1
|
Goel A, Tathireddy H, Wang SH, Vu HH, Puy C, Hinds MT, Zonies D, McCarty OJ, Shatzel JJ. Targeting the Contact Pathway of Coagulation for the Prevention and Management of Medical Device-Associated Thrombosis. Semin Thromb Hemost 2024; 50:989-997. [PMID: 37044117 PMCID: PMC11069398 DOI: 10.1055/s-0043-57011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Hemorrhage remains a major complication of anticoagulants, with bleeding leading to serious and even life-threatening outcomes in rare settings. Currently available anticoagulants target either multiple coagulation factors or specifically coagulation factor (F) Xa or thrombin; however, inhibiting these pathways universally impairs hemostasis. Bleeding complications are especially salient in the medically complex population who benefit from medical devices. Extracorporeal devices-such as extracorporeal membrane oxygenation, hemodialysis, and cardiac bypass-require anticoagulation for optimal use. Nonetheless, bleeding complications are common, and with certain devices, highly morbid. Likewise, pharmacologic prophylaxis to prevent thrombosis is not commonly used with many medical devices like central venous catheters due to high rates of bleeding. The contact pathway members FXI, FXII, and prekallikrein serve as a nexus, connecting biomaterial surface-mediated thrombin generation and inflammation, and may represent safe, druggable targets to improve medical device hemocompatibility and thrombogenicity. Recent in vivo and clinical data suggest that selectively targeting the contact pathway of coagulation through the inhibition of FXI and FXII can reduce the incidence of medical device-associated thrombotic events, and potentially systemic inflammation, without impairing hemostasis. In the following review, we will outline the current in vivo and clinical data encompassing the mechanism of action of drugs targeting the contact pathway. This new class of inhibitors has the potential to herald a new era of effective and low-risk anticoagulation for the management of patients requiring the use of medical devices.
Collapse
Affiliation(s)
- Abhishek Goel
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Harsha Tathireddy
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Si-Han Wang
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Helen H. Vu
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - David Zonies
- Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Owen J.T. McCarty
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Joseph J. Shatzel
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
2
|
Mäder J, Rolling CC, Voigtländer M, Schulenkorf A, Lehr C, Regenhardt J, Bokemeyer C, Beckmann L, Langer F. Effect of factor XI inhibition on tumor cell-induced coagulation activation. J Thromb Haemost 2024; 22:199-212. [PMID: 37751848 DOI: 10.1016/j.jtha.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/29/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Cancer-associated thrombosis is a frequent complication in patients with malignancies. While factor XI (FXI)/FXIa inhibition is efficacious in preventing postoperative venous thromboembolism, its role in tumor cell-induced coagulation is less defined. OBJECTIVES We thus aimed to provide mechanistic insights into FXI/FXIa inhibition in tumor cell-induced coagulation activation. METHODS Procoagulant activity (PCA) of 4 different tissue factor (TF) expressing tumor cell lines was analyzed by single-stage clotting and thrombin generation assay in the presence of a FXIa inhibitor, BMS-262084 (BMS), an inhibitory FXI antibody (anti-FXI), or peak and trough concentrations of rivaroxaban or tinzaparin. Further, tumor cell-induced platelet aggregation was recorded. Recombinant human TF served as positive control. RESULTS Although BMS and anti-FXI potently inhibited FXIa amidolytic activity, both inhibitors efficiently mitigated recombinant human TF- and tumor cell-induced fibrin clot formation and platelet aggregation only in the presence of low TF PCA. The anticoagulant effects showed an inverse correlation with the magnitude of cellular TF PCA expression. Similarly, BMS markedly interfered with tumor cell-induced thrombin generation, with the most prominent effects on peak and total thrombin. In addition, anticoagulant effects of FXIa inhibition by 10 μM BMS were in a similar range to those obtained by 600 nM rivaroxaban and 1.6 μM tinzaparin at low TF PCA levels. However, rivaroxaban and tinzaparin also exerted marked anticoagulant activity at high TF PCA levels. CONCLUSION Our findings indicate that FXI/FXIa inhibition interferes with tumor cell-induced coagulation activation only at low TF PCA expression levels, a finding with potential implications for future in vivo studies.
Collapse
Affiliation(s)
- Jonathan Mäder
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Christina C Rolling
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Minna Voigtländer
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Anita Schulenkorf
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Carina Lehr
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Judith Regenhardt
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Lennart Beckmann
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Florian Langer
- II. Medizinische Klinik und Poliklinik, Universitätsklinikum Eppendorf, Hamburg, Germany.
| |
Collapse
|
3
|
Li Y, Di C, Song S, Zhang Y, Lu Y, Liao J, Lei B, Zhong J, Guo K, Zhang N, Su S. Choroid plexus mast cells drive tumor-associated hydrocephalus. Cell 2023; 186:5719-5738.e28. [PMID: 38056463 DOI: 10.1016/j.cell.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/04/2023] [Accepted: 11/01/2023] [Indexed: 12/08/2023]
Abstract
Tumor-associated hydrocephalus (TAH) is a common and lethal complication of brain metastases. Although other factors beyond mechanical obstructions have been suggested, the exact mechanisms are unknown. Using single-nucleus RNA sequencing and spatial transcriptomics, we find that a distinct population of mast cells locate in the choroid plexus and dramatically increase during TAH. Genetic fate tracing and intracranial mast-cell-specific tryptase knockout showed that choroid plexus mast cells (CPMCs) disrupt cilia of choroid plexus epithelia via the tryptase-PAR2-FoxJ1 pathway and consequently increase cerebrospinal fluid production. Mast cells are also found in the human choroid plexus. Levels of tryptase in cerebrospinal fluid are closely associated with clinical severity of TAH. BMS-262084, an inhibitor of tryptase, can cross the blood-brain barrier, inhibit TAH in vivo, and alleviate mast-cell-induced damage of epithelial cilia in a human pluripotent stem-cell-derived choroid plexus organoid model. Collectively, we uncover the function of CPMCs and provide an attractive therapy for TAH.
Collapse
Affiliation(s)
- Yiye Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Can Di
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Shijian Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yubo Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yiwen Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jianyou Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Bingxi Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jian Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou 510080, China
| | - Kaihua Guo
- Department of Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou 510080, China; Department of Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Biotherapy Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
4
|
Kohs TCL, Fallon ME, Oseas EC, Healy LD, Tucker EI, Gailani D, McCarty OJT, Vandenbark AA, Offner H, Verbout NG. Pharmacological targeting of coagulation factor XI attenuates experimental autoimmune encephalomyelitis in mice. Metab Brain Dis 2023; 38:2383-2391. [PMID: 37341855 PMCID: PMC10530106 DOI: 10.1007/s11011-023-01251-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
Multiple sclerosis (MS) is the most common causes of non-traumatic disability in young adults worldwide. MS pathophysiologies include the formation of inflammatory lesions, axonal damage and demyelination, and blood brain barrier (BBB) disruption. Coagulation proteins, including factor (F)XII, can serve as important mediators of the adaptive immune response during neuroinflammation. Indeed, plasma FXII levels are increased during relapse in relapsing-remitting MS patients, and previous studies showed that reducing FXII levels was protective in a murine model of MS, experimental autoimmune encephalomyelitis (EAE). Our objective was to determine if pharmacological targeting of FXI, a major substrate of activated FXII (FXIIa), improves neurological function and attenuates CNS damage in the setting of EAE. EAE was induced in male mice using murine myelin oligodendrocyte glycoprotein peptides combined with heat-inactivated Mycobacterium tuberculosis and pertussis toxin. Upon onset of symptoms, mice were treated every other day intravenously with anti-FXI antibody, 14E11, or saline. Disease scores were recorded daily until euthanasia for ex vivo analyses of inflammation. Compared to the vehicle control, 14E11 treatment reduced the clinical severity of EAE and total mononuclear cells, including CD11b+CD45high macrophage/microglia and CD4+ T cell numbers in brain. Following pharmacological targeting of FXI, BBB disruption was reduced, as measured by decreased axonal damage and fibrin(ogen) accumulation in the spinal cord. These data demonstrate that pharmacological inhibition of FXI reduces disease severity, immune cell migration, axonal damage, and BBB disruption in mice with EAE. Thus, therapeutic agents targeting FXI and FXII may provide a useful approach for treating autoimmune and neurologic disorders.
Collapse
Affiliation(s)
- Tia C L Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA.
| | - Meghan E Fallon
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Ethan C Oseas
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Laura D Healy
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Erik I Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
- Aronora, Inc., Portland, OR, USA
| | - David Gailani
- Department of Pathology and Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
- Veterans Affairs Portland Health Care System, Portland, OR, USA
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Norah G Verbout
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
- Aronora, Inc., Portland, OR, USA
| |
Collapse
|
5
|
Roehrig S, Ackerstaff J, Jiménez Núñez E, Teller H, Ellerbrock P, Meier K, Heitmeier S, Tersteegen A, Stampfuss J, Lang D, Schlemmer KH, Schaefer M, Gericke KM, Kinzel T, Meibom D, Schmidt M, Gerdes C, Follmann M, Hillisch A. Design and Preclinical Characterization Program toward Asundexian (BAY 2433334), an Oral Factor XIa Inhibitor for the Prevention and Treatment of Thromboembolic Disorders. J Med Chem 2023; 66:12203-12224. [PMID: 37669040 PMCID: PMC10510402 DOI: 10.1021/acs.jmedchem.3c00795] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Indexed: 09/06/2023]
Abstract
Activated coagulation factor XI (FXIa) is a highly attractive antithrombotic target as it contributes to the development and progression of thrombosis but is thought to play only a minor role in hemostasis so that its inhibition may allow for decoupling of antithrombotic efficacy and bleeding time prolongation. Herein, we report our major efforts to identify an orally bioavailable, reversible FXIa inhibitor. Using a protein structure-based de novo design approach, we identified a novel micromolar hit with attractive physicochemical properties. During lead modification, a critical problem was balancing potency and absorption by focusing on the most important interactions of the lead series with FXIa while simultaneously seeking to improve metabolic stability and the cytochrome P450 interaction profile. In clinical trials, the resulting compound from our extensive research program, asundexian (BAY 2433334), proved to possess the desired DMPK properties for once-daily oral dosing, and even more importantly, the initial pharmacological hypothesis was confirmed.
