1
|
Zong T, Huang X, Zhou W, Hu Z, Jin L, Zhan P, Zhao Y, Sun J, Li G. Advances in the development of phosphodiesterase 5 inhibitors. Eur J Med Chem 2025; 287:117365. [PMID: 39947049 DOI: 10.1016/j.ejmech.2025.117365] [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: 12/19/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/24/2025]
Abstract
Phosphodiesterase 5 (PDE5) can hydrolyze cyclic guanosine monophosphate (cGMP), which is critical for maintaining various physiological processes in organisms. Currently, clinically approved indications for PDE5 inhibitors encompass therapeutic agents for erectile dysfunction (ED), symptoms associated with lower urinary tract symptoms (LUTS), and pulmonary artery hypertension (PAH). Despite the fact that the development of selective PDE5 inhibitors has been a significant focus in drug development for some time following the proven success of sildenafil as a PDE5 inhibitor for ED treatment, fewer than ten drugs in this therapeutic class have been marketed in the past 25 years, often accompanied by adverse effects. Therefore, the development of novel, isozyme-selective PDE5 inhibitors is highly warranted. In this review, we systematically summarize the research progress of PDE5 inhibitors over the past 20 years, focusing on the meticulously combing and categorizing the structures of PDE5 inhibitors and natural products exhibiting PDE5 inhibitory activities, along with their therapeutic potentials. We hope that this summary will aid in better understanding of PDE5 inhibitors and provide insights for developing novel therapies targeting PDE5.
Collapse
Affiliation(s)
- Tieqiang Zong
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin, 133002, China
| | - Xing Huang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, China
| | - Wei Zhou
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin, 133002, China
| | - Zhengyu Hu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin, 133002, China
| | - Long Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin, 133002, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, China
| | - Yuqing Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin, 133002, China.
| | - Jinfeng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin, 133002, China.
| | - Gao Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin, 133002, China.
| |
Collapse
|
2
|
Britto-Júnior J, Lima AT, Oliveira DL, Mariano FV, de Souza VB, Schenka AA, Peterson LW, Slot MS, Fjord R, Wang T, Antunes E, De Nucci G. 6-Cyanodopamine as an endogenous modulator of heart chronotropism and inotropism. Life Sci 2025; 366-367:123504. [PMID: 39983814 DOI: 10.1016/j.lfs.2025.123504] [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: 12/01/2024] [Revised: 01/22/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Rat isolated atria and ventricles release endothelium-derived 6-nitrodopamine, which induces potent positive chronotropic and inotropic responses. 6-Cyanodopamine is released from rabbit isolated atria and ventricles; however, it is not known whether this novel catecholamine has any action on the isolated heart. Therefore, it was investigated whether rat isolated ventricles release 6-cyanodopamine and its action on the rat isolated heart. Basal release of 6-cyanodopamine was assessed by LC-MS/MS method. Tyrosine hydroxylase expression was measured by both immunohistochemistry and fluorescence in situ hybridization (FISH). Chronotropic and inotropic effects were evaluated in isolated atria and Langendorff's preparation, respectively. Rat isolated ventricles presented basal release of 6-cyanodopamine, which was unaffected by pre-treatment with the voltage-gated sodium channel blocker tetrodotoxin. Immunohistochemistry and FISH assays identified tyrosine hydroxylase expression in both endothelium and cardiomyocytes. 6-Cyanodopamine at 10 and 100 pM significantly increased the atrial rate, which was maintained even at 30 min after washing the preparation. In the Langendorff's preparation, 1-min infusion of 6-cyanodopamine (10 and 100 pM) significantly increased heart frequency, left ventricular developed pressure (LVDP), and maximal rate of rise of the left ventricular pressure (dP/dtmax). Bolus injection of noradrenaline at 1 pmol had no effect on any of these parameters; however, in the presence of 6-cyanodopamine (0.01 pM), noradrenaline (1 pmol) significantly increased heart frequency, LVDP, and dP/dt(max). The results indicate that 6-cyanodopamine is a potent endogenous catecholamine mediating both chronotropism and inotropism in the rat isolated heart. 6-Cyanodopamine may have potential therapeutic effect in heart failure and may be useful as a biomarker of cardio-renal diseases.
Collapse
Affiliation(s)
- José Britto-Júnior
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Antonio Tiago Lima
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Denis Lima Oliveira
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Fernanda V Mariano
- Department of Pathology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Valéria Barbosa de Souza
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - André Almeida Schenka
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | | | | - Rebecca Fjord
- Department of Biology, Aarhus University, Aarhus, Denmark
| | - Tobias Wang
- Department of Biology, Aarhus University, Aarhus, Denmark
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (ICB-USP), São Paulo, Brazil
| |
Collapse
|
3
|
Buitrago L, Menezes MR, Larson C, Li J, Kartika T, Banerjee P, Glickman F, Coller B. Unbiased high-throughput screening of drug-repurposing libraries identifies small-molecule inhibitors of clot retraction. Blood Adv 2025; 9:1049-1068. [PMID: 39374578 PMCID: PMC11909436 DOI: 10.1182/bloodadvances.2024013810] [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: 05/29/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024] Open
Abstract
ABSTRACT Platelet clot retraction, the ultimate phase of platelet thrombus formation, is critical for clot stabilization. It requires functional αIIbβ3 receptors, fibrin, and the integrated actions of the actin-myosin contractile and cytoskeletal systems. Disturbances in clot retraction have been associated with both bleeding and thrombosis. We recently demonstrated that platelets treated with the αIIbβ3 antagonist peptide Arg-Gly-Asp-Trp, which eliminates fibrinogen-mediated platelet aggregation, are still able to retract clots. We have exploited this observation to develop an unbiased, functional high-throughput assay to identify small-molecule inhibitors of fibrin-mediated clot retraction adapted for a 384-well plate format. We tested 9710 compounds from drug-repurposing libraries (DRLs). These libraries contain compounds that are either US Food and Drug Administration approved or have undergone preclinical/clinical development. We identified 27 compounds from the Library of Pharmacologically Active Compounds library as inhibitors of clot retraction, of which 14 are known inhibitors of platelet function. From the DRLs, we identified 135 compounds (1.6% hit rate). After extensive curation, these compounds were categorized based on the activity of their reported target. Multiple kinase and phosphodiesterase inhibitors with known antiplatelet effects were identified, along with multiple deubiquitination and receptor inhibitors, as well as compounds that have not previously been reported to have antiplatelet activity. Studies of 1 of the deubiquitination inhibitors (degrasyn) suggest that its effects are downstream of thrombin-induced platelet-fibrinogen interactions and thus may permit the separation of platelet thrombin-induced aggregation-mediated events from clot retraction. Additional studies of the identified compounds may lead to novel mechanisms of inhibiting thrombosis.
Collapse
Affiliation(s)
- Lorena Buitrago
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY
| | - Miriam-Rose Menezes
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY
| | - Chloe Larson
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY
| | - Jihong Li
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY
| | - Thomas Kartika
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY
| | - Priyam Banerjee
- Bio-Imaging Resource Center, The Rockefeller University, New York, NY
| | - Fraser Glickman
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY
| | - Barry Coller
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY
| |
Collapse
|
4
|
O'Donoghue L, Hiebner D, Krishnankutty R, Schoen I, von Kriegsheim A, Smolenski A. Platelet inhibition by hypochlorous acid involves cAMP signalling. Cell Signal 2025; 127:111568. [PMID: 39689749 DOI: 10.1016/j.cellsig.2024.111568] [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: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024]
Abstract
Hypochlorous acid (HOCl), made by neutrophil-derived myeloperoxidase, has been suggested to inhibit platelets, however, the mechanisms involved have not been described. Here we confirm that HOCl exposure changes platelet morphology and inhibits platelet spreading and aggregation. HOCl effects could be reversed by glutathione suggesting a role for cysteine oxidation. Mass spectrometry-based proteomics of HOCl-exposed platelets revealed oxidised cysteine residues in 37 proteins including adenylate cyclase 6 and Rap1B. Adenylate cyclase is involved in the inhibitory cAMP pathway triggered by endothelium-derived prostacyclin and Rap1 is a small G protein required for integrin αIIbβ3 activation and platelet aggregation. We show that HOCl exposure stimulates cAMP production and phosphorylation of the cAMP-dependent protein kinase substrate VASP in platelets and transfected HEK293T cells. In addition, HOCl inhibited Rap1-GTP formation. These data suggest that HOCl inhibits platelets at least in part through the cAMP pathway and by regulating Rap1. Thus, this study provides new insights into HOCl mediated crosstalk between neutrophils and platelets.
Collapse
Affiliation(s)
- Lorna O'Donoghue
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Dublin 4, Belfield, Ireland; Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
| | - Dishon Hiebner
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland; UCD School of Chemical & Bioprocess Engineering, Engineering & Materials Science Centre University College Dublin, Dublin 4, Belfield, Ireland
| | - Roopesh Krishnankutty
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Ingmar Schoen
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
| | - Alex von Kriegsheim
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Albert Smolenski
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Dublin 4, Belfield, Ireland; Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland.
| |
Collapse
|
5
|
Ha SH, Lee YB, Kang HG, Choi KH, Kim BJ, Woo HG, Kwon HS, Song TJ, Kim BJ. Rationale and design of the efficacy and safety of combination of cilostazol and gingko biloba extract EGb 761 in patients with acute non-cardioembolic ischemic stroke (RENEW): A pilot and feasibility randomized controlled trial. J Stroke Cerebrovasc Dis 2025; 34:108105. [PMID: 39528058 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108105] [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: 09/18/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Dual antiplatelet therapy with aspirin and clopidogrel is the standard treatment for acute ischemic stroke (AIS). Cilostazol has emerged as a safe alternative with pleiotropic effects that prevent stroke without increasing the risk of bleeding and has been shown to reduce neurological deterioration (ND) in the acute phase. Ginkgo biloba extract (EGb 761) has also been reported to improve neurological impairment following AIS. This trial aimed to evaluate the efficacy and safety of the combination of cilostazol and EGb 761 in reducing early stroke recurrence and ND in patients with non-cardioembolic AIS. METHODS The RENEW trial is a prospective, randomized, active-controlled, double-blind, parallel, multicenter phase IV study. Five hundred patients with non-cardioembolic AIS presenting within 72 h of symptom onset will be randomized to receive either aspirin 100 mg and cilostazol 200 mg plus EGb 761 160 mg daily or aspirin 100 mg and clopidogrel 75 mg daily for 90 days. The primary outcomes included the combined ND rate during hospitalization and stroke recurrence within 90 days. Secondary outcomes included the rates of ND, recurrent AIS, hemorrhagic stroke, hemorrhagic transformation, functional outcomes (modified Rankin Scale 0-2), bleeding events, and changes in the dizziness handicap inventory scores. DISCUSSION The RENEW trial is expected to provide evidence for the safety and efficacy of combining aspirin, cilostazol, and EGb 761 as an alternative to standard therapy for the acute management of non-cardioembolic AIS. TRIAL REGISTRATION This trial was registered at ClinicalTrials.gov (NCT05445895).
Collapse
Affiliation(s)
- Sang Hee Ha
- Department of Neurology, Gil Medical Center, Gachon University, Incheon, South Korea
| | - Young Bae Lee
- Department of Neurology, Gil Medical Center, Gachon University, Incheon, South Korea
| | - Hyun Goo Kang
- Department of Neurology, Jeonbuk University College of Medicine, Jeonju, South Korea
| | - Kwang-Ho Choi
- Department of Neurology, Chonnam University College of Medicine, Gwang-ju, South Korea
| | - Beom Joon Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Ho Geol Woo
- Department of Neurology, Kyung Hee Medical Center, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Tae-Jin Song
- Department of Neurology, Seoul Hospital, College of Medicine, Ewha Womans University, Seoul, South Korea.
| | - Bum Joon Kim
- Department of Neurology, Asan Medical Center, University of Ulsan, Seoul, South Korea.
| |
Collapse
|
6
|
Cheng X, Ren Q, Zhi J, Chen Q, Luo K, Yu L, Jiao S. Effects of cilostazol on cognitive function and dementia risk: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e40668. [PMID: 39686486 DOI: 10.1097/md.0000000000040668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Cilostazol is an antiplatelet drug and is used for stroke prevention and symptomatic peripheral vascular disease. Studies have reported the effects of cilostazol on cognitive function, but the results are inconsistent and have not been systematically assessed. METHODS We systematically searched the PubMed, Embase, and Cochrane databases for relevant clinical studies. The primary outcome was the change in Mini-mental State Examination (MMSE) scores from baseline to the last available follow-up. The secondary outcome was dementia risk. Mean differences and 95% confidence intervals (CIs) were calculated for combining MMSE scores, and the pooled odds ratios and 95% CIs were used to calculate the association between use of cilostazol and dementia risk. RESULTS Overall, 8 eligible studies met inclusion criteria were pooled in meta-analysis. Though with a trend toward favoring cilostazol, the pooled changes in MMSE scores from baseline showed no significant difference in mild cognitive impairment and dementia patients (mean differences 1.02, 95% CI -0.53 to 2.57, P = .195). For secondary outcome, cilostazol reduced the risk of dementia in patients without prior history of dementia (pooled odds ratios 0.90; 95% CI 0.87 to 0.92; P = .000). CONCLUSION These results suggest the potential for cilostazol treatment in the suppression of cognitive decline and prevention of progression to dementia. However, the lack of blinding in most studies is likely to cause an overestimation of the effect sizes, and further well-designed studies are also needed.
