1
|
Yi JS, Perla S, Bennett AM. An Assessment of the Therapeutic Landscape for the Treatment of Heart Disease in the RASopathies. Cardiovasc Drugs Ther 2023; 37:1193-1204. [PMID: 35156148 DOI: 10.1007/s10557-022-07324-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2022] [Indexed: 12/14/2022]
Abstract
The RAS/mitogen-activated protein kinase (MAPK) pathway controls a plethora of developmental and post-developmental processes. It is now clear that mutations in the RAS-MAPK pathway cause developmental diseases collectively referred to as the RASopathies. The RASopathies include Noonan syndrome, Noonan syndrome with multiple lentigines, cardiofaciocutaneous syndrome, neurofibromatosis type 1, and Costello syndrome. RASopathy patients exhibit a wide spectrum of congenital heart defects (CHD), such as valvular abnormalities and hypertrophic cardiomyopathy (HCM). Since the cardiovascular defects are the most serious and recurrent cause of mortality in RASopathy patients, it is critical to understand the pathological signaling mechanisms that drive the disease. Therapies for the treatment of HCM and other RASopathy-associated comorbidities have yet to be fully realized. Recent developments have shown promise for the use of repurposed antineoplastic drugs that target the RAS-MAPK pathway for the treatment of RASopathy-associated HCM. However, given the impact of the RAS-MAPK pathway in post-developmental physiology, establishing safety and evaluating risk when treating children will be paramount. As such insight provided by preclinical and clinical information will be critical. This review will highlight the cardiovascular manifestations caused by the RASopathies and will discuss the emerging therapies for treatment.
Collapse
Affiliation(s)
- Jae-Sung Yi
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA.
- Yale Center for Molecular and Systems Metabolism, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
2
|
Sebastian SA, Padda I, Lehr EJ, Johal G. Aficamten: A Breakthrough Therapy for Symptomatic Obstructive Hypertrophic Cardiomyopathy. Am J Cardiovasc Drugs 2023; 23:519-532. [PMID: 37526885 DOI: 10.1007/s40256-023-00599-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/02/2023]
Abstract
Aficamten is a novel cardiac myosin inhibitor that has demonstrated its ability to safely lower left ventricular outflow tract (LVOT) gradients and improve heart failure symptoms in patients with obstructive hypertrophic cardiomyopathy (HCM). Based on the REDWOOD-HCM open label extension (OLE) study, participants receiving aficamten had significantly reduced resting and Valsalva LVOT gradient within 2 weeks after initiating treatment, with ongoing improvements over 24 weeks, and recent evidence suggests effects can sustain up to 48 weeks. While beta-blockers, calcium channel blockers, and disopyramide have shown some benefits in managing HCM, they have limited direct impact on the underlying disease process in patients with obstructive HCM. Aficamten achieves its therapeutic effect by reducing hypercontractility and improving diastolic function in obstructive HCM. Mavacamten was the first cardiac myosin inhibitor approved for symptomatic obstructive HCM. However, aficamten has a shorter human half-life (t1/2) and fewer drug-drug interactions, making it a preferable treatment option. This review evaluates the long-term clinical value and safety of aficamten in patients with obstructive HCM based on available data from completed and ongoing clinical trials. Additionally, the molecular basis of sarcomere-targeted therapy in reducing LVOT gradients is explored, and its potential in managing obstructive HCM is discussed.
Collapse
Affiliation(s)
- Sneha Annie Sebastian
- Department of Internal Medicine, Azeezia Medical College, Kollam, Kerala, India.
- , Airdrie, Canada.
