| | - Xiao Zhou
- Department of Respiratory Medicine, Zhongda Hospital of Southeast University, Nanjing, China
| | - Yingying Liu
- Department of Respiratory Medicine, Zhongda Hospital of Southeast University, Nanjing, China
| | - Zhixuan Chen
- Department of Respiratory Medicine, Zhongda Hospital of Southeast University, Nanjing, China
| | - Ziling Shen
- Department of Respiratory Medicine, Zhongda Hospital of Southeast University, Nanjing, China
| | - Pingsheng Chen
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Ruifeng Zhang
- Department of Respiratory Medicine, Zhongda Hospital of Southeast University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
34
|
Zhang M, Li H, Ma S, Li X, Xi L, Li Y, Zhang Z, Zhang S, Gao Q, Huang Q, Wan J, Xie W, Li J, Yang P, Zhang Y, Zhai Z. Serum proteome profiling reveals HGFA as a candidate biomarker for pulmonary arterial hypertension. Respir Res 2024; 25:418. [PMID: 39609799 PMCID: PMC11603967 DOI: 10.1186/s12931-024-03036-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 11/09/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Identification and validation of potential biomarkers could facilitate the identification of pulmonary arterial hypertension (PAH) and thus aid to study their roles in the disease process. METHODS We used the isobaric tag for relative and absolute quantitation approaches to compare the protein profiles between the serum of PAH patients and the controls. Bioinformatics analyses and enzyme-linked immunosorbent assay (ELISA) identification of PAH patients and the controls were performed to identify the potential biomarkers. The receiver operating characteristic curve (ROC) analysis was used to evaluate the diagnostic performance of these potential biomarkers. Mendelian randomization (MR) analysis further clarified the relationship between the potential biomarkers and PAH. Additionally, the expression levels of the potential biomarkers were further validated in two PAH animal models (monocrotaline-PH and Sugen5416 plus hypoxia-PH) using ELISA and reverse transcription-quantitative PCR (RT-qPCR). RESULTS We identified significant changes in three proteins including heparanase (HPSE), gelsolin (GSN), and hepatocyte growth factor activator (HGFA) in PAH patients. The ROC analysis showed that the areas under the curve of HPSE, GSN, and HGFA in differentiating PAH patients from controls were 0.769, 0.777, and 0.964, respectively. HGFA was correlated with multiple parameters of right ventricular functions in PAH patients. Besides proteomic analysis, we also used MR method to demonstrate the causal link between genetically reduced HGFA levels and an increased risk of PAH. In subsequent validation study in PAH animal models, the mRNA expression levels of HGFA in the lung tissues were significantly lower in PAH rat models than in controls. In the rat models, serum levels of HGFA were lower compared to the control group and showed a negative correlation with right ventricular systolic pressure. CONCLUSION The study demonstrated that HGFA might be a promising biomarker for noninvasive detection of PAH.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haobo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuangshuang Ma
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Xincheng Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Harbin Medical University, Harbin, China
| | - Linfeng Xi
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Yishan Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhu Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Shuai Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qian Gao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qiang Huang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jun Wan
- Department of Pulmonary and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wanmu Xie
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jifeng Li
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University; Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Beijing Chao-Yang Hospital, Capital Medical University; Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University; Department of Respiratory Disease, Capital Medical University, Beijing, China
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Yunxia Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Zhenguo Zhai
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
35
|
Lin Z, Zhuang J, He L, Zhu S, Kong W, Lu W, Zhang Z. Exploring Smad5: a review to pave the way for a deeper understanding of the pathobiology of common respiratory diseases. Mol Med 2024; 30:225. [PMID: 39578779 PMCID: PMC11585160 DOI: 10.1186/s10020-024-00961-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024] Open
Abstract
Smad5 (small mothers against decapentaplegic 5) protein is a receptor-regulated member of the Smad family proteins, mainly participating in the bone morphogenetic protein (BMP) signaling pathway in its phosphorylated form. This article will provide a detailed review of Smad5, focusing on its gene characteristics, protein structure, and subcellular localization properties. We will also explore the related signaling pathways and the mechanisms of Smad5 in respiratory diseases, including chronic obstructive pulmonary disease (COPD), bronchial asthma, pulmonary arterial hypertension(PAH), lung cancer, and idiopathic pulmonary fibrosis (IPF). Additionally, the review will cover aspects such as proliferation, differentiation, apoptosis, anti-fibrosis, and mitochondrial function metabolism. In addition, the review will cover aspects of proliferation, differentiation, apoptosis, anti-fibrosis and functional mitochondrial metabolism related to the above topics. Numerous studies suggest that Smad5 may play a unique and important role in the pathogenesis of respiratory system diseases. However, in previous research, Smad5 was mainly used to broadly determine the activation of the BMP signaling pathway, and its own function has not been given much attention. It is worth noting that Smad5 has distinct nuclear-cytoplasmic distribution characteristics different from Smad1 and Smad8. It can undergo significant nuclear-cytoplasmic shuttling when intracellular pH (pHi) changes, playing important roles in both the classical BMP signaling pathway and non-BMP signaling pathways. Given that Smad5 can move intracellularly in response to changes in physicochemical properties, its cellular localization may play a crucial role in the development of respiratory diseases. This article will explore the possibility that its distribution characteristics may be an important factor that is easily overlooked and not adequately considered in disease research.