Collapse
Affiliation(s)
- Xiaofang Cheng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Qiuxia Ren
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Jianxia Zhi
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Quanhui Chen
- Department of Gastroenterology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Kaifa Luo
- Department of Nephrology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Liheng Yu
- Department of Cardiology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Shusheng Jiao
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
7
|
Pantazopoulos D, Gouveri E, Rizzo M, Papanas N. Cilostazol for the treatment of distal symmetrical polyneuropathy in diabetes mellitus: Where do we stand? J Diabetes Complications 2024; 38:108905. [PMID: 39522391 DOI: 10.1016/j.jdiacomp.2024.108905] [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: 07/20/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Diabetic Neuropathy (DN) is one of the most frequent chronic complications of diabetes mellitus. Its commonest form, distal symmetrical polyneuropathy (DSPN), is characterised by slowly progressing length-dependent nerve damage in the lower limbs, increasing the risk of foot ulcerations and leading to symptoms like tingling, pain, or numbness. AIM The aim of this review was to discuss the utility of cilostazol, a phosphodiesterase inhibitor with known antiplatelet, vasodilatory, anti-inflammation properties, in the treatment of DSPN. RESULTS Preclinical studies in animals have demonstrated the ability of cilostazol to improve nerve function and to protect from peripheral nerve disruption and central sensitisation. However, clinical trials in humans are very sparse and have so far not been encouraging. CONCLUSIONS Further research is needed to fully understand the mechanisms and potential efficacy of cilostazol in treating DSPN.
Collapse
Affiliation(s)
- Dimitrios Pantazopoulos
- Diabetes Centre-Diabetic Foot Clinic, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| | - Evanthia Gouveri
- Diabetes Centre-Diabetic Foot Clinic, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), School of Medicine, University of Palermo, Palermo, Italy
| | - Nikolaos Papanas
- Diabetes Centre-Diabetic Foot Clinic, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece.
| |
Collapse
|
8
|
Nemr MTM, Abdelaziz MA, Teleb M, Elmasry AE, Elshaier YAAM. An overview on pharmaceutical applications of phosphodiesterase enzyme 5 (PDE5) inhibitors. Mol Divers 2024:10.1007/s11030-024-11016-2. [PMID: 39592536 DOI: 10.1007/s11030-024-11016-2] [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: 08/24/2024] [Accepted: 10/12/2024] [Indexed: 11/28/2024]
Abstract
Phosphodiesterase enzyme 5 (PDE5) inhibitors have emerged as one of the leading molecules for the treatment of erectile dysfunction (ED). PDE5 inhibitors are categorized structurally into several classes. PDE5 inhibitors have been a multidisciplinary endeavor that attracts the attention of researchers because of their multiple pharmaceutical applications. Beyond their action on ED, PDE5 inhibitors are widely used in treatment of benign prostatic hypertrophy (BPH), Eisenmenger's syndrome, Raynaud's Disease, Intrauterine growth retardation (IUGR), Mountain sickness, Bladder pain syndrome/interstitial cystitis (BPS/IC), pulmonary arterial hypertension and type II diabetes (insulin resistance). In addition, PDE5 inhibitors also show promising antiproliferative activity, anti-Alzheimer and COX-1/COX-2 inhibitory activity (anti-inflammatory). Pharmacokinetics, Pharmacogenetics and toxicity of PDE5 inhibitors were finally explored. The diverse therapeutic applications, the high feasibility of structural modification and the appropriate pharmacokinetic properties of PDE5 inhibitors have motivated researchers to develop new scaffolds that have been either under clinical trials or approved by FDA and utilize them to overcome some recent global concerns, such as COVID-19.
Collapse
Affiliation(s)
- Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini Street 11562, Cairo, Egypt.
| | | | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Alamein City, 5060310, Egypt
| | - Ahmed E Elmasry
- Organic & Medicinal Chemistry Department, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt
| | - Yaseen A A M Elshaier
- Organic & Medicinal Chemistry Department, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt.
| |
Collapse
|
9
|
Paronetto MP, Crescioli C. Rethinking of phosphodiesterase 5 inhibition: the old, the new and the perspective in human health. Front Endocrinol (Lausanne) 2024; 15:1461642. [PMID: 39355618 PMCID: PMC11442314 DOI: 10.3389/fendo.2024.1461642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/28/2024] [Indexed: 10/03/2024] Open
Abstract
The phosphodiesterases type 5 (PDE5) are catalytic enzymes converting the second messenger cyclic guanosine monophosphate (cGMP) to 5' GMP. While intracellular cGMP reduction is associated with several detrimental effects, cGMP stabilization associates with numerous benefits. The PDE5 specific inhibitors, PDE5i, found their explosive fortune as first-line treatment for erectile dysfunction (ED), due to their powerful vasoactive properties. The favorable effect for ED emerged as side-effect when PDE5i were originally proposed for coronary artery disease (CAD). From that point on, the use of PDE5i captured the attention of researchers, clinicians, and companies. Indeed, PDE5-induced intracellular cGMP stabilization offers a range of therapeutic opportunities associated not only with vasoactive effects, but also with immune regulatory/anti-inflammatory actions. Chronic inflammation is acknowledged as the common link underlying most non-communicable diseases, including metabolic and cardiac diseases, autoimmune and neurodegenerative disorders, cancer. In this scenario, the clinical exploitation of PDE5i is undeniably beyond ED, representing a potential therapeutic tool in several human diseases. This review aims to overview the biological actions exerted by PDE5i, focusing on their ability as modulators of inflammation-related human diseases, with particular attention to inflammatory-related disorders, like cardiac diseases and cancer.
Collapse
Affiliation(s)
- Maria Paola Paronetto
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Rome, Italy
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Clara Crescioli
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Rome, Italy
| |
Collapse
|
10
|
Martinez Bravo G, Annarapu G, Carmona E, Nawarskas J, Clark R, Novelli E, Mota Alvidrez RI. Platelets in Thrombosis and Atherosclerosis: A Double-Edged Sword. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1608-1621. [PMID: 38885926 PMCID: PMC11373056 DOI: 10.1016/j.ajpath.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
This review focuses on the dual role of platelets in atherosclerosis and thrombosis, exploring their involvement in inflammation, angiogenesis, and plaque formation, as well as their hemostatic and prothrombotic functions. Beyond their thrombotic functions, platelets engage in complex interactions with diverse cell types, influencing disease resolution and progression. The contribution of platelet degranulation helps in the formation of atheromatous plaque, whereas the reciprocal interaction with monocytes adds complexity. Alterations in platelet membrane receptors and signaling cascades contribute to advanced atherosclerosis, culminating in atherothrombotic events. Understanding these multifaceted roles of platelets will lead to the development of targeted antiplatelet strategies for effective cardiovascular disease prevention and treatment. Understanding platelet functions in atherosclerosis and atherothrombosis at different stages of disease will be critical for designing targeted treatments and medications to prevent or cure the disease Through this understanding, platelets can be targeted at specific times in the atherosclerosis process, possibly preventing the development of atherothrombosis.
Collapse
Affiliation(s)
| | - Gowtham Annarapu
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Emely Carmona
- School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - James Nawarskas
- Pharmaceutical Sciences-Pharmacy Practice, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | - Ross Clark
- Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico; Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico
| | - Enrico Novelli
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania; School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Roberto I Mota Alvidrez
- Biomedical Engineering Department, University of New Mexico, Albuquerque, New Mexico; Pharmaceutical Sciences-Pharmacy Practice, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico; Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico.
| |
Collapse
|
11
|
Ismail EA, El-Sakka AI. An overview of conventional and investigational phosphodiesterase 5 inhibitors for treating erectile dysfunction and other conditions. Expert Opin Investig Drugs 2024; 33:925-938. [PMID: 39096237 DOI: 10.1080/13543784.2024.2388569] [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: 07/01/2023] [Revised: 07/14/2024] [Accepted: 08/01/2024] [Indexed: 08/05/2024]
Abstract
INTRODUCTION There is a rising concern about developing innovative, efficacious PDE5I molecules that provide better safety, efficacy, and tolerability with less adverse effects. Innovative PDE5I with dual targets have also been defined in the literature. Additionally, some of PDE5I are able to selectively inhibit other enzymes such as histone deacetylase, acetylcholine esterase, and cyclooxygenase or act as nitric oxide donors. This review presents knowledge concerning the advanced trends and perspectives in using PDE5I in treatment of ED and other conditions. AREAS COVERED Pre-clinical and early clinical trials that investigated the safety, efficacy, and tolerability of novel PDE5I such as Udenafil, Mirodenafil, Lodenafil, Youkenafil, Celecoxib, and TPN729 in treatment of ED and other conditions. EXPERT OPINION Preclinical and limited early clinical studies of the new molecules of PDE5I have demonstrated encouraging results; however, safety, efficacy, and tolerability are still issues that necessitate further long-term multicenter clinical studies to ensure justification of their uses in treatment of ED and other conditions. Progress in molecular delivery techniques and tailored patient-specific management and additional therapeutic technology will dramatically improve care for ED and other conditions. The dream of ED and many other conditions becoming more effectively managed may be feasible in the near future.
Collapse
Affiliation(s)
- Ezzat A Ismail
- Department of Urology, Suez Canal University, Ismailia, Egypt
| | | |
Collapse
|
12
|
Kang MG, Ahn JH, Hwang JY, Hwang SJ, Koh JS, Park Y, Bae JS, Chun KJ, Kim JS, Kim JH, Chon MK. Long-acting cilostazol versus isosorbide mononitrate for patients with vasospastic angina: a randomized controlled trial. Coron Artery Dis 2024; 35:459-464. [PMID: 38595079 DOI: 10.1097/mca.0000000000001366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
BACKGROUND Cilostazol has a vasodilatory function that may be beneficial for patients with vasospastic angina (VSA). We conducted a randomized, open-label, controlled trial to compare the efficacy and safety of long-acting cilostazol and isosorbide mononitrate (ISMN) for VSA. METHODS The study included patients with confirmed VSA between September 2019 and May 2021. Participants were randomly assigned to receive long-acting cilostazol (test group, 200 mg once daily) or conventional ISMN therapy (control group, 20 mg twice daily) for 4 weeks. The clinical efficacy and safety were evaluated using weekly questionnaires. RESULTS Forty patients were enrolled in the study (long-acting cilostazol, n = 20; ISMN, n = 20). Baseline characteristics were balanced between the two groups. Long acting cilostazol showed better angina symptom control within the first week compared to ISMN [reduction of pain intensity score, 6.0 (4.0-8.0) vs. 4.0 (1.0-5.0), P = 0.005; frequency of angina symptom, 0 (0-2.0) vs. 2.0 (0-3.0), P = 0.027, respectively]. The rate of neurological adverse reactions was lower in the cilostazol group than in the ISMN group (headache or dizziness, 40 vs. 85%, P = 0.009; headache, 30 vs. 70%, P = 0.027). CONCLUSION Long-acting cilostazol provided comparable control of angina and fewer adverse neurologic reactions within 4 weeks compared to ISMN. Long-acting cilostazol provides more intensive control of angina within 1 week, suggesting that it may be an initial choice for the treatment of VSA.
