1
|
Lira AL, Taskin B, Puy C, Keshari RS, Silasi R, Pang J, Aslan JE, Shatzel JJ, Lorentz CU, Tucker EI, Schmaier AH, Gailani D, Lupu F, McCarty OJT. The physicochemical properties of lipopolysaccharide chemotypes regulate activation of the contact pathway of blood coagulation. J Biol Chem 2025; 301:108110. [PMID: 39706265 PMCID: PMC11773025 DOI: 10.1016/j.jbc.2024.108110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/15/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
Lipopolysaccharide (LPS) is the primary pathogenic factor in Gram-negative sepsis. While the presence of LPS in the bloodstream during infection is associated with disseminated intravascular coagulation, the mechanistic link between LPS and blood coagulation activation remains ill-defined. The contact pathway of coagulation-a series of biochemical reactions that initiates blood clotting when plasma factors XII (FXII) and XI (FXI), prekallikrein (PK), and high molecular weight kininogen interact with anionic surfaces-has been shown to be activated in Gram-negative septic patients. In this study, using an in vivo baboon model of Gram-negative Escherichia coli sepsis, we observed activation of the contact pathway including FXII, FXI, and PK. We examined whether E.coli LPS molecules could bind and activate contact pathway members by quantifying the interaction and activation of either FXII, FXI, or PK with each of the three chemotypes of LPS: O111:B4, O26:B6, or Rd2. The LPS chemotypes exhibited distinct physicochemical properties as aggregates and formed complexes with FXII, FXI, and PK. The LPS chemotype O26:B6 uniquely promoted the autoactivation of FXII to FXIIa and, in complex with FXIIa, promoted the cleavage of FXI and PK to generate FXIa and plasma kallikrein, respectively. Furthermore, in complex with the active forms of FXI or PK, LPS chemotypes were able to regulate the catalytic activity of FXIa and plasma kallikrein, respectively, despite the inability to promote the autoactivation of either zymogen. These data suggest that the procoagulant phenotype of E.coli is influenced by bacterial strain and the physicochemical properties of the LPS chemotypes.
Collapse
Affiliation(s)
- André L Lira
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.
| | - Berk Taskin
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Ravi S Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Division of Hematology and Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Christina U Lorentz
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Aronora, Inc, Portland, Oregon, USA
| | - Erik I Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Aronora, Inc, Portland, Oregon, USA
| | - Alvin H Schmaier
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - David Gailani
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
2
|
Lira AL, Kohs TC, Moellmer SA, Shatzel JJ, McCarty OJ, Puy C. Substrates, Cofactors, and Cellular Targets of Coagulation Factor XIa. Semin Thromb Hemost 2024; 50:962-969. [PMID: 36940715 PMCID: PMC11069399 DOI: 10.1055/s-0043-1764469] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023]
Abstract
Coagulation factor XI (FXI) has increasingly been shown to play an integral role in several physiologic and pathological processes. FXI is among several zymogens within the blood coagulation cascade that are activated by proteolytic cleavage, with FXI converting to the active serine protease form (FXIa). The evolutionary origins of FXI trace back to duplication of the gene that transcribes plasma prekallikrein, a key factor in the plasma kallikrein-kinin system, before further genetic divergence led to FXI playing a unique role in blood coagulation. While FXIa is canonically known for activating the intrinsic pathway of coagulation by catalyzing the conversion of FIX into FIXa, it is promiscuous in nature and has been shown to contribute to thrombin generation independent of FIX. In addition to its role in the intrinsic pathway of coagulation, FXI also interacts with platelets, endothelial cells, and mediates the inflammatory response through activation of FXII and cleavage of high-molecular-weight kininogen to generate bradykinin. In this manuscript, we critically review the current body of knowledge surrounding how FXI navigates the interplay of hemostasis, inflammatory processes, and the immune response and highlight future avenues for research. As FXI continues to be clinically explored as a druggable therapeutic target, understanding how this coagulation factor fits into physiological and disease mechanisms becomes increasingly important.
Collapse
Affiliation(s)
- André L. Lira
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Tia C.L. Kohs
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Samantha A. Moellmer
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Cristina Puy
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
3
|
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
|
4
|
Frunt R, El Otmani H, Smits S, Clark CC, Maas C. Factor XII contact activation can be prevented by targeting 2 unique patches in its epidermal growth factor-like 1 domain with a nanobody. J Thromb Haemost 2024; 22:2562-2575. [PMID: 38897387 DOI: 10.1016/j.jtha.2024.06.005] [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: 01/22/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Factor (F)XII triggers contact activation by binding to foreign surfaces, with the epidermal growth factor-like 1 (EGF-1) domain being the primary binding site. Blocking FXII surface-binding might hold therapeutic value to prevent medical device-induced thrombosis. OBJECTIVES To unravel and prevent EGF-1-mediated FXII surface-binding with a variable domain of heavy chain-only antibody (VHH). METHODS FXII variants with glutamine substitutions of 2 positively charged amino acid patches within the EGF-1 domain were created. Their role in FXII contact activation was assessed using kaolin pull-down experiments, amidolytic activity assays, and clotting assays. FXII EGF-1 domain-specific VHHs were raised to inhibit EGF-1-mediated FXII contact activation while preserving quiescence. RESULTS Two unique, positively charged patches in the EGF-1 domain were identified (upstream, 73K74K76K78H81K82H; downstream, 87K113K). Neutralizing the charge of both patches led to a 99% reduction in FXII kaolin binding, subsequent decrease in autoactivation of 94%, and prolongation of clot formation in activated partial thromboplastin time assays from 36 (±2) to 223 (±13) seconds. Three FXII EGF-1-specific VHHs were developed that are capable of inhibiting kaolin binding and subsequent contact system activation in plasma. The most effective VHH "F2" binds the positively charged patches and thereby dose-dependently extends activated partial thromboplastin time clotting times from 29 (±2) to 43 (±3) seconds without disrupting FXII quiescence. CONCLUSION The 2 unique, positively charged patches in FXII EGF-1 cooperatively mediate FXII surface-binding, making both patches crucial for contact activation. Targeting these with FXII EGF-1-specific VHHs can exclusively decrease FXII surface-binding and subsequent contact activation, while preserving zymogen quiescence. These patches thus have potential as druggable targets in preventing medical device-induced thrombosis.
Collapse
Affiliation(s)
- Rowan Frunt
- Central Diagnostic Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - Hinde El Otmani
- Central Diagnostic Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Simone Smits
- Central Diagnostic Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Chantal C Clark
- Center for Benign Hematology, Thrombosis and Hemostasis - Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Coen Maas
- Central Diagnostic Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
5
|
Kharnaf M, Zafar F, Hogue S, Rosenfeldt L, Cantrell RL, Sharma BK, Pearson A, Sprague C, Leino D, Abplanalp WA, Zelek WM, McCrae KR, Shim YJ, Morales D, Tweddell J, Qualls JE, Palumbo JS. Factor XII promotes the thromboinflammatory response in a rat model of venoarterial extracorporeal membrane oxygenation. J Thorac Cardiovasc Surg 2024; 168:e37-e53. [PMID: 37683721 PMCID: PMC10918029 DOI: 10.1016/j.jtcvs.2023.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/26/2023] [Accepted: 08/15/2023] [Indexed: 09/10/2023]
Abstract
BACKGROUND Factor XII (FXII) is a multifunctional protease capable of activating thrombotic and inflammatory pathways. FXII has been linked to thrombosis in extracorporeal membrane oxygenation (ECMO), but the role of FXII in ECMO-induced inflammatory complications has not been studied. We used novel gene-targeted FXII- deficient rats to evaluate the role of FXII in ECMO-induced thromboinflammation. METHODS FXII-deficient (FXII-/-) Sprague-Dawley rats were generated using CRISPR/Cas9. A minimally invasive venoarterial (VA) ECMO model was used to compare wild-type (WT) and FXII-/- rats in 2 separate experimental cohorts: rats placed on ECMO without pharmacologic anticoagulation and rats anticoagulated with argatroban. Rats were maintained on ECMO for 1 hour or until circuit failure occurred. Comparisons were made with unchallenged rats and rats that underwent a sham surgical procedure without ECMO. RESULTS FXII-/- rats were maintained on ECMO without pharmacologic anticoagulation with low resistance throughout the 1-hour experiment. In contrast, WT rats placed on ECMO without anticoagulation developed thrombotic circuit failure within 10 minutes. Argatroban provided a means to maintain WT and FXII-/- rats on ECMO for the 1-hour time frame without thrombotic complications. Analyses of these rats demonstrated that ECMO resulted in increased neutrophil migration into the liver that was significantly blunted by FXII deficiency. ECMO also resulted in increases in high molecular weight kininogen cleavage and complement activation that were abrogated by genetic deletion of FXII. CONCLUSIONS FXII initiates hemostatic system activation and key inflammatory sequelae in ECMO, suggesting that therapies targeting FXII could limit both thromboembolism and inopportune inflammatory complications in this setting.