Collapse
Affiliation(s)
- Zeqiang Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiayu Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lixia He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Siyuan Zhu
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weiguo Kong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Zili Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
36
|
Chen S, Bigdon S, Riether C, Ma X, Niu X, Häckel S, Li Z, Gantenbein B. The Role of the Bone Morphogenetic Protein Antagonist Noggin in Nucleus Pulposus Intervertebral Disc Cells. Int J Mol Sci 2024; 25:11803. [PMID: 39519354 PMCID: PMC11546912 DOI: 10.3390/ijms252111803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Low back pain (LBP) is a significant global health issue, contributing to disability and socioeconomic burdens worldwide. The degeneration of the human intervertebral disc (IVD) is a critical factor in the pathogenesis of LBP. Recent studies have emphasized the significance of a specific set of genes and extracellular matrix (ECM) in IVD health. In particular, Noggin has emerged as a critical gene due to its high expression levels in healthy nucleus pulposus cells (NPCs) observed in our previous research. In this study, it was hypothesized that decreased Noggin expression in NPCs is associated with IVD degeneration and contributes to LBP development. A lentivirus-mediated RNAi was applied to knock down Noggin expression in primary NPCs from six human donors. The NPCs after transduction were evaluated through cell viability analysis, XTT assay, and cell apoptosis analyses. After two weeks, a colony formation assay was used to examine the anchor-independent growth ability of transduced cells. At the transcript level, anabolic and catabolic markers were quantified using RT-qPCR. The results demonstrated that lentivirus-mediated downregulation of Noggin significantly inhibited cell proliferation, reduced cell viability, and suppressed colony formation, while inducing apoptosis in human NPCs in vitro. Notably, it disrupted cellular anabolic processes and promoted catabolic activity in human NPCs post-transduction. Our findings indicated that the degeneration of human IVD is possibly related to decreased Noggin expression in NPCs. This research provides valuable insights into the role of Noggin in IVD homeostasis and its implications in LBP treatment.
Collapse
Affiliation(s)
- Shuimu Chen
- Tissue Engineering for Orthopedics & Mechanobiology (TOM), Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, CH-3008 Bern, Switzerland;
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, CH-3012 Bern, Switzerland
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.B.); (S.H.)
| | - Sebastian Bigdon
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.B.); (S.H.)
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland;
- Department for BioMedical Research, University of Bern, CH-3008 Bern, Switzerland; (X.M.); (X.N.)
| | - Xiaochi Ma
- Department for BioMedical Research, University of Bern, CH-3008 Bern, Switzerland; (X.M.); (X.N.)
| | - Xiaoyi Niu
- Department for BioMedical Research, University of Bern, CH-3008 Bern, Switzerland; (X.M.); (X.N.)
| | - Sonja Häckel
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.B.); (S.H.)