Collapse
Affiliation(s)
- Min Gyu Kang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju
| | - Jong-Hwa Ahn
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Cardiovascular Center, Gyeongsang National University Changwon Hospital, Changwon
| | - Jin-Yong Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju
| | - Seok-Jae Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju
| | - Jin-Sin Koh
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju
| | - Yongwhi Park
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Cardiovascular Center, Gyeongsang National University Changwon Hospital, Changwon
| | - Jae Seok Bae
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Cardiovascular Center, Gyeongsang National University Changwon Hospital, Changwon
| | - Kook Jin Chun
- Department of Internal Medicine, Pusan National University School of Medicine and Cardiology, Cardiovascular Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Jeong Su Kim
- Department of Internal Medicine, Pusan National University School of Medicine and Cardiology, Cardiovascular Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - June Hong Kim
- Department of Internal Medicine, Pusan National University School of Medicine and Cardiology, Cardiovascular Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Min Ku Chon
- Department of Internal Medicine, Pusan National University School of Medicine and Cardiology, Cardiovascular Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| |
Collapse
|
13
|
Laudani C, Capodanno D, Angiolillo DJ. The pharmacology of antiplatelet agents for primary, secondary, and tertiary prevention of ischemic stroke. Expert Opin Pharmacother 2024; 25:1373-1390. [PMID: 39046451 DOI: 10.1080/14656566.2024.2385135] [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: 06/03/2024] [Revised: 07/06/2024] [Accepted: 07/23/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Ischemic etiology accounts for two thirds of all strokes in which platelet activation and aggregation play a major role. A variety of antiplatelet therapies have been tested for primary, secondary, and tertiary prevention, with certain patient subtypes benefiting more than others from a specific regimen. AREAS COVERED This review aims at synthetizing current evidence on pharmacology of antiplatelet agents approved for primary, secondary, and tertiary stroke prevention and their application among possible patient subtypes that may benefit more from their administration. EXPERT OPINION Management of ischemic stroke has largely evolved over the past decades. A better understanding of stroke pathophysiology has allowed to identify patients who can benefit most from antiplatelet therapies, with varying degrees of benefit depending on whether these agents are being used for primary, secondary, or tertiary prevention. Importantly, the antiplatelet treatment regimens currently available have expanded and no longer limited to aspirin but include other drugs such as P2Y12 and phosphodiesterase inhibitors, also used in combination, as well as precision medicine approaches using genetic testing aiming at optimizing the safety and efficacy in this population.
Collapse
Affiliation(s)
- Claudio Laudani
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "Rodolico - San Marco", University of Catania, Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "Rodolico - San Marco", University of Catania, Catania, Italy
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| |
Collapse
|
14
|
Khalil JS, Law R, Raslan Z, Cheah LT, Hindle MS, Aburima AA, Kearney MT, Naseem KM. Protein Kinase A Regulates Platelet Phosphodiesterase 3A through an A-Kinase Anchoring Protein Dependent Manner. Cells 2024; 13:1104. [PMID: 38994957 PMCID: PMC11240354 DOI: 10.3390/cells13131104] [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: 05/20/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
Platelet activation is critical for haemostasis, but if unregulated can lead to pathological thrombosis. Endogenous platelet inhibitory mechanisms are mediated by prostacyclin (PGI2)-stimulated cAMP signalling, which is regulated by phosphodiesterase 3A (PDE3A). However, spatiotemporal regulation of PDE3A activity in platelets is unknown. Here, we report that platelets possess multiple PDE3A isoforms with seemingly identical molecular weights (100 kDa). One isoform contained a unique N-terminal sequence that corresponded to PDE3A1 in nucleated cells but with negligible contribution to overall PDE3A activity. The predominant cytosolic PDE3A isoform did not possess the unique N-terminal sequence and accounted for >99% of basal PDE3A activity. PGI2 treatment induced a dose and time-dependent increase in PDE3A phosphorylation which was PKA-dependent and associated with an increase in phosphodiesterase enzymatic activity. The effects of PGI2 on PDE3A were modulated by A-kinase anchoring protein (AKAP) disruptor peptides, suggesting an AKAP-mediated PDE3A signalosome. We identified AKAP7, AKAP9, AKAP12, AKAP13, and moesin expressed in platelets but focussed on AKAP7 as a potential PDE3A binding partner. Using a combination of immunoprecipitation, proximity ligation techniques, and activity assays, we identified a novel PDE3A/PKA RII/AKAP7 signalosome in platelets that integrates propagation and termination of cAMP signalling through coupling of PKA and PDE3A.
Collapse
Affiliation(s)
- Jawad S. Khalil
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK; (J.S.K.); (Z.R.); (L.T.C.); (M.S.H.); (M.T.K.)
| | - Robert Law
- Hull York Medical School, University of Hull, Hull HU6 7EL, UK; (R.L.); (A.A.A.)
| | - Zaher Raslan
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK; (J.S.K.); (Z.R.); (L.T.C.); (M.S.H.); (M.T.K.)
| | - Lih T. Cheah
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK; (J.S.K.); (Z.R.); (L.T.C.); (M.S.H.); (M.T.K.)
| | - Matthew S. Hindle
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK; (J.S.K.); (Z.R.); (L.T.C.); (M.S.H.); (M.T.K.)
| | - Ahmed A. Aburima
- Hull York Medical School, University of Hull, Hull HU6 7EL, UK; (R.L.); (A.A.A.)
| | - Mark T. Kearney
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK; (J.S.K.); (Z.R.); (L.T.C.); (M.S.H.); (M.T.K.)
| | - Khalid M. Naseem
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK; (J.S.K.); (Z.R.); (L.T.C.); (M.S.H.); (M.T.K.)
| |
Collapse
|
15
|
Maded ZK, Sfar S, Taqa GAA, Lassoued MA, Ben Hadj Ayed O, Fawzi HA. Development and Optimization of Dipyridamole- and Roflumilast-Loaded Nanoemulsion and Nanoemulgel for Enhanced Skin Permeation: Formulation, Characterization, and In Vitro Assessment. Pharmaceuticals (Basel) 2024; 17:803. [PMID: 38931470 PMCID: PMC11207013 DOI: 10.3390/ph17060803] [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: 05/20/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
This study explores developing and optimizing a nanoemulsion (NE) system loaded with dipyridamole and roflumilast, aiming to improve skin penetration and retention. The NE formulation was further transformed into a nanoemulgel to enhance its application as a topical treatment for psoriasis. Solubility studies were conducted to select the oil, surfactant, and co-surfactant. Phase diagrams were constructed using the aqueous phase titration method. All the formulations were in nanoscale, and Formula (F2) (which contains oleic acid oil as the oil phase, a mixture of Surfactant Tween 80 and co-surfactant (ethanol) at a ratio of 1:2 in addition to distilled water as an aqueous phase in a ratio of 1:5:4, respectively) was the selected formula depending on the particle size, PDI, and zeta potential. Formula (F2) has the best ratio because it gives the smallest nanoemulsion globule size (particle size average of 167.1 nm), the best homogenicity (lowest PDI of 0.195), and the highest stability (higher zeta potential of -32.22). The selected formula was converted into a nanoemulgel by the addition of 0.5% (w/w) xanthan gum (average particle size of 172.7 nm) and the best homogenicity (lowest PDI of 0.121%) and highest stability (higher zeta potential of -28.31). In conclusion, the selected formula has accepted physical and chemical properties, which enhanced skin penetration.
Collapse
Affiliation(s)
- Zeyad Khalaf Maded
- Laboratory of Pharmaceutical, Chemical, and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia; (Z.K.M.); (M.A.L.); (O.B.H.A.)
| | - Souad Sfar
- Laboratory of Chemical, Galenic and Pharmacological Development of Medicines (LR12ES09), Faculty of Pharmacy of Monastir, University of Monastir, Monastir 5000, Tunisia;
| | - Ghada Abd Alrhman Taqa
- Department of Dental Basic Sciences, College of Dentistry, University of Mosul, Mosul 41002, Iraq;
| | - Mohamed Ali Lassoued
- Laboratory of Pharmaceutical, Chemical, and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia; (Z.K.M.); (M.A.L.); (O.B.H.A.)
| | - Olfa Ben Hadj Ayed
- Laboratory of Pharmaceutical, Chemical, and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia; (Z.K.M.); (M.A.L.); (O.B.H.A.)
| | - Hayder Adnan Fawzi
- Department of Pharmacy, Al Mustafa University College, Baghdad 10064, Iraq
| |
Collapse
|
16
|
Mollick T, Darekar S, Dalarun B, Plastino F, Zhang J, Fernández AP, Alkasalias T, André H, Laín S. Retinoblastoma vulnerability to combined de novo and salvage pyrimidine ribonucleotide synthesis pharmacologic blockage. Heliyon 2024; 10:e23831. [PMID: 38332874 PMCID: PMC10851301 DOI: 10.1016/j.heliyon.2023.e23831] [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: 05/12/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 02/10/2024] Open
Abstract
Retinoblastoma is an eye cancer that commonly affects young children. Despite significant advances, current treatments cause side effects even when administered locally, and patients may still have to undergo enucleation. This is particularly disheartening in cases of bilateral retinoblastoma. Hence, there is an urgent need for novel therapeutic strategies. Inhibitors of the enzyme dihydroorotate dehydrogenase (DHODH), which is involved in the de novo pyrimidine ribonucleotide synthesis pathway, have proven to be effective in preclinical trials against several cancers including pediatric cancers. Here we tested whether blocking pyrimidine ribonucleotide synthesis promotes retinoblastoma cell death. Cultured retinoblastoma cell lines were treated with small molecule inhibitors of DHODH alone or in combination with inhibitors of nucleoside uptake to also block the salvage pathway for pyrimidine ribonucleotide formation. On their own, DHODH inhibitors had a moderate killing effect. However, the combination with nucleoside uptake inhibitors greatly enhanced the effect of DHODH inhibition. In addition, we observed that pyrimidine ribonucleotide synthesis blockage can cause cell death in a p53 mutant retinoblastoma cell line derived from a patient with metastasis. Explaining these results, the analysis of a published patient cohort revealed that loss of chr16q22.2 (containing the DHODH gene) is amongst the most frequent alterations in retinoblastoma and that these tumors often show gains in chromosome regions expressing pyrimidine ribonucleotide salvage factors. Furthermore, these genome alterations associate with malignancy. These results indicate that targeting pyrimidine ribonucleotide synthesis may be an effective therapeutic strategy to consider as a treatment for retinoblastoma.
Collapse
Affiliation(s)
- Tanzina Mollick
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165, Solna, Stockholm, Sweden
| | - Suhas Darekar
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165, Solna, Stockholm, Sweden
| | - Basile Dalarun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165, Solna, Stockholm, Sweden
| | - Flavia Plastino
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Juan Zhang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165, Solna, Stockholm, Sweden
| | - Andres Pastor Fernández
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165, Solna, Stockholm, Sweden
| | - Twana Alkasalias
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165, Solna, Stockholm, Sweden
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Sonia Laín
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165, Solna, Stockholm, Sweden
| |
Collapse
|
17
|
Stanger L, Yamaguchi A, Holinstat M. Antiplatelet strategies: past, present, and future. J Thromb Haemost 2023; 21:3317-3328. [PMID: 38000851 PMCID: PMC10683860 DOI: 10.1016/j.jtha.2023.09.013] [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: 09/01/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 11/26/2023]
Abstract
Antiplatelet therapy plays a critical role in the prevention and treatment of major cardiovascular diseases triggered by thrombosis. Since the 1900s, significant progress in reducing morbidity and death caused by cardiovascular diseases has been made. However, despite the development and approval of drugs that specifically target the platelet, including inhibitors for cycloxygenase-1, P2Y12 receptor, integrin αIIbβ3, phosphodiesterases, and protease-activated receptor 1, the risk of recurrent thrombotic events remains high, and the increased risk of bleeding is a major concern. Scientific advances in our understanding of the role of platelets in haemostasis and thrombosis have revealed novel targets, such as protease-activated receptor 4 (PAR4), glycoprotein Ib (GPIb)-V-IX complex, glycoprotein VI, and 12-lipoxygenase. The antithrombotic effects and safety of the pharmacologic inhibition of these targets are currently under investigation in clinical studies. This review provides an overview of drugs in early development to target the platelet and those in current use in clinical practice. Furthermore, it describes the emerging drug targets being developed and studied to reduce platelet activity and outlines potential novel therapeutic targets in the platelet.
