1
|
Guo J, Zhou YL, Yang Y, Guo S, You E, Xie X, Jiang Y, Mao C, Xu HE, Zhang Y. Structural basis of tethered agonism and G protein coupling of protease-activated receptors. Cell Res 2024; 34:725-734. [PMID: 38997424 PMCID: PMC11443083 DOI: 10.1038/s41422-024-00997-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
Protease-activated receptors (PARs) are a unique group within the G protein-coupled receptor superfamily, orchestrating cellular responses to extracellular proteases via enzymatic cleavage, which triggers intracellular signaling pathways. Protease-activated receptor 1 (PAR1) is a key member of this family and is recognized as a critical pharmacological target for managing thrombotic disorders. In this study, we present cryo-electron microscopy structures of PAR1 in its activated state, induced by its natural tethered agonist (TA), in complex with two distinct downstream proteins, the Gq and Gi heterotrimers, respectively. The TA peptide is positioned within a surface pocket, prompting PAR1 activation through notable conformational shifts. Contrary to the typical receptor activation that involves the outward movement of transmembrane helix 6 (TM6), PAR1 activation is characterized by the simultaneous downward shift of TM6 and TM7, coupled with the rotation of a group of aromatic residues. This results in the displacement of an intracellular anion, creating space for downstream G protein binding. Our findings delineate the TA recognition pattern and highlight a distinct role of the second extracellular loop in forming β-sheets with TA within the PAR family, a feature not observed in other TA-activated receptors. Moreover, the nuanced differences in the interactions between intracellular loops 2/3 and the Gα subunit of different G proteins are crucial for determining the specificity of G protein coupling. These insights contribute to our understanding of the ligand binding and activation mechanisms of PARs, illuminating the basis for PAR1's versatility in G protein coupling.
Collapse
Affiliation(s)
- Jia Guo
- Department of Pharmacology and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center for Structural Pharmacology and Therapeutics Development, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- CAS Key Laboratory of Receptor Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yun-Li Zhou
- Department of Pharmacology and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yixin Yang
- Department of Pharmacology and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shimeng Guo
- CAS Key Laboratory of Receptor Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Erli You
- CAS Key Laboratory of Receptor Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xin Xie
- CAS Key Laboratory of Receptor Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Jiang
- Lingang Laboratory, Shanghai, China
| | - Chunyou Mao
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Center for Structural Pharmacology and Therapeutics Development, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang, China.
| | - H Eric Xu
- CAS Key Laboratory of Receptor Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Yan Zhang
- Department of Pharmacology and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Center for Structural Pharmacology and Therapeutics Development, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Gupta R, Lin M, Mehta A, Aedma SK, Shah R, Ranchal P, Vyas AV, Singh S, Kluck B, Combs WG, Patel NC. Protease-Activated Receptor Antagonist for Reducing Cardiovascular Events - A Review on Vorapaxar. Curr Probl Cardiol 2023; 48:101035. [PMID: 34718032 DOI: 10.1016/j.cpcardiol.2021.101035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/16/2021] [Indexed: 02/01/2023]
Abstract
Acute Coronary Syndrome (ACS) is a term that describes pathologies related to myocardial ischemia, and is comprised of unstable angina, non-ST elevation myocardial infarction, and ST elevation myocardial infarction. Urgent management of ACS is typically necessary to prevent future morbidity and mortality. Current medical recommendations of ACS management involve use of dual antiplatelet therapy, typically with aspirin and clopidogrel. However, newer therapies are being designed and researched to improve outcomes for patients with ACS. Vorapaxar is a novel antiplatelet therapy that inhibits thrombin-mediated platelet aggregation to prevent recurrence of ischemic events. It has been Food and Drug Administration approved for reduction of thrombotic cardiovascular events in patients with a history of MI or peripheral arterial disease with concomitant use of clopidogrel and/or aspirin, based upon the findings of the TRA 2°P-TIMI 50 trial. However, Vorapaxar was also found to have a significantly increased risk of bleeding, which must be considered when administering this drug. Based upon further subgroup analysis of both the TRA 2°P-TIMI 50 trial and TRACER trial, Vorapaxar was found to be potentially beneficial in patients with peripheral artery disease, coronary artery bypass grafting, and ischemic stroke. There are current trials in progress that are further evaluating the use of Vorapaxar in those conditions, and future research and trials are necessary to fully determine the utility of this drug.
Collapse
Affiliation(s)
- Rahul Gupta
- Department of Cardiology, Lehigh Valley Heart and Vascular Institute, Lehigh Valley Health Network, Allentown, PA.
| | - Muling Lin
- Department of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL
| | - Anila Mehta
- Department of Internal Medicine, Carle Foundation Hospital, Urbana, IL
| | - Surya K Aedma
- Department of Internal Medicine, Carle Foundation Hospital, Urbana, IL
| | - Rajendra Shah
- Vassar Brothers Medical Center, Nuvance Health, Poughkeepsie, NY
| | - Purva Ranchal
- Department of Internal Medicine, Boston University, Boston, MA
| | - Apurva V Vyas
- Department of Cardiology, Lehigh Valley Heart and Vascular Institute, Lehigh Valley Health Network, Allentown, PA
| | - Shailendra Singh
- Department of Cardiology, Lehigh Valley Heart and Vascular Institute, Lehigh Valley Health Network, Allentown, PA
| | - Bryan Kluck
- Department of Cardiology, Lehigh Valley Heart and Vascular Institute, Lehigh Valley Health Network, Allentown, PA
| | - William G Combs
- Department of Cardiology, Lehigh Valley Heart and Vascular Institute, Lehigh Valley Health Network, Allentown, PA
| | - Nainesh C Patel
- Department of Cardiology, Lehigh Valley Heart and Vascular Institute, Lehigh Valley Health Network, Allentown, PA
| |
Collapse
|
3
|
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
|
4
|
Jourdi G, Godier A, Lordkipanidzé M, Marquis-Gravel G, Gaussem P. Antiplatelet Therapy for Atherothrombotic Disease in 2022—From Population to Patient-Centered Approaches. Front Cardiovasc Med 2022; 9:805525. [PMID: 35155631 PMCID: PMC8832164 DOI: 10.3389/fcvm.2022.805525] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/06/2022] [Indexed: 12/20/2022] Open
Abstract
Antiplatelet agents, with aspirin and P2Y12 receptor antagonists as major key molecules, are currently the cornerstone of pharmacological treatment of atherothrombotic events including a variety of cardio- and cerebro-vascular as well as peripheral artery diseases. Over the last decades, significant changes have been made to antiplatelet therapeutic and prophylactic strategies. The shift from a population-based approach to patient-centered precision medicine requires greater awareness of individual risks and benefits associated with the different antiplatelet strategies, so that the right patient gets the right therapy at the right time. In this review, we present the currently available antiplatelet agents, outline different management strategies, particularly in case of bleeding or in perioperative setting, and develop the concept of high on-treatment platelet reactivity and the steps toward person-centered precision medicine aiming to optimize patient care.
Collapse
Affiliation(s)
- Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Georges Jourdi
| | - Anne Godier
- Université de Paris, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris, France
- Department of Anesthesiology and Critical Care, AP-HP, Université de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Guillaume Marquis-Gravel
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Pascale Gaussem
- Université de Paris, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris, France
- Service d'Hématologie Biologique, AP-HP, Université de Paris, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
5
|
Jourdi G, Lordkipanidzé M, Philippe A, Bachelot-Loza C, Gaussem P. Current and Novel Antiplatelet Therapies for the Treatment of Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms222313079. [PMID: 34884884 PMCID: PMC8658271 DOI: 10.3390/ijms222313079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Over the last decades, antiplatelet agents, mainly aspirin and P2Y12 receptor antagonists, have significantly reduced morbidity and mortality associated with arterial thrombosis. Their pharmacological characteristics, including pharmacokinetic/pharmacodynamics profiles, have been extensively studied, and a significant number of clinical trials assessing their efficacy and safety in various clinical settings have established antithrombotic efficacy. Notwithstanding, antiplatelet agents carry an inherent risk of bleeding. Given that bleeding is associated with adverse cardiovascular outcomes and mortality, there is an unmet clinical need to develop novel antiplatelet therapies that inhibit thrombosis while maintaining hemostasis. In this review, we present the currently available antiplatelet agents, with a particular focus on their targets, pharmacological characteristics, and patterns of use. We will further discuss the novel antiplatelet therapies in the pipeline, with the goal of improved clinical outcomes among patients with atherothrombotic diseases.
Collapse
Affiliation(s)
- Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada;
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Correspondence: (G.J.); (P.G.)
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada;
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Aurélien Philippe
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
- Service d’Hématologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Christilla Bachelot-Loza
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
| | - Pascale Gaussem
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
- Service d’Hématologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
- Correspondence: (G.J.); (P.G.)
| |
Collapse
|
6
|
Which proteinase-activated receptor-1 antagonist is better?: Evaluation of vorapaxar and parmodulin-2 effects on human left internal mammary artery endothelial function. Life Sci 2021; 286:120045. [PMID: 34653426 DOI: 10.1016/j.lfs.2021.120045] [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: 08/05/2021] [Revised: 09/26/2021] [Accepted: 10/06/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Endothelial dysfunction occurs as an early event in cardiovascular disease. Previously, vorapaxar, a proteinase-activated receptor-1 antagonist, was shown to cause endothelial damage in a cell culture study. Therefore, our study aimed to compare the effects of vorapaxar and parmodulin-2, proteinase-activated receptor-1 biased agonist, on human left internal mammary artery endothelial function in vitro. METHOD Isolated arteries were hung in the organ baths. Acetylcholine responses (10-11-10-6 M) were obtained in endothelium-intact tissues the following incubation with vorapaxar/parmodulin-2 (10-6 M) to determine the effects of these molecules on the endothelium-dependent relaxation. Subsequently, endothelium-dependent relaxation responses of tissues were investigated in the presence of L-NAME (10-4 M), L-arginine (10-5 M), indomethacin (10-5 M), and charybdotoxin-apamin (10-7 M) in addition to vorapaxar/parmodulin-2 incubation. Besides, the effect of these molecules on endothelium-independent relaxation response was evaluated with sodium nitroprusside (10-11-10-6 M). Finally, the sections of human arteries were imaged using a transmission electron microscope, and the integrity of the endothelial layer was evaluated. RESULTS We found that vorapaxar caused significant endothelial dysfunction by disrupting nitric oxide and endothelium-derived hyperpolarizing factor-dependent relaxation mechanisms. Parmodulin-2 did not cause endothelial damage. Neither vorapaxar nor parmodulin-2 disrupted endothelium-independent relaxation responses. The effect of vorapaxar on the endothelial layer was supported by the transmission electron microscope images. CONCLUSION Parmodulin-2 may be a better option than vorapaxar in treating cardiovascular diseases since it can inhibit PAR-1 without caused endothelial dysfunction.