- Graduate School for Health Sciences (GSH), University of Bern, CH-3012 Bern, Switzerland
| | - Zhen Li
- AO Research Institute Davos, CH-7270 Davos, Switzerland;
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopedics & Mechanobiology (TOM), Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, CH-3008 Bern, Switzerland;
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.B.); (S.H.)
| |
Collapse
|
37
|
Li Q, Li H, Zhu L, Zhang L, Zheng X, Hao Z. Growth Differentiation Factor 11 Evokes Lung Injury, Inflammation, and Fibrosis in Mice through the Activin A Receptor Type II-Like Kinase, 53kDa-Smad2/3 Signaling Pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2036-2058. [PMID: 39147236 DOI: 10.1016/j.ajpath.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/02/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024]
Abstract
Growth differentiation factor 11 (GDF11) belongs to the transforming growth factor beta superfamily and participates in various pathophysiological processes. Initially, GDF11 was suggested to act as a rejuvenator by improving age-related phenotypes of the heart, brain, and skeletal muscle in aged mice. Recent studies demonstrate that GDF11 also serves as an adverse risk factor for human frailty and diseases. However, the role of GDF11 in pulmonary fibrosis (PF) remains unclear. This study explored the role and signaling mechanisms of GDF11 in PF. GDF11 expression was markedly up-regulated in fibrotic lung tissues of both humans and mice. Intratracheal administration of commercial recombinant GDF11 caused lung injury, inflammation, and fibrogenesis in mice. Furthermore, adenovirus-mediated secretory expression of mature GDF11 was exacerbated, whereas full-length GDF11 or the GDF11 propeptide (GDF111-298) alleviated bleomycin-induced PF in mice. In in vitro experiments, GDF11 suppressed the growth of alveolar and bronchial epithelial cells (A549 and BEAS-2B) and human pulmonary microvascular endothelial cells, promoted fibroblast activation, and induced epithelial/endothelial-mesenchymal transition. These effects corresponded to the phosphorylation of Smad2/3, and blocking activin A receptor type II-like kinase, 53kDa (ALK5)-Smad2/3 signaling abolished the in vivo and in vitro effects of GDF11. In conclusion, these findings provide evidence that GDF11 acts as a potent injurious, proinflammatory, and profibrotic factor in the lungs via the ALK5-Smad2/3 pathway.
Collapse
Affiliation(s)
- Qian Li
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hanchao Li
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Zhu
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lijuan Zhang
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyan Zheng
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhiming Hao
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
38
|
Wang C, Liu X, Hu X, Wu T, Duan R. Therapeutic targeting of GDF11 in muscle atrophy: Insights and strategies. Int J Biol Macromol 2024; 279:135321. [PMID: 39236952 DOI: 10.1016/j.ijbiomac.2024.135321] [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: 02/19/2024] [Revised: 07/29/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
The exploration of novel therapeutic avenues for skeletal muscle atrophy is imperative due to its significant health impact. Recent studies have spotlighted growth differentiation factor 11 (GDF11), a TGFβ superfamily member, for its rejuvenating role in reversing age-related tissue dysfunction. This review synthesizes current findings on GDF11, elucidating its distinct biological functions and the ongoing debates regarding its efficacy in muscle homeostasis. By addressing discrepancies in current research outcomes and its ambiguous role due to its homological identity to myostatin, a negative regulator of muscle mass, this review aims to clarify the role of GDF11 in muscle homeostasis and its potential as a therapeutic target for muscle atrophy. Through a thorough examination of GDF11's mechanisms and effects, this review provides insights that could pave the way for innovative treatments for muscle atrophy, emphasizing the need and strategies to boost endogenous GDF11 levels for therapeutic potential.
Collapse
Affiliation(s)
- Chuanzhi Wang
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Xiaocao Liu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Xilong Hu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Tao Wu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Rui Duan
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| |
Collapse
|
39
|
Stump B, Waxman AB. Pulmonary Arterial Hypertension and TGF-β Superfamily Signaling: Focus on Sotatercept. BioDrugs 2024; 38:743-753. [PMID: 39292393 DOI: 10.1007/s40259-024-00680-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and progressive disease that continues to remain highly morbid despite multiple advances in medical therapies. There remains a persistent and desperate need to identify novel methods of treating and, ideally, reversing the pathologic vasculopathy that results in PAH development and progression. Sotatercept is a first-in-class fusion protein that is believed to primarily inhibit activin signaling resulting in decreased cell proliferation and differentiation, though the exact mechanism remains uncertain. Here, we review the currently available PAH therapies, data highlighting the importance of transforming growth factor-β (TGF-β) superfamily signaling in the development of PAH, and the published and on-going clinical trials evaluating sotatercept in the treatment of PAH. We will also discuss preclinical data supporting the potential use of the fusion protein KER-012 in the inhibition of aberrant TGF-β superfamily signaling to ameliorate the obstructive vasculopathy of PAH.