Collapse
Affiliation(s)
- Livia Stanger
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Adriana Yamaguchi
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Surgery, Division of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
18
|
Puertas-Umbert L, Alonso J, Hove-Madsen L, Martínez-González J, Rodríguez C. PDE4 Phosphodiesterases in Cardiovascular Diseases: Key Pathophysiological Players and Potential Therapeutic Targets. Int J Mol Sci 2023; 24:17017. [PMID: 38069339 PMCID: PMC10707411 DOI: 10.3390/ijms242317017] [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: 11/09/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
3',5'-cyclic adenosine monophosphate (cAMP) is a second messenger critically involved in the control of a myriad of processes with significant implications for vascular and cardiac cell function. The temporal and spatial compartmentalization of cAMP is governed by the activity of phosphodiesterases (PDEs), a superfamily of enzymes responsible for the hydrolysis of cyclic nucleotides. Through the fine-tuning of cAMP signaling, PDE4 enzymes could play an important role in cardiac hypertrophy and arrhythmogenesis, while it decisively influences vascular homeostasis through the control of vascular smooth muscle cell proliferation, migration, differentiation and contraction, as well as regulating endothelial permeability, angiogenesis, monocyte/macrophage activation and cardiomyocyte function. This review summarizes the current knowledge and recent advances in understanding the contribution of the PDE4 subfamily to cardiovascular function and underscores the intricate challenges associated with targeting PDE4 enzymes as a therapeutic strategy for the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Judith Alonso
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - Leif Hove-Madsen
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - José Martínez-González
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - Cristina Rodríguez
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| |
Collapse
|
19
|
Reimann MJ, Faisst DN, Knold M, Meurs KM, Stern JA, Cremer SE, Møller JE, Ljungvall I, Häggström J, Olsen LH. No impact of polymorphism in the phosphodiesterase 5A gene in Cavalier King Charles Spaniels on pimobendan-induced inhibition of platelet aggregation response. J Vet Intern Med 2023; 37:2145-2156. [PMID: 37743723 PMCID: PMC10658480 DOI: 10.1111/jvim.16871] [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: 03/30/2023] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND A variant in the canine phosphodiesterase (PDE) 5A gene (PDE5A:E90K) is associated with decreased concentrations of circulating cyclic guanosine monophosphate (cGMP) and response to PDE5 inhibitor treatment. Pimobendan is a PDE inhibitor recommended for medical treatment of certain stages of myxomatous mitral valve disease (MMVD) in dogs. HYPOTHESIS PDE5A:E90K polymorphism attenuates the inhibitory effect of pimobendan on in vitro platelet aggregation and increases basal platelet aggregation in Cavalier King Charles Spaniels (CKCS). Selected clinical variables (MMVD severity, sex, age, hematocrit, platelet count in platelet-rich plasma [PRP], and echocardiographic left ventricular fractional shortening [LV FS]) will not show an association with results. ANIMALS Fifty-two privately owned CKCS with no or preclinical MMVD. METHODS Using blood samples, we prospectively assessed PDE5A genotype using Sanger sequencing and adenosine diphosphate-induced platelet aggregation response (area under the curve [AUC], maximal aggregation [MaxA], and velocity [Vel]) with and without pimobendan using light transmission aggregometry. Dogs also underwent echocardiography. RESULTS Pimobendan inhibited platelet function as measured by AUC, MaxA, and Vel at a concentration of 10 μM (P < .0001) and Vel at 0.03 μM (P < .001). PDE5A:E90K polymorphism did not influence the inhibitory effect of pimobendan or basal platelet aggregation response. CONCLUSIONS AND CLINICAL IMPORTANCE The PDE5A:E90K polymorphism did not influence in vitro basal platelet aggregation response or the inhibitory effect of pimobendan on platelet aggregation in CKCS. Dogs with the PDE5A:E90K polymorphism did not appear to have altered platelet function or response to pimobendan treatment.
Collapse
Affiliation(s)
- Maria J. Reimann
- Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksbergDenmark
| | - Daniel N. Faisst
- Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksbergDenmark
| | - Mads Knold
- Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksbergDenmark
| | - Kathryn M. Meurs
- Department of Clinical SciencesNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Joshua A. Stern
- Department of Medicine and Epidemiology, School of Veterinary MedicineUniversity of California‐DavisDavisCaliforniaUSA
| | - Signe E. Cremer
- Department of Veterinary Clinical SciencesUniversity of CopenhagenFrederiksbergDenmark
| | - Jacob E. Møller
- Department of CardiologyCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | - Ingrid Ljungvall
- Department of Clinical SciencesSwedish University of Agricultural SciencesUppsalaSweden
| | - Jens Häggström
- Department of Clinical SciencesSwedish University of Agricultural SciencesUppsalaSweden
| | - Lisbeth H. Olsen
- Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksbergDenmark
| |
Collapse
|
20
|
Beura SK, Dhapola R, Panigrahi AR, Yadav P, Kumar R, Reddy DH, Singh SK. Antiplatelet drugs: Potential therapeutic options for the management of neurodegenerative diseases. Med Res Rev 2023; 43:1835-1877. [PMID: 37132460 DOI: 10.1002/med.21965] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/13/2023] [Accepted: 04/12/2023] [Indexed: 05/04/2023]
Abstract
The blood platelet plays an important role but often remains under-recognized in several vascular complications and associated diseases. Surprisingly, platelet hyperactivity and hyperaggregability have often been considered the critical risk factors for developing vascular dysfunctions in several neurodegenerative diseases (NDDs) like Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In addition, platelet structural and functional impairments promote prothrombotic and proinflammatory environment that can aggravate the progression of several NDDs. These findings provide the rationale for using antiplatelet agents not only to prevent morbidity but also to reduce mortality caused by NDDs. Therefore, we thoroughly review the evidence supporting the potential pleiotropic effects of several novel classes of synthetic antiplatelet drugs, that is, cyclooxygenase inhibitors, adenosine diphosphate receptor antagonists, protease-activated receptor blockers, and glycoprotein IIb/IIIa receptor inhibitors in NDDs. Apart from this, the review also emphasizes the recent developments of selected natural antiplatelet phytochemicals belonging to key classes of plant-based bioactive compounds, including polyphenols, alkaloids, terpenoids, and flavonoids as potential therapeutic candidates in NDDs. We believe that the broad analysis of contemporary strategies and specific approaches for plausible therapeutic treatment for NDDs presented in this review could be helpful for further successful research in this area.
Collapse
Affiliation(s)
- Samir K Beura
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Rishika Dhapola
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Abhishek R Panigrahi
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Pooja Yadav
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Reetesh Kumar
- Department of Agricultural Sciences, Institute of Applied Sciences and Humanities, GLA University, Mathura, Uttar Pradesh, India
| | - Dibbanti H Reddy
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Sunil K Singh
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| |
Collapse
|
21
|
Sim MMS, Shiferawe S, Wood JP. Novel strategies in antithrombotic therapy: targeting thrombosis while preserving hemostasis. Front Cardiovasc Med 2023; 10:1272971. [PMID: 37937289 PMCID: PMC10626538 DOI: 10.3389/fcvm.2023.1272971] [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: 08/04/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
Antithrombotic therapy is a delicate balance between the benefits of preventing a thrombotic event and the risks of inducing a major bleed. Traditional approaches have included antiplatelet and anticoagulant medications, require careful dosing and monitoring, and all carry some risk of bleeding. In recent years, several new targets have been identified, both in the platelet and coagulation systems, which may mitigate this bleeding risk. In this review, we briefly describe the current state of antithrombotic therapy, and then present a detailed discussion of the new generation of drugs that are being developed to target more safely existing or newly identified pathways, alongside the strategies to reverse direct oral anticoagulants, showcasing the breadth of approaches. Combined, these exciting advances in antithrombotic therapy bring us closer than we have ever been to the "holy grail" of the field, a treatment that separates the hemostatic and thrombotic systems, preventing clots without any concurrent bleeding risk.
Collapse
Affiliation(s)
- Martha M. S. Sim
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Semekidus Shiferawe
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Jeremy P. Wood
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
- Division of Cardiovascular Medicine Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
22
|
Kwon HW, Rhee MH, Shin JH. The Inhibitory Effects of Protaetia brevitarsis seulensis Larvae Extract on Human Platelet Aggregation and Glycoprotein IIb/IIIa Expression. Prev Nutr Food Sci 2023; 28:328-334. [PMID: 37842257 PMCID: PMC10567598 DOI: 10.3746/pnf.2023.28.3.328] [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: 04/03/2023] [Revised: 05/23/2023] [Accepted: 06/26/2023] [Indexed: 10/17/2023] Open
Abstract
The white-spotted flower chafer, Protaetia brevitarsis seulensis, is used as a traditional remedy against liver cirrhosis, hepatitis, and hepatic cancer. In this study, we investigated if P. brevitarsis extract (PBE) inhibited platelet aggregation via integrin αIIb/β3 regulation. We observed that PBE inhibited αIIb/β3 activation by regulating the cyclic nucleotides, cyclic adenosine monophosphate and cyclic guanosine monophosphate. Additionally, PBE affected phosphatidylinositol-3 kinase, Akt, SYK, glycogen synthase kinase-3α/β, cytosolic phospholipase A2, and p38 expression, which are signal transduction molecules expressed by platelets, and consequently suppressed αIIbβ3 activity and thromboxane A2 generation. Taken together, PBE showed strong antiplatelet effects and may be used to block thrombosis- and platelet-mediated cardiovascular diseases.
Collapse
Affiliation(s)
- Hyuk-Woo Kwon
- Department of Biomedical Laboratory Science, Far East University, Chungbuk 2760, Korea
- Microbiological Resource Research Institute, Far East University, Chungbuk 7601, Korea
| | - Man Hee Rhee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
- Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 19, Korea
| | - Jung-Hae Shin
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
- Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 19, Korea
| |
Collapse
|
23
|
Haroun EA, Mansour NO, Eltantawy A, Shams MEE. Effect of cilostazol on preventing paclitaxel-induced neuropathy in patients with breast cancer: A randomized controlled trial. Pharmacotherapy 2023; 43:872-882. [PMID: 37199288 DOI: 10.1002/phar.2830] [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: 03/10/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023]
Abstract
STUDY OBJECTIVE Paclitaxel-induced peripheral neuropathy is a significant clinical problem can markedly deteriorate patient's quality of life (QoL). Preclinical evidence exists about the preventive capacity of cilostazol against peripheral neuropathy. However, this hypothesis has not yet been clinically investigated. This proof-of-concept study evaluated the effect of cilostazol on the incidence of paclitaxel-induced peripheral neuropathy in patients with non-metastatic breast cancer. DESIGN This is a parallel randomized placebo-controlled trial. SETTING The Oncology Center at Mansoura University, Egypt. PATIENTS Patients with breast cancer scheduled to receive paclitaxel 175 mg/m2 biweekly. INTERVENTIONS Patients were randomized to either cilostazol group who received cilostazol tablets 100 mg BID, or to control group who received placebo instead. MEASUREMENTS The primary endpoint was the incidence of paclitaxel-induced neuropathy evaluated through common terminology criteria for adverse event (NCI-CTCAE) version 4. Secondary endpoints included assessment of the patient's QoL by the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (FACT-GOG-NTx) subscale. Exploratory outcome measures included changes in serum levels of biomarkers namely nerve growth factor (NGF), and neurofilament light chain (NfL). MAIN RESULTS The incidence of grade 2 and 3 peripheral neuropathies were significantly lower in the cilostazol group (40%) compared to the control group (86.7%) (p < 0.001). The incidence of clinically significant worsening in neuropathy-related QoL was higher in control group compared to the cilostazol group (p = 0.001). A higher percent increase from baseline in serum NGF was observed in the cilostazol group (p = 0.043). The circulating levels of NfL deemed comparable between the two arms at the end of the study (p = 0.593). CONCLUSION Adjunctive use of cilostazol is as a novel option that might reduce the incidence of paclitaxel-induced peripheral neuropathy and improve the patients' QoL. Future larger clinical trials are warranted to confirm these findings.
Collapse
Affiliation(s)
- Esraa A Haroun
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Noha O Mansour
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed Eltantawy
- Medical Oncology Unit, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Mohamed E E Shams
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
24
|
Kutryb-Zając B, Kawecka A, Nasadiuk K, Braczko A, Stawarska K, Caiazzo E, Koszałka P, Cicala C. Drugs targeting adenosine signaling pathways: A current view. Biomed Pharmacother 2023; 165:115184. [PMID: 37506580 DOI: 10.1016/j.biopha.2023.115184] [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: 05/18/2023] [Revised: 07/06/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Adenosine is an endogenous nucleoside that regulates many physiological and pathological processes. It is derived from either the intracellular or extracellular dephosphorylation of adenosine triphosphate and interacts with cell-surface G-protein-coupled receptors. Adenosine plays a substantial role in protecting against cell damage in areas of increased tissue metabolism and preventing organ dysfunction in pathological states. Targeting adenosine metabolism and receptor signaling may be an effective therapeutic approach for human diseases, including cardiovascular and central nervous system disorders, rheumatoid arthritis, asthma, renal diseases, and cancer. Several lines of evidence have shown that many drugs exert their beneficial effects by modulating adenosine signaling pathways but this knowledge urgently needs to be summarized, and most importantly, actualized. The present review collects pharmaceuticals and pharmacological or diagnostic tools that target adenosine signaling in their primary or secondary mode of action. We overviewed FDA-approved drugs as well as those currently being studied in clinical trials. Among them are already used in clinic A2A adenosine receptor modulators like istradefylline or regadenoson, but also plenty of anti-platelet, anti-inflammatory, or immunosuppressive, and anti-cancer drugs. On the other hand, we investigated dozens of specific adenosine pathway regulators that are tested in clinical trials to treat human infectious and noninfectious diseases. In conclusion, targeting purinergic signaling represents a great therapeutic challenge. The actual knowledge of the involvement of adenosinergic signaling as part of the mechanism of action of old drugs has open a path not only for drug-repurposing but also for new therapeutic strategies.