Collapse
Affiliation(s)
- Susanne Roehrig
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | | | | | | | | | | | - Stefan Heitmeier
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | - Adrian Tersteegen
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | - Jan Stampfuss
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | - Dieter Lang
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | | | | | - Kersten M. Gericke
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | | | - Daniel Meibom
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | - Martina Schmidt
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | - Christoph Gerdes
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | - Markus Follmann
- Pharmaceuticals, Research
and Development, Bayer AG, 42133 Wuppertal, Germany
| | | |
Collapse
|
6
|
Wichaiyo S, Parichatikanond W, Visansirikul S, Saengklub N, Rattanavipanon W. Determination of the Potential Clinical Benefits of Small Molecule Factor XIa Inhibitors in Arterial Thrombosis. ACS Pharmacol Transl Sci 2023; 6:970-981. [PMID: 37470020 PMCID: PMC10353063 DOI: 10.1021/acsptsci.3c00052] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Indexed: 07/21/2023]
Abstract
Anticoagulants are the mainstay for the prevention and treatment of thrombosis. However, bleeding complications remain a primary concern. Recent advances in understanding the contribution of activated factor XI (FXIa) in arterial thrombosis with a limited impact on hemostasis have led to the development of several FXIa-targeting modalities. Injectable agents including monoclonal antibodies and antisense oligonucleotides against FXIa have been primarily studied in venous thrombosis. The orally active small molecules that specifically inhibit the active site of FXIa are currently being investigated for their antithrombotic activity in both arteries and veins. This review focuses on a discussion of the potential clinical benefits of small molecule FXIa inhibitors, mainly asundexian and milvexian, in arterial thrombosis based on their pharmacological profiles and the compelling results of phase 2 clinical studies. The preclinical and epidemiological basis for the impact of FXIa in hemostasis and arterial thrombosis is also addressed. In recent clinical study results, asundexian appears to reduce ischemic events in patients with myocardial infarction and minor-to-moderate stroke, whereas milvexian possibly provides benefits in patients with minor stroke or high-risk transient ischemic attack (TIA). In addition, asundexian and milvexian had a minor impact on hemostasis even in combination with dual-antiplatelet therapy. Other orally active FXIa inhibitors also produce antithrombotic activity in vivo with low bleeding risk. Therefore, FXIa inhibitors might represent a new class of direct-acting oral anticoagulants (DOACs) for the treatment of thrombosis, although the explicit clinical positions of asundexian and milvexian in patients with ischemic stroke, high-risk TIA, and coronary artery disease require confirmation from the outcomes of ongoing phase 3 trials.
Collapse
Affiliation(s)
- Surasak Wichaiyo
- Department
of Pharmacology, Faculty of Pharmacy, Mahidol
University, Bangkok 10400, Thailand
- Centre
of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Warisara Parichatikanond
- Department
of Pharmacology, Faculty of Pharmacy, Mahidol
University, Bangkok 10400, Thailand
- Centre
of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Satsawat Visansirikul
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Nakkawee Saengklub
- Centre
of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Department
of Physiology, Faculty of Pharmacy, Mahidol
University, Bangkok 10400, Thailand
| | | |
Collapse
|
7
|
Xie Z, Meng Z, Yang X, Duan Y, Wang Q, Liao C. Factor XIa Inhibitors in Anticoagulation Therapy: Recent Advances and Perspectives. J Med Chem 2023; 66:5332-5363. [PMID: 37037122 DOI: 10.1021/acs.jmedchem.2c02130] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Factor XIa (FXIa) in the intrinsic pathway of the coagulation process has been proven to be an effective and safe target for anticoagulant discovery with limited or no bleeding. Numerous small-molecule FXIa inhibitors (SMFIs) with various scaffolds have been identified in the early stages of drug discovery. They have served as the foundation for the recent discovery of additional promising SMFIs with improved potency, selectivity, and pharmacokinetic profiles, some of which have entered clinical trials for the treatment of thrombosis. After reviewing the coagulation process and structure of FXIa, this perspective discusses the rational or structure-based design, discovery, structure-activity relationships, and development of SMFIs disclosed in recent years. Strategies for identifying more selective and druggable SMFIs are provided, paving the way for the design and discovery of more useful SMFIs for anticoagulation therapy.
Collapse
Affiliation(s)
- Zhouling Xie
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Zhiwei Meng
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Xiaoxiao Yang
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Yajun Duan
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Qin Wang
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Chenzhong Liao
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| |
Collapse
|
8
|
Wang X, Li Q, Du F, Shukla N, Nawrocki AR, Chintala M. Antithrombotic Effects of the Novel Small-Molecule Factor XIa Inhibitor Milvexian in a Rabbit Arteriovenous Shunt Model of Venous Thrombosis. TH OPEN 2023; 7:e97-e104. [PMID: 37101592 PMCID: PMC10125780 DOI: 10.1055/a-2061-3311] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/20/2023] [Indexed: 04/28/2023] Open
Abstract
Background Factor XIa (FXIa) is an emerging therapeutic target, and FXIa inhibition is a promising mechanism to improve therapeutic index over current anticoagulants. Milvexian (BMS-986177/JNJ-70033093) is an oral small-molecule FXIa inhibitor. Objective Milvexian's antithrombotic efficacy was characterized in a rabbit arteriovenous (AV) shunt model of venous thrombosis and compared with the factor Xa inhibitor apixaban and the direct thrombin inhibitor dabigatran. Methods The AV shunt model of thrombosis was conducted in anesthetized rabbits. Vehicle or drugs were administered as intravenous bolus plus a continuous infusion. Thrombus weight was the primary efficacy endpoint. Ex vivo activated partial thromboplastin time (aPTT), prothrombin time (PT), and thrombin time (TT) were measured as the pharmacodynamic responses. Results Milvexian dose dependently reduced thrombus weights by 34.3 ± 7.9, 51.6 ± 6.8 ( p < 0.01; n = 5), and 66.9 ± 4.8% ( p < 0.001; n = 6) versus vehicle at 0.25 + 0.17, 1.0 + 0.67, and 4.0 ± 2.68 mg/kg bolus + mg/kg/h infusion, respectively. Ex vivo clotting data supported a dose-dependent prolongation of aPTT (with 1.54-, 2.23-, and 3.12-fold increases from baseline upon the AV shunt start), but no changes in PT and TT. Dose-dependent inhibition in thrombus weight and clotting assays was also demonstrated for both apixaban and dabigatran as the references for the model validation. Conclusion Results demonstrate that milvexian is an effective anticoagulant for prevention of venous thrombosis in the rabbit model, which supports the utility of milvexian in venous thrombosis, as seen in the phase 2 clinical study.
Collapse
Affiliation(s)
- Xinkang Wang
- Cardiovascular & Metabolism Therapeutic Area, Janssen Research & Development, LLC, Spring House, Pennsylvania, United States
- Address for correspondence Xinkang Wang, PhD Janssen Research & Development, LLC1400 McKean Road, 42-2522, Spring House, PA 19002United States
| | - Qiu Li
- Cardiovascular & Metabolism Therapeutic Area, Janssen Research & Development, LLC, Spring House, Pennsylvania, United States
| | - Fuyong Du
- Cardiovascular & Metabolism Therapeutic Area, Janssen Research & Development, LLC, Spring House, Pennsylvania, United States
| | - Neetu Shukla
- Formulation, Janssen Research & Development, LLC, Spring House, Pennsylvania, United States
| | - Andrea R. Nawrocki
- Cardiovascular & Metabolism Therapeutic Area, Janssen Research & Development, LLC, Spring House, Pennsylvania, United States
| | - Madhu Chintala
- Cardiovascular & Metabolism Therapeutic Area, Janssen Research & Development, LLC, Spring House, Pennsylvania, United States
| |
Collapse
|
9
|
A Factor XIa Inhibitor Engineered from Banded Krait Venom Toxin: Efficacy and Safety in Rodent Models of Arterial and Venous Thrombosis. Biomedicines 2022; 10:biomedicines10071679. [PMID: 35884984 PMCID: PMC9312835 DOI: 10.3390/biomedicines10071679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/04/2022] Open
Abstract
Activated factor XI (FXIa) is an important antithrombotic drug target. Clinical and pre-clinical data have demonstrated that its inhibition attenuates thrombosis with minimal risk of excessive bleeding. We isolated Fasxiator from the venom of banded krait Bungarus fasciatus and subsequently engineered FasxiatorN17R,L19E, with improved affinity (Ki = 0.9 nM) and selectivity towards FXIa. Here, we assess the in vivo efficacy and bleeding risk of rFasxiatorN17R, L19E in pre-clinical animal models. Rats injected intravenously (i.v.) with bolus rFasxiatorN17R, L19E showed the specific in vivo attenuation of the intrinsic coagulation pathway, lasting for at least 60 min. We performed the in vivo dose-ranging experiments for rFasxiatorN17R, L19E as follows: FeCl3-induced carotid artery occlusion in rats (arterial thrombosis); inferior vena cava ligation in mice (venous thrombosis); tail bleeding time in both rats and mice (bleeding risk). Head-to-head comparisons were made using therapeutic dosages of unfractionated heparin (UFH) and low-molecular-weight heparin (LMWH) for arterial and venous thrombosis, respectively. In the arterial thrombosis model, 2 mg/kg i.v. rFasxiatorN17R,L19E achieved a similar antithrombotic efficacy to that of UFH, with >3-fold lower bleeding time. In the venous thrombosis model, the 10 mg/kg subcutaneous (s.c.) injection of rFasxiatorN17R,L19E achieved similar efficacy and bleeding levels to those of LMWH enoxaparin. Overall, rFasxiatorN17R,L19E represents a promising molecule for the development of FXIa-targeting anticoagulants.