Collapse
|
7
|
Multiparameter phenotyping of platelet reactivity for stratification of human cohorts. Blood Adv 2021; 5:4017-4030. [PMID: 34474473 PMCID: PMC8945618 DOI: 10.1182/bloodadvances.2020003261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 05/12/2021] [Indexed: 12/30/2022] Open
Abstract
Accurate and comprehensive assessment of platelet function across cohorts of donors may be key to understanding the risk of thrombotic events associated with cardiovascular disease, and, hence, to help personalize the application of antiplatelet drugs. However, platelet function tests can be difficult to perform and analyze; they also can be unreliable or uninformative and poorly standardized across studies. The Platelet Phenomic Analysis (PPAnalysis) assay and associated open-source software platform were developed in response to these challenges. PPAnalysis utilizes preprepared freeze-dried microtiter plates to provide a detailed characterization of platelet function. The automated analysis of the high-dimensional data enables the identification of subpopulations of donors with distinct platelet function phenotypes. Using this approach, we identified that the sensitivity of a donor's platelets to an agonist and their capacity to generate a functional response are distinct independent metrics of platelet reactivity. Hierarchical clustering of these metrics identified 6 subgroups with distinct platelet phenotypes within healthy cohorts, indicating that platelet reactivity does not fit into the traditional simple categories of "high" and "low" responders. These platelet phenotypes were found to exist in 2 independent cohorts of healthy donors and were stable on recall. PPAnalysis is a powerful tool for stratification of cohorts on the basis of platelet reactivity that will enable investigation of the causes and consequences of differences in platelet function and drive progress toward precision medicine.
Collapse
|
8
|
Abstract
Antiplatelet agents are the standard of practice in the management of atherosclerosis and acute coronary syndrome (ACS). In contrast to the available antiplatelet agents, vorapaxar represents a novel mechanism of action. It is an antagonist of the platelet protease-activated receptor-1 (PAR-1) and inhibits thrombin-induced and thrombin receptor agonist peptide (TRAP)- induced platelet aggregation. The TRA2○P-TIMI 50 trial led to the approval of vorapaxar by the Food and Drug Administration and European Medicines Agency for the reduction of thrombotic cardiovascular events in patients with a history of myocardial infarction (MI) or peripheral arterial disease. TRA2○P-TIMI 50 trial showed that the use of vorapaxar (2.5 mg once/daily) in addition to standard dual antiplatelet therapy (DAPT) with aspirin and a P2Y12 receptor inhibitor, was effective in the secondary prevention of recurrent thrombotic events among patients with previous atherothrombosis, particularly in patients with prior MI; at the expense of an increase in major bleeding. Another recently published VORA-PRATIC (Vorapaxar in Patients with Prior Myocardial Infarction Treated with prasugrel and ticagrelor) study showed that among post-MI patients treated with potent P2Y12 inhibitors (prasugrel or ticagrelor), vorapaxar reduced platelet-driven global thrombogenicity, an effect that persisted, albeit attenuated, in the absence of aspirin. The current review summarizes an up to date literature on pharmacokinetics, pharmacodynamics, and clinical efficacy of vorapaxar and proposes future directions of research.
Collapse
|
9
|
El Alaoui MZ, Guy A, Khalki L, Limami Y, Benomar A, Zaid N, Cherrah Y, Mekhfi H, Cadi R, Zaid Y. [Current antiplatelet agents, new inhibitors and therapeutic targets]. Med Sci (Paris) 2020; 36:348-357. [PMID: 32356711 DOI: 10.1051/medsci/2020061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular diseases are the leading cause of deaths in the world. Platelets play a major role in the occurrence of these diseases and the development of antiplatelet drugs is a priority in the fight against cardiovascular diseases-associated mortality. Aspirin and thienopyridine-based P2Y12 inhibitors are the main drugs currently used. These molecules target the initiation of platelets activation and are responsible for a moderate inhibitory action. Other antiplatelet agents, as glycoprotein (GP) IIb/IIIa antagonists, inhibit platelet aggregation independently of initial activation-associated pathways, but are responsible for increased hemorrhagic events. Regarding each antiplatelet agent's specific characteristics, the prescription of these drugs must take into account the type of cardiovascular event, the age of the patient, the past medical history, and the potential hemorrhagic adverse events. Thus, there is a need for the development of new molecules with a more targeted effect, maintaining optimal efficiency but with a reduction of the hemorrhagic risk, which is the principal limitation of these treatments.
Collapse
Affiliation(s)
| | - Alexandre Guy
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Maroc
| | - Loubna Khalki
- Research Center of Mohammed VI University of Health Sciences, Casablanca, Maroc
| | - Youness Limami
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Ali Benomar
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Nabil Zaid
- Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Maroc
| | - Yahia Cherrah
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Hassan Mekhfi
- Laboratory of Physiology, Genetic and Ethnopharmacology, Faculty of Sciences, Mohammed the First University, Oujda, Maroc
| | - Rachida Cadi
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Maroc
| | - Younes Zaid
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc - Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Maroc
| |
Collapse
|
10
|
Zhou P, Yin JX, Tao HL, Zhang HW. Pathogenesis and management of heparin-induced thrombocytopenia and thrombosis. Clin Chim Acta 2020; 504:73-80. [DOI: 10.1016/j.cca.2020.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 01/19/2023]
|
11
|
Tantry US, Bliden KP, Chaudhary R, Novakovic M, Rout A, Gurbel PA. Vorapaxar in the treatment of cardiovascular diseases. Future Cardiol 2020; 16:373-384. [PMID: 32308016 DOI: 10.2217/fca-2019-0090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vorapaxar specifically and effectively inhibits protease activated receptor-1 and may reduce thrombin-mediated ischemic events without interfering primary hemostasis. In the TRA-2P-TIMI 50 trial, vorapaxar reduced the risk of primary ischemic outcome but with increased bleeding risk. In the post hoc analysis, in patients with a history of myocardial infarction, peripheral artery disease, the net clinical outcome favored vorapaxar therapy with 10% reduction in cardiovascular death, myocardial infarction, stroke, urgent coronary revascularization and moderate or severe bleeding. Based on these favorable results, vorapaxar was approved for the reduction of thrombotic cardiovascular events in patients with prior myocardial infarction or with peripheral artery disease on top of standard antiplatelet therapy. A careful patient selection is needed to balance efficacy versus safety.
Collapse
Affiliation(s)
- Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, MD 21215, USA
| | - Kevin P Bliden
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, MD 21215, USA
| | - Rahul Chaudhary
- Division of Hospital Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Marko Novakovic
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, MD 21215, USA
| | - Amit Rout
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, MD 21215, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, MD 21215, USA
| |
Collapse
|
12
|
Bliden K, Chaudhary R, Kuliopulos A, Tran H, Taheri H, Tehrani B, Rosenblatt A, Navarese E, Tantry US, Gurbel PA. Effects of vorapaxar on clot characteristics, coagulation, inflammation, and platelet and endothelial function in patients treated with mono- and dual-antiplatelet therapy. J Thromb Haemost 2020; 18:23-35. [PMID: 31444884 PMCID: PMC6940524 DOI: 10.1111/jth.14616] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Vorapaxar is indicated with standard antiplatelet therapy (APT) in patients with a history of myocardial infarction (MI) or peripheral arterial disease (PAD). OBJECTIVES To evaluate the comparative effects of vorapaxar on platelet-fibrin clot characteristics (PFCC), coagulation, inflammation, and platelet and endothelial function during treatment with daily 81 mg aspirin (A), 75 mg clopidogrel (C), both (C + A), or neither. METHODS Thrombelastography, conventional platelet aggregation (PA), ex vivo endothelial function by ENDOPAT, coagulation, platelet activation/inflammation marked by urinary 11-dehydrothromboxane B2 (UTxB2 ) and safety were determined in patients who were APT naïve (n = 21), on C (n = 8), on A (n = 29), and on A + C (n = 23) during 1 month of vorapaxar therapy and 1 month of offset. RESULTS Vorapaxar had no effect on PFCC, ADP- or collagen-induced PA, thrombin time, fibrinogen, PT, PTT, von Willebrand factor (vWF), D-dimer, or endothelial function (P > .05 in all groups). Inhibition of SFLLRN (PAR-1 activating peptide)-stimulated PA by vorapaxar was accelerated by A + C at 2 hours (P < .05 versus other groups) with nearly complete inhibition by 30 days that persisted through 30 days after discontinuation in all groups (P < .001). SFLLRN-induced PA during offset was lower in APT patients versus APT-naïve patients (P < .05). Inhibition of UTxB2 was observed in APT-naive patients treated with vorapaxar (P < .05). Minor bleeding was only observed in C-treated patients. CONCLUSION Vorapaxar had no influence on PFCC measured by thrombelastography, coagulation, or endothelial function irrespective of APT. Inhibition of protease activated receptor (PAR)-1 mediated platelet aggregation by vorapaxar was accelerated by A + C and offset was prolonged by concomitant APT. Vorapaxar-induced anti-inflammatory effects were observed in non-aspirin-treated patients.
Collapse
Affiliation(s)
- Kevin Bliden
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA, USA
| | - Rahul Chaudhary
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, MA, USA
| | - Henry Tran
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA, USA
| | - Hamid Taheri
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA, USA
| | - Behnam Tehrani
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA, USA
| | - Arnold Rosenblatt
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA, USA
| | - Eliano Navarese
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA, USA
| | - Udaya S. Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Paul A. Gurbel
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA, USA
| |
Collapse
|
13
|
Xiang Q, Pang X, Liu Z, Yang G, Tao W, Pei Q, Cui Y. Progress in the development of antiplatelet agents: Focus on the targeted molecular pathway from bench to clinic. Pharmacol Ther 2019; 203:107393. [PMID: 31356909 DOI: 10.1016/j.pharmthera.2019.107393] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 07/10/2019] [Indexed: 12/22/2022]
Abstract
Antiplatelet drugs serve as a first-line antithrombotic therapy for the management of acute ischemic events and the prevention of secondary complications in vascular diseases. Numerous antiplatelet therapies have been developed; however, currently available agents are still associated with inadequate efficacy, risk of bleeding, and variability in individual response. Understanding the mechanisms of platelet involvement in thrombosis and the clinical development process of antiplatelet agents is critical for the discovery of novel agents. The functions of platelets in thrombosis are regulated by two major mechanisms: the interaction between surface receptors and their ligands, and the downstream intracellular signaling pathways. Recently, most of the progress made in antiplatelet drug development has been achieved with P2Y receptor antagonists. Additionally, the usage of GP IIb/IIIa receptor antagonists has decreased, because it is associated with a higher risk of bleeding and thrombocytopenia. Agents targeting other platelet surface receptors such as PARs, TP receptor, EP3 receptor, GPIb-IX-V receptor, P-selectin, as well as intracellular signaling factors, such as PI3Kβ, have been evaluated in an attempt to develop the next generation of antiplatelet drugs, reduce or eliminate interpatient variability of drug efficacy and significantly lower the risk of drug-induced bleeding. The aim of this review is to describe the pathways of platelet activation in thrombosis, and summarize the development process of antiplatelet agents, as well as the preclinical and clinical evaluations performed on these agents.