Collapse
Affiliation(s)
- Mousa Kharnaf
- The Heart Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Farhan Zafar
- The Heart Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Spencer Hogue
- The Heart Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Leah Rosenfeldt
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rachel L Cantrell
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Bal Krishan Sharma
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Amelia Pearson
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Cassandra Sprague
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Daniel Leino
- Department of Pathology, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - William A Abplanalp
- The Heart Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Wioleta M Zelek
- Systems Immunity Research Institute and Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Keith R McCrae
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio
| | - Young Jun Shim
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio
| | - David Morales
- The Heart Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - James Tweddell
- The Heart Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Joseph E Qualls
- Department of Biological Sciences, St Elizabeth College of Natural and Health Sciences, Thomas More University, Crestview Hills, Ky
| | - Joseph S Palumbo
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
6
|
Wan J, Dhrolia S, Kasthuri RR, Prokopenko Y, Ilich A, Saha P, Roest M, Wolberg AS, Key NS, Pawlinski R, Bendapudi PK, Mackman N, Grover SP. Plasma kallikrein supports FXII-independent thrombin generation in mouse whole blood. Blood Adv 2024; 8:3045-3057. [PMID: 38593231 PMCID: PMC11215197 DOI: 10.1182/bloodadvances.2024012613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024] Open
Abstract
ABSTRACT Plasma kallikrein (PKa) is an important activator of factor XII (FXII) of the contact pathway of coagulation. Several studies have shown that PKa also possesses procoagulant activity independent of FXII, likely through its ability to directly activate FIX. We evaluated the procoagulant activity of PKa using a mouse whole blood (WB) thrombin-generation (TG) assay. TG was measured in WB from PKa-deficient mice using contact pathway or extrinsic pathway triggers. PKa-deficient WB had significantly reduced contact pathway-initiated TG compared with that of wild-type controls and was comparable with that observed in FXII-deficient WB. PKa-deficient WB supported equivalent extrinsic pathway-initiated TG compared with wild-type controls. Consistent with the presence of FXII-independent functions of PKa, targeted blockade of PKa with either small molecule or antibody-based inhibitors significantly reduced contact pathway-initiated TG in FXII-deficient WB. Inhibition of activated FXII (FXIIa) using an antibody-based inhibitor significantly reduced TG in PKa-deficient WB, consistent with a PKa-independent function of FXIIa. Experiments using mice expressing low levels of tissue factor demonstrated that persistent TG present in PKa- and FXIIa-inhibited WB was driven primarily by endogenous tissue factor. Our work demonstrates that PKa contributes significantly to contact pathway-initiated TG in the complex milieu of mouse WB, and a component of this contribution occurs in an FXII-independent manner.
Collapse
Affiliation(s)
- Jun Wan
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
| | - Sophia Dhrolia
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rohan R. Kasthuri
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Yuriy Prokopenko
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Anton Ilich
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Prakash Saha
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, King’s College London, London, United Kingdom
| | - Mark Roest
- Synapse Research Institute, Maastricht, The Netherlands
| | - Alisa S. Wolberg
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nigel S. Key
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rafal Pawlinski
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Pavan K. Bendapudi
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Nigel Mackman
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Steven P. Grover
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
7
|
Prakash S, Mares AC, Porres-Aguilar M, Mukherjee D, Barnes GD. Factor XI/XIa inhibitors for the prevention and treatment of venous and arterial thromboembolism: A narrative review. Vasc Med 2024; 29:85-92. [PMID: 37947131 DOI: 10.1177/1358863x231206778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
During the past decade, direct oral anticoagulants (DOACs) have advanced and simplified the prevention and treatment of venous thromboembolism (VTE). However, there remains a high incidence of bleeds, which calls for agents that have a reduced risk of bleeding. Factor XI (FXI) deficiency is associated with lower rates of venous thrombosis and stroke compared to the general population with a lower risk of bleeding. In conjunction with this, phase 2 studies have demonstrated safety and the potential for reduced thrombotic events with FXI inhibitors as compared to currently available medications. The aim of this review is to summarize key data on the clinical pharmacology of FXI, the latest developments in clinical trials of FXI inhibitors, and to describe the efficacy and safety profiles of FXI inhibitors for the prevention of venous and arterial thromboembolism.
Collapse
Affiliation(s)
- Swathi Prakash
- Department of Internal Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Adriana C Mares
- Department of Internal Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Mateo Porres-Aguilar
- Department of Internal Medicine, Divisions of Hospital and Adult Thrombosis Medicine, Texas Tech University Health Sciences Center and Paul L Foster School of Medicine, El Paso, TX, USA
| | - Debabrata Mukherjee
- Division of Cardiovascular Diseases, Texas Tech University Health Sciences Center and Paul L Foster School of Medicine, El Paso, TX, USA
| | - Geoffrey D Barnes
- Department of Internal Medicine, Division of Cardiovascular Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Páramo JA, Marcos-Jubilar M. [Factor XI inhibitors: A new era in antithrombotic therapy]. Med Clin (Barc) 2024; 162:73-76. [PMID: 37863734 DOI: 10.1016/j.medcli.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 10/22/2023]
Affiliation(s)
- José A Páramo
- Servicio de Hematología y Hemoterapia, Clínica Universidad de Navarra, Pamplona, España.
| | - María Marcos-Jubilar
- Servicio de Hematología y Hemoterapia, Clínica Universidad de Navarra, Pamplona, España
| |
Collapse
|
9
|
Skoptsova AA, Geronikaki A, Novichikhina NP, Sulimov AV, Ilin IS, Sulimov VB, Bykov GA, Podoplelova NA, Pyankov OV, Shikhaliev KS. Design, Synthesis, and Evaluation of New Hybrid Derivatives of 5,6-Dihydro-4 H-pyrrolo[3,2,1- ij]quinolin-2(1 H)-one as Potential Dual Inhibitors of Blood Coagulation Factors Xa and XIa. Molecules 2024; 29:373. [PMID: 38257286 PMCID: PMC10818416 DOI: 10.3390/molecules29020373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular diseases caused by blood coagulation system disorders are one of the leading causes of morbidity and mortality in the world. Research shows that blood clotting factors are involved in these thrombotic processes. Among them, factor Xa occupies a key position in the blood coagulation cascade. Another coagulation factor, XIa, is also a promising target because its inhibition can suppress thrombosis with a limited contribution to normal hemostasis. In this regard, the development of dual inhibitors as new generation anticoagulants is an urgent problem. Here we report the synthesis and evaluation of novel potential dual inhibitors of coagulation factors Xa and XIa. Based on the principles of molecular design, we selected a series of compounds that combine in their structure fragments of pyrrolo[3,2,1-ij]quinolin-2-one and thiazole, connected through a hydrazine linker. The production of new hybrid molecules was carried out using a two-stage method. The reaction of 5,6-dihydropyrrolo[3,2,1-ij]quinoline-1,2-diones with thiosemicarbazide gave the corresponding hydrazinocarbothioamides. The reaction of the latter with DMAD led to the target methyl 2-(4-oxo-2-(2-(2-oxo-5,6-dihydro-4H-pyrrolo[3,2,1-ij]quinolin-1(2H)-ylidene)hydrazineyl)thiazol-5(4H)-ylidene)acetates in high yields. In vitro testing of the synthesized molecules revealed that ten of them showed high inhibition values for both the coagulation factors Xa and XIa, and the IC50 value for some compounds was also assessed. The resulting structures were also tested for their ability to inhibit thrombin.
Collapse
Affiliation(s)
- Anna A. Skoptsova
- Department of Organic Chemistry, Faculty of Chemistry, Voronezh State University, 1 Universitetskaya Sq., 394018 Voronezh, Russia; (A.A.S.); (N.P.N.)
| | - Athina Geronikaki
- School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Nadezhda P. Novichikhina
- Department of Organic Chemistry, Faculty of Chemistry, Voronezh State University, 1 Universitetskaya Sq., 394018 Voronezh, Russia; (A.A.S.); (N.P.N.)
| | - Alexey V. Sulimov
- Research Computing Center, Lomonosov Moscow State University, 119992 Moscow, Russia; (A.V.S.); (I.S.I.); (V.B.S.)
| | - Ivan S. Ilin
- Research Computing Center, Lomonosov Moscow State University, 119992 Moscow, Russia; (A.V.S.); (I.S.I.); (V.B.S.)
| | - Vladimir B. Sulimov
- Research Computing Center, Lomonosov Moscow State University, 119992 Moscow, Russia; (A.V.S.); (I.S.I.); (V.B.S.)
| | - Georgii A. Bykov
- Department of Biophysics at the Faculty of Physics, Lomonosov Moscow State University, 119992 Moscow, Russia;
| | | | - Oleg V. Pyankov
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Russia;
| | - Khidmet S. Shikhaliev
- Department of Organic Chemistry, Faculty of Chemistry, Voronezh State University, 1 Universitetskaya Sq., 394018 Voronezh, Russia; (A.A.S.); (N.P.N.)
| |
Collapse
|
10
|
Mohammed BM, Sun MF, Cheng Q, Litvak M, McCrae KR, Emsley J, McCarty OJT, Gailani D. High molecular weight kininogen interactions with the homologs prekallikrein and factor XI: importance to surface-induced coagulation. J Thromb Haemost 2024; 22:225-237. [PMID: 37813198 PMCID: PMC10841474 DOI: 10.1016/j.jtha.2023.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND In plasma, high molecular weight kininogen (HK) is either free or bound to prekallikrein (PK) or factor (F) XI (FXI). During contact activation, HK is thought to anchor PK and FXI to surfaces, facilitating their conversion to the proteases plasma kallikrein and FXIa. Mice lacking HK have normal hemostasis but are resistant to injury-induced arterial thrombosis. OBJECTIVES To identify amino acids on the HK-D6 domain involved in PK and FXI binding and study the importance of the HK-PK and HK-FXI interactions to coagulation. METHODS Twenty-four HK variants with alanine replacements spanning residues 542-613 were tested in PK/FXI binding and activated partial thromboplastin time clotting assays. Surface-induced FXI and PK activation in plasma were studied in the presence or absence of HK. Kng1-/- mice lacking HK were supplemented with human or murine HK and tested in an arterial thrombosis model. RESULTS Overlapping binding sites for PK and FXI were identified in the HK-D6 domain. HK variants with defects only in FXI binding corrected the activated partial thromboplastin time of HK-deficient plasma poorly compared to a variant defective only in PK-binding. In plasma, HK deficiency appeared to have a greater deleterious effect on FXI activation than PK activation. Human HK corrected the defect in arterial thrombus formation in HK-deficient mice poorly due to a specific defect in binding to mouse FXI. CONCLUSION Clinical observations indicate FXI is required for hemostasis, while HK is not. Yet, the HK-FXI interaction is required for contact activation-induced clotting in vitro and in vivo suggesting an important role in thrombosis and perhaps other FXI-related activities.