Collapse
Affiliation(s)
- Barbara Kutryb-Zając
- Department of Biochemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland.
| | - Ada Kawecka
- Department of Biochemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Khrystyna Nasadiuk
- Department of Biochemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Alicja Braczko
- Department of Biochemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Klaudia Stawarska
- Department of Biochemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Elisabetta Caiazzo
- Department of Pharmacy, School of Medicine, University of Naple Federico II, 80131 Naples, Italy
| | - Patrycja Koszałka
- Laboratory of Cell Biology and Immunology, Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology University of Gdańsk and Medical University of Gdańsk, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Carla Cicala
- Department of Pharmacy, School of Medicine, University of Naple Federico II, 80131 Naples, Italy
| |
Collapse
|
25
|
Gawaz M, Geisler T, Borst O. Current concepts and novel targets for antiplatelet therapy. Nat Rev Cardiol 2023; 20:583-599. [PMID: 37016032 DOI: 10.1038/s41569-023-00854-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
Platelets have a crucial role in haemostasis and atherothrombosis. Pharmacological control of platelet hyper-reactivity has become a cornerstone in the prevention of thrombo-ischaemic complications in atherosclerotic diseases. Current antiplatelet therapies substantially improve clinical outcomes in patients with coronary artery disease, but at the cost of increased risk of bleeding. Beyond their role in thrombosis, platelets are known to regulate inflammatory (thrombo-inflammatory) and microcirculatory pathways. Therefore, controlling platelet hyper-reactivity might have implications for both tissue inflammation (myocardial ischaemia) and vascular inflammation (vulnerable plaque formation) to prevent atherosclerosis. In this Review, we summarize the pathophysiological role of platelets in acute myocardial ischaemia, vascular inflammation and atherosclerotic progression. Furthermore, we highlight current clinical concepts of antiplatelet therapy that have contributed to improving patient care and have facilitated more individualized therapy. Finally, we discuss novel therapeutic targets and compounds for antiplatelet therapy that are currently in preclinical development, some of which have a more favourable safety profile than currently approved drugs with regard to bleeding risk. These novel antiplatelet targets might offer new strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- Meinrad Gawaz
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
26
|
Kubacka M, Mogilski S, Bednarski M, Pociecha K, Świerczek A, Nicosia N, Schabikowski J, Załuski M, Chłoń-Rzepa G, Hockemeyer J, Müller CE, Kieć-Kononowicz K, Kotańska M. Antiplatelet Effects of Selected Xanthine-Based Adenosine A 2A and A 2B Receptor Antagonists Determined in Rat Blood. Int J Mol Sci 2023; 24:13378. [PMID: 37686188 PMCID: PMC10487961 DOI: 10.3390/ijms241713378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The platelet aggregation inhibitory activity of selected xanthine-based adenosine A2A and A2B receptor antagonists was investigated, and attempts were made to explain the observed effects. The selective A2B receptor antagonist PSB-603 and the A2A receptor antagonist TB-42 inhibited platelet aggregation induced by collagen or ADP. In addition to adenosine receptor blockade, the compounds were found to act as moderately potent non-selective inhibitors of phosphodiesterases (PDEs). TB-42 showed the highest inhibitory activity against PDE3A along with moderate activity against PDE2A and PDE5A. The antiplatelet activity of PSB-603 and TB-42 may be due to inhibition of PDEs, which induces an increase in cAMP and/or cGMP concentrations in platelets. The xanthine-based adenosine receptor antagonists were found to be non-cytotoxic for platelets. Some of the compounds showed anti-oxidative properties reducing lipid peroxidation. These results may provide a basis for the future development of multi-target xanthine derivatives for the treatment of inflammation and atherosclerosis and the prevention of heart infarction and stroke.
Collapse
Affiliation(s)
- Monika Kubacka
- Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.K.); (S.M.)
| | - Szczepan Mogilski
- Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.K.); (S.M.)
| | - Marek Bednarski
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.B.); (N.N.)
| | - Krzysztof Pociecha
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (K.P.); (A.Ś.)
| | - Artur Świerczek
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (K.P.); (A.Ś.)
| | - Noemi Nicosia
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.B.); (N.N.)
- Division of Neuroscience, Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Jakub Schabikowski
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (J.S.); (M.Z.); (K.K.-K.)
| | - Michał Załuski
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (J.S.); (M.Z.); (K.K.-K.)
| | - Grażyna Chłoń-Rzepa
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Kraków, Poland;
| | - Jörg Hockemeyer
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, An der Immenburg 4, D-53121 Bonn, Germany; (J.H.); (C.E.M.)
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, An der Immenburg 4, D-53121 Bonn, Germany; (J.H.); (C.E.M.)
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (J.S.); (M.Z.); (K.K.-K.)
| | - Magdalena Kotańska
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.B.); (N.N.)
| |
Collapse
|
27
|
Hong HJ, Nam GS, Nam KS. Daidzein Inhibits Human Platelet Activation by Downregulating Thromboxane A 2 Production and Granule Release, Regardless of COX-1 Activity. Int J Mol Sci 2023; 24:11985. [PMID: 37569361 PMCID: PMC10418957 DOI: 10.3390/ijms241511985] [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: 06/19/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Platelets play crucial roles in cardiovascular diseases (CVDs) by regulating hemostasis and blood coagulation at sites of blood vessel damage. Accumulating evidence indicates daidzein inhibits platelet activation, but the mechanism involved has not been elucidated. Thus, in this study, we investigated the mechanism responsible for the inhibition of collagen-induced platelet aggregation by daidzein. We found that in collagen-induced platelets, daidzein suppressed the production of thromboxane A2 (TXA2), a molecule involved in platelet activation and aggregation, by inhibiting the cytosolic phospholipase A2 (cPLA2) signaling pathway. However, daidzein did not affect cyclooxygenase-1 (COX-1). Furthermore, daidzein attenuated the PI3K/PDK1/Akt/GSK3αβ and MAPK (p38, ERK) signaling pathways, increased the phosphorylation of inositol trisphosphate receptor1 (IP3R1) and vasodilator-stimulated phosphoprotein (VASP), and increased the level of cyclic adenosine monophosphate (cAMP). These results suggest that daidzein inhibits granule release (ATP, serotonin, P-selectin), integrin αIIbβ3 activation, and clot retraction. Taken together, our study demonstrates that daidzein inhibits collagen-induced platelet aggregation and suggests that daidzein has therapeutic potential for the treatment of platelet aggregation-related diseases such as atherosclerosis and thrombosis.
Collapse
Affiliation(s)
- Hyun-Jin Hong
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea;
| | - Gi-Suk Nam
- Department of Biomedical Laboratory Science, Honam University, 120, Honamdae-gil, Gwangsan-gu, Gwangju 62399, Republic of Korea
| | - Kyung-Soo Nam
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea;
| |
Collapse
|
28
|
Altê GA, Rodrigues ALS. Exploring the Molecular Targets for the Antidepressant and Antisuicidal Effects of Ketamine Enantiomers by Using Network Pharmacology and Molecular Docking. Pharmaceuticals (Basel) 2023; 16:1013. [PMID: 37513925 PMCID: PMC10383558 DOI: 10.3390/ph16071013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Ketamine, a racemic mixture of esketamine (S-ketamine) and arketamine (R-ketamine), has received particular attention for its rapid antidepressant and antisuicidal effects. NMDA receptor inhibition has been indicated as one of the main mechanisms of action of the racemic mixture, but other pharmacological targets have also been proposed. This study aimed to explore the possible multiple targets of ketamine enantiomers related to their antidepressant and antisuicidal effects. To this end, targets were predicted using Swiss Target Prediction software for each ketamine enantiomer. Targets related to depression and suicide were collected by the Gene Cards database. The intersections of targets were analyzed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Network pharmacology analysis was performed using Gene Mania and Cytoscape software. Molecular docking was used to predict the main targets of the network. The results indicated that esketamine and arketamine share some biological targets, particularly NMDA receptor and phosphodiesterases 3A, 7A, and 5A but have specific molecular targets. While esketamine is predicted to interact with the GABAergic system, arketamine may interact with macrophage migration inhibitory factor (MIF). Both ketamine enantiomers activate neuroplasticity-related signaling pathways and show addiction potential. Our results identified novel, poorly explored molecular targets that may be related to the beneficial effects of esketamine and arketamine against depression and suicide.
Collapse
Affiliation(s)
- Glorister A Altê
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis 88037-000, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis 88037-000, SC, Brazil
| |
Collapse
|
29
|
Twine CP, Kakkos SK, Aboyans V, Baumgartner I, Behrendt CA, Bellmunt-Montoya S, Jilma B, Nordanstig J, Saratzis A, Reekers JA, Zlatanovic P, Antoniou GA, de Borst GJ, Bastos Gonçalves F, Chakfé N, Coscas R, Dias NV, Hinchliffe RJ, Kolh P, Lindholt JS, Mees BME, Resch TA, Trimarchi S, Tulamo R, Vermassen FEG, Wanhainen A, Koncar I, Fitridge R, Matsagkas M, Valgimigli M. Editor's Choice - European Society for Vascular Surgery (ESVS) 2023 Clinical Practice Guidelines on Antithrombotic Therapy for Vascular Diseases. Eur J Vasc Endovasc Surg 2023; 65:627-689. [PMID: 37019274 DOI: 10.1016/j.ejvs.2023.03.042] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 04/05/2023]
|
30
|
Korb VG, Schultz IC, Beckenkamp LR, Wink MR. A Systematic Review of the Role of Purinergic Signalling Pathway in the Treatment of COVID-19. Int J Mol Sci 2023; 24:ijms24097865. [PMID: 37175571 PMCID: PMC10178215 DOI: 10.3390/ijms24097865] [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: 01/09/2023] [Revised: 03/27/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global health concern. Three years since its origin, despite the approval of vaccines and specific treatments against this new coronavirus, there are still high rates of infection, hospitalization, and mortality in some countries. COVID-19 is characterised by a high inflammatory state and coagulation disturbances that may be linked to purinergic signalling molecules such as adenosine triphosphate (ATP), adenosine diphosphate (ADP), adenosine (ADO), and purinergic receptors (P1 and P2). These nucleotides/nucleosides play important roles in cellular processes, such as immunomodulation, blood clot formation, and vasodilation, which are affected during SARS-CoV-2 infection. Therefore, drugs targeting this purinergic pathway, currently used for other pathologies, are being evaluated in preclinical and clinical trials for COVID-19. In this review, we focus on the potential of these drugs to control the release, degradation, and reuptake of these extracellular nucleotides and nucleosides to treat COVID-19. Drugs targeting the P1 receptors could have therapeutic efficacy due to their capacity to modulate the cytokine storm and the immune response. Those acting in P2X7, which is linked to NLRP3 inflammasome activation, are also valuable candidates as they can reduce the release of pro-inflammatory cytokines. However, according to the available preclinical and clinical data, the most promising medications to be used for COVID-19 treatment are those that modulate platelets behaviour and blood coagulation factors, mainly through the P2Y12 receptor.
Collapse
Affiliation(s)
- Vitoria Guero Korb
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil
| | - Iago Carvalho Schultz
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil
| | - Liziane Raquel Beckenkamp
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil
| | - Márcia Rosângela Wink
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Sala 304 Centro, Porto Alegre 90050-170, RS, Brazil
| |
Collapse
|
31
|
Callegari K, Dash S, Uchida H, Shingai Y, Liu C, Khodarkovskaya A, Lee Y, Ito A, Lopez A, Zhang T, Xiang J, Kluk MJ, Sanchez T. Molecular profiling of the stroke-induced alterations in the cerebral microvasculature reveals promising therapeutic candidates. Proc Natl Acad Sci U S A 2023; 120:e2205786120. [PMID: 37058487 PMCID: PMC10120001 DOI: 10.1073/pnas.2205786120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 02/08/2023] [Indexed: 05/11/2023] Open
Abstract
Stroke-induced cerebral microvascular dysfunction contributes to aggravation of neuronal injury and compromises the efficacy of current reperfusion therapies. Understanding the molecular alterations in cerebral microvessels in stroke will provide original opportunities for scientific investigation of novel therapeutic strategies. Toward this goal, using a recently optimized method which minimizes cell activation and preserves endothelial cell interactions and RNA integrity, we conducted a genome-wide transcriptomic analysis of cerebral microvessels in a mouse model of stroke and compared these transcriptomic alterations with the ones observed in human, nonfatal, brain stroke lesions. Results from these unbiased comparative analyses have revealed the common alterations in mouse stroke microvessels and human stroke lesions and identified shared molecular features associated with vascular disease (e.g., Serpine1/Plasminogen Activator Inhibitor-1, Hemoxygenase-1), endothelial activation (e.g., Angiopoietin-2), and alterations in sphingolipid metabolism and signaling (e.g., Sphigosine-1-Phosphate Receptor 2). Sphingolipid profiling of mouse cerebral microvessels validated the transcript data and revealed the enrichment of sphingomyelin and sphingoid species in the cerebral microvasculature compared to brain and the stroke-induced increase in ceramide species. In summary, our study has identified novel molecular alterations in several microvessel-enriched, translationally relevant, and druggable targets, which are potent modulators of endothelial function. Our comparative analyses have revealed the presence of molecular features associated with cerebral microvascular dysfunction in human chronic stroke lesions. The results shared here provide a detailed resource for therapeutic discovery of candidates for neurovascular protection in stroke and potentially, other pathologies exhibiting cerebral microvascular dysfunction.