| | - Inderbir Padda
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, NY, USA
| | - Eric J Lehr
- Division of Cardiac Surgery, Swedish Heart and Vascular Institute, Seattle, WA, USA
| | - Gurpreet Johal
- Department of Cardiology, University of Washington, Valley Medical Center, Seattle, WA, USA
| |
Collapse
|
3
|
Rao SJ, Forst B, Kanwal AK, Kanwal A, Aronow WS, Naidu SS. Cardiac myosin inhibitors for hypertrophic cardiomyopathy: shedding light on their clinical potential. Expert Opin Investig Drugs 2023; 32:1-4. [PMID: 36625220 DOI: 10.1080/13543784.2023.2166825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Shiavax J Rao
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Beani Forst
- Department of Medicine, Creighton University Arizona Health Education Alliance, Phoenix, Az, USA
| | | | - Arjun Kanwal
- Department of Cardiology, Westchester Medical Center, Valhalla, NY, USA
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center and Department of Medicine, New York Medical College, Valhalla, NY, USA
| | - Srihari S Naidu
- Hypertrophic Cardiomyopathy Center, Cardiac Catheterization Laboratory, Department of Cardiology, Westchester Medical Center and Department of Medicine, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
4
|
Forouzandehmehr M, Paci M, Koivumäki JT, Hyttinen J. Altered contractility in mutation-specific hypertrophic cardiomyopathy: A mechano-energetic in silico study with pharmacological insights. Front Physiol 2022; 13:1010786. [PMID: 36388127 PMCID: PMC9659818 DOI: 10.3389/fphys.2022.1010786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/14/2022] [Indexed: 07/25/2023] Open
Abstract
Introduction: Mavacamten (MAVA), Blebbistatin (BLEB), and Omecamtiv mecarbil (OM) are promising drugs directly targeting sarcomere dynamics, with demonstrated efficacy against hypertrophic cardiomyopathy (HCM) in (pre)clinical trials. However, the molecular mechanism affecting cardiac contractility regulation, and the diseased cell mechano-energetics are not fully understood yet. Methods: We present a new metabolite-sensitive computational model of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) electromechanics to investigate the pathology of R403Q HCM mutation and the effect of MAVA, BLEB, and OM on the cell mechano-energetics. Results: We offer a mechano-energetic HCM calibration of the model, capturing the prolonged contractile relaxation due to R403Q mutation (∼33%), without assuming any further modifications such as an additional Ca2+ flux to the thin filaments. The HCM model variant correctly predicts the negligible alteration in ATPase activity in R403Q HCM condition compared to normal hiPSC-CMs. The simulated inotropic effects of MAVA, OM, and BLEB, along with the ATPase activities in the control and HCM model variant agree with in vitro results from different labs. The proposed model recapitulates the tension-Ca2+ relationship and action potential duration change due to 1 µM OM and 5 µM BLEB, consistently with in vitro data. Finally, our model replicates the experimental dose-dependent effect of OM and BLEB on the normalized isometric tension. Conclusion: This work is a step toward deep-phenotyping the mutation-specific HCM pathophysiology, manifesting as altered interfilament kinetics. Accordingly, the modeling efforts lend original insights into the MAVA, BLEB, and OM contributions to a new interfilament balance resulting in a cardioprotective effect.
Collapse
|
5
|
Singh RR, Slater RE, Wang J, Wang C, Guo Q, Motani AS, Hartman JJ, Sadayappan S, Ason BL. Distinct Mechanisms for Increased Cardiac Contraction Through Selective Alteration of Either Myosin or Troponin Activity. JACC Basic Transl Sci 2022; 7:1021-1037. [DOI: 10.1016/j.jacbts.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 10/14/2022]
|
6
|
Morelli C, Ingrasciotta G, Jacoby D, Masri A, Olivotto I. Sarcomere protein modulation: The new frontier in cardiovascular medicine and beyond. Eur J Intern Med 2022; 102:1-7. [PMID: 35534374 DOI: 10.1016/j.ejim.2022.04.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 01/10/2023]
Abstract
Over the past decade, the constant progress in science and technologies has provided innovative drug molecules that address specific disease mechanisms thus opening the era of drugs targeting the underlying pathophysiology of the disease. In this scenario, a new paradigm of modulation has emerged, following the development of small molecules capable of interfering with sarcomere contractile proteins. Potential applications include heart muscle disease and various forms of heart failure, although promising targets also include conditions affecting the skeletal muscle, such as degenerative neuromuscular diseases. In cardiac patients, a cardiac myosin stimulator, omecamtiv mecarbil, has shown efficacy in heart failure with reduced systolic function, lowering heart failure related events or cardiovascular death, while two inhibitors, mavacamten and aficamten, in randomized trials targeting hypertrophic cardiomyopathy, have been shown to reduce hypercontractility and left ventricular outflow obstruction improving functional capacity. Based on years of intensive basic and translational research, these agents are the prototypes of active pipelines promising to deliver an array of molecules in the near future. We here review the available evidence and future perspectives of myosin modulation in cardiovascular medicine.
Collapse
Affiliation(s)
- Cristina Morelli
- Azienda Ospedaliera Universitaria Careggi and University of Florence, Florence, Italy
| | - Gessica Ingrasciotta
- Azienda Ospedaliera Universitaria Careggi and University of Florence, Florence, Italy
| | - Daniel Jacoby
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University, New Haven, CT, USA
| | - Ahmad Masri
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Iacopo Olivotto
- Azienda Ospedaliera Universitaria Careggi and University of Florence, Florence, Italy.