Collapse
Affiliation(s)
- Qian Xiang
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing 100034, China
| | - Xiaocong Pang
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing 100034, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Research Center of Drug Clinical Evaluation of Central South University, 138 TongZiPo Road, Changsha, Hunan 410013, China
| | - Weikang Tao
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Research Center of Drug Clinical Evaluation of Central South University, 138 TongZiPo Road, Changsha, Hunan 410013, China
| | - Qi Pei
- Shanghai Hengrui Pharmaceuticals Co., 279 Wenjing Road, Shanghai, China
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing 100034, China.
| |
Collapse
|
14
|
Park CM, Lee S, Song J, Lee J. Discovery of ( E)‐5,5‐Difluoro‐1‐[2‐[5‐(3‐fluorophenyl)pyridin‐2‐yl]vinyl]octahydrospiro(indene‐2,5′‐oxazolidin)‐2′‐one as a PAR1 Antagonist. B KOREAN CHEM SOC 2019. [DOI: 10.1002/bkcs.11747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Chul Min Park
- Division of Drug Discovery ResearchKorea Research Institute of Chemical Technology Deajeon 34114 South Korea
| | - Sunkyung Lee
- Division of Drug Discovery ResearchKorea Research Institute of Chemical Technology Deajeon 34114 South Korea
- Korea University of Science and Technology Daejeon 34114 South Korea
| | - Jong‐Hwan Song
- Division of Drug Discovery ResearchKorea Research Institute of Chemical Technology Deajeon 34114 South Korea
| | - Joo‐Youn Lee
- Division of Drug Discovery ResearchKorea Research Institute of Chemical Technology Deajeon 34114 South Korea
| |
Collapse
|
15
|
|
16
|
Pultar J, Wadowski PP, Panzer S, Gremmel T. Oral antiplatelet agents in cardiovascular disease. VASA 2018; 48:291-302. [PMID: 30324870 DOI: 10.1024/0301-1526/a000753] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antiplatelet agents significantly reduce mortality and morbidity in ischemic heart disease, cerebrovascular disease and peripheral artery disease (PAD), and are therefore part of guideline-driven daily medical treatment in these patients. Due to its beneficial effects in the secondary prevention of atherothrombotic events, aspirin remains the most frequently prescribed antiplatelet agent in cardiovascular disease. In patients with acute coronary syndromes (ACS) and in those undergoing angioplasty with stent implantation dual antiplatelet therapy with aspirin and an adenosine diphosphate (ADP) receptor antagonist is indicated. The development of the newer ADP P2Y12 inhibitors prasugrel and ticagrelor has further improved prognosis in ACS patients compared to clopidogrel. Moreover, vorapaxar allows the inhibition of platelet activation by thrombin via protease-activated receptor-1 and has been approved for the use in patients with PAD and in those with a history of myocardial infarction. This review article summarizes the current evidence on oral antiplatelet agents in cardiovascular disease. Keywords: Aspirin, clopidogrel, prasugrel, ticagrelor, vorapaxar, cardiovascular disease.
Collapse
Affiliation(s)
- Joseph Pultar
- 1 Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,a Joseph Pultar and Patricia P. Wadowski share first authorship
| | - Patricia P Wadowski
- 1 Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,a Joseph Pultar and Patricia P. Wadowski share first authorship
| | - Simon Panzer
- 2 Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Gremmel
- 1 Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,3 Department of Internal Medicine, Cardiology and Nephrology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| |
Collapse
|
17
|
Yeung J, Li W, Holinstat M. Platelet Signaling and Disease: Targeted Therapy for Thrombosis and Other Related Diseases. Pharmacol Rev 2018; 70:526-548. [PMID: 29925522 PMCID: PMC6013590 DOI: 10.1124/pr.117.014530] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Platelets are essential for clotting in the blood and maintenance of normal hemostasis. Under pathologic conditions such as atherosclerosis, vascular injury often results in hyperactive platelet activation, resulting in occlusive thrombus formation, myocardial infarction, and stroke. Recent work in the field has elucidated a number of platelet functions unique from that of maintaining hemostasis, including regulation of tumor growth and metastasis, inflammation, infection, and immune response. Traditional therapeutic targets for inhibiting platelet activation have primarily been limited to cyclooxygenase-1, integrin αIIbβ3, and the P2Y12 receptor. Recently identified signaling pathways regulating platelet function have made it possible to develop novel approaches for pharmacological intervention in the blood to limit platelet reactivity. In this review, we cover the newly discovered roles for platelets as well as their role in hemostasis and thrombosis. These new roles for platelets lend importance to the development of new therapies targeted to the platelet. Additionally, we highlight the promising receptor and enzymatic targets that may further decrease platelet activation and help to address the myriad of pathologic conditions now known to involve platelets without significant effects on hemostasis.
Collapse
Affiliation(s)
- Jennifer Yeung
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| | - Wenjie Li
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| | - Michael Holinstat
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
18
|
Protease-Activated Receptor 1 Inhibitors: Novel Antiplatelet Drugs in Prevention of Atherothrombosis. Am J Ther 2018; 24:e730-e736. [PMID: 26398717 DOI: 10.1097/mjt.0000000000000347] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Protease-activated receptor (PAR)-1 inhibitors have recently become popular in the use of atherosclerosis among clinicians. Atherosclerosis can cause cardiovascular and cerebrovascular events leading to one of the major causes of mortality worldwide. Thrombin-mediated platelets can cause atherosclerotic plaques, and these platelets are activated by thrombin through the PAR-1. Vorapaxar and atopaxar are novel antiplatelet drugs that inhibit the thrombin-induced platelet activation by antagonizing the PAR-1. The objective of this article is to review the mechanism of action of vorapaxar and atopaxar and explain the rationale for using them in atherothrombosis patients including myocardial infarction, peripheral arterial disease, and stroke.
Collapse
|
19
|
Abstract
Platelets play a key role in the pathophysiology of coronary artery disease and acute coronary syndromes. Our understanding of platelet function in thrombus formation has increased considerably, resulting in the development of clinically effective treatment strategies and identification of new targets. An underappreciated platelet function is their contribution toward acute and chronic inflammatory processes including atherogenesis. In this review, we discuss the role of platelets in atherosclerosis and thrombosis, platelet function testing, and the pharmacology of currently available antiplatelet drugs.
Collapse
|
20
|
Wong PC, Seiffert D, Bird JE, Watson CA, Bostwick JS, Giancarli M, Allegretto N, Hua J, Harden D, Guay J, Callejo M, Miller MM, Lawrence RM, Banville J, Guy J, Maxwell BD, Priestley ES, Marinier A, Wexler RR, Bouvier M, Gordon DA, Schumacher WA, Yang J. Blockade of protease-activated receptor-4 (PAR4) provides robust antithrombotic activity with low bleeding. Sci Transl Med 2018; 9:9/371/eaaf5294. [PMID: 28053157 DOI: 10.1126/scitranslmed.aaf5294] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 09/23/2016] [Indexed: 12/21/2022]
Abstract
Antiplatelet agents are proven efficacious treatments for cardiovascular and cerebrovascular diseases. However, the existing drugs are compromised by unwanted and sometimes life-threatening bleeding that limits drug usage or dosage. There is a substantial unmet medical need for an antiplatelet drug with strong efficacy and low bleeding risk. Thrombin is a potent platelet agonist that directly induces platelet activation via the G protein (heterotrimeric guanine nucleotide-binding protein)-coupled protease-activated receptors PAR1 and PAR4. A PAR1 antagonist is approved for clinical use, but its use is limited by a substantial bleeding risk. Conversely, the potential of PAR4 as an antiplatelet target has not been well characterized. Using anti-PAR4 antibodies, we demonstrated a low bleeding risk and an effective antithrombotic profile with PAR4 inhibition in guinea pigs. Subsequently, high-throughput screening and an extensive medicinal chemistry effort resulted in the discovery of BMS-986120, an orally active, selective, and reversible PAR4 antagonist. In a cynomolgus monkey arterial thrombosis model, BMS-986120 demonstrated potent and highly efficacious antithrombotic activity. BMS-986120 also exhibited a low bleeding liability and a markedly wider therapeutic window compared to the standard antiplatelet agent clopidogrel tested in the same nonhuman primate model. These preclinical findings define the biological role of PAR4 in mediating platelet aggregation. In addition, they indicate that targeting PAR4 is an attractive antiplatelet strategy with the potential to treat patients at a high risk of atherothrombosis with superior safety compared with the current standard of care.