Collapse
Affiliation(s)
- Bassem M Mohammed
- Edward A. Doisy Research Center, Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, St. Louis, Missouri, USA.
| | - Mao-Fu Sun
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Qiufang Cheng
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maxim Litvak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Keith R McCrae
- Department of Hematology and Oncology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jonas Emsley
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Owen J T McCarty
- Department of Biomedical Engineering, Division of Hematology/Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
11
|
Padilla S, Prado R, Anitua E. An evolutionary history of F12 gene: Emergence, loss, and vulnerability with the environment as a driver. Bioessays 2023; 45:e2300077. [PMID: 37750435 DOI: 10.1002/bies.202300077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
In the context of macroevolutionary transitions, environmental changes prompted vertebrates already bearing genetic variations to undergo gradual adaptations resulting in profound anatomical, physiological, and behavioral adaptations. The emergence of new genes led to the genetic variation essential in metazoan evolution, just as was gene loss, both sources of genetic variation resulting in adaptive phenotypic diversity. In this context, F12-coding protein with defense and hemostatic roles emerged some 425 Mya, and it might have contributed in aquatic vertebrates to the transition from water-to-land. Conversely, the F12 loss in marine, air-breathing mammals like cetaceans has been associated with phenotypic adaptations in some terrestrial mammals in their transition to aquatic lifestyle. More recently, the advent of technological innovations in western lifestyle with blood-contacting devices and harmful environmental nanoparticles, has unfolded new roles of FXII. Environment operates as either a positive or a relaxed selective pressure on genes, and consequently genes are selected or lost. FXII, an old dog facing environmental novelties can learn new tricks and teach us new therapeutic avenues.
Collapse
Affiliation(s)
- Sabino Padilla
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Roberto Prado
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Eduardo Anitua
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| |
Collapse
|
12
|
Tamargo J, Agewall S, Borghi C, Ceconi C, Cerbai E, Dan GA, Ferdinandy P, Grove EL, Rocca B, Sulzgruber P, Semb AG, Sossalla S, Niessner A, Kaski JC, Dobrev D. New pharmacological agents and novel cardiovascular pharmacotherapy strategies in 2022. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2023; 9:pvad034. [PMID: 37169875 PMCID: PMC10236523 DOI: 10.1093/ehjcvp/pvad034] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/10/2023] [Accepted: 05/10/2023] [Indexed: 05/13/2023]
Abstract
Cardiovascular diseases (CVD) remain the leading cause of death worldwide and pharmacotherapy of most of them is suboptimal. Thus, there is a clear unmet clinical need to develop new pharmacological strategies with greater efficacy and better safety profiles. In this review, we summarize the most relevant advances in cardiovascular pharmacology in 2022 including the approval of first-in-class drugs that open new avenues for the treatment of obstructive hypertrophic cardiomyopathy (mavacamten), type 2 diabetes mellitus (tirzepatide), and heart failure (HF) independent of left ventricular ejection fraction (sodium-glucose cotransporter 2 inhibitors). We also dealt with fixed dose combination therapies repurposing different formulations of "old" drugs with well-known efficacy and safety for the treatment of patients with acute decompensated HF (acetazolamide plus loop diuretics), atherosclerotic cardiovascular disease (moderate-dose statin plus ezetimibe), Marfan syndrome (angiotensin receptor blockers plus β-blockers), and secondary cardiovascular prevention (i.e. low-dose aspirin, ramipril and atorvastatin), thereby filling existing gaps in knowledge, and opening new avenues for the treatment of CVD. Clinical trials confirming the role of dapagliflozin in patients with HF and mildly reduced or preserved ejection fraction, long-term evolocumab to reduce the risk of cardiovascular events, vitamin K antagonists for stroke prevention in patients with rheumatic heart disease-associated atrial fibrillation, antibiotic prophylaxis in patients at high risk for infective endocarditis before invasive dental procedures, and vutrisiran for the treatment of hereditary transthyretin-related amyloidosis with polyneuropathy were also reviewed. Finally, we briefly discuss recent clinical trials suggesting that FXIa inhibitors may have the potential to uncouple thrombosis from hemostasis and attenuate/prevent thromboembolic events with minimal disruption of hemostasis.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense, Instituto de Investigación Sanitaria Gregorio Marañón, Plaza de Ramón y Cajal s/n, Madrid 28040, Spain
| | - Stefan Agewall
- Department of Cardiology, Oslo University Hospital and Institute of Clinical Medicine, Oslo University, Oslo, Norvay
| | - Claudio Borghi
- Department of Cardiovascular Medicine, University of Bologna-IRCCS AOU S. Orsola, Bologna, Italy
| | - Claudio Ceconi
- Unit of Cardiologia, ASST Garda, Desenzano del Garda, Italy
| | - Elisabetta Cerbai
- Department Neurofarba, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Gheorghe A Dan
- “Carol Davila” University of Medicine, Colentina University Hospital, Bucharest, Romania
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Erik Lerkevang Grove
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Bianca Rocca
- Section of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - Patrick Sulzgruber
- Department of Medicine, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Anne Grete Semb
- Preventive Cario-Rheuma Clinic, Division of Research and Innovation, REMEDY Centre, Diakonhjemmet Hospital, Oslo, Norway
| | - Samuel Sossalla
- Department of Internal Medicine II, University Regensburg, Regensburg, Germany
| | - Alexander Niessner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Juan Carlos Kaski
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Dobromir Dobrev
- Institute of Pharmacology, West-German Heart and Vascular Centre, University Duisburg-Essen, Essen, Germany
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
13
|
Tweddell JS, Kharnaf M, Zafar F, Riggs KW, Reagor JA, Monia BP, Revenko A, Leino DG, Owens AP, Martin JK, Gourley B, Rosenfeldt L, Palumbo JS. Targeting the contact system in a rabbit model of extracorporeal membrane oxygenation. Blood Adv 2023; 7:1404-1417. [PMID: 36240297 PMCID: PMC10139951 DOI: 10.1182/bloodadvances.2022007586] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022] Open
Abstract
Previous studies suggested that contact pathway factors drive thrombosis in mechanical circulation. We used a rabbit model of veno-arterial extracorporeal circulation (VA-ECMO) to evaluate the role of factors XI and XII in ECMO-associated thrombosis and organ damage. Factors XI and XII (FXI, FXII) were depleted using established antisense oligonucleotides before placement on a blood-primed VA-ECMO circuit. Decreasing FXII or FXI to < 5% of baseline activity significantly prolonged ECMO circuit lifespan, limited the development of coagulopathy, and prevented fibrinogen consumption. Histological analysis suggested that FXII depletion mitigated interstitial pulmonary edema and hemorrhage whereas heparin and FXI depletion did not. Neither FXI nor FXII depletion was associated with significant hemorrhage in other organs. In vitro analysis showed that membrane oxygenator fibers (MOFs) alone are capable of driving significant thrombin generation in a FXII- and FXI-dependent manner. MOFs also augment thrombin generation triggered by low (1 pM) or high (5 pM) tissue factor concentrations. However, only FXI elimination completely prevented the increase in thrombin generation driven by MOFs, suggesting MOFs augment thrombin-mediated FXI activation. Together, these results suggest that therapies targeting FXII or FXI limit thromboembolic complications associated with ECMO. Further studies are needed to determine the contexts wherein targeting FXI and FXII, either alone or in combination, would be most beneficial in ECMO. Moreover, studies are also needed to determine the potential mechanisms coupling FXII to end-organ damage in ECMO.
Collapse
Affiliation(s)
- James S. Tweddell
- The Heart Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Mousa Kharnaf
- The Heart Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Farhan Zafar
- The Heart Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kyle W. Riggs
- The Heart Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | - James A. Reagor
- The Heart Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | | | | | - Daniel G. Leino
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | - A. Phillip Owens
- Department of Internal Medicine, The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Janine K. Martin
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Benjamin Gourley
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Leah Rosenfeldt
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| | - Joseph S. Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
14
|
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: 1.0] [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
|
15
|
Zhou S, Zhao W, Hu J, Mao C, Zhou M. Application of Nanotechnology in Thrombus Therapy. Adv Healthc Mater 2023; 12:e2202578. [PMID: 36507827 DOI: 10.1002/adhm.202202578] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/26/2022] [Indexed: 12/14/2022]
Abstract
A thrombus is a blood clot that forms in the lumen of an artery or vein, restricting blood flow and causing clinical symptoms. Thrombosis is associated with many life-threatening cardiovascular diseases. However, current clinical therapeutic technologies still have many problems in targeting, enrichment, penetration, and safety to meet the thrombosis treatment needs. Therefore, researchers devote themselves to developing nanosystems loaded with antithrombotic drugs to address this paradox in recent years. Herein, the existing thrombosis treatment technologies are first reviewed; and then, their advantages and disadvantages are outlined based on a brief discussion of thrombosis's definition and formation mechanism. Furthermore, the need and application cases for introducing nanotechnology are discussed, focusing on thrombus-specific targeted ligand modification technology and microenvironment-triggered responsive drug release technology. Then, nanomaterials that can be used to design antithrombotic nanotherapeutic systems are summarized. Moreover, a variety of drug delivery technologies driven by nanomotors in thrombosis therapy is also introduced. Last of all, a prospective discussion on the future development of nanotechnology for thrombosis therapy is highlighted.