Collapse
|
10
|
Nopp S, Kraemmer D, Ay C. Factor XI Inhibitors for Prevention and Treatment of Venous Thromboembolism: A Review on the Rationale and Update on Current Evidence. Front Cardiovasc Med 2022; 9:903029. [PMID: 35647061 PMCID: PMC9133368 DOI: 10.3389/fcvm.2022.903029] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
Although anticoagulation therapy has evolved from non-specific drugs (i.e., heparins and vitamin K antagonists) to agents that directly target specific coagulation factors (i.e., direct oral anticoagulants, argatroban, fondaparinux), thrombosis remains a leading cause of death worldwide. Direct oral anticoagulants (i.e., factor IIa- and factor Xa-inhibitors) now dominate clinical practice because of their favorable pharmacological profile and ease of use, particularly in venous thromboembolism (VTE) treatment and stroke prevention in atrial fibrillation. However, despite having a better safety profile than vitamin K antagonists, their bleeding risk is not insignificant. This is true for all currently available anticoagulants, and a high bleeding risk is considered a contraindication to anticoagulation. As a result, ongoing research focuses on developing future anticoagulants with an improved safety profile. Several promising approaches to reduce the bleeding risk involve targeting the intrinsic (or contact activation) pathway of coagulation, with the ultimate goal of preventing thrombosis without impairing hemostasis. Based on epidemiological data on hereditary factor deficiencies and preclinical studies factor XI (FXI) emerged as the most promising candidate target. In this review, we highlight unmet clinical needs of anticoagulation therapy, outlay the rationale and evidence for inhibiting FXI, discuss FXI inhibitors in current clinical trials, conduct an exploratory meta-analysis on their efficacy and safety, and provide an outlook on the potential clinical application of these novel anticoagulants.
Collapse
Affiliation(s)
| | | | - Cihan Ay
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
11
|
Dilger AK, Pabbisetty KB, Corte JR, De Lucca I, Fang T, Yang W, Pinto DJP, Wang Y, Zhu Y, Mathur A, Li J, Hou X, Smith D, Sun D, Zhang H, Krishnananthan S, Wu DR, Myers JE, Sheriff S, Rossi KA, Chacko S, Zheng JJ, Galella MA, Ziemba T, Dierks EA, Bozarth JM, Wu Y, Crain E, Wong PC, Luettgen JM, Wexler RR, Ewing WR. Discovery of Milvexian, a High-Affinity, Orally Bioavailable Inhibitor of Factor XIa in Clinical Studies for Antithrombotic Therapy. J Med Chem 2022; 65:1770-1785. [PMID: 34494428 DOI: 10.1021/acs.jmedchem.1c00613] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Factor XIa (FXIa) is an enzyme in the coagulation cascade thought to amplify thrombin generation but has a limited role in hemostasis. From preclinical models and human genetics, an inhibitor of FXIa has the potential to be an antithrombotic agent with superior efficacy and safety. Reversible and irreversible inhibitors of FXIa have demonstrated excellent antithrombotic efficacy without increased bleeding time in animal models (Weitz, J. I., Chan, N. C. Arterioscler. Thromb. Vasc. Biol. 2019, 39 (1), 7-12). Herein, we report the discovery of a novel series of macrocyclic FXIa inhibitors containing a pyrazole P2' moiety. Optimization of the series for (pharmacokinetic) PK properties, free fraction, and solubility resulted in the identification of milvexian (BMS-986177/JNJ-70033093, 17, FXIa Ki = 0.11 nM) as a clinical candidate for the prevention and treatment of thromboembolic disorders, suitable for oral administration.
Collapse
Affiliation(s)
- Andrew K Dilger
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Kumar B Pabbisetty
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - James R Corte
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Indawati De Lucca
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Tianan Fang
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Wu Yang
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Donald J P Pinto
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Yufeng Wang
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Yeheng Zhu
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Arvind Mathur
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Jianqing Li
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Xiaoping Hou
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Daniel Smith
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Dawn Sun
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Huiping Zhang
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Subramaniam Krishnananthan
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Dauh-Rurng Wu
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joseph E Myers
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Steven Sheriff
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Karen A Rossi
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Silvi Chacko
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joanna J Zheng
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Michael A Galella
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Theresa Ziemba
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Elizabeth A Dierks
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Jeffrey M Bozarth
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Yiming Wu
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Earl Crain
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Pancras C Wong
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joseph M Luettgen
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Ruth R Wexler
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - William R Ewing
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| |
Collapse
|
12
|
WPK5, a Novel Kunitz-Type Peptide from the Leech Whitmania pigra Inhibiting Factor XIa, and Its Loop-Replaced Mutant to Improve Potency. Biomedicines 2021; 9:biomedicines9121745. [PMID: 34944561 PMCID: PMC8698482 DOI: 10.3390/biomedicines9121745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
Kunitz-type proteins or peptides have been found in many blood-sucking animals, but the identity of them in leeches remained elusive. In the present study, five Kunitz-type peptides named WPK1-WPK5 were identified from the leech Whitmania pigra. Recombinant WPK1-WPK5 were expressed in Pichia pastoris GS115, and their inhibitory activity against Factor XIa (FXIa) was tested. WPK5 showed inhibitory activity against FXIa with an IC50 value of 978.20 nM. To improve its potency, the loop replacement strategy was used. The loop 1 (TGPCRSNLER) and loop 2 (QYGGC) in WPK5 were replaced by loop 1 (TGPCRAMISR) and loop 2 (FYGGC) in PN2KPI, respectively, and the resulting peptide named WPK5-Mut showed an IC50 value of 8.34 nM to FXIa, which is about 100-fold the potency of FXIa compared to that of WPK5. WPK5-Mut was further evaluated for its extensive bioactivity in vitro and in vivo. It dose-dependently prolonged APTT on both murine plasma and human plasma, and potently inhibited FeCl3-induced carotid artery thrombosis in mice at a dose of 1.5 mg/kg. Additionally, WPK5-Mut did not show significant bleeding risk at a dose of 6 mg/kg. Together, these results showed that WPK5-Mut is a promising candidate for the development of an antithrombotic drug.
Collapse
|
13
|
Wong PC, Quan ML. Improved efficacy/safety profile of factor XIa inhibitor BMS-724296 versus factor Xa inhibitor apixaban and thrombin inhibitor dabigatran in cynomolgus monkeys. Res Pract Thromb Haemost 2021; 5:e12524. [PMID: 34095733 PMCID: PMC8162232 DOI: 10.1002/rth2.12524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/31/2022] Open
Abstract
Background Inhibition of activated factor XI (FXIa) is a promising antithrombotic drug target. BMS-724296 is a selective, reversible, small-molecule inhibitor of human FXIa (Ki 0.3 nM). Objectives This study assessed effects of BMS-724296 versus standard-of-care oral anticoagulants apixaban (activated factor X inhibitor) and dabigatran (thrombin inhibitor) on arterial thrombosis, kidney bleeding time (KBT), and clotting time (CT) in nonhuman primate (NHP) cynomolgus monkey models. Methods Carotid artery thrombosis was produced by electrical stimulation in anesthetized NHPs. Hemostasis was assessed with a provoked KBT model. Thrombosis, KBT, and CT were monitored. Vehicle and various doses of BMS-724296, apixaban, and dabigatran were administered as bolus (intravenous [i.v.]) followed by infusion starting 30 minutes before initiation of thrombosis and continued until the experiment's end (n = 3-8/group). Primary end points included thrombus weight reduction (TWR), KBT, and CT (activated partial thromboplastin time [aPTT], prothrombin time [PT], and thrombin time [TT]). Results BMS-724296 at 0.025 + 0.05, 0.05 + 0.1, 0.102 + 0.2, and 0.4 + 0.8 mg/kg+mg/kg/h i.v. (bolus + infusion) reduced thrombus weight by 0 ± 0, 35 ± 7*, 72 ± 4*, and 86 ± 4%*, respectively (*P < .05 vs vehicle; n = 5-6/group). BMS-724296 at the highest dose (0.4 + 0.8 mg/kg+mg/kg/h) did not increase KBT compared to vehicle (109 ± 6 vs 113 ± 20 seconds, respectively) and increased ex vivo aPTT by 2.9 ± 0.1-fold without changing PT and TT. In companion NHP studies, high doses of apixaban and dabigatran produced similar TWR as BMS-724296, but increased KBT 4.3 ± 0.5-fold and 5.8 ± 0.5-fold, respectively (n = 3-4/group). Conclusions BMS-724296 produced similar antithrombotic efficacy as apixaban and dabigatran but with no increase in KBT in NHPs. These findings suggest that FXIa inhibitors may provide safe and effective antithrombotic therapy.
Collapse
Affiliation(s)
- Pancras C Wong
- Cardiovascular & Fibrosis Drug Discovery Biology Bristol Myers Squibb Princeton NJ USA
| | - Mimi L Quan
- Cardiovascular & Fibrosis Drug Discovery Biology Bristol Myers Squibb Princeton NJ USA
| |
Collapse
|
14
|
Demoulin S, Godfroid E, Hermans C. Dual inhibition of factor XIIa and factor XIa as a therapeutic approach for safe thromboprotection. J Thromb Haemost 2021; 19:323-329. [PMID: 33047454 DOI: 10.1111/jth.15130] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/11/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022]
Abstract
Clinical practice shows that a critical unmet need in the field of medical device-associated thrombosis prevention is the availability of an anticoagulant therapy without hemorrhagic risk. In the quest for new drugs that are at least as effective as those currently available, while avoiding bleeding complications, molecules that target nearly every step of the coagulation pathway have been developed. Among these molecules, inhibitors of factor XII (FXII) or factor XI (FXI) are promising alternatives as deficiencies in these factors protect against thrombosis without causing spontaneous hemorrhage, as revealed by epidemiological and preclinical data. Ixodes ricinus-contact phase inhibitor (Ir-CPI), a new anticoagulant candidate with an innovative mechanism of action could be this ideal anticoagulant agent for safe prevention from clotting on medical devices. This protein, which selectively binds to FXIIa, FXIa, and plasma kallikrein and inhibits the reciprocal activation of FXII, prekallikrein, and FXI in human plasma, was shown to prevent thrombosis in an ovine cardiopulmonary bypass system associated with cardiac surgeries. Furthermore, as opposed to unfractionated heparin, Ir-CPI appears to be devoid of bleeding risk. This review outlines the rationale for targeting upstream coagulation factors in order to prevent medical device-associated thrombosis; examines the novel approaches under development; and focuses on Ir-CPI, which shows promising properties in the field of thrombosis prevention.