Collapse
Affiliation(s)
- Pancras C Wong
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA.
| | - Dietmar Seiffert
- Bristol-Myers Squibb Company, Route 206 and Province Line Road, Princeton, NJ 08543, USA
| | - J Eileen Bird
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - Carol A Watson
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - Jeffrey S Bostwick
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - Mary Giancarli
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - Nick Allegretto
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - Ji Hua
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - David Harden
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - Jocelyne Guay
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Mario Callejo
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Michael M Miller
- Bristol-Myers Squibb Company, Route 206 and Province Line Road, Princeton, NJ 08543, USA
| | | | - Jacques Banville
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Julia Guy
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Brad D Maxwell
- Bristol-Myers Squibb Company, Route 206 and Province Line Road, Princeton, NJ 08543, USA
| | - E Scott Priestley
- Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, NJ 08540, USA
| | - Anne Marinier
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Ruth R Wexler
- Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, NJ 08540, USA
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec H3C 3J7, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - David A Gordon
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - William A Schumacher
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| | - Jing Yang
- Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA
| |
Collapse
|
21
|
Morikawa Y, Kato H, Kashiwagi H, Nishiura N, Akuta K, Honda S, Kanakura Y, Tomiyama Y. Protease-activated receptor-4 (PAR4) variant influences on platelet reactivity induced by PAR4-activating peptide through altered Ca 2+ mobilization and ERK phosphorylation in healthy Japanese subjects. Thromb Res 2018; 162:44-52. [DOI: 10.1016/j.thromres.2017.12.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/09/2017] [Accepted: 12/22/2017] [Indexed: 11/29/2022]
|
22
|
French SL, Hamilton JR. Drugs targeting protease-activated receptor-4 improve the anti-thrombotic therapeutic window. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:464. [PMID: 29285497 DOI: 10.21037/atm.2017.09.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Shauna L French
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - Justin R Hamilton
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| |
Collapse
|
23
|
Affiliation(s)
- Anastazia Kei
- a Department of Internal Medicine , University of Ioannina Medical School , Ioannina , Greece
| | - Moses Elisaf
- a Department of Internal Medicine , University of Ioannina Medical School , Ioannina , Greece
| |
Collapse
|
24
|
van den Eshof BL, Hoogendijk AJ, Simpson PJ, van Alphen FPJ, Zanivan S, Mertens K, Meijer AB, van den Biggelaar M. Paradigm of Biased PAR1 (Protease-Activated Receptor-1) Activation and Inhibition in Endothelial Cells Dissected by Phosphoproteomics. Arterioscler Thromb Vasc Biol 2017; 37:1891-1902. [PMID: 28818855 DOI: 10.1161/atvbaha.117.309926] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/03/2017] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Thrombin is the key serine protease of the coagulation cascade and mediates cellular responses by activation of PARs (protease-activated receptors). The predominant thrombin receptor is PAR1, and in endothelial cells (ECs), thrombin dynamically regulates a plethora of phosphorylation events. However, it has remained unclear whether thrombin signaling is exclusively mediated through PAR1. Furthermore, mechanistic insight into activation and inhibition of PAR1-mediated EC signaling is lacking. In addition, signaling networks of biased PAR1 activation after differential cleavage of the PAR1 N terminus have remained an unresolved issue. APPROACH AND RESULTS Here, we used a quantitative phosphoproteomics approach to show that classical and peptide activation of PAR1 induce highly similar signaling, that low thrombin concentrations initiate only limited phosphoregulation, and that the PAR1 inhibitors vorapaxar and parmodulin-2 demonstrate distinct antagonistic properties. Subsequent analysis of the thrombin-regulated phosphosites in the presence of PAR1 inhibitors revealed that biased activation of PAR1 is not solely linked to a specific G-protein downstream of PAR1. In addition, we showed that only the canonical thrombin PAR1 tethered ligand induces extensive early phosphoregulation in ECs. CONCLUSIONS Our study provides detailed insight in the signaling mechanisms downstream of PAR1. Our data demonstrate that thrombin-induced EC phosphoregulation is mediated exclusively through PAR1, that thrombin and thrombin-tethered ligand peptide induce similar phosphoregulation, and that only canonical PAR1 cleavage by thrombin generates a tethered ligand that potently induces early signaling. Furthermore, platelet PAR1 inhibitors directly affect EC signaling, indicating that it will be a challenge to design a PAR1 antagonist that will target only those pathways responsible for tissue pathology.
Collapse
Affiliation(s)
- Bart L van den Eshof
- From the Department Plasma Proteins (B.L.v.d.E., A.J.H., P.J.S., K.M., A.B.M., M.v.d.B.), Department of Research Facilities (F.P.J.v.A., A.B.M.), Sanquin Research, Amsterdam, The Netherlands; Tumour Microenvironment and Proteomics Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom (S.Z.); Tumour Microenvironment and Proteomics Laboratory, Institute of Cancer Sciences, University of Glasgow, United Kingdom (S.Z.); Department Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, The Netherlands (K.M., A.B.M.)
| | - Arie J Hoogendijk
- From the Department Plasma Proteins (B.L.v.d.E., A.J.H., P.J.S., K.M., A.B.M., M.v.d.B.), Department of Research Facilities (F.P.J.v.A., A.B.M.), Sanquin Research, Amsterdam, The Netherlands; Tumour Microenvironment and Proteomics Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom (S.Z.); Tumour Microenvironment and Proteomics Laboratory, Institute of Cancer Sciences, University of Glasgow, United Kingdom (S.Z.); Department Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, The Netherlands (K.M., A.B.M.)
| | - Pelle J Simpson
- From the Department Plasma Proteins (B.L.v.d.E., A.J.H., P.J.S., K.M., A.B.M., M.v.d.B.), Department of Research Facilities (F.P.J.v.A., A.B.M.), Sanquin Research, Amsterdam, The Netherlands; Tumour Microenvironment and Proteomics Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom (S.Z.); Tumour Microenvironment and Proteomics Laboratory, Institute of Cancer Sciences, University of Glasgow, United Kingdom (S.Z.); Department Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, The Netherlands (K.M., A.B.M.)
| | - Floris P J van Alphen
- From the Department Plasma Proteins (B.L.v.d.E., A.J.H., P.J.S., K.M., A.B.M., M.v.d.B.), Department of Research Facilities (F.P.J.v.A., A.B.M.), Sanquin Research, Amsterdam, The Netherlands; Tumour Microenvironment and Proteomics Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom (S.Z.); Tumour Microenvironment and Proteomics Laboratory, Institute of Cancer Sciences, University of Glasgow, United Kingdom (S.Z.); Department Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, The Netherlands (K.M., A.B.M.)
| | - Sara Zanivan
- From the Department Plasma Proteins (B.L.v.d.E., A.J.H., P.J.S., K.M., A.B.M., M.v.d.B.), Department of Research Facilities (F.P.J.v.A., A.B.M.), Sanquin Research, Amsterdam, The Netherlands; Tumour Microenvironment and Proteomics Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom (S.Z.); Tumour Microenvironment and Proteomics Laboratory, Institute of Cancer Sciences, University of Glasgow, United Kingdom (S.Z.); Department Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, The Netherlands (K.M., A.B.M.)
| | - Koen Mertens
- From the Department Plasma Proteins (B.L.v.d.E., A.J.H., P.J.S., K.M., A.B.M., M.v.d.B.), Department of Research Facilities (F.P.J.v.A., A.B.M.), Sanquin Research, Amsterdam, The Netherlands; Tumour Microenvironment and Proteomics Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom (S.Z.); Tumour Microenvironment and Proteomics Laboratory, Institute of Cancer Sciences, University of Glasgow, United Kingdom (S.Z.); Department Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, The Netherlands (K.M., A.B.M.)
| | - Alexander B Meijer
- From the Department Plasma Proteins (B.L.v.d.E., A.J.H., P.J.S., K.M., A.B.M., M.v.d.B.), Department of Research Facilities (F.P.J.v.A., A.B.M.), Sanquin Research, Amsterdam, The Netherlands; Tumour Microenvironment and Proteomics Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom (S.Z.); Tumour Microenvironment and Proteomics Laboratory, Institute of Cancer Sciences, University of Glasgow, United Kingdom (S.Z.); Department Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, The Netherlands (K.M., A.B.M.)
| | - Maartje van den Biggelaar
- From the Department Plasma Proteins (B.L.v.d.E., A.J.H., P.J.S., K.M., A.B.M., M.v.d.B.), Department of Research Facilities (F.P.J.v.A., A.B.M.), Sanquin Research, Amsterdam, The Netherlands; Tumour Microenvironment and Proteomics Laboratory, Cancer Research UK Beatson Institute, Glasgow, United Kingdom (S.Z.); Tumour Microenvironment and Proteomics Laboratory, Institute of Cancer Sciences, University of Glasgow, United Kingdom (S.Z.); Department Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, The Netherlands (K.M., A.B.M.).
| |
Collapse
|
25
|
Liu J, Sun B, Zhao X, Xing J, Gao Y, Chang W, Ji J, Zheng H, Cui C, Ji A, Lou H. Discovery of Potent Orally Active Protease-Activated Receptor 1 (PAR1) Antagonists Based on Andrographolide. J Med Chem 2017; 60:7166-7185. [DOI: 10.1021/acs.jmedchem.7b00951] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Jun Liu
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Bin Sun
- National
Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Xiaoyu Zhao
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Jie Xing
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Yanhui Gao
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Wenqiang Chang
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Hongbo Zheng
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Changyi Cui
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Aiguo Ji
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
- Weihai
International Biotechnology Research and Development Centre, Shandong University, Weihai 264209, China
| | - Hongxiang Lou
- School
of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
26
|
Flaumenhaft R, De Ceunynck K. Targeting PAR1: Now What? Trends Pharmacol Sci 2017; 38:701-716. [PMID: 28558960 PMCID: PMC5580498 DOI: 10.1016/j.tips.2017.05.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 12/30/2022]
Abstract
Protease-activated receptors (PARs) are a ubiquitously expressed class of G-protein-coupled receptors (GPCRs) that enable cells to respond to proteases in the extracellular environment in a nuanced and dynamic manner. PAR1 is the archetypal family member and has been the object of large-scale drug development programs since the 1990s. Vorapaxar and drotrecogin-alfa are approved PAR1-targeted therapeutics, but safety concerns have limited the clinical use of vorapaxar and questions regarding the efficacy of drotrecogin-alfa led to its withdrawal from the market. New understanding of mechanisms of PAR1 function, discovery of improved strategies for modifying PAR1 function, and identification of novel indications for PAR1 modulators have provided new opportunities for therapies targeting PAR1. In this review, we critically evaluate prospects for the next generation of PAR1-targeted therapeutics.
Collapse
Affiliation(s)
- Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Karen De Ceunynck
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
27
|
Abuqayyas S, Raju S, Bartholomew JR, Abu Hweij R, Mehta AC. Management of antithrombotic agents in patients undergoing flexible bronchoscopy. Eur Respir Rev 2017; 26:26/145/170001. [PMID: 28724561 DOI: 10.1183/16000617.0001-2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/09/2017] [Indexed: 12/16/2022] Open
Abstract
Bleeding is one of the most feared complications of flexible bronchoscopy. Although infrequent, it can be catastrophic and result in fatal outcomes. Compared to other endoscopic procedures, the risk of morbidity and mortality from the bleeding is increased, as even a small amount of blood can fill the tracheobronchial tree and lead to respiratory failure. Patients using antithrombotic agents (ATAs) have higher bleeding risk. A thorough understanding of the different ATAs is critical to manage patients during the peri-procedural period. A decision to stop an ATA before bronchoscopy should take into account a variety of factors, including indication for its use and the type of procedure. This article serves as a detailed review on the different ATAs, their pharmacokinetics and the pre- and post-bronchoscopy management of patients receiving these medications.