Collapse
Affiliation(s)
- Keri Callegari
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Sabyasachi Dash
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Hiroki Uchida
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Yuto Shingai
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Catherine Liu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Anne Khodarkovskaya
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Yunkyoung Lee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Akira Ito
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Amanda Lopez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY10065
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY10065
| | - Michael J. Kluk
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Teresa Sanchez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065
| |
Collapse
|
32
|
Gui X, Chu X, Du Y, Wang Y, Zhang S, Ding Y, Tong H, Xu M, Li Y, Ju W, Sun Z, Li Z, Zeng L, Xu K, Qiao J. Impaired Platelet Function and Thrombus Formation in PDE5A-Deficient Mice. Thromb Haemost 2023; 123:207-218. [PMID: 36252813 DOI: 10.1055/a-1962-1613] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Intracellular cyclic GMP (cGMP) inhibits platelet function. Platelet cGMP levels are controlled by phosphodiesterase 5A (PDE5A)-mediated degradation. However, the exact role of PDE5A in platelet function and thrombus formation remains poorly understood. In this study, we characterized the role of PDE5A in platelet activation and function. Platelets were isolated from wild type or PDE5A-/- mice to measure platelet aggregation, activation, phosphatidylserine exposure (annexin-V binding), reactive oxygen species (ROS) generation, platelet spreading as well as clot retraction. Cytosolic calcium mobilization was measured using Fluo-4 AM by a microplate reader. Western blot was used to measure the phosphorylation of VASP, ERK1/2, p38, JNK, and AKT. FeCl3-induced arterial thrombosis and venous thrombosis were assessed to evaluate the in vivo hemostatic function and thrombus formation. Additionally, in vitro thrombus formation was assessed in a microfluidic whole-blood perfusion assay. PDE5A-deficient mice presented significantly prolonged tail bleeding time and delayed arterial and venous thrombus formation. PDE5A deficiency significantly inhibited platelet aggregation, ATP release, P-selectin expression, and integrin aIIbb3 activation. In addition, an impaired spreading on collagen or fibrinogen and clot retraction was observed in PDE5A-deficient platelets. Moreover, PDE5A deficiency reduced phosphatidylserine exposure, calcium mobilization, ROS production, and increased intracellular cGMP level along with elevated VASP phosphorylation and reduced phosphorylation of ERK1/2, p38, JNK, and AKT. In conclusion, PDE5A modulates platelet activation and function and thrombus formation, indicating that therapeutically targeting it might be beneficial for the treatment of thrombotic diseases.
Collapse
Affiliation(s)
- Xiang Gui
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Xiang Chu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yuwei Du
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yuhan Wang
- School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Sixuan Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yangyang Ding
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Huan Tong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Mengdi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yue Li
- School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Zengtian Sun
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China.,School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| |
Collapse
|
33
|
Singla A, Dadario NB, Singla A, Greenberg P, Yan R, Nanda A, Boison D, Malhotra R, Patel S, Nipun S, Maninderpal K, Castro D, Bdiiwi S, Boktor H, Kyi HH, Sutherland A, Patrawalla A, Ly K, Xie Y, Sonig A, Khandelwal P, Liu J, Koziol J, Finkle D, Subanna S, Libutti SK. A randomized controlled trial to evaluate outcomes with Aggrenox in patients with SARS-CoV-2 infection. PLoS One 2023; 18:e0274243. [PMID: 36716303 PMCID: PMC9886260 DOI: 10.1371/journal.pone.0274243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/23/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is an immunoinflammatory and hypercoagulable state that contributes to respiratory distress, multi-organ dysfunction, and mortality. Dipyridamole, by increasing extracellular adenosine, has been postulated to be protective for COVID-19 patients through its immunosuppressive, anti-inflammatory, anti-coagulant, vasodilatory, and anti-viral actions. Likewise, low-dose aspirin has also demonstrated protective effects for COVID-19 patients. This study evaluated the effect of these two drugs formulated together as Aggrenox in hospitalized COVID-19 patients. METHODS In an open-label, single site randomized controlled trial (RCT), hospitalized COVID-19 patients were assigned to adjunctive Aggrenox (Dipyridamole ER 200mg/ Aspirin 25mg orally/enterally) with standard of care treatment compared to standard of care treatment alone. Primary endpoint was illness severity according to changes on the eight-point COVID ordinal scale, with levels of 1 to 8 where higher scores represent worse illness. Secondary endpoints included all-cause mortality and respiratory failure. Outcomes were measured through days 14, 28, and/or hospital discharge. RESULTS From October 1, 2020 to April 30, 2021, a total of 98 patients, who had a median [IQR] age of 57 [47, 62] years and were 53.1% (n = 52) female, were randomized equally between study groups (n = 49 Aggrenox plus standard of care versus n = 49 standard of care alone). No clinically significant differences were found between those who received adjunctive Aggrenox and the control group in terms of illness severity (COVID ordinal scale) at days 14 and 28. The overall mortality through day 28 was 6.1% (3 patients, n = 49) in the Aggrenox group and 10.2% (5 patients, n = 49) in the control group (OR [95% CI]: 0.40 [0.04, 4.01], p = 0.44). Respiratory failure through day 28 occurred in 4 (8.3%, n = 48) patients in the Aggrenox group and 7 (14.6%, n = 48) patients in the standard of care group (OR [95% CI]: 0.21 [0.02, 2.56], p = 0.22). A larger decrease in the platelet count and blood glucose levels, and larger increase in creatinine and sodium levels within the first 7 days of hospital admission were each independent predictors of 28-day mortality (p < 0.05). CONCLUSION In this study of hospitalized patients with COVID-19, while the outcomes of COVID illness severity, odds of mortality, and chance of respiratory failure were better in the Aggrenox group compared to standard of care alone, the data did not reach statistical significance to support the standard use of adjuvant Aggrenox in such patients.
Collapse
Affiliation(s)
- Amit Singla
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Nicholas B. Dadario
- Department of Neurological Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Ashima Singla
- Department of OBGYN, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Patricia Greenberg
- Biostatistics and Epidemiology Services Center (RUBIES), Rutgers School of Public Health, Rutgers University, Piscataway, New Jersey, United States of America
| | - Rachel Yan
- Department of Neurological Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Anil Nanda
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
- Department of Neurological Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Detlev Boison
- Department of Neurological Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, United States of America
| | - Rakesh Malhotra
- Department of Medicine, Division of Nephrology, UCSD, San Diego, California, United States of America
| | - Sunil Patel
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Suri Nipun
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Kaur Maninderpal
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Dorothy Castro
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Sanaa Bdiiwi
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Hala Boktor
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Htay Htay Kyi
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Anne Sutherland
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Amee Patrawalla
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Kevin Ly
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Yingda Xie
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Ashish Sonig
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Priyank Khandelwal
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - James Liu
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Joseph Koziol
- Department of Neurological Surgery, Saint Barnabas Medical Center, Livingston, New Jersey, United States of America
| | - Diana Finkle
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Sara Subanna
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Steven K. Libutti
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| |
Collapse
|
34
|
Kloner RA, Stanek E, Crowe CL, Singhal M, Pepe RS, Bradsher J, Rosen RC. Effect of phosphodiesterase type 5 inhibitors on major adverse cardiovascular events and overall mortality in a large nationwide cohort of men with erectile dysfunction and cardiovascular risk factors: A retrospective, observational study based on healthcare claims and national death index data. J Sex Med 2023; 20:38-48. [PMID: 36897243 DOI: 10.1093/jsxmed/qdac005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/23/2022] [Accepted: 09/12/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Treatment with phosphodiesterase type 5 inhibitors (PDE-5is) is effective in treating erectile dysfunction (ED). AIM The objective of this study was to determine the effect of PDE-5is on the incidence of major adverse cardiovascular (CV) events (MACE; composite outcome of CV death, hospitalization for myocardial infarction, coronary revascularization, stroke, heart failure, and unstable angina pectoris) and overall mortality. METHODS A retrospective observational cohort study was conducted in a large US claims database in men with ≥1 diagnosis of ED without prior MACE within 1 year, from January 1, 2006, to October 31, 2020. The exposed group had ≥1 claim for PDE-5i and the unexposed group had no claims for PDE-5i, and the groups were matched up to 1:4 on baseline risk variables. OUTCOME The primary outcome was MACE and the secondary outcomes were overall mortality and individual components of MACE, determined by multivariable Cox proportional hazard modeling. RESULTS Matched plus multivariable analyses showed that MACE was lower by 13% in men exposed (n = 23 816) to PDE-5is (hazard ratio [HR] 0.87; 95% CI 0.79-0.95; P = .001) vs nonexposure (n = 48 682) over mean follow-up periods of 37 and 29 months, respectively, with lower incidence of coronary revascularization (HR 0.85; 95% CI 0.73-0.98; P = .029), heart failure (HR 0.83; 95% CI 0.72-0.97; P = .016), unstable angina (HR 0.78; 95% CI 0.64-0.96; P = .021), and CV death (HR 0.61; 95% CI 0.41-0.90; P = .014) with PDE-5i exposure. Phosphodiesterase type 5 inhibitor-exposed men had a 25% lower incidence of overall mortality (HR 0.75; 95% CI 0.65-0.87; P < .001). Men without coronary artery disease (CAD) but with CV risk factors at baseline showed a similar pattern. In the main study cohort, men in the highest quartile of PDE-5i exposure had the lowest incidence of MACE (HR 0.45; 95% CI 0.37-0.54; P < .001) and overall mortality (HR 0.51; 95% CI 0.37-0.71; P < .001) vs the lowest exposure quartile. In a subgroup with baseline type 2 diabetes (n = 6503), PDE-5i exposure was associated with a lower MACE risk (HR 0.79; 95% CI 0.64-0.97; P = .022). CLINICAL IMPLICATIONS PDE-5is may have cardioprotective effects. STRENGTHS AND LIMITATIONS Strengths are the large numbers of participants and consistency of the data; limitations include the retrospective nature of the study and unknown confounders. CONCLUSIONS In a large population of US men with ED, PDE-5i exposure was associated with lower incidence of MACE, CV death, and overall mortality risk compared to non-exposure. Risk reduction correlated with PDE-5i exposure level.
Collapse
Affiliation(s)
- Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, United States.,Keck School of Medicine, Department of Medicine, Division of Cardiovascular Medicine, Los Angeles, CA, United States
| | - Eric Stanek
- HealthCore Inc., Wilmington, DE, United States.,Anthem, Inc., Indianapolis, IN, United States
| | | | | | | | - Julia Bradsher
- Huntington Medical Research Institutes, Pasadena, CA, United States
| | - Raymond C Rosen
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, San Francisco, CA, United States
| |
Collapse
|
35
|
Rahn C, Bakuradze T, Stegmüller S, Galan J, Niesen S, Winterhalter P, Richling E. Polyphenol-Rich Beverage Consumption Affecting Parameters of the Lipid Metabolism in Healthy Subjects. Int J Mol Sci 2023; 24:ijms24010841. [PMID: 36614281 PMCID: PMC9821765 DOI: 10.3390/ijms24010841] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023] Open
Abstract
Polyphenols are a diverse and widely distributed class of secondary metabolites, which possess numerous beneficial properties including a modulation of glucose and lipid metabolism. This placebo-controlled human intervention study was performed to explore effects of polyphenol-rich beverage (PRB) uptake on lipid metabolism, as well as DNA integrity. In this case, 36 healthy men were randomly divided to consume either 750 mL of a PRB (containing 51% chokeberry, cranberry, and pomegranate) or a placebo drink daily for eight weeks. Only PRB consumption was found to decrease fat and protein intakes significantly compared to the preceding one-week washout period. During the intervention with PRB an increased fat-free mass was shown after four weeks, whereas a significant elevation in body weight and leptin was observed in placebo group. Blood lipids were not significantly altered after PRB consumption, while triglyceride levels increased after placebo drink intake. In platelets, a significant inhibition of phosphodiesterase (PDE) activity was observed, more pronounced in test group. Consuming the PRB decreased total DNA strand breaks in whole blood as well as H2O2-induced breaks in isolated lymphocytes. Overall, our study suggested beneficial effects on lipid metabolism by reduced energy intake, modulation of biomarkers such as PDE activity and improved DNA integrity associated with PRB consumption.