Collapse
Affiliation(s)
| | | | - Cédric Hermans
- Division of Hematology, Hemostasis and Thrombosis Unit, Saint-Luc University Hospital, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
15
|
Fredenburgh JC, Weitz JI. New anticoagulants: Moving beyond the direct oral anticoagulants. J Thromb Haemost 2021; 19:20-29. [PMID: 33047462 DOI: 10.1111/jth.15126] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 01/23/2023]
Abstract
Although anticoagulants have been in use for more than 80 years, heparin and vitamin K antagonists were the sole available options until recently. Although these agents revolutionized the prevention and treatment of thrombotic diseases, their use has been hampered by the necessity for coagulation monitoring and by bleeding complications resulting in part from their multiple sites of action. Owing to advances in basic science, animal models, and epidemiology, the arsenal of available anticoagulants has expanded in the past two decades. This evolution has yielded many novel compounds that target single coagulation enzymes. Initially, thrombin and factor Xa were targeted because of their critical roles in coagulation. However, attention has now shifted to compounds that target upstream reactions, particularly those catalyzed by factors XIIa and XIa, which are part of the contact system. This shift is predicated on epidemiological and experimental evidence suggesting that these factors are more important for thrombosis than for hemostasis. With the goal of developing a new class of anticoagulants associated with a lower risk of bleeding than currently available agents, dozens of drugs targeting the contact system are now in development. This article focuses on the rationale, development, and testing of these new agents with a concentration on those that have reached or completed phase 2 evaluation for at least one indication.
Collapse
Affiliation(s)
- James C Fredenburgh
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
- Departments of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jeffrey I Weitz
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
- Departments of Medicine, McMaster University, Hamilton, ON, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
16
|
EP-7041, a Factor XIa Inhibitor as a Potential Antithrombotic Strategy in Extracorporeal Membrane Oxygenation: A Brief Report. Crit Care Explor 2020; 2:e0196. [PMID: 32984829 PMCID: PMC7498136 DOI: 10.1097/cce.0000000000000196] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objectives: Current approaches to systemic antithrombotic therapy in support of extracorporeal membrane oxygenation are limited and are hampered by both thrombotic and hemorrhagic complications. An alternative approach is needed. Design: Inhibition of coagulation factor XI/activated factor XI is an appealing pathway for antithrombotic support of extracorporeal membrane oxygenation. Selective inhibition of the contact pathway of coagulation could reduce bleeding risk, and because factor XI is linked with the inflammatory and complement systems, it can also be viewed as a biologically plausible target for the prevention of abnormal thrombosis during extracorporeal membrane oxygenation. Conclusions: We introduce initial information on EP-7041, a parenteral, potent, and selective, small-molecule activated factor XIa inhibitor with pharmacodynamic and pharmacokinetic characteristics that appear well suited for use in a critical care environment.
Collapse
|
17
|
Boothello RS, Sankaranarayanan NV, Afosah DK, Karuturi R, Al-Horani RA, Desai UR. Studies on fragment-based design of allosteric inhibitors of human factor XIa. Bioorg Med Chem 2020; 28:115762. [PMID: 32992249 DOI: 10.1016/j.bmc.2020.115762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/24/2020] [Accepted: 09/06/2020] [Indexed: 12/16/2022]
Abstract
Human factor XIa (hFXIa) has emerged as an attractive target for development of new anticoagulants that promise higher level of safety. Different strategies have been adopted so far for the design of anti-hFXIa molecules including competitive and non-competitive inhibition. Of these, allosteric dysfunction of hFXIa's active site is especially promising because of the possibility of controlled reduction in activity that may offer a route to safer anticoagulants. In this work, we assess fragment-based design approach to realize a group of novel allosteric hFXIa inhibitors. Starting with our earlier discovery that sulfated quinazolinone (QAO) bind in the heparin-binding site of hFXIa, we developed a group of two dozen dimeric sulfated QAOs with intervening linkers that displayed a progressive variation in inhibition potency. In direct opposition to the traditional wisdom, increasing linker flexibility led to higher potency, which could be explained by computational studies. Sulfated QAO 19S was identified as the most potent and selective inhibitor of hFXIa. Enzyme inhibition studies revealed that 19S utilizes a non-competitive mechanism of action, which was supported by fluorescence studies showing a classic sigmoidal binding profile. Studies with selected mutants of hFXIa indicated that sulfated QAOs bind in heparin-binding site of the catalytic domain of hFXIa. Overall, the approach of fragment-based design offers considerable promise for designing heparin-binding site-directed allosteric inhibitors of hFXIa.
Collapse
Affiliation(s)
- Rio S Boothello
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States; Hunter Holmes McGuire Medical Center, Richmond, VA 23249, United States
| | - Nehru Viji Sankaranarayanan
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States
| | - Daniel K Afosah
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States
| | - Rajesh Karuturi
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States
| | - Rami A Al-Horani
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States; Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, United States
| | - Umesh R Desai
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States.
| |
Collapse
|
18
|
Cave BE, Shah SP. Turning Up to Eleven: Factor XI Inhibitors as Novel Agents to Maximize Safety and Maintain Efficacy in Thromboembolic Disease. Curr Probl Cardiol 2020; 46:100696. [PMID: 32994051 DOI: 10.1016/j.cpcardiol.2020.100696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/23/2020] [Indexed: 11/16/2022]
Abstract
Within the past decade nonvitamin K oral anticoagulants have emerged as the standard of care for the prevention and treatment of thromboembolic disorders, however safety of anticoagulants remain a concern for many patients and providers. There exists new interest in factor XI inhibition as novel therapeutic target based on observations of lower thrombotic rates and without significant bleed risk in individuals with inherited factor XI deficiency. Several classes of factor XI inhibitors including antisense oligonucleotides, monoclonal antibodies, and small molecule inhibitors have undergone preclinical studies and clinical trials in humans. Both osocimab and IONIS-FXI have been evaluated in patients undergoing orthopedic surgery and demonstrated superiority to enoxaparin without increasing major bleeding. Future studies with both these agents are ongoing, as well as the continued development of other inhibitors of factor XI. Early data regarding factor XI inhibition is encouraging as a potent anticoagulant and may offer a safer alternative compared to therapeutic currently available in contemporary practice for thromboembolic disease.
Collapse
|
19
|
La CC, Takeuchi LE, Abbina S, Vappala S, Abbasi U, Kizhakkedathu JN. Targeting Biological Polyanions in Blood: Strategies toward the Design of Therapeutics. Biomacromolecules 2020; 21:2595-2621. [DOI: 10.1021/acs.biomac.0c00654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
20
|
Orally bioavailable amine-linked macrocyclic inhibitors of factor XIa. Bioorg Med Chem Lett 2020; 30:126949. [PMID: 31932224 DOI: 10.1016/j.bmcl.2020.126949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/23/2019] [Accepted: 01/01/2020] [Indexed: 11/20/2022]
Abstract
The discovery of orally bioavailable FXIa inhibitors has been a challenge. Herein, we describe our efforts to address this challenge by optimization of our imidazole-based macrocyclic series. Our optimization strategy focused on modifications to the P2 prime, macrocyclic amide linker, and the imidazole scaffold. Replacing the amide of the macrocyclic linker with amide isosteres led to the discovery of substituted amine linkers which not only maintained FXIa binding affinity but also improved oral exposure in rats. Combining the optimized macrocyclic amine linker with a pyridine scaffold afforded compounds 23 and 24 that were orally bioavailable, single-digit nanomolar FXIa inhibitors with excellent selectivity against relevant blood coagulation enzymes.
Collapse
|
21
|
Abstract
Activation of the intrinsic pathway of coagulation contributes to the pathogenesis of arterial and venous thrombosis. Critical insights into the involvement of intrinsic pathway factors have been derived from the study of gene-specific knockout animals and targeted inhibitors. Importantly, preclinical studies have indicated that targeting components of this pathway, including FXI (factor XI), FXII, and PKK (prekallikrein), reduces thrombosis with no significant effect on protective hemostatic pathways. This review highlights the advances made from studying the intrinsic pathway using gene-specific knockout animals and inhibitors in models of arterial and venous thrombosis. Development of inhibitors of activated FXI and FXII may reduce thrombosis with minimal increases in bleeding compared with current anticoagulant drugs.