Collapse
Affiliation(s)
- Sami Abuqayyas
- Internal Medicine Institute, Cleveland Clinic, Cleveland, OH, USA.,Both authors contributed equally
| | - Shine Raju
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA.,Both authors contributed equally
| | | | - Roulan Abu Hweij
- Internal Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Atul C Mehta
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
28
|
Gryka RJ, Buckley LF, Anderson SM. Vorapaxar: The Current Role and Future Directions of a Novel Protease-Activated Receptor Antagonist for Risk Reduction in Atherosclerotic Disease. Drugs R D 2017; 17:65-72. [PMID: 28063023 PMCID: PMC5318326 DOI: 10.1007/s40268-016-0158-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Introduction Despite the current standard of care, patients with cardiovascular disease remain at a high risk for recurrent events. Inhibition of thrombin-mediated platelet activation through protease-activated receptor-1 antagonism may provide reductions in atherosclerotic disease beyond those achievable with the current standard of care. Objective Our primary objective is to evaluate the clinical literature regarding the role of vorapaxar (Zontivity™) in the reduction of cardiovascular events in patients with a history of myocardial infarction and peripheral artery disease. In particular, we focus on the potential future directions for protease-activating receptor antagonists in the treatment of a broad range of atherosclerotic diseases. Data Sources A literature search of PubMed and EBSCO was conducted to identify randomized clinical trials from August 2005 to June 2016 using the search terms: ‘vorapaxar’, ‘SCH 530348’, ‘protease-activated receptor-1 antagonist’, and ‘Zontivity™’. Bibliographies were searched and additional resources were obtained. Results Vorapaxar is a first-in-class, protease-activated receptor-1 antagonist. The Thrombin Receptor Antagonist for Clinical Event Reduction (TRACER) trial did not demonstrate a significant reduction in a broad primary composite endpoint. However, the Thrombin-Receptor Antagonist in Secondary Prevention of Atherothrombotic Ischemic Events (TRA 2°P-TIMI 50) trial examined a more traditional composite endpoint and found a significant benefit with vorapaxar. Vorapaxar significantly increased bleeding compared with standard care. Ongoing trials will help define the role of vorapaxar in patients with peripheral arterial disease, patients with diabetes mellitus, and other important subgroups. The use of multivariate modeling may enable the identification of subgroups with maximal benefit and minimal harm from vorapaxar. Conclusion Vorapaxar provides clinicians with a novel mechanism of action to further reduce the burden of ischemic heart disease. Identification of patients with a high ischemic risk and low bleeding risk would enable clinicians to maximize the utility of this unique agent. Electronic supplementary material The online version of this article (doi:10.1007/s40268-016-0158-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rebecca J Gryka
- Pharmaceutical Sciences Department, School of Pharmacy, Cedarville University, 251 North Main Street, Cedarville, OH, 45314, USA.
| | - Leo F Buckley
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Cedarville, OH, USA
| | - Sarah M Anderson
- Center for Pharmacy Care, Duquesne University, Pittsburgh, PA, USA
| |
Collapse
|
29
|
Anderson MS, Kosoglou T, Statkevich P, Li J, Rotonda J, Meehan AG, Cutler DL. No Pharmacokinetic Drug-Drug Interaction Between Prasugrel and Vorapaxar Following Multiple-Dose Administration in Healthy Volunteers. Clin Pharmacol Drug Dev 2017; 7:143-150. [PMID: 28403576 PMCID: PMC5811915 DOI: 10.1002/cpdd.354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/06/2017] [Indexed: 11/11/2022]
Abstract
Vorapaxar is a first-in-class antagonist of the protease-activated receptor-1, the primary thrombin receptor on human platelets, which mediates the downstream effects of thrombin in hemostasis and thrombosis. Prasugrel is a platelet inhibitor that acts as a P2Y12 receptor antagonist through an active metabolite, R-138727. This study investigated the interaction of these 2 platelet antagonists when coadministered. This was a randomized, open-label, multiple-dose study in 54 healthy volunteers consisting of a fixed-sequence crossover and a parallel group design. In sequence 1, 36 subjects received prasugrel 60 mg on day 1 and then prasugrel 10 mg once daily on days 2 to 7, followed by vorapaxar 40 mg and prasugrel 10 mg on day 8 and then vorapaxar 2.5 mg and prasugrel 10 mg orally once daily on days 9 to 28. In sequence 2, 18 subjects received vorapaxar 40 mg on day 1 and then vorapaxar 2.5 mg once daily on days 2 to 21. The geometric mean ratios (90% confidence intervals) for AUCτ and Cmax of coadministration/monotherapy for vorapaxar (0.93 ng·h/mL[0.85-1.02 ng·h/mL] and 0.95 ng/mL [0.86-1.05 ng/mL]) and R-138727 (0.91 ng·h/mL [0.85- 0.99 ng·h/mL] and 1.02 ng/mL [0.89-1.17 ng/mL]) were within prespecified bounds, demonstrating the absence of a pharmacokinetic interaction between vorapaxar and prasugrel. There was no specific safety or tolerability risk associated with multiple-dose coadministration of vorapaxar and prasugrel. In conclusion, in this study in healthy volunteers, there was no pharmacokinetic drug-drug interaction between vorapaxar and prasugrel. Multiple-dose coadministration of the 2 drugs was generally well tolerated.
Collapse
Affiliation(s)
| | | | | | - Jing Li
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | | |
Collapse
|
30
|
Abstract
BACKGROUND Vorapaxar, a novel antiplatelet thrombin PAR-1 inhibitor, is currently approved for post myocardial infarction and peripheral artery disease indications with concomitant use of clopidogrel and/or aspirin. The vorapaxar safety profile was acceptable. However, aside from heightened bleeding risks, excesses of solid cancers and diplopia, there were more amyotrophic lateral sclerosis (ALS) diagnoses after vorapaxar. STUDY QUESTION To assess the Food and Drug Administration (FDA) reviews on the potential association of vorapaxar with ALS. STUDY DESIGN The review the public FDA records on reported adverse events after vorapaxar. MEASURES AND OUTCOMES Incidence of ALS after vorapaxar and placebo. RESULTS The ALS risk appears very small, about 1 case per 10,000 treated subjects, but quite probable. Indeed, there were overall 2 placebo and 4 vorapaxar ALS incidences in the Phase III clinical trials. CONCLUSIONS Potential adverse association of vorapaxar with ALS risks may be related to off-target neuronal PAR receptor(s) blockade beyond platelet inhibition.
Collapse
|
31
|
Perez-Rivera JA, Monedero-Campo J, Cieza-Borrella C, Ruiz-Perez P. Pharmacokinetic drug evaluation of vorapaxar for secondary prevention after acute coronary syndrome. Expert Opin Drug Metab Toxicol 2017; 13:339-350. [PMID: 28135897 DOI: 10.1080/17425255.2017.1289175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Vorapaxar is the first protease-activated receptor-1 inhibitor approved for clinical use. Its main indication is the reduction in thrombotic cardiovascular events in patients with previous myocardial infarction or symptomatic peripheral artery disease. Areas covered: This article reviews the pharmacokinetics of vorapaxar and its potential use in secondary prevention after an acute coronary syndrome. Expert opinion: Vorapaxar inhibits platelet aggregation mediated by thrombin. This effect is carried out without affecting to coagulation parameters and bleeding times. This drug has showed a significant reduction of cardiovascular events in patients with chronic atherosclerosis but not during the admission for an acute coronary syndrome. The rate of major bleeding found in patients treated with vorapaxar in randomized trials was consistently higher than placebo in most of the analyzed subgroups. For this reason, cautious evaluation of risk-benefit profiles should be required before prescribing this drug.
Collapse
Affiliation(s)
- Jose-Angel Perez-Rivera
- a Department of Cardiology , University Hospital of Burgos , Burgos , Spain.,b Universidad Isabel I , Burgos , Spain
| | | | - Clara Cieza-Borrella
- c Division of Genetics and Epidemiology , The Institute of Cancer Research , London , UK
| | - Pablo Ruiz-Perez
- a Department of Cardiology , University Hospital of Burgos , Burgos , Spain
| |
Collapse
|
32
|
Sharma A, Helft G, Garg A, Agrawal S, Chatterjee S, Lavie CJ, Goel S, Mukherjee D, Marmur JD. Safety and efficacy of vorapaxar in secondary prevention of atherosclerotic disease: A meta-analysis of randomized control trials. Int J Cardiol 2017; 227:617-624. [DOI: 10.1016/j.ijcard.2016.10.088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 10/28/2016] [Indexed: 10/20/2022]
|
33
|
Proteinase-activated receptors (PARs) as targets for antiplatelet therapy. Biochem Soc Trans 2016; 44:606-12. [PMID: 27068977 DOI: 10.1042/bst20150282] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Indexed: 01/07/2023]
Abstract
Since the identification of the proteinase-activated receptor (PAR) family as mediators of serine protease activity in the 1990s, there has been tremendous progress in the elucidation of their pathophysiological roles. The development of drugs that target PARs has been the focus of many laboratories for the potential treatment of thrombosis, cancer and other inflammatory diseases. Understanding the mechanisms of PAR activation and G protein signalling pathways evoked in response to the growing list of endogenous proteases has yielded great insight into receptor regulation at the molecular level. This has led to the development of new selective modulators of PAR activity, particularly PAR1. The mixed success of targeting PARs has been best exemplified in the context of inhibiting PAR1 as a new antiplatelet therapy. The development of the competitive PAR1 antagonist, vorapaxar (Zontivity), has clearly shown the value in targeting PAR1 in acute coronary syndrome (ACS); however the severity of associated bleeding with this drug has limited its use in the clinic. Due to the efficacy of thrombin acting via PAR1, strategies to selectively inhibit specific PAR1-mediated G protein signalling pathways or to target the second thrombin platelet receptor, PAR4, are being devised. The rationale behind these alternative approaches is to bias downstream thrombin activity via PARs to allow for inhibition of pro-thrombotic pathways but maintain other pathways that may preserve haemostatic balance and improve bleeding profiles for widespread clinical use. This review summarizes the structural determinants that regulate PARs and the modulators of PAR activity developed to date.