Collapse
Affiliation(s)
- Shuyin Zhou
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.,Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Wenbo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing, 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Min Zhou
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
16
|
Koulas I, Spyropoulos AC. A Review of FXIa Inhibition as a Novel Target for Anticoagulation. Hamostaseologie 2023; 43:28-36. [PMID: 36807817 DOI: 10.1055/a-1984-7021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Limitations of vitamin K antagonists as chronic oral anticoagulant therapy have largely been supplanted by direct factor IIa and factor Xa inhibitor oral anticoagulants with similar efficacy but an overall better safety profile, lack of routine monitoring, and very limited drug-drug interactions compared with agents such as warfarin. However, an increased risk of bleeding remains even with these new-generation oral anticoagulants in fragile patient populations, in patients requiring dual or triple antithrombotic therapy, or high bleed risk surgeries. Epidemiologic data in patients with hereditary factor XI deficiency and preclinical studies support the notion that factor XIa inhibitors have the ability to be an effective but potentially safer alternative to existing anticoagulants, based on their ability to prevent thrombosis directly within the intrinsic pathway without affecting hemostatic mechanisms. As such, various types of factor XIa inhibitors have been studied in early phase clinical studies, including inhibitors of the biosynthesis of factor XIa with antisense oligonucleotides or direct inhibitors of factor XIa using small peptidomimetic molecules, monoclonal antibodies, aptamers, or natural inhibitors. In this review, we discuss how different types of factor XIa inhibitors work and present findings from recently published Phase II clinical trials across multiple indications, including stroke prevention in atrial fibrillation, dual pathway inhibition with concurrent antiplatelets post-myocardial infarction, and thromboprophylaxis of orthopaedic surgery patients. Finally, we refer to ongoing Phase III clinical trials of factor XIa inhibitors and their potential to provide definitive answers regarding their safety and efficacy in preventing thromboembolic events in specific patient groups.
Collapse
Affiliation(s)
- Ioannis Koulas
- The Institute of Health Systems Science at the Feinstein Institutes for Medical Research, Manhasset, New York, United States
| | - Alex C Spyropoulos
- The Institute of Health Systems Science at the Feinstein Institutes for Medical Research, Manhasset, New York, United States.,Northwell Health at Lenox Hill Hospital, New York, New York, United States.,The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, United States
| |
Collapse
|
17
|
Luo Q, Wenzel P. Properdin (factor P) as a new target cleaved by factor XIa: Intrinsic coagulation at the crossroads with inflammation. Res Pract Thromb Haemost 2022; 6:e12835. [PMID: 36349265 PMCID: PMC9634820 DOI: 10.1002/rth2.12835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Qi Luo
- Center for Thrombosis and Hemostasis MainzUniversity Medical Center MainzMainzGermany
- Department of Biochemistry, CARIM School for Cardiovascular ResearchMaastricht UniversityMaastrichtThe Netherlands
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis MainzUniversity Medical Center MainzMainzGermany
- Department of Biochemistry, CARIM School for Cardiovascular ResearchMaastricht UniversityMaastrichtThe Netherlands
- Center for Cardiology, Cardiology IUniversity Medical Center MainzMainzGermany
- DZHK (German Center for Cardiovascular Research), Partner Site Rhine MainUniversity Medical Center MainzMainzGermany
| |
Collapse
|
18
|
Santagata D, Cammà G, Donadini MP, Squizzato A, Ageno W. Current and emerging drug strategies for the prevention of venous thromboembolism in acutely ill medical inpatients. Expert Opin Pharmacother 2022; 23:1651-1665. [PMID: 36154548 DOI: 10.1080/14656566.2022.2128757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Venous thromboembolism (VTE) is a common complication in patients hospitalized for acute medical illnesses. Therefore, medical inpatients require a careful VTE and bleeding risk assessment to drive optimal strategies for VTE prevention. Low molecular weight heparin and fondaparinux have long been used for inhospital prophylaxis for patients at increased risk of VTE. The selection of patients who require post-discharge prophylaxis, and the role of direct oral anticoagulants remain debated. New molecules currently under development may contribute to improve the risk benefit of VTE prevention in this setting. AREAS COVERED This text summarizes the evidence on approved treatments and on other drugs for the prevention of VTE in acutely ill medical patients. The main focus is on their pharmacological proprieties, clinical efficacy and safety, and the current license approved by the FDA (Food and Drug Administration) and EMA (European Medicines Agency), giving the readers a way to compare available drugs to date. The trials presented consider both inhospital and extended prophylaxis. EXPERT OPINION Thanks to the potentially favorable safety profile, factor XI inhibitors may play a role in the prevention of VTE in this setting. The expert opinion section discusses pharmacological properties, prophylaxis trials, and potential clinical applications of this novel class of drugs.
Collapse
Affiliation(s)
- D Santagata
- Department of Medicine and Surgery, Research Center on Thromboembolic Disorders and Antithrombotic Therapies, University of Insubria, Via Gucciardini 9, 21100, Varese and Como, Italy
| | - G Cammà
- Department of Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Curore, Largo Francesco Vito 1, 00139, Rome, Italy
| | - M P Donadini
- Department of Medicine and Surgery, Research Center on Thromboembolic Disorders and Antithrombotic Therapies, University of Insubria, Via Gucciardini 9, 21100, Varese and Como, Italy
| | - A Squizzato
- Department of Medicine and Surgery, Research Center on Thromboembolic Disorders and Antithrombotic Therapies, University of Insubria, Via Ravona 20 San Fermo della Battaglia (Como), 22042 Como, Italy
| | - W Ageno
- Department of Medicine and Surgery, Research Center on Thromboembolic Disorders and Antithrombotic Therapies, University of Insubria, Via Gucciardini 9, 21100, Varese and Como, Italy
| |
Collapse
|
19
|
Association of FXI activity with thrombo-inflammation, extracellular matrix, lipid metabolism and apoptosis in venous thrombosis. Sci Rep 2022; 12:9761. [PMID: 35697739 PMCID: PMC9192691 DOI: 10.1038/s41598-022-13174-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/17/2022] [Indexed: 12/31/2022] Open
Abstract
Animal experiments and early phase human trials suggest that inhibition of factor XIa (FXIa) safely prevents venous thromboembolism (VTE), and specific murine models of sepsis have shown potential efficacy in alleviating cytokine storm. These latter findings support the role of FXI beyond coagulation. Here, we combine targeted proteomics, machine learning and bioinformatics, to discover associations between FXI activity (FXI:C) and the plasma protein profile of patients with VTE. FXI:C was measured with a modified activated partial prothrombin time (APTT) clotting time assay. Proximity extension assay-based protein profiling was performed on plasma collected from subjects from the Genotyping and Molecular Phenotyping of Venous Thromboembolism (GMP-VTE) Project, collected during an acute VTE event (n = 549) and 12-months after (n = 187). Among 444 proteins investigated, N = 21 and N = 66 were associated with FXI:C during the acute VTE event and at 12 months follow-up, respectively. Seven proteins were identified as FXI:C-associated at both time points. These FXI-related proteins were enriched in immune pathways related to causes of thrombo-inflammation, extracellular matrix interaction, lipid metabolism, and apoptosis. The results of this study offer important new avenues for future research into the multiple properties of FXI, which are of high clinical interest given the current development of FXI inhibitors.
Collapse
|
20
|
Poenou G, Dumitru Dumitru T, Lafaie L, Mismetti V, Heestermans M, Bertoletti L. Factor XI Inhibition for the Prevention of Venous Thromboembolism: An Update on Current Evidence and Future perspectives. Vasc Health Risk Manag 2022; 18:359-373. [PMID: 35707632 PMCID: PMC9191224 DOI: 10.2147/vhrm.s331614] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/30/2022] [Indexed: 12/18/2022] Open
Abstract
During the past decade, emergence of direct oral anticoagulants (DOACs) has drastically improved the prevention of thrombosis. However, several unmet needs prevail in the field of thrombosis prevention, even in the DOACs’ era. The use of DOACs is still constrained and the drugs cannot be administered in every clinical scenario, such as an increased anticoagulant-associated bleeding risk, particularly in some specific populations (cancer – notably those with gastrointestinal or genitourinary cancer – and frail patients), the impossibility to be used in certain patients (eg, end-stage kidney failure during hemodialysis, pregnancy and breastfeeding), and their lack of efficacy in certain clinical scenarios (eg, mechanical heart valves, triple-positive antiphospholipid syndrome). Efforts to find a factor that upon antagonization prevents thrombosis but spares haemostasis have resulted in the identification of coagulation factor XI (FXI) as a therapeutic target. After briefly recapitulating the role of factor XI in the balance of haemostasis, we propose a narrative review of the key data published to date with compounds targeting factor XI to prevent thrombosis as well as the main ongoing clinical studies, opening up prospects for improving the care of patients requiring thrombosis prevention.