Collapse
|
40
|
Madonna R, Biondi F. Perspectives on Sotatercept in Pulmonary Arterial Hypertension. J Clin Med 2024; 13:6463. [PMID: 39518603 PMCID: PMC11547004 DOI: 10.3390/jcm13216463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Sotatercept acts as a type IIA-Fc activin receptor, thereby scavenging free activin from its physiological membrane receptor. Through this type of action, sotaterpect leads to a rebalancing of the proliferation and antiproliferation pathways of pulmonary smooth muscle cells in response to bone morphogenic protein (BMP). Although sotatercept has been approved as the fourth pillar of therapy for group 1 pulmonary arterial hypertension (PAH) in the United States and Europe, several studies are ongoing to broaden the application of the drug to non-Group 1 PAH. We provide an overview of the clinical and preclinical evidence of targeting the activation pathway by sotatercept in the treatment of PAH. We also discuss other potential applications of sotatercept in the context of pulmonary hypertension other than PAH group 1.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Cardiology Division, Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56124 Pisa, Italy;
- Cardiology Division, Pisa University Hospital, Via Paradisa, 2, 56124 Pisa, Italy
| | - Filippo Biondi
- Cardiology Division, Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56124 Pisa, Italy;
| |
Collapse
|
41
|
Tsai J, Malik S, Tjen-A-Looi SC. Pulmonary Hypertension: Pharmacological and Non-Pharmacological Therapies. Life (Basel) 2024; 14:1265. [PMID: 39459565 PMCID: PMC11509317 DOI: 10.3390/life14101265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Pulmonary hypertension (PH) is a severe and chronic disease characterized by increased pulmonary vascular resistance and remodeling, often precipitating right-sided heart dysfunction and death. Although the condition is progressive and incurable, current therapies for the disease focus on multiple different drugs and general supportive therapies to manage symptoms and prolong survival, ranging from medications more specific to pulmonary arterial hypertension (PAH) to exercise training. Moreover, there are multiple studies exploring novel experimental drugs and therapies including unique neurostimulation, to help better manage the disease. Here, we provide a narrative review focusing on current PH treatments that target multiple underlying biochemical mechanisms, including imbalances in vasoconstrictor-vasodilator and autonomic nervous system function, inflammation, and bone morphogenic protein (BMP) signaling. We also focus on the potential of novel therapies for managing PH, focusing on multiple types of neurostimulation including acupuncture. Lastly, we also touch upon the disease's different subgroups, clinical presentations and prognosis, diagnostics, demographics, and cost.
Collapse
Affiliation(s)
- Jason Tsai
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| | | | - Stephanie C. Tjen-A-Looi
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| |
Collapse
|
42
|
Steinhauser ML, Maron BA. Viewing Pulmonary Arterial Hypertension Pathogenesis and Opportunities for Disease-Modifying Therapy Through the Lens of Biomass. JACC Basic Transl Sci 2024; 9:1252-1263. [PMID: 39534642 PMCID: PMC11551874 DOI: 10.1016/j.jacbts.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 11/16/2024]
Abstract
Fibroproliferative remodeling of distal pulmonary arterioles is a cornerstone characteristic of pulmonary arterial hypertension (PAH). Data from contemporary quantitative imaging suggest that anabolic synthesis of macromolecular substrate, defined here as biomass, is the proximate event that causes vascular remodeling via pathogenic changes to DNA, collagen, cytoskeleton, and lipid membranes. Modifying biomass is achievable but requires tilting the balance in favor of endogenous degradation over synthetic pathways in order to advance the first-ever disease-modifying PAH pharmacotherapy. Viewing PAH pathobiology through the lens of biomass represents an opportunity to decipher novel determinants of disease inception and inform interventions that induce reverse remodeling.