| |
Collapse
|
7
|
Longobardi S, Sher A, Niederer SA. Quantitative mapping of force-pCa curves to whole heart contraction and relaxation. J Physiol 2022; 600:3497-3516. [PMID: 35737959 PMCID: PMC9540007 DOI: 10.1113/jp283352] [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: 05/21/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Abstract The force–pCa (F–pCa) curve is used to characterize steady‐state contractile properties of cardiac muscle cells in different physiological, pathological and pharmacological conditions. This provides a reduced preparation in which to isolate sarcomere mechanisms. However, it is unclear how changes in the F–pCa curve impact emergent whole‐heart mechanics quantitatively. We study the link between sarcomere and whole‐heart function using a multiscale mathematical model of rat biventricular mechanics that describes sarcomere, tissue, anatomy, preload and afterload properties quantitatively. We first map individual cell‐level changes in sarcomere‐regulating parameters to organ‐level changes in the left ventricular function described by pressure–volume loop characteristics (e.g. end‐diastolic and end‐systolic volumes, ejection fraction and isovolumetric relaxation time). We next map changes in the sarcomere‐regulating parameters to changes in the F–pCa curve. We demonstrate that a change in the F–pCa curve can be caused by multiple different changes in sarcomere properties. We demonstrate that changes in sarcomere properties cause non‐linear and, importantly, non‐monotonic changes in left ventricular function. As a result, a change in sarcomere properties yielding changes in the F–pCa curve that improve contractility does not guarantee an improvement in whole‐heart function. Likewise, a desired change in whole‐heart function (i.e. ejection fraction or relaxation time) is not caused by a unique shift in the F–pCa curve. Changes in the F–pCa curve alone cannot be used to predict the impact of a compound on whole‐heart function.
![]() Key points The force–pCa (F–pCa) curve is used to assess myofilament calcium sensitivity after pharmacological modulation and to infer pharmacological effects on whole‐heart function. We demonstrate that there is a non‐unique mapping from changes in F–pCa curves to changes in left ventricular (LV) function. The effect of changes in F–pCa on LV function depend on the state of the heart and could be different for different pathological conditions. Screening of compounds to impact whole‐heart function by F–pCa should be combined with active tension and calcium transient measurements to predict better how changes in muscle function will impact whole‐heart physiology.
Collapse
Affiliation(s)
- Stefano Longobardi
- Cardiac Electromechanics Research Group, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Anna Sher
- Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Steven A Niederer
- Cardiac Electromechanics Research Group, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
8
|
Trujillo AS, Hsu KH, Viswanathan MC, Cammarato A, Bernstein SI. The R369 Myosin Residue within Loop 4 Is Critical for Actin Binding and Muscle Function in Drosophila. Int J Mol Sci 2022; 23:ijms23052533. [PMID: 35269675 PMCID: PMC8910226 DOI: 10.3390/ijms23052533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
The myosin molecular motor interacts with actin filaments in an ATP-dependent manner to yield muscle contraction. Myosin heavy chain residue R369 is located within loop 4 at the actin-tropomyosin interface of myosin's upper 50 kDa subdomain. To probe the importance of R369, we introduced a histidine mutation of that residue into Drosophila myosin and implemented an integrative approach to determine effects at the biochemical, cellular, and whole organism levels. Substituting the similarly charged but bulkier histidine residue reduces maximal actin binding in vitro without affecting myosin ATPase activity. R369H mutants exhibit impaired flight ability that is dominant in heterozygotes and progressive with age in homozygotes. Indirect flight muscle ultrastructure is normal in mutant homozygotes, suggesting that assembly defects or structural deterioration of myofibrils are not causative of reduced flight. Jump ability is also reduced in homozygotes. In contrast to these skeletal muscle defects, R369H mutants show normal heart ultrastructure and function, suggesting that this residue is differentially sensitive to perturbation in different myosin isoforms or muscle types. Overall, our findings indicate that R369 is an actin binding residue that is critical for myosin function in skeletal muscles, and suggest that more severe perturbations at this residue may cause human myopathies through a similar mechanism.
Collapse
Affiliation(s)
- Adriana S. Trujillo
- Department of Biology, Molecular Biology Institute, Heart Institute, San Diego State University, San Diego, CA 92182, USA; (A.S.T.); (K.H.H.)
| | - Karen H. Hsu
- Department of Biology, Molecular Biology Institute, Heart Institute, San Diego State University, San Diego, CA 92182, USA; (A.S.T.); (K.H.H.)
| | - Meera C. Viswanathan
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD 21205, USA; (M.C.V.); (A.C.)
| | - Anthony Cammarato
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD 21205, USA; (M.C.V.); (A.C.)
| | - Sanford I. Bernstein
- Department of Biology, Molecular Biology Institute, Heart Institute, San Diego State University, San Diego, CA 92182, USA; (A.S.T.); (K.H.H.)