Collapse
Affiliation(s)
- Celina Rahn
- Division of Food Chemistry and Toxicology, Department of Chemistry, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Erwin-Schrödinger-Straße 52, D-67663 Kaiserslautern, Germany
| | - Tamara Bakuradze
- Division of Food Chemistry and Toxicology, Department of Chemistry, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Erwin-Schrödinger-Straße 52, D-67663 Kaiserslautern, Germany
| | - Simone Stegmüller
- Division of Food Chemistry and Toxicology, Department of Chemistry, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Erwin-Schrödinger-Straße 52, D-67663 Kaiserslautern, Germany
| | - Jens Galan
- Medical Institute, Hochgewanne 19, D-67269 Grünstadt, Germany
| | - Sonja Niesen
- Institute of Food Chemistry, Technische Universität Braunschweig, Schleinitzstraße 20, D-38106 Braunschweig, Germany
| | - Peter Winterhalter
- Institute of Food Chemistry, Technische Universität Braunschweig, Schleinitzstraße 20, D-38106 Braunschweig, Germany
| | - Elke Richling
- Division of Food Chemistry and Toxicology, Department of Chemistry, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Erwin-Schrödinger-Straße 52, D-67663 Kaiserslautern, Germany
- Correspondence: ; Tel.: +49-631-205-4061
| |
Collapse
|
36
|
Novel approaches to antiplatelet therapy. Biochem Pharmacol 2022; 206:115297. [DOI: 10.1016/j.bcp.2022.115297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/20/2022]
|
37
|
Kwon HW, Kim SD, Rhee MH, Shin JH. Pharmacological Actions of 5-Hydroxyindolin-2 on Modulation of Platelet Functions and Thrombus Formation via Thromboxane A 2 Inhibition and cAMP Production. Int J Mol Sci 2022; 23:ijms232314545. [PMID: 36498873 PMCID: PMC9739977 DOI: 10.3390/ijms232314545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Platelets play a very significant role in hemostasis while simultaneously posing a risk for the development of various cardiovascular diseases. Platelet-mediated issues can occur in blood vessels and trigger various medical problems. Therefore, controlling platelet function is important in the prevention of thrombosis. In this regard, we need to find compounds that provide potent antiplatelet activity with minimum side effects. Therefore, we examined the effect of 5-hydroxyindolin-2-one isolated from Protaetia brevitarsis larvae having antiplatelet properties and investigated different pathways that mediate the antiplatelet activity. We examined the effect of 5-hydroxyindolin-2-one (5-HI) on the regulation of phosphoproteins, thromboxane A2 generation, and integrin αIIbβ3 action. Our data showed that human platelet aggregation was inhibited by 5-HI (75, 100, 150, 200 μM) without cytotoxicity, and it suppressed intracellular Ca2+ concentration through the regulation of inositol 1, 4, 5-triphosphate receptor I (Ser1756) and extracellular signal-regulated kinase (ERK). Moreover, collagen-elevated thromboxane A2 production and αIIbβ3 action were inhibited by 5-HI through the regulation of cytosolic phospholipase A2 (cPLA2), mitogen-activated protein kinase p38 (p38MAPK), vasodilator-stimulated phosphoprotein (VASP), phosphoinositide 3-kinase (PI3K), and Akt (protein kinase B). Therefore, we suggested that 5-HI could be a potential substance for the prevention of thrombosis-mediated thrombosis.
Collapse
Affiliation(s)
- Hyuk-Woo Kwon
- Department of Biomedical Laboratory Science, Far East University, Eumseong 27601, Republic of Korea
- Department of Microbiological Resource Research Institute, Far East University, Eumseong 27601, Republic of Korea
| | - Sung Dae Kim
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Man Hee Rhee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
- Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Correspondence: author: (M.H.R.); (J.-H.S.)
| | - Jung-Hae Shin
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
- Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Correspondence: author: (M.H.R.); (J.-H.S.)
| |
Collapse
|
38
|
Degjoni A, Campolo F, Stefanini L, Venneri MA. The NO/cGMP/PKG pathway in platelets: The therapeutic potential of PDE5 inhibitors in platelet disorders. J Thromb Haemost 2022; 20:2465-2474. [PMID: 35950928 PMCID: PMC9805178 DOI: 10.1111/jth.15844] [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: 05/20/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 01/09/2023]
Abstract
Platelets are the "guardians" of the blood circulatory system. At sites of vessel injury, they ensure hemostasis and promote immunity and vessel repair. However, their uncontrolled activation is one of the main drivers of thrombosis. To keep circulating platelets in a quiescent state, the endothelium releases platelet antagonists including nitric oxide (NO) that acts by stimulating the intracellular receptor guanylyl cyclase (GC). The latter produces the second messenger cyclic guanosine-3',5'-monophosphate (cGMP) that inhibits platelet activation by stimulating protein kinase G, which phosphorylates hundreds of intracellular targets. Intracellular cGMP pools are tightly regulated by a fine balance between GC and phosphodiesterases (PDEs) that are responsible for the hydrolysis of cyclic nucleotides. Phosphodiesterase type 5 (PDE5) is a cGMP-specific PDE, broadly expressed in most tissues in humans and rodents. In clinical practice, PDE5 inhibitors (PDE5i) are used as first-line therapy for erectile dysfunction, pulmonary artery hypertension, and lower urinary tract symptoms. However, several studies have shown that PDE5i may ameliorate the outcome of various other conditions, like heart failure and stroke. Interestingly, NO donors and cGMP analogs increase the capacity of anti-platelet drugs targeting the purinergic receptor type Y, subtype 12 (P2Y12) receptor to block platelet aggregation, and preclinical studies have shown that PDE5i inhibits platelet functions. This review summarizes the molecular mechanisms underlying the effect of PDE5i on platelet activation and aggregation focusing on the therapeutic potential of PDE5i in platelet disorders, and the outcomes of a combined therapy with PDE5i and NO donors to inhibit platelet activation.
Collapse
Affiliation(s)
- Anisa Degjoni
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Federica Campolo
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Lucia Stefanini
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Mary Anna Venneri
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| |
Collapse
|
39
|
Abstract
INTRODUCTION Platelets play a key role in arterial thrombosis and antiplatelet therapy is pivotal in the treatment of cardiovascular disease. Current antiplatelet drugs target different pathways of platelet activation and show specific pharmacodynamic and pharmacokinetic characteristics, implicating clinically relevant drug-drug interactions. AREAS COVERED This article reviews the role of platelets in hemostasis and cardiovascular thrombosis, and discusses the key pharmacodynamics, drug-drug interactions and reversal strategies of clinically used antiplatelet drugs. EXPERT OPINION Antiplatelet therapies target distinct pathways of platelet activation: thromboxane A2 synthesis, adenosine diphosphate-mediated signaling, integrin αIIbβ3 (GPIIb/IIIa), thrombin-mediated platelet activation via the PAR1 receptor and phosphodiesterases. Key clinical drug-drug interactions of antiplatelet agents involve acetylsalicylic acid - ibuprofen, clopidogrel - omeprazole, and morphine - oral P2Y12 inhibitors, all of which lead to an attenuated antiplatelet effect. Platelet function and genetic testing and the use of scores (ARC-HBR, PRECISE-DAPT, ESC ischemic risk definition) may contribute to a more tailored antiplatelet therapy. High on-treatment platelet reactivity presents a key problem in the acute management of ST-elevation myocardial infarction (STEMI). A treatment strategy involving early initiation of an intravenous antiplatelet agent may be able to bridge the gap of insufficient platelet inhibition in high ischemic risk patients with STEMI.
Collapse
Affiliation(s)
- Georg Gelbenegger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Quantification of cyclic AMP and cyclic GMP levels in Krebs-Henseleit solution by LC-MS/MS: application in washed platelet aggregation samples. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1211:123472. [DOI: 10.1016/j.jchromb.2022.123472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022]
|
41
|
Altered platelet functions during treatment with apremilast for psoriatic arthritis: A case report. Curr Res Transl Med 2022; 70:103358. [PMID: 35724504 DOI: 10.1016/j.retram.2022.103358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/05/2022] [Indexed: 01/31/2023]
|
42
|
Falcinelli E, Petito E, Gresele P. The role of platelets, neutrophils and endothelium in COVID-19 infection. Expert Rev Hematol 2022; 15:727-745. [PMID: 35930267 DOI: 10.1080/17474086.2022.2110061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION COVID-19 is associated to an increased risk of thrombosis, as a result of a complex process that involves the activation of vascular and circulating cells, the release of soluble inflammatory and thrombotic mediators and blood clotting activation. AREAS COVERED This article reviews the pathophysiological role of platelets, neutrophils and the endothelium, and of their interactions, in the thrombotic complications of COVID-19 patients, and the current and future therapeutic approaches targeting these cell types. EXPERT OPINION Virus-induced platelet, neutrophil and endothelial cell changes are crucial triggers of the thrombotic complications and of the adverse evolution of COVID-19. Both the direct interaction with the virus and the associated cytokine storm concur to trigger cell activation in a classical thromboinflammatory vicious circle. Although heparin has proven to be an effective prophylactic and therapeutic weapon for the prevention and treatment of COVID-19-associated thrombosis, it acts downstream of the cascade of events triggered by SARS-CoV-2. The identification of specific molecular targets interrupting the thromboinflammatory cascade upstream, and more specifically acting either on the interaction of SARS-CoV-2 with blood and vascular cells or on the specific signalling mechanisms associated with their COVID-19-associated activation, might theoretically offer greater protection with potentially lesser side effects.
Collapse
Affiliation(s)
- E Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - E Petito
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - P Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
43
|
Zhang Y, Benz P, Stehle D, Yang S, Kurz H, Feil S, Nagel G, Feil R, Gao S, Bender M. Optogenetic manipulation of cyclic guanosine monophosphate to probe phosphodiesterase activities in megakaryocytes. Open Biol 2022; 12:220058. [PMID: 35975649 PMCID: PMC9382455 DOI: 10.1098/rsob.220058] [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] [Indexed: 11/23/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP) signalling plays a fundamental role in many cell types, including platelets. cGMP has been implicated in platelet formation, but mechanistic detail about its spatio-temporal regulation in megakaryocytes (MKs) is lacking. Optogenetics is a technique which allows spatio-temporal manipulation of molecular events in living cells or organisms. We took advantage of this method and expressed a photo-activated guanylyl cyclase, Blastocladiella emersonii Cyclase opsin (BeCyclop), after viral-mediated gene transfer in bone marrow (BM)-derived MKs to precisely light-modulate cGMP levels. BeCyclop-MKs showed a significantly increased cGMP concentration after illumination, which was strongly dependent on phosphodiesterase (PDE) 5 activity. This finding was corroborated by real-time imaging of cGMP signals which revealed that pharmacological PDE5 inhibition also potentiated nitric oxide-triggered cGMP generation in BM MKs. In summary, we established for the first-time optogenetics in primary MKs and show that PDE5 is the predominant PDE regulating cGMP levels in MKs. These findings also demonstrate that optogenetics allows for the precise manipulation of MK biology.
Collapse
Affiliation(s)
- Yujing Zhang
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Pascal Benz
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Shang Yang
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Hendrikje Kurz
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Georg Nagel
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Shiqiang Gao
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Markus Bender
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| |
Collapse
|
44
|
Leonardi GR, Lescano CH, Costa JL, Mazetto B, Orsi FA, Monica FZ. Adenosine diphosphate-induced aggregation is enhanced in platelets obtained from patients with thrombotic primary antiphospholipid syndrome (t-PAPS): Role of P2Y 12 -cAMP signaling pathway. J Thromb Haemost 2022; 20:1699-1711. [PMID: 35395698 DOI: 10.1111/jth.15724] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/22/2022] [Accepted: 04/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Thrombotic antiphospholipid syndrome (t-PAPS) is characterized by arterial, venous, or microvascular occlusions, which are explained, in part, by the presence of antiphospholipid (aPL) antibodies. Although there is much evidence indicating that isolated aPL antibodies increase the activity of platelets obtained from healthy volunteers, platelet function in t-PAPS has not been as widely studied. OBJECTIVE To evaluate platelet reactivity in t-PAPS patients. METHODS Platelet aggregation, protein expression, and cyclic nucleotide levels were carried out in platelet rich plasma (PRP) or washed platelets (WPs) obtained from t-PAPS or healthy volunteers. RESULTS ADP-induced aggregation was significantly higher in PRP obtained from t-PAPS than obtained from the control. The protein expression of P2Y12 receptor and Gs alpha was significantly higher and lower, respectively in WPs from t-PAPS patients. In PRP incubated with iloprost or sodium nitroprusside, the residual platelet reactivity induced by ADP was still higher in PRP from t-PAPS than from the control. Lower intracellular levels of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP) were observed in unstimulated PRP from t-PAPS patients. The protein expression of soluble guanylate cyclase subunits and phosphodiesterases types 3 and 5 did not differ. The antiplatelet activity of ticagrelor was similar between the groups and cilostazol significantly potentiated this response. Isolated aPL antibodies obtained from t-PAPS patients potentiated ADP-induced aggregation in healthy platelets but did not affect the inhibitory responses induced by iloprost or sodium nitroprusside. CONCLUSIONS The overexpression of P2Y12 receptor, accompanied by lower levels of cAMP and cGMP levels produced greater amplitude of ADP aggregation in platelets from t-PAPS patients.