Collapse
Affiliation(s)
- Matthew L. Steinhauser
- Division of Cardiovascular Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bradley A. Maron
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- University of Maryland-Institute for Health Computing, Bethesda, Maryland, USA
| |
Collapse
|
43
|
Savale L, Tu L, Normand C, Boucly A, Sitbon O, Montani D, Olsson KM, Park DH, Fuge J, Kamp JC, Humbert M, Hoeper MM, Guignabert C. Effect of sotatercept on circulating proteomics in pulmonary arterial hypertension. Eur Respir J 2024; 64:2401483. [PMID: 39227073 PMCID: PMC11525346 DOI: 10.1183/13993003.01483-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024]
Abstract
Alterations in specific signalling pathways within the bone morphogenetic protein/transforming growth factor-β (BMP/TGF-β) family, involving several precisely regulated activator or inhibitor ligands, have been identified as pathogenic drivers of pulmonary arterial hypertension (PAH). These alterations, particularly affecting BMPRII and activin-dependent pathways, have led to innovative therapies, notably the development of sotatercept [1, 2]. Sotatercept, a fusion protein of the extracellular domain of human ACTRIIA and the Fc domain of human IgG1, has shown promising results in improving key clinical, functional, and haemodynamic parameters in PAH patients, as evidenced by positive results in the phase 2 PULSAR and phase 3 STELLAR trials [3, 4]. This progress was partly based on preclinical studies showing that reducing activin-induced Smad2/3 phosphorylation levels, by suppressing activin production in mice [5] or using soluble receptors in rats [6, 7], can attenuate pulmonary vascular remodelling. Despite these advancements, the precise mechanisms of action of these approaches in humans and rodents need to be better understood to enhance these valuable tools. Sotatercept raises several critical questions regarding its mechanism of action, and a deeper understanding could reveal the pathophysiological mechanisms of PAH, leading to more effective therapeutic approaches. Proteomic analysis of circulating biomarkers reveals that sotatercept's impact extends beyond activins to influence BMP-9 and BMP-10, along with essential metabolic and inflammatory factors https://bit.ly/3Z5AZJ3
Collapse
Affiliation(s)
- Laurent Savale
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - Ly Tu
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
| | - Corinne Normand
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
| | - Athénaïs Boucly
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - Olivier Sitbon
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - David Montani
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - Karen M Olsson
- Department for Respiratory Medicine and Infectious Diseases and German Centre of Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Da-Hee Park
- Department for Respiratory Medicine and Infectious Diseases and German Centre of Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Jan Fuge
- Department for Respiratory Medicine and Infectious Diseases and German Centre of Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Jan C Kamp
- Department for Respiratory Medicine and Infectious Diseases and German Centre of Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Marc Humbert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - Marius M Hoeper
- Department for Respiratory Medicine and Infectious Diseases and German Centre of Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Christophe Guignabert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
| |
Collapse
|
44
|
Guignabert C, Aman J, Bonnet S, Dorfmüller P, Olschewski AJ, Pullamsetti S, Rabinovitch M, Schermuly RT, Humbert M, Stenmark KR. Pathology and pathobiology of pulmonary hypertension: current insights and future directions. Eur Respir J 2024; 64:2401095. [PMID: 39209474 PMCID: PMC11533988 DOI: 10.1183/13993003.01095-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 09/04/2024]
Abstract
In recent years, major advances have been made in the understanding of the cellular and molecular mechanisms driving pulmonary vascular remodelling in various forms of pulmonary hypertension, including pulmonary arterial hypertension, pulmonary hypertension associated with left heart disease, pulmonary hypertension associated with chronic lung disease and hypoxia, and chronic thromboembolic pulmonary hypertension. However, the survival rates for these different forms of pulmonary hypertension remain unsatisfactory, underscoring the crucial need to more effectively translate innovative scientific knowledge into healthcare interventions. In these proceedings of the 7th World Symposium on Pulmonary Hypertension, we delve into recent developments in the field of pathology and pathophysiology, prioritising them while questioning their relevance to different subsets of pulmonary hypertension. In addition, we explore how the latest omics and other technological advances can help us better and more rapidly understand the myriad basic mechanisms contributing to the initiation and progression of pulmonary vascular remodelling. Finally, we discuss strategies aimed at improving patient care, optimising drug development, and providing essential support to advance research in this field.