Collapse
Affiliation(s)
- Steven P Grover
- From the Division of Hematology and Oncology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill
| | - Nigel Mackman
- From the Division of Hematology and Oncology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill
| |
Collapse
|
22
|
Corte JR, Pinto DJP, Fang T, Osuna H, Yang W, Wang Y, Lai A, Clark CG, Sun JH, Rampulla R, Mathur A, Kaspady M, Neithnadka PR, Li YXC, Rossi KA, Myers JE, Sheriff S, Lou Z, Harper TW, Huang C, Zheng JJ, Bozarth JM, Wu Y, Wong PC, Crain EJ, Seiffert DA, Luettgen JM, Lam PYS, Wexler RR, Ewing WR. Potent, Orally Bioavailable, and Efficacious Macrocyclic Inhibitors of Factor XIa. Discovery of Pyridine-Based Macrocycles Possessing Phenylazole Carboxamide P1 Groups. J Med Chem 2019; 63:784-803. [DOI: 10.1021/acs.jmedchem.9b01768] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- James R. Corte
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Donald J. P. Pinto
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Tianan Fang
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Honey Osuna
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Wu Yang
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Yufeng Wang
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Amy Lai
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Charles G. Clark
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Jung-Hui Sun
- Research and Development, Bristol-Myers Squibb Company, US Rt. 206 & Province Line Road, Princeton, New Jersey 08540, United States
| | - Richard Rampulla
- Research and Development, Bristol-Myers Squibb Company, US Rt. 206 & Province Line Road, Princeton, New Jersey 08540, United States
| | - Arvind Mathur
- Research and Development, Bristol-Myers Squibb Company, US Rt. 206 & Province Line Road, Princeton, New Jersey 08540, United States
| | - Mahammed Kaspady
- Bristol-Myers Squibb Research Center, Syngene International Pvt. Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra−Jigani Road, Bangalore 560 100, India
| | - Premsai Rai Neithnadka
- Bristol-Myers Squibb Research Center, Syngene International Pvt. Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra−Jigani Road, Bangalore 560 100, India
| | - Yi-Xin Cindy Li
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Karen A. Rossi
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Joseph E. Myers
- Research and Development, Bristol-Myers Squibb Company, US Rt. 206 & Province Line Road, Princeton, New Jersey 08540, United States
| | - Steven Sheriff
- Research and Development, Bristol-Myers Squibb Company, US Rt. 206 & Province Line Road, Princeton, New Jersey 08540, United States
| | - Zhen Lou
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Timothy W. Harper
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Christine Huang
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Joanna J. Zheng
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Jeffrey M. Bozarth
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Yiming Wu
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Pancras C. Wong
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Earl J. Crain
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Dietmar A. Seiffert
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Joseph M. Luettgen
- Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08543, United States
| | - Patrick Y. S. Lam
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - Ruth R. Wexler
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| | - William R. Ewing
- Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States
| |
Collapse
|
23
|
Núñez-Navarro NE, Santana FM, Parra LP, Zacconi FC. Surfing the Blood Coagulation Cascade: Insight into the Vital Factor Xa. Curr Med Chem 2019; 26:3175-3200. [PMID: 29376487 DOI: 10.2174/0929867325666180125165340] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/28/2017] [Accepted: 01/11/2018] [Indexed: 01/19/2023]
Abstract
Factor Xa (FXa) plays a key role in haemostasis, it is a central part of the blood coagulation cascade which catalyzes the production of thrombin and leads to clot formation and wound closure. Therefore, FXa is an attractive target for the development of new anticoagulant agents. In this review, we will first describe the molecular features of this fundamental protein in order to understand its mechanism of action, an essential background for the design of novel inhibitors by means of synthetic organic chemistry or using peptides obtained from recombinant methodologies. Then, we will review the current state of the synthesis of novel direct FXa inhibitors along with their mechanisms of action. Finally, approved reversal agents that aid in maintaining blood haemostasis by using these commercial drugs will also be discussed.
Collapse
Affiliation(s)
- Nicolás E Núñez-Navarro
- Department of Organic Chemistry, Faculty of Chemistry, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Department of Chemical and Bioprocesses Engineering, School of Engineering, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Fabián M Santana
- Department of Organic Chemistry, Faculty of Chemistry, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Loreto P Parra
- Department of Chemical and Bioprocesses Engineering, School of Engineering, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Flavia C Zacconi
- Department of Organic Chemistry, Faculty of Chemistry, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Research Center for Nanotechnology and Advanced Materials "CIEN-UC", Pontificia Universidad Catolica de Chile, Santiago, Chile
| |
Collapse
|
24
|
Székely O, Borgi M, Lip GYH. Factor XI inhibition fulfilling the optimal expectations for ideal anticoagulation. Expert Opin Emerg Drugs 2019; 24:55-61. [DOI: 10.1080/14728214.2019.1591368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Orsolya Székely
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Marco Borgi
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Gregory Y. H. Lip
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Aalborg Thrombosis Research Unit, Aalborg University, Aalborg, Denmark
| |
Collapse
|
25
|
Al-Horani RA, Afosah DK. Recent advances in the discovery and development of factor XI/XIa inhibitors. Med Res Rev 2018; 38:1974-2023. [PMID: 29727017 PMCID: PMC6173998 DOI: 10.1002/med.21503] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 03/09/2018] [Accepted: 04/04/2018] [Indexed: 12/12/2022]
Abstract
Factor XIa (FXIa) is a serine protease homodimer that belongs to the intrinsic coagulation pathway. FXIa primarily catalyzes factor IX activation to factor IXa, which subsequently activates factor X to factor Xa in the common coagulation pathway. Growing evidence suggests that FXIa plays an important role in thrombosis with a relatively limited contribution to hemostasis. Therefore, inhibitors targeting factor XI (FXI)/FXIa system have emerged as a paradigm-shifting strategy so as to develop a new generation of anticoagulants to effectively prevent and/or treat thromboembolic diseases without the life-threatening risk of internal bleeding. Several inhibitors of FXI/FXIa proteins have been discovered or designed over the last decade including polypeptides, active site peptidomimetic inhibitors, allosteric inhibitors, antibodies, and aptamers. Antisense oligonucleotides (ASOs), which ultimately reduce the hepatic biosynthesis of FXI, have also been introduced. A phase II study, which included patients undergoing elective primary unilateral total knee arthroplasty, revealed that a specific FXI ASO effectively protects patients against venous thrombosis with a relatively limited risk of bleeding. Initial findings have also demonstrated the potential of FXI/FXIa inhibitors in sepsis, listeriosis, and arterial hypertension. This review highlights various chemical, biochemical, and pharmacological aspects of FXI/FXIa inhibitors with the goal of advancing their development toward clinical use.
Collapse
Affiliation(s)
- Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, Louisiana 70125
| | - Daniel K. Afosah
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23219
| |
Collapse
|
26
|
Quan ML, Pinto DJP, Smallheer JM, Ewing WR, Rossi KA, Luettgen JM, Seiffert DA, Wexler RR. Factor XIa Inhibitors as New Anticoagulants. J Med Chem 2018; 61:7425-7447. [PMID: 29775297 DOI: 10.1021/acs.jmedchem.8b00173] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
With the introduction of thrombin and factor Xa inhibitors to the oral anticoagulant market, significant improvements in both efficacy and safety have been achieved. Early clinical and preclinical data suggest that inhibitors of factor XIa can provide a still safer alternative, with expanded efficacy for arterial indications. This Perspective provides an overview of target rationale and details of the discovery and development of inhibitors of factor XIa as next generation antithrombotic agents.
Collapse
Affiliation(s)
- Mimi L Quan
- Research and Development , Bristol-Myers Squibb Company , P.O. Box 5400, Princeton , New Jersey 08543 , United States
| | - Donald J P Pinto
- Research and Development , Bristol-Myers Squibb Company , P.O. Box 5400, Princeton , New Jersey 08543 , United States
| | - Joanne M Smallheer
- Research and Development , Bristol-Myers Squibb Company , P.O. Box 5400, Princeton , New Jersey 08543 , United States
| | - William R Ewing
- Research and Development , Bristol-Myers Squibb Company , P.O. Box 5400, Princeton , New Jersey 08543 , United States
| | - Karen A Rossi
- Research and Development , Bristol-Myers Squibb Company , P.O. Box 5400, Princeton , New Jersey 08543 , United States
| | - Joseph M Luettgen
- Research and Development , Bristol-Myers Squibb Company , P.O. Box 5400, Princeton , New Jersey 08543 , United States
| | - Dietmar A Seiffert
- Research and Development , Bristol-Myers Squibb Company , P.O. Box 5400, Princeton , New Jersey 08543 , United States
| | - Ruth R Wexler
- Research and Development , Bristol-Myers Squibb Company , P.O. Box 5400, Princeton , New Jersey 08543 , United States
| |
Collapse
|
27
|
Pinto DJP, Orwat MJ, Smith LM, Quan ML, Lam PYS, Rossi KA, Apedo A, Bozarth JM, Wu Y, Zheng JJ, Xin B, Toussaint N, Stetsko P, Gudmundsson O, Maxwell B, Crain EJ, Wong PC, Lou Z, Harper TW, Chacko SA, Myers JE, Sheriff S, Zhang H, Hou X, Mathur A, Seiffert DA, Wexler RR, Luettgen JM, Ewing WR. Discovery of a Parenteral Small Molecule Coagulation Factor XIa Inhibitor Clinical Candidate (BMS-962212). J Med Chem 2017; 60:9703-9723. [DOI: 10.1021/acs.jmedchem.7b01171] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Donald J. P. Pinto
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Michael J. Orwat
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Leon M. Smith
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Mimi L. Quan
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Patrick Y. S. Lam
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Karen A. Rossi
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Atsu Apedo
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Jeffrey M. Bozarth
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Yiming Wu
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joanna J. Zheng
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Baomin Xin
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Nathalie Toussaint
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Paul Stetsko
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Olafur Gudmundsson
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Brad Maxwell
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Earl J. Crain
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Pancras C. Wong
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Zhen Lou
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Timothy W. Harper
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Silvi A. Chacko
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joseph E. Myers
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Steven Sheriff
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Huiping Zhang
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Xiaoping Hou
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Arvind Mathur
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Dietmar A. Seiffert
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Ruth R. Wexler
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joseph M. Luettgen
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - William R. Ewing
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| |
Collapse
|
28
|
Corte JR, Yang W, Fang T, Wang Y, Osuna H, Lai A, Ewing WR, Rossi KA, Myers JE, Sheriff S, Lou Z, Zheng JJ, Harper TW, Bozarth JM, Wu Y, Luettgen JM, Seiffert DA, Quan ML, Wexler RR, Lam PY. Macrocyclic inhibitors of Factor XIa: Discovery of alkyl-substituted macrocyclic amide linkers with improved potency. Bioorg Med Chem Lett 2017; 27:3833-3839. [DOI: 10.1016/j.bmcl.2017.06.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/21/2017] [Indexed: 10/19/2022]
|
29
|
Corte JR, Fang T, Osuna H, Pinto DJP, Rossi KA, Myers JE, Sheriff S, Lou Z, Zheng JJ, Harper TW, Bozarth JM, Wu Y, Luettgen JM, Seiffert DA, Decicco CP, Wexler RR, Quan ML. Structure-Based Design of Macrocyclic Factor XIa Inhibitors: Discovery of the Macrocyclic Amide Linker. J Med Chem 2017; 60:1060-1075. [DOI: 10.1021/acs.jmedchem.6b01460] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- James R. Corte
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Tianan Fang
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Honey Osuna
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Donald J. P. Pinto
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Karen A. Rossi
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Joseph E. Myers
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Steven Sheriff
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Zhen Lou
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Joanna J. Zheng
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Timothy W. Harper
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Jeffrey M. Bozarth
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Yiming Wu
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Joseph M. Luettgen
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Dietmar A. Seiffert
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Carl P. Decicco
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Ruth R. Wexler
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| | - Mimi L. Quan
- Research and Development, Bristol-Myers Squibb Company, P.O. Box
5400, Princeton, New Jersey 08543, United States
| |
Collapse
|
30
|
Abstract
INTRODUCTION Anticoagulants are the mainstay for prevention and/or treatment of thrombotic disorders. Each clinically used anticoagulant is associated with significant adverse consequences, especially bleeding. Factor XIa (FXIa), a key factor involved in the amplification of procoagulation signal, has been suggested as a major target for anticoagulant drug discovery because of reduced risk of bleeding. AREAS COVERED Our literature search uncovered dozens of industrial and academic patents on the discovery of novel FXIa/FXI inhibitors. Small peptidomimetics, sulfated glycosaminoglycan mimetics, polypeptides, antisense oligonucleotides, and monoclonal antibodies have been developed as inhibitors of FXIa. Although many agents are in early discovery/development phases, the activity and safety of a few have been evaluated in various animal models and in humans. EXPERT OPINION FXIa is a promising drug target for development of effective anticoagulants with limited bleeding complications. Literature reveals a major trend in the number of patent applications over the last three years. These inhibitors exploit different approaches for target inhibition. Allosteric modulation of FXIa and biosynthetic inhibition of FXI are mechanistically unique. Despite initial results in patients undergoing knee anthroplasty as with antisense oligonucleotides, major advances should be realized, particularly with respect to pharmacokinetics, for FXI/FXIa inhibitors to enter the clinic.