Collapse
|
34
|
Tricoci P. Protease-Activated Receptor-1 Antagonists Post-Percutaneous Coronary Intervention. Interv Cardiol Clin 2016; 6:57-66. [PMID: 27886823 DOI: 10.1016/j.iccl.2016.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Thrombin is a potent platelet agonist, and protease-activated receptor-1 (PAR-1) is the main thrombin receptor in human platelets and thrombin. PAR-1 antagonism has attracted interest as a potential therapeutic target to reduce atherothrombotic events in patients with atherosclerotic disease, especially coronary artery disease. In this review, the author describes the rationale of PAR-1 antagonism for the reduction of atherothrombotic events and reviews the key phase 3 trial results, with special attention to analyses in percutaneous coronary intervention patients.
Collapse
Affiliation(s)
- Pierluigi Tricoci
- Division of Cardiology, Duke Clinical Research Institute, Duke University Medical Center, 2400 Pratt Street, 0311 Terrace Level, Box 3850 DUMC, Durham, NC 27705, USA.
| |
Collapse
|
35
|
Hamilton JR, Trejo J. Challenges and Opportunities in Protease-Activated Receptor Drug Development. Annu Rev Pharmacol Toxicol 2016; 57:349-373. [PMID: 27618736 DOI: 10.1146/annurev-pharmtox-011613-140016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protease-activated receptors (PARs) are a unique class of G protein-coupled receptors (GPCRs) that transduce cellular responses to extracellular proteases. PARs have important functions in the vasculature, inflammation, and cancer and are important drug targets. A unique feature of PARs is their irreversible proteolytic mechanism of activation that results in the generation of a tethered ligand that cannot diffuse away. Despite the fact that GPCRs have proved to be the most successful class of druggable targets, the development of agents that target PARs specifically has been challenging. As a consequence, researchers have taken a remarkable diversity of approaches to develop pharmacological entities that modulate PAR function. Here, we present an overview of the diversity of therapeutic agents that have been developed against PARs. We further discuss PAR biased signaling and the influence of receptor compartmentalization, posttranslational modifications, and dimerization, which are important considerations for drug development.
Collapse
Affiliation(s)
- Justin R Hamilton
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|
36
|
Park CM, Baek S, Kim S, Song JH, Lee S, Kim M. Trans-fused 5-[(tert-Butoxtycarbonyl)amino]octahydroindenes as a protease activated receptor-1 (PAR1) antagonist. Arch Pharm Res 2016; 39:1275-95. [PMID: 27406140 DOI: 10.1007/s12272-016-0788-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/28/2016] [Indexed: 12/01/2022]
Abstract
Protease activated receptor 1 (PAR1) has been considered as a promising antiplatelet target to prevent thrombotic cardiovascular events in patients with prior myocardial infarction or peripheral arterial diseases. Previously, we found a series of octahydroindene analogues to have high potency on PAR1 and no significant cytotoxicity but poor metabolic stability in human and rat liver microsomes. We designed and synthesized substituted analogues of octahydroindenes at C5 or C6 aiming to improvement of metabolic stability, and identified that trans-fused 5-[(tert-butoxtycarbonyl)amino]octahydroindene analogues showed improved metabolic stability with maintaining good activity on PAR1. Especially, 2-methanesulfonate 57 (IC50 = 0.006 μM; R50 = 126.3 min in human, 83.3 min in rat), sulfamate 58 (IC50 = 0.020 μM; R50 = 52.8 min in human, 106.0 min in rat), and N-(cyclopropyl)methylsufonamide 63 (IC50 = 0.010 μM; R50 = 51.4 min in human, 90.5 min in rat) exhibited excellent activity and metabolic stability both on human and rat liver microsomes, comparable to those obtained for varapaxar (IC50 = 0.0015 μM; R50 = 83.2 min in human, 32.4 min in rat). Additionally, these compounds (57, 58, and 63) represented significant efficacy (IC50 = 0.0022, 0.0062, and 0.015 μM, each) in human washed platelet aggregation (WPA) assay without cytotoxicity and CYP3A4 inhibitory activity.
Collapse
Affiliation(s)
- Chul Min Park
- Research Center for Medicinal Chemistry, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong, Daejeon, 305-343, Republic of Korea
| | - Songhyun Baek
- Research Center for Medicinal Chemistry, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong, Daejeon, 305-343, Republic of Korea.,Korea University of Science and Technology, 141 Gajeong-ro, Yuseong, Daejeon, 305-343, Republic of Korea
| | - Seongwoo Kim
- Research Center for Medicinal Chemistry, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong, Daejeon, 305-343, Republic of Korea.,Department of Chemistry, Chungbuk National University, Chungdae-ro, Seowon-gu, Cheongju-si, 28644, Chungbuk, Republic of Korea
| | - Jong-Hwan Song
- Research Center for Medicinal Chemistry, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong, Daejeon, 305-343, Republic of Korea
| | - Sunkyung Lee
- Research Center for Medicinal Chemistry, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong, Daejeon, 305-343, Republic of Korea. .,Korea University of Science and Technology, 141 Gajeong-ro, Yuseong, Daejeon, 305-343, Republic of Korea.
| | - Min Kim
- Department of Chemistry, Chungbuk National University, Chungdae-ro, Seowon-gu, Cheongju-si, 28644, Chungbuk, Republic of Korea
| |
Collapse
|
37
|
Ungar L, Rodriguez F, Mahaffey KW. Vorapaxar: emerging evidence and clinical questions in a new era of PAR-1 inhibition. Coron Artery Dis 2016; 27:604-15. [PMID: 27398626 DOI: 10.1097/mca.0000000000000409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Despite the use of therapies recommended in practice guidelines for secondary prevention in patients with atherosclerotic coronary artery disease, the residual risk for cardiovascular events remains high. Some of the residual risk is believed to result from incomplete platelet inhibition with current therapy. Vorapaxar is a first-in-class, novel antiplatelet agent that acts by antagonizing the PAR-1 receptor, inhibiting thrombin-mediated platelet activation. Vorapaxar was recently approved by the Food and Drug Administration for secondary prevention of cardiovascular events in patients with a history of myocardial infarction or peripheral artery disease who do not have a history of transient ischemic attack or stroke. We review the data from two key phase III cardiovascular outcome trials with vorapaxar: TRACER and TRA 2P-TIMI 50. We will focus on identifying the key patient populations that should be identified for treatment, highlight practical clinical issues when prescribing vorapaxar, and review unanswered questions. Vorapaxar should be considered in patients at high risk for recurrent ischemic events and low risk of bleeding.
Collapse
Affiliation(s)
- Leo Ungar
- aDepartment of Medicine bDivision of Cardiovascular Medicine and Stanford Center for Clinical Research, Stanford University, Stanford, California, USA
| | | | | |
Collapse
|
38
|
Pharmacology: a new bleeding issue. Br Dent J 2016; 217:54-6. [PMID: 25060431 DOI: 10.1038/sj.bdj.2014.602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
39
|
Kraft WK, Gilmartin JH, Chappell DL, Gheyas F, Walker BM, Nagalla S, Naik UP, Horrow JC, Wrishko RE, Zhang S, Anderson MS. Effect of Vorapaxar Alone and in Combination with Aspirin on Bleeding Time and Platelet Aggregation in Healthy Adult Subjects. Clin Transl Sci 2016; 9:221-7. [PMID: 27304196 PMCID: PMC5351335 DOI: 10.1111/cts.12405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/17/2016] [Accepted: 05/12/2016] [Indexed: 11/25/2022] Open
Abstract
The effect of the protease‐activated receptor‐1 (PAR‐1) antagonist vorapaxar on human bleeding time is not known. This was a randomized, two‐period, open‐label trial in healthy men (n = 31) and women (n = 5). In period 1, subjects received 81 mg aspirin q.d. or a vorapaxar regimen achieving steady‐state plasma concentrations equivalent to chronic 2.5 mg q.d. doses, for 7 days. In period 2, each group added 7 days of the therapy alternate to that of period 1 without washout. Bleeding time and platelet aggregation using arachidonic acid, ADP, and TRAP agonists were assessed. Bleeding time geometric mean ratio (90% CI) for vorapaxar/baseline was 1.01 (0.88–1.15), aspirin/baseline was 1.32 (1.15–1.51), vorapaxar + aspirin/vorapaxar was 1.47 (1.26–1.70), and vorapaxar + aspirin/aspirin was 1.12 (0.96–1.30). Unlike aspirin, vorapaxar did not prolong bleeding time compared with baseline. Bleeding time following administration of vorapaxar with aspirin was similar to that following aspirin alone.
Collapse
Affiliation(s)
- W K Kraft
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | - F Gheyas
- Merck & Co, Kenilworth, New Jersey, USA
| | | | - S Nagalla
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - U P Naik
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | - S Zhang
- Merck & Co, Kenilworth, New Jersey, USA
| | | |
Collapse
|
40
|
Abstract
Antiplatelet therapy reduces the risks for cardiovascular morbidity and mortality in patients with atherosclerotic disease, and it is also beneficial in managing peripheral arterial disease (PAD). These agents work through various therapeutic pathways to achieve antithrombotic effects. Although single- or two-drug regimens have been deployed to prevent vascular events, approximately 10% of the patients with acute coronary syndrome remain at risk for recurrent thrombotic events and may need a more aggressive preventative strategy. Vorapaxar offers a unique mechanism for platelet inhibition via the antagonism of protease-activated receptor-1. It is approved for the reduction of thrombotic cardiovascular events in patients with a history of myocardial infarction (MI) or PAD. This new drug approval was mainly based on the results from subgroup analyses from a large landmark trial (Thrombin Receptor Antagonist in Secondary Prevention of Atherothrombotic Ischemic Events-Thrombolysis in Myocardial Infarction 50), which found that vorapaxar reduces the rate of the combined end point of cardiovascular death, MI, stroke, and urgent coronary revascularization when used in addition to aspirin and/or clopidogrel in patients without a history of stroke. In this study, vorapaxar was discontinued in patients with a history of stroke due to excessive risk for intracranial hemorrhage after 2 years of therapy. As an adjunctive therapy to standard regimens, vorapaxar provides a greater net clinical benefit in MI patients who are at a lower risk for bleeding. In patients with PAD, it reduces the rates of recurrent acute limb ischemia with rehospitalization or peripheral revascularization. The most concerning adverse effect is bleeding. Vorapaxar should not be used in patients with a history of stroke, transient ischemic attack, intracranial hemorrhage, or active pathological bleeding. The risks and benefits of adding vorapaxar to intensify antiplatelet regimens should be assessed in individual patients to aim for additional therapeutic outcomes with minimal bleeding risks.