Collapse
Affiliation(s)
- Christophe Guignabert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Sébastien Bonnet
- Pulmonary Hypertension research group, Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Peter Dorfmüller
- Department of Pathology, University Hospital Giessen/Marburg, Giessen, Germany
| | - Andrea J Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Soni Pullamsetti
- Max Planck Institute for Heart and Lung Research Bad Nauheim, Bad Nauheim, Germany
- Department of Internal Medicine, German Center for Lung Research (DZL) Cardio-Pulmonary Institute (CPI)
- Universities of Giessen and Marburg Lung Centre, Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Marlene Rabinovitch
- BASE Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ralph T Schermuly
- Department of Internal Medicine, German Center for Lung Research (DZL) Cardio-Pulmonary Institute (CPI)
| | - Marc Humbert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - Kurt R Stenmark
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado, Denver, CO, USA
| |
Collapse
|
45
|
Corboz MR, Nguyen TL, Stautberg A, Cipolla D, Perkins WR, Chapman RW. Current Overview of the Biology and Pharmacology in Sugen/Hypoxia-Induced Pulmonary Hypertension in Rats. J Aerosol Med Pulm Drug Deliv 2024; 37:241-283. [PMID: 39388691 PMCID: PMC11502635 DOI: 10.1089/jamp.2024.0016] [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: 04/03/2024] [Accepted: 07/03/2024] [Indexed: 10/12/2024] Open
Abstract
The Sugen 5416/hypoxia (Su/Hx) rat model of pulmonary arterial hypertension (PAH) demonstrates most of the distinguishing features of PAH in humans, including increased wall thickness and obstruction of the small pulmonary arteries along with plexiform lesion formation. Recently, significant advancement has been made describing the epidemiology, genomics, biochemistry, physiology, and pharmacology in Su/Hx challenge in rats. For example, there are differences in the overall reactivity to Su/Hx challenge in different rat strains and only female rats respond to estrogen treatments. These conditions are also encountered in human subjects with PAH. Also, there is a good translation in both the biochemical and metabolic pathways in the pulmonary vasculature and right heart between Su/Hx rats and humans, particularly during the transition from the adaptive to the nonadaptive phase of right heart failure. Noninvasive techniques such as echocardiography and magnetic resonance imaging have recently been used to evaluate the progression of the pulmonary vascular and cardiac hemodynamics, which are important parameters to monitor the efficacy of drug treatment over time. From a pharmacological perspective, most of the compounds approved clinically for the treatment of PAH are efficacious in Su/Hx rats. Several compounds that show efficacy in Su/Hx rats have advanced into phase II/phase III studies in humans with positive results. Results from these drug trials, if successful, will provide additional treatment options for patients with PAH and will also further validate the excellent translation that currently exists between Su/Hx rats and the human PAH condition.
Collapse
|
46
|
Boutel M, Dara A, Arvanitaki A, Deuteraiou C, Mytilinaiou M, Dimitroulas T. Towards a Better Prognosis in Patients with Systemic Sclerosis-Related Pulmonary Arterial Hypertension: Recent Developments and Perspectives. J Clin Med 2024; 13:5834. [PMID: 39407897 PMCID: PMC11477739 DOI: 10.3390/jcm13195834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Precapillary pulmonary hypertension (PH) is a significant complication of systemic sclerosis (SSc). It represents one of the leading causes of morbidity and mortality, correlating with a significantly dismal prognosis and quality of life. Despite advancements in the management of patients with pulmonary arterial hypertension associated with SSc (SSc-PAH), no significant improvement has been reported in survival of patients with precapillary SSc-PH associated with extensive lung parenchyma disease. International expert consensus and guidelines for the management of PH recommend annual screening of SSc patients for early detection of pre-capillary PH. The implementation of screening algorithms capable of identifying patients with a high likelihood of developing PH could help limit unnecessary right-heart catheterization procedures and prevent significant delay in diagnosis. Furthermore, early initiation of up-front combination targeted therapy in patients with PAH has shown increase in survival rates, indicating that timely and aggressive medical therapy is key for stabilizing and even improving functional class, hemodynamic parameters and 6 min walking distance (6MWD) in this population. Further research is warranted into the benefit of PAH-targeted therapies in patients with PH associated with lung disease. Lastly, we discuss the potential role of immunosuppression using biologic agents in the therapeutic management of precapillary PH in SSc patients.
Collapse
Affiliation(s)
- Maria Boutel
- Fourth Department of Internal Medicine, Hippokration University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (M.B.); (A.D.); (C.D.); (M.M.); (T.D.)
| | - Athanasia Dara
- Fourth Department of Internal Medicine, Hippokration University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (M.B.); (A.D.); (C.D.); (M.M.); (T.D.)
| | - Alexandra Arvanitaki
- Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’s NHS Foundation Trust, Imperial College, London SW3 6NP, UK
- First Department of Cardiology, AHEPA University Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Cleopatra Deuteraiou
- Fourth Department of Internal Medicine, Hippokration University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (M.B.); (A.D.); (C.D.); (M.M.); (T.D.)
| | - Maria Mytilinaiou
- Fourth Department of Internal Medicine, Hippokration University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (M.B.); (A.D.); (C.D.); (M.M.); (T.D.)