Collapse
Affiliation(s)
- Guilherme Ruiz Leonardi
- Department of Translation Medicine (Pharmacology), Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Caroline Honaiser Lescano
- Department of Translation Medicine (Pharmacology), Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Jose Luiz Costa
- Campinas Poison Control Center, University of Campinas, Campinas, Brazil
| | - Bruna Mazetto
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Fernanda Andrade Orsi
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
- Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Fabiola Zakia Monica
- Department of Translation Medicine (Pharmacology), Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| |
Collapse
|
45
|
Handler SS, Jin J, Ogawa MT, Feinstein JA, Lo C. Abnormal platelet aggregation in pediatric pulmonary hypertension. Pulm Circ 2022; 12:e12104. [PMID: 35864911 PMCID: PMC9294293 DOI: 10.1002/pul2.12104] [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: 08/03/2021] [Revised: 05/24/2022] [Accepted: 06/16/2022] [Indexed: 11/14/2022] Open
Abstract
Endogenous prostacyclin stimulates pulmonary vasodilation and inhibits platelet aggregation. For the synthetic analog treprostinil, used in the treatment of pulmonary hypertension (PH), conflicting, anecdotal evidence exists regarding its effects on clinically relevant platelet function. This study investigated whether treprostinil therapy results in inhibition of platelet aggregation in pediatric PH patients. This is a single institution, prospective, cohort study. Pediatric patients ≤18 years of age on medical therapy for PH underwent platelet function testing by light transmission aggregometry with U-46619-a stable analog of endoperoxide prostaglandin H2, exhibiting properties similar to thromboxane A2 (TXA2). Results were compared for those on continuous treprostinil therapy (TRE) versus those on other, non-prostacyclin therapies (non-TRE). Thirty-five patients were enrolled: 18 in the TRE group and 17 in the non-TRE group. There was no difference in platelet aggregation abnormalities between the two groups: 44% (n = 8) in the TRE group and 41% (n = 7) in the non-TRE group were abnormal. Furthermore, subgroup analysis showed no difference based on treprostinil dosing. This study demonstrated similar, moderately high rates of abnormal platelet aggregation in pediatric PH patients on continuous treprostinil therapy compared to those on other, non-prostacyclin therapies. The high rate of abnormal platelet aggregation in the entire cohort, however, warrants follow-up study to identify a potential inherent risk in this population.
Collapse
Affiliation(s)
- Stephanie S. Handler
- Department of Pediatrics (Cardiology)Medical College of WisconsinMilwaukeeWisconsinUSA
| | - Jing Jin
- Clinical LaboratoriesStanford Health CarePalo AltoCaliforniaUSA
| | - Michelle T. Ogawa
- Department of Pediatrics (Cardiology)Stanford UniversityStanfordCaliforniaUSA
| | - Jeffrey A. Feinstein
- Department of Pediatrics (Cardiology)Stanford UniversityStanfordCaliforniaUSA
- Department of BioengineeringStanford UniversityStanfordCaliforniaUSA
| | - Clara Lo
- Department of Pediatrics (Hematology)Stanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
46
|
Phosphodiesterase (PDE) III inhibitor, Cilostazol, improved memory impairment in aluminum chloride-treated rats: modulation of cAMP/CREB pathway. Inflammopharmacology 2022; 30:2477-2488. [PMID: 35727381 DOI: 10.1007/s10787-022-01010-1] [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: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/05/2022]
Abstract
The most prevalent type of dementia is Alzheimer's disease (AD), which is currently incurable. Existing treatments for Alzheimer's disease, such as acetylcholinesterase inhibitors, are only effective for symptom relief. Disease-modifying medications for Alzheimer's disease are desperately required, given the enormous burdens that the disease places on individuals and communities. Phosphodiesterase (PDE) inhibitors are gaining a lot of attention in the research community because of their potential in treating age-related cognitive decline. Cilostazol is a selective PDE III inhibitor used as antiplatelet agent through cAMP response element-binding (CREB) protein phosphorylation pathway (cAMP/CREB). The neuroprotective effect of cilostazol in AD-like cognitive decline in rats was investigated in this study. After 2 months of intraperitoneal administration of 10 mg/kg aluminum chloride, Morris water maze and Y-maze (behavioral tests) were performed. After that, histological and biochemical examinations of the hippocampal region were carried out. Aluminum chloride-treated rats showed histological, biochemical, and behavioral changes similar to Alzheimer's disease. Cilostazol improved rats' behavioral and histological conditions, raised neprilysin level while reduced levels of amyloid-beta protein and phosphorylated tau protein. It also decreased the hippocampal levels of tumor necrosis factor-alpha, nuclear factor-kappa B, FAS ligand, acetylcholinesterase content, and malondialdehyde. These outcomes demonstrate the protective activity of cilostazol versus aluminum-induced memory impairment.
Collapse
|
47
|
Huang MX, Chen YQ, Liu RD, Huang Y, Zhang C. Discovery of Dipyridamole Analogues with Enhanced Metabolic Stability for the Treatment of Idiopathic Pulmonary Fibrosis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113452. [PMID: 35684390 PMCID: PMC9182104 DOI: 10.3390/molecules27113452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/15/2022] [Accepted: 05/19/2022] [Indexed: 11/28/2022]
Abstract
Dipyridamole, apart from its well-known antiplatelet and phosphodiesterase inhibitory activities, is a promising old drug for the treatment of pulmonary fibrosis. However, dipyridamole shows poor pharmacokinetic properties with a half-life (T1/2) of 7 min in rat liver microsomes (RLM). To improve the metabolic stability of dipyridamole, a series of pyrimidopyrimidine derivatives have been designed with the assistance of molecular docking. Among all the twenty-four synthesized compounds, compound (S)-4h showed outstanding metabolic stability (T1/2 = 67 min) in RLM, with an IC50 of 332 nM against PDE5. Furthermore, some interesting structure–activity relationships (SAR) were explained with the assistance of molecular docking.
Collapse
Affiliation(s)
- Meng-Xing Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.-X.H.); (Y.-Q.C.); (R.-D.L.); (Y.H.)
| | - Yan-Quan Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.-X.H.); (Y.-Q.C.); (R.-D.L.); (Y.H.)
| | - Run-Duo Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.-X.H.); (Y.-Q.C.); (R.-D.L.); (Y.H.)
| | - Yue Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.-X.H.); (Y.-Q.C.); (R.-D.L.); (Y.H.)
| | - Chen Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.-X.H.); (Y.-Q.C.); (R.-D.L.); (Y.H.)
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
- Correspondence:
| |
Collapse
|
48
|
A Review on Phosphodiesterase-5 Inhibitors as a Topical Therapy for Erectile Dysfunction. Sex Med Rev 2022; 10:376-391. [DOI: 10.1016/j.sxmr.2022.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 11/18/2022]
|
49
|
Chai E, Chen J, Li C, Zhang X, Fan Z, Yang S, Zhao K, Li W, Xiao Z, Zhang Y, Tang F. The Efficacy and Safety of Cilostazol vs. Aspirin for Secondary Stroke Prevention: A Systematic Review and Meta-Analysis. Front Neurol 2022; 13:814654. [PMID: 35242099 PMCID: PMC8885726 DOI: 10.3389/fneur.2022.814654] [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: 11/14/2021] [Accepted: 01/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background Cilostazol is often used in Asia-Pacific countries for stroke prevention. The current systematic review and meta-analysis aimed to evaluate the effectiveness, safety, and adverse outcomes of cilostazol monotherapy compared to aspirin monotherapy for secondary stroke prevention. Methods The researchers conducted a comprehensive research in multiple databases (PubMed, Embase, and Cochrane library) of randomized controlled trials from conception to December 2020. The primary efficacy outcome was the occurrence of any stroke, the primary safety outcome was the bleeding risk, and the primary adverse outcome was the rate of headache and dizziness. The Mantel-Haenszel method was used to calculate a random-effects prediction. Cilostazol and aspirin were compared using a pooled risk assessment with 95% CIs. Results Six studies involving 5,617 patients were included in this review. Compared with aspirin monotherapy, cilostazol was associated with significantly lower rates of any strokes (RR: 0.67; 95% CI: 0.55–0.82) and significantly lower bleeding rates [risk ratio (RR): 0.53; 95% CI: 0.37–0.74]. However, compared with aspirin monotherapy, cilostazol was associated with significantly higher rates of headache (RR: 1.77; 95% CI: 1.41–2.20) and dizziness (RR: 1.28; 95% CI: 1.08–1.52). Conclusions Consistent with previous studies, cilostazol monotherapy is superior to aspirin monotherapy in reducing the rate of any strokes and the bleeding risk after having a stroke. However, the use of cilostazol monotherapy is associated with several adverse life outcomes such as headaches and dizziness.
Collapse
Affiliation(s)
- Erqing Chai
- Cerebrovascular Disease Center, Gansu Provincial Hospital, Lanzhou, China.,Emergency General Hospital, Beijing, China
| | - Jinhua Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Changqing Li
- Cerebrovascular Disease Center, Gansu Provincial Hospital, Lanzhou, China
| | - Xue Zhang
- Intensive Care Unit 1, Gansu Provincial Hospital, Lanzhou, China
| | - Zhiqiang Fan
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shijie Yang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Kaixuan Zhao
- School of Clinical Medicine, Ningxia Medical University, Ningxia, China
| | - Wei Li
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zaixing Xiao
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yichuan Zhang
- School of Clinical Medicine, Ningxia Medical University, Ningxia, China
| | - Futian Tang
- Key Laboratory of Gansu Digestive System Tumor, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
50
|
Zhao Y, Zhou P, Gao W, Zhong H, Chen Y, Chen W, Waresi M, Xie K, Shi H, Gong H, He G, Qiu Z, Luo X, Li J. Cilostazol combined with P2Y 12 receptor inhibitors: A substitute antiplatelet regimen for aspirin-intolerant patients undergoing percutaneous coronary stent implantation. Clin Cardiol 2022; 45:189-197. [PMID: 35120275 PMCID: PMC8860475 DOI: 10.1002/clc.23787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/28/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Background Cilostazol combined with P2Y12 receptor inhibitor has been used as a substitute regimen for aspirin‐intolerant patients undergoing percutaneous coronary stent implantation on a small scale. Its exact impact on platelet functions and clinical benefits of aspirin‐intolerant patients is unknown. Hypothesis Cilostazol combined with P2Y12 receptor inhibitors could be used as a substitute antiplatelet regimen for aspirin‐intolerant patients undergoing percutaneous coronary stent implantation. Methods In this multicenter prospective cohort trial, patients undergoing elective percutaneous coronary stent implantation were assigned to the cilostazol group (cilostazol plus P2Y12 receptor inhibitors), based on aspirin intolerance criteria, or the aspirin group (aspirin plus P2Y12 receptor inhibitors). Platelet PAC‐1, CD62p, and vasodilator‐stimulated phosphoprotein phosphorylation (VASP‐P) were detected by flow cytometry. The primary endpoints were major adverse cardiovascular and cerebrovascular events (MACCE) including all‐cause death, acute myocardial infarction, emerging arrhythmia, nonfatal stroke, and heart failure. The secondary endpoints were the Bleeding Academic Research Consortium (BARC) bleeding events. Results One hundred and fifty‐four aspirin‐intolerant percutaneous coronary stent implantation patients and 154 matched aspirin‐tolerant patients from a total of 2059 percutaneous coronary stent implantation patients were enrolled. The relative activation level of PAC‐1, CD62p, and platelet reaction index reflected by the VASP‐P test were similar in the two groups (p > .05). After 12 months of follow‐up, the incidence of all‐cause death was 1.9% in the cilostazol group and 1.3% in the aspirin group (risk ratio [RR], 1.500; 95% confidence interval [CI], 0.254–8.852; p = 1.000); the incidence of acute myocardial infarction was 0.6% in the cilostazol group and 1.3% in the aspirin group (RR, 0.500; 95% CI, 0.046–5.457; p = 1.000). No significant difference was seen in other MACCE events, or in any types of BARC bleeding events. Conclusions Cilostazol combined with P2Y12 inhibitors was not inferior to aspirin‐based standard therapy and could be used as a reasonable substitute antiplatelet regimen for aspirin‐intolerant patients undergoing percutaneous coronary stent implantation, but again with limitations, which required a larger sample and longer follow‐up to confirm its efficacy.
Collapse
Affiliation(s)
- Yikai Zhao
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Peng Zhou
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Wen Gao
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Haoxuan Zhong
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Yufei Chen
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Wei Chen
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Maieryemu Waresi
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Kun Xie
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Haiming Shi
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Hui Gong
- Department of CardiologyJinshan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Guibin He
- Department of CardiologyLuodian HospitalShanghaiPeople's Republic of China
| | - Zhaohui Qiu
- Department of CardiologyTongren Hospital of Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Xinping Luo
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| | - Jian Li
- Department of CardiologyHuashan Hospital of Fudan UniversityShanghaiPeople's Republic of China
| |
Collapse
|