Collapse
Affiliation(s)
- Geraldine Poenou
- Therapeutic and Vascular Medicine Department, University Hospital of Saint Etienne, Saint Etienne, France
| | - Teona Dumitru Dumitru
- Therapeutic and Vascular Medicine Department, University Hospital of Saint Etienne, Saint Etienne, France
- Internal Medicine Department, University Hospital Santa Lucía, Cartagena, Murcia, Spain
- Catholic University San Antonio, Murcia, Spain
| | - Ludovic Lafaie
- Geriatric Department, University Hospital of Saint Etienne, Saint Etienne, France
- INSERM, UMR1059, Haemostasis and Vascular Dysfunction Team, Jean Monnet University, Saint-Etienne, F-42055, France
| | - Valentine Mismetti
- INSERM, UMR1059, Haemostasis and Vascular Dysfunction Team, Jean Monnet University, Saint-Etienne, F-42055, France
- Pneumology Department, University Hospital of Saint Etienne, Saint Etienne, France
| | - Marco Heestermans
- INSERM, UMR1059, Haemostasis and Vascular Dysfunction Team, Jean Monnet University, Saint-Etienne, F-42055, France
- Auvergne-Rhône-Alpes French Blood Donation Agency, Saint-Etienne, F-42100, France
| | - Laurent Bertoletti
- Therapeutic and Vascular Medicine Department, University Hospital of Saint Etienne, Saint Etienne, France
- INSERM, UMR1059, Haemostasis and Vascular Dysfunction Team, Jean Monnet University, Saint-Etienne, F-42055, France
- INSERM, CIC-1408, University Hospital of Saint Etienne, Saint Etienne, France
- Correspondence: Laurent Bertoletti, Therapeutic and Vascular Medicine Department, University Hospital of Saint Etienne, Saint Etienne, France, Tel +33477827771, Fax +33477820482, Email
| |
Collapse
|
21
|
Mosconi MG, Paciaroni M, Ageno W. Investigational drugs for ischemic stroke: what's in the clinical development pipeline for acute phase and prevention? Expert Opin Investig Drugs 2022; 31:645-667. [PMID: 35486110 DOI: 10.1080/13543784.2022.2072725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Stroke is a leading cause of disability and mortality and its burden expected to increase. The only approved drug for acute ischemic stroke is the intravenous thrombolytic alteplase. The risk of bleeding complications is one of the reasons for the undertreatment of eligible patients. Numerous drugs are currently being developed to improve safety-efficacy. AREAS COVERED We reviewed literature from January 1st, 2000, to 15th January 2022 for the development and testing of novel drugs with the aim of targeting treatment at prevention of ischemic stroke: PubMed, MEDLINE, Google Scholar, and ClinicalTrial.gov. EXPERT OPINION The pathophysiology of ischemic stroke involves multiple pathways causing cerebral artery obstruction and brain tissue ischemia. Data suggest that tenecteplase is a more promising fibrinolytic agent with a superior efficacy-safety profile, compared to the currently approved alteplase. Current guidelines consider a short-term cycle of mannitol or hypertonic saline to be advisable in patients with space-occupying hemispheric infarction. Regarding primary and secondary prevention, research is primarily focused on identifying mechanisms to improve the safety-efficacy profile using a "hemostasis-sparing" approach. Further evaluation on those agents that have already shown promise for their risk/benefit profiles, would benefit greatly a neurologist's capacity to successfully prevent and treat ischemic stroke patients.
Collapse
Affiliation(s)
- Maria Giulia Mosconi
- Emergency and vascular medicine Stroke Unit University of Perugia, Perugia, Italy
| | - Maurizio Paciaroni
- Emergency and vascular medicine Stroke Unit University of Perugia, Perugia, Italy
| | - Walter Ageno
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
22
|
Abstract
Thrombin is a multifunctional serine protease generated in injured cells. The generation of thrombin in coagulation plays a central role in the functioning of haemostasis. The last enzyme in the coagulation cascade is thrombin, with the function of cleaving fibrinogen to fibrin, which forms the fibrin clot of a haemostatic plug. Although thrombin primarily converts fibrinogen to fibrin, it also has many other positive regulatory effects on coagulation. Thrombin has procoagulant, inflammatory, cellular proliferation and anticoagulant effects. In coagulation system, thrombin has two very distinct roles. Firstly, it acts as a procoagulant when it converts fibrinogen into an insoluble fibrin clot, activates factor (F) XIII, activates thrombin activatable fibrinolysis inhibitor (TAFI) and activates FV, FVIII and FXI. Thrombin also enhances platelet adhesion by inactivating a disintegrin and metalloprotease with thrombospondin type1 motif (ADAMTS13). However, when thrombin activates protein C, it acts as an anticoagulant. A natural anticoagulant pathway that supplies regulation of the blood coagulation system contains protein C, which is the key component. This is accomplished by the specific proteolytic inactivation of FV and FVIII. In this review, the multiple roles of thrombin in the haemostatic response to injury are studied in addition to the cofactors that determine thrombin activity and how thrombin activity is thought to be coordinated.
Collapse
|
23
|
The versatile role of the contact system in cardiovascular disease, inflammation, sepsis and cancer. Biomed Pharmacother 2021; 145:112429. [PMID: 34801854 DOI: 10.1016/j.biopha.2021.112429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 11/24/2022] Open
Abstract
The human contact system consists of plasma proteins, which - after contact to foreign surfaces - are bound to them, thereby activating the zymogens of the system into enzymes. This activation mechanism gave the system its name - contact system. It is considered as a procoagulant and proinflammatory response mechanism, as activation finally leads to the generation of fibrin and bradykinin. To date, no physiological processes have been described that are mediated by contact activation. However, contact system factors play a pathophysiological role in numerous diseases, such as cardiovascular diseases, arthritis, colitis, sepsis, and cancer. Contact system factors are therefore an interesting target for new therapeutic options in different clinical conditions.
Collapse
|
24
|
Narducci ML, Patrono C. Abelacimab and factor XI inhibition: a novel mechanism for the prevention of venous thromboembolism. Eur Heart J 2021; 42:4109-4110. [PMID: 34519340 DOI: 10.1093/eurheartj/ehab590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Maria Lucia Narducci
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A.Gemelli IRCCS, Catholic University School of Medicine, Largo A. Gemelli, 8, Rome 00168, Italy
| | - Carlo Patrono
- Department of Pharmacology, Fondazione Policlinico Universitario A.Gemelli IRCCS, Catholic University School of Medicine, Largo A. Gemelli, 8, Rome 00168, Italy
| |
Collapse
|
25
|
Identification of the factor XII contact activation site enables sensitive coagulation diagnostics. Nat Commun 2021; 12:5596. [PMID: 34552086 PMCID: PMC8458485 DOI: 10.1038/s41467-021-25888-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/31/2021] [Indexed: 11/08/2022] Open
Abstract
Contact activation refers to the process of surface-induced activation of factor XII (FXII), which initiates blood coagulation and is captured by the activated partial thromboplastin time (aPTT) assay. Here, we show the mechanism and diagnostic implications of FXII contact activation. Screening of recombinant FXII mutants identified a continuous stretch of residues Gln317-Ser339 that was essential for FXII surface binding and activation, thrombin generation and coagulation. Peptides spanning these 23 residues competed with surface-induced FXII activation. Although FXII mutants lacking residues Gln317-Ser339 were susceptible to activation by plasmin and plasma kallikrein, they were ineffective in supporting arterial and venous thrombus formation in mice. Antibodies raised against the Gln317-Ser339 region induced FXII activation and triggered controllable contact activation in solution leading to thrombin generation by the intrinsic pathway of coagulation. The antibody-activated aPTT allows for standardization of particulate aPTT reagents and for sensitive monitoring of coagulation factors VIII, IX, XI.
Collapse
|
26
|
Zhang H, Pan D, Shen W. Genetic analysis of a pedigree with hereditary coagulation factor XII deficiency. Blood Coagul Fibrinolysis 2021; 32:406-410. [PMID: 33973893 DOI: 10.1097/mbc.0000000000001044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The aim of this study was to elucidate the molecular defects of a Chinese family with hereditary coagulation factor XII (FXII) deficiency. The FXII activity (FXII:C) and FXII antigen (FXII:Ag) levels were measured by clotting assay and ELISA, respectively. To identify mutations, the F12 gene sequencing was carried out. ClustalX-2.1-win and four online bioinformatics tools were applied to study the conservatism and harm of the mutation. The proband's FXII:C and FXII:Ag were 3 and 4%, respectively. Sequencing analysis revealed compound heterozygous mutations, including the deletion mutation (c.130delG) resulting in p.E26Sfs∗50 and the missense mutation (c.1561G>A) resulting in p.E502K. Bioinformatics indicated that mutations probably disrupt the function of the FXII protein. The c.130delG heterozygous deletion variation and the c.1561G>A heterozygous missense variation were responsible for the reduction of FXII:C in this family, of which c.130delG was first reported in the world.