Collapse
|
41
|
Norman JE, Cunningham MR, Jones ML, Walker ME, Westbury SK, Sessions RB, Mundell SJ, Mumford AD. Protease-Activated Receptor 4 Variant p.Tyr157Cys Reduces Platelet Functional Responses and Alters Receptor Trafficking. Arterioscler Thromb Vasc Biol 2016; 36:952-60. [DOI: 10.1161/atvbaha.115.307102] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/22/2016] [Indexed: 01/05/2023]
Abstract
Objective—
Protease-activated receptor 4 (PAR4) is a key regulator of platelet reactivity and is encoded by
F2RL3
, which has abundant rare missense variants. We aimed to provide proof of principle that rare
F2LR3
variants potentially affect platelet reactivity and responsiveness to PAR1 antagonist drugs and to explore underlying molecular mechanisms.
Approach and Results—
We identified 6 rare
F2RL3
missense variants in 236 cardiac patients, of which the variant causing a tyrosine 157 to cysteine substitution (Y157C) was predicted computationally to have the greatest effect on PAR4 structure. Y157C platelets from 3 cases showed reduced responses to PAR4-activating peptide and to α-thrombin compared with controls, but no reduction in responses to PAR1-activating peptide. Pretreatment with the PAR1 antagonist vorapaxar caused lower residual α-thrombin responses in Y157C platelets than in controls, indicating greater platelet inhibition. HEK293 cells transfected with a PAR4 Y157C expression construct had reduced PAR4 functional responses, unchanged total PAR4 expression but reduced surface expression. PAR4 Y157C was partially retained in the endoplasmic reticulum and displayed an expression pattern consistent with defective
N
-glycosylation. Mutagenesis of Y322, which is the putative hydrogen bond partner of Y157, also reduced PAR4 surface expression in HEK293 cells.
Conclusions—
Reduced PAR4 responses associated with Y157C result from aberrant anterograde surface receptor trafficking, in part, because of disrupted intramolecular hydrogen bonding. Characterization of PAR4 Y157C establishes that rare
F2RL3
variants have the potential to markedly alter platelet PAR4 reactivity particularly after exposure to therapeutic PAR1 antagonists.
Collapse
Affiliation(s)
- Jane E. Norman
- From the School of Clinical Sciences (J.E.N., M.E.W., S.K.W., A.D.M.), School of Cellular and Molecular Medicine (M.L.J., A.D.M.), School of Biochemistry (R.B.S.), and School of Physiology and Pharmacology (S.J.M.), University of Bristol, Bristol, United Kingdom; and Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom (M.R.C.)
| | - Margaret R. Cunningham
- From the School of Clinical Sciences (J.E.N., M.E.W., S.K.W., A.D.M.), School of Cellular and Molecular Medicine (M.L.J., A.D.M.), School of Biochemistry (R.B.S.), and School of Physiology and Pharmacology (S.J.M.), University of Bristol, Bristol, United Kingdom; and Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom (M.R.C.)
| | - Matthew L. Jones
- From the School of Clinical Sciences (J.E.N., M.E.W., S.K.W., A.D.M.), School of Cellular and Molecular Medicine (M.L.J., A.D.M.), School of Biochemistry (R.B.S.), and School of Physiology and Pharmacology (S.J.M.), University of Bristol, Bristol, United Kingdom; and Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom (M.R.C.)
| | - Mary E. Walker
- From the School of Clinical Sciences (J.E.N., M.E.W., S.K.W., A.D.M.), School of Cellular and Molecular Medicine (M.L.J., A.D.M.), School of Biochemistry (R.B.S.), and School of Physiology and Pharmacology (S.J.M.), University of Bristol, Bristol, United Kingdom; and Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom (M.R.C.)
| | - Sarah K. Westbury
- From the School of Clinical Sciences (J.E.N., M.E.W., S.K.W., A.D.M.), School of Cellular and Molecular Medicine (M.L.J., A.D.M.), School of Biochemistry (R.B.S.), and School of Physiology and Pharmacology (S.J.M.), University of Bristol, Bristol, United Kingdom; and Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom (M.R.C.)
| | - Richard B. Sessions
- From the School of Clinical Sciences (J.E.N., M.E.W., S.K.W., A.D.M.), School of Cellular and Molecular Medicine (M.L.J., A.D.M.), School of Biochemistry (R.B.S.), and School of Physiology and Pharmacology (S.J.M.), University of Bristol, Bristol, United Kingdom; and Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom (M.R.C.)
| | - Stuart J. Mundell
- From the School of Clinical Sciences (J.E.N., M.E.W., S.K.W., A.D.M.), School of Cellular and Molecular Medicine (M.L.J., A.D.M.), School of Biochemistry (R.B.S.), and School of Physiology and Pharmacology (S.J.M.), University of Bristol, Bristol, United Kingdom; and Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom (M.R.C.)
| | - Andrew D. Mumford
- From the School of Clinical Sciences (J.E.N., M.E.W., S.K.W., A.D.M.), School of Cellular and Molecular Medicine (M.L.J., A.D.M.), School of Biochemistry (R.B.S.), and School of Physiology and Pharmacology (S.J.M.), University of Bristol, Bristol, United Kingdom; and Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom (M.R.C.)
| |
Collapse
|
42
|
Behm MO, Kosoglou T, Miltenburg AMM, Li J, Statkevich P, Johnson-Levonas AO, Martinho M, Fackler P. The Absence of a Clinically Significant Effect of Food on the Single Dose Pharmacokinetics of Vorapaxar, a PAR-1 Antagonist, in Healthy Adult Subjects. Clin Pharmacol Drug Dev 2016; 2:310-5. [PMID: 27121935 DOI: 10.1002/cpdd.38] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 03/11/2013] [Indexed: 11/06/2022]
Abstract
In this open-label, randomized, 2-period crossover study, 16 healthy subjects received a single oral 2.5-mg dose of vorapaxar in the fed (i.e., standardized high-fat breakfast) and fasted (i.e., an overnight fast) state with a 6-week washout. Plasma samples for vorapaxar assay were obtained pre-dose and up to 72 hours post-dose. Least squares (LS) geometric mean AUC0-72 hr and Cmax were analyzed by ANOVA. If 90% confidence intervals (CI) for the geometric mean ratios (GMRs; fed/fasted) of AUC0-72 hr and Cmax were within the 50-200% range, then food was deemed not to have a clinically important effect. The LS geometric mean (90% CI) AUC0-72 hr and Cmax of vorapaxar in the fasted state were 314 (284-348) ng hr/mL and 23.4 (20.7-26.4) ng/mL, respectively. The GMRs (fed/fasted) and 90% CIs for AUC0-72 hr and Cmax were 96.9 (92.2-102) and 79.1 (67.6-92.5), respectively. Vorapaxar was generally safe and well tolerated in the presence and absence of food. Concomitant food decreased the rate (i.e., 21% reduction in Cmax and 45-min delay in Tmax ) with no effect on the extent of vorapaxar absorption when administered as a single 2.5-mg dose. Thus, vorapaxar can be administered without regard to food.
Collapse
Affiliation(s)
| | | | | | - Jing Li
- Merck & Co., Inc., Whitehouse Station, NJ, USA
| | | | | | | | | |
Collapse
|
43
|
Kehinde O, Kunle R. Vorapaxar: A novel agent to be considered in the secondary prevention of myocardial infarction. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2016; 8:98-105. [PMID: 27134460 PMCID: PMC4832913 DOI: 10.4103/0975-7406.171690] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 08/21/2015] [Accepted: 09/17/2015] [Indexed: 11/29/2022] Open
Abstract
Patients receiving therapy for the secondary prevention of myocardial infarction (MI) are still at high risk of a major cardiovascular event or death despite the use of currently available treatment strategy. Vorapaxar, an oral protease-activated receptor antagonist, is a novel antiplatelet drug that has been recently approved to provide further risk reduction. The results of two Phase III trials (thrombin receptor antagonists for clinical event reduction and the TRA 2°P-TIMI 50) have showed that vorapaxar, in addition to standard of care therapy, has the potential to provide further risk reduction in patients with prior MI. A search was made on PubMed on articles related to clinical trials and clinical consideration with the use of vorapaxar. This review article summarizes the results of Phase II trials, Phase III trials, subgroup analysis, precautions, and drug interaction with the use of vorapaxar.
Collapse
Affiliation(s)
- Obamiro Kehinde
- Division of Pharmacy, School of Medicine, University of Tasmania, Australia
| | - Rotimi Kunle
- Department of Pharmacology, College of Medicine, University of Lagos, Nigeria
| |
Collapse
|
44
|
The Ratio of ADP- to TRAP-Induced Platelet Aggregation Quantifies P2Y12-Dependent Platelet Inhibition Independently of the Platelet Count. PLoS One 2016; 11:e0149053. [PMID: 26885820 PMCID: PMC4757031 DOI: 10.1371/journal.pone.0149053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/25/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study aimed to assess the association of clinical factors with P2Y12-dependent platelet inhibition as monitored by the ratio of ADP- to TRAP-induced platelet aggregation and conventional ADP-induced aggregation, respectively. BACKGROUND Controversial findings to identify and overcome high platelet reactivity (HPR) after coronary stent-implantation and to improve clinical outcome by tailored anti-platelet therapy exist. Monitoring anti-platelet therapy ex vivo underlies several confounding parameters causing that ex vivo platelet aggregation might not reflect in vivo platelet inhibition. METHODS In a single centre observational study, multiple electrode aggregometry was performed in whole blood of patients after recent coronary stent-implantation. Relative ADP-induced aggregation (r-ADP-agg) was defined as the ratio of ADP- to TRAP- induced aggregation reflecting the individual degree of P2Y12-mediated platelet reactivity. RESULTS Platelet aggregation was assessed in 359 patients. Means (± SD) of TRAP-, ADP-induced aggregation and r-ADP-agg were 794 ± 239 AU*min, 297 ± 153 AU*min and 37 ± 14%, respectively. While ADP- and TRAP-induced platelet aggregation correlated significantly with platelet count (ADP: r = 0.302; p<0.001; TRAP: r = 0.509 p<0.001), r-ADP-agg values did not (r = -0.003; p = 0.960). These findings were unaltered in multivariate analyses adjusting for a range of factors potentially influencing platelet aggregation. The presence of an acute coronary syndrome and body weight were found to correlate with both ADP-induced platelet aggregation and r-ADP-agg. CONCLUSION The ratio of ADP- to TRAP-induced platelet aggregation quantifies P2Y12-dependent platelet inhibition independently of the platelet count in contrast to conventional ADP-induced aggregation. Furthermore, r-ADP-agg was associated with the presence of an acute coronary syndrome and body weight as well as ADP-induced aggregation. Thus, the r-ADP-agg is a more valid reflecting platelet aggregation and potentially prognosis after coronary stent-implantation in P2Y12-mediated HPR than conventional ADP-induced platelet aggregation.