| | - Theodoros Dimitroulas
- Fourth Department of Internal Medicine, Hippokration University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (M.B.); (A.D.); (C.D.); (M.M.); (T.D.)
| |
Collapse
|
47
|
Gomez-Arroyo J, Houweling AC, Bogaard HJ, Aman J, Kitzmiller JA, Porollo A, Dooijes D, Meijboom LJ, Hale P, Pauciulo MW, Hong J, Zhu N, Welch C, Shen Y, Zacharias WJ, McCormack FX, Aldred MA, Weirauch MT, Graf S, Rhodes C, Chung WK, Whitsett JA, Martin LJ, Kalinichenko VV, Nichols WC. Role of Forkhead box F1 in the Pathobiology of Pulmonary Arterial Hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.611448. [PMID: 39345371 PMCID: PMC11429893 DOI: 10.1101/2024.09.18.611448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Rationale Approximately 80% of patients with non-familial pulmonary arterial hypertension (PAH) lack identifiable pathogenic genetic variants. While most genetic studies of PAH have focused on predicted loss-of-function variants, recent approaches have identified ultra-rare missense variants associated with the disease. FOXF1 encodes a highly conserved transcription factor, essential for angiogenesis and vasculogenesis in human and mouse lungs. Objectives We identified a rare FOXF1 missense coding variant in two unrelated probands with PAH. FOXF1 is an evolutionarily conserved transcription factor required for lung vascular development and vascular integrity. Our aims were to determine the frequency of FOXF1 variants in larger PAH cohorts compared to the general population, study FOXF1 expression in explanted lung tissue from PAH patients versus control (failed-donor) lungs, and define potential downstream targets linked to PAH development. Methods Three independent, international, multicenter cohorts were analyzed to evaluate the frequency of FOXF1 rare variants. Various composite prediction models assessed the deleteriousness of individual variants. Bulk RNA sequencing datasets from human explanted lung tissues were compared to failed-donor controls to determine FOXF1 expression. Bioinformatic tools identified putative FOXF1 binding targets, which were orthogonally validated using mouse ChIP-seq datasets. Measurements and Main Results Seven novel or ultra-rare missense coding variants were identified across three patient cohorts in different regions of the FOXF1 gene, including the DNA binding domain. FOXF1 expression was dysregulated in PAH lungs, correlating with disease severity. Histological analysis showed heterogeneous FOXF1 expression, with the lowest levels in phenotypically abnormal endothelial cells within complex vascular lesions in PAH samples. A hybrid bioinformatic approach identified FOXF1 downstream targets potentially involved in PAH pathogenesis, including BMPR2 . Conclusions Large genomic and transcriptomic datasets suggest that decreased FOXF1 expression or predicted dysfunction is associated with PAH.
Collapse
|
48
|
Veisman I, Massey WJ, Goren I, Liu W, Chauhan G, Rieder F. Muscular hyperplasia in Crohn's disease strictures: through thick and thin. Am J Physiol Cell Physiol 2024; 327:C671-C683. [PMID: 38912732 PMCID: PMC11427014 DOI: 10.1152/ajpcell.00307.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Fibrostenosing Crohn's disease (CD) represents a challenging clinical condition characterized by the development of symptomatic strictures within the gastrointestinal tract. Despite therapeutic advancements in managing inflammation, the progression of fibrostenotic complications remains a significant concern, often necessitating surgical intervention. Recent investigations have unveiled the pivotal role of smooth muscle cell hyperplasia in driving luminal narrowing and clinical symptomatology. Drawing parallels to analogous inflammatory conditions affecting other organs, such as the airways and blood vessels, sheds light on common underlying mechanisms of muscular hyperplasia. This review synthesizes current evidence to elucidate the mechanisms underlying smooth muscle cell proliferation in CD-associated strictures, offering insights into potential therapeutic targets. By highlighting the emerging significance of muscle thickening as a novel therapeutic target, this review aims to inform future research endeavors and clinical strategies with the goal to mitigate the burden of fibrostenotic complications in CD and other conditions.