Collapse
Affiliation(s)
- Rami A Al-Horani
- a Department of Medicinal Chemistry & Institute for Structural Biology , Drug Discovery and Development, Virginia Commonwealth University , Richmond , VA 23219 , USA
| | - Umesh R Desai
- a Department of Medicinal Chemistry & Institute for Structural Biology , Drug Discovery and Development, Virginia Commonwealth University , Richmond , VA 23219 , USA
| |
Collapse
|
31
|
Lee MH, Song HY, Kim H, Park KE, Kim J, Park TK, Kim YJ, Kang NS. The Discovery of New Human Coagulation Factor XIa (FXIa) Inhibitors by Synthesis, Biological Evaluation, and Structure-based Modeling. B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Myeong Hwi Lee
- Graduate School of New Drug Discovery and Development; Chungnam National University; Daejeon 305-764 Korea
| | | | | | | | | | | | | | - Nam Sook Kang
- Graduate School of New Drug Discovery and Development; Chungnam National University; Daejeon 305-764 Korea
| |
Collapse
|
32
|
Corte JR, Fang T, Pinto DJ, Orwat MJ, Rendina AR, Luettgen JM, Rossi KA, Wei A, Ramamurthy V, Myers JE, Sheriff S, Narayanan R, Harper TW, Zheng JJ, Li YX, Seiffert DA, Wexler RR, Quan ML. Orally bioavailable pyridine and pyrimidine-based Factor XIa inhibitors: Discovery of the methyl N-phenyl carbamate P2 prime group. Bioorg Med Chem 2016; 24:2257-72. [DOI: 10.1016/j.bmc.2016.03.062] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/23/2016] [Accepted: 03/30/2016] [Indexed: 12/12/2022]
|
33
|
Wong PC, Quan ML, Watson CA, Crain EJ, Harpel MR, Rendina AR, Luettgen JM, Wexler RR, Schumacher WA, Seiffert DA. In vitro, antithrombotic and bleeding time studies of BMS-654457, a small-molecule, reversible and direct inhibitor of factor XIa. J Thromb Thrombolysis 2015; 40:416-23. [DOI: 10.1007/s11239-015-1258-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
34
|
A clinical and pharmacologic assessment of once-daily versus twice-daily dosing for rivaroxaban. J Thromb Thrombolysis 2015; 38:137-49. [PMID: 24293106 DOI: 10.1007/s11239-013-1029-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Altering doses and regimens of a drug has consequences for the drug's pharmacokinetic and pharmacodynamic profile. Based on a half-life of 5-13 h, it is expected that the Factor Xa inhibitor rivaroxaban would be best suited to a twice-daily rather than a once-daily dose regimen. However, although rivaroxaban is used as a twice-daily regimen for the initial treatment of venous thromboembolism (VTE) and secondary prevention after acute coronary syndromes, the approved dosing is once-daily for prevention of VTE after orthopaedic surgery, long-term secondary prevention of VTE and stroke prevention in patients with non-valvular atrial fibrillation. Rivaroxaban dosing was based on the evaluation of the efficacy and safety of several rivaroxaban doses and regimens in phase II trials. A clear overall advantage of twice-daily dosing compared with once-daily dosing was not documented for indications for which once-daily dosing was subsequently selected. Once-daily dosing was therefore selected for these indications because it is expected to be associated with better compliance than twice-daily dosing, and potentially, with improved outcomes. These studies and data obtained with another Factor Xa inhibitor, edoxaban, in addition to previous experience with low molecular weight heparins, indicate that the clinical impact of once-daily versus twice-daily doses on outcome in terms of efficacy and safety cannot be reliably predicted from pharmacology data, e.g. elimination half-life, obtained during pre-clinical and early phase I clinical studies but rather should be ascertained empirically in phase II and III clinical trials.
Collapse
|
35
|
Corte JR, Fang T, Hangeland JJ, Friends TJ, Rendina AR, Luettgen JM, Bozarth JM, Barbera FA, Rossi KA, Wei A, Ramamurthy V, Morin PE, Seiffert DA, Wexler RR, Quan ML. Pyridine and pyridinone-based factor XIa inhibitors. Bioorg Med Chem Lett 2015; 25:925-30. [DOI: 10.1016/j.bmcl.2014.12.050] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 11/24/2022]
|
36
|
Fjellström O, Akkaya S, Beisel HG, Eriksson PO, Erixon K, Gustafsson D, Jurva U, Kang D, Karis D, Knecht W, Nerme V, Nilsson I, Olsson T, Redzic A, Roth R, Sandmark J, Tigerström A, Öster L. Creating novel activated factor XI inhibitors through fragment based lead generation and structure aided drug design. PLoS One 2015; 10:e0113705. [PMID: 25629509 PMCID: PMC4309560 DOI: 10.1371/journal.pone.0113705] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/27/2014] [Indexed: 11/19/2022] Open
Abstract
Activated factor XI (FXIa) inhibitors are anticipated to combine anticoagulant and profibrinolytic effects with a low bleeding risk. This motivated a structure aided fragment based lead generation campaign to create novel FXIa inhibitor leads. A virtual screen, based on docking experiments, was performed to generate a FXIa targeted fragment library for an NMR screen that resulted in the identification of fragments binding in the FXIa S1 binding pocket. The neutral 6-chloro-3,4-dihydro-1H-quinolin-2-one and the weakly basic quinolin-2-amine structures are novel FXIa P1 fragments. The expansion of these fragments towards the FXIa prime side binding sites was aided by solving the X-ray structures of reported FXIa inhibitors that we found to bind in the S1-S1'-S2' FXIa binding pockets. Combining the X-ray structure information from the identified S1 binding 6-chloro-3,4-dihydro-1H-quinolin-2-one fragment and the S1-S1'-S2' binding reference compounds enabled structure guided linking and expansion work to achieve one of the most potent and selective FXIa inhibitors reported to date, compound 13, with a FXIa IC50 of 1.0 nM. The hydrophilicity and large polar surface area of the potent S1-S1'-S2' binding FXIa inhibitors compromised permeability. Initial work to expand the 6-chloro-3,4-dihydro-1H-quinolin-2-one fragment towards the prime side to yield molecules with less hydrophilicity shows promise to afford potent, selective and orally bioavailable compounds.