Collapse
Affiliation(s)
- Haiyue Zhang
- Department of Clinical Laboratory, The First Hospital of Jiaxing & The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | | | | |
Collapse
|
27
|
Coagulation factor XII contributes to hemostasis when activated by soil in wounds. Blood Adv 2021; 4:1737-1745. [PMID: 32339233 DOI: 10.1182/bloodadvances.2019000425] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 03/24/2020] [Indexed: 01/14/2023] Open
Abstract
Bleeding is a common contributor to death and morbidity in animals and provides strong selective pressure for the coagulation system to optimize hemostasis for diverse environments. Although coagulation factor XII (FXII) is activated by nonbiologic surfaces, such as silicates, which leads to blood clotting in vitro, it is unclear whether FXII contributes to hemostasis in vivo. Humans and mice lacking FXII do not appear to bleed more from clean wounds than their counterparts with normal FXII levels. We tested the hypothesis that soil, a silicate-rich material abundant in the environment and wounds of terrestrial mammals, is a normal and potent activator of FXII and coagulation. Blood loss was compared between wild-type (WT) and FXII-knocked out (FXII-/-) mice after soil or exogenous tissue factor was applied to transected tails. The activation of FXII and other components of the coagulation and contact system was assessed with in vitro coagulation and enzyme assays. Soils were analyzed by time-of-flight secondary ionization mass spectrometry and dynamic light scattering. Soil reduced blood loss in WT mice, but not FXII-/- mice. Soil accelerated clotting of blood plasma from humans and mice in a FXII-dependent manner, but not plasma from a cetacean or a bird, which lack FXII. The procoagulant activity of 13 soils strongly correlated with the surface concentration of silicon, but only moderately correlated with the ζ potential. FXII augments coagulation in soil-contaminated wounds of terrestrial mammals, perhaps explaining why this protein has a seemingly minor role in hemostasis in clean wounds.
Collapse
|
28
|
Kallikrein directly interacts with and activates Factor IX, resulting in thrombin generation and fibrin formation independent of Factor XI. Proc Natl Acad Sci U S A 2021; 118:2014810118. [PMID: 33397811 PMCID: PMC7826336 DOI: 10.1073/pnas.2014810118] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Prekallikrein (PK) is a zymogen that is converted to kallikrein (PKa) by factor (F)XIIa. PK and FXII reciprocally activate each other; the resulting FXIIa initiates activation of the coagulation system via the cleavage of FXI to FXIa, which then activates FIX. This manuscript describes a novel high-affinity binding interaction between FIX(a) and PK(a) and reports that PKa can dose- and time-dependently activate FIX to generate FIXa, resulting in thrombin generation and clot formation independent of FXIa. Characterization of the kinetics of FIX activation reveal that PKa is a more significant activator of FIX than previously considered. This work highlights a new amendment to the coagulation cascade where PKa can directly activate FIX. Kallikrein (PKa), generated by activation of its precursor prekallikrein (PK), plays a role in the contact activation phase of coagulation and functions in the kallikrein-kinin system to generate bradykinin. The general dogma has been that the contribution of PKa to the coagulation cascade is dependent on its action on FXII. Recently this dogma has been challenged by studies in human plasma showing thrombin generation due to PKa activity on FIX and also by murine studies showing formation of FIXa-antithrombin complexes in FXI deficient mice. In this study, we demonstrate high-affinity binding interactions between PK(a) and FIX(a) using surface plasmon resonance and show that these interactions are likely to occur under physiological conditions. Furthermore, we directly demonstrate dose- and time-dependent cleavage of FIX by PKa in a purified system by sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis and chromogenic assays. By using normal pooled plasma and a range of coagulation factor-deficient plasmas, we show that this action of PKa on FIX not only results in thrombin generation, but also promotes fibrin formation in the absence of FXII or FXI. Comparison of the kinetics of either FXIa- or PKa-induced activation of FIX suggest that PKa could be a significant physiological activator of FIX. Our data indicate that the coagulation cascade needs to be redefined to indicate that PKa can directly activate FIX. The circumstances that drive PKa substrate specificity remain to be determined.
Collapse
|
29
|
Davoine C, Fillet M, Pochet L. Capillary electrophoresis as a fragment screening tool to cross-validate hits from chromogenic assay: Application to FXIIa. Talanta 2021; 226:122163. [PMID: 33676706 DOI: 10.1016/j.talanta.2021.122163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/20/2020] [Accepted: 01/25/2021] [Indexed: 10/22/2022]
Abstract
In this study, a partial-filling affinity capillary electrophoresis (pf-ACE) method was developed for the cross-validation of fragment hits revealed by chromogenic factor XIIa (FXIIa) assay. Chromogenic assay produces false positives, mainly due to spectrophotometric interferences and sample purity issues. pf-ACE was selected as counter-screening technology because of its separative character and the fact that the target does not have to be attached or tagged. The effects of protein plug length, applied voltage and composition of the running buffer were examined and optimized. Detection limit in terms of dissociation constant was estimated at 400 μM. The affinity evaluation was performed close to physiological conditions (pH 7.4, ionic strength 0.13 mol L-1) in a poly (ethylene oxide)-coated capillary of 75 μm internal diameter x 33 cm length with an applied voltage of 3 kV. This method uncovered chromogenic assay's false positives due to zinc contamination. Moreover, pf-ACE supported the evaluation of compounds absorbing at 405 nm.
Collapse
Affiliation(s)
- C Davoine
- Namur Medicine & Drug Innovation Center (NAMEDIC - NARILIS), University of Namur, Rue de Bruxelles 61, 5000, Namur, Belgium; Laboratory for the Analysis of Medicines (LAM), Department of Pharmacy, CIRM, University of Liege, Place du 20 Août 7, 4000, Liège, Belgium
| | - M Fillet
- Laboratory for the Analysis of Medicines (LAM), Department of Pharmacy, CIRM, University of Liege, Place du 20 Août 7, 4000, Liège, Belgium
| | - L Pochet
- Namur Medicine & Drug Innovation Center (NAMEDIC - NARILIS), University of Namur, Rue de Bruxelles 61, 5000, Namur, Belgium.
| |
Collapse
|
30
|
Abstract
Despite advances in anticoagulant therapy, thrombosis remains the leading cause of morbidity and mortality worldwide. Heparin and vitamin K antagonists (VKAs), the first anticoagulants to be used successfully for the prevention and treatment of thrombosis, are associated with a risk of bleeding. These agents target multiple coagulation factors. Thus, by activating antithrombin, heparin mainly inhibits factor Xa and thrombin, whereas VKAs lower the levels of the vitamin K-dependent clotting factors. Direct oral anticoagulants, which have replaced VKAs for many indications, inhibit only factor Xa or thrombin. Although the direct oral anticoagulants are associated with less bleeding than VKAs, bleeding remains their major side effect. Epidemiological and animal studies have identified factor XI as a target for potentially safer anticoagulant drugs because factor XI deficiency or inhibition protects against thrombosis and is associated with little or no bleeding. Several factor XI-directed strategies are currently under investigation. This article (1) reviews the rationale for the development of factor XI inhibitors, (2) identifies the agents in most advanced stages of development, (3) describes the results of completed clinical trials and provides a summary of those underway, and (4) highlights the opportunities and challenges for this next generation of anticoagulants.
Collapse
Affiliation(s)
- James C Fredenburgh
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jeffrey I Weitz
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Anticoagulation with vitamin-K antagonists or direct oral anticoagulants is associated with a significant risk of bleeding. There is a major effort underway to develop antithrombotic drugs that have a smaller impact on hemostasis. The plasma contact proteins factor XI (FXI) and factor XII (FXII) have drawn considerable interest because they contribute to thrombosis but have limited roles in hemostasis. Here, we discuss results of preclinical and clinical trials supporting the hypothesis that the contact system contributes to thromboembolic disease. RECENT FINDINGS Numerous compounds targeting FXI or FXII have shown antithrombotic properties in preclinical studies. In phase 2 studies, drugs-targeting FXI or its protease form FXIa compared favorably with standard care for venous thrombosis prophylaxis in patients undergoing knee replacement. While less work has been done with FXII inhibitors, they may be particularly useful for limiting thrombosis in situations where blood comes into contact with artificial surfaces of medical devices. SUMMARY Inhibitors of contact activation, and particularly of FXI, are showing promise for prevention of thromboembolic disease. Larger studies are required to establish their efficacy, and to establish that they are safer than current therapy from a bleeding standpoint.
Collapse
|
32
|
Deguchi H, Morla S, Griffin JH. Novel blood coagulation molecules: Skeletal muscle myosin and cardiac myosin. J Thromb Haemost 2021; 19:7-19. [PMID: 32920971 PMCID: PMC7819347 DOI: 10.1111/jth.15097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
Essentials Striated muscle myosins can promote prothrombin activation by FXa or FVa inactivation by APC. Cardiac myosin and skeletal muscle myosin are pro-hemostatic in murine tail cut bleeding models. Infused cardiac myosin exacerbates myocardial injury caused by myocardial ischemia reperfusion. Skeletal muscle myosin isoforms that circulate in human plasma can be grouped into 3 phenotypes. ABSTRACT: Two striated muscle myosins, namely skeletal muscle myosin (SkM) and cardiac myosin (CM), may potentially contribute to physiologic mechanisms for regulation of thrombosis and hemostasis. Thrombin is generated from activation of prothrombin by the prothrombinase (IIase) complex comprising factor Xa, factor Va, and Ca++ ions located on surfaces where these factors are assembled. We discovered that SkM and CM, which are abundant motor proteins in skeletal and cardiac muscles, can provide a surface for thrombin generation by the prothrombinase complex without any apparent requirement for phosphatidylserine or lipids. These myosins can also provide a surface that supports the inactivation of factor Va by activated protein C/protein S, resulting in negative feedback downregulation of thrombin generation. Although the physiologic significance of these reactions remains to be established for humans, substantive insights may be gleaned from murine studies. In mice, exogenously infused SkM and CM can promote hemostasis as they are capable of reducing tail cut bleeding. In a murine myocardial ischemia-reperfusion injury model, exogenously infused CM exacerbates myocardial infarction damage. Studies of human plasmas show that SkM antigen isoforms of different MWs circulate in human plasma, and they can be used to identify three plasma SkM phenotypes. A pilot clinical study showed that one SkM isoform pattern appeared to be linked to isolated pulmonary embolism. These discoveries enable multiple preclinical and clinical studies of SkM and CM, which should provide novel mechanistic insights with potential translational relevance for the roles of CM and SkM in the pathobiology of hemostasis and thrombosis.