Collapse
Affiliation(s)
- Ido Veisman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - William J Massey
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Idan Goren
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Weiwei Liu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
- Cleveland Clinic Program for Global Translational Inflammatory Bowel Diseases (GRID), Cleveland, Ohio, United States
| |
Collapse
|
49
|
Preston IR, Lewis D, Gomberg-Maitland M. Using Sotatercept in the Care of Patients With Pulmonary Arterial Hypertension. Chest 2024; 166:604-611. [PMID: 39004216 DOI: 10.1016/j.chest.2024.06.3801] [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: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 07/16/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease of the pulmonary microvasculature leading to elevated precapillary pulmonary hypertension. Pulmonary vascular remodeling, a characteristic of PAH, is driven by dysfunctions in the signaling between the pulmonary smooth muscle and endothelial cells with abnormalities that affect cell proliferation and immune dysregulation. Sotatercept, an activin signaling inhibitor, has recently been approved by the US Food and Drug Administration for the treatment of PAH based on two pivotal clinical trials. Evidence-based clinical trials have provided a framework to guide clinicians treating the disease; however, they are not tailored to the individual patient. Often, recommendations from these data are unclear or too general, due to remaining gaps in knowledge. In this edition of "How I Do It," we provide a case-based discussion of common clinical decisions regarding diagnostic testing, choice of first-line agents, escalation of therapy, potential timing of sotatercept, safety awareness, practical use, potential management changes, and the future use of sotatercept in other pulmonary hypertension cohorts.
Collapse
Affiliation(s)
- Ioana R Preston
- Pulmonary Critical Care and Sleep Division, Tufts University School of Medicine, Boston, MA.
| | - Denise Lewis
- Division of Cardiology, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Mardi Gomberg-Maitland
- Division of Cardiology, George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
50
|
Seidinger A, Roberts R, Bai Y, Müller M, Pfeil E, Matthey M, Rieck S, Alenfelder J, König GM, Pfeifer A, Kostenis E, Klinke A, Fleischmann BK, Wenzel D. Pharmacological Gq inhibition induces strong pulmonary vasorelaxation and reverses pulmonary hypertension. EMBO Mol Med 2024; 16:1930-1956. [PMID: 38977926 PMCID: PMC11319782 DOI: 10.1038/s44321-024-00096-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 07/10/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease with limited survival. Herein, we propose the pharmacological inhibition of Gq proteins as a novel concept to counteract pulmonary vasoconstriction and proliferation/migration of pulmonary artery smooth muscle cells (PASMCs) in PAH. We demonstrate that the specific pan-Gq inhibitor FR900359 (FR) induced a strong vasorelaxation in large and small pulmonary arteries in mouse, pig, and human subjects ex vivo. Vasorelaxation by FR proved at least as potent as the currently used triple therapy. We also provide in vivo evidence that local pulmonary application of FR prevented right ventricular systolic pressure increase in healthy mice as well as in mice suffering from hypoxia (Hx)-induced pulmonary hypertension (PH). In addition, we demonstrate that chronic application of FR prevented and also reversed Sugen (Su)Hx-induced PH in mice. We also demonstrate that Gq inhibition reduces proliferation and migration of PASMCs in vitro. Thus, our work illustrates a dominant role of Gq proteins for pulmonary vasoconstriction as well as remodeling and proposes direct Gq inhibition as a powerful pharmacological strategy in PH.
Collapse
Affiliation(s)
- Alexander Seidinger
- Institute of Physiology, Department of Systems Physiology, Medical Faculty, Ruhr University of Bochum, Bochum, Germany
| | - Richard Roberts
- Pharmacology Research Group, University Hospital of Nottingham, Nottingham, UK
| | - Yan Bai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Marion Müller
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr University of Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr University of Bochum, Bad Oeynhausen, Germany
| | - Eva Pfeil
- Molecular-, Cellular-, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Institute of Physiology, Department of Systems Physiology, Medical Faculty, Ruhr University of Bochum, Bochum, Germany
| | - Sarah Rieck
- Institute of Physiology I, Life&Brain Center, Medical Faculty, University of Bonn, Bonn, Germany
| | - Judith Alenfelder
- Molecular-, Cellular-, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Gabriele M König
- Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Evi Kostenis
- Molecular-, Cellular-, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Anna Klinke
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr University of Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr University of Bochum, Bad Oeynhausen, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life&Brain Center, Medical Faculty, University of Bonn, Bonn, Germany
| | - Daniela Wenzel
- Institute of Physiology, Department of Systems Physiology, Medical Faculty, Ruhr University of Bochum, Bochum, Germany.
- Institute of Physiology I, Life&Brain Center, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|