Collapse
|
45
|
Serebruany VL, Choi SY, Kim MH. The FDA review on data quality and conduct in vorapaxar trials: Much better than in PLATO, but still not perfect. Int J Cardiol 2016; 205:13-16. [PMID: 26709134 DOI: 10.1016/j.ijcard.2015.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/01/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND/AIMS Vorapaxar, a novel antiplatelet thrombin PAR-1 inhibitor, has been evaluated in TRA2P and TRACER trials. The drug is currently approved for post-myocardial infarction and peripheral artery disease indications with concomitant use of clopidogrel and/or aspirin. The FDA ruled that the overall vorapaxar data quality was acceptable, but conducted the sensitivity analyses for potential censoring. This was unusual, intriguing, and directly related to the challenged quality of ticagrelor dataset in PLATO in the previous New Drug Application for an oral antiplatelet agent submitted to the same Agency. METHODS Hence, we compared the FDA-confirmed evidence of conduct and data quality in vorapaxar (TRA2P, and TRACER) with those of ticagrelor (PLATO) trials. RESULTS The FDA provides a detailed report on information censoring, and follow-up completeness for 3 trials. TRA2P and TRACER used independent CRO for site monitoring, exhibit no heterogeneity in trial results dependent on geography, and consistent adjudication results with much less censoring than in PLATO. CONCLUSION The data quality and trial conduct in vorapaxar trials were better than testing ticagrelor in PLATO, however, there is still some room for improvement especially with regard to follow-up completeness, and less information censoring.
Collapse
Affiliation(s)
| | - Sun Young Choi
- Clinical Trial Center, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Moo Hyun Kim
- Clinical Trial Center, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
46
|
Schaff M, Gachet C, Mangin PH. [Anti-platelets without a bleeding risk: novel targets and strategies]. Biol Aujourdhui 2016; 209:211-28. [PMID: 26820829 DOI: 10.1051/jbio/2015023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Indexed: 01/29/2023]
Abstract
Anti-platelet agents such as aspirin, clopidogrel and antagonists of integrin αIIbβ3 allowed to efficiently reduce morbidity and mortality associated with arterial thrombosis. A major limit of these drugs is that they increase the risk of bleeding. During the last few years, several innovative anti-thrombotic strategies with a potentially low bleeding risk were proposed. These approaches target the collagen receptor glycoprotein (GP) VI, the GPIb/von Willebrand factor axis, the thrombin receptor PAR-1, the activated form of integrin αIIbβ3 or the ADP receptor P2Y1. While an antagonist of PAR-1 was recently marketed, the clinical proofs of the efficiency and safety of the other agents remain to be established. This review evaluates these new anti-platelet approaches toward safer anti-thrombotic therapies.
Collapse
Affiliation(s)
- Mathieu Schaff
- Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australie
| | - Christian Gachet
- UMR_S949, INSERM, Etablissement Français du Sang (EFS)-Alsace, Université de Strasbourg, Strasbourg, France
| | - Pierre Henri Mangin
- UMR_S949, INSERM, Etablissement Français du Sang (EFS)-Alsace, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
47
|
Vorapaxar: a review of its use in the long-term secondary prevention of atherothrombotic events. Drugs 2016; 75:797-808. [PMID: 25895464 DOI: 10.1007/s40265-015-0387-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Vorapaxar (Zontivity®) is a first-in-class, potent and orally-active protease-activated receptor 1 (PAR-1) antagonist that blocks thrombin-mediated platelet activation without interfering with thrombin-mediated fibrin deposition. The long-term efficacy of once-daily vorapaxar added to standard antiplatelet therapy (aspirin with or without clopidogrel) in the secondary prevention of atherothrombotic events in patients with a history of myocardial infarction (MI), ischaemic stroke or peripheral arterial disease was investigated in the large, multinational TRA 2°P-TIMI 50 trial. Compared with placebo, vorapaxar significantly reduced the risk of the composite endpoints of cardiovascular (CV) death, MI or stroke, and CV death, MI, stroke or urgent coronary revascularization in the overall trial population. Vorapaxar also significantly reduced the risk of these composite endpoints in the subgroup of patients with prior MI (the largest qualifying disease cohort) and the subset of post-MI patients with no history of stroke or transient ischaemic attack (TIA). Vorapaxar significantly increased the risk of GUSTO moderate and/or severe bleeding in the overall trial population and all key subgroups (including post-MI patients with no history of stroke or TIA). Vorapaxar also significantly increased the risk of intracranial haemorrhage (ICH) in the overall trial population and the subgroup of patients with prior stroke, but not the subgroup of post-MI patients or the subset of post-MI patients with no history of stroke or TIA. Based on these results, vorapaxar has been approved in the EU as an adjunctive treatment for the secondary prevention of atherothrombotic events in patients with prior MI who do not have a history of stroke, TIA or ICH.
Collapse
|
48
|
Serebruany VL, Fortmann SD. The FDA report on vorapaxar in the elderly: A convoluted dilemma. Int J Cardiol 2015; 201:601-3. [DOI: 10.1016/j.ijcard.2015.08.177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/21/2015] [Indexed: 10/23/2022]
|
49
|
Abstract
Dual antiplatelet therapy with aspirin, a platelet cyclooxygenase-1 inhibitor and P2Y12 receptor blockers, remains the major drug strategy to prevent ischemic event occurrence in patients with acute coronary syndromes and in patients undergoing coronary stenting, but there some limitations that can be overcome by targeting novel targets. Unlike direct thrombin inhibitors that bind directly to thrombin, targeting the platelet thrombin receptor, protease activated receptor (PAR)-1, may offer a better choice for the attenuation of atherosclerosis progression, thrombus-mediated ischemic events and restenosis without interfering with primary hemostasis. Vorapaxar - a synthetic analogue of himbacine, is a high affinity and highly selective PAR-1 antagonist that can effectively inhibit thrombin-induced platelet aggregation. In the TRACER trial, the addition of vorapaxar to standard therapy in patients with non-stent thrombosis-elevation- acute coronary syndromes did not significantly reduce the primary composite end point occurrence of cardiovascular (CV) death, myocardial infarction (MI), stroke, hospitalization for ischemia, or urgent revascularization, but significantly increased the GUSTO moderate and severe bleeding (p < 0.001) and intracranial hemorrhage (ICH). In the TRA 2°P-TIMI 50 trial, in patients with a history of MI and peripheral arterial disease (PAD) (67% of the total population), the end point of CV death, MI, or stroke was significantly (20%) reduced with vorapaxar whereas GUSTO moderate or severe bleeding was increased (1.5-fold), but not ICH or fatal bleeding and the net clinical outcome favoring the vorapaxar therapy. Based on these favorable results, the FDA approved vorapaxar for the reduction of thrombotic cardiovascular events in patients with prior MI or with PAD for long term therapy. A careful patient selection is needed to balance efficacy versus safety. At this time, patients with high risk for recurrent ischemic event occurrence such as patients with diabetes mellitus and previous MI can be safely treated with vorapaxar for long-term therapy.
Collapse
Affiliation(s)
- Udaya S Tantry
- a "Inova Center for Thrombosis Research and Drug Development" Inova Heart and Vascular Institute , Fairfax , VA , USA
| | - Fang Liu
- a "Inova Center for Thrombosis Research and Drug Development" Inova Heart and Vascular Institute , Fairfax , VA , USA.,b Department of Neurology , Beijing Hospital , Beijing 100730 , China
| | - Gailing Chen
- a "Inova Center for Thrombosis Research and Drug Development" Inova Heart and Vascular Institute , Fairfax , VA , USA.,c Department of Cardiology , China-Japan Friendship Hospital , Beijing 100029 , China
| | - Paul A Gurbel
- a "Inova Center for Thrombosis Research and Drug Development" Inova Heart and Vascular Institute , Fairfax , VA , USA
| |
Collapse
|
50
|
Serebruany VL, Fortmann SD, Kim MH. Should the Dose of Antiplatelet Drugs Be Adjusted for Body Weight? The Example of Vorapaxar. Cardiology 2015; 133:69-72. [PMID: 26488178 DOI: 10.1159/000440798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/28/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND In contrast to the vast majority of pharmaceuticals on the market, antiplatelet agents are widely prescribed in a uniform, 'one size fits all' manner, without conventional dose adjustments. However, strong evidence yielded from clinical trials repeatedly suggests that patients with a low body weight (LBW), the elderly and those with renal or hepatic impairment may benefit from reduced doses, while younger, heavier patients, males and diabetics may benefit from a dose escalation. Vorapaxar, a thrombin protease-activated receptor-1 inhibitor, has been tested in the TRA2P and TRACER clinical trials, but its efficacy and safety in patients with a LBW is unclear. OBJECTIVE To determine the impact of LBW on primary end point rates (PER) and bleeding risk after vorapaxar, as yielded from the TRA2P and TRACER secondary FDA review. RESULTS The LBW (<60 kg) groups in TRA2P (n = 1,852; 7%) and TRACER (n = 1,046; 8%) were small. However, the PER repeatedly suggested inferiority of vorapaxar over placebo in both the successful TRA2P study (10.6 vs. 8.4%; p = 0.012) and the failed TRACER study (19.3 vs. 18.2%; p = not significant). In TRA2P, the PER monotonically escalated with increasing weight for placebo, while those in the vorapaxar arm formed a flat U- or J-shaped distribution across the weight quintiles. In TRACER, the PER by weight quintile appear much higher, but also more random than in TRA2P. The bleeding rates in TRA2P were higher for the 2 lowest-weight quintiles with both placebo and vorapaxar. In TRACER, bleeding rates were more than doubled when compared to TRA2P, and they varied little by weight quintile, with a slight decrease for the heaviest patients in the placebo population and being the highest in the 2 lowest-weight quintiles after vorapaxar. CONCLUSION The FDA analyses revealed no definite proof that LBW is associated with reduced efficacy of vorapaxar. While these data are striking, they can be explained by better outcomes in LBW placebo patients already sufficiently treated with dual-antiplatelet therapy. In contrast to efficacy, both TRA2P and TRACER definitely suggest that bleeding rates after vorapaxar are higher in patients with LBW. Dose adjustment for antiplatelet agents may soon become a reality.
Collapse
Affiliation(s)
- Victor L Serebruany
- HeartDrug™ Research Laboratories, Johns Hopkins University, Towson, Md., USA
| | | | | |
Collapse
|