Collapse
Affiliation(s)
- Hiroshi Deguchi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Shravan Morla
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Division of Hematology, Department of Medicine, University of California, San Diego, CA, USA
| |
Collapse
|
33
|
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: 45] [Impact Index Per Article: 11.3] [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
|
34
|
Factor XII/XIIa inhibitors: Their discovery, development, and potential indications. Eur J Med Chem 2020; 208:112753. [DOI: 10.1016/j.ejmech.2020.112753] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022]
|
35
|
Aymonnier K, Kawecki C, Arocas V, Boulaftali Y, Bouton MC. Serpins, New Therapeutic Targets for Hemophilia. Thromb Haemost 2020; 121:261-269. [PMID: 32987444 DOI: 10.1055/s-0040-1716751] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hemostasis is a tightly regulated process characterized by a finely tuned balance between procoagulant and anticoagulant systems. Among inherited hemostatic conditions, hemophilia is one of the most well-known bleeding disorders. Hemophilia A (HA) and B (HB) are due to deficiencies in coagulation factor VIII (FVIII) or FIX, respectively, leading to unwanted bleeding. Until recently, hemophilia treatment has consisted of prophylactic replacement therapy using plasma-derived or recombinant FVIII in cases of HA or FIX in cases of HB. Because FVIII and FIX deficiencies lead to an imbalance between procoagulant and anticoagulant systems, a recent upcoming strategy implies blocking of endogenous anticoagulant proteins to compensate for the procoagulant factor deficit, thus restoring hemostatic equilibrium. Important physiological proteins of the anticoagulant pathways belong to the serpin (serine protease inhibitor) family and, recently, different experimental and clinical studies have demonstrated that targeting natural serpins could decrease bleeding in hemophilia. Here, we aim to review the different, recent studies demonstrating that blocking serpins such as antithrombin, protein Z-dependent protease inhibitor, and protease nexin-1 or modifying a serpin like α1-antitrypsin could rebalance coagulation in hemophilia. Furthermore, we underline the potential therapeutic use of serpins for the treatment of hemophilia.
Collapse
Affiliation(s)
- Karen Aymonnier
- INSERM U1148-LVTS, Université de Paris, Paris, France.,CHU Xavier Bichat, Paris, France
| | - Charlotte Kawecki
- INSERM U1148-LVTS, Université de Paris, Paris, France.,INSERM U1176-HITh, Université Paris-Sud (Université Paris-Saclay), Le Kremlin-Bicêtre, France
| | - Véronique Arocas
- INSERM U1148-LVTS, Université de Paris, Paris, France.,CHU Xavier Bichat, Paris, France
| | - Yacine Boulaftali
- INSERM U1148-LVTS, Université de Paris, Paris, France.,CHU Xavier Bichat, Paris, France
| | | |
Collapse
|
36
|
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: 0.4] [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
|
37
|
Mohammed BM, Monroe DM, Gailani D. Mouse models of hemostasis. Platelets 2020; 31:417-422. [PMID: 31992118 PMCID: PMC7244364 DOI: 10.1080/09537104.2020.1719056] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 02/02/2023]
Abstract
Hemostasis is the normal process that produces a blood clot at a site of vascular injury. Mice are widely used to study hemostasis and abnormalities of blood coagulation because their hemostatic system is similar in most respects to that of humans, and their genomes can be easily manipulated to create models of inherited human coagulation disorders. Two of the most widely used techniques for assessing hemostasis in mice are the tail bleeding time (TBT) and saphenous vein bleeding (SVB) models. Here we discuss the use of these methods in the evaluation of hemostasis, and the advantages and limits of using mice as surrogates for studying hemostasis in humans.
Collapse
Affiliation(s)
- Bassem M. Mohammed
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
- Department of Pathology and Immunology, Washington University, St. Louis, MO
| | - Dougald M. Monroe
- UNC Blood Research Center and Hematology/Oncology, University of North Carolina, Chapel Hill, NC
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
| |
Collapse
|
38
|
Noubouossie DF, Henderson MW, Mooberry M, Ilich A, Ellsworth P, Piegore M, Skinner SC, Pawlinski R, Welsby I, Renné T, Hoffman M, Monroe DM, Key NS. Red blood cell microvesicles activate the contact system, leading to factor IX activation via 2 independent pathways. Blood 2020; 135:755-765. [PMID: 31971571 PMCID: PMC7059516 DOI: 10.1182/blood.2019001643] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 01/08/2020] [Indexed: 01/01/2023] Open
Abstract
Storage lesion-induced, red cell-derived microvesicles (RBC-MVs) propagate coagulation by supporting the assembly of the prothrombinase complex. It has also been reported that RBC-MVs initiate coagulation via the intrinsic pathway. To elucidate the mechanism(s) of RBC-MV-induced coagulation activation, the ability of storage lesion-induced RBC-MVs to activate each zymogen of the intrinsic pathway was assessed in a buffer system. Simultaneously, the thrombin generation (TG) assay was used to assess their ability to initiate coagulation in plasma. RBC-MVs directly activated factor XII (FXII) or prekallikrein, but not FXI or FIX. RBC-MVs initiated TG in normal pooled plasma and in FXII- or FXI-deficient plasma, but not in FIX-deficient plasma, suggesting an alternate pathway that bypasses both FXII and FXI. Interestingly, RBC-MVs generated FIXa in a prekallikrein-dependent manner. Similarly, purified kallikrein activated FIX in buffer and initiated TG in normal pooled plasma, as well as FXII- or FXI-deficient plasma, but not FIX-deficient plasma. Dual inhibition of FXIIa by corn trypsin inhibitor and kallikrein by soybean trypsin inhibitor was necessary for abolishing RBC-MV-induced TG in normal pooled plasma, whereas kallikrein inhibition alone was sufficient to abolish TG in FXII- or FXI-deficient plasma. Heating RBC-MVs at 60°C for 15 minutes or pretreatment with trypsin abolished TG, suggesting the presence of MV-associated proteins that are essential for contact activation. In summary, RBC-MVs activate both FXII and prekallikrein, leading to FIX activation by 2 independent pathways: the classic FXIIa-FXI-FIX pathway and direct kallikrein activation of FIX. These data suggest novel mechanisms by which RBC transfusion mediates inflammatory and/or thrombotic outcomes.
Collapse
Affiliation(s)
| | - Michael W Henderson
- UNC Blood Research Center, and
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Anton Ilich
- Department of Medicine
- UNC Blood Research Center, and
| | - Patrick Ellsworth
- Department of Medicine
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Mark Piegore
- Department of Medicine
- UNC Blood Research Center, and
| | - Sarah C Skinner
- Department of Medicine
- UNC Blood Research Center, and
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Ian Welsby
- Department of Anesthesiology, Duke University, Durham, NC
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; and
| | - Maureane Hoffman
- Department of Pathology, Veteran Affairs Medical Center, Durham, NC
| | | | - Nigel S Key
- Department of Medicine
- UNC Blood Research Center, and
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
39
|
Wallisch M, Lorentz CU, Lakshmanan HHS, Johnson J, Carris MR, Puy C, Gailani D, Hinds MT, McCarty OJT, Gruber A, Tucker EI. Antibody inhibition of contact factor XII reduces platelet deposition in a model of extracorporeal membrane oxygenator perfusion in nonhuman primates. Res Pract Thromb Haemost 2020; 4:205-216. [PMID: 32110750 PMCID: PMC7040549 DOI: 10.1002/rth2.12309] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/20/2019] [Accepted: 12/27/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The contact factor XII (FXII) activates upon contact with a variety of charged surfaces. Activated FXII (FXIIa) activates factor XI, which activates factor IX, resulting in thrombin generation, platelet activation, and fibrin formation. In both in vitro and in vivo rabbit models, components of medical devices, including extracorporeal oxygenators, are known to incite fibrin formation in a FXII-dependent manner. Since FXII has no known role in hemostasis and its inhibition is therefore likely a safe antithrombotic approach, we investigated whether FXII inhibition also reduces accumulation of platelets in extracorporeal oxygenators. OBJECTIVES We aimed to determine the effect of FXII inhibition on platelet deposition in perfused extracorporeal membrane oxygenators in nonhuman primates. METHODS A potent FXII neutralizing monoclonal antibody, 5C12, was administered intravenously to block contact activation in baboons. Extracorporeal membrane oxygenators were temporarily deployed into chronic arteriovenous access shunts. Radiolabeled platelet deposition in oxygenators was quantified in real time using gamma camera imaging. Biochemical assays were performed to characterize the method of action of 5C12. RESULTS The anti-FXII monoclonal antibody 5C12 recognized both the alpha and beta forms of human and baboon FXII by binding to the protease-containing domain, and inhibited FXIIa activity. Administration of 5C12 to baboons reduced platelet deposition and fibrin formation in the extracorporeal membrane oxygenators, in both the presence and absence of systemic low-dose unfractionated heparin. The antiplatelet dose of 5C12 did not cause measurable increases in template bleeding times in baboons. CONCLUSIONS FXII represents a possible therapeutic and safe target for reducing platelet deposition and fibrin formation during medical interventions including extracorporeal membrane oxygenation.