Collapse
Affiliation(s)
- Ola Fjellström
- Medicinal Chemistry, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
- * E-mail:
| | - Sibel Akkaya
- Medicinal Chemistry, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Hans-Georg Beisel
- Medicinal Chemistry, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | | | - Karl Erixon
- Medicinal Chemistry, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - David Gustafsson
- Bioscience, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Ulrik Jurva
- Drug Metabolism and Pharmacokinetics, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Daiwu Kang
- Bioscience, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - David Karis
- Medicinal Chemistry, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Wolfgang Knecht
- Bioscience, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Viveca Nerme
- Bioscience, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Ingemar Nilsson
- Medicinal Chemistry, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Thomas Olsson
- Medicinal Chemistry, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Alma Redzic
- Medicinal Chemistry, Cardiovascular & Metabolic Diseases Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Robert Roth
- Discovery Sciences, AstraZeneca R&D, Mölndal, Sweden
| | | | | | - Linda Öster
- Discovery Sciences, AstraZeneca R&D, Mölndal, Sweden
| |
Collapse
|
37
|
Hangeland JJ, Friends TJ, Rossi KA, Smallheer JM, Wang C, Sun Z, Corte JR, Fang T, Wong PC, Rendina AR, Barbera FA, Bozarth JM, Luettgen JM, Watson CA, Zhang G, Wei A, Ramamurthy V, Morin PE, Bisacchi GS, Subramaniam S, Arunachalam P, Mathur A, Seiffert DA, Wexler RR, Quan ML. Phenylimidazoles as Potent and Selective Inhibitors of Coagulation Factor XIa with in Vivo Antithrombotic Activity. J Med Chem 2014; 57:9915-32. [DOI: 10.1021/jm5010607] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jon J. Hangeland
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Todd J. Friends
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Karen A. Rossi
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joanne M. Smallheer
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Cailan Wang
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Zhong Sun
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - James R. Corte
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Tianan Fang
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Pancras C. Wong
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Alan R. Rendina
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Frank A. Barbera
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Jeffrey M. Bozarth
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joseph M. Luettgen
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Carol A. Watson
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Ge Zhang
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Anzhi Wei
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Vidhyashankar Ramamurthy
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Paul E. Morin
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Gregory S. Bisacchi
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Srinath Subramaniam
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Piramanayagam Arunachalam
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Arvind Mathur
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Dietmar A. Seiffert
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Ruth R. Wexler
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Mimi L. Quan
- Research and Development, Bristol-Myers Squibb,
P.O. Box 5400, Princeton, New Jersey 08543, United States
| |
Collapse
|
38
|
Schürmann D, Herzog E, Raquet E, Nolte MW, May F, Müller-Cohrs J, Björkqvist J, Dickneite G, Pragst I. C1-esterase inhibitor treatment: preclinical safety aspects on the potential prothrombotic risk. Thromb Haemost 2014; 112:960-71. [PMID: 25103795 PMCID: PMC6374987 DOI: 10.1160/th13-06-0469] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 06/16/2014] [Indexed: 11/05/2022]
Abstract
Human plasma-derived C1-esterase inhibitor (C1-INH) is an efficacious and safe treatment for hereditary angioedema. However, thrombotic events in subjects treated with C1-INH at recommended or off-label, high doses have been reported. In this study, we addressed the potential prothrombotic risk of C1-INH treatment in high doses using a non-clinical rabbit model. Following intravenous infusion of C1-INH to rabbits at doses up to 800 IU/kg, the exposure and the pharmacodynamic efficacy of C1-INH in rabbits were confirmed by activity measurements of C1-esterase, and coagulation factors XIa and XIIa, respectively. Potential prothrombotic effects were assessed following induction of venous and arterial thrombosis using in vivo models of venous and arterial stasis, complemented by various in vitro assays of coagulation markers. Administration of C1-INH at doses up to 800 IU/kg did not potentiate thrombus formation during venous stasis. In contrast, inhibition of arterial occlusion was observed upon C1-INH administration when compared with isotonic saline treatment, indicating antithrombotic rather than prothrombotic activity of high dose C1-INH treatment in vivo. This was further confirmed in vitro by decreased thrombin generation, increased activated partial thromboplastin time, clotting time and clot formation time, and inhibition of platelet aggregation. No relevant changes in fibrinolysis or in the levels of thrombin-antithrombin complexes, and prothrombin fragment 1+2 were observed upon high dose C1-INH treatment. The data suggest that treatment of healthy rabbits with high doses of C1-INH could potentially inhibit coagulation and thrombus formation rather than induce a prothrombotic risk.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ingo Pragst
- Dr. Ingo Pragst, CSL Behring GmbH, Marburg, Emil-von-Behring Straße 76, 35041 Marburg, Germany, Tel.: +49 6421 39 2799, Fax: +49 6421 39 5310, E-mail:
| |
Collapse
|
39
|
Qiao JX, Wang TC, Hiebert S, Hu CH, Schumacher WA, Spronk SA, Clark CG, Han Y, Hua J, Price LA, Shen H, Chacko SA, Everlof G, Bostwick JS, Steinbacher TE, Li YX, Huang CS, Seiffert DA, Rehfuss R, Wexler RR, Lam PYS. 4-Benzothiazole-7-hydroxyindolinyl diaryl ureas are potent P2Y1 antagonists with favorable pharmacokinetics: low clearance and small volume of distribution. ChemMedChem 2014; 9:2327-43. [PMID: 24989964 DOI: 10.1002/cmdc.201402141] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Indexed: 11/10/2022]
Abstract
Current antithrombotic discovery efforts target compounds that are highly efficacious in thrombus reduction with less bleeding liability than the standard of care. Preclinical data suggest that P2Y1 antagonists may have lower bleeding liabilities than P2Y12 antagonists while providing similar antithrombotic efficacy. This article describes our continuous SAR efforts in a series of 7-hydroxyindolinyl diaryl ureas. When dosed orally, 4-trifluoromethyl-7-hydroxy-3,3-dimethylindolinyl analogue 4 was highly efficacious in a model of arterial thrombosis in rats with limited bleeding. The chemically labile CF3 group in 4 was then transformed to various groups via a novel one-step synthesis, yielding a series of potent P2Y1 antagonists. Among them, the 4-benzothiazole-substituted indolines had desirable PK properties in rats, specifically, low clearance and small volume of distribution. In addition, compound 40 had high i.v. exposure and modest bioavailability, giving it the best overall profile.
Collapse
Affiliation(s)
- Jennifer X Qiao
- Medicinal Chemistry, Molecular Sciences and Candidate Optimization, Bristol-Myers Squibb Company, Rt. 206 and Province Line Road, Princeton, NJ 08543 (USA).
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Al-Horani RA, Desai UR. Designing allosteric inhibitors of factor XIa. Lessons from the interactions of sulfated pentagalloylglucopyranosides. J Med Chem 2014; 57:4805-18. [PMID: 24844380 PMCID: PMC4216218 DOI: 10.1021/jm500311e] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
We recently introduced sulfated pentagalloylglucopyranoside
(SPGG)
as an allosteric inhibitor of factor XIa (FXIa) (Al-Horani et al.,2013, 56, 867–87823316863). To better understand the SPGG–FXIa interaction,
we utilized eight SPGG variants and a range of biochemical techniques.
The results reveal that SPGG’s sulfation level moderately affected
FXIa inhibition potency and selectivity over thrombin and factor Xa.
Variation in the anomeric configuration did not affect potency. Interestingly,
zymogen factor XI bound SPGG with high affinity, suggesting its possible
use as an antidote. Acrylamide quenching experiments suggested that
SPGG induced significant conformational changes in the active site
of FXIa. Inhibition studies in the presence of heparin showed marginal
competition with highly sulfated SPGG variants but robust competition
with less sulfated variants. Resolution of energetic contributions
revealed that nonionic forces contribute nearly 87% of binding energy
suggesting a strong possibility of specific interaction. Overall,
the results indicate that SPGG may recognize more than one anion-binding,
allosteric site on FXIa. An SPGG molecule containing approximately
10 sulfate groups on positions 2 through 6 of the pentagalloylglucopyranosyl
scaffold may be the optimal FXIa inhibitor for further preclinical
studies.
Collapse
Affiliation(s)
- Rami A Al-Horani
- Department of Medicinal Chemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University , 800 E. Leigh Street, Suite 212, Richmond, Virginia 23219, United States
| | | |
Collapse
|
41
|
Inhibition of Factor XI activity as a promising antithrombotic strategy. Drug Discov Today 2014; 19:1435-9. [PMID: 24794465 DOI: 10.1016/j.drudis.2014.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 04/28/2014] [Indexed: 11/21/2022]
Abstract
Prevention and treatment of thromboembolic disorders with minimal bleeding risk remains a significant unmet medical need. Studies in Factor XI (FXI)-deficient humans and experimental animal models suggest that targeting FXI in humans provides antithrombotic benefits with reduced bleeding liability compared with current standard of care. In this review, we describe an exciting era in the discovery and development of antithrombotic agents as multiple therapeutic modalities for FXI(a) inhibition progress through preclinical and clinical development.
Collapse
|
42
|
Geddings JE, Mackman N. New players in haemostasis and thrombosis. Thromb Haemost 2014; 111:570-4. [PMID: 24573314 DOI: 10.1160/th13-10-0812] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 02/10/2014] [Indexed: 01/02/2023]
Abstract
The blood coagulation cascade is essential for haemostasis, but excessive activation can cause thrombosis. Importantly, recent studies have identified factors that contribute to thrombosis but not haemostasis. These include factor XII (FXII), tissue factor-positive microparticles (MPs) and neutrophil extracellular traps (NETs). Studies have shown that FXII plays a role in thrombosis but not haemostasis. FXII is activated in vivo by a variety of negatively-charged polyphosphates, which include extracellular RNA, DNA and inorganic polyphosphate (PolyP) that are released during cell damage and infection. These findings have led to the development of nucleic acid-binding polymers as a new class of anticoagulant drug. Other studies have analysed the role of MPs in experimental thrombosis. MPs are small membrane vesicles released from activated or apoptotic cells. We and others have found that tissue factor-positive MPs enhance thrombosis in mouse models and are elevated in the plasma of pancreatic cancer patients. Finally, NETs have been shown to contribute to experimental venous thrombosis in mouse models and are present in human thrombi. NETs are composed of chromatin fibers that are released from neutrophils undergoing cell death. NETs can capture platelets and increase fibrin deposition. The recent advances in our understanding of the factors contributing to thrombosis in animal models provide new opportunities for the development of safer anticoagulant drugs.