- Correspondence:
| |
Collapse
|
9
|
Lewalle A, Campbell KS, Campbell SG, Milburn GN, Niederer SA. Functional and structural differences between skinned and intact muscle preparations. J Gen Physiol 2022; 154:e202112990. [PMID: 35045156 PMCID: PMC8929306 DOI: 10.1085/jgp.202112990] [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: 07/03/2021] [Accepted: 12/16/2021] [Indexed: 11/20/2022] Open
Abstract
Myofilaments and their associated proteins, which together constitute the sarcomeres, provide the molecular-level basis for contractile function in all muscle types. In intact muscle, sarcomere-level contraction is strongly coupled to other cellular subsystems, in particular the sarcolemmal membrane. Skinned muscle preparations (where the sarcolemma has been removed or permeabilized) are an experimental system designed to probe contractile mechanisms independently of the sarcolemma. Over the last few decades, experiments performed using permeabilized preparations have been invaluable for clarifying the understanding of contractile mechanisms in both skeletal and cardiac muscle. Today, the technique is increasingly harnessed for preclinical and/or pharmacological studies that seek to understand how interventions will impact intact muscle contraction. In this context, intrinsic functional and structural differences between skinned and intact muscle pose a major interpretational challenge. This review first surveys measurements that highlight these differences in terms of the sarcomere structure, passive and active tension generation, and calcium dependence. We then highlight the main practical challenges and caveats faced by experimentalists seeking to emulate the physiological conditions of intact muscle. Gaining an awareness of these complexities is essential for putting experiments in due perspective.
Collapse
Affiliation(s)
- Alex Lewalle
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Kenneth S. Campbell
- Department of Physiology and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY
| | - Stuart G. Campbell
- Departments of Biomedical Engineering and Cellular and Molecular Physiology, Yale University, New Haven, CT
| | - Gregory N. Milburn
- Department of Physiology and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY
| | - Steven A. Niederer
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| |
Collapse
|
10
|
Chuang C, Collibee S, Ashcraft L, Wang W, Vander Wal M, Wang X, Hwee DT, Wu Y, Wang J, Chin ER, Cremin P, Zamora J, Hartman J, Schaletzky J, Wehri E, Robertson LA, Malik FI, Morgan BP. Discovery of Aficamten (CK-274), a Next-Generation Cardiac Myosin Inhibitor for the Treatment of Hypertrophic Cardiomyopathy. J Med Chem 2021; 64:14142-14152. [PMID: 34606259 DOI: 10.1021/acs.jmedchem.1c01290] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hypercontractility of the cardiac sarcomere may be essential for the underlying pathological hypertrophy and fibrosis in genetic hypertrophic cardiomyopathies. Aficamten (CK-274) is a novel cardiac myosin inhibitor that was discovered from the optimization of indoline compound 1. The important advancement of the optimization was discovery of an Indane analogue (12) with a less restrictive structure-activity relationship that allowed for the rapid improvement of drug-like properties. Aficamten was designed to provide a predicted human half-life (t1/2) appropriate for once a day (qd) dosing, to reach steady state within two weeks, to have no substantial cytochrome P450 induction or inhibition, and to have a wide therapeutic window in vivo with a clear pharmacokinetic/pharmacodynamic relationship. In a phase I clinical trial, aficamten demonstrated a human t1/2 similar to predictions and was able to reach steady state concentration within the desired two-week window.
Collapse
Affiliation(s)
- Chihyuan Chuang
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Scott Collibee
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Luke Ashcraft
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Wenyue Wang
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Mark Vander Wal
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Xiaolin Wang
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Darren T Hwee
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Yangsong Wu
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Jingying Wang
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Eva R Chin
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Peadar Cremin
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Jeanelle Zamora
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - James Hartman
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Julia Schaletzky
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Eddie Wehri
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Laura A Robertson
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Fady I Malik
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Bradley P Morgan
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| |
Collapse
|
11
|
Lekaditi D, Sakellaropoulos S. Myosin Modulators: The New Era of Medical Therapy for Systolic Heart Failure and Hypertrophic Cardiomyopathy. Cardiol Res 2021; 12:146-148. [PMID: 34046107 PMCID: PMC8139755 DOI: 10.14740/cr1243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/13/2021] [Indexed: 11/11/2022] Open
Abstract
A new uprising pharmacological treatment for systolic heart failure and hypertrophic cardiomyopathy demonstrates very promising results the last years. Myosin modulators have already been tested in numerous studies. Myosin inhibitor (mavacamten) and myosin stimulator, (omecamtiv mecarbil) exhibit their effect by improving clinical outcomes, as well as reducing morbidity and mortality. More studies are however required for further evaluation and particularly effect on sarcomeric level. Side effects of both pharmacological agents have been described and should always be close monitored. Cardiopulmonary exercise test has a pivotal role by means of assessing treatment efficacy.
Collapse
Affiliation(s)
- Dimitra Lekaditi
- Department of Pediatrics, Kantonsspital Aarau, Aarau, Switzerland
| | | |
Collapse
|