Collapse
Affiliation(s)
- Michael Wallisch
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
- Aronora, Inc.PortlandORUSA
| | - Christina U. Lorentz
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
- Aronora, Inc.PortlandORUSA
| | | | - Jennifer Johnson
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
| | - Marschelle R. Carris
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
- Aronora, Inc.PortlandORUSA
| | - Cristina Puy
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
| | - David Gailani
- Department of Pathology, Microbiology, and ImmunologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Monica T. Hinds
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
| | - Owen J. T. McCarty
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
- Division of Hematology & Medical OncologyDepartment of MedicineOregon Health & Science UniversityPortlandORUSA
| | - András Gruber
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
- Aronora, Inc.PortlandORUSA
- Division of Hematology & Medical OncologyDepartment of MedicineOregon Health & Science UniversityPortlandORUSA
| | - Erik I. Tucker
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandORUSA
- Aronora, Inc.PortlandORUSA
| |
Collapse
|
40
|
Visser M, van Oerle R, ten Cate H, Laux V, Mackman N, Heitmeier S, Spronk HM. Plasma Kallikrein Contributes to Coagulation in the Absence of Factor XI by Activating Factor IX. Arterioscler Thromb Vasc Biol 2020; 40:103-111. [DOI: 10.1161/atvbaha.119.313503] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objectives:
FXIa (factor XIa) induces clot formation, and human congenital FXI deficiency protects against venous thromboembolism and stroke. In contrast, the role of FXI in hemostasis is rather small, especially compared with FIX deficiency. Little is known about the cause of the difference in phenotypes associated with FIX deficiency and FXI deficiency. We speculated that activation of FIX via the intrinsic coagulation is not solely dependent on FXI(a; activated FXI) and aimed at identifying an FXI-independent FIX activation pathway.
Approach and Results:
We observed that ellagic acid and long-chain polyphosphates activated the coagulation system in FXI-deficient plasma, as could be demonstrated by measurement of thrombin generation, FIXa-AT (antithrombin), and FXa-AT complex levels, suggesting an FXI bypass route of FIX activation. Addition of a specific PKa (plasma kallikrein) inhibitor to FXI-deficient plasma decreased thrombin generation, prolonged activated partial thromboplastin time, and diminished FIXa-AT and FXa-AT complex formation, indicating that PKa plays a role in the FXI bypass route of FIX activation. In addition, FIXa-AT complex formation was significantly increased in
F11
−/−
mice treated with ellagic acid or long-chain polyphosphates compared with controls and this increase was significantly reduced by inhibition of PKa.
Conclusions:
We demonstrated that activation of FXII leads to thrombin generation via FIX activation by PKa in the absence of FXI. These findings may, in part, explain the different phenotypes associated with FIX and FXI deficiencies.
Collapse
Affiliation(s)
- Mayken Visser
- From the Bayer AG, Cardiovascular Research, Wuppertal, Germany (M.V., V.L., S.H.)
- Laboratory for Clinical Thrombosis and Haemostasis, Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (M.V., R.v.O., H.t.C., H.M.H.S.)
| | - René van Oerle
- Laboratory for Clinical Thrombosis and Haemostasis, Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (M.V., R.v.O., H.t.C., H.M.H.S.)
| | - Hugo ten Cate
- Laboratory for Clinical Thrombosis and Haemostasis, Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (M.V., R.v.O., H.t.C., H.M.H.S.)
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Germany (H.t.C.)
| | - Volker Laux
- From the Bayer AG, Cardiovascular Research, Wuppertal, Germany (M.V., V.L., S.H.)
| | - Nigel Mackman
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Stefan Heitmeier
- From the Bayer AG, Cardiovascular Research, Wuppertal, Germany (M.V., V.L., S.H.)
| | - Henri M.H. Spronk
- Laboratory for Clinical Thrombosis and Haemostasis, Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (M.V., R.v.O., H.t.C., H.M.H.S.)
| |
Collapse
|
41
|
From multi-target anticoagulants to DOACs, and intrinsic coagulation factor inhibitors. Blood Rev 2020; 39:100615. [DOI: 10.1016/j.blre.2019.100615] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 08/08/2019] [Accepted: 08/27/2019] [Indexed: 01/10/2023]
|
42
|
Al-Horani RA. Factor XI(a) inhibitors for thrombosis: an updated patent review (2016-present). Expert Opin Ther Pat 2019; 30:39-55. [PMID: 31847619 DOI: 10.1080/13543776.2020.1705783] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Anticoagulation without bleeding is an ideal goal in treating thrombosis, however, this goal has not been achieved. All current anticoagulants are associated with significant bleeding which limits their safe use. Genetic and pharmacological findings indicate that factor XIa is a key player in thrombosis, yet it is a relatively marginal one in hemostasis. Thus, factor XIa and its zymogen offer a unique opportunity to develop anticoagulants with low bleeding risk.Areas covered: A survey of patent literature has retrieved more than 50 patents on the discovery of novel therapeutics targeting factor XI(a) since 2016. Small molecules, monoclonal antibodies, oligonucleotides, and polypeptides have been developed to inhibit factor XI(a). Many inhibitors are in early development and few have been evaluated in clinical trials.Expert opinion: Factor XI(a) is being actively pursued as a drug target for the development of effective and safer anticoagulants. Although many patents claiming factor XI(a) inhibitors were filed prior to 2016, recent literature reveals a moderately declining trend. Nevertheless, more agents have entered different levels of clinical trials. These agents exploit diverse mechanistic strategies for inhibition. Although further development is warranted, reaching one or more of these agents to the clinic will transform the anticoagulation therapy.
Collapse
Affiliation(s)
- Rami A Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, USA
| |
Collapse
|
43
|
Mohammed BM, Cheng Q, Matafonov A, Verhamme IM, Emsley J, McCrae KR, McCarty OJT, Gruber A, Gailani D. A non-circulating pool of factor XI associated with glycosaminoglycans in mice. J Thromb Haemost 2019; 17:1449-1460. [PMID: 31125187 PMCID: PMC6768408 DOI: 10.1111/jth.14494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 05/20/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The homologous plasma proteins prekallikrein and factor XI (FXI) circulate as complexes with high molecular weight kininogen. Although evidence supports an interaction between the prekallikrein-kininogen complexes and vascular endothelium, there is conflicting information regarding FXI binding to endothelium. OBJECTIVE To study the interaction between FXI and blood vessels in mice. METHODS C57Bl/6 wild-type or F11-/- mice in which variants of FXI were expressed by hydrodynamic tail vein injection, received intravenous infusions of saline, heparin, polyphosphates, protamine, or enzymes that digest glycosaminoglycans (GAGs). Blood was collected after infusion and plasma was analyzed by western blot for FXI. RESULTS AND CONCLUSIONS Plasma FXI increased 5- to 10-fold in wild-type mice after infusion of heparin, polyphosphates, protamine, or GAG-digesting enzymes, but not saline. Similar treatments resulted in a much smaller change in plasma FXI levels in rats, and infusions of large boluses of heparin did not change FXI levels appreciably in baboons or humans. The releasable FXI fraction was reconstituted in F11-/- mice by expressing murine FXI, but not human FXI. We identified a cluster of basic residues on the apple 4 domain of mouse FXI that is not present in other species. Replacing the basic residues with alanine prevented the interaction of mouse FXI with blood vessels, whereas introducing the basic residues into human FXI allowed it to bind to blood vessels. Most FXI in mice is noncovalently associated with GAGs on blood vessel endothelium and does not circulate in plasma.
Collapse
Affiliation(s)
- Bassem M. Mohammed
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
- Department of Clinical Pharmacy, School of Pharmacy, Cairo University, Cairo, Egypt
| | - Qiufang Cheng
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Anton Matafonov
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Ingrid M. Verhamme
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Jonas Emsley
- Center for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Keith R. McCrae
- Department of Hematology and Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Owen J. T. McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Division of Hematology/ Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Andras Gruber
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Division of Hematology/ Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
44
|
Raghunathan V, Zilberman‐Rudenko J, Olson SR, Lupu F, McCarty OJT, Shatzel JJ. The contact pathway and sepsis. Res Pract Thromb Haemost 2019; 3:331-339. [PMID: 31294319 PMCID: PMC6611366 DOI: 10.1002/rth2.12217] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/15/2019] [Indexed: 12/12/2022] Open
Abstract
The contact pathway factors XI (FXI) and XII (FXII) have been demonstrated to be largely dispensable for hemostasis, as their absence results in a mild to absent bleeding diathesis. A growing body of literature, however, suggests that the contact pathway contributes to the pathologic host response to certain infectious organisms that produces the often-fatal syndrome known as sepsis. The contact pathway factors serve as a central node connecting inflammation to coagulation, and may offer a potentially safe therapeutic target to mitigate the morbidity and mortality associated with sepsis. Herein, we summarize published in vivo and in vitro data that have explored the roles of the contact pathway in sepsis, and discuss potential clinical applications of novel FXI- and FXII-inhibiting drugs currently under investigation.
Collapse
Affiliation(s)
- Vikram Raghunathan
- Division of Hematology‐Medical OncologyKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | | | - Sven R. Olson
- Division of Hematology‐Medical OncologyKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
| | - Florea Lupu
- Cardiovascular Biology Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Owen J. T. McCarty
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
| | - Joseph J. Shatzel
- Division of Hematology‐Medical OncologyKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
45
|
Li X, Wood JP. Studies into prekallikrein activation pave the way for new avenues of antithrombotic research. J Thromb Haemost 2019; 17:717-719. [PMID: 30916449 DOI: 10.1111/jth.14435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/21/2019] [Indexed: 01/25/2023]
Affiliation(s)
- Xian Li
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Jeremy P Wood
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, Kentucky, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
46
|
|