Collapse
Affiliation(s)
| | - Nigel Mackman
- Nigel Mackman, PhD, Division of Hematology/Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA, Tel.: +1 919 843 3961, Fax: +1 919 966 7639, E-mail:
| |
Collapse
|
43
|
Leung PY, Hurst S, Berny-Lang MA, Verbout NG, Gailani D, Tucker EI, Wang RK, McCarty OJT, Gruber A. Inhibition of Factor XII-Mediated Activation of Factor XI Provides Protection Against Experimental Acute Ischemic Stroke in Mice. Transl Stroke Res 2014; 3:381-9. [PMID: 23634198 DOI: 10.1007/s12975-012-0186-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Blood coagulation factor XI (FXI) is an established risk factor for acute ischemic stroke (AIS) and thrombosis, but is also needed for normal hemostasis. Contact factor XII (FXII), an upstream activator of FXI, also contributes to experimental stroke, but is not required for hemostasis. We investigated whether selectively inhibiting FXII-mediated FXI activation, while leaving other FXI and FXII functions intact, could improve the outcome of experimental AIS in mice. Twenty-four hours before induction of AIS by placement of a filament into the internal carotid artery for 60 min, mice were anticoagulated with an antibody directed against the apple 2 domain of FXI. This antibody selectively reduces the prothrombotic activation of FXI by FXIIa but does not affect activated FXI or hemostatic activation of FXI by thrombin, thus leaving hemostasis intact in mice and primates. In this model of AIS/reperfusion injury, mice that received the antibody before AIS displayed less ischemic damage, manifested as reduced cerebral infarction and fibrin deposition (thrombosis), increased cortical reperfusion, and improved neurological behavior. Further, the antibody-anticoagulated mice had no detectable hemostasis impairment. Consistent with the neuroprotective phenotype of FXII-deficient mice, our data suggest that a single molecular event, FXII-mediated FXI activation, contributes to the development of experimental AIS.
Collapse
Affiliation(s)
- Philberta Y Leung
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; Aronora, LLC, Beaverton, OR 97006, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Quan ML, Wong PC, Wang C, Woerner F, Smallheer JM, Barbera FA, Bozarth JM, Brown RL, Harpel MR, Luettgen JM, Morin PE, Peterson T, Ramamurthy V, Rendina AR, Rossi KA, Watson CA, Wei A, Zhang G, Seiffert D, Wexler RR. Tetrahydroquinoline Derivatives as Potent and Selective Factor XIa Inhibitors. J Med Chem 2014; 57:955-69. [DOI: 10.1021/jm401670x] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Mimi L. Quan
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Pancras C. Wong
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Cailan Wang
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Francis Woerner
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Joanne M. Smallheer
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Frank A. Barbera
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Jeffrey M. Bozarth
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Randi L. Brown
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Mark R. Harpel
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Joseph M. Luettgen
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Paul E. Morin
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Tara Peterson
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Vidhyashankar Ramamurthy
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Alan R. Rendina
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Karen A. Rossi
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Carol A. Watson
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Anzhi Wei
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Ge Zhang
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Dietmar Seiffert
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| | - Ruth R. Wexler
- Discovery Chemistry and Cardiovascular
Biology, Research and Development, Bristol-Myers Squibb Company, 311 Pennington-Rocky
Hill Road, Pennington, New
Jersey 08543, United States
| |
Collapse
|
45
|
Qiao JX, Wang TC, Ruel R, Thibeault C, L'Heureux A, Schumacher WA, Spronk SA, Hiebert S, Bouthillier G, Lloyd J, Pi Z, Schnur DM, Abell LM, Hua J, Price LA, Liu E, Wu Q, Steinbacher TE, Bostwick JS, Chang M, Zheng J, Gao Q, Ma B, McDonnell PA, Huang CS, Rehfuss R, Wexler RR, Lam PYS. Conformationally constrained ortho-anilino diaryl ureas: discovery of 1-(2-(1'-neopentylspiro[indoline-3,4'-piperidine]-1-yl)phenyl)-3-(4-(trifluoromethoxy)phenyl)urea, a potent, selective, and bioavailable P2Y1 antagonist. J Med Chem 2013; 56:9275-95. [PMID: 24164581 DOI: 10.1021/jm4013906] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Preclinical antithrombotic efficacy and bleeding models have demonstrated that P2Y1 antagonists are efficacious as antiplatelet agents and may offer a safety advantage over P2Y12 antagonists in terms of reduced bleeding liabilities. In this article, we describe the structural modification of the tert-butyl phenoxy portion of lead compound 1 and the subsequent discovery of a novel series of conformationally constrained ortho-anilino diaryl ureas. In particular, spiropiperidine indoline-substituted diaryl ureas are described as potent, orally bioavailable small-molecule P2Y1 antagonists with improved activity in functional assays and improved oral bioavailability in rats. Homology modeling and rat PK/PD studies on benchmark compound 3l will also be presented. Compound 3l was our first P2Y1 antagonist to demonstrate a robust oral antithrombotic effect with mild bleeding liability in the rat thrombosis and hemostasis models.
Collapse
Affiliation(s)
- Jennifer X Qiao
- Research and Development, Bristol-Myers Squibb Company , 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mehta AY, Jin Y, Desai UR. An update on recent patents on thrombin inhibitors (2010 – 2013). Expert Opin Ther Pat 2013; 24:47-67. [DOI: 10.1517/13543776.2014.845169] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
47
|
Bolton SA, Sutton JC, Anumula R, Bisacchi GS, Jacobson B, Slusarchyk WA, Treuner UD, Wu SC, Zhao G, Pi Z, Sheriff S, Smirk RA, Bisaha S, Cheney DL, Wei A, Schumacher WA, Hartl KS, Liu E, Zahler R, Seiler SM. Discovery of nonbenzamidine factor VIIa inhibitors using a biaryl acid scaffold. Bioorg Med Chem Lett 2013; 23:5239-43. [DOI: 10.1016/j.bmcl.2013.06.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/05/2013] [Accepted: 06/11/2013] [Indexed: 10/26/2022]
|
48
|
Denas G, Pengo V. Investigational anticoagulants for hematological conditions: a new generation of therapies. Expert Opin Investig Drugs 2013; 22:1281-94. [PMID: 23876036 DOI: 10.1517/13543784.2013.821463] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The introduction of novel anticoagulants has had contrasting effects on the agents in the pipeline, fueling the development of some and sinking the others. The complexity of the coagulation cascade offers interesting inhibition choices that might become valid treatment options. AREAS COVERED This review will highlight some of the anticoagulants in the pipeline. Following the success of the direct thrombin and FXa inhibitors already in the market, new agents are being tested. These include AZD0837, betrixaban, letaxaban, darexaban, and LY517717. Targeting other components of the hemostatic pathway might lead to better safety profiles without influencing efficacy. Inhibitors to FVIIa-tissue factor (FVIIa/TF) complex, FIX, FXI, and FXII are being assessed. New inspiring inhibitors are antisense oligonucleotides (ASOs) and aptamers. These are highly specific agents with readily reversible effect and might be engineered to inhibit any coagulation factor. Currently tested ASOs and aptamers are inhibitors of FXI, FXII, thrombin, FIXa, and platelet GPIV. EXPERT OPINION Some of the agents in the pipeline offer valid treatment option for long-term therapy, overcoming some of the drawbacks of the novel anticoagulants. Research is being driven by an expanding market in the anticoagulation field that has been unexploited for a long time.
Collapse
Affiliation(s)
- Gentian Denas
- Padua University Hospital, Cardiology Clinic, Department of Cardiac, Thoracic, and Vascular Sciences , Via Giustiniani 2, 35128 Padova , Italy +390498215658 ; +390498215658 ;
| | | |
Collapse
|
49
|
Sugita C, Yamashita A, Matsuura Y, Iwakiri T, Okuyama N, Matsuda S, Matsumoto T, Inoue O, Harada A, Kitazawa T, Hattori K, Shima M, Asada Y. Elevated plasma factor VIII enhances venous thrombus formation in rabbits: contribution of factor XI, von Willebrand factor and tissue factor. Thromb Haemost 2013; 110:62-75. [PMID: 23636277 DOI: 10.1160/th13-01-0069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 04/12/2013] [Indexed: 01/13/2023]
Abstract
Elevated plasma levels of factor VIII (FVIII) are associated with increased risk of deep venous thrombosis. The aim of this study is to elucidate how elevated FVIII levels affect venous thrombus formation and propagation in vivo. We examined rabbit plasma FVIII activity, plasma thrombin generation, whole blood coagulation, platelet aggregation and venous wall thrombogenicity before and one hour after an intravenous infusion of recombinant human FVIII (rFVIII). Venous thrombus induced by the endothelial denudation of rabbit jugular veins was histologically assessed. Thrombus propagation was evaluated as indocyanine green fluorescence intensity. Argatroban, a thrombin inhibitor, and neutralised antibodies for tissue factor (TF), factor XI (FXI), and von Willebrand factor (VWF) were infused before or after thrombus induction to investigate their effects on venous thrombus formation or propagation. Recombinant FVIII (100 IU/kg) increased rabbit plasma FVIII activity two-fold and significantly enhanced whole blood coagulation and total plasma thrombin generation, but did not affect initial thrombin generation time, platelet aggregation and venous wall thrombogenicity. The rFVIII infusion also increased the size of venous thrombus 1 hour after thrombus induction. Argatroban and the antibodies for TF, FXI or VWF inhibited such enhanced thrombus formation and all except TF suppressed thrombus propagation. In conclusion, elevated plasma FVIII levels enhance venous thrombus formation and propagation. Excess thrombin generation by FXI and VWF-mediated FVIII recruitment appear to contribute to the growth of FVIII-driven venous thrombus.
Collapse
Affiliation(s)
- Chihiro Sugita
- Department of Pathology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Antithrombotic activity of HY023016, a novel Dabigatran prodrug evaluated in animal thrombosis models. Thromb Res 2013; 131:425-35. [DOI: 10.1016/j.thromres.2013.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 02/15/2013] [Accepted: 03/01/2013] [Indexed: 11/23/2022]
|